Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 43
Filtrar
1.
Dev Cell ; 56(5): 602-612.e4, 2021 03 08.
Artigo em Inglês | MEDLINE | ID: mdl-33636105

RESUMO

Tissue-specific DNA methylation patterns are created by transcription factors that recruit methylation and demethylation enzymes to cis-regulatory elements. To date, it is not known whether transcription factors are needed to continuously maintain methylation profiles in development and mature tissues or whether they only establish these marks during organ development. We queried the role of the pioneer factor FoxA in generating hypomethylated DNA at liver enhancers. We discovered a set of FoxA-binding sites that undergo regional, FoxA-dependent demethylation during organ development. Conditional ablation of FoxA genes in the adult liver demonstrated that continued FoxA presence was not required to maintain the hypomethylated state, even when massive cell proliferation was induced. This study provides strong evidence for the stable, epigenetic nature of tissue-specific DNA methylation patterns directed by lineage-determining transcription factors during organ development.


Assuntos
Diferenciação Celular , Metilação de DNA , Elementos Facilitadores Genéticos , Epigênese Genética , Fator 3-alfa Nuclear de Hepatócito/fisiologia , Fator 3-beta Nuclear de Hepatócito/fisiologia , Fígado/metabolismo , Animais , Sítios de Ligação , Cromatina/química , Cromatina/genética , Cromatina/metabolismo , Desmetilação , Regulação da Expressão Gênica no Desenvolvimento , Fígado/embriologia , Masculino , Camundongos , Camundongos Knockout
2.
Mol Med Rep ; 22(6): 5155-5162, 2020 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-33174054

RESUMO

Polycystic ovary syndrome (PCOS) is one of the most common endocrine metabolic disorders characterized by hyperandrogenism, polycystic ovaries and ovulatory dysfunction. Several studies have suggested that the aberrant expression of microRNAs (miRNAs/miRs) plays an important role in the pathogenesis of PCOS; however, the role and underlying mechanisms of miR­132 in the development of PCOS remain unclear. In the present study, the expression of miR­132 in granulosa cells (GCs) derived from 26 patients with PCOS and 30 healthy controls was detected by reverse transcription­quantitative PCR (RT­qPCR). The apoptosis of GCs was examined using a TUNEL assay. The human ovarian granulosa­like tumor cell line, KGN, was cultured for Cell Counting Kit­8 assays following the overexpression or knockdown of miR­132. TargetScan was applied to identify the potential targets of miR­132, which was further verified by a luciferase assay, RT­qPCR and western blotting. The expression of miR­132 was decreased in GCs from patients with PCOS. Moreover, the GCs of patients with PCOS exhibited significantly increased apoptotic nuclei. Furthermore, the overexpression of miR­132 inhibited the viability of KGN cells. In addition, the results verified that miR­132 directly targeted forkhead box protein A1 (Foxa1), the knockdown of which suppressed KGN cell viability. On the whole, the findings of the present study demonstrated that miR­132 inhibited cell viability and induced apoptosis by directly interacting with Foxa1. Thus, miR­132 may be a potential target for the treatment of patients with PCOS.


Assuntos
Células da Granulosa/metabolismo , MicroRNAs/genética , Síndrome do Ovário Policístico/genética , Regiões 3' não Traduzidas/genética , Apoptose/genética , Linhagem Celular Tumoral , Proliferação de Células/genética , Sobrevivência Celular/genética , Feminino , Células da Granulosa/fisiologia , Fator 3-alfa Nuclear de Hepatócito/metabolismo , Fator 3-alfa Nuclear de Hepatócito/fisiologia , Humanos , MicroRNAs/metabolismo , Ativação Transcricional/genética
3.
Breast Cancer Res Treat ; 183(1): 41-48, 2020 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-32572714

RESUMO

BACKGROUND: Multi-gene expression assays have been developed with the aim of predicting late recurrence in patients with estrogen receptor (ER)-positive breast cancer. However, establishment of alternative markers based on immunohistochemistry is also important for achieving practical use. Based on our previous study, forkhead box A1 (FOXA1) protein was tested as a potentially useful predictive marker for late recurrence. METHODS: 117 patients with ER-positive HER2-negative invasive breast cancer who developed distant metastasis following curative surgery were retrospectively investigated. We also evaluated responsiveness to endocrine therapy according to FOXA1 expression. Furthermore, publicly available mRNA microarray data were analyzed to examine patterns of metastasis according to FOXA1 mRNA expression, employing the Kaplan-Meier plotter. RESULTS: High expression of FOXA1 was an independent factor predicting long disease-free survival (DFS), along with small tumor size (p = 0.010 and 0.016, respectively). Discrimination of DFS was improved by combining these two factors, i.e., patients with FOXA1-high small tumors had the longest DFS while those with FOXA1-low large tumors had the shortest DFS. Moreover, we revealed that risk of distant metastasis started to increase after the completion of adjuvant endocrine therapy in patients with FOXA1-high tumors. CONCLUSION: Among patients who developed distant metastasis, those with FOXA1-high tumors had significantly longer DFS. We believe our data to raise the possibility of FOXA1 being a useful predictive marker for late recurrence and to provide new insights into the biology of FOXA1-high breast cancers.


Assuntos
Neoplasias da Mama/metabolismo , Estrogênios , Fator 3-alfa Nuclear de Hepatócito/biossíntese , Proteínas de Neoplasias/biossíntese , Neoplasias Hormônio-Dependentes/metabolismo , Adulto , Idoso , Neoplasias da Mama/química , Neoplasias da Mama/genética , Intervalo Livre de Doença , Feminino , Regulação Neoplásica da Expressão Gênica , Fator 3-alfa Nuclear de Hepatócito/genética , Fator 3-alfa Nuclear de Hepatócito/fisiologia , Humanos , Estimativa de Kaplan-Meier , Pessoa de Meia-Idade , Invasividade Neoplásica , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/fisiologia , Neoplasias Hormônio-Dependentes/química , Neoplasias Hormônio-Dependentes/genética , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , RNA Neoplásico/biossíntese , RNA Neoplásico/genética , Receptor ErbB-2/análise , Receptores de Estrogênio/análise , Recidiva , Fatores de Tempo
4.
Inflamm Res ; 69(7): 645-656, 2020 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-32342116

RESUMO

OBJECTIVE AND DESIGN: Nowadays, sepsis-induced acute kidney injury (AKI) has gradually become a global problem for its high incidence and increasing mortality. Previous study has reported lncRNA ENST00000452391.1 in sepsis patients. However, its potential biological function and downstream molecular mechanism are still mysterious. METHODS AND RESULTS: Our study found that it was upregulated in sepsis-induced AKI patients, so it was identified as "sepsis-induced kidney injury associated transcript 1 (SIKIAT1)". We used lipopolysaccharide (LPS) stimulated HK-2 cells as an in vitro model to demonstrated that SIKIAT1 acts as a ceRNA for miR-96-3p to enhance FOXA1 expression and promote HK-2 cell apoptosis. CONCLUSION: Therefore, it could be a potential biomarker and therapeutic target for sepsis-induced AKI in the development of disease.


Assuntos
Apoptose/fisiologia , Fator 3-alfa Nuclear de Hepatócito/fisiologia , Nefropatias/etiologia , MicroRNAs/fisiologia , Sepse/complicações , Linhagem Celular , Regulação da Expressão Gênica , Técnicas de Silenciamento de Genes , Fator 3-alfa Nuclear de Hepatócito/genética , Humanos , Rim/efeitos dos fármacos , Rim/metabolismo , Rim/patologia , Nefropatias/patologia , Lipopolissacarídeos/farmacologia , MicroRNAs/genética , RNA Longo não Codificante/genética , RNA Longo não Codificante/fisiologia , Sepse/genética , Sepse/patologia , Transfecção
5.
Endocrinology ; 158(11): 4076-4092, 2017 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-28938408

RESUMO

Successful pregnancy relies on dynamic control of cell signaling to achieve uterine receptivity and the necessary biological changes required for endometrial decidualization, embryo implantation, and fetal development. Glucocorticoids are master regulators of intracellular signaling and can directly regulate embryo implantation and endometrial remodeling during murine pregnancy. In immortalized human uterine cells, we have shown that glucocorticoids and estradiol (E2) coregulate thousands of genes. Recently, glucocorticoids and E2 were shown to coregulate the expression of Left-right determination factor 1 (LEFTY1), previously implicated in the regulation of decidualization. To elucidate the molecular mechanism by which glucocorticoids and E2 regulate the expression of LEFTY1, immortalized and primary human endometrial cells were evaluated for gene expression and receptor recruitment to regulatory regions of the LEFTY1 gene. Glucocorticoid administration induced expression of LEFTY1 messenger RNA and protein and recruitment of the glucocorticoid receptor (GR) and activated polymerase 2 to the promoter of LEFTY1. Glucocorticoid-mediated recruitment of GR was dependent on pioneer factors FOXA1 and FOXA2. E2 was found to antagonize glucocorticoid-mediated induction of LEFTY1 by reducing recruitment of GR, FOXA1, FOXA2, and activated polymerase 2 to the LEFTY1 promoter. Gene expression analysis identified several genes whose glucocorticoid-dependent induction required FOXA1 and FOXA2 in endometrial cells. These results suggest a molecular mechanism by which E2 antagonizes GR-dependent induction of specific genes by preventing the recruitment of the pioneer factors FOXA1 and FOXA2 in a physiologically relevant model.


Assuntos
Endométrio/efeitos dos fármacos , Endométrio/metabolismo , Glucocorticoides/farmacologia , Fator 3-alfa Nuclear de Hepatócito/fisiologia , Fator 3-beta Nuclear de Hepatócito/fisiologia , Receptores de Glucocorticoides/fisiologia , Células Cultivadas , Dexametasona/farmacologia , Implantação do Embrião/efeitos dos fármacos , Implantação do Embrião/genética , Estradiol/farmacologia , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Fator 3-alfa Nuclear de Hepatócito/metabolismo , Fator 3-beta Nuclear de Hepatócito/metabolismo , Humanos , Fatores de Determinação Direita-Esquerda/genética , Regiões Promotoras Genéticas/efeitos dos fármacos , Receptores de Glucocorticoides/metabolismo , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética
6.
Oncogene ; 36(28): 4072-4080, 2017 07 13.
Artigo em Inglês | MEDLINE | ID: mdl-28319070

RESUMO

Neuroendocrine prostate cancer (NEPC) has increasingly become a clinical challenge. The mechanisms by which neuroendocrine (NE) cells arises from prostate adenocarcinoma cells are poorly understood. FOXA1 is a transcription factor of the forkhead family that is required for prostate epithelial differentiation. In this study, we demonstrated that FOXA1 loss drives NE differentiation, demarcated by phenotypical changes and NEPC marker expressions. Mechanistically, this is mediated by FOXA1 binding to the promoter of interleukin 8 (IL-8), a chemokine previously shown elevated in NEPC, to directly inhibit its expression. Further, IL-8 upregulation activates the MAPK/ERK pathway, leading to ERK phosphorylation and enolase 2 (ENO2) expression. IL-8 knockdown or ERK inhibition, on the other hand, abolished FOXA1 loss-induced NE differentiation. Analysis of xenograft mouse models confirmed FOXA1 loss in NEPC tumors relative to its adenocarcinoma counterparts. Importantly, FOXA1 is downregulated in human NEPC tumors compared to primary and castration-resistant prostate cancers, and its expression is negatively correlated with that of ENO2. These findings indicate that FOXA1 transcriptionally suppresses IL-8, the expression of which would otherwise stimulate the MAPK/ERK pathway to promote NE differentiation of prostate cancer cells. Our data strongly suggest that FOXA1 loss may play a significant role in enabling prostate cancer progression to NEPC, whereas IL-8 and MAPK/ERK pathways may be promising targets for therapeutic intervention.


Assuntos
Adenocarcinoma/genética , Diferenciação Celular/genética , Fator 3-alfa Nuclear de Hepatócito/fisiologia , Células Neuroendócrinas/fisiologia , Tumores Neuroendócrinos/genética , Neoplasias da Próstata/genética , Adenocarcinoma/patologia , Carcinoma Neuroendócrino/genética , Carcinoma Neuroendócrino/patologia , Linhagem Celular Tumoral , Progressão da Doença , Regulação para Baixo/genética , Humanos , Sistema de Sinalização das MAP Quinases/fisiologia , Masculino , Tumores Neuroendócrinos/patologia , Neoplasias da Próstata/patologia
7.
Cancer Res ; 76(19): 5881-5893, 2016 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-27496708

RESUMO

Glucuronidation is an enzymatic process that terminally inactivates steroid hormones, including estrogens and androgens, thereby influencing carcinogenesis in hormone-dependent cancers. While estrogens drive breast carcinogenesis via the estrogen receptor alpha (ERα), androgens play a critical role as prohormones for estrogen biosynthesis and ligands for the androgen receptor (AR). In this study, the expression and regulation of two androgen-inactivating enzymes, the UDP-glucuronosyltransferases UGT2B15 and UGT2B17, was assessed in breast cancer. In large clinical cohorts, high UGT2B15 and UGT2B17 levels positively influenced disease-specific survival in distinct molecular subgroups. Expression of these genes was highest in cases positive for ERα. In cell line models, ERα, AR, and the transcription factor FOXA1 cooperated to increase transcription via tandem binding events at their proximal promoters. ERα activity was dependent on FOXA1, facilitated by AR activation, and potently stimulated by estradiol as well as estrogenic metabolites of 5α-dihydrotestosterone. AR activity was mediated via binding to an estrogen receptor half-site 3' to the FOXA1 and ERα-binding sites. Although AR and FOXA1 bound the UGT promoters in AR-positive/ERα-negative breast cancer cell lines, androgen treatment did not influence basal transcription levels. Ex vivo culture of human breast tissue and ERα+ tumors provided evidence for upregulation of UGT2B15 and UGT2B17 by estrogen or androgen treatment. ERα binding was evident at the promoters of these genes in a small cohort of primary tumors and distant metastases. Collectively, these data provide insight into sex steroid receptor-mediated regulation of androgen-inactivating enzymes in ERα+ breast cancer, which may have subtype-specific consequences for disease progression and outcomes. Cancer Res; 76(19); 5881-93. ©2016 AACR.


Assuntos
Neoplasias da Mama/enzimologia , Receptor alfa de Estrogênio/análise , Glucuronosiltransferase/fisiologia , Antígenos de Histocompatibilidade Menor/fisiologia , Receptores Androgênicos/análise , Anilidas/farmacologia , Neoplasias da Mama/patologia , Linhagem Celular Tumoral , Feminino , Glucuronosiltransferase/genética , Fator 3-alfa Nuclear de Hepatócito/fisiologia , Humanos , Antígenos de Histocompatibilidade Menor/genética , Regiões Promotoras Genéticas , Receptor ErbB-2/análise
8.
PLoS One ; 11(3): e0151785, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26986977

RESUMO

BACKGROUND: Standardly collected clinical and pathological patient information has demonstrated only moderate ability to predict risk of biochemical recurrence (BCR) of prostate cancer in men undergoing salvage radiation therapy (SRT) for a rising PSA after radical prostatectomy (RP). Although elevated FOXA1 staining has been associated with poor patient outcomes following RP, it has not been studied in the specific setting of SRT after RP. The aim of this study was to evaluate the association between FOXA1 staining level and BCR after SRT for recurrent prostate cancer. METHODS: A total of 141 men who underwent SRT at our institution were included. FOXA1 staining levels in primary tumor samples were detected using immunohistochemistry. FOXA1 staining percentage and intensity were measured and multiplied together to obtain a FOXA1 H-score (range 0-12) which was our primary staining measure. P-values ≤ 0.0056 were considered as statistically significant after applying a Bonferroni correction for multiple comparisons. RESULTS: There was not a significant association between FOXA1 H-score and risk of BCR when considering H-score as an ordinal variable or as a categorical variable (all P ≥ 0.090). Similarly, no significant associations with BCR were observed for FOXA1 staining percentage or staining intensity (all P ≥ 0.14). CONCLUSIONS: FOXA1 staining level does not appear to have a major impact on risk of BCR after SRT.


Assuntos
Fator 3-alfa Nuclear de Hepatócito/fisiologia , Recidiva Local de Neoplasia/patologia , Próstata/patologia , Neoplasias da Próstata/radioterapia , Idoso , Corantes/uso terapêutico , Terapia Combinada , Humanos , Masculino , Recidiva Local de Neoplasia/radioterapia , Próstata/efeitos da radiação , Prostatectomia , Neoplasias da Próstata/mortalidade , Neoplasias da Próstata/patologia , Neoplasias da Próstata/cirurgia , Fatores de Risco , Terapia de Salvação/métodos , Análise de Sobrevida
9.
Oncogene ; 35(33): 4335-44, 2016 08 18.
Artigo em Inglês | MEDLINE | ID: mdl-26751772

RESUMO

Hormonal regulation of gene expression by androgen receptor (AR) is tightly controlled by many transcriptional cofactors, including pioneer factors FOXA1 and GATA2, which, however, exhibit distinct expression patterns and functional roles in prostate cancer. Here, we examined how FOXA1, GATA2 and AR crosstalk and regulate hormone-dependent gene expression in prostate cancer cells. Chromatin immunoprecipitation sequencing analysis revealed that FOXA1 reprograms both AR and GATA2 cistrome by preferably recruiting them to FKHD-containing genomic sites. By contrast, GATA2 is unable to shift AR or FOXA1 to GATA motifs. Rather, GATA2 co-occupancy enhances AR and FOXA1 binding to nearby ARE and FKHD sites, respectively. Similarly, AR increases, but not reprograms, GATA2 and FOXA1 cistromes. Concordantly, GATA2 and AR strongly enhance the transcriptional program of each other, whereas FOXA1 regulates GATA2- and AR-mediated gene expression in a context-dependent manner due to its reprogramming effects. Taken together, our data delineated for the first time the distinct mechanisms by which GATA2 and FOXA1 regulate AR cistrome and suggest that FOXA1 acts upstream of GATA2 and AR in determining hormone-dependent gene expression in prostate cancer.


Assuntos
Fator de Transcrição GATA2/fisiologia , Regulação Neoplásica da Expressão Gênica , Fator 3-alfa Nuclear de Hepatócito/fisiologia , Neoplasias da Próstata/genética , Receptores Androgênicos/fisiologia , Linhagem Celular Tumoral , Humanos , Masculino , Transcrição Gênica
10.
Nucleic Acids Res ; 44(9): 4105-22, 2016 05 19.
Artigo em Inglês | MEDLINE | ID: mdl-26743006

RESUMO

Identifying prostate cancer-driving transcription factors (TFs) in addition to the androgen receptor promises to improve our ability to effectively diagnose and treat this disease. We employed an integrative genomics analysis of master TFs CREB1 and FoxA1 in androgen-dependent prostate cancer (ADPC) and castration-resistant prostate cancer (CRPC) cell lines, primary prostate cancer tissues and circulating tumor cells (CTCs) to investigate their role in defining prostate cancer gene expression profiles. Combining genome-wide binding site and gene expression profiles we define CREB1 as a critical driver of pro-survival, cell cycle and metabolic transcription programs. We show that CREB1 and FoxA1 co-localize and mutually influence each other's binding to define disease-driving transcription profiles associated with advanced prostate cancer. Gene expression analysis in human prostate cancer samples found that CREB1/FoxA1 target gene panels predict prostate cancer recurrence. Finally, we showed that this signaling pathway is sensitive to compounds that inhibit the transcription co-regulatory factor MED1. These findings not only reveal a novel, global transcriptional co-regulatory function of CREB1 and FoxA1, but also suggest CREB1/FoxA1 signaling is a targetable driver of prostate cancer progression and serves as a biomarker of poor clinical outcomes.


Assuntos
Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/fisiologia , Fator 3-alfa Nuclear de Hepatócito/fisiologia , Recidiva Local de Neoplasia/metabolismo , Neoplasias da Próstata/metabolismo , Idoso , Idoso de 80 Anos ou mais , Sequência de Bases , Sítios de Ligação , Biomarcadores Tumorais , Linhagem Celular Tumoral , Sequência Consenso , Intervalo Livre de Doença , Regulação Neoplásica da Expressão Gênica , Ontologia Genética , Humanos , Estimativa de Kaplan-Meier , Masculino , Subunidade 1 do Complexo Mediador/metabolismo , Pessoa de Meia-Idade , Recidiva Local de Neoplasia/genética , Recidiva Local de Neoplasia/mortalidade , Prognóstico , Modelos de Riscos Proporcionais , Neoplasias da Próstata/genética , Neoplasias da Próstata/mortalidade , Neoplasias da Próstata/patologia , Inibidores de Proteínas Quinases/farmacologia , Transdução de Sinais , Transcrição Gênica
11.
Proc Natl Acad Sci U S A ; 112(35): E4929-38, 2015 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-26283356

RESUMO

Midbrain dopaminergic (mDA) neurons are implicated in cognitive functions, neuropsychiatric disorders, and pathological conditions; hence understanding genes regulating their homeostasis has medical relevance. Transcription factors FOXA1 and FOXA2 (FOXA1/2) are key determinants of mDA neuronal identity during development, but their roles in adult mDA neurons are unknown. We used a conditional knockout strategy to specifically ablate FOXA1/2 in mDA neurons of adult mice. We show that deletion of Foxa1/2 results in down-regulation of tyrosine hydroxylase, the rate-limiting enzyme of dopamine (DA) biosynthesis, specifically in dopaminergic neurons of the substantia nigra pars compacta (SNc). In addition, DA synthesis and striatal DA transmission were reduced after Foxa1/2 deletion. Furthermore, the burst-firing activity characteristic of SNc mDA neurons was drastically reduced in the absence of FOXA1/2. These molecular and functional alterations lead to a severe feeding deficit in adult Foxa1/2 mutant mice, independently of motor control, which could be rescued by L-DOPA treatment. FOXA1/2 therefore control the maintenance of molecular and physiological properties of SNc mDA neurons and impact on feeding behavior in adult mice.


Assuntos
Dopamina/metabolismo , Comportamento Alimentar , Fator 3-alfa Nuclear de Hepatócito/fisiologia , Fator 3-beta Nuclear de Hepatócito/fisiologia , Neurônios/metabolismo , Animais , Encéfalo/citologia , Encéfalo/metabolismo , Deleção de Genes , Fator 3-alfa Nuclear de Hepatócito/genética , Fator 3-beta Nuclear de Hepatócito/genética , Camundongos , Camundongos Knockout , Neurônios/citologia , RNA Mensageiro/genética
12.
Oncogene ; 34(34): 4509-18, 2015 Aug 20.
Artigo em Inglês | MEDLINE | ID: mdl-25435372

RESUMO

Liver receptor homolog-1 (LRH-1, NR5A2) is an orphan nuclear receptor that has an essential role in cancer progression, notably in breast cancer. Although its role in promoting cancer cell proliferation and migration is well documented, the molecular basis is not completely established. Here, we report that LRH-1 inhibition affects two- and three-dimensional cell proliferation of different types of breast cancer cells, including estrogen receptor α (ERα)-positive and triple-negative cells. This phenotype is accompanied by the upregulation of the cyclin-dependent kinase inhibitor CDKN1A (aka p21(CIP1/WAF1)) in a p53-independent manner. Chromatin immunoprecipitation analysis shows that LRH-1 cooperates with FOXA1 and binds directly to CDKN1A promoter and a distal regulatory region found at -62 kb from its transcriptional start sites, allowing repression of CDKN1A transcription. LRH-1 or FOXA1 depletion induces CDKN1A upregulation by removing histone deacetylase 2 from the promoter and distal regulatory elements and permitting histone acetylation in these regions. Analysis of breast cancer samples reveals that a high LRH-1 level is inversely correlated with CDKN1A expression in breast cancer patients and is associated with poor prognosis. This study reveals a novel mechanism of control of cell proliferation by LRH-1 regulating CDKN1A transcription in breast cancer cells, independent of ERα and p53 status. Targeting LRH-1 may provide an attractive prospect for treatment of tumors that are resistant to hormonal and targeted therapy.


Assuntos
Neoplasias da Mama/patologia , Proliferação de Células , Inibidor de Quinase Dependente de Ciclina p21/genética , Regulação Neoplásica da Expressão Gênica , Receptores Citoplasmáticos e Nucleares/fisiologia , Neoplasias da Mama/metabolismo , Neoplasias da Mama/mortalidade , Linhagem Celular Tumoral , Receptor alfa de Estrogênio/análise , Feminino , Fator 3-alfa Nuclear de Hepatócito/fisiologia , Histona Desacetilase 2/fisiologia , Humanos , Proteína Supressora de Tumor p53/fisiologia
13.
Nucleic Acids Res ; 42(18): 11339-48, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25223786

RESUMO

Chromatin constitutes a repressive barrier to the process of ligand-dependent transcriptional activity of nuclear receptors. Nucleosomes prevent the binding of estrogen receptor α (ERα) in absence of ligand and thus represent an important level of transcriptional regulation. Here, we show that in breast cancer MCF-7 cells, TLE3, a co-repressor of the Groucho/Grg/TLE family, interacts with FoxA1 and is detected at regulatory elements of ERα target genes in absence of estrogen. As a result, the chromatin is maintained in a basal state of acetylation, thus preventing ligand-independent activation of transcription. In absence of TLE3, the basal expression of ERα target genes induced by E2 is increased. At the TFF1 gene, the recruitment of TLE3 to the chromatin is FoxA1-dependent and prevents ERα and RNA polymerase II recruitment to TFF1 gene regulatory elements. Moreover, the interaction of TLE3 with HDAC2 results in the maintenance of acetylation at a basal level. We also provide evidence that TLE3 is recruited at several other regulatory elements of ERα target genes and is probably an important co-regulator of the E2 signaling pathway. In sum, our results describe a mechanism by which TLE3 affects ligand dependency in ERα-regulated gene expression via its binding restricting function and its role in gene regulation by histone acetylation.


Assuntos
Proteínas Correpressoras/metabolismo , Receptor alfa de Estrogênio/metabolismo , Regulação da Expressão Gênica , Linhagem Celular , Cromatina/metabolismo , Proteínas Correpressoras/fisiologia , Fator 3-alfa Nuclear de Hepatócito/fisiologia , Histona Desacetilases/metabolismo , Histonas/metabolismo , Humanos , Células MCF-7 , Elementos Reguladores de Transcrição , Transdução de Sinais , Transcrição Gênica
14.
Endocrinology ; 155(10): 3781-92, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25057789

RESUMO

The Forkhead box A transcription factors are major regulators of glucose homeostasis. They show both distinct and redundant roles during pancreas development and in adult mouse ß-cells. In vivo ablation studies have revealed critical implications of Foxa1 on glucagon biosynthesis and requirement of Foxa2 in α-cell terminal differentiation. In order to examine the respective role of these factors in mature α-cells, we used small interfering RNA (siRNA) directed against Foxa1 and Foxa2 in rat primary pancreatic α-cells and rodent α-cell lines leading to marked decreases in Foxa1 and Foxa2 mRNA levels and proteins. Both Foxa1 and Foxa2 control glucagon gene expression specifically through the G2 element. Although we found that Foxa2 controls the expression of the glucagon, MafB, Pou3f4, Pcsk2, Nkx2.2, Kir6.2, and Sur1 genes, Foxa1 only regulates glucagon gene expression. Interestingly, the Isl1 and Gipr genes were not controlled by either Foxa1 or Foxa2 alone but by their combination. Foxa1 and Foxa2 directly activate and bind the promoter region the Nkx2.2, Kir6.2 and Sur1, Gipr, Isl1, and Pou3f4 genes. We also demonstrated that glucagon secretion is affected by the combined effects of Foxa1 and Foxa2 but not by either one alone. Our results indicate that Foxa1 and Foxa2 control glucagon biosynthesis and secretion as well as α-cell differentiation with both common and unique target genes.


Assuntos
Diferenciação Celular/genética , Células Secretoras de Glucagon/fisiologia , Glucagon/biossíntese , Glucagon/metabolismo , Fator 3-alfa Nuclear de Hepatócito/fisiologia , Fator 3-beta Nuclear de Hepatócito/fisiologia , Animais , Sítios de Ligação/genética , Diferenciação Celular/efeitos dos fármacos , Células Cultivadas , Cricetinae , Fator 3-alfa Nuclear de Hepatócito/antagonistas & inibidores , Fator 3-beta Nuclear de Hepatócito/antagonistas & inibidores , Proteína Homeobox Nkx-2.2 , Masculino , Regiões Promotoras Genéticas , RNA Interferente Pequeno/farmacologia , Ratos
15.
BMC Genomics ; 15: 520, 2014 Jun 24.
Artigo em Inglês | MEDLINE | ID: mdl-24962896

RESUMO

BACKGROUND: The ZNF217 gene, encoding a C2H2 zinc finger protein, is located at 20q13 and found amplified and overexpressed in greater than 20% of breast tumors. Current studies indicate ZNF217 drives tumorigenesis, yet the regulatory mechanisms of ZNF217 are largely unknown. Because ZNF217 associates with chromatin modifying enzymes, we postulate that ZNF217 functions to regulate specific gene signaling networks. Here, we present a large-scale functional genomic analysis of ZNF217, which provides insights into the regulatory role of ZNF217 in MCF7 breast cancer cells. RESULTS: ChIP-seq analysis reveals that the majority of ZNF217 binding sites are located at distal regulatory regions associated with the chromatin marks H3K27ac and H3K4me1. Analysis of ChIP-seq transcription factor binding sites shows clustering of ZNF217 with FOXA1, GATA3 and ERalpha binding sites, supported by the enrichment of corresponding motifs for the ERalpha-associated cis-regulatory sequences. ERalpha expression highly correlates with ZNF217 in lysates from breast tumors (n = 15), and ERalpha co-precipitates ZNF217 and its binding partner CtBP2 from nuclear extracts. Transcriptome profiling following ZNF217 depletion identifies differentially expressed genes co-bound by ZNF217 and ERalpha; gene ontology suggests a role for ZNF217-ERalpha in expression programs associated with ER+ breast cancer studies found in the Molecular Signature Database. Data-mining of expression data from breast cancer patients correlates ZNF217 with reduced overall survival. CONCLUSIONS: Our genome-wide ZNF217 data suggests a functional role for ZNF217 at ERalpha target genes. Future studies will investigate whether ZNF217 expression contributes to aberrant ERalpha regulatory events in ER+ breast cancer and hormone resistance.


Assuntos
Neoplasias da Mama/metabolismo , Cromatina/metabolismo , Receptor alfa de Estrogênio/genética , Transativadores/fisiologia , Sítios de Ligação , Neoplasias da Mama/genética , Neoplasias da Mama/mortalidade , Análise por Conglomerados , Feminino , Fator de Transcrição GATA3/fisiologia , Regulação Neoplásica da Expressão Gênica , Genes Neoplásicos , Genoma Humano , Fator 3-alfa Nuclear de Hepatócito/fisiologia , Humanos , Estimativa de Kaplan-Meier , Células MCF-7 , Ligação Proteica , Transcriptoma
16.
Development ; 141(7): 1514-25, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24598166

RESUMO

The larval epidermis of Xenopus is a bilayered epithelium, which is an excellent model system for the study of the development and function of mucosal and mucociliary epithelia. Goblet cells develop in the outer layer while multiciliated cells and ionocytes sequentially intercalate from the inner to the outer layer. Here, we identify and characterise a fourth cell type, the small secretory cell (SSC). We show that the development of these cells is controlled by the transcription factor Foxa1 and that they intercalate into the outer layer of the epidermis relatively late, at the same time as embryonic hatching. Ultrastructural and molecular characterisation shows that these cells have an abundance of large apical secretory vesicles, which contain highly glycosylated material, positive for binding of the lectin, peanut agglutinin, and an antibody to the carbohydrate epitope, HNK-1. By specifically depleting SSCs, we show that these cells are crucial for protecting the embryo against bacterial infection. Mass spectrometry studies show that SSCs secrete a glycoprotein similar to Otogelin, which may form the structural component of a mucus-like protective layer, over the surface of the embryo, and several potential antimicrobial substances. Our study completes the characterisation of all the epidermal cell types in the early tadpole epidermis and reinforces the suitability of this system for the in vivo study of complex epithelia, including investigation of innate immune defences.


Assuntos
Epiderme/embriologia , Epiderme/imunologia , Células Caliciformes/imunologia , Imunidade Inata/fisiologia , Xenopus/embriologia , Xenopus/microbiologia , Animais , Diferenciação Celular/fisiologia , Cílios/imunologia , Embrião não Mamífero , Epiderme/metabolismo , Glicoproteínas/análise , Glicoproteínas/metabolismo , Fator 3-alfa Nuclear de Hepatócito/fisiologia , Íons/metabolismo , Larva , Muco/química , Muco/metabolismo , Via Secretória/imunologia , Vesículas Secretórias/imunologia , Vesículas Secretórias/metabolismo , Xenopus/imunologia
17.
BMC Cancer ; 14: 78, 2014 Feb 11.
Artigo em Inglês | MEDLINE | ID: mdl-24512546

RESUMO

BACKGROUND: Increasing evidence suggests that forkhead box A1 (FOXA1) is frequently dysregulated in many types of human cancers. However, the exact function and mechanism of FOXA1 in human endometrial cancer (EC) remains unclear. METHODS: FOXA1 expression, androgen receptor (AR) expression, and the relationships of these two markers with clinicopathological factors were determined by immunohistochemistry analysis. FOXA1 and AR were up-regulated by transient transfection with plasmids, and were down-regulated by transfection with siRNA or short hairpin RNA (shRNA). The effects of FOXA1 depletion and FOXA1 overexpression on AR-mediated transcription as well as Notch pathway and their impact on EC cell proliferation were examined by qRT-PCR, western blotting, co-immunoprecipitation, ChIP-PCR, MTT, colony-formation, and xenograft tumor-formation assays. RESULTS: We found that the expression of FOXA1 and AR in ECs was significantly higher than that in a typical hyperplasia and normal tissues. FOXA1 expression was significantly correlated with AR expression in clinical tissues. High FOXA1 levels positively correlated with pathological grade and depth of myometrial invasion in EC. High AR levels also positively correlated with pathological grade in EC. Moreover, the expression of XBP1, MYC, ZBTB16, and UHRF1, which are downstream targets of AR, was promoted by FOXA1 up-regulation or inhibited by FOXA1 down-regulation. Co-immunoprecipitation showed that FOXA1 interacted with AR in EC cells. ChIP-PCR assays showed that FOXA1 and AR could directly bind to the promoter and enhancer regions upstream of MYC. Mechanistic investigation revealed that over-expression of Notch1 and Hes1 proteins by FOXA1 could be reversed by AR depletion. In addition, we showed that down-regulation of AR attenuated FOXA1-up-regulated cell proliferation. However, AR didn't influence the promotion effect of FOXA1 on cell migration and invasion. In vivo xenograft model, FOXA1 knockdown reduced the rate of tumor growth. CONCLUSIONS: These results suggest that FOXA1 promotes cell proliferation by AR and activates Notch pathway. It indicated that FOXA1 and AR may serve as potential gene therapy in EC.


Assuntos
Proliferação de Células , Neoplasias do Endométrio/metabolismo , Fator 3-alfa Nuclear de Hepatócito/fisiologia , Receptores Androgênicos/metabolismo , Receptores Notch/metabolismo , Transdução de Sinais/fisiologia , Animais , Linhagem Celular Tumoral , Neoplasias do Endométrio/patologia , Feminino , Humanos , Camundongos , Camundongos Nus , Regulação para Cima/fisiologia , Ensaios Antitumorais Modelo de Xenoenxerto/métodos
18.
Asian Pac J Cancer Prev ; 15(1): 11-6, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24528009

RESUMO

Accurate diagnosis and proper monitoring of cancer patients remain important obstacles for successful cancer treatment. The search for cancer biomarkers can aid in more accurate prediction of clinical outcome and may also reveal novel predictive factors and therapeutic targets. One such prognostic marker seems to be FOXA1. Many studies have shown that FOXA1 is strongly expressed in a vast majority of cancers, including breast cancer, in which high expression is associated with a good prognosis. In this review, we summarize the role of this transcription factor in the development and prognosis of breast cancer in the hope of providing insights into utility of FOXA1 as a novel biomarker.


Assuntos
Biomarcadores Tumorais/fisiologia , Neoplasias da Mama/metabolismo , Fator 3-alfa Nuclear de Hepatócito/fisiologia , Biomarcadores Tumorais/análise , Neoplasias da Mama/química , Feminino , Fator 3-alfa Nuclear de Hepatócito/análise , Humanos , Prognóstico
19.
Oncol Rep ; 31(3): 1225-34, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24452315

RESUMO

Mechanisms governing the function of Forkhead-box A1 (FOXA1), a member of the FOX class of transcription factors, have been extensively studied. However, little is known about the activities and expression pattern of FOXA1 in endometrial cancer (EC). In the present study, we investigated the level of FOXA1 in multiple human EC cell lines and clinical samples by immunohistochemistry, qRT-PCR and Western blot analysis. FOXA1 overexpression was observed in estrogen receptor (ER)α-positive EC cell lines (P=0.0048). In endometrial tissues, FOXA1 was significantly upregulated in both normal endometrium and well-differentiated endometrial cancer tissues (P<0.001). Functional analyses of FOXA1 were evaluated by MTT, plate colony formation and Transwell assay. The results revealed that forced expression of FOXA1 inhibited EC cell proliferation, whereas FOXA1 depletion promoted cell viability and was associated with tumorigenesis. The nude mouse tumor xenograft assay also confirmed that ablation of FOXA1 expression promoted cell proliferation. Furthermore, we found that knockdown of FOXA1 decreased the expression of ERα, and FOXA1 interacted with this receptor in the EC cell lines. Collectively, these experiments suggest that FOXA1 is a tumor suppressor in EC and has a possible interaction with ERα.


Assuntos
Carcinoma Endometrioide/metabolismo , Neoplasias do Endométrio/metabolismo , Receptor alfa de Estrogênio/metabolismo , Fator 3-alfa Nuclear de Hepatócito/fisiologia , Animais , Carcinoma Endometrioide/patologia , Linhagem Celular Tumoral , Movimento Celular , Proliferação de Células , Neoplasias do Endométrio/patologia , Feminino , Humanos , Camundongos , Camundongos Nus , Pessoa de Meia-Idade , Invasividade Neoplásica , Transplante de Neoplasias , Receptor Cross-Talk , Transdução de Sinais , Carga Tumoral
20.
EMBO J ; 33(4): 312-26, 2014 Feb 18.
Artigo em Inglês | MEDLINE | ID: mdl-24451200

RESUMO

Androgen receptor (AR) binds male sex steroids and mediates physiological androgen actions in target tissues. ChIP-seq analyses of AR-binding events in murine prostate, kidney and epididymis show that in vivo AR cistromes and their respective androgen-dependent transcription programs are highly tissue specific mediating distinct biological pathways. This high order of tissue specificity is achieved by the use of exclusive collaborating factors in the three androgen-responsive tissues. We find two novel collaborating factors for AR signaling in vivo--Hnf4α (hepatocyte nuclear factor 4α) in mouse kidney and AP-2α (activating enhancer binding protein 2α) in mouse epididymis--that define tissue-specific AR recruitment. In mouse prostate, FoxA1 serves for the same purpose. FoxA1, Hnf4α and AP-2α motifs are over-represented within unique AR-binding loci, and the cistromes of these factors show substantial overlap with AR-binding events distinct to each tissue type. These licensing or pioneering factors are constitutively bound to chromatin and guide AR to specific genomic loci upon hormone exposure. Collectively, liganded receptor and its DNA-response elements are required but not sufficient for establishment of tissue-specific transcription programs.


Assuntos
Epididimo/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Fator 3-alfa Nuclear de Hepatócito/fisiologia , Fator 4 Nuclear de Hepatócito/fisiologia , Rim/metabolismo , Próstata/metabolismo , Receptores Androgênicos/metabolismo , Testosterona/farmacologia , Fator de Transcrição AP-2/fisiologia , Animais , Linhagem Celular , Cromatina/metabolismo , Imunoprecipitação da Cromatina , Epididimo/citologia , Perfilação da Expressão Gênica , Masculino , Camundongos , Camundongos Endogâmicos ICR , Orquiectomia , Especificidade de Órgãos , Ligação Proteica , Interferência de RNA , RNA Interferente Pequeno/farmacologia , Proteínas de Ligação a Tacrolimo/biossíntese , Proteínas de Ligação a Tacrolimo/genética , Testosterona/fisiologia , Transcrição Gênica
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...