Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
1.
Proc Natl Acad Sci U S A ; 116(35): 17429-17437, 2019 08 27.
Artículo en Inglés | MEDLINE | ID: mdl-31409715

RESUMEN

Gastrointestinal (GI) syndrome is a serious side effect and dose-limiting toxicity observed in patients undergoing lower-abdominal radiotherapy. Previous mouse studies show that p53 gene dosage determines susceptibility to GI syndrome development. However, the translational relevance of p53 activity has not been addressed. Here, we used a knock-in mouse in which the p53-Mdm2 negative feedback loop is genetically disrupted. These mice retain biallelic p53 and thus, normal basal p53 levels and activity. However, due to the lack of p53-mediated Mdm2 transcription, irradiated Mdm2P2/P2 mice exhibit enhanced acute p53 activity, which protects them from GI failure. Intestinal crypt cells residing in the +4 and higher positions exhibit decreased apoptosis, increased p21 expression, and hyperproliferation to reinstate intestinal integrity. Correspondingly, pharmacological augmentation of p53 activity in wild-type mice with an Mdm2 inhibitor protects against GI toxicity without affecting therapeutic outcome. Our results suggest that transient disruption of the p53-Mdm2 interaction to enhance p53 activity could be a viable prophylactic strategy for alleviating GI syndrome in patients undergoing radiotherapy.


Asunto(s)
Enfermedades Gastrointestinales/etiología , Tracto Gastrointestinal/metabolismo , Tracto Gastrointestinal/efectos de la radiación , Traumatismos por Radiación/metabolismo , Radiación Ionizante , Proteína p53 Supresora de Tumor/metabolismo , Animales , Apoptosis/genética , Línea Celular Tumoral , Modelos Animales de Enfermedad , Enfermedades Gastrointestinales/metabolismo , Enfermedades Gastrointestinales/mortalidad , Enfermedades Gastrointestinales/patología , Tracto Gastrointestinal/patología , Humanos , Ratones , Ratones Noqueados , Modelos Biológicos , Traumatismos por Radiación/genética , Traumatismos por Radiación/mortalidad , Traumatismos por Radiación/patología , Traumatismos Experimentales por Radiación , Proteína p53 Supresora de Tumor/genética , Proteínas ras/genética , Proteínas ras/metabolismo
2.
Proc Natl Acad Sci U S A ; 116(3): 960-969, 2019 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-30593561

RESUMEN

DICER1 gene alterations and decreased expression are associated with developmental disorders and diseases in humans. Oscillation of Dicer1 phosphorylation and dephosphorylation regulates its function during the oocyte-to-embryo transition in Caenorhabditis elegans Dicer1 is also phosphorylated upon FGF stimulation at conserved serines in mouse embryonic fibroblasts and HEK293 cells. However, whether phosphorylation of Dicer1 has a role in mammalian development remains unknown. To investigate the consequence of constitutive phosphorylation, we generated phosphomimetic knock-in mouse models by replacing conserved serines 1712 and 1836 with aspartic acids individually or together. Dicer1S1836D/S1836D mice display highly penetrant postnatal lethality, and the few survivors display accelerated aging and infertility. Homozygous dual-phosphomimetic Dicer1 augments these defects, alters metabolism-associated miRNAs, and causes a hypermetabolic phenotype. Thus, constitutive phosphorylation of Dicer1 results in multiple pathologic processes in mice, indicating that phosphorylation tightly regulates Dicer1 function and activity in mammals.


Asunto(s)
Envejecimiento , ARN Helicasas DEAD-box , Homocigoto , Mutación Missense , Ribonucleasa III , Envejecimiento/genética , Envejecimiento/metabolismo , Sustitución de Aminoácidos , Animales , Caenorhabditis elegans , Proteínas de Caenorhabditis elegans , ARN Helicasas DEAD-box/genética , ARN Helicasas DEAD-box/metabolismo , Femenino , Técnicas de Sustitución del Gen , Células HEK293 , Humanos , Masculino , Ratones , Fosforilación/genética , Ribonucleasa III/genética , Ribonucleasa III/metabolismo
3.
Genes Dev ; 28(16): 1739-51, 2014 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-25128494

RESUMEN

The ubiquitin proteasome pathway is critical in restraining the activities of the p53 tumor suppressor. Numerous E3 and E4 ligases regulate p53 levels. Additionally, deubquitinating enzymes that modify p53 directly or indirectly also impact p53 function. When alterations of these proteins result in increased p53 activity, cells arrest in the cell cycle, senesce, or apoptose. On the other hand, alterations that result in decreased p53 levels yield tumor-prone phenotypes. This review focuses on the physiological relevance of these important regulators of p53 and their therapeutic implications.


Asunto(s)
Complejo de la Endopetidasa Proteasomal/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Ubiquitina/metabolismo , Animales , Humanos , Estabilidad Proteica , Proteínas Proto-Oncogénicas c-mdm2/metabolismo , Proteína p53 Supresora de Tumor/genética , Ubiquitinación
4.
Proc Natl Acad Sci U S A ; 115(9): 2198-2203, 2018 02 27.
Artículo en Inglés | MEDLINE | ID: mdl-29440484

RESUMEN

TP53 mutations occur in ∼50% of all human tumors, with increased frequency in aggressive cancers that are notoriously difficult to treat. Additionally, p53 missense mutations are remarkably predictive of refractoriness to chemo/radiotherapy in various malignancies. These observations have led to the development of mutant p53-targeting agents that restore p53 function. An important unknown is which p53-mutant tumors will respond to p53 reactivation-based therapies. Here, we found a heterogeneous impact on therapeutic response to p53 restoration, suggesting that it will unlikely be effective as a monotherapy. Through gene expression profiling of p53R172H -mutant lymphomas, we identified retinoic acid receptor gamma (RARγ) as an actionable target and demonstrated that pharmacological activation of RARγ with a synthetic retinoid sensitizes resistant p53-mutant lymphomas to p53 restoration, while additively improving outcome and survival in inherently sensitive tumors.


Asunto(s)
Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Neoplasias Experimentales/tratamiento farmacológico , Retinoides/farmacología , Tamoxifeno/farmacología , Proteína p53 Supresora de Tumor/metabolismo , Animales , Antineoplásicos Hormonales/administración & dosificación , Antineoplásicos Hormonales/farmacocinética , Antineoplásicos Hormonales/farmacología , Resistencia a Antineoplásicos , Sinergismo Farmacológico , Ratones , Ratones Endogámicos , Mutación Missense , Retinoides/administración & dosificación , Retinoides/farmacocinética , Tamoxifeno/administración & dosificación , Tamoxifeno/farmacocinética , Transcriptoma , Factor de Necrosis Tumoral alfa/metabolismo , Proteína p53 Supresora de Tumor/genética
5.
Genes Dev ; 27(17): 1857-67, 2013 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-23973961

RESUMEN

The p53-Mdm2 feedback loop is perceived to be critical for regulating stress-induced p53 activity and levels. However, this has never been tested in vivo. Using a genetically engineered mouse with mutated p53 response elements in the Mdm2 P2 promoter, we show that feedback loop-deficient Mdm2(P2/P2) mice are viable and aphenotypic and age normally. p53 degradation kinetics after DNA damage in radiosensitive tissues remains similar to wild-type controls. Nonetheless, DNA damage response is elevated in Mdm2(P2/P2) mice. Enhanced p53-dependent apoptosis sensitizes hematopoietic stem cells (HSCs), causing drastic myeloablation and lethality. These results suggest that while basal Mdm2 levels are sufficient to regulate p53 in most tissues under homeostatic conditions, the p53-Mdm2 feedback loop is critical for regulating p53 activity and sustaining HSC function after DNA damage. Therefore, transient disruption of p53-Mdm2 interaction could be explored as a potential adjuvant/therapeutic strategy for targeting stem cells in hematological malignancies.


Asunto(s)
Daño del ADN/genética , Retroalimentación Fisiológica , Longevidad/genética , Proteínas Proto-Oncogénicas c-mdm2/genética , Proteínas Proto-Oncogénicas c-mdm2/metabolismo , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo , Animales , Apoptosis/genética , Daño del ADN/efectos de la radiación , Técnicas de Sustitución del Gen , Células Madre Hematopoyéticas/efectos de la radiación , Ratones , Ratones Endogámicos C57BL , Mutación/genética , Regiones Promotoras Genéticas/genética , Unión Proteica/genética , Desnaturalización Proteica/efectos de la radiación , Estabilidad Proteica/efectos de la radiación , Tolerancia a Radiación/genética , Radiación Ionizante , Rayos Ultravioleta
6.
J Pathol ; 241(4): 501-510, 2017 03.
Artículo en Inglés | MEDLINE | ID: mdl-27925213

RESUMEN

The p53 inhibitor Mdm4 is present at high levels in multiple human cancers. Overexpression of Mdm4 in mice drives the spontaneous development of mostly lymphomas and sarcomas. In this study, we explored the ability of Mdm4 to cooperate with lesions in tumour development. The Mdm4 transgene contributed to mammary tumour development in a BALB/cJ background. High levels of Mdm4 enhanced tumour development in a mutant p53R172H heterozygous background, and reduced the need to lose the wild-type p53 allele, as compared with mice heterozygous only for the p53R172H mutation. Additionally, high levels of Mdm4 cooperated with an oncogenic K-ras mutation to drive lung tumourigenesis in vivo. Finally, we examined p53-independent functions of Mdm4 by studying the contribution of Mdm4 to tumour development in the absence of p53. Whereas the overall survival times of p53-null mice with and without the Mdm4 transgene were similar, male mice with both alterations showed significantly shorter survival than p53-null male mice, and showed differences in tumour spectrum, demonstrating a p53-independent function of Mdm4 in tumourigenesis. Furthermore, p53-null mice with the highest level of Mdm4 tended to have multiple tumours. Thus, a detailed analysis of Mdm4 transgenic mice in various genetic backgrounds shows synergy in tumour development in vivo. Mdm4 may thus serve as a therapeutic target in cancers. Copyright © 2016 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.


Asunto(s)
Carcinogénesis/genética , Linfoma/genética , Proteínas Proto-Oncogénicas/genética , Sarcoma/genética , Proteína p53 Supresora de Tumor/genética , Animales , Regulación Neoplásica de la Expresión Génica , Humanos , Pérdida de Heterocigocidad , Linfoma/patología , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Mutación , Proteínas Proto-Oncogénicas/metabolismo , Sarcoma/patología , Transgenes , Proteína p53 Supresora de Tumor/metabolismo
7.
Proc Natl Acad Sci U S A ; 111(30): 11145-50, 2014 Jul 29.
Artículo en Inglés | MEDLINE | ID: mdl-25024203

RESUMEN

p53(R172H/+) mice inherit a p53 mutation found in Li-Fraumeni syndrome and develop metastatic tumors at much higher frequency than p53(+/-) mice. To explore the mutant p53 metastatic phenotype, we used expression arrays to compare primary osteosarcomas from p53(R172H/+) mice with metastasis to osteosarcomas from p53(+/-) mice lacking metastasis. For this study, 213 genes were differentially expressed with a P value <0.05. Of particular interest, Pla2g16, which encodes a phospholipase that catalyzes phosphatidic acid into lysophosphatidic acid and free fatty acid (both implicated in metastasis), was increased in p53(R172H/+) osteosarcomas. Functional analyses showed that Pla2g16 knockdown decreased migration and invasion in mutant p53-expressing cells, and vice versa: overexpression of Pla2g16 increased the invasion of p53-null cells. Furthermore, Pla2g16 levels were increased upon expression of mutant p53 in both mouse and human osteosarcoma cell lines, indicating that Pla2g16 is a downstream target of the mutant p53 protein. ChIP analysis revealed that several mutant p53 proteins bind the Pla2g16 promoter at E26 transformation-specific (ETS) binding motifs and knockdown of ETS2 suppressed mutant p53 induction of Pla2g16. Thus, our study identifies a phospholipase as a transcriptional target of mutant p53 that is required for metastasis.


Asunto(s)
Neoplasias Óseas/metabolismo , Síndrome de Li-Fraumeni/metabolismo , Mutación , Osteosarcoma/metabolismo , Fosfolipasas A2 Calcio-Independiente/biosíntesis , Proteínas Supresoras de Tumor/biosíntesis , Animales , Neoplasias Óseas/genética , Neoplasias Óseas/patología , Línea Celular Tumoral , Humanos , Síndrome de Li-Fraumeni/genética , Síndrome de Li-Fraumeni/patología , Ratones , Ratones Mutantes , Invasividad Neoplásica , Osteosarcoma/genética , Osteosarcoma/patología , Fosfolipasas A2 Calcio-Independiente/genética , Elementos de Respuesta , Proteínas Supresoras de Tumor/genética
8.
J Hepatol ; 62(2): 371-9, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25281858

RESUMEN

BACKGROUND & AIMS: Aberrantly high expression of TRIM24 occurs in human cancers, including hepatocellular carcinoma. In contrast, TRIM24 in the mouse is reportedly a liver-specific tumour suppressor. To address this dichotomy and to uncover direct regulatory functions of TRIM24 in vivo, we developed a new mouse model that lacks expression of all Trim24 isoforms, as the previous model expressed normal levels of Trim24 lacking only exon 4. METHODS: To produce germline-deleted Trim24(dlE1) mice, deletion of the promoter and exon 1 of Trim24 was induced in Trim24(LoxP) mice by crossing with a zona pellucida 3-Cre line for global deletion. Liver-specific deletion (Trim24(hep)) was achieved by crossing with an albumin-Cre line. Phenotypic analyses were complemented by protein, gene-specific and global RNA expression analyses and quantitative chromatin immunoprecipitation. RESULTS: Global loss of Trim24 disrupted hepatic homeostasis in 100% of mice with highly significant, decreased expression of oxidation/reduction, steroid, fatty acid, and lipid metabolism genes, as well as increased expression of genes involved in unfolded protein response, endoplasmic reticulum stress and cell cycle pathways. Trim24(dlE1/dlE1) mice have markedly depleted visceral fat and, like Trim24(hep/hep) mice, spontaneously develop hepatic lipid-filled lesions, steatosis, hepatic injury, fibrosis and hepatocellular carcinoma. CONCLUSIONS: TRIM24, an epigenetic co-regulator of transcription, directly and indirectly represses hepatic lipid accumulation, inflammation, fibrosis and damage in the murine liver. Complete loss of Trim24 offers a model of human non-alcoholic fatty liver disease, steatosis, fibrosis and development of hepatocellular carcinoma in the absence of high-fat diet or obesity.


Asunto(s)
Carcinoma Hepatocelular/genética , Hígado Graso/genética , Regulación Neoplásica de la Expresión Génica , Lípidos/análisis , Neoplasias Hepáticas Experimentales/genética , Proteínas Nucleares/genética , ARN Neoplásico/genética , Factores de Transcripción/genética , Animales , Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/patología , Progresión de la Enfermedad , Hígado Graso/metabolismo , Hígado Graso/patología , Humanos , Hígado/metabolismo , Hígado/patología , Neoplasias Hepáticas Experimentales/metabolismo , Neoplasias Hepáticas Experimentales/patología , Ratones , Ratones Noqueados , Proteínas Nucleares/biosíntesis , Reacción en Cadena de la Polimerasa , Factores de Transcripción/biosíntesis
9.
Nat Genet ; 38(11): 1341-7, 2006 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-17033624

RESUMEN

Accumulating evidence converges on the possibility that chromosomes interact with each other to regulate transcription in trans. To systematically explore the epigenetic dimension of such interactions, we devised a strategy termed circular chromosome conformation capture (4C). This approach involves a circularization step that enables high-throughput screening of physical interactions between chromosomes without a preconceived idea of the interacting partners. Here we identify 114 unique sequences from all autosomes, several of which interact primarily with the maternally inherited H19 imprinting control region. Imprinted domains were strongly overrepresented in the library of 4C sequences, further highlighting the epigenetic nature of these interactions. Moreover, we found that the direct interaction between differentially methylated regions was linked to epigenetic regulation of transcription in trans. Finally, the patterns of interactions specific to the maternal H19 imprinting control region underwent reprogramming during in vitro maturation of embryonic stem cells. These observations shed new light on development, cancer epigenetics and the evolution of imprinting.


Asunto(s)
Cromosomas/química , Clonación Molecular/métodos , Epigénesis Genética/fisiología , Regulación de la Expresión Génica/genética , Animales , Animales Recién Nacidos , Sitios de Unión , Factor de Unión a CCCTC , Cromatina/química , Cromatina/metabolismo , Proteínas de Unión al ADN/metabolismo , Células Madre Embrionarias , Impresión Genómica/fisiología , Hígado/metabolismo , Ratones , Ratones Transgénicos , Modelos Biológicos , Conformación de Ácido Nucleico , Análisis de Secuencia por Matrices de Oligonucleótidos/métodos , ARN Largo no Codificante , ARN no Traducido/genética , Proteínas Represoras/metabolismo , Transactivadores
10.
Blood ; 120(26): 5118-27, 2012 Dec 20.
Artículo en Inglés | MEDLINE | ID: mdl-23018641

RESUMEN

Aberrations in the p53 tumor suppressor pathway are associated with hematologic malignancies. p53-dependent cell cycle control, senescence, and apoptosis functions are actively involved in maintaining hematopoietic homeostasis under normal and stress conditions. Whereas loss of p53 function promotes leukemia and lymphoma development in humans and mice, increased p53 activity inhibits hematopoietic stem cell function and results in myelodysplasia. Thus, exquisite regulation of p53 activity is critical for homeostasis. Most of our understanding of p53 function in hematopoiesis is derived from genetically engineered mice. Here we summarize some of these models, the various mechanisms that disrupt the regulation of p53 activity, and their relevance to human disease.


Asunto(s)
Enfermedades Hematológicas/genética , Hematopoyesis/genética , Ratones , Modelos Animales , Proteína p53 Supresora de Tumor/fisiología , Animales , Enfermedades Hematológicas/sangre , Enfermedades Hematológicas/patología , Hematopoyesis/fisiología , Humanos , Aprendizaje , Modelos Biológicos , Transducción de Señal/genética , Transducción de Señal/fisiología , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo
11.
Proc Natl Acad Sci U S A ; 108(29): 11995-2000, 2011 Jul 19.
Artículo en Inglés | MEDLINE | ID: mdl-21730132

RESUMEN

Mdm2 and Mdm4 are homologous RING domain-containing proteins that negatively regulate the tumor suppressor p53 under physiological and stress conditions. The RING domain of Mdm2 encodes an E3-ubiquitin ligase that promotes p53 degradation. In addition, Mdm2 and Mdm4 interact through their respective RING domains. The in vivo significance of Mdm2-Mdm4 heterodimerization in regulation of p53 function is unknown. In this study, we generated an Mdm4 conditional allele lacking the RING domain to investigate its role in Mdm2 and p53 regulation. Our results demonstrate that homozygous deletion of the Mdm4 RING domain results in prenatal lethality. Mechanistically, Mdm2-Mdm4 heterodimerization is critical for inhibiting lethal p53 activation during early embryogenesis. However, Mdm2-Mdm4 interaction is dispensable for regulating p53 activity as well as the stability of Mdm2 and p53 at later stages of development. We propose that Mdm4 is a key cofactor of Mdm2 that inhibits p53 activity primarily during early embryogenesis but is dispensable for regulating p53 and Mdm2 stability in the adult mouse.


Asunto(s)
Desarrollo Embrionario/fisiología , Regulación del Desarrollo de la Expresión Génica/genética , Multimerización de Proteína/genética , Proteínas Proto-Oncogénicas c-mdm2/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Animales , Southern Blotting , Radioisótopos de Cesio , Cartilla de ADN/genética , Desarrollo Embrionario/genética , Genotipo , Ratones , Ratones Transgénicos , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas c-mdm2/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Ubiquitina-Proteína Ligasas/genética , Ubiquitinación
12.
Nat Genet ; 36(10): 1105-10, 2004 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-15361875

RESUMEN

Chromatin insulators demarcate expression domains by blocking the cis effects of enhancers or silencers in a position-dependent manner. We show that the chromatin insulator protein CTCF carries a post-translational modification: poly(ADP-ribosyl)ation. Chromatin immunoprecipitation analysis showed that a poly(ADP-ribosyl)ation mark, which exclusively segregates with the maternal allele of the insulator domain in the H19 imprinting control region, requires the bases that are essential for interaction with CTCF. Chromatin immunoprecipitation-on-chip analysis documented that the link between CTCF and poly(ADP-ribosyl)ation extended to more than 140 mouse CTCF target sites. An insulator trap assay showed that the insulator function of most of these CTCF target sites is sensitive to 3-aminobenzamide, an inhibitor of poly(ADP-ribose) polymerase activity. We suggest that poly(ADP-ribosyl)ation imparts chromatin insulator properties to CTCF at both imprinted and nonimprinted loci, which has implications for the regulation of expression domains and their demise in pathological lesions.


Asunto(s)
Cromatina/metabolismo , Proteínas de Unión al ADN/metabolismo , Poli Adenosina Difosfato Ribosa/metabolismo , Proteínas Represoras/metabolismo , Animales , Factor de Unión a CCCTC , Cromatina/genética , Proteínas de Unión al ADN/genética , Epigénesis Genética , Femenino , Regulación de la Expresión Génica , Impresión Genómica , Humanos , Factor II del Crecimiento Similar a la Insulina/genética , Masculino , Análisis de Secuencia por Matrices de Oligonucleótidos , Procesamiento Proteico-Postraduccional , ARN Largo no Codificante , ARN no Traducido/genética , Proteínas Represoras/genética , Transcripción Genética
13.
Cell Death Differ ; 30(4): 897-905, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-36755072

RESUMEN

Transcription factors regulate hundreds of genes and p53 is no exception. As a stress responsive protein, p53 transactivates an array of downstream targets which define its role in maintaining physiological functions of cells/tissues. Despite decades of studies, our understanding of the p53 in vivo transcriptional program is still incomplete. Here we discuss some of the physiological stressors that activate p53, the pathological and physiological implications of p53 activation and the molecular profiling of the p53 transcriptional program in maintaining tissue homeostasis. We argue that the p53 transcriptional program is spatiotemporally regulated in a tissue-specific manner and define a p53 target signature that faithfully depicts p53 activity. We further emphasize that additional in vivo studies are needed to refine the p53 transactivation profile to harness it for therapeutic purposes.


Asunto(s)
Proteína p53 Supresora de Tumor , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo , Especificidad de Órganos , Activación Transcripcional/genética
14.
Cancer Discov ; 13(5): 1230-1249, 2023 05 04.
Artículo en Inglés | MEDLINE | ID: mdl-37067911

RESUMEN

Cancer-related alterations of the p53 tetramerization domain (TD) abrogate wild-type (WT) p53 function. They result in a protein that preferentially forms monomers or dimers, which are also normal p53 states under basal cellular conditions. However, their physiologic relevance is not well understood. We have established in vivo models for monomeric and dimeric p53, which model Li-Fraumeni syndrome patients with germline p53 TD alterations. p53 monomers are inactive forms of the protein. Unexpectedly, p53 dimers conferred some tumor suppression that is not mediated by canonical WT p53 activities. p53 dimers upregulate the PPAR pathway. These activities are associated with lower prevalence of thymic lymphomas and increased CD8+ T-cell differentiation. Lymphomas derived from dimeric p53 mice show cooperating alterations in the PPAR pathway, further implicating a role for these activities in tumor suppression. Our data reveal novel functions for p53 dimers and support the exploration of PPAR agonists as therapies. SIGNIFICANCE: New mouse models with TP53R342P (monomer) or TP53A347D (dimer) mutations mimic Li-Fraumeni syndrome. Although p53 monomers lack function, p53 dimers conferred noncanonical tumor-suppressive activities. We describe novel activities for p53 dimers facilitated by PPARs and propose these are "basal" p53 activities. See related commentary by Stieg et al., p. 1046. See related article by Choe et al., p. 1250. This article is highlighted in the In This Issue feature, p. 1027.


Asunto(s)
Síndrome de Li-Fraumeni , Animales , Ratones , Síndrome de Li-Fraumeni/genética , Síndrome de Li-Fraumeni/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Activación Transcripcional , Receptores Activados del Proliferador del Peroxisoma/genética , Receptores Activados del Proliferador del Peroxisoma/metabolismo , Muerte Celular
15.
Cancer Res ; 82(7): 1313-1320, 2022 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-35078816

RESUMEN

Murine double minute 2 (Mdm2) is the principal E3-ubiquitin ligase for p53 and contains a C2H2C4 type RING domain wherein the last cysteine residue is followed by an evolutionarily conserved 13 amino acid C-terminal tail. Previous studies have indicated that integrity of the C-terminal tail is critical for Mdm2 function. Recently, a mutation extending the MDM2 length by five amino acids was identified and associated with enhanced p53 response in fibroblasts and premature aging in a human patient. To investigate the importance of the conserved Mdm2 C-terminal length on p53 regulatory function in vivo, we engineered three novel mouse alleles using CRISPR-Cas9 technology. Genetic studies with these murine models showed that curtailing Mdm2 C-terminal length by even a single amino acid leads to p53-dependent embryonic lethality. Extension of the Mdm2 C-terminal length by five amino acids (QLTCL) yielded viable mice that are smaller in size, exhibit fertility problems, and have a shortened life span. Analysis of early passage mouse embryonic fibroblasts indicated impaired Mdm2 function correlates with enhanced p53 activity under stress conditions. Furthermore, analysis in mice showed tissue-specific alterations in p53 target gene expression and enhanced radiosensitivity. These results confirm the physiological importance of the evolutionarily conserved Mdm2 C-terminus in regulating p53 functions. SIGNIFICANCE: This in vivo study highlights that alterations to the C-terminus of Mdm2 perturb its regulation of the tumor suppressor p53.


Asunto(s)
Fibroblastos , Proteínas Proto-Oncogénicas c-mdm2 , Animales , Fibroblastos/metabolismo , Ratones , Mutación , Unión Proteica , Proteínas Proto-Oncogénicas c-mdm2/genética , Proteínas Proto-Oncogénicas c-mdm2/metabolismo , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo
16.
Cancer Res ; 82(10): 1926-1936, 2022 05 16.
Artículo en Inglés | MEDLINE | ID: mdl-35320355

RESUMEN

The majority of TP53 missense mutations identified in cancer patients are in the DNA-binding domain and are characterized as either structural or contact mutations. These missense mutations exhibit inhibitory effects on wild-type p53 activity. More importantly, these mutations also demonstrate gain-of-function (GOF) activities characterized by increased metastasis, poor prognosis, and drug resistance. To better understand the activities by which TP53 mutations, identified in Li-Fraumeni syndrome, contribute to tumorigenesis, we generated mice harboring a novel germline Trp53R245W allele (contact mutation) and compared them with existing models with Trp53R172H (structural mutation) and Trp53R270H (contact mutation) alleles. Thymocytes from heterozygous mice showed that all three hotspot mutations exhibited similar inhibitory effects on wild-type p53 transcription in vivo, and tumors from these mice had similar levels of loss of heterozygosity. However, the overall survival of Trp53R245W/+ and Trp53R270H/+ mice, but not Trp53R172H/+ mice, was significantly shorter than that of Trp53+/- mice, providing strong evidence for p53-mutant-specific GOF contributions to tumor development. Furthermore, Trp53R245W/+ and Trp53R270H/+ mice had more osteosarcoma metastases than Trp53R172H/+ mice, suggesting that these two contact mutants have stronger GOF in driving osteosarcoma metastasis. Transcriptomic analyses using RNA sequencing data from Trp53R172H/+, Trp53R245W/+, and Trp53R270H/+ primary osteosarcomas in comparison with Trp53+/- indicated that GOF of the three mutants was mediated by distinct pathways. Thus, both the inhibitory effect of mutant over wild-type p53 and GOF activities of mutant p53 contributed to tumorigenesis in vivo. Targeting p53 mutant-specific pathways may be important for therapeutic outcomes in osteosarcoma. SIGNIFICANCE: p53 hotspot mutants inhibit wild-type p53 similarly but differ in their GOF activities, with stronger tumor-promoting activity in contact mutants and distinct protein partners of each mutant driving tumorigenesis and metastasis.


Asunto(s)
Mutación con Ganancia de Función , Osteosarcoma , Proteína p53 Supresora de Tumor , Animales , Neoplasias Óseas/genética , Neoplasias Óseas/metabolismo , Neoplasias Óseas/patología , Carcinogénesis/genética , Línea Celular Tumoral , Transformación Celular Neoplásica , Ratones , Osteosarcoma/genética , Osteosarcoma/metabolismo , Osteosarcoma/patología , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo
17.
Cancer Res ; 79(10): 2662-2668, 2019 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-30914430

RESUMEN

Dicer1 functions as a tumor suppressor in mouse models. In humans, somatic mutations are associated with many cancers in adults, and patients with DICER1 syndrome with DICER1 germline mutations are susceptible to childhood cancers. Dicer is phosphorylated by the ERK-MAP kinase pathway and because this pathway is activated in human cancers, we asked whether phosphorylated Dicer1 contributed to tumor development. In human endometrioid cancers, we discovered that phosphorylated DICER1 is significantly associated with invasive disease. To test a direct involvement of Dicer1 phosphorylation in tumor development, we studied mice with phosphomimetic alterations at the two conserved serines phosphorylated by ERK and discovered that a phosphomimetic Dicer1 drives tumor development and dissemination in two independent murine cancer models (KRas+/LA1 and p53+/- ). Our findings demonstrate that phosphomimetic Dicer1 promotes tumor development and invasion. SIGNIFICANCE: This work highlights the relevance of Dicer1 phosphorylation in mammalian tumor development and dissemination.


Asunto(s)
Carcinogénesis/genética , ARN Helicasas DEAD-box/genética , Neoplasias/genética , Neoplasias/patología , Ribonucleasa III/genética , Animales , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Sistema de Señalización de MAP Quinasas/genética , Ratones , Ratones Endogámicos C57BL , Invasividad Neoplásica/genética , Invasividad Neoplásica/patología , Fosforilación/genética , Transducción de Señal/genética
18.
Mol Cancer Res ; 16(10): 1523-1529, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-29903771

RESUMEN

Mutations in the death domain-associated protein (DAXX) have been recently identified in a substantial proportion of human pancreatic neuroendocrine tumors (PanNETs). Remarkably, however, little is known about the physiologic role(s) of DAXX despite in vitro studies suggesting potential functions. Most prominently, and supported by tumor sequencing data, DAXX functions in concert with alpha thalassemia/mental retardation X-linked (ATRX) as a histone chaperone complex for the H3.3 variant. Studies have also identified potential roles in apoptosis, transcription, and negative regulation of the p53 tumor suppressor pathway. Herein, a mouse modeling approach was used to specifically address the latter and no significant genetic interaction between Daxx and the p53 pathway was determined. The embryonic lethal phenotype of Daxx loss is not p53-dependent. In addition, Daxx heterozygosity does not sensitize mice to a sublethal dose of ionizing radiation or alter the survival or tumor phenotype of Mdm2 transgenic mice. However, the data support a tumor suppressor role for DAXX as low-dose ionizing radiation produced a higher proportion of carcinomas in Daxx heterozygous mice than wild-type controls.Implications: While DAXX has important in vivo functions, they are independent of an inhibitory role on the p53 tumor suppressor pathway. Mol Cancer Res; 16(10); 1523-9. ©2018 AACR.


Asunto(s)
Proteínas Portadoras/genética , Péptidos y Proteínas de Señalización Intracelular/genética , Tumores Neuroendocrinos/genética , Proteínas Nucleares/genética , Neoplasias Pancreáticas/genética , Proteína p53 Supresora de Tumor/genética , Animales , Apoptosis/genética , Proteínas Co-Represoras , Modelos Animales de Enfermedad , Chaperonas de Histonas/genética , Histonas/genética , Humanos , Ratones , Ratones Transgénicos , Chaperonas Moleculares , Tumores Neuroendocrinos/patología , Tumores Neuroendocrinos/radioterapia , Neoplasias Pancreáticas/patología , Neoplasias Pancreáticas/radioterapia , Proteínas Proto-Oncogénicas c-mdm2/genética , Tolerancia a Radiación/genética , Transducción de Señal/efectos de la radiación , Proteína Nuclear Ligada al Cromosoma X/genética
19.
Nat Commun ; 9(1): 3953, 2018 09 27.
Artículo en Inglés | MEDLINE | ID: mdl-30262850

RESUMEN

TP53 mutations are the most frequent genetic alterations in breast cancer and are associated with more aggressive disease and worse overall survival. We have created two conditional mutant Trp53 alleles in the mouse that allow expression of Trp53R172H or Trp53R245W missense mutations in single cells surrounded by a normal stroma and immune system. Mice with Trp53 mutations in a few breast epithelial cells develop breast cancers with high similarity to human breast cancer including triple negative. p53R245W tumors are the most aggressive and exhibit metastases to lung and liver. Development of p53R172H breast tumors with some metastases requires additional hits. Sequencing of primary tumors and metastases shows p53R245W drives a parallel evolutionary pattern of metastases. These in vivo models most closely simulate the genesis of human breast cancer and will thus be invaluable in testing novel therapeutic options.


Asunto(s)
Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Neoplasias Mamarias Animales/genética , Neoplasias Mamarias Animales/patología , Mutación/genética , Proteína p53 Supresora de Tumor/genética , Alelos , Animales , Carcinogénesis/genética , Carcinogénesis/patología , Femenino , Heterogeneidad Genética , Humanos , Ratones Endogámicos C57BL , Metástasis de la Neoplasia
20.
Mol Cell Biol ; 24(8): 3497-504, 2004 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-15060168

RESUMEN

The differentially methylated imprinting control region (ICR) region upstream of the H19 gene regulates allelic Igf2 expression by means of a methylation-sensitive chromatin insulator function. We have previously shown that maternal inheritance of mutated (three of the four) target sites for the 11-zinc finger protein CTCF leads to loss of Igf2 imprinting. Here we show that a mutation in only CTCF site 4 also leads to robust activation of the maternal Igf2 allele despite a noticeably weaker interaction in vitro of site 4 DNA with CTCF compared to other ICR sites, sites 1 and 3. Moreover, maternally inherited sites 1 to 3 become de novo methylated in complex patterns in subpopulations of liver and heart cells with a mutated site 4, suggesting that the methylation privilege status of the maternal H19 ICR allele requires an interdependence between all four CTCF sites. In support of this conclusion, we show that CTCF molecules bind to each other both in vivo and in vitro, and we demonstrate strong interaction between two CTCF-DNA complexes, preassembled in vitro with sites 3 and 4. We propose that the CTCF sites may cooperate to jointly maintain both methylation-free status and insulator properties of the maternal H19 ICR allele. Considering many other CTCF targets, we propose that site-specific interactions between various DNA-bound CTCF molecules may provide general focal points in the organization of looped chromatin domains involved in gene regulation.


Asunto(s)
Proteínas de Unión al ADN/genética , Impresión Genómica , Factor II del Crecimiento Similar a la Insulina/genética , Mutación , ARN no Traducido/genética , Secuencias Reguladoras de Ácidos Nucleicos , Proteínas Represoras/genética , Animales , Factor de Unión a CCCTC , Islas de CpG , Metilación de ADN , Proteínas de Unión al ADN/metabolismo , Femenino , Regulación de la Expresión Génica , Factor II del Crecimiento Similar a la Insulina/metabolismo , Masculino , Metilación , Ratones , Unión Proteica , ARN Largo no Codificante , Proteínas Represoras/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA