Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 56
Filtrar
1.
EMBO J ; 38(20): e103322, 2019 10 15.
Artigo em Inglês | MEDLINE | ID: mdl-31553097

RESUMO

Loss of tumor suppression by the p53 protein involves altered or abrogated transcriptional activity resulting in a failure to mediate wild-type cellular responses including cell cycle arrest, senescence, and apoptosis. Timofeev et al (2019) make the fascinating finding that a novel p53 cooperativity mutation devoid of DNA binding results in no tumor suppression but surprising retention of an apoptotic response to chemotherapy and other treatments. This shows a need for rethinking how mutant p53-driven tumors are treated in the clinic.


Assuntos
Apoptose , Proteína Supressora de Tumor p53/genética , Carcinogênese , Pontos de Checagem do Ciclo Celular , Humanos , Mutação
2.
Genes Dev ; 27(17): 1868-85, 2013 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-24013501

RESUMO

The p53 tumor suppressor is a transcription factor that mediates varied cellular responses. The C terminus of p53 is subjected to multiple and diverse post-translational modifications. An attractive hypothesis is that differing sets of combinatorial modifications therein determine distinct cellular outcomes. To address this in vivo, a Trp53(ΔCTD/ΔCTD) mouse was generated in which the endogenous p53 is targeted and replaced with a truncated mutant lacking the C-terminal 24 amino acids. These Trp53(ΔCTD/ΔCTD) mice die within 2 wk post-partum with hematopoietic failure and impaired cerebellar development. Intriguingly, the C terminus acts via three distinct mechanisms to control p53-dependent gene expression depending on the tissue. First, in the bone marrow and thymus, the C terminus dampens p53 activity. Increased senescence in the Trp53(ΔCTD/ΔCTD) bone marrow is accompanied by up-regulation of Cdkn1 (p21). In the thymus, the C-terminal domain negatively regulates p53-dependent gene expression by inhibiting promoter occupancy. Here, the hyperactive p53(ΔCTD) induces apoptosis via enhanced expression of the proapoptotic Bbc3 (Puma) and Pmaip1 (Noxa). In the liver, a second mechanism prevails, since p53(ΔCTD) has wild-type DNA binding but impaired gene expression. Thus, the C terminus of p53 is needed in liver cells at a step subsequent to DNA binding. Finally, in the spleen, the C terminus controls p53 protein levels, with the overexpressed p53(ΔCTD) showing hyperactivity for gene expression. Thus, the C terminus of p53 regulates gene expression via multiple mechanisms depending on the tissue and target, and this leads to specific phenotypic effects in vivo.


Assuntos
Regulação da Expressão Gênica , Genes p53/genética , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/metabolismo , Animais , Apoptose/genética , Células da Medula Óssea/citologia , Células da Medula Óssea/metabolismo , Senescência Celular/genética , Cerebelo/crescimento & desenvolvimento , Cerebelo/metabolismo , Técnicas de Introdução de Genes , Crescimento e Desenvolvimento/genética , Fígado/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Mutação/genética , Ligação Proteica , Processamento de Proteína Pós-Traducional , Deleção de Sequência/genética , Timócitos/citologia , Timócitos/metabolismo , Fatores de Tempo
3.
Genes Dev ; 26(14): 1533-45, 2012 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-22802528

RESUMO

The p53 tumor suppressor protein is a transcription factor that exerts its effects on the cell cycle via regulation of gene expression. Although the mechanism of p53-dependent transcriptional activation has been well-studied, the molecular basis for p53-mediated repression has been elusive. The E2F family of transcription factors has been implicated in regulation of cell cycle-related genes, with E2F6, E2F7, and E2F8 playing key roles in repression. In response to cellular DNA damage, E2F7, but not E2F6 or E2F8, is up-regulated in a p53-dependent manner, with p53 being sufficient to increase expression of E2F7. Indeed, p53 occupies the promoter of the E2F7 gene after genotoxic stress, consistent with E2F7 being a novel p53 target. Ablation of E2F7 expression abrogates p53-dependent repression of a subset of its targets, including E2F1 and DHFR, in response to DNA damage. Furthermore, E2F7 occupancy of the E2F1 and DHFR promoters is detected, and expression of E2F7 is sufficient to inhibit cell proliferation. Taken together, these results show that p53-dependent transcriptional up-regulation of its target, E2F7, leads to repression of relevant gene expression. In turn, this E2F7-dependent mechanism contributes to p53-dependent cell cycle arrest in response to DNA damage.


Assuntos
Dano ao DNA , Fator de Transcrição E2F7/metabolismo , Transcrição Gênica , Proteína Supressora de Tumor p53/metabolismo , Regulação para Cima , Pontos de Checagem do Ciclo Celular/genética , Linhagem Celular Tumoral , Fator de Transcrição E2F7/genética , Humanos , Elementos de Resposta , Tetra-Hidrofolato Desidrogenase/biossíntese , Tetra-Hidrofolato Desidrogenase/genética , Proteína Supressora de Tumor p53/genética
4.
Genes Dev ; 24(15): 1580-9, 2010 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-20679392

RESUMO

Mdm2 has been well characterized as a negative regulator of the tumor suppressor p53. Recent studies have shown that Mdm2 is activated in response to a variety of oncogenic pathways independent of p53. Although its role as an oncogene via suppression of p53 function remains clear, growing evidence argues for p53-independent effects, as well as the remarkable possibility that Mdm2 has tumor suppressor functions in the appropriate context. Hence, Mdm2 is proving to be a key player in human cancer in its own right, and thus an important target for therapeutic intervention.


Assuntos
Genes Supressores de Tumor , Oncogenes , Proteínas Proto-Oncogênicas c-mdm2/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Animais , Apoptose , Diferenciação Celular , Proliferação de Células , Células Epiteliais/citologia , Regulação Neoplásica da Expressão Gênica , Humanos , Proteínas Proto-Oncogênicas c-mdm2/genética , Proteína Supressora de Tumor p53/genética
5.
Genes Dev ; 23(3): 261-4, 2009 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-19204113

RESUMO

The p53 tumor suppressor has been reported to be expressed as multiple isoforms. In this issue of Genes & Development, Chen and colleagues (pp. 278-290) use a zebrafish model to show that one such isoform, Delta113p53, is a target for transcriptional activation by p53 and can, in turn, inhibit the activity of the full-length p53 protein, establishing a novel negative feedback loop centered on the p53 locus itself. The challenge will be to determine whether these intriguing results in zebrafish will pave the way for new insights into several perplexing issues in p53 biology that will impact human disease.


Assuntos
Proteína Supressora de Tumor p53/metabolismo , Proteínas de Peixe-Zebra/metabolismo , Peixe-Zebra/genética , Peixe-Zebra/metabolismo , Sequência de Aminoácidos , Animais , Genes p53 , Humanos , Dados de Sequência Molecular , Isoformas de Proteínas/química , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Homologia de Sequência de Aminoácidos , Ativação Transcricional , Proteína Supressora de Tumor p53/química , Proteína Supressora de Tumor p53/genética , Peixe-Zebra/crescimento & desenvolvimento , Proteínas de Peixe-Zebra/química , Proteínas de Peixe-Zebra/genética
6.
EMBO Rep ; 14(5): 414-21, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-23588418

RESUMO

In response to cellular stress signals, the tumour suppressor p53 accumulates and triggers a host of antineoplastic responses. For instance, DNA damage activates two main p53-dependent responses: cell cycle arrest and attendant DNA repair or apoptosis (cell death). It is broadly accepted that, in response to DNA damage, the function of p53 as a sequence-specific transcription factor is crucial for tumour suppression. The molecular determinants, however, that favour the initiation of either a p53-dependent cell cycle arrest (life) or apoptotic (death) transcriptional programme remain elusive. Gaining a clear understanding of the mechanisms controlling cell fate determination by p53 could lead to the identification of molecular targets for therapy, which could selectively sensitize cancer cells to apoptosis. This review summarizes the literature addressing this important question in the field. Special emphasis is given to the role of the p53 response element, post-translational modifications and protein-protein interactions on cell fate decisions made by p53 in response to DNA damage.


Assuntos
Apoptose , Proteína Supressora de Tumor p53/fisiologia , Animais , Pontos de Checagem do Ciclo Celular , Regulação da Expressão Gênica , Humanos , Neoplasias/metabolismo , Regiões Promotoras Genéticas , Processamento de Proteína Pós-Traducional , Estresse Fisiológico , Transcrição Gênica
7.
Subcell Biochem ; 85: 235-46, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25201198

RESUMO

Mdm2 is best known as the primary negative regulator of p53, but a growing body of evidence suggests that Mdm2 also has a number of functions independent of its role in regulating p53. Although these functions are not yet well-characterized, they have been implicated in regulating of a number of cellular processes, including cell-cycle control, apoptosis, differentiation, genome stability, and transcription, among others. It appears that Mdm2 exerts these functions through a surprisingly wide variety of mechanisms. For example, it has been shown that Mdm2 can ubiquitinate alternative targets, can stimulate the activity of transcription factors, and can directly bind to mRNA to regulate its stability. Dysregulation of p53-independent functions could be responsible for the oncogenic properties of Mdm2 seen even in the absence of p53, and may explain why approximately 10 % of human tumors overexpress Mdm2 instead of inactivating p53 through other mechanisms. As the p53-independent functions of Mdm2 present novel targets for potential therapeutic interventions, fully characterizing these cellular and pathogenic roles of Mdm2 will be important in the study of tumor biology and the treatment of cancer.


Assuntos
Genes p53 , Proteínas Proto-Oncogênicas c-mdm2/fisiologia , Carcinogênese , Instabilidade Genômica , Humanos , Neoplasias/genética , Neoplasias/metabolismo , Neoplasias/patologia , Biossíntese de Proteínas , Transcrição Gênica , Ubiquitinação
9.
J Biol Chem ; 287(26): 22397-407, 2012 Jun 22.
Artigo em Inglês | MEDLINE | ID: mdl-22514277

RESUMO

The p53 gene encodes a transcription factor that is composed of several functional domains: the N-terminal transactivation domain, the central sequence-specific DNA binding domain, the tetramerization domain, and the highly basic C-terminal regulatory domain (CTD). The p53 CTD is a nonspecific DNA binding domain that is subject to extensive post-translational modifications. However, the functional significance of the p53 CTD remains unclear. The role of this domain in the regulation of p53 functions is explored by comparing the activity of ectopically expressed wild-type (WT) p53 protein to that of a truncated mutant lacking the 24 terminal amino acids (Δ24). Using quantitative real time PCR and chromatin Immuno-Precipitation experiments, a p53 CTD deletion is shown to alter the p53-dependent induction of a subset of its target genes due to impaired specific DNA binding. Moreover, p53-induced growth arrest and apoptosis both require an intact p53 CTD. These data indicate that the p53 CTD is a positive regulator of p53 tumor suppressor functions.


Assuntos
Regulação da Expressão Gênica , Proteína Supressora de Tumor p53/química , Aminoácidos/química , Apoptose , Bromodesoxiuridina/farmacologia , Linhagem Celular , Linhagem Celular Tumoral , Linhagem da Célula , Imunoprecipitação da Cromatina , DNA/metabolismo , Humanos , Microscopia de Fluorescência/métodos , Plasmídeos/metabolismo , Ligação Proteica , Estrutura Terciária de Proteína , Transcrição Gênica , Proteína Supressora de Tumor p53/metabolismo
10.
Biomedicines ; 11(4)2023 Apr 11.
Artigo em Inglês | MEDLINE | ID: mdl-37189770

RESUMO

TP53 mutations are less frequent in myelodysplastic syndromes (MDS) and acute myeloid leukemia (AML) than in solid tumors, except in secondary and therapy-related MDS/AMLs, and in cases with complex monosomal karyotype. As in solid tumors, missense mutations predominate, with the same hotspot mutated codons (particularly codons 175, 248, 273). As TP53-mutated MDS/AMLs are generally associated with complex chromosomal abnormalities, it is not always clear when TP53 mutations occur in the pathophysiological process. It is also uncertain in these MDS/AML cases, which often have inactivation of both TP53 alleles, if the missense mutation is only deleterious through the absence of a functional p53 protein, or through a potential dominant-negative effect, or finally a gain-of-function effect of mutant p53, as demonstrated in some solid tumors. Understanding when TP53 mutations occur in the disease course and how they are deleterious would help to design new treatments for those patients who generally show poor response to all therapeutic approaches.

11.
Cell Rep ; 42(3): 112216, 2023 03 28.
Artigo em Inglês | MEDLINE | ID: mdl-36924496

RESUMO

Thymus and spleen, in contrast to liver, are radiosensitive tissues in which p53-dependent apoptosis is triggered after whole-body radiation in vivo. Combined RNA sequencing (RNA-seq) and chromatin immunoprecipitation sequencing (ChIP-seq) analyses of radiation-treated mouse organs identifies both shared and tissue-specific p53 transcriptional responses. As expected, the p53 targets shared among thymus and spleen are enriched in apoptotic targets. The inability to upregulate these genes in the liver is not due to reduced gene occupancy. Use of an engineered mouse model shows that deletion of the C terminus of p53 can confer radiation-induced expression of p53 apoptotic targets in the liver with concomitant increased cell death. Global RNA-seq analysis reveals that an additional role of the C terminus is also needed for transcriptional activation of liver-specific p53 targets. It is hypothesized that both suppression of apoptotic gene expression combined with enhanced activation of liver-specific targets confers tissue-specific radio-resistance.


Assuntos
Sequenciamento de Cromatina por Imunoprecipitação , Proteína Supressora de Tumor p53 , Animais , Camundongos , Proteína Supressora de Tumor p53/metabolismo , RNA-Seq , Ativação Transcricional , Tolerância a Radiação
12.
Nat Cancer ; 4(4): 564-581, 2023 04.
Artigo em Inglês | MEDLINE | ID: mdl-36973430

RESUMO

Although the gain of function (GOF) of p53 mutants is well recognized, it remains unclear whether different p53 mutants share the same cofactors to induce GOFs. In a proteomic screen, we identified BACH1 as a cellular factor that recognizes the p53 DNA-binding domain depending on its mutation status. BACH1 strongly interacts with p53R175H but fails to effectively bind wild-type p53 or other hotspot mutants in vivo for functional regulation. Notably, p53R175H acts as a repressor for ferroptosis by abrogating BACH1-mediated downregulation of SLC7A11 to enhance tumor growth; conversely, p53R175H promotes BACH1-dependent tumor metastasis by upregulating expression of pro-metastatic targets. Mechanistically, p53R175H-mediated bidirectional regulation of BACH1 function is dependent on its ability to recruit the histone demethylase LSD2 to target promoters and differentially modulate transcription. These data demonstrate that BACH1 acts as a unique partner for p53R175H in executing its specific GOFs and suggest that different p53 mutants induce their GOFs through distinct mechanisms.


Assuntos
Mutação com Ganho de Função , Proteína Supressora de Tumor p53 , Regulação para Baixo , Mutação com Ganho de Função/genética , Mutação , Proteômica , Proteína Supressora de Tumor p53/genética , Fatores de Transcrição de Zíper de Leucina Básica/metabolismo
13.
Cancer Discov ; 13(12): 2632-2651, 2023 12 12.
Artigo em Inglês | MEDLINE | ID: mdl-37676642

RESUMO

TP53 mutations are frequent in esophageal squamous cell carcinoma (ESCC) and other SCCs and are associated with a proclivity for metastasis. Here, we report that colony-stimulating factor-1 (CSF-1) expression is upregulated significantly in a p53-R172H-dependent manner in metastatic lung lesions of ESCC. The p53-R172H-dependent CSF-1 signaling, through its cognate receptor CSF-1R, increases tumor cell invasion and lung metastasis, which in turn is mediated in part through Stat3 phosphorylation and epithelial-to-mesenchymal transition (EMT). In Trp53R172H tumor cells, p53 occupies the Csf-1 promoter. The Csf-1 locus is enriched with histone 3 lysine 27 acetylation (H3K27ac), which is likely permissive for fostering an interaction between bromodomain-containing domain 4 (BRD4) and p53-R172H to regulate Csf-1 transcription. Inhibition of BRD4 not only reduces tumor invasion and lung metastasis but also reduces circulating CSF-1 levels. Overall, our results establish a novel p53-R172H-dependent BRD4-CSF-1 axis that promotes ESCC lung metastasis and suggest avenues for therapeutic strategies for this difficult-to-treat disease. SIGNIFICANCE: The invasion-metastasis cascade is a recalcitrant barrier to effective cancer therapy. We establish that the p53-R172H-dependent BRD4-CSF-1 axis is a mediator of prometastatic properties, correlates with patient survival and tumor stages, and its inhibition significantly reduces tumor cell invasion and lung metastasis. This axis can be exploited for therapeutic advantage. This article is featured in Selected Articles from This Issue, p. 2489.


Assuntos
Neoplasias Esofágicas , Carcinoma de Células Escamosas do Esôfago , Neoplasias Pulmonares , Humanos , Linhagem Celular Tumoral , Movimento Celular , Proliferação de Células , Transição Epitelial-Mesenquimal/genética , Neoplasias Esofágicas/genética , Carcinoma de Células Escamosas do Esôfago/genética , Mutação com Ganho de Função , Regulação Neoplásica da Expressão Gênica , Neoplasias Pulmonares/genética , Fator Estimulador de Colônias de Macrófagos/genética , Fator Estimulador de Colônias de Macrófagos/metabolismo , Mutação , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/metabolismo
14.
EMBO Rep ; 11(3): 220-5, 2010 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-20134482

RESUMO

The F-box protein Skp2 and its isoform Skp2B are both overexpressed in breast cancers. Skp2 alters the activity of p53 by inhibiting its interaction with p300 and by promoting p300 degradation. Here, we report that Skp2B also attenuates the activity of p53; however, this effect is independent of p300, suggesting that another mechanism might be involved. Prohibitin, a protein reported to activate p53, was isolated in a two-hybrid screen with the carboxy-terminal domain unique to Skp2B. We observed that prohibitin is a new substrate of Skp2B and that the degradation of prohibitin is responsible for the attenuated activity of p53 in cells overexpressing Skp2B. Furthermore, we show that the activity of p53 is reduced in the mammary glands of Skp2B transgenic mice. This study indicates that both Skp2 and Skp2B attenuate p53 activity through different pathways, suggesting that amplification of the Skp2 locus represents a powerful mechanism to attenuate p53 function in cancer.


Assuntos
Regulação da Expressão Gênica , Proteínas Repressoras/antagonistas & inibidores , Proteínas Quinases Associadas a Fase S/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Animais , Linhagem Celular Tumoral , Proteína p300 Associada a E1A/metabolismo , Humanos , Camundongos , Camundongos Transgênicos , Modelos Biológicos , Proibitinas , Ligação Proteica , Estrutura Terciária de Proteína , RNA Interferente Pequeno/metabolismo , Técnicas do Sistema de Duplo-Híbrido
15.
EMBO Rep ; 10(1): 71-8, 2009 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19079133

RESUMO

Ku80 is important in the repair of DNA double-strand breaks by its essential function in non-homologous end-joining. The absence of Ku80 causes the accumulation of DNA damage and leads to premature ageing in mice. We showed that mouse embryonic fibroblasts (MEFs) from ku80(-/-) mice senesced rapidly with elevated levels of p53 and p21. Deletion of p21 delayed the early senescence phenotype in ku80(-/-) MEFs, despite an otherwise intact response of p53. In contrast to ku80(-/-)p53(-/-) mice, which die rapidly primarily from lymphomas, there was no significant increase in tumorigenesis in ku80(-/-)p21(-/-) mice. However, ku80(-/-)p21(-/-) mice showed no improvement with respect to rough fur coat or osteopaenia, and even showed a shortened lifespan compared with ku80(-/-) mice. These results show that the increased lifespan of ku80(-/-) MEFs owing to the loss of p21 is not associated with an improvement of the premature ageing phenotypes of ku80(-/-) mice observed at the organismal level.


Assuntos
Envelhecimento/fisiologia , Antígenos Nucleares/metabolismo , Senescência Celular/fisiologia , Inibidor de Quinase Dependente de Ciclina p21/deficiência , Inibidor de Quinase Dependente de Ciclina p21/metabolismo , Proteínas de Ligação a DNA/deficiência , Proteínas de Ligação a DNA/metabolismo , Animais , Antígenos Nucleares/genética , Linhagem Celular , Transformação Celular Neoplásica/genética , Transformação Celular Neoplásica/metabolismo , Transformação Celular Neoplásica/patologia , Inibidor de Quinase Dependente de Ciclina p21/genética , Proteínas de Ligação a DNA/genética , Fibroblastos , Fase G1 , Deleção de Genes , Autoantígeno Ku , Camundongos , Camundongos Knockout , Fenótipo , Taxa de Sobrevida , Proteína Supressora de Tumor p53/metabolismo
16.
Cancer Res ; 81(7): 1633-1634, 2021 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-34003788

RESUMO

Mdm2 and MdmX are two closely related proteins that have been well-characterized as negative regulators of the tumor suppressor p53. Their interplay and especially respective roles in ubiquitination and subsequent degradation of p53 have lacked clarity. Yang and colleagues now demonstrate an obligate role for MdmX in recruitment of the E2 ubiquitin ligase UbcH5c to the Mdm2-MdmX hetero-oligomer. The use of elegant genetically engineered mouse models ensures the biological relevance of their findings that have important implications for targeted therapies involving these key players in the p53 pathway.See related article by Yang et al., Cancer Res 2021;81:898-909.


Assuntos
Proteínas de Ciclo Celular , Proteína Supressora de Tumor p53 , Animais , Camundongos , Proteínas Nucleares/metabolismo , Ligação Proteica , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Proto-Oncogênicas c-mdm2/genética , Proteínas Proto-Oncogênicas c-mdm2/metabolismo , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/metabolismo
17.
Methods Mol Biol ; 2267: 159-163, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33786790

RESUMO

RNA interference (RNAi) is a cellular process involved in the silencing of genes, which makes RNAi important for observing and understanding the function of specific gene products. Short interfering RNA (siRNA) pathway is a RNAi pathway, where exogenous double stranded RNA is introduced to the cell and cleaved by an endoribonuclease, Dicer, to form siRNA, which interacts with a protein complex to scan mRNAs to bind to its complementary sequence. The binding of the siRNA to its complementary mRNA, the mRNA is cleaved and degraded by the cell, significantly reducing the levels of the target protein product. The discovery of this mechanism made it a powerful tool to use as a technique for therapeutics, agricultural biology, and cellular and molecular biology.


Assuntos
Técnicas de Silenciamento de Genes/métodos , Interferência de RNA , Animais , Ciclo Celular , Linhagem Celular , Humanos , RNA Mensageiro/genética , RNA Interferente Pequeno/genética
18.
Cancer Res ; 81(4): 935-944, 2021 02 15.
Artigo em Inglês | MEDLINE | ID: mdl-33323382

RESUMO

p53 is a short-lived protein with low basal levels under normal homeostasis conditions. However, upon DNA damage, levels of p53 dramatically increase for its activation. Although robust stabilization of p53 serves as a "trademark" for DNA damage responses, the requirement for such dramatic protein stabilization in tumor suppression has not been well addressed. Here we generated a mutant p53KQ mouse where all the C-terminal domain lysine residues were mutated to glutamines (K to Q mutations at K367, K369, K370, K378, K379, K383, and K384) to mimic constitutive acetylation of the p53 C-terminus. Because of p53 activation, p53KQ/KQ mice were perinatal lethal, yet this lethality was averted in p53KQ/- mice, which displayed normal postnatal development. Nevertheless, p53KQ/- mice died prematurely due to anemia and hematopoiesis failure. Further analyses indicated that expression of the acetylation-mimicking p53 mutant in vivo induces activation of p53 targets in various tissues without obviously increasing p53 levels. In the well-established pancreatic ductal adenocarcinoma (PDAC) mouse model, expression of the acetylation-mimicking p53-mutant protein effectively suppressed K-Ras-induced PDAC development in the absence of robust p53 stabilization. Together, our results provide proof-of-principle evidence that p53-mediated transcriptional function and tumor suppression can be achieved independently of its robust stabilization and reveal an alternative approach to activate p53 function for therapeutic purposes. SIGNIFICANCE: Although robust p53 stabilization is critical for acute p53 responses such as DNA damage, this study underscores the important role of low basal p53 protein levels in p53 activation and tumor suppression.


Assuntos
Proteína Supressora de Tumor p53/metabolismo , Proteína Supressora de Tumor p53/fisiologia , Acetilação , Animais , Apoptose/genética , Carcinoma Ductal Pancreático/genética , Carcinoma Ductal Pancreático/metabolismo , Carcinoma Ductal Pancreático/patologia , Células Cultivadas , Dano ao DNA/genética , Genes Supressores de Tumor/fisiologia , Lisina/metabolismo , Camundongos , Camundongos Transgênicos , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Proteínas Mutantes/fisiologia , Mutação , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/patologia , Processamento de Proteína Pós-Traducional/genética , Estabilidade Proteica , Ativação Transcricional/genética , Proteína Supressora de Tumor p53/química
19.
Nat Commun ; 12(1): 484, 2021 01 20.
Artigo em Inglês | MEDLINE | ID: mdl-33473123

RESUMO

The tumor suppressor p53 integrates stress response pathways by selectively engaging one of several potential transcriptomes, thereby triggering cell fate decisions (e.g., cell cycle arrest, apoptosis). Foundational to this process is the binding of tetrameric p53 to 20-bp response elements (REs) in the genome (RRRCWWGYYYN0-13RRRCWWGYYY). In general, REs at cell cycle arrest targets (e.g. p21) are of higher affinity than those at apoptosis targets (e.g., BAX). However, the RE sequence code underlying selectivity remains undeciphered. Here, we identify molecular mechanisms mediating p53 binding to high- and low-affinity REs by showing that key determinants of the code are embedded in the DNA shape. We further demonstrate that differences in minor/major groove widths, encoded by G/C or A/T bp content at positions 3, 8, 13, and 18 in the RE, determine distinct p53 DNA-binding modes by inducing different Arg248 and Lys120 conformations and interactions. The predictive capacity of this code was confirmed in vivo using genome editing at the BAX RE to interconvert the DNA-binding modes, transcription pattern, and cell fate outcome.


Assuntos
Diferenciação Celular/genética , Diferenciação Celular/fisiologia , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/metabolismo , Apoptose/genética , Ciclo Celular , Pontos de Checagem do Ciclo Celular , Linhagem Celular , DNA/química , Proteínas de Ligação a DNA , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , Modelos Moleculares , Conformação Molecular , Ligação Proteica/genética , Elementos de Resposta
20.
Mol Cancer Res ; 19(9): 1522-1533, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-34045312

RESUMO

p53 mutations that result in loss of transcriptional activity are commonly found in numerous types of cancer. While the majority of these are missense mutations that map within the central DNA-binding domain, truncations and/or frameshift mutations can also occur due to various nucleotide substitutions, insertions, or deletions. These changes result in mRNAs containing premature stop codons that are translated into a diverse group of C-terminally truncated proteins. Here we characterized three p53 frameshift mutant proteins expressed from the endogenous TP53 locus in U2OS osteosarcoma and HCT116 colorectal cancer cell lines. These mutants retain intact DNA-binding domains but display altered oligomerization properties. Despite their abnormally high expression levels, they are mostly transcriptionally inactive and unable to initiate a stimuli-induced transcriptional program characteristic of wild-type p53. However, one of these variant p53 proteins, I332fs*14, which resembles naturally expressed TAp53 isoforms ß and γ, retains some residual antiproliferative activity and can induce cellular senescence in HCT116 cells. Cells expressing this mutant also display decreased motility in migration assays. Hence, this p53 variant exhibits a combination of loss-of-gain and gain-of-function characteristics, distinguishing it from both wild type p53 and p53 loss. IMPLICATIONS: p53 frameshift mutants display a mixture of residual antiproliferative and neomorphic functions that may be differentially exploited for targeted therapy.


Assuntos
Biomarcadores Tumorais/genética , Neoplasias Colorretais/patologia , Mutação da Fase de Leitura , Regulação Neoplásica da Expressão Gênica , Mutação com Perda de Função , Osteossarcoma/patologia , Proteína Supressora de Tumor p53/genética , Apoptose , Neoplasias Ósseas/genética , Neoplasias Ósseas/patologia , Ciclo Celular , Movimento Celular , Proliferação de Células , Neoplasias Colorretais/genética , Humanos , Osteossarcoma/genética , Células Tumorais Cultivadas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA