Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Cell ; 157(3): 595-610, 2014 Apr 24.
Artículo en Inglés | MEDLINE | ID: mdl-24766807

RESUMEN

PTEN dysfunction plays a crucial role in the pathogenesis of hereditary and sporadic cancers. Here, we show that PTEN homodimerizes and, in this active conformation, exerts lipid phosphatase activity on PtdIns(3,4,5)P3. We demonstrate that catalytically inactive cancer-associated PTEN mutants heterodimerize with wild-type PTEN and constrain its phosphatase activity in a dominant-negative manner. To study the consequences of homo- and heterodimerization of wild-type and mutant PTEN in vivo, we generated Pten knockin mice harboring two cancer-associated PTEN mutations (PtenC124S and PtenG129E). Heterozygous Pten(C124S/+) and Pten(G129E/+) cells and tissues exhibit increased sensitivity to PI3-K/Akt activation compared to wild-type and Pten(+/-) counterparts, whereas this difference is no longer apparent between Pten(C124S/-) and Pten(-/-) cells. Notably, Pten KI mice are more tumor prone and display features reminiscent of complete Pten loss. Our findings reveal that PTEN loss and PTEN mutations are not synonymous and define a working model for the function and regulation of PTEN.


Asunto(s)
Fosfohidrolasa PTEN/genética , Fosfohidrolasa PTEN/metabolismo , Transducción de Señal , Animales , Embrión de Mamíferos/citología , Femenino , Humanos , Pérdida de Heterocigocidad , Masculino , Ratones , Mutación , Multimerización de Proteína , Proteínas Proto-Oncogénicas c-akt/metabolismo
2.
Cell ; 154(2): 311-324, 2013 Jul 18.
Artículo en Inglés | MEDLINE | ID: mdl-23830207

RESUMEN

Tumor cells metastasize to distant organs through genetic and epigenetic alterations, including changes in microRNA (miR) expression. Here we find miR-22 triggers epithelial-mesenchymal transition (EMT), enhances invasiveness and promotes metastasis in mouse xenografts. In a conditional mammary gland-specific transgenic (TG) mouse model, we show that miR-22 enhances mammary gland side-branching, expands the stem cell compartment, and promotes tumor development. Critically, miR-22 promotes aggressive metastatic disease in MMTV-miR-22 TG mice, as well as compound MMTV-neu or -PyVT-miR-22 TG mice. We demonstrate that miR-22 exerts its metastatic potential by silencing antimetastatic miR-200 through direct targeting of the TET (Ten eleven translocation) family of methylcytosine dioxygenases, thereby inhibiting demethylation of the mir-200 promoter. Finally, we show that miR-22 overexpression correlates with poor clinical outcomes and silencing of the TET-miR-200 axis in patients. Taken together, our findings implicate miR-22 as a crucial epigenetic modifier and promoter of EMT and breast cancer stemness toward metastasis.


Asunto(s)
Neoplasias de la Mama/patología , Ensamble y Desensamble de Cromatina , Transición Epitelial-Mesenquimal , Regulación Neoplásica de la Expresión Génica , MicroARNs/metabolismo , Metástasis de la Neoplasia , Células Madre Neoplásicas/metabolismo , 5-Metilcitosina/análogos & derivados , Animales , Neoplasias de la Mama/metabolismo , Citosina/análogos & derivados , Citosina/metabolismo , Humanos , Ratones , Ratones Transgénicos , Trasplante de Neoplasias , Proteínas Proto-Oncogénicas/metabolismo , Interferencia de ARN , Trasplante Heterólogo
3.
Cell ; 147(2): 382-95, 2011 Oct 14.
Artículo en Inglés | MEDLINE | ID: mdl-22000016

RESUMEN

We recently proposed that competitive endogenous RNAs (ceRNAs) sequester microRNAs to regulate mRNA transcripts containing common microRNA recognition elements (MREs). However, the functional role of ceRNAs in cancer remains unknown. Loss of PTEN, a tumor suppressor regulated by ceRNA activity, frequently occurs in melanoma. Here, we report the discovery of significant enrichment of putative PTEN ceRNAs among genes whose loss accelerates tumorigenesis following Sleeping Beauty insertional mutagenesis in a mouse model of melanoma. We validated several putative PTEN ceRNAs and further characterized one, the ZEB2 transcript. We show that ZEB2 modulates PTEN protein levels in a microRNA-dependent, protein coding-independent manner. Attenuation of ZEB2 expression activates the PI3K/AKT pathway, enhances cell transformation, and commonly occurs in human melanomas and other cancers expressing low PTEN levels. Our study genetically identifies multiple putative microRNA decoys for PTEN, validates ZEB2 mRNA as a bona fide PTEN ceRNA, and demonstrates that abrogated ZEB2 expression cooperates with BRAF(V600E) to promote melanomagenesis.


Asunto(s)
Proteínas de Homeodominio/genética , Melanoma/genética , Fosfohidrolasa PTEN/genética , Fosfohidrolasa PTEN/metabolismo , Proteínas Proto-Oncogénicas B-raf/genética , ARN Mensajero/metabolismo , Proteínas Represoras/genética , Regiones no Traducidas 3' , Animales , Modelos Animales de Enfermedad , Proteínas de Homeodominio/metabolismo , Humanos , Ratones , MicroARNs/metabolismo , Mutagénesis Insercional , Proteínas Represoras/metabolismo , Caja Homeótica 2 de Unión a E-Box con Dedos de Zinc
4.
MMWR Morb Mortal Wkly Rep ; 70(46): 1613-1616, 2021 Nov 19.
Artículo en Inglés | MEDLINE | ID: mdl-34793414

RESUMEN

Surges in COVID-19 cases have stressed hospital systems, negatively affected health care and public health infrastructures, and degraded national critical functions (1,2). Resource limitations, such as available hospital space, staffing, and supplies led some facilities to adopt crisis standards of care, the most extreme operating condition for hospitals, in which the focus of medical decision-making shifted from achieving the best outcomes for individual patients to addressing the immediate care needs of larger groups of patients (3). When hospitals deviated from conventional standards of care, many preventive and elective procedures were suspended, leading to the progression of serious conditions among some persons who would have benefitted from earlier diagnosis and intervention (4). During March-May 2020, U.S. emergency department visits declined by 23% for heart attacks, 20% for strokes, and 10% for diabetic emergencies (5). The Cybersecurity & Infrastructure Security Agency (CISA) COVID Task Force* examined the relationship between hospital strain and excess deaths during July 4, 2020-July 10, 2021, to assess the impact of COVID-19 surges on hospital system operations and potential effects on other critical infrastructure sectors and national critical functions. The study period included the months during which the highly transmissible SARS-CoV-2 B.1.617.2 (Delta) variant became predominant in the United States.† The negative binomial regression model used to calculate estimated deaths predicted that, if intensive care unit (ICU) bed use nationwide reached 75% capacity an estimated 12,000 additional excess deaths would occur nationally over the next 2 weeks. As hospitals exceed 100% ICU bed capacity, 80,000 excess deaths would be expected in the following 2 weeks. This analysis indicates the importance of controlling case growth and subsequent hospitalizations before severe strain. State, local, tribal, and territorial leaders could evaluate ways to reduce strain on public health and health care infrastructures, including implementing interventions to reduce overall disease prevalence such as vaccination and other prevention strategies, as well as ways to expand or enhance capacity during times of high disease prevalence.


Asunto(s)
COVID-19/epidemiología , Hospitales/estadística & datos numéricos , Mortalidad/tendencias , Pandemias , Adulto , Ocupación de Camas/estadística & datos numéricos , COVID-19/mortalidad , COVID-19/terapia , Humanos , Unidades de Cuidados Intensivos/estadística & datos numéricos , Estados Unidos/epidemiología
5.
Proc Natl Acad Sci U S A ; 113(38): 10583-8, 2016 09 20.
Artículo en Inglés | MEDLINE | ID: mdl-27601662

RESUMEN

The transcriptional repressor Capicua (Cic) controls tissue patterning and restricts organ growth, and has been recently implicated in several cancers. Cic has emerged as a primary sensor of signaling downstream of the receptor tyrosine kinase (RTK)/extracellular signal-regulated kinase (ERK) pathway, but how Cic activity is regulated in different cellular contexts remains poorly understood. We found that the kinase Minibrain (Mnb, ortholog of mammalian DYRK1A), acting through the adaptor protein Wings apart (Wap), physically interacts with and phosphorylates the Cic protein. Mnb and Wap inhibit Cic function by limiting its transcriptional repressor activity. Down-regulation of Cic by Mnb/Wap is necessary for promoting the growth of multiple organs, including the wings, eyes, and the brain, and for proper tissue patterning in the wing. We have thus uncovered a previously unknown mechanism of down-regulation of Cic activity by Mnb and Wap, which operates independently from the ERK-mediated control of Cic. Therefore, Cic functions as an integrator of upstream signals that are essential for tissue patterning and organ growth. Finally, because DYRK1A and CIC exhibit, respectively, prooncogenic vs. tumor suppressor activities in human oligodendroglioma, our results raise the possibility that DYRK1A may also down-regulate CIC in human cells.


Asunto(s)
Tipificación del Cuerpo/genética , Proteínas de Drosophila/genética , Drosophila/genética , Proteínas HMGB/genética , Proteínas Serina-Treonina Quinasas/genética , Proteínas Tirosina Quinasas/genética , Proteínas Represoras/genética , Proteínas Adaptadoras Transductoras de Señales/genética , Animales , Drosophila/crecimiento & desarrollo , Proteínas de Drosophila/biosíntesis , Regulación del Desarrollo de la Expresión Génica , Proteínas HMGB/biosíntesis , Humanos , Neoplasias/genética , Fosforilación , Proteínas Serina-Treonina Quinasas/biosíntesis , Proteínas Represoras/biosíntesis , Alas de Animales/crecimiento & desarrollo , Quinasas DyrK
6.
Dev Biol ; 422(1): 33-46, 2017 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-27940159

RESUMEN

The dmrt1 (doublesex and mab-3 related transcription factor 1) gene is a key regulator of sex determination and/or gonadal sex differentiation across metazoan animals. This is unusual given that sex determination genes are typically not well conserved. The mechanisms by which zebrafish sex is determined have remained elusive due to the lack of sex chromosomes and the complex polygenic nature of sex determination in domesticated strains. To investigate the role of dmrt1 in zebrafish sex determination and gonad development, we isolated mutations disrupting this gene. We found that the majority of dmrt1 mutant fish develop as fertile females suggesting a complete male-to-female sex reversal in mutant animals that would have otherwise developed as males. A small percentage of mutant animals became males, but were sterile and displayed testicular dysgenesis. Therefore zebrafish dmrt1 functions in male sex determination and testis development. Mutant males had aberrant gonadal development at the onset of gonadal sex-differentiation, displaying reduced oocyte apoptosis followed by development of intersex gonads and failed testis morphogenesis and spermatogenesis. By contrast, female ovaries developed normally. We found that Dmrt1 is necessary for normal transcriptional regulation of the amh (anti-Müllerian hormone) and foxl2 (forkhead box L2) genes, which are thought to be important for male or female sexual development respectively. Interestingly, we identified one dmrt1 mutant allele that co-operates with a linked segregation distorter locus to generate an apparent XY sex determination mechanism. We conclude that dmrt1 is dispensable for ovary development but necessary for testis development in zebrafish, and that dmrt1 promotes male development by transcriptionally regulating male and female genes as has been described in other animals. Furthermore, the strong sex-ratio bias caused by dmrt1 reduction-of-function points to potential mechanisms through which sex chromosomes may evolve.


Asunto(s)
Desarrollo Sexual , Testículo/embriología , Factores de Transcripción/fisiología , Pez Cebra/embriología , Animales , Femenino , Proteína Forkhead Box L2 , Factores de Transcripción Forkhead/análisis , Regulación del Desarrollo de la Expresión Génica , Masculino , Cromosomas Sexuales , Procesos de Determinación del Sexo , Diferenciación Sexual , Factores de Transcripción/genética , Proteínas de Pez Cebra/análisis
7.
Reproduction ; 157(4): 383-398, 2018 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-30763280

RESUMEN

Germ cell differentiation and maintenance relies on complex regulation of mitotic and meiotic progression. Cyclin-dependent kinases (CDKs) and their activating cyclin partners are known to have specialized roles in regulating cell cycle progression across tissues, including germ cells. Very little is known about CDK/cyclin function in zebrafish or the regulation of germ cell maintenance and differentiation. In a forward genetic screen for gonadogenesis defects in zebrafish, a mutation disrupting cdk21 (cyclin-dependent kinase 21) was identified, which caused gonad hypoplasia, reduced fertility and failure of female sex specification. The cdk21 gene is unique to fishes, though the encoded protein is related to the D-cyclin partners Cdk4 and Cdk6, which are known G1 cell cycle regulators. In the testis, cdk21 mutant germ cells exhibited cell cycle defects such as diminished proliferation, prolonged meiosis and delayed sperm differentiation. Furthermore, cdk21 mutants failed to maintain germ cells following breeding. Based on these findings, we propose that cdk21 regulates spermatogonial proliferation, progression through meiosis and germline stem cell activation in the testis. In addition, we investigated cdk4 and cdk6 in zebrafish development and found that each has distinct expression patterns in the gonads. Mutant analysis demonstrated that cdk6 was necessary for viability beyond larval stages. In contrast, cdk4 mutants were viable but were all male with low breeding success and sperm overabundance. Our analysis demonstrated that zebrafish harbor three genes of the cdk4/6 family, cdk4, cdk6 and cdk21, with cdk21 having an essential role in germ cell development in the testis.


Asunto(s)
Proliferación Celular , Quinasas Ciclina-Dependientes/metabolismo , Células Germinativas/fisiología , Meiosis , Proteínas de Pez Cebra/metabolismo , Pez Cebra/fisiología , Animales , Quinasas Ciclina-Dependientes/genética , Ciclinas/metabolismo , Femenino , Fase G1 , Células Germinativas/citología , Masculino , Oogénesis , Fosforilación , Espermatogénesis , Proteínas de Pez Cebra/genética
10.
Nat Genet ; 50(2): 206-218, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-29335545

RESUMEN

Lipids, either endogenously synthesized or exogenous, have been linked to human cancer. Here we found that PML is frequently co-deleted with PTEN in metastatic human prostate cancer (CaP). We demonstrated that conditional inactivation of Pml in the mouse prostate morphs indolent Pten-null tumors into lethal metastatic disease. We identified MAPK reactivation, subsequent hyperactivation of an aberrant SREBP prometastatic lipogenic program, and a distinctive lipidomic profile as key characteristic features of metastatic Pml and Pten double-null CaP. Furthermore, targeting SREBP in vivo by fatostatin blocked both tumor growth and distant metastasis. Importantly, a high-fat diet (HFD) induced lipid accumulation in prostate tumors and was sufficient to drive metastasis in a nonmetastatic Pten-null mouse model of CaP, and an SREBP signature was highly enriched in metastatic human CaP. Thus, our findings uncover a prometastatic lipogenic program and lend direct genetic and experimental support to the notion that a Western HFD can promote metastasis.


Asunto(s)
Lipogénesis/genética , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/patología , Proteínas de Unión a los Elementos Reguladores de Esteroles/fisiología , Animales , Línea Celular Tumoral , Proliferación Celular/genética , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/metabolismo , Humanos , Masculino , Redes y Vías Metabólicas/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Metástasis de la Neoplasia , Células PC-3 , Fosfohidrolasa PTEN/genética , Neoplasias de la Próstata/metabolismo , Proteínas de Unión a los Elementos Reguladores de Esteroles/genética
11.
PLoS One ; 12(10): e0185292, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-29023511

RESUMEN

The nuclear landscape plays an important role in the regulation of tissue and positional specific genes in embryonic and developing cells. Changes in this landscape can be dynamic, and are associated with the differentiation of cells during embryogenesis, and the de-differentiation of cells during induced pluripotent stem cell (iPSC) formation and in many cancers. However, tools to quantitatively characterize these changes are limited, especially in the in vivo context, where numerous tissue types are present and cells are arranged in multiple layers. Previous tools have been optimized for the monolayer nature of cultured cells. Therefore, we present a new algorithm to quantify the condensation of chromatin in two in vivo systems. We first developed this algorithm to quantify changes in chromatin compaction and validated it in differentiating spermatids in zebrafish testes. Our algorithm successfully detected the typical increase in chromatin compaction as these cells differentiate. We then employed the algorithm to quantify the changes that occur in amphibian limb cells as they participate in a regenerative response. We observed that the chromatin in the limb cells de-compacts as they contribute to the regenerating organ. We present this new tool as an open sourced software that can be readily accessed and optimized to quantify chromatin compaction in complex multi-layered samples.


Asunto(s)
Algoritmos , Núcleo Celular/metabolismo , Cromatina/química , Extremidades/embriología , Células Madre Pluripotentes Inducidas/metabolismo , Pez Cebra/genética , Ambystoma mexicanum , Animales , Diferenciación Celular , Células Cultivadas , Cromatina/metabolismo , Ensamble y Desensamble de Cromatina , Desarrollo Embrionario/fisiología , Células Madre Pluripotentes Inducidas/citología , Pez Cebra/crecimiento & desarrollo , Pez Cebra/metabolismo
12.
Oncotarget ; 5(4): 894-900, 2014 Feb 28.
Artículo en Inglés | MEDLINE | ID: mdl-24658595

RESUMEN

The PI3K/AKT pathway governs a plethora of cellular processes, including cell growth, proliferation, and metabolism, in response to growth factors and cytokines. By acting as a unique lipid phosphatase converting phosphatidylinositol-3,4,5,- trisphosphate (PIP3) to phosphatidylinositol-4,5,-bisphosphate (PIP2), phosphatase and tensin homolog (PTEN) acts as the major cellular suppressor of PI3K signaling and AKT activation. Recently, PI3K mutations and loss/mutation of PTEN have been characterized in human gallbladder tumors; whether aberrant PTEN/PI3K pathway plays a causal role in gallbladder carcinogenesis, however, remains unknown. Herein we show that in mice, deregulation of PI3K/AKT signaling is sufficient to transform gallbladder epithelial cells and trigger fully penetrant, highly proliferative gallbladder tumors characterized by high levels of phospho-AKT. Histopathologically, these mouse tumors faithfully resemble human adenomatous gallbladder lesions. The identification of PI3K pathway deregulation as both an early event in the neoplastic transformation of the gallbladder epithelium and a main mechanism of tumor growth in Pten heterozygous and Pten mutant mouse models provides a new framework for studying in vivo the efficacy of target therapies directed against the PI3K pathway, as advanced metastatic tumors are often addicted to "trunkular" mutations.


Asunto(s)
Neoplasias de la Vesícula Biliar/enzimología , Neoplasias de la Vesícula Biliar/patología , Fosfatidilinositol 3-Quinasas/metabolismo , Animales , Carcinogénesis , Procesos de Crecimiento Celular/fisiología , Neoplasias de la Vesícula Biliar/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Fosfohidrolasa PTEN/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal
13.
Cancer Discov ; 4(8): 896-904, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24866151

RESUMEN

UNLABELLED: Prostate cancer is the most prevalent cancer in males, and treatment options are limited for advanced forms of the disease. Loss of the PTEN and TP53 tumor suppressor genes is commonly observed in prostate cancer, whereas their compound loss is often observed in advanced prostate cancer. Here, we show that PARP inhibition triggers a p53-dependent cellular senescence in a PTEN-deficient setting in the prostate. Surprisingly, we also find that PARP-induced cellular senescence is morphed into an apoptotic response upon compound loss of PTEN and p53. We further show that superactivation of the prosurvival PI3K-AKT signaling pathway limits the efficacy of a PARP single-agent treatment, and that PARP and PI3K inhibitors effectively synergize to suppress tumorigenesis in human prostate cancer cell lines and in a Pten/Trp53-deficient mouse model of advanced prostate cancer. Our findings, therefore, identify a combinatorial treatment with PARP and PI3K inhibitors as an effective option for PTEN-deficient prostate cancer. SIGNIFICANCE: The paucity of therapeutic options in advanced prostate cancer displays an urgent need for the preclinical assessment of novel therapeutic strategies. We identified differential therapeutic vulnerabilities that emerge upon the loss of both PTEN and p53, and observed that combined inhibition of PARP and PI3K provides increased efficacy in hormone-insensitive advanced prostate cancer.


Asunto(s)
Elafina/genética , Fosfohidrolasa PTEN/genética , Poli(ADP-Ribosa) Polimerasas/genética , Neoplasias de la Próstata/tratamiento farmacológico , Proteína p53 Supresora de Tumor/genética , Animales , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Senescencia Celular/efectos de los fármacos , Elafina/antagonistas & inhibidores , Humanos , Masculino , Ratones , Terapia Molecular Dirigida , Inhibidores de Poli(ADP-Ribosa) Polimerasas , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/patología
14.
Cell Stem Cell ; 13(1): 87-101, 2013 Jul 03.
Artículo en Inglés | MEDLINE | ID: mdl-23827711

RESUMEN

MicroRNAs are frequently deregulated in cancer. Here we show that miR-22 is upregulated in myelodysplastic syndrome (MDS) and leukemia and its aberrant expression correlates with poor survival. To explore its role in hematopoietic stem cell function and malignancy, we generated transgenic mice conditionally expressing miR-22 in the hematopoietic compartment. These mice displayed reduced levels of global 5-hydroxymethylcytosine (5-hmC) and increased hematopoietic stem cell self-renewal accompanied by defective differentiation. Conversely, miR-22 inhibition blocked proliferation in both mouse and human leukemic cells. Over time, miR-22 transgenic mice developed MDS and hematological malignancies. We also identify TET2 as a key target of miR-22 in this context. Ectopic expression of TET2 suppressed the miR-22-induced phenotypes. Downregulation of TET2 protein also correlated with poor clinical outcomes and miR-22 overexpression in MDS patients. Our results therefore identify miR-22 as a potent proto-oncogene and suggest that aberrations in the miR-22/TET2 regulatory network are common in hematopoietic malignancies.


Asunto(s)
Transformación Celular Neoplásica/patología , Proteínas de Unión al ADN/metabolismo , Regulación Neoplásica de la Expresión Génica , Neoplasias Hematológicas/patología , Células Madre Hematopoyéticas/citología , MicroARNs/fisiología , Síndromes Mielodisplásicos/patología , Proteínas Proto-Oncogénicas/metabolismo , Animales , Western Blotting , Diferenciación Celular , Proliferación Celular , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/metabolismo , Proteínas de Unión al ADN/genética , Dioxigenasas , Citometría de Flujo , Técnica del Anticuerpo Fluorescente , Neoplasias Hematológicas/genética , Neoplasias Hematológicas/mortalidad , Células Madre Hematopoyéticas/metabolismo , Humanos , Técnicas para Inmunoenzimas , Luciferasas/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Síndromes Mielodisplásicos/genética , Síndromes Mielodisplásicos/mortalidad , Proto-Oncogenes Mas , Proteínas Proto-Oncogénicas/genética , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Tasa de Supervivencia
15.
Nat Genet ; 45(7): 747-55, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23727860

RESUMEN

Here we report an integrated analysis that leverages data from treatment of genetic mouse models of prostate cancer along with clinical data from patients to elucidate new mechanisms of castration resistance. We show that castration counteracts tumor progression in a Pten loss-driven mouse model of prostate cancer through the induction of apoptosis and proliferation block. Conversely, this response is bypassed with deletion of either Trp53 or Zbtb7a together with Pten, leading to the development of castration-resistant prostate cancer (CRPC). Mechanistically, the integrated acquisition of data from mouse models and patients identifies the expression patterns of XAF1, XIAP and SRD5A1 as a predictive and actionable signature for CRPC. Notably, we show that combined inhibition of XIAP, SRD5A1 and AR pathways overcomes castration resistance. Thus, our co-clinical approach facilitates the stratification of patients and the development of tailored and innovative therapeutic treatments.


Asunto(s)
Antagonistas de Andrógenos/uso terapéutico , Andrógenos/metabolismo , Neoplasias de la Próstata/terapia , Terapias en Investigación , Animales , Antineoplásicos/uso terapéutico , Benzamidas , Línea Celular Tumoral , Evaluación Preclínica de Medicamentos , Humanos , Masculino , Ratones , Ratones Transgénicos , Modelos Biológicos , Nitrilos , Orquiectomía , Fosfohidrolasa PTEN/genética , Feniltiohidantoína/análogos & derivados , Feniltiohidantoína/uso terapéutico , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/patología , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Investigación Biomédica Traslacional/métodos , Insuficiencia del Tratamiento
16.
Nat Genet ; 45(7): 739-746, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23727861

RESUMEN

Zbtb7a has previously been described as a powerful proto-oncogene. Here we unexpectedly demonstrate that Zbtb7a has a critical oncosuppressive role in the prostate. Prostate-specific inactivation of Zbtb7a leads to a marked acceleration of Pten loss-driven prostate tumorigenesis through bypass of Pten loss-induced cellular senescence (PICS). We show that ZBTB7A physically interacts with SOX9 and functionally antagonizes its transcriptional activity on key target genes such as MIA, which is involved in tumor cell invasion, and H19, a long noncoding RNA precursor for an RB-targeting microRNA. Inactivation of Zbtb7a in vivo leads to Rb downregulation, PICS bypass and invasive prostate cancer. Notably, we found that ZBTB7A is genetically lost, as well as downregulated at both the mRNA and protein levels, in a subset of human advanced prostate cancers. Thus, we identify ZBTB7A as a context-dependent cancer gene that can act as an oncogene in some contexts but also has oncosuppressive-like activity in PTEN-null tumors.


Asunto(s)
Senescencia Celular/genética , Proteínas de Unión al ADN/fisiología , Genes Supresores de Tumor , Neoplasias de la Próstata/patología , Factor de Transcripción SOX9/genética , Factores de Transcripción/fisiología , Animales , Línea Celular Tumoral , Transformación Celular Neoplásica/genética , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Regulación hacia Abajo/genética , Femenino , Regulación Neoplásica de la Expresión Génica/fisiología , Genes Supresores de Tumor/fisiología , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Modelos Biológicos , Invasividad Neoplásica , Fosfohidrolasa PTEN/genética , Fosfohidrolasa PTEN/metabolismo , Neoplasias de la Próstata/genética , Proto-Oncogenes Mas , Factor de Transcripción SOX9/metabolismo , Factor de Transcripción SOX9/fisiología , Transducción de Señal/genética , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
17.
Cell Stem Cell ; 10(3): 284-98, 2012 Mar 02.
Artículo en Inglés | MEDLINE | ID: mdl-22385656

RESUMEN

Transcription factors required for formation of embryonic tissues often maintain their expression in adult stem cell populations, but whether their function remains equivalent is not clear. Here we demonstrate critical and distinct roles for Sall4 in development of embryonic germ cells and differentiation of postnatal spermatogonial progenitor cells (SPCs). In differentiating SPCs, Sall4 levels transiently increase and Sall4 physically interacts with Plzf, a transcription factor exclusively required for adult stem cell maintenance. Mechanistically, Sall4 sequesters Plzf to noncognate chromatin domains to induce expression of Kit, a target of Plzf-mediated repression required for differentiation. Plzf in turn antagonizes Sall4 function by displacing Sall4 from cognate chromatin to induce Sall1 expression. Taken together, these data suggest that transcription factors required for embryonic tissue development postnatally take on distinct roles through interaction with opposing factors, which hence define properties of the adult stem cell compartment.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Células Germinativas/citología , Factores de Transcripción de Tipo Kruppel/metabolismo , Células Madre/metabolismo , Factores de Transcripción/metabolismo , Animales , Western Blotting , Células Cultivadas , Proteínas de Unión al ADN/genética , Embrión de Mamíferos , Femenino , Fibroblastos/citología , Fibroblastos/metabolismo , Fibroblastos/fisiología , Citometría de Flujo , Regulación del Desarrollo de la Expresión Génica , Factores de Transcripción de Tipo Kruppel/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Proteína de la Leucemia Promielocítica con Dedos de Zinc , Células Madre/citología , Factores de Transcripción/genética
18.
Cancer Res ; 70(15): 6225-32, 2010 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-20631067

RESUMEN

Latent endometrial carcinoma precancers are normal-appearing endometrial glands with sporadic loss of tumor suppressor gene function such as PTEN. Progression to carcinoma is inefficient and requires additional genetic damage that creates a histologic precursor lesion called endometrial intraepithelial neoplasia (EIN). In this study, we examined loss of PAX2 expression, a gene required for embryonic uterine development, during endometrial carcinogenesis. Normal proliferative, EIN, and malignant (endometrial adenocarcinoma) endometrial tissues were immunostained for PTEN and PAX2. Proliferative samples with loss of protein in at least one gland were scored as latent precancers. EIN and cancer lesions were scored by the majority pattern. Overall prevalence and topography of joint PAX2-PTEN expression loss was examined. The prevalence of PAX2 protein loss in the sequence of normal to precancer to cancer was 36%, 71%, and 77%, respectively, and for PTEN, it was 49%, 44%, and 68%, respectively. The normal endometrial prevalence of PAX2- or PTEN-deficient latent precancers was unaffected by biopsy indication, but increased significantly with age. Coincident loss of PAX2 and PTEN expression in an individual normal endometrium was seen in 21% of patients, but usually involved different glands. Coincident loss was more common in precancers (31%) and carcinoma (55%), in which case, both markers were protein null in an overlapping clonal distribution. PAX2 and PTEN protein loss occurs independently and accumulates with increasing age in latent precancers of normal premenopausal endometrium. Loss of function of both genes in an overlapping distribution characterizes the clinical emergence of a premalignant lesion which is carried forward to carcinoma.


Asunto(s)
Neoplasias Endometriales/genética , Factor de Transcripción PAX2/genética , Fosfohidrolasa PTEN/genética , Lesiones Precancerosas/genética , Adulto , Neoplasias Endometriales/metabolismo , Neoplasias Endometriales/patología , Femenino , Genes Supresores de Tumor , Humanos , Inmunohistoquímica , Persona de Mediana Edad , Factor de Transcripción PAX2/biosíntesis , Factor de Transcripción PAX2/deficiencia , Fosfohidrolasa PTEN/biosíntesis , Fosfohidrolasa PTEN/deficiencia , Lesiones Precancerosas/metabolismo , Lesiones Precancerosas/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA