Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 36
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
J Biol Chem ; 300(2): 105629, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38199563

RESUMEN

In contrast to stage-specific transcription factors, the role of ubiquitous transcription factors in neuronal development remains a matter of scrutiny. Here, we demonstrated that a ubiquitous factor NF-Y is essential for neural progenitor maintenance during brain morphogenesis. Deletion of the NF-YA subunit in neural progenitors by using nestin-cre transgene in mice resulted in significant abnormalities in brain morphology, including a thinner cerebral cortex and loss of striatum during embryogenesis. Detailed analyses revealed a progressive decline in multiple neural progenitors in the cerebral cortex and ganglionic eminences, accompanied by induced apoptotic cell death and reduced cell proliferation. In neural progenitors, the NF-YA short isoform lacking exon 3 is dominant and co-expressed with cell cycle genes. ChIP-seq analysis from the cortex during early corticogenesis revealed preferential binding of NF-Y to the cell cycle genes, some of which were confirmed to be downregulated following NF-YA deletion. Notably, the NF-YA short isoform disappears and is replaced by its long isoform during neuronal differentiation. Forced expression of the NF-YA long isoform in neural progenitors resulted in a significant decline in neuronal count, possibly due to the suppression of cell proliferation. Collectively, we elucidated a critical role of the NF-YA short isoform in maintaining neural progenitors, possibly by regulating cell proliferation and apoptosis. Moreover, we identified an isoform switch in NF-YA within the neuronal lineage in vivo, which may explain the stage-specific role of NF-Y during neuronal development.


Asunto(s)
Factor de Unión a CCAAT , Corteza Cerebral , Animales , Ratones , Factor de Unión a CCAAT/genética , Factor de Unión a CCAAT/metabolismo , Corteza Cerebral/citología , Corteza Cerebral/crecimiento & desarrollo , Corteza Cerebral/metabolismo , Regulación de la Expresión Génica , Neurogénesis , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Factores de Transcripción/metabolismo
2.
Cancer ; 124(18): 3693-3705, 2018 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-30289966

RESUMEN

BACKGROUND: Patients with advanced primary and recurrent salivary duct carcinoma (SDC), a rare and lethal malignancy, have limited therapeutic options. Novel small-molecule agents aimed at targeting critical signaling associated with SDC tumorigenesis may lead to new therapeutic options for patients with these tumors. The human epidermal growth factor receptor 2 (HER2)/phosphoinositide 3-kinase (PI3K) axis, an important oncogenic pathway, has been targeted for therapy in several solid tumors. Currently, little is known about the role and clinical implications of alterations of the HER2/PI3K pathway in patients with SDC. METHODS: The authors investigated the clinicopathologic features, genetic alterations, and expression of key members of the HER2/PI3K pathway in 43 primary tumors and conducted in vitro functional and targeted drug-response analyses on cell lines derived from salivary epithelial carcinomas. RESULTS: In primary tumors, loss of phosphatase and tensin homolog (PTEN) expression was identified in 22 of 43 tumors (51%), overexpression of HER2 was observed in 12 of 43 tumors (28%), and phosphatidylinositol-4,5-bisphosphate 3-kinase catalytic subunit alpha (PIK3CA) mutations were identified in 12 of 43 tumors (28%). Phosphorylated protein kinase B (p-AKT) was highly expressed in most tumors. Most tumors (70%) displayed mutually exclusive alterations of PI3K members, whereas 8 tumors (19%) had 2 or more concurrent abnormalities. In vitro studies demonstrated a direct association between PTEN loss and PI3K pathway activation and evidence of response to combined PI3Kα and PI3Kß and/or pan-PI3K inhibitors. CONCLUSIONS: The current analyses reveal frequent PTEN loss and mutually exclusive alterations of key PI3K pathway members in SDC and demonstrate in vitro evidence of a response to pan-PI3K inhibitors. These results provide a framework for a biomarker-based substratification of patients with SDC in future targeted therapy. Cancer 2018;124:3523-32. © 2018 American Cancer Society.


Asunto(s)
Carcinoma Ductal/terapia , Terapia Molecular Dirigida/métodos , Fosfohidrolasa PTEN/genética , Fosfatidilinositol 3-Quinasas/genética , Receptor ErbB-2/genética , Neoplasias de las Glándulas Salivales/terapia , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/metabolismo , Carcinoma Ductal/genética , Fosfatidilinositol 3-Quinasa Clase I/genética , Fosfatidilinositol 3-Quinasa Clase I/metabolismo , Eliminación de Gen , Frecuencia de los Genes , Células HEK293 , Humanos , Mutación , Fosfatidilinositol 3-Quinasas/metabolismo , Receptor ErbB-2/metabolismo , Medición de Riesgo , Neoplasias de las Glándulas Salivales/genética , Transducción de Señal/genética , Transcriptoma , Células Tumorales Cultivadas
3.
Mol Cell Proteomics ; 14(3): 471-83, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25527621

RESUMEN

A distinct feature of human prostate cancer (PCa) is the development of osteoblastic (bone-forming) bone metastases. Metastatic growth in the bone is supported by factors secreted by PCa cells that activate signaling networks in the tumor microenvironment that augment tumor growth. To better understand these signaling networks and identify potential targets for therapy of bone metastases, we characterized the secretome of a patient-derived xenograft, MDA-PCa-118b (PCa-118b), generated from osteoblastic bone lesion. PCa-118b induces osteoblastic tumors when implanted either in mouse femurs or subcutaneously. To study signaling molecules critical to these unique tumor/microenvironment-mediated events, we performed mass spectrometry on conditioned media of isolated PCa-118b tumor cells, and identified 26 secretory proteins, such as TGF-ß2, GDF15, FGF3, FGF19, CXCL1, galectins, and ß2-microglobulin, which represent both novel and previously published secreted proteins. RT-PCR using human versus mouse-specific primers showed that TGFß2, GDF15, FGF3, FGF19, and CXCL1 were secreted from PCa-118b cells. TGFß2, GDF15, FGF3, and FGF19 function as both autocrine and paracrine factors on tumor cells and stromal cells, that is, endothelial cells and osteoblasts. In contrast, CXCL1 functions as a paracrine factor through the CXCR2 receptor expressed on endothelial cells and osteoblasts. Thus, our study reveals a complex PCa bone metastasis secretome with paracrine and autocrine signaling functions that mediate cross-talk among multiple cell types within the tumor microenvironment.


Asunto(s)
Neoplasias Óseas/metabolismo , Neoplasias Óseas/secundario , Proteínas de Neoplasias/metabolismo , Neoplasias de la Próstata/metabolismo , Proteómica/métodos , Microambiente Tumoral , Animales , Neoplasias Óseas/patología , Comunicación Celular , Línea Celular Tumoral , Medios de Cultivo Condicionados/metabolismo , Humanos , Masculino , Ratones , Ratones SCID , Proteínas de Neoplasias/genética , Trasplante de Neoplasias , Neoplasias de la Próstata/patología , Transducción de Señal , Células del Estroma/fisiología
4.
Nucleic Acids Res ; 43(13): 6257-69, 2015 Jul 27.
Artículo en Inglés | MEDLINE | ID: mdl-26040697

RESUMEN

Roles for SOX9 have been extensively studied in development and particular emphasis has been placed on SOX9 roles in cell lineage determination in a number of discrete tissues. Aberrant expression of SOX9 in many cancers, including colorectal cancer, suggests roles in these diseases as well and recent studies have suggested tissue- and context-specific roles of SOX9. Our genome wide approach by chromatin immunoprecipitation sequencing (ChIP-seq) in human colorectal cancer cells identified a number of physiological targets of SOX9, including ubiquitously expressed cell cycle regulatory genes, such as CCNB1 and CCNB2, CDK1, and TOP2A. These novel high affinity-SOX9 binding peaks precisely overlapped with binding sites for histone-fold NF-Y transcription factor. Furthermore, our data showed that SOX9 is recruited by NF-Y to these promoters of cell cycle regulatory genes and that SOX9 is critical for the full function of NF-Y in activation of the cell cycle genes. Mutagenesis analysis and in vitro binding assays provided additional evidence to show that SOX9 affinity is through NF-Y and that SOX9 DNA binding domain is not necessary for SOX9 affinity to those target genes. Collectively, our results reveal possibly a context-dependent, non-classical regulatory role for SOX9.


Asunto(s)
Factor de Unión a CCAAT/metabolismo , Neoplasias Colorrectales/genética , Regulación Neoplásica de la Expresión Génica , Factor de Transcripción SOX9/metabolismo , Activación Transcripcional , Sitios de Unión , Línea Celular Tumoral , Neoplasias Colorrectales/metabolismo , Genoma Humano , Humanos , Regiones Promotoras Genéticas , Factor de Transcripción SOX9/fisiología
5.
Nucleic Acids Res ; 38(18): 6018-28, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20484372

RESUMEN

Mutations in SOX9, a gene essential for chondrocyte differentiation cause the human disease campomelic dysplasia (CD). To understand how SOX9 activates transcription, we characterized the DNA binding and cell-free transcription ability of wild-type SOX9 and a dimerization domain SOX9 mutant. Whereas formation of monomeric mutant SOX9-DNA complex increased linearly with increasing SOX9 concentrations, formation of a wild-type SOX9-DNA dimeric complex increased more slowly suggesting a more sigmoidal-type progression. Stability of SOX9-DNA complexes, however, was unaffected by the dimerization mutation. Both wild-type and mutant SOX9 activated transcription of a naked Col2a1 DNA template. However, after nucleosomal assembly, only wild-type and not the mutant was able to remodel chromatin and activate transcription of this template. Using a cell line, in which the Col2a1 vector was stably integrated, no differences were seen in the interactions of wild-type and mutant SOX9 with the chromatin of the Col2a1 vector using ChIP. However, the mutant was unable to activate transcription in agreement with in vitro results. We hypothesize that the SOX9 dimerization domain is necessary to remodel the Col2a1 chromatin in order to allow transcription to take place. These results further clarify the mechanism that accounts for CD in patients harboring SOX9 dimerization domain mutations.


Asunto(s)
Condrocitos/metabolismo , Ensamble y Desensamble de Cromatina , Factor de Transcripción SOX9/genética , Factor de Transcripción SOX9/metabolismo , Activación Transcripcional , Animales , Línea Celular , Cromatina/metabolismo , Colágeno Tipo II/genética , ADN/metabolismo , Dimerización , Elementos de Facilitación Genéticos , Humanos , Mutación , Estructura Terciaria de Proteína , Proteínas Recombinantes/metabolismo , Factor de Transcripción SOX9/química
6.
Cell Rep ; 39(1): 110595, 2022 04 05.
Artículo en Inglés | MEDLINE | ID: mdl-35385726

RESUMEN

Bioinformatic analysis of 94 patient-derived xenografts (PDXs), cell lines, and organoids (PCOs) identifies three intrinsic transcriptional subtypes of metastatic castration-resistant prostate cancer: androgen receptor (AR) pathway + prostate cancer (PC) (ARPC), mesenchymal and stem-like PC (MSPC), and neuroendocrine PC (NEPC). A sizable proportion of castration-resistant and metastatic stage PC (M-CRPC) cases are admixtures of ARPC and MSPC. Analysis of clinical datasets and mechanistic studies indicates that MSPC arises from ARPC as a consequence of therapy-induced lineage plasticity. AR blockade with enzalutamide induces (1) transcriptional silencing of TP53 and hence dedifferentiation to a hybrid epithelial and mesenchymal and stem-like state and (2) inhibition of BMP signaling, which promotes resistance to AR inhibition. Enzalutamide-tolerant LNCaP cells re-enter the cell cycle in response to neuregulin and generate metastasis in mice. Combined inhibition of HER2/3 and AR or mTORC1 exhibits efficacy in models of ARPC and MSPC or MSPC, respectively. These results define MSPC, trace its origin to therapy-induced lineage plasticity, and reveal its sensitivity to HER2/3 inhibition.


Asunto(s)
Antineoplásicos , Neoplasias de la Próstata Resistentes a la Castración , Transducción de Señal , Animales , Antineoplásicos/farmacología , Benzamidas , Carcinoma Neuroendocrino , Línea Celular Tumoral , Plasticidad de la Célula/efectos de los fármacos , Plasticidad de la Célula/fisiología , Resistencia a Antineoplásicos , Humanos , Masculino , Ratones , Células Madre Neoplásicas/efectos de los fármacos , Células Madre Neoplásicas/metabolismo , Nitrilos , Feniltiohidantoína , Neoplasias de la Próstata Resistentes a la Castración/tratamiento farmacológico , Neoplasias de la Próstata Resistentes a la Castración/genética , Neoplasias de la Próstata Resistentes a la Castración/metabolismo , Receptores Androgénicos/efectos de los fármacos , Receptores Androgénicos/metabolismo , Microambiente Tumoral/efectos de los fármacos , Microambiente Tumoral/fisiología
7.
Prostate ; 71(8): 846-56, 2011 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-21456067

RESUMEN

BACKGROUND: Small-cell carcinoma (SCC) of the prostate is an AR-negative variant of prostate cancer found at progression in 10-20% of castrate-resistant disease. Its finding predicts a distinct clinical course and a poor prognosis. Large-cell neuroendocrine carcinoma (LCNEC) is a much rarer variant that behaves similarly to SCC. The biological mechanisms that drive these disease variants are poorly understood. METHODS: Eight tumor fragments from the salvage pelvic exenteration specimen of a patient with castrate-resistant prostate carcinoma were subcutaneously implanted into 6- to 8-week-old male CB17 SCID mice. Serial tissue sections and tissue microarrays of the resulting MDA PCa 144 xenograft lines were used for histopathologic and immunohistochemical characterization of the xenografts and their tissue of origin. RNA from two representative xenograft sublines was used for gene-expression profiling. RESULTS: All eight fragments formed tumors: four of the MDA PCa 144 xenograft sublines had morphologic characteristics of SCC and four, of LCNEC. All retained high fidelity to their parent tumor tissue, which remained stable through serial passages. Morphological transitions in the specimen of origin suggested LCNEC represents an intermediate step between adenocarcinoma and SCC. Over 2,500 genes were differentially expressed between the SCC (MDA PCa 144-13) and the LCNEC (MDA PCa 144-4) sublines and enriched in "Nervous System Development" Gene Ontology subtree. CONCLUSION: The eight xenograft models described represent the spectrum of neuroendocrine carcinomas in prostate cancer and will be valuable preclinical tools to study the pathogenesis of and therapy targets for this increasingly recognized subset of lethal prostate cancer.


Asunto(s)
Carcinoma de Células Grandes/patología , Carcinoma Neuroendocrino/patología , Carcinoma de Células Pequeñas/patología , Neoplasias de la Próstata/patología , Ensayos Antitumor por Modelo de Xenoinjerto , Anciano , Animales , Antineoplásicos/uso terapéutico , Carcinoma de Células Grandes/tratamiento farmacológico , Carcinoma de Células Grandes/radioterapia , Carcinoma Neuroendocrino/tratamiento farmacológico , Carcinoma Neuroendocrino/radioterapia , Carcinoma de Células Pequeñas/tratamiento farmacológico , Carcinoma de Células Pequeñas/radioterapia , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Humanos , Masculino , Ratones , Ratones SCID , Antígeno Prostático Específico , Neoplasias de la Próstata/tratamiento farmacológico , Neoplasias de la Próstata/radioterapia
8.
BMC Cancer ; 10: 599, 2010 Nov 02.
Artículo en Inglés | MEDLINE | ID: mdl-21044312

RESUMEN

BACKGROUND: The genetic control of prostate cancer development is poorly understood. Large numbers of gene-expression datasets on different aspects of prostate tumorigenesis are available. We used these data to identify and prioritize candidate genes associated with the development of prostate cancer and bone metastases. Our working hypothesis was that combining meta-analyses on different but overlapping steps of prostate tumorigenesis will improve identification of genes associated with prostate cancer development. METHODS: A Z score-based meta-analysis of gene-expression data was used to identify candidate genes associated with prostate cancer development. To put together different datasets, we conducted a meta-analysis on 3 levels that follow the natural history of prostate cancer development. For experimental verification of candidates, we used in silico validation as well as in-house gene-expression data. RESULTS: Genes with experimental evidence of an association with prostate cancer development were overrepresented among our top candidates. The meta-analysis also identified a considerable number of novel candidate genes with no published evidence of a role in prostate cancer development. Functional annotation identified cytoskeleton, cell adhesion, extracellular matrix, and cell motility as the top functions associated with prostate cancer development. We identified 10 genes--CDC2, CCNA2, IGF1, EGR1, SRF, CTGF, CCL2, CAV1, SMAD4, and AURKA--that form hubs of the interaction network and therefore are likely to be primary drivers of prostate cancer development. CONCLUSIONS: By using this large 3-level meta-analysis of the gene-expression data to identify candidate genes associated with prostate cancer development, we have generated a list of candidate genes that may be a useful resource for researchers studying the molecular mechanisms underlying prostate cancer development.


Asunto(s)
Regulación Neoplásica de la Expresión Génica , Neoplasias de la Próstata/metabolismo , Algoritmos , Neoplasias Óseas/secundario , Perfilación de la Expresión Génica , Humanos , Masculino , Modelos Estadísticos , Metástasis de la Neoplasia , Fenotipo , Neoplasias de la Próstata/patología
9.
Sci Rep ; 10(1): 21714, 2020 12 10.
Artículo en Inglés | MEDLINE | ID: mdl-33303918

RESUMEN

A heterotrimeric transcription factor NF-Y is crucial for cell-cycle progression in various types of cells. In contrast, studies using NF-YA knockout mice have unveiled its essential role in endoplasmic reticulum (ER) homeostasis in neuronal cells. However, whether NF-Y modulates a different transcriptome to mediate distinct cellular functions remains obscure. Here, we knocked down NF-Y in two types of neuronal cells, neuro2a neuroblastoma cells and mouse brain striatal cells, and performed gene expression profiling. We found that down-regulated genes preferentially contained NF-Y-binding motifs in their proximal promoters, and notably enriched genes related to ER functions rather than those for cell cycle. This contrasts with the profiling data of HeLa and embryonic stem cells in which distinct down-regulation of cell cycle-related genes was observed. Clustering analysis further identified several functional clusters where populations of the down-regulated genes were highly distinct. Further analyses using chromatin immunoprecipitation and RNA-seq data revealed that the transcriptomic difference was not correlated with DNA binding of NF-Y but with splicing of NF-YA. These data suggest that neuronal cells have a different type of transcriptome in which ER-related genes are dominantly modulated by NF-Y, and imply that NF-YA splicing alteration could be involved in this cell type-specific gene modulation.


Asunto(s)
Factor de Unión a CCAAT/genética , Factor de Unión a CCAAT/fisiología , Ciclo Celular/genética , Neuronas/fisiología , Transcriptoma/genética , Empalme Alternativo , Animales , Retículo Endoplásmico/genética , Perfilación de la Expresión Génica , Células HeLa , Homeostasis/genética , Humanos , Ratones , Neuronas/metabolismo , Empalme del ARN
10.
Nat Commun ; 10(1): 2571, 2019 06 12.
Artículo en Inglés | MEDLINE | ID: mdl-31189930

RESUMEN

While the antiandrogen enzalutamide (Enz) extends the castration resistant prostate cancer (CRPC) patients' survival an extra 4.8 months, it might also result in some adverse effects via inducing the neuroendocrine differentiation (NED). Here we found that lncRNA-p21 is highly expressed in the NEPC patients derived xenograft tissues (NEPC-PDX). Results from cell lines and human clinical sample surveys also revealed that lncRNA-p21 expression is up-regulated in NEPC and Enz treatment could increase the lncRNA-p21 to induce the NED. Mechanism dissection revealed that Enz could promote the lncRNA-p21 transcription via altering the androgen receptor (AR) binding to different androgen-response-elements, which switch the EZH2 function from histone-methyltransferase to non-histone methyltransferase, consequently methylating the STAT3 to promote the NED. Preclinical studies using the PDX mouse model proved that EZH2 inhibitor could block the Enz-induced NED. Together, these results suggest targeting the Enz/AR/lncRNA-p21/EZH2/STAT3 signaling may help urologists to develop a treatment for better suppression of the human CRPC progression.


Asunto(s)
Antagonistas de Andrógenos/efectos adversos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Tumores Neuroendocrinos/patología , Feniltiohidantoína/análogos & derivados , Neoplasias de la Próstata Resistentes a la Castración/patología , ARN Largo no Codificante/metabolismo , Animales , Benzamidas , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/genética , Línea Celular Tumoral , Progresión de la Enfermedad , Proteína Potenciadora del Homólogo Zeste 2/metabolismo , Humanos , Masculino , Ratones , Ratones SCID , Células Neuroendocrinas/efectos de los fármacos , Células Neuroendocrinas/patología , Tumores Neuroendocrinos/tratamiento farmacológico , Tumores Neuroendocrinos/genética , Nitrilos , Feniltiohidantoína/efectos adversos , Próstata/citología , Próstata/efectos de los fármacos , Próstata/patología , Neoplasias de la Próstata Resistentes a la Castración/tratamiento farmacológico , Neoplasias de la Próstata Resistentes a la Castración/genética , Factor de Transcripción STAT3/metabolismo , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Regulación hacia Arriba/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto
11.
Mol Imaging Biol ; 21(1): 86-94, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-29748904

RESUMEN

PURPOSE: Androgen receptor (AR) signaling affects prostate cancer (PCa) growth, metabolism, and progression. Often, PCa progresses from androgen-sensitive to castration-resistant prostate cancer (CRPC) following androgen-deprivation therapy. Clinicopathologic and genomic characterizations of CRPC tumors lead to subdividing CRPC into two subtypes: (1) AR-dependent CRPC containing dysregulation of AR signaling alterations in AR such as amplification, point mutations, and/or generation of splice variants in the AR gene; and (2) an aggressive variant PCa (AVPC) subtype that is phenotypically similar to small cell prostate cancer and is defined by chemotherapy sensitivity, gain of neuroendocrine or pro-neural marker expression, loss of AR expression, and combined alterations of PTEN, TP53, and RB1 tumor suppressors. Previously, we reported patient-derived xenograft (PDX) animal models that contain characteristics of these CRPC subtypes. In this study, we have employed the PDX models to test metabolic alterations in the CRPC subtypes. PROCEDURES: Mass spectrometry and nuclear magnetic resonance analysis along with in vivo hyperpolarized 1-[13C]pyruvate spectroscopy experiments were performed on prostate PDX animal models. RESULTS: Using hyperpolarized 1-[13C]pyruvate conversion to 1-[13C]lactate in vivo as well as lactate measurements ex vivo, we have found increased lactate production in AR-dependent CRPC PDX models even under low-hormone levels (castrated mouse) compared to AR-negative AVPC PDX models. CONCLUSIONS: Our analysis underscores the potential of hyperpolarized metabolic imaging in determining the underlying biology and in vivo phenotyping of CRPC.


Asunto(s)
Ácido Láctico/metabolismo , Imagen por Resonancia Magnética/métodos , Espectroscopía de Resonancia Magnética/métodos , Neoplasias de la Próstata Resistentes a la Castración/diagnóstico , Ácido Pirúvico/metabolismo , Receptores Androgénicos/metabolismo , Animales , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Xenoinjertos , Humanos , Aumento de la Imagen/métodos , Ácido Láctico/análisis , Masculino , Ratones , Ratones SCID , Invasividad Neoplásica , Próstata/química , Próstata/diagnóstico por imagen , Próstata/metabolismo , Próstata/patología , Neoplasias de la Próstata Resistentes a la Castración/metabolismo , Neoplasias de la Próstata Resistentes a la Castración/patología , Ácido Pirúvico/análisis , Transducción de Señal/fisiología , Células Tumorales Cultivadas
12.
J Cell Biochem ; 104(5): 1708-23, 2008 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-18348279

RESUMEN

Previously the analysis of promoters regulated by endoplasmic reticulum (ER) stress identified a composite promoter element, ERSE that interacts with both CBF/NF-Y (CBF) and ATF6(N) transcription factors. This prompted us to investigate the underlying mechanism by which CBF, a ubiquitously binding transcription factor, specifically controls transcription activation during ER stress. The in vitro DNA binding study performed using purified recombinant proteins revealed that CBF specifically recruits ATF6(N) to ERSE DNA but it does not interact with ATF6(N) in absence of DNA binding. Inhibition of CBF binding resulted in a significant reduction of optimal transcription activation of cellular genes during ER stress. Analysis of cellular promoters by ChIP demonstrated that CBF is needed for recruitment of both ATF6(N) and TBP but not for either acetylation of histone H3-K9 or trimethylation of histone H3-K4 during ER stress. Together these study results reveal that CBF controls ER stress-inducible transcription through recruitment of both ATF6(N) and TBP but not through chromatin modifications. Our observations are in agreement with the results of recently published studies that have shown that CBF controls transcription of varieties of inducible promoters through recruitment of general transcription factors but not through acetylation of histone H4. These findings provide a paradigm of the function of CBF in inducible transcription.


Asunto(s)
Factor de Transcripción Activador 6/metabolismo , Factor de Unión a CCAAT/metabolismo , Retículo Endoplásmico/patología , Proteína de Unión a TATA-Box/metabolismo , Transcripción Genética , Secuencia de Bases , Inmunoprecipitación de Cromatina , ADN/metabolismo , Chaperón BiP del Retículo Endoplásmico , Regulación Neoplásica de la Expresión Génica , Células HeLa , Proteínas de Choque Térmico/genética , Humanos , Chaperonas Moleculares/genética , Datos de Secuencia Molecular , Regiones Promotoras Genéticas/genética , Unión Proteica , ARN Interferente Pequeño/metabolismo , Proteínas Recombinantes/aislamiento & purificación , Proteínas Recombinantes/metabolismo , Activación Transcripcional/genética
13.
Nucleic Acids Res ; 34(21): 6272-85, 2006.
Artículo en Inglés | MEDLINE | ID: mdl-17098936

RESUMEN

Previous studies showed that binding of the CBF/NF-Y (CBF) transcription factor to cellular promoters is essential for cell proliferation. This observation prompted us to investigate the function of CBF in relation to cell cycle progression and in cell-cycle-regulated transcription. In this study, we used a tetracycline-inducible adenoviral vector to express a truncated CBF-B subunit, Bdbd, lacking a transcription activation domain in various mammalian cell lines. The Bdbd polypeptide interacts with cellular CBF-A/CBF-C and binds to promoters containing CBF-binding sites. Interestingly, expression of Bdbd in various mammalian cells resulted in the inhibition of cell proliferation and specific cell cycle arrest at G2/M phase. Gene expression analysis demonstrated that the expression of Bdbd strongly suppressed cell cycle-dependent transcription activation of Cyclin B1, Aurora A and CDK1 genes, key regulators for cell cycle progression at G2/M phase. Chromatin immunoprecipitation analysis showed that Bdbd significantly inhibited binding of TATA-binding protein, TBP to both Cyclin B1 and Aurora A promoters, but did not inhibit binding of E2F3 activator to Cyclin B1 promoter. This study suggested that the activation domain of CBF-B plays an essential role in the transcription activation of Cyclin B1 and Aurora A genes at G2/M phase, thus regulating cell cycle progression at G2/M phase.


Asunto(s)
Factor de Unión a CCAAT/metabolismo , División Celular/genética , Ciclina B/genética , Fase G2/genética , Proteínas Serina-Treonina Quinasas/genética , Activación Transcripcional , Adenoviridae/genética , Aurora Quinasas , Factor de Unión a CCAAT/antagonistas & inhibidores , Factor de Unión a CCAAT/genética , Ciclina B1 , Vectores Genéticos , Células HeLa , Humanos , Regiones Promotoras Genéticas , Estructura Terciaria de Proteína , Eliminación de Secuencia , Proteína de Unión a TATA-Box/metabolismo , Tetraciclina/farmacología
14.
Front Oncol ; 8: 69, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29600194

RESUMEN

Combined loss of tumor suppressors (TSPs), PTEN, TP53, and RB1, is highly associated with small cell carcinoma of prostate phenotype. Recent genomic studies of human tumors as well as analyses in mouse genetic models have revealed a unique role for these TSPs in dictating epithelial lineage plasticity-a phenomenon that plays a critical role in the development of aggressive variant prostate cancer (PCa) and associated androgen therapy resistance. Here, we summarize recently published key observations on this topic and hypothesize a possible mechanism by which concurrent loss of TSPs could potentially regulate the PCa disease phenotype.

15.
Biochim Biophys Acta Gene Regul Mech ; 1860(5): 598-603, 2017 May.
Artículo en Inglés | MEDLINE | ID: mdl-27815195

RESUMEN

The CCAAT-binding factor CBF/NF-Y is needed for cell proliferation and early embryonic development. NF-Y can regulate the expression of different cell type-specific genes that are activated by various physiological signaling pathways. Dysregulation of NF-Y was observed in pathogenic conditions in humans such as scleroderma, neurodegenerative disease, and cancer. Conditional inactivation of the NF-YA gene in mice demonstrated that NF-Y activity is essential for normal tissue homeostasis, survival, and metabolic function. Altogether, NF-Y is an essential transcription factor that plays a critical role in mammalian development, from the early stages to adulthood, and in human pathogenesis. This article is part of a Special Issue entitled: Nuclear Factor Y in Development and Disease, edited by Prof. Roberto Mantovani.


Asunto(s)
Factor de Unión a CCAAT/metabolismo , Proteínas de Neoplasias/metabolismo , Neoplasias Experimentales/metabolismo , Enfermedades Neurodegenerativas/metabolismo , Esclerodermia Difusa/metabolismo , Animales , Factor de Unión a CCAAT/genética , Humanos , Ratones , Proteínas de Neoplasias/genética , Neoplasias Experimentales/genética , Enfermedades Neurodegenerativas/genética , Esclerodermia Difusa/genética
16.
Cell Rep ; 18(8): 1970-1981, 2017 02 21.
Artículo en Inglés | MEDLINE | ID: mdl-28228262

RESUMEN

Cell division cycle 6 (CDC6), an androgen receptor (AR) target gene, is implicated in regulating DNA replication and checkpoint mechanisms. CDC6 expression is increased during prostate cancer (PCa) progression and positively correlates with AR in PCa tissues. AR or CDC6 knockdown, together with AZD7762, a Chk1/2 inhibitor, results in decreased TopBP1-ATR-Chk1 signaling and markedly increased ataxia-telangiectasia-mutated (ATM) phosphorylation, a biomarker of DNA damage, and synergistically increases treatment efficacy. Combination treatment with the AR signaling inhibitor enzalutamide (ENZ) and the Chk1/2 inhibitor AZD7762 demonstrates synergy with regard to inhibition of AR-CDC6-ATR-Chk1 signaling, ATM phosphorylation induction, and apoptosis in VCaP (mutant p53) and LNCaP-C4-2b (wild-type p53) cells. CDC6 overexpression significantly reduced ENZ- and AZD7762-induced apoptosis. Additive or synergistic therapeutic activities are demonstrated in AR-positive animal xenograft models. These findings have important clinical implications, since they introduce a therapeutic strategy for AR-positive, metastatic, castration-resistant PCa, regardless of p53 status, through targeting AR-CDC6-ATR-Chk1 signaling.


Asunto(s)
Antagonistas de Receptores Androgénicos/farmacología , Proteínas de Ciclo Celular/metabolismo , Quinasa 1 Reguladora del Ciclo Celular (Checkpoint 1)/metabolismo , Daño del ADN/fisiología , Proteínas Nucleares/metabolismo , Neoplasias de la Próstata/metabolismo , Receptores Androgénicos/metabolismo , Animales , Apoptosis/efectos de los fármacos , Proteínas de la Ataxia Telangiectasia Mutada/metabolismo , Biomarcadores/metabolismo , Puntos de Control del Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Daño del ADN/efectos de los fármacos , Replicación del ADN/efectos de los fármacos , Proteínas de Unión al ADN/metabolismo , Humanos , Masculino , Ratones , Ratones Desnudos , Fosforilación/efectos de los fármacos , Neoplasias de la Próstata/tratamiento farmacológico , Transducción de Señal/efectos de los fármacos , Tiofenos/farmacología , Urea/análogos & derivados , Urea/farmacología
17.
Dev Cell ; 41(5): 467-480.e3, 2017 06 05.
Artículo en Inglés | MEDLINE | ID: mdl-28586644

RESUMEN

Prostate cancer (PCa) bone metastasis is frequently associated with bone-forming lesions, but the source of the osteoblastic lesions remains unclear. We show that the tumor-induced bone derives partly from tumor-associated endothelial cells that have undergone endothelial-to-osteoblast (EC-to-OSB) conversion. The tumor-associated osteoblasts in PCa bone metastasis specimens and patient-derived xenografts (PDXs) were found to co-express endothelial marker Tie-2. BMP4, identified in PDX-conditioned medium, promoted EC-to-OSB conversion of 2H11 endothelial cells. BMP4 overexpression in non-osteogenic C4-2b PCa cells led to ectopic bone formation under subcutaneous implantation. Tumor-induced bone was reduced in trigenic mice (Tie2cre/Osxf/f/SCID) with endothelial-specific deletion of osteoblast cell-fate determinant OSX compared with bigenic mice (Osxf/f/SCID). Thus, tumor-induced EC-to-OSB conversion is one mechanism that leads to osteoblastic bone metastasis of PCa.


Asunto(s)
Neoplasias Óseas/secundario , Diferenciación Celular , Endotelio Vascular/patología , Osteoblastos/patología , Neoplasias de la Próstata/patología , Animales , Biomarcadores de Tumor , Proteína Morfogenética Ósea 4/genética , Proteína Morfogenética Ósea 4/metabolismo , Neoplasias Óseas/genética , Neoplasias Óseas/metabolismo , Medios de Cultivo Condicionados/farmacología , Endotelio Vascular/metabolismo , Humanos , Masculino , Ratones , Ratones SCID , Ratones Transgénicos , Estadificación de Neoplasias , Osteoblastos/metabolismo , Pronóstico , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/metabolismo , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
18.
Nucleic Acids Res ; 32(12): 3632-41, 2004.
Artículo en Inglés | MEDLINE | ID: mdl-15243141

RESUMEN

To understand the role of the CCAAT-binding factor, CBF, in transcription, we developed a strategy to purify the heterotrimeric CBF complex from HeLa cell extracts using two successive immunoaffinity chromatography steps. Here we show that the p32 protein, previously identified as the ASF/SF2 splicing factor-associated protein, copurified with the CBF complex. Studies of protein-protein interaction demonstrated that p32 interacts specifically with CBF-B subunit and also associates with CBF-DNA complex. Cellular localization by immunofluorescence staining revealed that p32 is present in the cell throughout the cytosol and nucleus, whereas CBF is present primarily in the nucleus. A portion of the p32 colocalizes with CBF-B in the nucleus. Interestingly, reconstitution of p32 in an in vitro transcription reaction demonstrated that p32 specifically inhibits CBF-mediated transcription activation. Altogether, our study identified p32 as a novel and specific corepressor of CBF-mediated transcription activation in vitro.


Asunto(s)
Factor de Unión a CCAAT/antagonistas & inhibidores , Receptores de Hialuranos , Glicoproteínas de Membrana , Receptores de Complemento/fisiología , Proteínas Represoras/fisiología , Activación Transcripcional , Factor de Unión a CCAAT/análisis , Factor de Unión a CCAAT/metabolismo , Proteínas Portadoras , Núcleo Celular/química , Cromatografía de Afinidad , Células HeLa , Humanos , Sustancias Macromoleculares , Proteínas Mitocondriales , Subunidades de Proteína/metabolismo , Receptores de Complemento/análisis , Receptores de Complemento/metabolismo , Proteínas Recombinantes/metabolismo , Proteínas Represoras/análisis , Proteínas Represoras/metabolismo
19.
Cancer Res ; 63(23): 8167-72, 2003 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-14678971

RESUMEN

To understand the physiological function of the mammalian heterotrimeric CCAAT binding factor CBF, also known as NF-Y, we have generated a conditional Cbf-b mouse mutant by introducing loxP sites in the murine Cbf-b/Nf-ya gene. Controlled expression of Cre recombinase deletes the gene in vivo, which leads to a loss of DNA binding by the CBF complex and hence CBF-mediated transcription. Deletion of both Cbf-b alleles causes early embryo lethality, indicating that CBF activity is essential for early mouse development. In primary cultures of mouse embryonic fibroblasts, conditional inactivation of CBF results in a block in cell proliferation and inhibition of S phase or DNA synthesis, which is followed by induction of apoptosis. We conclude that the CBF transcription factor complex is essential for cell proliferation and viability.


Asunto(s)
Factor de Unión a CCAAT/fisiología , Ratones/embriología , Alelos , Animales , Apoptosis/genética , Apoptosis/fisiología , Factor de Unión a CCAAT/genética , Factor de Unión a CCAAT/metabolismo , Ciclo Celular/genética , Ciclo Celular/fisiología , División Celular/genética , División Celular/fisiología , Eliminación de Gen , Ratones/genética , Ratones Endogámicos C57BL , Fase S/genética , Fase S/fisiología , Transcripción Genética
20.
Sci Rep ; 6: 34575, 2016 Sep 30.
Artículo en Inglés | MEDLINE | ID: mdl-27687130

RESUMEN

The mammalian central nervous system (CNS) contains various types of neurons with different neuronal functions. In contrast to established roles of cell type-specific transcription factors on neuronal specification and maintenance, whether ubiquitous transcription factors have conserved or differential neuronal function remains uncertain. Here, we revealed that inactivation of a ubiquitous factor NF-Y in different sets of neurons resulted in cell type-specific neuropathologies and gene downregulation in mouse CNS. In striatal and cerebellar neurons, NF-Y inactivation led to ubiquitin/p62 pathologies with downregulation of an endoplasmic reticulum (ER) chaperone Grp94, as we previously observed by NF-Y deletion in cortical neurons. In contrast, NF-Y inactivation in motor neurons induced neuronal loss without obvious protein deposition. Detailed analysis clarified downregulation of another ER chaperone Grp78 in addition to Grp94 in motor neurons, and knockdown of both ER chaperones in motor neurons recapitulated the pathology observed after NF-Y inactivation. Finally, additional downregulation of Grp78 in striatal neurons suppressed ubiquitin accumulation induced by NF-Y inactivation, implying that selective ER chaperone downregulation mediates different neuropathologies. Our data suggest distinct roles of NF-Y in protein homeostasis and neuronal maintenance in the CNS by differential regulation of ER chaperone expression.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA