Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 76
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Proc Natl Acad Sci U S A ; 118(4)2021 01 26.
Artículo en Inglés | MEDLINE | ID: mdl-33468664

RESUMEN

We have shown previously that phosphorylation of Mdm2 by ATM and c-Abl regulates Mdm2-p53 signaling and alters the effects of DNA damage in mice, including bone marrow failure and tumorigenesis induced by ionizing radiation. Here, we examine the physiological effects of Mdm2 phosphorylation by Akt, another DNA damage effector kinase. Surprisingly, Akt phosphorylation of Mdm2 does not alter the p53-mediated effects of ionizing radiation in cells or mice but regulates the p53 response to oxidative stress. Akt phosphorylation of Mdm2 serine residue 183 increases nuclear Mdm2 stability, decreases p53 levels, and prevents senescence in primary cells exposed to reactive oxidative species (ROS). Using multiple mouse models of ROS-induced cancer, we show that Mdm2 phosphorylation by Akt reduces senescence to promote KrasG12D-driven lung cancers and carcinogen-induced papilloma and hepatocellular carcinomas. Collectively, we document a unique physiologic role for Akt-Mdm2-p53 signaling in regulating cell growth and tumorigenesis in response to oxidative stress.


Asunto(s)
Neoplasias Inducidas por Radiación/genética , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-mdm2/genética , Proteínas Proto-Oncogénicas p21(ras)/genética , Proteína p53 Supresora de Tumor/genética , Animales , Apoptosis/efectos de los fármacos , Apoptosis/efectos de la radiación , Carcinogénesis/efectos de los fármacos , Carcinogénesis/genética , Carcinogénesis/efectos de la radiación , Carcinógenos/toxicidad , Carcinoma Hepatocelular/inducido químicamente , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/virología , Proliferación Celular/genética , Senescencia Celular/efectos de los fármacos , Senescencia Celular/efectos de la radiación , Daño del ADN/genética , Daño del ADN/efectos de la radiación , Modelos Animales de Enfermedad , Humanos , Neoplasias Hepáticas/inducido químicamente , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/patología , Neoplasias Hepáticas/virología , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patología , Ratones , Neoplasias Inducidas por Radiación/patología , Estrés Oxidativo/efectos de los fármacos , Estrés Oxidativo/efectos de la radiación , Papillomaviridae/patogenicidad , Fosforilación/efectos de los fármacos , Fosforilación/efectos de la radiación , Radiación Ionizante , Especies Reactivas de Oxígeno/metabolismo
2.
Nature ; 511(7507): 86-9, 2014 Jul 03.
Artículo en Inglés | MEDLINE | ID: mdl-24870238

RESUMEN

In female mice, two forms of X-chromosome inactivation (XCI) ensure the selective silencing of female sex chromosomes during mouse embryogenesis. Beginning at the four-cell stage, imprinted XCI (iXCI) exclusively silences the paternal X chromosome. Later, around implantation, epiblast cells of the inner cell mass that give rise to the embryo reactivate the paternal X chromosome and undergo a random form of XCI (rXCI). Xist, a long non-coding RNA crucial for both forms of XCI, is activated by the ubiquitin ligase RLIM (also known as Rnf12). Although RLIM is required for triggering iXCI in mice, its importance for rXCI has been controversial. Here we show that RLIM levels are downregulated in embryonic cells undergoing rXCI. Using mouse genetics we demonstrate that female cells lacking RLIM from pre-implantation stages onwards show hallmarks of XCI, including Xist clouds and H3K27me3 foci, and have full embryogenic potential. These results provide evidence that RLIM is dispensable for rXCI, indicating that in mice an RLIM-independent mechanism activates Xist in the embryo proper.


Asunto(s)
Estratos Germinativos/embriología , Estratos Germinativos/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Inactivación del Cromosoma X/genética , Animales , Regulación hacia Abajo , Implantación del Embrión , Embrión de Mamíferos/embriología , Embrión de Mamíferos/metabolismo , Femenino , Histonas/química , Histonas/metabolismo , Hibridación Fluorescente in Situ , Lisina/metabolismo , Metilación , Ratones , Ratones Noqueados , ARN Largo no Codificante/genética , Ubiquitina-Proteína Ligasas/genética
3.
Proc Natl Acad Sci U S A ; 113(52): 15024-15029, 2016 12 27.
Artículo en Inglés | MEDLINE | ID: mdl-27956626

RESUMEN

The p53 tumor suppressor acts as a guardian of the genome by preventing the propagation of DNA damage-induced breaks and mutations to subsequent generations of cells. We have previously shown that phosphorylation of the Mdm2 oncoprotein at Ser394 by the ATM kinase is required for robust p53 stabilization and activation in cells treated with ionizing radiation, and that loss of Mdm2 Ser394 phosphorylation leads to spontaneous tumorigenesis and radioresistance in Mdm2S394A mice. Previous in vitro data indicate that the c-Abl kinase phosphorylates Mdm2 at the neighboring residue (Tyr393) in response to DNA damage to regulate p53-dependent apoptosis. In this present study, we have generated an Mdm2 mutant mouse (Mdm2Y393F) to determine whether c-Abl phosphorylation of Mdm2 regulates the p53-mediated DNA damage response or p53 tumor suppression in vivo. The Mdm2Y393F mice develop accelerated spontaneous and oncogene-induced tumors, yet display no defects in p53 stabilization and activity following acute genotoxic stress. Although apoptosis is unaltered in these mice, they recover more rapidly from radiation-induced bone marrow ablation and are more resistant to whole-body radiation-induced lethality. These data reveal an in vivo role for c-Abl phosphorylation of Mdm2 in regulation of p53 tumor suppression and bone marrow failure. However, c-Abl phosphorylation of Mdm2 Tyr393 appears to play a lesser role in governing Mdm2-p53 signaling than ATM phosphorylation of Mdm2 Ser394. Furthermore, the effects of these phosphorylation events on p53 regulation are not additive, as Mdm2Y393F/S394A mice and Mdm2S394A mice display similar phenotypes.


Asunto(s)
Regulación Neoplásica de la Expresión Génica , Neoplasias/metabolismo , Proteínas Proto-Oncogénicas c-abl/metabolismo , Proteínas Proto-Oncogénicas c-mdm2/química , Tolerancia a Radiación , Proteína p53 Supresora de Tumor/metabolismo , Alelos , Animales , Apoptosis , Daño del ADN , Exones , Femenino , Fibroblastos/metabolismo , Perfilación de la Expresión Génica , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Neoplasias/genética , Neoplasias/radioterapia , Fosforilación , Procesamiento Proteico-Postraduccional , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Proto-Oncogénicas c-mdm2/metabolismo , Transducción de Señal
4.
Dev Biol ; 419(2): 311-320, 2016 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-27609454

RESUMEN

Fidelity of histone gene expression is important for normal cell growth and differentiation that is stringently controlled during development but is compromised during tumorigenesis. Efficient production of histones for packaging newly replicated DNA is particularly important for proper cell division and epigenetic control during the initial pre-implantation stages of embryonic development. Here, we addressed the unresolved question of when the machinery for histone gene transcription is activated in the developing zygote to accommodate temporal demands for histone gene expression. We examined induction of Histone Nuclear Factor P (HINFP), the only known transcription factor required for histone H4 gene expression, that binds directly to a unique H4 promoter-specific element to regulate histone H4 transcription. We show that Hinfp gene transcripts are stored in oocytes and maternally transmitted to the zygote. Transcripts from the paternal Hinfp gene, which reflect induction of zygotic gene expression, are apparent at the 4- to 8-cell stage, when most maternal mRNA pools are depleted. Loss of Hinfp expression due to gene ablation reduces cell numbers in E3.5 stage embryos and compromises implantation. Reduced cell proliferation is attributable to severe reduction in histone mRNA levels accompanied by reduced cell survival and genomic damage as measured by cleaved Caspase 3 and phospho-H2AX staining, respectively. We conclude that transmission of maternal Hinfp transcripts and zygotic activation of the Hinfp gene together are necessary to control H4 gene expression in early pre-implantation embryos in order to support normal embryonic development.


Asunto(s)
Desarrollo Embrionario , Regulación del Desarrollo de la Expresión Génica , Histonas/biosíntesis , ARN Mensajero Almacenado/genética , Proteínas Represoras/fisiología , Cigoto/metabolismo , Animales , Blastocisto/fisiología , Caspasa 3/metabolismo , Implantación del Embrión/fisiología , Desarrollo Embrionario/fisiología , Femenino , Genes Reporteros , Histonas/genética , Histonas/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Regiones Promotoras Genéticas , ARN Mensajero/metabolismo , Proteínas Represoras/deficiencia , Proteínas Represoras/genética
5.
Nature ; 467(7318): 977-81, 2010 Oct 21.
Artículo en Inglés | MEDLINE | ID: mdl-20962847

RESUMEN

Two forms of X-chromosome inactivation (XCI) ensure the selective silencing of female sex chromosomes during mouse embryogenesis. Imprinted XCI begins with the detection of Xist RNA expression on the paternal X chromosome (Xp) at about the four-cell stage of embryonic development. In the embryonic tissues of the inner cell mass, a random form of XCI occurs in blastocysts that inactivates either Xp or the maternal X chromosome (Xm). Both forms of XCI require the non-coding Xist RNA that coats the inactive X chromosome from which it is expressed. Xist has crucial functions in the silencing of X-linked genes, including Rnf12 (refs 3, 4) encoding the ubiquitin ligase RLIM (RING finger LIM-domain-interacting protein). Here we show, by targeting a conditional knockout of Rnf12 to oocytes where RLIM accumulates to high levels, that the maternal transmission of the mutant X chromosome (Δm) leads to lethality in female embryos as a result of defective imprinted XCI. We provide evidence that in Δm female embryos the initial formation of Xist clouds and Xp silencing are inhibited. In contrast, embryonic stem cells lacking RLIM are able to form Xist clouds and silence at least some X-linked genes during random XCI. These results assign crucial functions to the maternal deposit of Rnf12/RLIM for the initiation of imprinted XCI.


Asunto(s)
Cromosomas de los Mamíferos/genética , Impresión Genómica , Madres , Proteínas Represoras/metabolismo , Inactivación del Cromosoma X/genética , Cromosoma X/genética , Animales , Animales Congénicos , Blastocisto/metabolismo , Línea Celular , Pérdida del Embrión/genética , Padre , Femenino , Silenciador del Gen , Masculino , Ratones , Ratones Transgénicos , ARN Largo no Codificante , ARN no Traducido/genética , Proteínas Represoras/deficiencia , Proteínas Represoras/genética , Ubiquitina-Proteína Ligasas
6.
Cancer Cell ; 12(4): 342-54, 2007 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-17936559

RESUMEN

The tumor suppressor p53 is a transcription factor that responds to cellular stresses by initiating cell cycle arrest or apoptosis. One transcriptional target of p53 is Mdm2, an E3 ubiquitin ligase that interacts with p53 to promote its proteasomal degradation in a negative feedback regulatory loop. Here we show that the wild-type p53-induced phosphatase 1 (Wip1), or PPM1D, downregulates p53 protein levels by stabilizing Mdm2 and facilitating its access to p53. Wip1 interacts with and dephosphorylates Mdm2 at serine 395, a site phosphorylated by the ATM kinase. Dephosphorylated Mdm2 has increased stability and affinity for p53, facilitating p53 ubiquitination and degradation. Thus, Wip1 acts as a gatekeeper in the Mdm2-p53 regulatory loop by stabilizing Mdm2 and promoting Mdm2-mediated proteolysis of p53.


Asunto(s)
Fibroblastos/metabolismo , Osteosarcoma/metabolismo , Fosfoproteínas Fosfatasas/metabolismo , Proteínas Proto-Oncogénicas c-mdm2/metabolismo , Transducción de Señal , Proteína p53 Supresora de Tumor/metabolismo , Animales , Proteínas de la Ataxia Telangiectasia Mutada , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Línea Celular Tumoral , Daño del ADN , Proteínas de Unión al ADN/deficiencia , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Fibroblastos/enzimología , Fibroblastos/efectos de la radiación , Homeostasis , Humanos , Ratones , Ratones Noqueados , Mutación , Osteosarcoma/enzimología , Osteosarcoma/genética , Fosfoproteínas Fosfatasas/deficiencia , Fosfoproteínas Fosfatasas/genética , Fosforilación , Complejo de la Endopetidasa Proteasomal/metabolismo , Proteína Fosfatasa 2C , Proteínas Serina-Treonina Quinasas/deficiencia , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-mdm2/genética , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Serina/metabolismo , Transducción de Señal/efectos de la radiación , Factores de Tiempo , Transcripción Genética , Transfección , Proteína p53 Supresora de Tumor/genética , Proteínas Supresoras de Tumor/deficiencia , Proteínas Supresoras de Tumor/genética , Proteínas Supresoras de Tumor/metabolismo , Ubiquitina/metabolismo , Ubiquitina Tiolesterasa/metabolismo , Peptidasa Específica de Ubiquitina 7
7.
J Biol Chem ; 288(29): 21307-21319, 2013 Jul 19.
Artículo en Inglés | MEDLINE | ID: mdl-23720736

RESUMEN

Osteosarcoma (OS) is a primary bone tumor that is most prevalent during adolescence. RUNX2, which stimulates differentiation and suppresses proliferation of osteoblasts, is deregulated in OS. Here, we define pathological roles of RUNX2 in the etiology of OS and mechanisms by which RUNX2 expression is stimulated. RUNX2 is often highly expressed in human OS biopsies and cell lines. Small interference RNA-mediated depletion of RUNX2 inhibits growth of U2OS OS cells. RUNX2 levels are inversely linked to loss of p53 (which predisposes to OS) in distinct OS cell lines and osteoblasts. RUNX2 protein levels decrease upon stabilization of p53 with the MDM2 inhibitor Nutlin-3. Elevated RUNX2 protein expression is post-transcriptionally regulated and directly linked to diminished expression of several validated RUNX2 targeting microRNAs in human OS cells compared with mesenchymal progenitor cells. The p53-dependent miR-34c is the most significantly down-regulated RUNX2 targeting microRNAs in OS. Exogenous supplementation of miR-34c markedly decreases RUNX2 protein levels, whereas 3'-UTR reporter assays establish RUNX2 as a direct target of miR-34c in OS cells. Importantly, Nutlin-3-mediated stabilization of p53 increases expression of miR-34c and decreases RUNX2. Thus, a novel p53-miR-34c-RUNX2 network controls cell growth of osseous cells and is compromised in OS.


Asunto(s)
Neoplasias Óseas/metabolismo , Subunidad alfa 1 del Factor de Unión al Sitio Principal/metabolismo , MicroARNs/metabolismo , Osteosarcoma/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Animales , Neoplasias Óseas/genética , Neoplasias Óseas/patología , Ciclo Celular/genética , Ciclo Celular/efectos de la radiación , Línea Celular Tumoral , Proliferación Celular/efectos de la radiación , Subunidad alfa 1 del Factor de Unión al Sitio Principal/genética , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Daño del ADN , Regulación hacia Abajo/genética , Regulación hacia Abajo/efectos de la radiación , Rayos gamma , Regulación Neoplásica de la Expresión Génica/efectos de la radiación , Humanos , Ratones , Osteosarcoma/genética , Osteosarcoma/patología , Estabilidad Proteica/efectos de la radiación , ARN Mensajero/genética , ARN Mensajero/metabolismo , Proteína p14ARF Supresora de Tumor/metabolismo , Proteína p53 Supresora de Tumor/deficiencia
8.
Dev Biol ; 371(1): 77-85, 2012 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-22939930

RESUMEN

Aurora A is a mitotic kinase essential for cell proliferation. In mice, ablation of Aurora A results in mitotic arrest and pre-implantation lethality, preventing studies at later stages of development. Here we report the effects of Aurora A ablation on embryo patterning at early post-implantation stages. Inactivation of Aurora A in the epiblast or visceral endoderm layers of the conceptus leads to apoptosis and inhibition of embryo growth, causing lethality and resorption at approximately E9.5. The effects on embryo patterning, however, depend on the tissue affected by the mutation. Embryos with an epiblast ablation of Aurora A properly establish the anteroposterior axis but fail to progress through gastrulation. In contrast, mutation of Aurora A in the visceral endoderm, leads to posteriorization of the conceptus or failure to elongate the anteroposterior axis. Injection of ES cells into Aurora A epiblast knockout blastocysts reconstitutes embryonic development to E9.5, indicating that the extra-embryonic tissues in these mutant embryos can sustain development to organogenesis stages. Our results reveal new ways to induce apoptosis and to ablate cells in a tissue-specific manner in vivo. Moreover, they show that epiblast-ablated embryos can be used to test the potency of stem cells.


Asunto(s)
Tipificación del Cuerpo/genética , Embrión de Mamíferos/embriología , Endodermo/embriología , Estratos Germinativos/embriología , Proteínas Serina-Treonina Quinasas/deficiencia , Animales , Apoptosis/genética , Aurora Quinasa A , Aurora Quinasas , Cartilla de ADN/genética , Células Madre Embrionarias/metabolismo , Técnica del Anticuerpo Fluorescente , Técnicas de Inactivación de Genes , Hibridación in Situ , Ratones , Proteínas Serina-Treonina Quinasas/genética , beta-Galactosidasa
9.
Circulation ; 125(22): 2762-71, 2012 Jun 05.
Artículo en Inglés | MEDLINE | ID: mdl-22550155

RESUMEN

BACKGROUND: High shear force critically regulates platelet adhesion and thrombus formation during ischemic vascular events. To identify genetic factors that influence platelet thrombus formation under high shear stress, we performed a genome-wide association study and confirmatory experiments in human and animal platelets. METHODS AND RESULTS: Closure times in the shear-dependent platelet function analyzer (PFA)-100 were measured on healthy, nondiabetic European Americans (n=125) and blacks (n=116). A genome-wide association (P<5×10(-8)) was identified with 2 single-nucleotide polymorphisms within the SVIL gene (chromosome 10p11.23) in African Americans but not European Americans. Microarray analyses of human platelet RNA demonstrated the presence of SVIL isoform 1 (supervillin) but not muscle-specific isoforms 2 and 3 (archvillin, SmAV). SVIL mRNA levels were associated with SVIL genotypes (P≤0.02) and were inversely correlated with PFA-100 closure times (P<0.04) and platelet volume (P<0.02). Leukocyte-depleted platelets contained abundant levels of the ≈205-kDa supervillin polypeptide. To assess functionality, mice lacking platelet supervillin were generated and back-crossed onto a C57BL/6 background. Compared with controls, murine platelets lacking supervillin were larger by flow cytometry and confocal microscopy and exhibited enhanced platelet thrombus formation under high-shear but not low-shear conditions. CONCLUSIONS: We show for the first time that (1) platelets contain supervillin; (2) platelet thrombus formation in the PFA-100 is associated with human SVIL variants and low SVIL expression; and (3) murine platelets lacking supervillin exhibit enhanced platelet thrombus formation at high shear stress. These data are consistent with an inhibitory role for supervillin in platelet adhesion and arterial thrombosis.


Asunto(s)
Plaquetas/fisiología , Estudio de Asociación del Genoma Completo , Proteínas de la Membrana/fisiología , Proteínas de Microfilamentos/fisiología , Adhesividad Plaquetaria/fisiología , Estrés Mecánico , Trombosis/fisiopatología , Adulto , Negro o Afroamericano/genética , Animales , Plaquetas/citología , Tamaño de la Célula , Femenino , Genotipo , Humanos , Masculino , Proteínas de la Membrana/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas de Microfilamentos/genética , Persona de Mediana Edad , Modelos Animales , Polimorfismo de Nucleótido Simple/genética , Población Blanca/genética
10.
Cancer Cell ; 7(4): 294-5, 2005 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-15837618

RESUMEN

SNF5 is a core subunit of the SWI/SNF chromatin-remodeling complex. Mammalian SNF5 is essential for normal cell viability, and loss or mutation of the human SNF gene is the molecular basis for familial malignant rhabdoid tumorigenesis. Previous studies have suggested that SNF5 suppresses cancer by signaling through the p16Ink4a and retinoblastoma tumor suppressors to negatively regulate cell cycle progression from G0/G1 into S phase. A recent paper in Genes & Development (Vries et al., 2005) reports that human SNF5 also signals via the p16INK4a-Rb-E2F pathway to regulate chromosomal stability, suggesting a new function for this chromatin remodeling protein in tumor suppression.


Asunto(s)
Ciclo Celular/fisiología , Ensamble y Desensamble de Cromatina/fisiología , Inestabilidad Cromosómica/fisiología , Proteínas de Unión al ADN/fisiología , Aneuploidia , Animales , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Proteínas de Ciclo Celular/fisiología , Proteínas Cromosómicas no Histona/química , Ciclina D1/metabolismo , Quinasa 4 Dependiente de la Ciclina , Inhibidor p16 de la Quinasa Dependiente de Ciclina/metabolismo , Quinasas Ciclina-Dependientes/metabolismo , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Factores de Transcripción E2F , Humanos , Modelos Biológicos , Fosforilación , Poliploidía , Proteínas Proto-Oncogénicas/metabolismo , Proteína de Retinoblastoma/metabolismo , Proteína de Retinoblastoma/fisiología , Tumor Rabdoide/genética , Tumor Rabdoide/metabolismo , Tumor Rabdoide/fisiopatología , Proteína SMARCB1 , Factores de Transcripción/química , Factores de Transcripción/metabolismo , Proteínas Supresoras de Tumor/genética , Proteínas Supresoras de Tumor/metabolismo , Proteínas Supresoras de Tumor/fisiología
11.
Proc Natl Acad Sci U S A ; 107(25): 11423-8, 2010 Jun 22.
Artículo en Inglés | MEDLINE | ID: mdl-20534538

RESUMEN

Ing4 is a member of the inhibitor of growth (ING) family of chromatin-modifying proteins. Biochemical experiments indicate that Ing4 is a subunit of the HB01-JADE-hEAF6 histone acetyltransferase complex responsible for most nucleosomal histone H4 acetylation in eukaryotes, and transfection studies suggest that Ing4 may regulate a wide variety of cellular processes, including DNA repair, apoptosis, cell-cycle regulation, metastasis, angiogenesis, and tumor suppression. However, in vivo evidence for a physiological role for Ing4 in cell-growth regulation is lacking. We have generated Ing4-deficient mice to explore the role of Ing4 in development, tumorigenesis, and in NF-kappaB signaling. Ing4-null mice develop normally and are viable. Although mice deficient for Ing4 fail to form spontaneous tumors, they are hypersensitive to LPS treatment and display elevated cytokine responses. Macrophages isolated from Ing4-null mice have increased levels of nuclear p65/RelA protein, resulting in increased RelA binding to NF-kappaB target promoters and up-regulation of cytokine gene expression. However, increased promoter occupancy by RelA in LPS-stimulated, Ing4-null cells does not always correlate with increased NF-kappaB target-gene expression, as RelA activation of a subset of cytokine promoters also requires Ing4 for proper histone H4 acetylation. Furthermore, activation of the IkappaB alpha promoter by RelA is also Ing4-dependent, and LPS-stimulated, Ing4-null cells have reduced levels of IkappaB alpha promoter H4 acetylation and IkappaB gene expression. Thus, Ing4 negatively regulates the cytokine-mediated inflammatory response in mice by facilitating NF-kappaB activation of IkappaB promoters, thereby suppressing nuclear RelA levels and the activation of select NF-kappaB target cytokines.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Proteínas de Homeodominio/metabolismo , FN-kappa B/metabolismo , Regiones Promotoras Genéticas , Proteínas Supresoras de Tumor/metabolismo , Animales , Núcleo Celular/metabolismo , Cruzamientos Genéticos , Citocinas/metabolismo , Inflamación , Lipopolisacáridos/metabolismo , Macrófagos/metabolismo , Ratones , Ratones Endogámicos C57BL , Modelos Biológicos , Transducción de Señal , Factor de Transcripción ReIA/metabolismo
12.
J Neurosci ; 31(33): 12029-35, 2011 Aug 17.
Artículo en Inglés | MEDLINE | ID: mdl-21849563

RESUMEN

Cyclin-dependent kinase 5 (Cdk5) and its activator p35 have been implicated in drug addiction, neurodegenerative diseases such as Alzheimer's, learning and memory, and synapse maturation and plasticity. However, the molecular mechanisms by which Cdk5 regulates synaptic plasticity are still unclear. PSD-95 is a major postsynaptic scaffolding protein of glutamatergic synapses that regulates synaptic strength and plasticity. PSD-95 is ubiquitinated by the ubiquitin E3 ligase Mdm2, and rapid and transient PSD-95 ubiquitination has been implicated in NMDA receptor-induced AMPA receptor endocytosis. Here we demonstrate that genetic or pharmacological reduction of Cdk5 activity increases the interaction of Mdm2 with PSD-95 and enhances PSD-95 ubiquitination without affecting PSD-95 protein levels in vivo in mice, suggesting a nonproteolytic function of ubiquitinated PSD-95 at synapses. We show that PSD-95 ubiquitination correlates with increased interaction with ß-adaptin, a subunit of the clathrin adaptor protein complex AP-2. This interaction is increased by genetic reduction of Cdk5 activity or NMDA receptor stimulation and is dependent on Mdm2. Together these results support a function for Cdk5 in regulating PSD-95 ubiquitination and its interaction with AP-2 and suggest a mechanism by which PSD-95 may regulate NMDA receptor-induced AMPA receptor endocytosis.


Asunto(s)
Quinasa 5 Dependiente de la Ciclina/fisiología , Guanilato-Quinasas/metabolismo , Proteínas de la Membrana/metabolismo , Neuronas/metabolismo , Ubiquitinación/fisiología , Animales , Homólogo 4 de la Proteína Discs Large , Femenino , Hipocampo/enzimología , Hipocampo/metabolismo , Masculino , Ratones , Ratones Noqueados , Neuronas/enzimología , Técnicas de Cultivo de Órganos , Mapeo de Interacción de Proteínas
13.
Dev Biol ; 353(1): 1-9, 2011 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-21334322

RESUMEN

The p53 transcription factor is activated by various types of cell stress or DNA damage and induces the expression of genes that control cell growth and inhibit tumor formation. Analysis of mice that express mutant forms of p53 suggest that inappropriate p53 activation can alter tissue homeostasis and life span, connecting p53 tumor suppressor functions with accelerated aging. However, other mouse models that display increased levels of wildtype p53 in various tissues fail to corroborate a link between p53 and aging phenotypes, possibly due to the retention of signaling pathways that negatively regulate p53 activity in these models. In this present study, we have generated mice lacking Mdm2 in the epidermis. Deletion of Mdm2, the chief negative regulator of p53, induced an aging phenotype in the skin of mice, including thinning of the epidermis, reduced wound healing, and a progressive loss of fur. These phenotypes arise due to an induction of p53-mediated senescence in epidermal stem cells and a gradual loss of epidermal stem cell function. These results reveal that activation of endogenous p53 by ablation of Mdm2 can induce accelerated aging phenotypes in mice.


Asunto(s)
Senescencia Celular , Células Epidérmicas , Proteínas Proto-Oncogénicas c-mdm2/fisiología , Transducción de Señal/fisiología , Envejecimiento de la Piel , Células Madre/fisiología , Proteína p53 Supresora de Tumor/fisiología , Animales , Inhibidor p16 de la Quinasa Dependiente de Ciclina/análisis , Ratones , Fenotipo
14.
J Biol Chem ; 286(34): 30057-70, 2011 Aug 26.
Artículo en Inglés | MEDLINE | ID: mdl-21676869

RESUMEN

The Runt-related transcription factor, Runx2, is essential for osteogenesis and is controlled by both distal (P1) and proximal (P2) promoters. To understand Runx2 function requires determination of the spatiotemporal activity of P1 and P2 to Runx2 protein production. We generated a mouse model in which the P1-derived transcript was replaced with a lacZ reporter allele, resulting in loss of P1-derived protein while simultaneously allowing discrimination between the activities of the two promoters. Loss of P1-driven expression causes developmental defects with cleidocranial dysplasia-like syndromes that persist in the postnatal skeleton. P1 activity is robust in preosteogenic mesenchyme and at the onset of bone formation but decreases as bone matures. Homozygous Runx2-P1(lacZ/lacZ) mice have a normal life span but exhibit severe osteopenia and compromised bone repair in adult mice because of osteoblastic defects and not increased osteoclastic resorption. Gene expression profiles of bone, immunohistochemical studies, and ex vivo differentiation using calvarial osteoblasts and marrow stromal cells identified mechanisms for the skeletal phenotype. The findings indicate that P1 promoter activity is necessary for generating a threshold level of Runx2 protein to commit sufficient osteoprogenitor numbers for normal bone formation. P1 promoter function is not compensated via the P2 promoter. However, the P2 transcript with compensatory mechanisms from bone morphogenetic protein (BMP) and Wnt signaling is adequate for mineralization of the bone tissue that does form. We conclude that selective utilization of the P1 and P2 promoters enables the precise spatiotemporal expression of Runx2 necessary for normal skeletogenesis and the maintenance of bone mass in the adult.


Asunto(s)
Calcificación Fisiológica/fisiología , Subunidad alfa 1 del Factor de Unión al Sitio Principal/biosíntesis , Regulación de la Expresión Génica/fisiología , Osteoblastos/metabolismo , Osteogénesis/fisiología , Regiones Promotoras Genéticas/fisiología , Células Madre/metabolismo , Animales , Enfermedades del Desarrollo Óseo/genética , Enfermedades del Desarrollo Óseo/metabolismo , Enfermedades Óseas Metabólicas/genética , Enfermedades Óseas Metabólicas/metabolismo , Regeneración Ósea/fisiología , Subunidad alfa 1 del Factor de Unión al Sitio Principal/genética , Ratones , Ratones Transgénicos
15.
Hum Mol Genet ; 19(6): 1048-57, 2010 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-20035012

RESUMEN

Runx1 is a key hematopoietic transcription factor required for definitive hematopoiesis and is a frequent target of leukemia-related chromosomal translocations. The resulting fusion proteins, while retaining DNA binding activity, display loss of subnuclear targeting and associated transactivation functions encoded by the C-terminus of the protein. To define the precise contribution of the Runx1 C-terminus in development and leukemia, we created a knock-in mouse with a C-terminal truncation by introducing a single nucleic acid substitution in the native Runx1 locus. This mutation (Runx1(Q307X)) models genetic lesions observed in patients with leukemia and myeloproliferative disorders. The Runx1(Q307X) homozygous mouse exhibits embryonic lethality at E12.5 due to central nervous system hemorrhages and a complete lack of hematopoietic stem cell function. While able to bind DNA, Runx1(Q307X) is unable to activate target genes, resulting in deregulation of various hematopoietic markers. Thus, we demonstrate that the subnuclear targeting and transcriptional regulatory activities of the Runx1 C-terminus are critical for hematopoietic development. We propose that compromising the C-terminal functions of Runx1 is a common mechanism for the pathological consequences of a variety of somatic mutations and Runx1-related leukemic fusion proteins observed in human patients.


Asunto(s)
Núcleo Celular/metabolismo , Subunidad alfa 2 del Factor de Unión al Sitio Principal/química , Subunidad alfa 2 del Factor de Unión al Sitio Principal/metabolismo , Hematopoyesis , Activación Transcripcional/genética , Animales , Línea Celular Tumoral , Cruzamientos Genéticos , Embrión de Mamíferos/anomalías , Embrión de Mamíferos/metabolismo , Femenino , Regulación del Desarrollo de la Expresión Génica , Genotipo , Hematopoyesis/genética , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/metabolismo , Heterocigoto , Humanos , Masculino , Ratones , Proteínas Mutantes/química , Proteínas Mutantes/metabolismo , Mutación/genética , Unión Proteica , Estructura Terciaria de Proteína , Transporte de Proteínas , Relación Estructura-Actividad
16.
Cancer Cell ; 4(5): 349-60, 2003 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-14667502

RESUMEN

Wnt5a is a member of the Wnt family of secreted glycoproteins that play essential organizing roles in development. Similar to other Wnt members, Wnt5a can upregulate cell proliferation and has been proposed to have oncogenic function. Here we report that Wnt5a signals through the noncanonical Wnt/Ca++ pathway to suppress cyclin D1 expression and negatively regulate B cell proliferation in a cell-autonomous manner. Wnt5a hemizygous mice develop myeloid leukemias and B cell lymphomas that are clonal in origin and display loss of Wnt5a function in tumor tissues. Furthermore, analysis of human primary leukemias reveals deletion of the WNT5A gene and/or loss of WNT5A expression in a majority of the patient samples. These results demonstrate that Wnt5a suppresses hematopoietic malignancies.


Asunto(s)
Linfocitos B/metabolismo , División Celular/fisiología , Leucemia Mieloide/metabolismo , Linfoma de Células B/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Animales , Calcio/metabolismo , Células Cultivadas , Ciclina D1/metabolismo , Citometría de Flujo , Sistema Hematopoyético/metabolismo , Sistema Hematopoyético/fisiopatología , Humanos , Interleucina-7/metabolismo , Leucemia Mieloide/patología , Pérdida de Heterocigocidad/fisiología , Tejido Linfoide/fisiopatología , Linfoma de Células B/patología , Ratones , Ratones Noqueados , Proteínas Proto-Oncogénicas/genética , Transducción de Señal/fisiología , Trasplante Heterólogo , Proteínas Wnt , Proteína Wnt-5a
17.
Proc Natl Acad Sci U S A ; 106(30): 12359-64, 2009 Jul 28.
Artículo en Inglés | MEDLINE | ID: mdl-19590016

RESUMEN

Competency for DNA replication is functionally coupled to the activation of histone gene expression at the onset of S phase to form chromatin. Human histone nuclear factor P (HiNF-P; gene symbol HINFP) bound to its cyclin E/cyclin-dependent kinase 2 (CDK2) responsive coactivator p220(NPAT) is a key regulator of multiple human histone H4 genes that encode a major subunit of the nucleosome. Induction of the histone H4 transcription factor (HINFP)/p220(NPAT) coactivation complex occurs in parallel with the CDK-dependent release of pRB from E2F at the restriction point. Here, we show that the downstream CDK-dependent cell cycle effector HINFP is genetically required and, in contrast to the CDK2/cyclin E complex, cannot be compensated. We constructed a mouse Hinfp-null mutation and found that heterozygous Hinfp mice survive, indicating that 1 allele suffices for embryogenesis. Homozygous loss-of-function causes embryonic lethality: No homozygous Hinfp-null mice are obtained at or beyond embryonic day (E) 6.5. In blastocyst cultures, Hinfp-null embryos exhibit a delay in hatching, abnormal growth, and loss of histone H4 gene expression. Our data indicate that the CDK2/cyclin E/p220(NPAT)/HINFP/histone gene signaling pathway at the G1/S phase transition is an essential, nonredundant cell cycle regulatory mechanism that is established early in embryogenesis.


Asunto(s)
Ciclo Celular/fisiología , Ciclina E/metabolismo , Quinasa 2 Dependiente de la Ciclina/metabolismo , Histonas/metabolismo , Proteínas Represoras/metabolismo , Animales , Blastocisto/citología , Blastocisto/metabolismo , Western Blotting , Ciclo Celular/genética , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Células Cultivadas , Ciclina E/genética , Quinasa 2 Dependiente de la Ciclina/genética , Embrión de Mamíferos/citología , Embrión de Mamíferos/embriología , Embrión de Mamíferos/metabolismo , Femenino , Fibroblastos/citología , Fibroblastos/metabolismo , Fase G1/genética , Fase G1/fisiología , Regulación del Desarrollo de la Expresión Génica , Histonas/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Proteínas Represoras/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Fase S/genética , Fase S/fisiología , Transducción de Señal/genética , Transducción de Señal/fisiología , Factores de Tiempo
18.
Dev Biol ; 340(1): 10-21, 2010 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-20079730

RESUMEN

MicroRNA attenuation of protein translation has emerged as an important regulator of mesenchymal cell differentiation into the osteoblast lineage. A compelling question is the extent to which miR biogenesis is obligatory for bone formation. Here we show conditional deletion of the Dicer enzyme in osteoprogenitors by Col1a1-Cre compromised fetal survival after E14.5. A mechanism was associated with the post-commitment stage of osteoblastogenesis, demonstrated by impaired ECM mineralization and reduced expression of mature osteoblast markers during differentiation of mesenchymal cells of ex vivo deleted Dicer(c/c). In contrast, in vivo excision of Dicer by Osteocalcin-Cre in mature osteoblasts generated a viable mouse with a perinatal phenotype of delayed bone mineralization which was resolved by 1 month. However, a second phenotype of significantly increased bone mass developed by 2 months, which continued up to 8 months in long bones and vertebrae, but not calvariae. Cortical bone width and trabecular thickness in Dicer(Deltaoc/Deltaoc) was twice that of Dicer(c/c) controls. Normal cell and tissue organization was observed. Expression of osteoblast and osteoclast markers demonstrated increased coupled activity of both cell types. We propose that Dicer generated miRs are essential for two periods of bone formation, to promote osteoblast differentiation before birth, and control bone accrual in the adult.


Asunto(s)
Diferenciación Celular , ARN Helicasas DEAD-box/genética , Endorribonucleasas/genética , Osteoblastos/metabolismo , Osteogénesis/fisiología , Células Madre/citología , Animales , Senescencia Celular , Colágeno Tipo I/genética , Colágeno Tipo I/metabolismo , Cadena alfa 1 del Colágeno Tipo I , ARN Helicasas DEAD-box/metabolismo , Embrión de Mamíferos/metabolismo , Endorribonucleasas/metabolismo , Genes Letales , Ratones , Ratones Endogámicos , ARN Mensajero/metabolismo , Ribonucleasa III , Células Madre/metabolismo
19.
J Cell Physiol ; 226(5): 1399-406, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-20945399

RESUMEN

Dicer, an enzyme involved in microRNA maturation, is required for proper embryo gastrulation and tissue morphogenesis during mammalian development. Using primary cultures of fibroblasts and pre-adipocytes, we have previously shown that Dicer is essential for early stages of adipogenic cell differentiation. In this study, we have utilized Dicer-conditional mice to explore a role for Dicer and microRNA biogenesis in the terminal differentiation of adipocytes in vivo and in the formation of white and brown adipose tissue. Deletion of Dicer in differentiated adipocytes in Dicer-conditional, aP2-Cre transgenic mice reduced the level of various adipogenic-associated transcripts and inhibited lipogenesis in white adipocytes, resulting in a severe depletion of white adipose tissue in mice. In contrast, Dicer was not required in vivo for lipogenesis in brown adipose or for brown fat formation. However, Dicer deletion in brown adipose did decrease the expression of genes involved in thermoregulation. The results of our study provide genetic evidence of a role for microRNA molecules in regulating adipogenesis and reveal distinct requirements for Dicer in the formation of white and brown adipose tissue.


Asunto(s)
Adipocitos/enzimología , Adipogénesis , Tejido Adiposo Pardo/enzimología , Tejido Adiposo Blanco/enzimología , ARN Helicasas DEAD-box/metabolismo , Endorribonucleasas/metabolismo , MicroARNs/metabolismo , Adipocitos/patología , Adipogénesis/genética , Tejido Adiposo Pardo/crecimiento & desarrollo , Tejido Adiposo Pardo/patología , Tejido Adiposo Blanco/crecimiento & desarrollo , Tejido Adiposo Blanco/patología , Factores de Edad , Animales , Regulación de la Temperatura Corporal/genética , ARN Helicasas DEAD-box/deficiencia , ARN Helicasas DEAD-box/genética , Endorribonucleasas/deficiencia , Endorribonucleasas/genética , Proteínas de Unión a Ácidos Grasos/genética , Regulación del Desarrollo de la Expresión Génica , Integrasas/genética , Lipogénesis/genética , Ratones , Ratones Noqueados , Regiones Promotoras Genéticas , ARN Mensajero/metabolismo , Ribonucleasa III
20.
Hum Mol Genet ; 18(3): 556-68, 2009 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-19028669

RESUMEN

Cleidocranial dysplasia (CCD) in humans is an autosomal-dominant skeletal disease that results from mutations in the bone-specific transcription factor RUNX2 (CBFA1/AML3). However, distinct RUNX2 mutations in CCD do not correlate with the severity of the disease. Here we generated a new mouse model with a hypomorphic Runx2 mutant allele (Runx2(neo7)), in which only part of the transcript is processed to full-length (wild-type) Runx2 mRNA. Homozygous Runx2(neo7/neo7) mice express a reduced level of wild-type Runx2 mRNA (55-70%) and protein. This mouse model allowed us to establish the minimal requirement of functional Runx2 for normal bone development. Runx2(neo7/neo7) mice have grossly normal skeletons with no abnormalities observed in the growth plate, but do exhibit developmental defects in calvaria and clavicles that persist through post-natal growth. Clavicle defects are caused by disrupted endochondral bone formation during embryogenesis. These hypomorphic mice have altered calvarial bone volume, as observed by histology and microCT imaging, and decreased expression of osteoblast marker genes. The bone phenotype of the heterozygous mice, which have 79-84% of wild-type Runx2 mRNA, is normal. These results show there is a critical gene dosage requirement of functional Runx2 for the formation of intramembranous bone tissues during embryogenesis. A decrease to 70% of wild-type Runx2 levels results in the CCD syndrome, whereas levels >79% produce a normal skeleton. Our findings suggest that the range of bone phenotypes in CCD patients is attributable to quantitative reduction in the functional activity of RUNX2.


Asunto(s)
Displasia Cleidocraneal/metabolismo , Subunidad alfa 1 del Factor de Unión al Sitio Principal/metabolismo , Animales , Desarrollo Óseo , Células Cultivadas , Clavícula/anomalías , Clavícula/embriología , Clavícula/crecimiento & desarrollo , Clavícula/metabolismo , Displasia Cleidocraneal/embriología , Displasia Cleidocraneal/genética , Subunidad alfa 1 del Factor de Unión al Sitio Principal/genética , Femenino , Placa de Crecimiento/crecimiento & desarrollo , Placa de Crecimiento/metabolismo , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Osteoblastos/metabolismo , Fenotipo , Cráneo/anomalías , Cráneo/embriología , Cráneo/crecimiento & desarrollo , Cráneo/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA