Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Endocr Relat Cancer ; 24(7): 365-378, 2017 07.
Artículo en Inglés | MEDLINE | ID: mdl-28487351

RESUMEN

In utero exposure to bisphenol A (BPA) increases mammary cancer susceptibility in offspring. High-fat diet is widely believed to be a risk factor of breast cancer. The objective of this study was to determine whether maternal exposure to BPA in addition to high-butterfat (HBF) intake during pregnancy further influences carcinogen-induced mammary cancer risk in offspring, and its dose-response curve. In this study, we found that gestational HBF intake in addition to a low-dose BPA (25 µg/kg BW/day) exposure increased mammary tumor incidence in a 50-day-of-age chemical carcinogen administration model and altered mammary gland morphology in offspring in a non-monotonic manner, while shortening tumor-free survival time compared with the HBF-alone group. In utero HBF and BPA exposure elicited differential effects at the gene level in PND21 mammary glands through DNA methylation, compared with HBF intake in the absence of BPA. Top HBF + BPA-dysregulated genes (ALDH1B1, ASTL, CA7, CPLX4, KCNV2, MAGEE2 and TUBA3E) are associated with poor overall survival in The Cancer Genomic Atlas (TCGA) human breast cancer cohort (n = 1082). Furthermore, the prognostic power of the identified genes was further enhanced in the survival analysis of Caucasian patients with estrogen receptor-positive tumors. In conclusion, concurrent HBF dietary and a low-dose BPA exposure during pregnancy increases mammary tumor incidence in offspring, accompanied by alterations in mammary gland development and gene expression, and possibly through epigenetic reprogramming.


Asunto(s)
Compuestos de Bencidrilo/toxicidad , Neoplasias Mamarias Experimentales/etiología , Fenoles/toxicidad , Animales , Compuestos de Bencidrilo/administración & dosificación , Dieta Alta en Grasa , Femenino , Glándulas Mamarias Animales/efectos de los fármacos , Neoplasias Mamarias Experimentales/inducido químicamente , Neoplasias Mamarias Experimentales/patología , Fenoles/administración & dosificación , Distribución Aleatoria , Ratas , Riesgo
2.
Oncol Lett ; 14(6): 7443-7448, 2017 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-29344186

RESUMEN

Klotho ß (KLB) is a single-pass transmembrane protein measuring 1,043 amino acids in length that shares 41.2% homology with klotho α (KLA). KLB is a co-receptor and key regulator of the fibroblast growth factor receptor 4 (FGFR4) pathway. KLB interacts with FGFR4 to induce apoptosis and inhibit the proliferation of hepatoma cells, and KLA has been demonstrated to be a tumor suppressor in human breast cancer; however, little is known regarding the role of KLB in breast cancer. In the present study, through an immunohistochemical analysis of invasive ductal carcinoma tissue arrays, low KLB expression was identified in invasive ductal carcinoma samples compared with paired adjacent non-tumorous breast tissues (82 cases). In invasive ductal carcinoma tissues, KLB expression was negatively associated with pathological grade and lymph node metastasis. In 42 cases of paired microdissected breast specimens, the condition of the KLB gene allele was examined to determine the loss of heterozygosity (LOH), and selective LOH was identified at the KLB locus in 57.1% of primary tumors. These data suggest that KLB may be associated with the progression and metastasis of invasive ductal carcinoma, and therefore have clinical and therapeutic importance.

3.
Endocrinology ; 156(11): 3984-95, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26248216

RESUMEN

Bisphenol A (BPA) is a ubiquitous endocrine disruptor exerting lifelong effects on gene expression in rodent prostate cancer (PCa) models. Here, we aimed to determine whether epigenetic events mediating the action of BPA on human prostaspheres enriched in epithelial stem-like/progenitor cells is linked to PCa. We performed genome-wide transcriptome and methylome analyses to identify changes in prostaspheres treated with BPA (10 nM, 200 nM, and 1000 nM) or estradiol-17ß (E2) (0.1 nM) for 7 days and validated changes in expression, methylation, and histone marks in parallel-treated prostaspheres. BPA/E2-treatment altered expression of 91 genes but not the methylation status of 485,000 CpG sites in BPA/E2-treated prostaspheres. A panel of 26 genes was found repressed in all treatment groups. Fifteen of them were small nucleolar RNAs with C/D motif (SNORDs), which are noncoding, small nucleolar RNAs known to regulate ribosomal RNA assembly and function. Ten of the most down-regulated SNORDs were further studied. All 10 were confirmed repressed by BPA, but only 3 ratified as E2-repressed. SNORD suppression showed no correlation with methylation status changes in CpG sites in gene regulatory regions. Instead, BPA-induced gene silencing was found to associate with altered recruitments of H3K9me3, H3K4me3, and H3K27me3 to 5'-regulatory/exonic sequences of 5 SNORDs. Expression of 4 out of these 5 SNORDs (SNORD59A, SNORD82, SNORD116, and SNORD117) was shown to be reduced in PCa compared with adjacent normal tissue. This study reveals a novel and unique action of BPA in disrupting expression of PCa-associated SNORDs and a putative mechanism for reprogramming the prostasphere epigenome via histone modification.


Asunto(s)
Compuestos de Bencidrilo/farmacología , Código de Histonas/efectos de los fármacos , Fenoles/farmacología , Próstata/efectos de los fármacos , ARN no Traducido/genética , Transcriptoma/efectos de los fármacos , Análisis por Conglomerados , Islas de CpG/genética , Metilación de ADN/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Epigénesis Genética/efectos de los fármacos , Células Epiteliales/efectos de los fármacos , Células Epiteliales/metabolismo , Estradiol/farmacología , Estrógenos/farmacología , Estrógenos no Esteroides/farmacología , Histonas/metabolismo , Humanos , Lisina/metabolismo , Masculino , Metilación/efectos de los fármacos , Próstata/citología , Próstata/metabolismo , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/patología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Células Madre/efectos de los fármacos , Células Madre/metabolismo
4.
Am J Pathol ; 177(6): 2860-9, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21037081

RESUMEN

Genome-wide association studies highlight the importance of the fibroblast growth factor (FGF) receptor as a risk factor for breast cancer development. In particular, FGFR4 has been implicated in membrane ruffling, cancer cell invasiveness, and clinical chemoresistance in breast cancer. In this work, we studied FGFR4 in both human breast cancers and cell lines. We examined primary human microdissected breast samples for FGFR4 mutations, polymorphisms, loss of heterozygosity (LOH), and DNA methylation status. We identified no activating somatic mutations of FGFR4; however, we did identify a high proportion of the FGFR4-R388 heterozygous germline polymorphism. Analysis of paired microdissected samples uncovered selective LOH at the FGFR4 locus in 50% of primary tumors. This LOH involved the FGFR4-WT allele as frequently as the cancer progression-associated FGFR4-G388R polymorphic allele. Further, we identified DNA methylation in one-third of cases that targeted the FGFR4-WT allele more often and occurred more frequently either in concert with or exclusively in lymph node metastases. The role of DNA methylation in silencing the FGFR4-WT allele was supported by azacytidine treatment findings and was also confirmed in mouse xenograft studies, demonstrating selective FGFR4-WT allelic methylation with corresponding gene down-regulation. These findings support a growth advantage function for FGFR4-R388 and underscore the complex role of DNA methylation and LOH in determining the penetrance of allelic selection in breast cancer progression. These findings therefore have critical therapeutic importance.


Asunto(s)
Neoplasias de la Mama/genética , Carcinoma Ductal de Mama/genética , Metilación de ADN , Pérdida de Heterocigocidad , Receptor Tipo 4 de Factor de Crecimiento de Fibroblastos/genética , Selección Genética , Adulto , Anciano , Anciano de 80 o más Años , Alelos , Animales , Azacitidina/análogos & derivados , Azacitidina/farmacología , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Carcinoma Ductal de Mama/metabolismo , Carcinoma Ductal de Mama/patología , Línea Celular Tumoral , Metilación de ADN/efectos de los fármacos , Metilación de ADN/genética , Decitabina , Femenino , Mutación de Línea Germinal/fisiología , Humanos , Pérdida de Heterocigocidad/genética , Ratones , Ratones SCID , Persona de Mediana Edad , Polimorfismo de Nucleótido Simple/fisiología , Receptor Tipo 4 de Factor de Crecimiento de Fibroblastos/metabolismo , Selección Genética/genética , Selección Genética/fisiología , Ensayos Antitumor por Modelo de Xenoinjerto
5.
Am J Pathol ; 176(5): 2333-43, 2010 May.
Artículo en Inglés | MEDLINE | ID: mdl-20348248

RESUMEN

Recent genome-wide association studies have identified single nucleotide polymorphisms (SNPs) in the gene encoding fibroblast growth factor receptor 2 (FGFR2) as a risk factor for breast cancer. We examined the relationship between these intron 2 SNPs and gene expression in breast carcinomas. Primary breast tissue showed a common occurrence of these SNPs accompanied by FGFR2 expression in normal ductal epithelium. Unexpectedly, we found that FGFR2 mRNA and protein levels were reduced in microdissected cancer cells when compared with paired normal breast epithelium. FGFR2 down-regulation was associated with DNA methylation and loss-of-heterozygosity. Where FGFR2-IIIb was expressed in tumor cells, it was accompanied by up-regulation of the RNA-binding proteins ESRP1/2, consistent with splicing of this isoform. Reduction in FGFR2 was associated with re-expression of its putative target melanoma-associated antigen (MAGE-A) in primary carcinoma cells. Conversely, forced expression or activation of FGFR2-IIIb resulted in MAGE-A silencing. These data provide the first evidence for FGFR2 down-regulation in breast carcinomas harboring intron 2 SNPs. Our findings underscore the significance of epigenetic and somatic changes that can potentially modify the effects of germline polymorphisms in determining FGFR2 gene expression.


Asunto(s)
Neoplasias de la Mama/metabolismo , Carcinoma/metabolismo , Regulación Neoplásica de la Expresión Génica , Proteínas de Neoplasias/biosíntesis , Fragmentos de Péptidos/biosíntesis , Receptor Tipo 2 de Factor de Crecimiento de Fibroblastos/metabolismo , Línea Celular Tumoral , Metilación de ADN , Femenino , Perfilación de la Expresión Génica , Silenciador del Gen , Predisposición Genética a la Enfermedad , Humanos , Intrones , ARN Mensajero/metabolismo , Receptor Tipo 2 de Factor de Crecimiento de Fibroblastos/genética , Factores de Riesgo
6.
Mol Cell Endocrinol ; 326(1-2): 66-70, 2010 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-20060434

RESUMEN

Pituitary tumorigenesis infrequently involves intragenic mutations of oncogenes or tumor suppressor genes (Asa and Ezzat, 1998, 2002). Increasing evidence suggests that signals implicated in pituitary development may be relevant to neoplastic changes. Equally as important, accumulating evidence suggests that in contrast to other forms of solid neoplasia, pituitary tumors are frequently associated with epigenetic changes. In this review we discuss evolving models of epigenetic control and use members of the fibroblast growth factor receptor (FGF) receptor family as examples of dysregulated signals in pituitary tumorigenesis (Asa and Ezzat, 2009).


Asunto(s)
Ensamble y Desensamble de Cromatina/genética , Epigénesis Genética/genética , Histonas/metabolismo , Neoplasias Hipofisarias/genética , Animales , Metilación de ADN , Histonas/genética , Humanos , Ratones , Hipófisis/metabolismo , Hipófisis/patología , Neoplasias Hipofisarias/patología , Receptores de Factores de Crecimiento de Fibroblastos/metabolismo
7.
Mol Endocrinol ; 23(9): 1397-405, 2009 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19497954

RESUMEN

Recent genome-wide association studies have identified fibroblast growth factor receptor (FGFR)2 as one of a few candidate genes linked with breast cancer susceptibility. In particular, the disease-predisposing allele of FGFR2 is inherited as a 7.5-kb region within intron 2 that harbors eight single nucleotide polymorphisms. The relationship between these single nucleotide polymorphisms and FGFR2 gene expression remains unclear. Here we show the common occurrence of polymorphisms within the intron 2 region in a panel of 10 breast cancer cell lines. High FGFR2-expressing cell lines such as MCF-7 cells displayed polymorphic sequences with constitutive histone acetylation at multiple intron 2 sequences harboring putative transcription binding sites. Knockdown of Runx2 or CCAAT enhancer binding protein beta in these cells resulted in diminished endogenous FGFR2 gene expression. In contrast FGFR2-negative MDA-231 cells were wild type and showed evidence of histone 3/4 deacetylation at the rs2981578, rs10736303, and rs7895676 disease-associated alleles that harbor binding sites for Runx2, estrogen receptor, and CCAAT enhancer binding protein beta, respectively. Histone deacetylation inhibition with trichostatin A resulted in enhanced acetylation at these intron 2 sites, an effect associated with robust FGFR2 reexpression. Isoform analysis proved reexpression of the FGFR2-IIIc variant the splicing of which was positively influenced by trichostatin A-mediated recruitment of the Fas-activated serine/threonine phosphoprotein survival protein. Our findings highlight the potential role of histone acetylation in modulating access to selected polymorphic sites within intron 2 as well as downstream splicing sites in generating variable FGFR2 levels and isoforms in breast cancer.


Asunto(s)
Empalme Alternativo , Neoplasias de la Mama/metabolismo , Histonas/química , Intrones , Neoplasias Mamarias Animales/metabolismo , Polimorfismo Genético , Receptor Tipo 2 de Factor de Crecimiento de Fibroblastos/genética , Acetilación , Animales , Neoplasias de la Mama/genética , Línea Celular Tumoral , Femenino , Humanos , Ratones , Ratones SCID , Trasplante de Neoplasias , Isoformas de Proteínas
8.
J Clin Endocrinol Metab ; 93(9): 3610-7, 2008 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-18544619

RESUMEN

CONTEXT AND OBJECTIVE: Epigenetic dysregulation is implicated in pituitary neoplasia as the cause of silencing of several tumor suppressor genes. However, the upstream mediators of such events remain unknown. DESIGN: We examined the three members of the DNA methyltransferase (DNMT) enzyme family in normal and neoplastic human and mouse pituitary cells. SETTING: This study was performed at a university-affiliated cancer research institute. MAIN OUTCOME MEASURES: Gene expression, promoter DNA methylation, histone modifications, and cell proliferation were determined. RESULTS: In contrast to DNMT1 and DNMT3a, DNMT3b was expressed at relatively higher levels in neoplastic pituitary cells. However, examination of the human DNMT3b 5' region showed uniformly low DNA methylation levels with little difference between normal and tumor samples. Through pharmacological methylation inhibition or histone deacetylation inhibition, we identified that DNMT3b gene expression is subject to histone modifications. Down-regulation of DNMT3b resulted in induction of retinoblastoma, p21, and p27, and reduction in cell proliferation. These targeted effects were associated with enhanced histone 3 acetylation and diminished histone methylation. CONCLUSION: Our findings identify DNMT3b as a putative mediator of epigenetic control through histone modifications of gene expression in pituitary cells.


Asunto(s)
Cromatina/metabolismo , ADN (Citosina-5-)-Metiltransferasas/fisiología , Epigénesis Genética , Silenciador del Gen , Histonas/metabolismo , Hipófisis/metabolismo , Animales , Azacitidina/análogos & derivados , Azacitidina/farmacología , Células Cultivadas , ADN (Citosina-5-)-Metiltransferasas/antagonistas & inhibidores , ADN (Citosina-5-)-Metiltransferasas/genética , ADN (Citosina-5-)-Metiltransferasas/metabolismo , Decitabina , Epigénesis Genética/fisiología , Silenciador del Gen/fisiología , Inhibidores de Histona Desacetilasas , Histona Desacetilasas/metabolismo , Humanos , Ácidos Hidroxámicos/farmacología , Masculino , Ratones , Procesamiento Proteico-Postraduccional/fisiología , Regulación hacia Arriba/efectos de los fármacos , ADN Metiltransferasa 3B
9.
Clin Cancer Res ; 14(7): 1984-96, 2008 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-18381936

RESUMEN

PURPOSE: Four members of the fibroblast growth factor receptor (FGFR) family transduce signals of a diverse group of FGF ligands. The FGFR2-IIIb isoform is abundantly present in the normal pituitary gland with contrasting down-regulation in neoplastic pituitary cells. cDNA profiling identified the cancer-testis antigen melanoma-associated antigen A3 (MAGE-A3) as a putative target negatively regulated by FGFR2. EXPERIMENTAL DESIGN: Comparisons were made between normal and neoplastic human and mouse pituitary cells. Gene expression was examined by reverse transcription-PCR, DNA methylation was determined by methylation-specific PCR and combined bisulfite restriction analysis, and histone modification marks were identified by chromatin immunoprecipitation. RESULTS: Normal human pituitary tissue that expresses FGFR2-IIIb does not express MAGE-A3; in contrast, pituitary tumors that are FGFR2 negative show abundant MAGE-A3 mRNA expression. MAGE-A3 expression correlates with the presence and extent of DNA promoter methylation; more frequent and higher-degree methylation is present in the normal gland compared with pituitary tumors. Conversely, pituitary tumors are hypomethylated, particularly in females where MAGE-A3 expression is nearly thrice higher than in males. Estradiol treatment induces MAGE-A3 through enhanced histone 3 acetylation and diminished methylation. The effects of estradiol are directly opposed by FGF7/FGFR2-IIIb. Down-regulation of MAGE-A3 results in p53 transcriptional induction, also through reciprocal histone acetylation and methylation modifications. CONCLUSIONS: These findings highlight MAGE-A3 as a target of FGFR2-IIIb and estrogen action and provide evidence for a common histone-modifying network in the control of the balance between opposing signals.


Asunto(s)
Antígenos de Neoplasias/metabolismo , Estrógenos/metabolismo , Factor 2 de Crecimiento de Fibroblastos/metabolismo , Histonas/genética , Proteínas de Neoplasias/metabolismo , Neoplasias Hipofisarias/metabolismo , Animales , Antígenos de Neoplasias/genética , Western Blotting , Proliferación Celular , Metilación de ADN , Femenino , Expresión Génica , Regulación Neoplásica de la Expresión Génica , Humanos , Inmunohistoquímica , Inmunoprecipitación , Masculino , Ratones , Proteínas de Neoplasias/genética , Análisis de Secuencia por Matrices de Oligonucleótidos , Neoplasias Hipofisarias/genética , ARN Mensajero/análisis , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
11.
Blood ; 110(12): 4047-54, 2007 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-17875808

RESUMEN

The oncogene c-maf is frequently overexpressed in multiple myeloma cell lines and patient samples and contributes to increased cellular proliferation in part by inducing cyclin D2 expression. To identify regulators of c-maf, we developed a chemical screen in NIH3T3 cells stably overexpressing c-maf and the cyclin D2 promoter driving luciferase. From a screen of 2400 off-patent drugs and chemicals, we identified glucocorticoids as c-maf-dependent inhibitors of cyclin D2 transactivation. In multiple myeloma cell lines, glucocorticoids reduced levels of c-maf protein without influencing corresponding mRNA levels. Subsequent studies demonstrated that glucocorticoids increased ubiquitination-dependent degradation of c-maf and up-regulated ubiquitin C mRNA. Moreover, ectopic expression of ubiquitin C recapitulated the effects of glucocorticoids, demonstrating regulation of c-maf protein through the abundance of the ubiquitin substrate. Thus, using a chemical biology approach, we identified a novel mechanism of action of glucocorticoids and a novel mechanism by which levels of c-maf protein are regulated by the abundance of the ubiquitin substrate.


Asunto(s)
Glucocorticoides/farmacología , Complejo de la Endopetidasa Proteasomal/metabolismo , Proteínas Proto-Oncogénicas c-maf/metabolismo , Ubiquitina/metabolismo , Ubiquitinación/efectos de los fármacos , Regulación hacia Arriba/efectos de los fármacos , Animales , Ciclina D2 , Ciclinas/genética , Ciclinas/metabolismo , Evaluación Preclínica de Medicamentos , Glucocorticoides/química , Luciferasas/genética , Luciferasas/metabolismo , Ratones , Células 3T3 NIH , Regiones Promotoras Genéticas/genética , Activación Transcripcional/efectos de los fármacos
12.
Clin Cancer Res ; 13(16): 4713-20, 2007 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-17699848

RESUMEN

PURPOSE: Fibroblast growth factor (FGF) signals play fundamental roles in development and tumorigenesis. Thyroid cancer is an example of a tumor with nonoverlapping genetic mutations that up-regulate mitogen-activated protein kinase. We reported recently that FGF receptor 2 (FGFR2) is down-regulated through extensive DNA promoter methylation in thyroid cancer. Reexpression of the FGFR2-IIIb isoform impedes signaling upstream of the BRAF/mitogen-activated protein kinase pathway to interrupt tumor progression. In this analysis, we examined a novel target of FGFR2-IIIb signaling, melanoma-associated antigen-A3 and A6 (MAGE-A3/6). EXPERIMENTAL DESIGN: cDNA microarray analysis was done on human WRO thyroid cancer cells transfected with FGFR2-IIIb or empty vector. Identified gene target was confirmed by reverse transcription-PCR and Western blotting. Gene regulation was examined by treatment of WRO cells with the methylation inhibitor 5'-azacytidine followed by methylation-specific PCR and reverse transcription-PCR and by chromatin immunoprecipitation. RESULTS: Gene expression profiling identified the cancer/testis antigen MAGE-A3/6 as a novel target of FGFR2-IIIb signaling. MAGE-A3/6 regulation was mediated through DNA methylation and chromatin modifications. In particular, FGF7/FGFR2-IIIb activation resulted in histone 3 methylation and deacetylation associated with the MAGE-A3/6 promoter to down-regulate gene expression. CONCLUSIONS: These data unmask a complex repertoire of epigenetically controlled signals that govern FGFR2-IIIb and MAGE-A3/6 expression. Our findings provide insights into the interrelationship between novel tumor markers that may also represent overlapping therapeutic targets.


Asunto(s)
Antígenos de Neoplasias/genética , Histonas/metabolismo , Proteínas de Neoplasias/genética , Receptor Tipo 2 de Factor de Crecimiento de Fibroblastos/fisiología , Neoplasias de la Tiroides/metabolismo , Línea Celular Tumoral , Metilación de ADN , Epigénesis Genética , Factor 7 de Crecimiento de Fibroblastos/fisiología , Regulación Neoplásica de la Expresión Génica , Humanos , Análisis de Secuencia por Matrices de Oligonucleótidos , Regiones Promotoras Genéticas , Isoformas de Proteínas/fisiología , Transducción de Señal , Neoplasias de la Tiroides/genética
13.
Am J Pathol ; 170(5): 1618-28, 2007 May.
Artículo en Inglés | MEDLINE | ID: mdl-17456767

RESUMEN

Four members of the fibroblast growth factor receptor (FGFR) family of tyrosine kinases transduce signals of a diverse group of more than 23 fibroblast growth factor (FGF) ligands. Each prototypic receptor is composed of three immunoglobulin-like extracellular domains, two of which are involved in ligand binding. Alternative RNA splicing of one of two exons results in two different forms of the second half of the third immunoglobulin-like domain, the IIIb or IIIc isoforms. The contribution of each receptor and their isoforms in tumorigenesis remains unknown. In the pituitary, FGFR2 is expressed primarily as the IIIb isoform in normal adenohypophysial cells. In contrast, FGFR2 is significantly down-regulated in mouse corticotroph AtT20 tumor cells where the 5' promoter is methylated. Treatment of AtT20 cells with 5'-azacytidine resulted in FGFR2 re-expression, mainly as the FGFR2-IIIb isoform. Chromatin immunoprecipitation revealed evidence of histone methylation, but not of deacetylation, in the silencing of FGFR2 in AtT20 cells. Exposure of these cells to the cognate FGFR2-IIIb ligand FGF-7 resulted in diminished Rb phosphorylation and accumulation of p21 and p27, indicating diminished cell cycle progression. Examination of primary human pituitary adenomas revealed FGFR2 down-regulation in 52% (11 of 21) of samples and FGFR2 promoter DNA methylation in 45% (10 of 22) of samples. These data highlight the contribution from DNA and histone methylation as epigenetic mechanisms responsible for FGFR2 silencing in pituitary neoplasia.


Asunto(s)
Metilación de ADN , Silenciador del Gen , Neoplasias Hipofisarias/genética , Receptor Tipo 2 de Factor de Crecimiento de Fibroblastos/genética , Empalme Alternativo , Animales , Secuencia de Bases , Western Blotting , Femenino , Histonas/genética , Humanos , Inmunohistoquímica , Inmunoprecipitación , Masculino , Ratones , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Receptor Tipo 2 de Factor de Crecimiento de Fibroblastos/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
14.
Mol Endocrinol ; 21(5): 1205-15, 2007 May.
Artículo en Inglés | MEDLINE | ID: mdl-17341593

RESUMEN

The transcription factor Ikaros (Ik) is at the center of a functionally diverse chromatin-remodeling network that is critical for the development and regulation of both the immune and endocrine systems. Dominant negative forms of Ik result in neoplastic growth in mouse genetic studies and have been identified in human tumors. Ik modulates chromatin accessibility through associations with members of the NURD complex including histone deacetylase complexes. We show here that Ik expression in mouse pituitary corticotroph cells is itself regulated through histone modifications as well as DNA methylation. Examination of primary human pituitary specimens also identified a correlation of loss of Ik expression with the presence of DNA methylation in the untranslated exon 1 CpG island. These findings have important implications for the understanding of Ikaros' role in epigenetic functions and suggest a potential role for demethylating agents in the treatment of related disorders.


Asunto(s)
ADN/genética , Regulación de la Expresión Génica , Factor de Transcripción Ikaros/genética , Hipófisis/fisiología , Animales , Secuencia de Bases , Metilación de ADN , Exones , Femenino , Inhibidores de Histona Desacetilasas , Histona Desacetilasas/metabolismo , Humanos , Masculino , Ratones , Datos de Secuencia Molecular , ARN Mensajero/genética , ARN Mensajero/aislamiento & purificación
15.
Mol Endocrinol ; 20(11): 2976-86, 2006 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-16873443

RESUMEN

We reported the expression of the lymphoid zinc finger transcription factor Ikaros (Ik) in the endocrine pituitary gland, where the usual isoforms, Ik1 and Ik2, are thought to play multiple physiological roles. The gene is alternatively spliced to yield a dominant negative isoform, Ik6, in nearly half of human pituitary tumors. We show here that the tumor-specific truncated Ik6 isoform promotes pituitary tumor AtT20 corticotroph and GH4 mammosomatotroph cell growth, evidenced by increased S-phase entry, colony formation in soft agar, and growth of xenografts in vivo. Ik6-mediated cell growth was associated with enhanced protection against apoptosis and up-regulation of the antiapoptotic factor Bcl-XL but not the related Bcl-2 family member. The effect of Ik6 on Bcl-XL induction was not reproduced by small interfering RNA-mediated Ik-down-regulation, indicating that this effect is not mediated entirely by disruption of Ik1 action. In cotransfection studies, Ik1 attenuated and Ik6 enhanced Bcl-XL promoter activity. The effect of Ik6 was mimicked by histone deacetylation inhibition but not by methylation inhibition. Furthermore, chromatin immunoprecipitation confirmed the ability of Ik6 to selectively acetylate histone 3 sites but not influence methylation of the Bcl-XL promoter. Thus, the contribution of Ik6 to tumor pathogenesis involves up-regulation of an antiapoptotic signal generated through selective acetylation of the Bcl-XL promoter.


Asunto(s)
Factor de Transcripción Ikaros/metabolismo , Factor de Transcripción Ikaros/fisiología , Hipófisis/citología , Hipófisis/metabolismo , Regiones Promotoras Genéticas , Proteína bcl-X/genética , Acetilación , Animales , Apoptosis/fisiología , Secuencia de Bases , Proliferación Celular , Supervivencia Celular , Células Cultivadas , Femenino , Histonas/metabolismo , Ratones , Ratones SCID , Datos de Secuencia Molecular , Proteínas de Neoplasias/metabolismo , Proteínas de Neoplasias/fisiología , Isoformas de Proteínas/metabolismo , Isoformas de Proteínas/fisiología , Ratas , Somatotrofos/metabolismo , Activación Transcripcional , Proteína bcl-X/metabolismo
16.
Am J Pathol ; 164(6): 2003-12, 2004 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-15161636

RESUMEN

Estrogen receptor (ER)-beta is thought to exert anti-proliferative effects in the normal prostate but supports prostate cancer (PCa) cell survival. We previously reported that the receptor's expression declined as PCa developed in the gland but reappeared in lymph node and bone metastases. To investigate whether hypermethylation was the underlying mechanism for these phenomena, we first identified two CpG islands (CGIs) encompassing 41 CpG dinucleotides, located separately in the untranslated exon 0N and the promoter region of ER-beta. Using immunostained, laser capture-microdissected samples from 56 clinical specimens, we demonstrated an inverse relationship exists between the extent of ER-beta CGI methylation and receptor expression in normal, hyperplastic, premalignant, and malignant foci of the prostate and in lymph node and bone metastases. Treatment of PCa cell lines (LNCaP and DU145), that express little ER-beta mRNA, with a demethylating agent increased levels of receptor expression thus corroborating our in vivo findings that methylation is involved in ER-beta silencing. Methylation centers in the promoter region and exon 0N were identified by hierarchical cluster analysis of bisulfite sequencing data obtained from 710 alleles. Methylation at these centers was insignificant in normal epithelium, reached 80 to 90% in grade 4/5 PCa, but declined to less than 20% in bone metastases. In addition, progressive methylation spreading from the exonic CGI to the promoter CGI, which correlated with loss of ER-beta expression, was detected in microdissected samples and in cell cultures. Using a new class of methylated oligonucleotides that mediate sequence-specific methylation in cellulo, we demonstrated that methylation of the promoter CGI, but not the exonic CGIs, led to transcriptional inactivation of ER-beta. Our results present the first evidence that epigenetic regulation of ER-beta is a reversible and tumor stage-specific process and that gene silencing via methylated oligonucleotides may have therapeutic potential in the treatment of advanced PCa.


Asunto(s)
Metilación de ADN , Neoplasias de la Próstata/patología , Receptores de Estrógenos/genética , Secuencia de Bases , Línea Celular Tumoral , Cartilla de ADN , Fosfatos de Dinucleósidos/análisis , Receptor beta de Estrógeno , Humanos , Masculino , Datos de Secuencia Molecular , Metástasis de la Neoplasia/genética , Metástasis de la Neoplasia/patología , Estadificación de Neoplasias , Lesiones Precancerosas/genética , Lesiones Precancerosas/patología , Regiones Promotoras Genéticas , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/cirugía , TATA Box
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...