Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Exp Mol Med ; 53(11): 1781-1791, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-34845330

RESUMO

Vascular calcification increases morbidity and mortality in patients with cardiovascular and renal diseases. Previously, we reported that histone deacetylase 1 prevents vascular calcification, whereas its E3 ligase, mouse double minute 2 homolog (MDM2), induces vascular calcification. In the present study, we identified the upstream regulator of MDM2. By utilizing cellular models and transgenic mice, we confirmed that E3 ligase activity is required for vascular calcification. By promoter analysis, we found that both msh homeobox 1 (Msx1) and msh homeobox 2 (Msx2) bound to the MDM2 promoter region, which resulted in transcriptional activation of MDM2. The expression levels of both Msx1 and Msx2 were increased in mouse models of vascular calcification and in calcified human coronary arteries. Msx1 and Msx2 potentiated vascular calcification in cellular and mouse models in an MDM2-dependent manner. Our results establish a novel role for MSX1/MSX2 in the transcriptional activation of MDM2 and the resultant increase in MDM2 E3 ligase activity during vascular calcification.


Assuntos
Proteínas de Homeodomínio/metabolismo , Fator de Transcrição MSX1/metabolismo , Proteínas Proto-Oncogênicas c-mdm2/genética , Ubiquitina-Proteína Ligases/genética , Calcificação Vascular/etiologia , Calcificação Vascular/metabolismo , Animais , Biomarcadores , Cálcio/metabolismo , Modelos Animais de Doenças , Suscetibilidade a Doenças , Regulação da Expressão Gênica , Técnicas de Silenciamento de Genes , Humanos , Masculino , Camundongos , Camundongos Knockout , Modelos Biológicos , Mutação , Regiões Promotoras Genéticas , Proteínas Proto-Oncogênicas c-mdm2/metabolismo , Elementos de Resposta , Ubiquitina-Proteína Ligases/metabolismo , Calcificação Vascular/patologia
2.
Nat Commun ; 7: 10492, 2016 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-26832969

RESUMO

Vascular calcification (VC) is often associated with cardiovascular and metabolic diseases. However, the molecular mechanisms linking VC to these diseases have yet to be elucidated. Here we report that MDM2-induced ubiquitination of histone deacetylase 1 (HDAC1) mediates VC. Loss of HDAC1 activity via either chemical inhibitor or genetic ablation enhances VC. HDAC1 protein, but not mRNA, is reduced in cell and animal calcification models and in human calcified coronary artery. Under calcification-inducing conditions, proteasomal degradation of HDAC1 precedes VC and it is mediated by MDM2 E3 ubiquitin ligase that initiates HDAC1 K74 ubiquitination. Overexpression of MDM2 enhances VC, whereas loss of MDM2 blunts it. Decoy peptide spanning HDAC1 K74 and RG 7112, an MDM2 inhibitor, prevent VC in vivo and in vitro. These results uncover a previously unappreciated ubiquitination pathway and suggest MDM2-mediated HDAC1 ubiquitination as a new therapeutic target in VC.


Assuntos
Histona Desacetilase 1/metabolismo , Proteínas Proto-Oncogênicas c-mdm2/metabolismo , Calcificação Vascular/metabolismo , Animais , Cálcio , Regulação da Expressão Gênica , Histona Desacetilase 1/genética , Humanos , Masculino , Camundongos , Músculo Liso Vascular/citologia , Proteínas Proto-Oncogênicas c-mdm2/genética , Ratos , Ubiquitinação
3.
Birth Defects Res A Clin Mol Teratol ; 94(9): 729-37, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22815056

RESUMO

BACKGROUND: Ly-1 antibody reactive clone (LYAR) is a nucleolar zinc finger protein that has been implicated in cell growth, self-renewal of embryonic stem cells, and medulloblastoma. To test whether LYAR is critical for cell growth and development, we generated Lyar mutant mice. METHODS: Mice carrying the mutant Lyar(gt) allele were generated from embryonic stem cells that contained a gene-trap insertion in the Lyar gene. Phenotypic analyses were performed on Lyar mutant mice and mouse embryonic fibroblasts. Lyar(gt/gt) mice were crossed to mice lacking the p53 tumor suppressor protein and Lyar/p53 compound mutants scored for external abnormalities. RESULTS: Lyar(gt/gt) homozygotes are viable, fertile, and indistinguishable from wild type littermates. However, the growth of Lyar(+/gt) and Lyar(gt/gt) mouse embryonic fibroblasts (MEFs) was impaired, coincident with an increase in the steady-state level of p53 and a key p53 effector of growth arrest, p21, suggesting that a cellular stress response is triggered in the absence of a wild type level of LYAR. Remarkably, the majority of Lyar(+/gt) and Lyar(gt/gt) female mice lacking p53 mice failed to survive. The neural tube defect (NTD) exencephaly was observed in ≈26% and ≈61% of female Lyar(+/gt;) p53(-/-) and Lyar(gt/gt;) p53(-/-) embryos, respectively. CONCLUSIONS: Lyar/p53 mutant mice represent a new digenic model of NTDs. Furthermore, these studies identify Lyar as a novel candidate gene for a role in human NTDs. These results provide new data to support the idea that loss of a p53-mediated developmental checkpoint may increase the risk of NTDs owing to some germline mutations.


Assuntos
Proteínas de Ligação a DNA/genética , Efeito Fundador , Mutação , Defeitos do Tubo Neural/genética , Proteínas Nucleares/genética , Proteína Supressora de Tumor p53/genética , Animais , Ciclo Celular/genética , Proliferação de Células , Modelos Animais de Doenças , Células-Tronco Embrionárias/metabolismo , Células-Tronco Embrionárias/patologia , Feminino , Fibroblastos/metabolismo , Fibroblastos/patologia , Genótipo , Homozigoto , Humanos , Camundongos , Camundongos Transgênicos , Mutagênese Insercional , Defeitos do Tubo Neural/patologia , Fenótipo , Fatores Sexuais
4.
Neurosci Lett ; 513(1): 106-10, 2012 Mar 28.
Artigo em Inglês | MEDLINE | ID: mdl-22343310

RESUMO

A critical component of the cellular stress response, the p53 tumor suppressor protein must be functional for many cancer therapies to be effective. Adjuvant therapies that augment p53 function are predicted to sensitize tumor cells to cancer therapies that rely upon p53 for their efficacy. Of those strategies currently being explored to enhance p53 function, inhibition of the ubiquitin ligase, MDM2, a negative regulator of p53, has shown promise. Here, we investigated whether MDM2 antagonism might be effective in inducing cell death in human medulloblastoma (MB) cells. Nutlin-3, a small-molecule inhibitor of MDM2, potently induced apoptosis in MB cells with wild-type TP53. Moreover, nutlin-3 potentiated p53 activation and growth impairment of MB cells in combination with the classic DNA-damaging agent doxorubicin. Together, these results support the concept that MDM2 antagonists may be therapeutically beneficial for patients with MB tumors.


Assuntos
Neoplasias Encefálicas/patologia , Imidazóis/farmacologia , Meduloblastoma/patologia , Piperazinas/farmacologia , Proteínas Proto-Oncogênicas c-mdm2/antagonistas & inibidores , Antibióticos Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Western Blotting , Ciclo Celular/efeitos dos fármacos , Morte Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Corantes , Doxorrubicina/farmacologia , Humanos , Sais de Tetrazólio , Tiazóis , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/fisiologia
5.
Genes Cancer ; 2(4): 431-42, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21779511

RESUMO

The p53 tumor suppressor potently limits the growth of immature and mature neurons under conditions of cellular stress. Although loss of p53 function contributes to the pathogenesis of central nervous system (CNS) tumors, excessive p53 function is implicated in neural tube defects, embryonic lethality, and neuronal degeneration. Thus, p53 function must be tightly controlled. The anti-proliferative properties of p53 are mediated, in part, through the induction of apoptosis, cell cycle arrest, and senescence. Although there is still much to be learned about the role of p53 in these processes, recent evidence supports exciting new roles for p53 in a wide range of processes, including neural precursor cell self-renewal, differentiation, and cell fate decisions. Understanding the full repertoire of p53 function in CNS development and tumorigenesis may provide us with novel points of therapeutic intervention for human diseases of the CNS.

6.
Blood ; 118(13): 3622-33, 2011 Sep 29.
Artigo em Inglês | MEDLINE | ID: mdl-21788341

RESUMO

Reduced gene dosage of ribosomal protein subunits has been implicated in 5q- myelodysplastic syndrome and Diamond Blackfan anemia, but the cellular and pathophysiologic defects associated with these conditions are enigmatic. Using conditional inactivation of the ribosomal protein S6 gene in laboratory mice, we found that reduced ribosomal protein gene dosage recapitulates cardinal features of the 5q- syndrome, including macrocytic anemia, erythroid hypoplasia, and megakaryocytic dysplasia with thrombocytosis, and that p53 plays a critical role in manifestation of these phenotypes. The blood cell abnormalities are accompanied by a reduction in the number of HSCs, a specific defect in late erythrocyte development, and suggest a disease-specific ontogenetic pathway for megakaryocyte development. Further studies of highly purified HSCs from healthy patients and from those with myelodysplastic syndrome link reduced expression of ribosomal protein genes to decreased RBC maturation and suggest an underlying and common pathophysiologic pathway for additional subtypes of myelodysplastic syndrome.


Assuntos
Dosagem de Genes , Síndromes Mielodisplásicas/genética , Proteínas Ribossômicas/genética , Proteína Supressora de Tumor p53/metabolismo , Animais , Regulação para Baixo/genética , Feminino , Dosagem de Genes/genética , Dosagem de Genes/fisiologia , Predisposição Genética para Doença , Masculino , Camundongos , Camundongos Endogâmicos C3H , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Modelos Biológicos , Síndromes Mielodisplásicas/etiologia , Síndromes Mielodisplásicas/metabolismo , Proteínas Ribossômicas/metabolismo , Fatores de Risco , Proteína Supressora de Tumor p53/fisiologia
7.
PLoS One ; 6(3): e17884, 2011 Mar 18.
Artigo em Inglês | MEDLINE | ID: mdl-21437245

RESUMO

Disruption of cerebellar granular neuronal precursor (GNP) maturation can result in defects in motor coordination and learning, or in medulloblastoma, the most common childhood brain tumor. The Sonic Hedgehog (Shh) pathway is important for GNP proliferation; however, the factors regulating the extent and timing of GNP proliferation, as well as GNP differentiation and migration are poorly understood. The p53 tumor suppressor has been shown to negatively regulate the activity of the Shh effector, Gli1, in neural stem cells; however, the contribution of p53 to the regulation of Shh signaling in GNPs during cerebellar development has not been determined. Here, we exploited a hypomorphic allele of Mdm2 (Mdm2(puro)), which encodes a critical negative regulator of p53, to alter the level of wild-type MDM2 and p53 in vivo. We report that mice with reduced levels of MDM2 and increased levels of p53 have small cerebella with shortened folia, reminiscent of deficient Shh signaling. Indeed, Shh signaling in Mdm2-deficient GNPs is attenuated, concomitant with decreased expression of the Shh transducers, Gli1 and Gli2. We also find that Shh stimulation of GNPs promotes MDM2 accumulation and enhances phosphorylation at serine 166, a modification known to increase MDM2-p53 binding. Significantly, loss of MDM2 in Ptch1(+/-) mice, a model for Shh-mediated human medulloblastoma, impedes cerebellar tumorigenesis. Together, these results place MDM2 at a major nexus between the p53 and Shh signaling pathways in GNPs, with key roles in cerebellar development, GNP survival, cerebellar foliation, and MB tumorigenesis.


Assuntos
Neoplasias Cerebelares/patologia , Cerebelo/crescimento & desenvolvimento , Cerebelo/metabolismo , Proteínas Hedgehog/metabolismo , Lesões Pré-Cancerosas/metabolismo , Proteínas Proto-Oncogênicas c-mdm2/metabolismo , Proteína Supressora de Tumor p53/antagonistas & inibidores , Envelhecimento/patologia , Alelos , Animais , Apoptose , Proliferação de Células , Sobrevivência Celular , Neoplasias Cerebelares/metabolismo , Cerebelo/anormalidades , Cerebelo/patologia , Humanos , Fatores de Transcrição Kruppel-Like/metabolismo , Camundongos , Células-Tronco Neurais/metabolismo , Células-Tronco Neurais/patologia , Neurônios/metabolismo , Neurônios/patologia , Receptores Patched , Receptor Patched-1 , Lesões Pré-Cancerosas/patologia , Proteínas Proto-Oncogênicas c-mdm2/genética , Receptores de Superfície Celular/metabolismo , Transdução de Sinais , Proteína Supressora de Tumor p53/metabolismo , Proteína GLI1 em Dedos de Zinco , Proteína Gli2 com Dedos de Zinco
8.
Genes Cancer ; 1(5): 465-479, 2010 May.
Artigo em Inglês | MEDLINE | ID: mdl-20927200

RESUMO

Deregulated c-Myc is associated with a wide range of human cancers. In many cell types, overexpression of c-Myc potently promotes cell growth and proliferation concomitant with the induction of apoptosis. Secondary genetic events that shift this balance either by increasing growth and proliferation or limiting apoptosis are likely to cooperate with c-Myc in tumorigenesis. Here, the authors have performed large-scale insertional mutagenesis in Eµ-c-myc mice that, through mdm2 loss of function mutations, are sensitized to apoptosis. The authors chose to use this genetic background based on the hypothesis that the high level of apoptosis induced by c-Myc overexpression in MDM2-deficient mice would act as a rate-limiting barrier for lymphoma development. As a result, it was predicted that the spectrum of retroviral insertions would be shifted toward loci that harbor antiapoptotic genes. Nine novel common insertion sites (CISs) specific to mice with this sensitized genetic background were identified, suggesting the presence of novel antiapoptotic cancer genes. Moreover, cross-comparing the data to the Retroviral Tagged Cancer Gene Database, the authors identified an additional 23 novel CISs. Here, evidence is presented that 2 genes, ppp1r16b and hdac6, identified at CISs, are bona fide cellular oncogenes. This study highlights the power of combining unique sensitized genetic backgrounds with large-scale mutagenesis as an approach for identifying novel cancer genes.

9.
Nature ; 444(7115): 61-6, 2006 Nov 02.
Artigo em Inglês | MEDLINE | ID: mdl-17080083

RESUMO

Most human tumours have genetic mutations in their Rb and p53 pathways, but retinoblastoma is thought to be an exception. Studies suggest that retinoblastomas, which initiate with mutations in the gene retinoblastoma 1 (RB1), bypass the p53 pathway because they arise from intrinsically death-resistant cells during retinal development. In contrast to this prevailing theory, here we show that the tumour surveillance pathway mediated by Arf, MDM2, MDMX and p53 is activated after loss of RB1 during retinogenesis. RB1-deficient retinoblasts undergo p53-mediated apoptosis and exit the cell cycle. Subsequently, amplification of the MDMX gene and increased expression of MDMX protein are strongly selected for during tumour progression as a mechanism to suppress the p53 response in RB1-deficient retinal cells. Our data provide evidence that the p53 pathway is inactivated in retinoblastoma and that this cancer does not originate from intrinsically death-resistant cells as previously thought. In addition, they support the idea that MDMX is a specific chemotherapeutic target for treating retinoblastoma.


Assuntos
Proteínas Nucleares/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Retinoblastoma/metabolismo , Proteína Supressora de Tumor p53/antagonistas & inibidores , Proteína Supressora de Tumor p53/metabolismo , Animais , Proteínas de Ciclo Celular , Morte Celular , Divisão Celular , Dano ao DNA , Amplificação de Genes/genética , Humanos , Imidazóis/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Proteínas Nucleares/antagonistas & inibidores , Proteínas Nucleares/genética , Piperazinas/metabolismo , Proteínas Proto-Oncogênicas/antagonistas & inibidores , Proteínas Proto-Oncogênicas/genética , Ratos , Ratos Sprague-Dawley , Retina/metabolismo , Retinoblastoma/genética , Retinoblastoma/patologia , Proteína do Retinoblastoma/deficiência , Proteína do Retinoblastoma/genética , Proteína do Retinoblastoma/metabolismo , Proteína Supressora de Tumor p14ARF/metabolismo
10.
Cell Cycle ; 5(7): 714-7, 2006 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-16582632

RESUMO

The prevalence of mutations that inactivate the p53 tumor suppressor gene in human cancers reveals the importance of p53 in preventing cancer. Recent progress has generated increased enthusiasm for re-activating p53 in tumors with mutant p53 proteins as well as for increasing p53 function in tumors expressing wild-type p53 that is inhibited in trans. However, excessive p53 activity can be detrimental to the host, potentially limiting the utility of p53 activation as a therapeutic strategy. For example, uncontrolled p53 activity is lethal to the murine embryo, and p53 has been associated with increased aging in people and mice. Here we review the literature linking p53 to aging and discuss reports demonstrating that p53 can suppress tumor formation without accelerating aging. We raise the possibility that activation of p53 remains a promising strategy for cancer chemoprevention and therapy even if, under some circumstances, p53 might accelerate aging.


Assuntos
Envelhecimento/metabolismo , Neoplasias/metabolismo , Neoplasias/patologia , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/metabolismo , Envelhecimento/genética , Senilidade Prematura/genética , Animais , Predisposição Genética para Doença , Humanos , Neoplasias/genética , Proteínas Proto-Oncogênicas c-mdm2/metabolismo
11.
Genes Dev ; 20(1): 16-21, 2006 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-16391230

RESUMO

The p53 inhibitor murine double-minute gene 2 (Mdm2) is a target for potential cancer therapies, however increased p53 function can be lethal. To directly address whether reduced Mdm2 function can inhibit tumorigenesis without causing detrimental side effects, we exploited a hypomorphic murine allele of mdm2 to compare the effects of decreased levels of Mdm2 and hence increased p53 activity on tumorigenesis and life span in mice. Here we report that mice with decreased levels of Mdm2 are resistant to tumor formation yet do not age prematurely, supporting the notion that Mdm2 is a promising target for cancer therapeutics.


Assuntos
Envelhecimento/metabolismo , Neoplasias Intestinais/patologia , Proteínas Proto-Oncogênicas c-mdm2/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Animais , Osso e Ossos/metabolismo , Osso e Ossos/patologia , Neoplasias Intestinais/metabolismo , Contagem de Leucócitos , Camundongos , Camundongos Mutantes , Proteínas Proto-Oncogênicas c-mdm2/genética , Pele/metabolismo , Pele/patologia , Taxa de Sobrevida , Proteína Supressora de Tumor p53/genética
12.
Mol Cell Biol ; 25(16): 6990-7004, 2005 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-16055712

RESUMO

The corepressor mSin3A is the core component of a chromatin-modifying complex that is recruited by multiple gene-specific transcriptional repressors. In order to understand the role of mSin3A during development, we generated constitutive germ line as well as conditional msin3A deletions. msin3A deletion in the developing mouse embryo results in lethality at the postimplantation stage, demonstrating that it is an essential gene. Blastocysts derived from preimplantation msin3A null embryos and mouse embryo fibroblasts (MEFs) lacking msin3A display a significant reduction in cell division. msin3A null MEFs also show mislocalization of the heterochromatin protein, HP1alpha, without alterations in global histone acetylation. Heterozygous msin3A(+/-) mice with a systemic twofold decrease in mSin3A protein develop splenomegaly as well as kidney disease indicative of a disruption of lymphocyte homeostasis. Conditional deletion of msin3A from developing T cells results in reduced thymic cellularity and a fivefold decrease in the number of cytotoxic (CD8) T cells, while helper (CD4) T cells are unaffected. We show that CD8 development is dependent on mSin3A at a step downstream of T-cell receptor signaling and that loss of mSin3A specifically decreases survival of double-positive and CD8 T cells. Thus, msin3A is a pleiotropic gene which, in addition to its role in cell cycle progression, is required for the development and homeostasis of cells in the lymphoid lineage.


Assuntos
Cromatina/metabolismo , Proteínas Repressoras/fisiologia , Linfócitos T/citologia , Animais , Apoptose , Blastocisto , Western Blotting , Linfócitos T CD4-Positivos/metabolismo , Linfócitos T CD8-Positivos/metabolismo , Ciclo Celular , Diferenciação Celular , Linhagem da Célula , Proliferação de Células , Células Cultivadas , Cromatina/química , Homólogo 5 da Proteína Cromobox , Proteínas Cromossômicas não Histona/metabolismo , Éxons , Fibroblastos/citologia , Fibroblastos/metabolismo , Citometria de Fluxo , Deleção de Genes , Regulação da Expressão Gênica no Desenvolvimento , Genótipo , Glomerulonefrite Membranosa , Heterocromatina/metabolismo , Heterozigoto , Camundongos , Camundongos Transgênicos , Modelos Biológicos , Modelos Genéticos , Recombinação Genética , Complexo Correpressor Histona Desacetilase e Sin3 , Esplenomegalia , Linfócitos T/metabolismo , Linfócitos T Citotóxicos/citologia , Timo/citologia , Fatores de Tempo
13.
Mol Cell Biol ; 24(1): 186-91, 2004 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-14673154

RESUMO

Tumor suppressor proteins must be exquisitely regulated since they can induce cell death while preventing cancer. For example, the p19(ARF) tumor suppressor (p14(ARF) in humans) appears to stimulate the apoptotic function of the p53 tumor suppressor to prevent lymphomagenesis and carcinogenesis induced by oncogene overexpression. Here we present a genetic approach to defining the role of p19(ARF) in regulating the apoptotic function of p53 in highly proliferating, homeostatic tissues. In contrast to our expectation, p19(ARF) did not activate the apoptotic function of p53 in lymphocytes or epithelial cells. These results demonstrate that the mechanisms that control p53 function during homeostasis differ from those that are critical for tumor suppression. Moreover, the Mdm2/p53/p19(ARF) pathway appears to exist only under very restricted conditions.


Assuntos
Homeostase/fisiologia , Proteínas Nucleares , Proteínas Proto-Oncogênicas/metabolismo , Proteína Supressora de Tumor p14ARF/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Animais , Peso Corporal/genética , Inibidor p16 de Quinase Dependente de Ciclina , Epitélio/metabolismo , Intestino Delgado/metabolismo , Camundongos , Camundongos Transgênicos , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas c-mdm2 , Proteína Supressora de Tumor p14ARF/genética
14.
Mol Cell Biol ; 23(2): 462-72, 2003 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-12509446

RESUMO

The function of the p53 tumor suppressor protein must be highly regulated because p53 can cause cell death and prevent tumorigenesis. In cultured cells, the p90MDM2 protein blocks the transcriptional activation domain of p53 and also stimulates the degradation of p53. Here we provide the first conclusive demonstration that p90MDM2 constitutively regulates p53 activity in homeostatic tissues. Mice with a hypomorphic allele of mdm2 revealed a heretofore unknown role for mdm2 in lymphopoiesis and epithelial cell survival. Phenotypic analyses revealed that both the transcriptional activation and apoptotic functions of p53 were increased in these mice. However, the level of p53 protein was not coordinately increased, suggesting that p90MDM2 can inhibit the transcriptional activation and apoptotic functions of p53 in a manner independent of degradation. Cre-mediated deletion of mdm2 caused a greater accumulation of p53, demonstrating that p90MDM2 constitutively regulates both the activity and the level of p53 in homeostatic tissues. The observation that only a subset of tissues with activated p53 underwent apoptosis indicates that factors other than p90(MDM2) determine the physiological consequences of p53 activation. Furthermore, reduction of mdm2 in vivo resulted in radiosensitivity, highlighting the importance of mdm2 as a potential target for adjuvant cancer therapies.


Assuntos
Regulação Neoplásica da Expressão Gênica , Linfócitos/citologia , Proteínas Nucleares , Proteínas Proto-Oncogênicas/fisiologia , Proteína Supressora de Tumor p53/metabolismo , Alelos , Animais , Apoptose , Northern Blotting , Western Blotting , Peso Corporal/genética , Morte Celular , Células Cultivadas , Relação Dose-Resposta à Radiação , Citometria de Fluxo , Deleção de Genes , Genótipo , Células-Tronco Hematopoéticas , Marcação In Situ das Extremidades Cortadas , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Modelos Genéticos , Testes de Precipitina , Estrutura Terciária de Proteína , Proteínas Proto-Oncogênicas c-mdm2 , RNA Mensageiro/metabolismo , Fatores de Tempo , Distribuição Tecidual , Transcrição Gênica , Ativação Transcricional , Proteína Supressora de Tumor p53/biossíntese , Proteína Supressora de Tumor p53/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...