Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
1.
J Nat Med ; 75(3): 590-601, 2021 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-33713277

RESUMO

Colorectal cancer stem cells (CSCs) have the potential for self-renewal, proliferation, and differentiation. And LGR5 is a stem cell marker gene of colorectal cancer. Curcumin can suppress oncogenicity of many cancer cells, yet the effect and mechanism of curcumin in LGR5(+) colorectal cancer stem cells (CSCs) have not been studied. In this study, we studied the effect of curcumin on LGR5(+) colorectal CSCs using the experiments of tumorsphere formation, cell viability and cell apoptosis. Then autophagy analysis, RNA-Seq, and real-time PCR were used to identify the mechanism responsible for the inhibition of LGR5(+) colorectal CSCs. Our results showed that curcumin inhibited tumorsphere formation, decreased cell viability in a dose-dependent manner, and also promoted apoptosis of LGR5(+) colorectal CSCs. Next, we found curcumin induced autophagy of LGR5(+) colorectal CSCs. When LGR5(+) colorectal CSCs were co-treated with curcumin and the autophagy inhibitor (hydroxychloroquine), curcumin-induced cell proliferation inhibition decreased. In addition, we also found that curcumin inhibited the extracellular matrix (ECM)-receptor interaction pathway via the downregulation of the following genes: GP1BB, COL9A3, COMP, AGRN, ITGB4, LAMA5, COL2A1, ITGB6, ITGA1, and TNC. Further, these genes were transcriptionally regulated by TFAP2A, and the high expression of TFAP2A was associated with poor prognosis in colorectal cancer. In conclusion, curcumin suppressed LGR5(+) colorectal CSCs, potentially by inducing autophagy and repressing the oncogenic TFAP2A-mediated ECM pathway.


Assuntos
Autofagia , Neoplasias Colorretais , Curcumina/farmacologia , Células-Tronco Neoplásicas/efeitos dos fármacos , Fator de Transcrição AP-2/antagonistas & inibidores , Apoptose , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular , Matriz Extracelular , Humanos , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , Esferoides Celulares , Fator de Transcrição AP-2/metabolismo , Fator de Transcrição AP-2/farmacologia
2.
Mol Cell Biochem ; 476(2): 1051-1061, 2021 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-33170430

RESUMO

Long non-coding RNAs (lncRNAs) are known as crucial regulators in the development of OC. In the current study, we aim to explore the function and molecular mechanism of lncRNA DLEU1 in OC. Quantitative real-time polymerase chain reaction (qRT-PCR) was applied to determine the expression of DLEU1, miR-429, and TFAP2A in OC cells and tissues. The relationship among DLEU1, miR-429, and TFAP2A was tested by dual-luciferase reporter (DLR) assay. Besides, the proliferative, migratory and invasive abilities of OC cells were analyzed by MTT, wound healing, and transwell assays, respectively. Western blot was performed to determine the protein expression of TFAP2A. The expression of lncRNA DLEU1 and TFAP2A were upregulated, and miR-429 was downregulated in OC tissues. Silencing of DLEU1 inhibited the proliferation, migration, and invasion of OC cells. Bioinformation and DLR assay showed that DLEU1 acted as the sponge for miR-429. Moreover, miR-429 could directly target TFAP2A and inhibit the proliferation, migration, and invasion of OC cells. Moreover, we observed a negative correlation between miR-429 and DLEU1, and between miR-429 and TFAP2A in OC tissues. The transfection of miR-429 inhibitor or pcDNA-TFAP2A reversed the inhibitory effects of si-DLEU1 on the proliferation, migration, and invasion of OC cells. Silencing of DLEU1 inhibited the proliferation, migration, and invasion of OC cells by regulating miR-429/TFAP2A axis, indicating a potential therapeutic target for OC.


Assuntos
Carcinoma Epitelial do Ovário/prevenção & controle , Regulação Neoplásica da Expressão Gênica , Inativação Gênica , MicroRNAs/genética , Neoplasias Ovarianas/prevenção & controle , RNA Longo não Codificante/antagonistas & inibidores , Fator de Transcrição AP-2/antagonistas & inibidores , Adulto , Carcinoma Epitelial do Ovário/genética , Carcinoma Epitelial do Ovário/metabolismo , Carcinoma Epitelial do Ovário/patologia , Linhagem Celular Tumoral , Movimento Celular , Proliferação de Células , Biologia Computacional/métodos , Feminino , Humanos , Pessoa de Meia-Idade , Neoplasias Ovarianas/genética , Neoplasias Ovarianas/metabolismo , Neoplasias Ovarianas/patologia , RNA Longo não Codificante/genética , Transdução de Sinais , Fator de Transcrição AP-2/metabolismo , Regulação para Cima
3.
Eur Rev Med Pharmacol Sci ; 24(9): 4745-4755, 2020 05.
Artigo em Inglês | MEDLINE | ID: mdl-32432738

RESUMO

OBJECTIVE: Ferroptosis is a recently identified form of controlled cell death generally associated with the accumulation of lipid-associated reactive oxygen species (ROS). However, the molecular mechanisms underlying ferroptosis have not been established. MATERIALS AND METHODS: Microarray expression data for three human gallbladder carcinoma (GBC) and matched non-tumour specimens were downloaded from the Gene Expression Omnibus (GEO) repository. Candidate genes were filtered using bioinformatic analysis. After cell transfection, candidate gene impacts on cell proliferation, migration, invasion and ferroptosis (ferrous iron (Fe2+) and malondialdehyde (MDA) levels) were assessed. RESULTS: We screened 626 differentially expressed genes (DEGs) including 465 that were downregulated and 161 that were upregulated in the three tissue pairs. These DEGs were used to construct a protein-protein interaction (PPI) network. Functional enrichment analysis revealed the top three modules in the network and four hub genes. Transcription factor AP-2 alpha (TFAP2A) was screened and showed overexpression in The Cancer Genome Atlas (TCGA) digestive system tumour data and a relationship with clinical survival. In vitro, GBC exhibited upregulated expression of TFAP2A, whose inhibition reduced GBC cell proliferation, migration, and invasion. Fe2+ and MDA levels were elevated. Moreover, Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis revealed TFAP2A enrichment in oxidative stress. Subsequent experiments demonstrated that TFAP2A silencing attenuated the expression of key genes associated with oxidative stress such as heme oxygenase 1 (HO-1), nuclear factor erythroid 2 like 2 (Nrf2), ferritin heavy chain 1 (FTH1) and NAD(P)H quinone dehydrogenase 1 (NQO1). CONCLUSIONS: Bioinformatic and experimental analyses reveal that TFAP2A plays a vital role in ferroptosis and hence is a potential therapeutic target for GBC treatment.


Assuntos
Ferroptose/fisiologia , Neoplasias da Vesícula Biliar/metabolismo , Fator 2 Relacionado a NF-E2/biossíntese , Fator de Transcrição AP-2/biossíntese , Linhagem Celular Transformada , Linhagem Celular Tumoral , Neoplasias da Vesícula Biliar/patologia , Redes Reguladoras de Genes/fisiologia , Humanos , Transdução de Sinais/fisiologia , Fator de Transcrição AP-2/antagonistas & inibidores
4.
Cell Death Dis ; 10(6): 397, 2019 05 21.
Artigo em Inglês | MEDLINE | ID: mdl-31113934

RESUMO

Thyroid cancer is commonly seen in the clinic with a rapidly increasing incidence globally. COX-2 overexpression correlates with the pathologic type of thyroid carcinoma, and it has been suggested that COX-2 overexpression is associated with a poor prognosis. However, little is known about its upstream regulatory mechanism. Bioinformatics suggested that transcription factor AP-2 beta (TFAP2B) might specifically bind to the COX-2 promoter, which was confirmed by biotin-labeled COX-2 promoter pulldown and luciferase reporter assays. We performed western blot and immunohistochemical staining to detect the expression of TFAP2B/COX-2 in thyroid cancer tissues (T) and the matched adjacent noncarcinoma tissues (ANT), and investigated the relationship between TFAP2B/COX-2 expression and clinical pathological factors in thyroid cancer patients. Afterward, MTS, colony formation, cell-apoptosis assay, transwell-invasion and scratch assays were performed to examine the proliferation, apoptosis, invasion, and migration of thyroid cancer cells with TFAP2B knocked down or overexpressed. The mouse xenograft experiment was performed to study in vivo the proliferation of thyroid cancer cells with TFAP2B knocked down or overexpressed. We found that TFAP2B bound to the promoter of COX-2 to activate its expression. Western blot and immunohistochemistry showed that TFAP2B/COX-2 was highly expressed in thyroid cancer, and high TFAP2B and COX-2 expression was associated with aggressive clinicopathological features in thyroid cancer. TFAP2B mediated thyroid cancer cell proliferation, apoptosis, invasion, and migration via the COX-2 signaling pathway in vitro and in vivo. TFAP2B bound to the promoter of COX-2 to activate its expression, indicating that TFAP2B is a critical regulatory molecule in the COX-2 signaling pathway that promoted tumor progression in thyroid cancer.


Assuntos
Ciclo-Oxigenase 2/metabolismo , Neoplasias da Glândula Tireoide/patologia , Fator de Transcrição AP-2/metabolismo , Animais , Linhagem Celular Tumoral , Movimento Celular , Proliferação de Células , Ciclo-Oxigenase 2/genética , Inibidores de Ciclo-Oxigenase 2/uso terapêutico , Proteínas do Olho/metabolismo , Feminino , Humanos , Masculino , Camundongos , Camundongos Nus , Pessoa de Meia-Idade , Fatores de Crescimento Neural/metabolismo , Regiões Promotoras Genéticas , Interferência de RNA , RNA Interferente Pequeno/metabolismo , RNA Interferente Pequeno/uso terapêutico , Serpinas/metabolismo , Transdução de Sinais , Neoplasias da Glândula Tireoide/tratamento farmacológico , Neoplasias da Glândula Tireoide/metabolismo , Fator de Transcrição AP-2/antagonistas & inibidores , Fator de Transcrição AP-2/genética , Fator A de Crescimento do Endotélio Vascular/metabolismo
5.
Biochem J ; 475(11): 1965-1977, 2018 06 11.
Artigo em Inglês | MEDLINE | ID: mdl-29760237

RESUMO

AP-2 gamma (AP-2γ) is a transcription factor that plays pivotal roles in breast cancer biology. To search for small molecule inhibitors of AP-2γ, we performed a high-throughput fluorescence anisotropy screen and identified a polyoxometalate compound with Wells-Dawson structure K6[P2Mo18O62] (Dawson-POM) that blocks the DNA-binding activity of AP-2γ. We showed that this blocking activity is due to the direct binding of Dawson-POM to AP-2γ. We also provided evidence to show that Dawson-POM decreases AP-2γ-dependent transcription similar to silencing the gene. Finally, we demonstrated that Dawson-POM contains anti-proliferative and pro-apoptotic effects in breast cancer cells. In summary, we identified the first small molecule inhibitor of AP-2γ and showed Dawson-POM-mediated inhibition of AP-2γ as a potential avenue for cancer therapy.


Assuntos
Apoptose/efeitos dos fármacos , Neoplasias da Mama/metabolismo , Bibliotecas de Moléculas Pequenas/farmacologia , Fator de Transcrição AP-2/antagonistas & inibidores , Compostos de Tungstênio/farmacologia , Neoplasias da Mama/genética , Neoplasias da Mama/fisiopatologia , Proliferação de Células/efeitos dos fármacos , Feminino , Humanos , Cinética , Bibliotecas de Moléculas Pequenas/química , Bibliotecas de Moléculas Pequenas/metabolismo , Fator de Transcrição AP-2/genética , Fator de Transcrição AP-2/metabolismo , Compostos de Tungstênio/química , Compostos de Tungstênio/metabolismo
6.
J Biol Chem ; 293(22): 8342-8361, 2018 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-29674317

RESUMO

Epithelia contribute to physical barriers that protect internal tissues from the external environment and also support organ structure. Accordingly, establishment and maintenance of epithelial architecture are essential for both embryonic development and adult physiology. Here, using gene knockout and knockdown techniques along with gene profiling, we show that extracellular signal-regulated kinase 3 (ERK3), a poorly characterized atypical mitogen-activated protein kinase (MAPK), regulates the epithelial architecture in vertebrates. We found that in Xenopus embryonic epidermal epithelia, ERK3 knockdown impairs adherens and tight-junction protein distribution, as well as tight-junction barrier function, resulting in epidermal breakdown. Moreover, in human epithelial breast cancer cells, inhibition of ERK3 expression induced thickened epithelia with aberrant adherens and tight junctions. Results from microarray analyses suggested that transcription factor AP-2α (TFAP2A), a transcriptional regulator important for epithelial gene expression, is involved in ERK3-dependent changes in gene expression. Of note, TFAP2A knockdown phenocopied ERK3 knockdown in both Xenopus embryos and human cells, and ERK3 was required for full activation of TFAP2A-dependent transcription. Our findings reveal that ERK3 regulates epithelial architecture, possibly together with TFAP2A.


Assuntos
Neoplasias da Mama/patologia , Embrião não Mamífero/enzimologia , Células Epiteliais/química , Proteína Quinase 6 Ativada por Mitógeno/metabolismo , Fator de Transcrição AP-2/metabolismo , Xenopus laevis/fisiologia , Animais , Neoplasias da Mama/enzimologia , Sistemas CRISPR-Cas , Adesão Celular , Membrana Celular , Células Cultivadas , Embrião não Mamífero/citologia , Células Epiteliais/enzimologia , Células Epiteliais/patologia , Feminino , Células Hep G2 , Humanos , Proteína Quinase 6 Ativada por Mitógeno/antagonistas & inibidores , Proteína Quinase 6 Ativada por Mitógeno/genética , Junções Íntimas , Fator de Transcrição AP-2/antagonistas & inibidores , Fator de Transcrição AP-2/genética , Xenopus laevis/embriologia
7.
Lab Invest ; 98(1): 117-129, 2018 01.
Artigo em Inglês | MEDLINE | ID: mdl-29035379

RESUMO

Transcription factor AP-2ß (TFAP2B) regulates embryonic organ development and is overexpressed in alveolar rhabdomyosarcoma, a rare childhood malignancy. Gene expression profiling has implicated AP-2ß in breast cancer (BC). This study characterizes AP-2ß expression in the mammary gland and in BC. AP-2ß protein expression was assessed in the normal mammary gland epithelium, in various reactive, metaplastic and pre-invasive neoplastic lesions and in two clinical BC cohorts comprising >2000 patients. BCs from various genetically engineered mouse (GEM) models were also evaluated. Human BC cell lines served as functional models to study siRNA-mediated inhibition of AP-2ß. The normal mammary gland epithelium showed scattered AP-2ß-positive cells in the luminal cell layer. Various reactive and pre-invasive neoplastic lesions, including apocrine metaplasia, usual ductal hyperplasia and lobular carcinoma in situ (LCIS) showed enhanced AP-2ß expression. Cases of ductal carcinoma in situ (DCIS) were more often AP-2ß-negative (P<0.001). In invasive BC cohorts, AP-2ß-positivity was associated with the lobular BC subtype (P<0.001), loss of E-cadherin (P<0.001), a positive estrogen receptor (ER) status (P<0.001), low Ki67 (P<0.001), low/intermediate Oncotype DX recurrence scores (P<0.001), and prolonged event-free survival (P=0.003). BCs from GEM models were all AP-2ß-negative. In human BC cell lines, AP-2ß expression was independent from ER-signaling. SiRNA-mediated inhibition of AP-2ß diminished proliferation of lobular BC cell lines in vitro. In summary, AP-2ß is a new mammary epithelial differentiation marker. Its expression is preferentially retained and enhanced in LCIS and invasive lobular BC and has prognostic implications. Our findings indicate that AP-2ß controls tumor cell proliferation in this slow-growing BC subtype.


Assuntos
Carcinoma de Mama in situ/metabolismo , Neoplasias da Mama/metabolismo , Carcinoma Lobular/metabolismo , Regulação Neoplásica da Expressão Gênica , Glândulas Mamárias Humanas/metabolismo , Proteínas de Neoplasias/metabolismo , Fator de Transcrição AP-2/metabolismo , Animais , Biomarcadores Tumorais/metabolismo , Carcinoma de Mama in situ/patologia , Carcinoma de Mama in situ/cirurgia , Neoplasias da Mama/sangue , Neoplasias da Mama/patologia , Neoplasias da Mama/cirurgia , Carcinoma Lobular/patologia , Carcinoma Lobular/cirurgia , Linhagem Celular Tumoral , Proliferação de Células , Estudos de Coortes , Intervalo Livre de Doença , Feminino , Seguimentos , Humanos , Glândulas Mamárias Humanas/patologia , Glândulas Mamárias Humanas/cirurgia , Camundongos Transgênicos , Gradação de Tumores , Invasividade Neoplásica , Proteínas de Neoplasias/genética , Intervalo Livre de Progressão , Interferência de RNA , Fator de Transcrição AP-2/antagonistas & inibidores , Fator de Transcrição AP-2/química , Fator de Transcrição AP-2/genética
8.
Oncotarget ; 7(3): 2985-3001, 2016 Jan 19.
Artigo em Inglês | MEDLINE | ID: mdl-26672764

RESUMO

Melatonin, a molecule produced throughout the animal and plant kingdoms, and berberine, a plant derived agent, both exhibit antitumor and multiple biological and pharmacological effects, but they have never been combined altogether for the inhibition of human lung cancers. In this study, we investigated the role and underlying mechanisms of melatonin in the regulation of antitumor activity of berberine in lung cancer cells. Treatment with melatonin effectively increased the berberine-mediated inhibitions of cell proliferation, colony formation and cell migration, thereby enhancing the sensitivities of lung cancer cells to berberine. Melatonin also markedly increased apoptosis induced by berberine. Further mechanism study showed that melatonin promoted the cleavage of caspse-9 and PARP, enhanced the inhibition of Bcl2, and triggered the releasing of cytochrome C (Cyto C), thereby increasing the berberine-induced apoptosis. Melatonin also enhanced the berberine-mediated inhibition of telomerase reverses transcriptase (hTERT) by down-regulating the expression of AP-2ß and its binding on hTERT promoter. Moreover, melatonin enhanced the berberine-mediated inhibition of cyclooxygenase 2 (COX-2) by inhibiting the nuclear translocation of NF-κB and its binding on COX-2 promoter. Melatonin also increased the berberine-mediated inhibition of the phosphorylated Akt and ERK. Collectively, our results demonstrated that melatonin enhanced the antitumor activity of berberine by activating caspase/Cyto C and inhibiting AP-2ß/hTERT, NF-κB/COX-2 and Akt/ERK signaling pathways. Our findings provide new insights in exploring the potential therapeutic strategies and novel targets for lung cancer treatment.


Assuntos
Antineoplásicos/farmacologia , Berberina/farmacologia , Inibidores de Ciclo-Oxigenase 2/farmacologia , MAP Quinases Reguladas por Sinal Extracelular/antagonistas & inibidores , Neoplasias Pulmonares/tratamento farmacológico , Melatonina/farmacologia , NF-kappa B/antagonistas & inibidores , Proteínas Proto-Oncogênicas c-akt/antagonistas & inibidores , Telomerase/antagonistas & inibidores , Fator de Transcrição AP-2/antagonistas & inibidores , Transporte Ativo do Núcleo Celular , Animais , Protocolos de Quimioterapia Combinada Antineoplásica , Apoptose/efeitos dos fármacos , Caspase 9/metabolismo , Linhagem Celular Tumoral , Ciclo-Oxigenase 2/metabolismo , Citocromos c/metabolismo , Proteínas de Ligação a DNA/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , NF-kappa B/metabolismo , Poli(ADP-Ribose) Polimerases/metabolismo , Regiões Promotoras Genéticas/genética , Proteínas Proto-Oncogênicas c-bcl-2/antagonistas & inibidores , Transdução de Sinais/efeitos dos fármacos , Fator de Transcrição AP-2/biossíntese , Ensaios Antitumorais Modelo de Xenoenxerto
9.
Cell Rep ; 10(8): 1398-409, 2015 Mar 03.
Artigo em Inglês | MEDLINE | ID: mdl-25732829

RESUMO

A challenge of genome annotation is the identification of genes performing specific biological functions. Here, we propose a phylogenetic approach that utilizes RNA-seq data to infer the historical relationships among cell types and to trace the pattern of gene-expression changes on the tree. The hypothesis is that gene-expression changes coincidental with the origin of a cell type will be important for the function of the derived cell type. We apply this approach to the endometrial stromal cells (ESCs), which are critical for the initiation and maintenance of pregnancy. Our approach identified well-known regulators of ESCs, PGR and FOXO1, as well as genes not yet implicated in female fertility, including GATA2 and TFAP2C. Knockdown analysis confirmed that they are essential for ESC differentiation. We conclude that phylogenetic analysis of cell transcriptomes is a powerful tool for discovery of genes performing cell-type-specific functions.


Assuntos
Endométrio/citologia , Células Estromais/metabolismo , Diferenciação Celular , Linhagem Celular , Análise por Conglomerados , Feminino , Proteína Forkhead Box O1 , Fatores de Transcrição Forkhead/antagonistas & inibidores , Fatores de Transcrição Forkhead/genética , Fatores de Transcrição Forkhead/metabolismo , Fator de Transcrição GATA2/antagonistas & inibidores , Fator de Transcrição GATA2/genética , Fator de Transcrição GATA2/metabolismo , Humanos , Miofibroblastos/citologia , Miofibroblastos/metabolismo , Gravidez , Análise de Componente Principal , Interferência de RNA , RNA Interferente Pequeno/metabolismo , Análise de Sequência de RNA , Células Estromais/citologia , Fator de Transcrição AP-2/antagonistas & inibidores , Fator de Transcrição AP-2/genética , Fator de Transcrição AP-2/metabolismo
10.
Mol Cancer ; 13: 89, 2014 Apr 26.
Artigo em Inglês | MEDLINE | ID: mdl-24766673

RESUMO

BACKGROUND: TFAP2B is a member of the AP2 transcription factor family, which orchestrates a variety of cell processes. However, the roles of TFAP2B in regulating carcinogenesis remain largely unknown. Here, we investigated the regulatory effects of TFAP2B on lung adenocarcinomas growth and identified the underlying mechanisms of actions in non-small cell lung cancer (NSCLC) cells. METHODS: We first examined the expression of TFAP2B in lung cancer cell lines and tumor tissues. We also analyzed the prognostic predicting value of TFAP2B in lung adenocarcinomas. Then we investigated the molecular mechanisms by which TFAP2B knockdown or overexpression regulated lung cancer cell growth, angiogenesis and apoptosis, and further confirmed the role of TFAP2B in tumor growth in a lung cancer xenograft mouse model. RESULTS: TFAP2B was highly expressed in NSCLC cell lines and tumor tissues. Strong TFAP2B expression showed a positive correlation with the poor prognoses of patients with lung adenocarcinomas (P < 0.001). TFAP2B knockdown by siRNA significantly inhibited cell growth and induced apoptosis in NSCLC cells in vitro and in a lung cancer subcutaneous xenograft model, whereas TFAP2B overexpression promoted cell growth. The observed regulation of cell growth was accompanied by the TFAP2B-mediated modulation of the ERK/p38, caspase/cytochrome-c and VEGF/PEDF-dependent signaling pathways in NSCLC cells. CONCLUSIONS: These results indicate that TFAP2B plays a critical role in regulating lung adenocarcinomas growth and could serve as a promising therapeutic target for lung cancer treatment.


Assuntos
Adenocarcinoma/genética , MAP Quinases Reguladas por Sinal Extracelular/genética , Proteínas do Olho/genética , Regulação Neoplásica da Expressão Gênica , Neoplasias Pulmonares/genética , Fatores de Crescimento Neural/genética , Serpinas/genética , Fator de Transcrição AP-2/genética , Fator A de Crescimento do Endotélio Vascular/genética , Adenocarcinoma/irrigação sanguínea , Adenocarcinoma/mortalidade , Adenocarcinoma/patologia , Idoso , Animais , Apoptose , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Proteínas do Olho/metabolismo , Feminino , Humanos , Neoplasias Pulmonares/irrigação sanguínea , Neoplasias Pulmonares/mortalidade , Neoplasias Pulmonares/patologia , Masculino , Camundongos , Pessoa de Meia-Idade , Transplante de Neoplasias , Neovascularização Patológica/prevenção & controle , Fatores de Crescimento Neural/metabolismo , Prognóstico , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Serpinas/metabolismo , Transdução de Sinais , Análise de Sobrevida , Fator de Transcrição AP-2/antagonistas & inibidores , Fator de Transcrição AP-2/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo
11.
J Neurochem ; 129(1): 72-84, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24188130

RESUMO

Retinal ganglion cells transmit the visual signal from the retina to the brain. We have previously shown that the activator protein 2 (AP-2)δ (TFAP2D) transcription factor is expressed in one third of ganglion cells in developing retina suggesting a specialized role for these AP-2δ-expressing cells. Here, we address the role of AP-2δ in retina by in ovo electroporation of RCAS/AP-2δ retroviral constructs into the eyes of chick embryos at day 2 of gestation. Ectopic expression of AP-2δ does not affect lineage differentiation in the developing retina. However, immunostaining of retinal tissue with markers associated with axonal growth such as growth-associated protein 43 and polysialic acid-neural cell adhesion molecule (PSA-NCAM) demonstrates axonal misrouting and abnormal axonal bundling. Treatment of AP-2δ-misexpressing retinal cell cultures with endoneuraminidase, an enzyme that removes PSA from NCAM, decreases AP-2δ-induced axonal bundling. Our data suggest a role for AP-2δ in polysialylation of NCAM, with ectopic expression of AP-2δ resulting in premature bundling of emerging axons and misrouting of axons. We propose that expression of AP-2δ in a subset of ganglion cells contributes to the fine-tuning of axonal growth in the developing retina.


Assuntos
Axônios/fisiologia , Coristoma , Regulação da Expressão Gênica no Desenvolvimento , Molécula L1 de Adesão de Célula Nervosa/biossíntese , Retina/embriologia , Retina/metabolismo , Ácidos Siálicos/biossíntese , Fator de Transcrição AP-2/biossíntese , Animais , Embrião de Galinha , Galinhas , Glicosídeo Hidrolases/farmacologia , Molécula L1 de Adesão de Célula Nervosa/antagonistas & inibidores , Retina/efeitos dos fármacos , Ácidos Siálicos/antagonistas & inibidores , Fator de Transcrição AP-2/antagonistas & inibidores
12.
Cancer Prev Res (Phila) ; 7(2): 266-77, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24335623

RESUMO

TFAP2A is a transcription factor that orchestrates a variety of cell processes, including cell growth and tissue differentiation. However, the regulation of TFAP2A in human nasopharyngeal carcinoma tumorigenesis and its precise mechanism of action remain largely unknown. In this study, we investigated the biologic role and clinical significance of TFAP2A in nasopharyngeal carcinoma growth and progression and identified the underlying molecular mechanisms. We found that TFAP2A was highly expressed in various nasopharyngeal carcinoma cell lines and tumor tissue specimens and was significantly correlated with hypoxia-inducible factor-1α (HIF-1α) expression. A positive correlation of TFAP2A overexpression with advanced tumor stage, local invasion, clinical progression, and poor prognosis of patients with nasopharyngeal carcinomas were also observed. Moreover, we found that knockdown of TFAP2A expression by siRNA significantly inhibited tumor cell growth in nasopharyngeal carcinoma cell lines and in a subcutaneous xenograft mouse model by targeting the HIF-1α-mediated VEGF/pigment epithelium-derived factor (PEDF) signaling pathway. Treatment of nasopharyngeal carcinoma cells with TFAP2A siRNA dramatically inhibited the expression and the release of VEGF protein but did not change the level of PEDF protein, resulting in a significant reduction of the ratio of VEGF/PEDF. Pretreatment with a HIF-1α siRNA did not significantly change the TFAP2A siRNA-mediated inhibition in cell viability. Our results indicate that TFAP2A regulates nasopharyngeal carcinoma growth and survival through the modulation of the HIF-1α-mediated VEGF/PEDF signaling pathway, and suggest that TFAP2A could be a potential prognostic biomarker and therapeutic target for nasopharyngeal carcinoma treatment.


Assuntos
Proliferação de Células , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Neoplasias Nasofaríngeas/patologia , Fator de Transcrição AP-2/fisiologia , Adulto , Idoso , Animais , Carcinoma , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/genética , Feminino , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Masculino , Camundongos , Pessoa de Meia-Idade , Carcinoma Nasofaríngeo , Neoplasias Nasofaríngeas/genética , RNA Interferente Pequeno/farmacologia , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética , Fator de Transcrição AP-2/antagonistas & inibidores , Ensaios Antitumorais Modelo de Xenoenxerto , Adulto Jovem
13.
Acta Pharmacol Sin ; 34(9): 1192-200, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23685957

RESUMO

AIM: To investigate the effects of curcumin on proliferation and apoptosis in testicular cancer cells in vitro and to investigate its molecular mechanisms of action. METHODS: NTera-2 human malignant testicular germ cell line and F9 mouse teratocarcinoma stem cell line were used. The anti-proliferative effect was examined using MTT and colony formation assays. Hoechst 33258 staining, TUNEL and Annexin V-FITC/PI staining assays were used to analyze cell apoptosis. Protein expression was examined with Western blot analysis and immunocytochemical staining. RESULTS: Curcumin (5, 10 and 15 µmol/L) inhibited the viability of NTera-2 cells in dose- and time-dependent manners. Curcumin significantly inhibited the colony formation in both NTera-2 and F9 cells. Curcumin dose-dependently induced apoptosis of NTera-2 cells by reducing FasL expression and Bcl-2-to-Bax ratio, and activating caspase-9, -8 and -3. Furthermore, curcumin dose-dependently reduced the expression of AP transcription factor AP-2γ in NTera-2 cells, whereas the pretreatment with the proteasome inhibitor MG132 blocked both the curcumin-induced reduction of AP-2γ and antiproliferative effect. Curcumin inhibited ErbB2 expression, and decreased the phosphorylation of Akt and ERK in NTera-2 cells. CONCLUSION: Curcumin induces apoptosis and inhibits proliferation in NTera-2 cells via the inhibition of AP-2γ-mediated downstream cell survival signaling pathways.


Assuntos
Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Curcumina/farmacologia , Neoplasias Embrionárias de Células Germinativas , Neoplasias Testiculares , Fator de Transcrição AP-2/antagonistas & inibidores , Fator de Transcrição AP-2/farmacologia , Animais , Antineoplásicos/uso terapêutico , Apoptose/fisiologia , Curcumina/uso terapêutico , Humanos , Camundongos , Neoplasias Embrionárias de Células Germinativas/tratamento farmacológico , Neoplasias Embrionárias de Células Germinativas/patologia , Neoplasias Testiculares/tratamento farmacológico , Neoplasias Testiculares/patologia
14.
Mol Med Rep ; 7(3): 909-14, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23292806

RESUMO

Aspirin has been reported to trigger apoptosis in various cancer cell lines. However, the detailed mechanisms involved remain elusive. The present study aimed to investigate whether aspirin plays a role in apoptosis of MDA-MB-453 cells. The effect of aspirin on the proliferation of human MDA-MB-453 cells breast cancer cells was evaluated using MTT assay, flow cytometry and western blotting. The present study reports that aspirin induces the apoptosis of MDA­MB­453 breast cancer cells which was attributed to the increased expression and activation of caspase­3. Moreover, AP­2α, a transcription factor highly expressed in MDA­MB­453 cells, was identified as a negative regulator of caspase­3 transcription and AP­2α was attenuated following aspirin treatment. Therefore, aspirin may increase the expression of caspase­3 by inducing the degradation of AP­2α, which increases activated caspase­3 expression, thereby triggering apoptosis in MDA­MB­453 cells. Thus, aspirin may be used in breast cancer therapy.


Assuntos
Anti-Inflamatórios não Esteroides/toxicidade , Apoptose/efeitos dos fármacos , Aspirina/toxicidade , Caspase 3/metabolismo , Fator de Transcrição AP-2/metabolismo , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Caspase 3/genética , Linhagem Celular Tumoral , Feminino , Humanos , Regiões Promotoras Genéticas , Complexo de Endopeptidases do Proteassoma/metabolismo , Ligação Proteica , Interferência de RNA , RNA Interferente Pequeno/metabolismo , Fator de Transcrição AP-2/antagonistas & inibidores , Fator de Transcrição AP-2/genética , Regulação para Cima/efeitos dos fármacos
15.
Nucleic Acids Res ; 40(15): 7190-206, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22573176

RESUMO

The p63 transcription factor (TP63) is critical in development, growth and differentiation of stratifying epithelia. This is highlighted by the severity of congenital abnormalities caused by TP63 mutations in humans, the dramatic phenotypes in knockout mice and de-regulation of TP63 expression in neoplasia altering the tumour suppressive roles of the TP53 family. In order to define the normal role played by TP63 and provide the basis for better understanding how this network is perturbed in disease, we used chromatin immunoprecipitation combined with massively parallel sequencing (ChIP-seq) to identify >7500 high-confidence TP63-binding regions across the entire genome, in primary human neonatal foreskin keratinocytes (HFKs). Using integrative strategies, we demonstrate that only a subset of these sites are bound by TP53 in response to DNA damage. We identify a role for TP63 in transcriptional regulation of multiple genes genetically linked to cleft palate and identify AP-2alpha (TFAP2A) as a co-regulator of a subset of these genes. We further demonstrate that AP-2gamma (TFAP2C) can bind a subset of these regions and that acute depletion of either TFAP2A or TFAP2C alone is sufficient to reduce terminal differentiation of organotypic epidermal skin equivalents, indicating overlapping physiological functions with TP63.


Assuntos
Células Epidérmicas , Queratinócitos/metabolismo , Fator de Transcrição AP-2/metabolismo , Fatores de Transcrição/metabolismo , Proteínas Supressoras de Tumor/metabolismo , Sítios de Ligação , Diferenciação Celular , Células Cultivadas , Fissura Palatina/genética , Regulação da Expressão Gênica , Genoma Humano , Humanos , Queratinócitos/citologia , Anotação de Sequência Molecular , Elementos Reguladores de Transcrição , Fator de Transcrição AP-2/antagonistas & inibidores , Proteína Supressora de Tumor p53/metabolismo
16.
EMBO J ; 30(13): 2569-81, 2011 May 13.
Artigo em Inglês | MEDLINE | ID: mdl-21572391

RESUMO

Oestrogen receptor α (ERα) is key player in the progression of breast cancer. Recently, the cistrome and interactome of ERα were mapped in breast cancer cells, revealing the importance of spatial organization in oestrogen-mediated transcription. However, the underlying mechanism of this process is unclear. Here, we show that ERα binding sites (ERBS) identified from the Chromatin Interaction Analysis-Paired End DiTag of ERα are enriched for AP-2 motifs. We demonstrate the transcription factor, AP-2γ, which has been implicated in breast cancer oncogenesis, binds to ERBS in a ligand-independent manner. Furthermore, perturbation of AP-2γ expression impaired ERα DNA binding, long-range chromatin interactions, and gene transcription. In genome-wide analyses, we show that a large number of AP-2γ and ERα binding events converge together across the genome. The majority of these shared regions are also occupied by the pioneer factor, FoxA1. Molecular studies indicate there is functional interplay between AP-2γ and FoxA1. Finally, we show that most ERBS associated with long-range chromatin interactions are colocalized with AP-2γ and FoxA1. Together, our results suggest AP-2γ is a novel collaborative factor in ERα-mediated transcription.


Assuntos
Cromatina/metabolismo , Receptor alfa de Estrogênio/fisiologia , Fator de Transcrição AP-2/fisiologia , Transcrição Gênica , Sítios de Ligação/genética , Cromatina/genética , Imunoprecipitação da Cromatina , Análise por Conglomerados , Receptor alfa de Estrogênio/antagonistas & inibidores , Receptor alfa de Estrogênio/genética , Receptor alfa de Estrogênio/metabolismo , Perfilação da Expressão Gênica , Técnicas de Silenciamento de Genes , Humanos , Análise em Microsséries , Modelos Biológicos , Ligação Proteica/efeitos dos fármacos , Ligação Proteica/genética , Ligação Proteica/fisiologia , RNA Interferente Pequeno/farmacologia , Elementos de Resposta/genética , Fator de Transcrição AP-2/antagonistas & inibidores , Fator de Transcrição AP-2/genética , Transcrição Gênica/genética , Transcrição Gênica/fisiologia , Células Tumorais Cultivadas
17.
FEBS Lett ; 584(16): 3615-9, 2010 Aug 20.
Artigo em Inglês | MEDLINE | ID: mdl-20659467

RESUMO

The role of transcription factor activator protein-2beta (AP-2beta) in foam cell formation was investigated. Uptake assay of DiI-labeled oxidized low density lipoprotein and the lipid quantitative analysis by high performance liquid chromatography showed AP-2beta promoted the lipid accumulation. The quantitative real-time RT-PCR and Western blot indicated that AP-2beta upregulated the expression of CD36. Luciferase assay, electrophoretic mobility shift assay and chromatin immunoprecipitation confirmed that AP-2beta bound to AP-2 binding sites in CD36 promoter region and enhanced the promoter activity of CD36 gene. We conclude that AP-2beta may function in foam cell formation through enhancing the transcription and expression of CD36 followed by increased lipid uptake.


Assuntos
Antígenos CD36/genética , Metabolismo dos Lipídeos , Macrófagos/metabolismo , Fator de Transcrição AP-2/metabolismo , Animais , Aterosclerose , Sequência de Bases , Sítios de Ligação/genética , Linhagem Celular , DNA/genética , DNA/metabolismo , Células Espumosas/metabolismo , Humanos , Lipoproteínas LDL/metabolismo , Camundongos , Dados de Sequência Molecular , Mutação , Regiões Promotoras Genéticas , RNA Interferente Pequeno/genética , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Homologia de Sequência do Ácido Nucleico , Fator de Transcrição AP-2/antagonistas & inibidores , Fator de Transcrição AP-2/genética , Transcrição Gênica
18.
Genes Chromosomes Cancer ; 49(10): 948-62, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-20629094

RESUMO

The TFAP2C transcription factor is involved in mammary development, differentiation, and oncogenesis. Previous studies established a role for TFAP2C in the regulation of ESR1 (ERalpha) and ERBB2 (Her2) in breast carcinomas. However, the role of TFAP2C in different breast cancer phenotypes has not been examined in detail. To develop a more complete characterization of TFAP2C target genes, ChIP-seq with anti-TFAP2C antibody and expression arrays with TFAP2C knock down were analyzed in MCF-7 breast carcinoma cells. Genomic sequences common to the ChIP-seq data set defined the consensus sequence for TFAP2C chromatin binding as the nine base sequence SCCTSRGGS (S = G/C, r = A/G), which closely matches the previously defined optimal in vitro binding site. Comparing expression arrays before and after knock down of TFAP2C with ChIP-seq data demonstrated a conservative estimate that 8% of genes altered by TFAP2C expression are primary target genes and includes genes that are both induced and repressed by TFAP2C. A set of 447 primary target genes of TFAP2C was identified, which included ESR1 (ERalpha), FREM2, RET, FOXA1, WWOX, GREB1, MYC, and members of the retinoic acid response pathway. The identification of ESR1, WWOX, GREB1, and FOXA1 as primary targets confirmed the role of TFAP2C in hormone response. TFAP2C plays a critical role in gene regulation in hormone responsive breast cancer and its target genes are different than for the Her2 breast cancer phenotype.


Assuntos
Biomarcadores Tumorais/genética , Neoplasias da Mama/genética , Regulação Neoplásica da Expressão Gênica , Neoplasias Hormônio-Dependentes/genética , Regiões Promotoras Genéticas/genética , Fator de Transcrição AP-2/genética , Biomarcadores Tumorais/metabolismo , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Linhagem Celular Tumoral , Imunoprecipitação da Cromatina , Feminino , Perfilação da Expressão Gênica , Humanos , Neoplasias Hormônio-Dependentes/metabolismo , Neoplasias Hormônio-Dependentes/patologia , Análise de Sequência com Séries de Oligonucleotídeos , RNA Mensageiro/genética , RNA Interferente Pequeno/farmacologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fator de Transcrição AP-2/antagonistas & inibidores , Fator de Transcrição AP-2/metabolismo
19.
Carcinogenesis ; 31(5): 918-26, 2010 May.
Artigo em Inglês | MEDLINE | ID: mdl-20123756

RESUMO

A common feature shared between several human cancer-associated viruses, such as Epstein-Barr virus, Hepatitis B virus and Hepatitis C virus, and Human papillomavirus (HPV) is the ability to reduce the expression of cellular E-cadherin. Since E-cadherin is used by Langerhans cells to move through the stratified epithelium, its reduction may affect the efficiency by which the immune system responds to HPV infection and the length of persistent HPV infections. We observed that the E7 protein of this virus (HPV16) is most efficient at reducing E-cadherin levels. This E7 activity is independent of retinoblastoma protein or AP-2alpha degradation. Instead it is associated with augmentation of cellular DNA methyltransferase I (Dnmt1) activity. Significantly, inhibition of Dnmt activity re-established E-cadherin levels of the cells, presenting the possibility that similar epigenetic intervention clinically may be a way to re-establish the influx of Langerhans cells into infected epithelium to counteract HPV persistence.


Assuntos
Caderinas/análise , Epigênese Genética , Proteínas E7 de Papillomavirus/fisiologia , Caderinas/genética , Caderinas/metabolismo , DNA (Citosina-5-)-Metiltransferase 1 , DNA (Citosina-5-)-Metiltransferases/análise , DNA (Citosina-5-)-Metiltransferases/antagonistas & inibidores , DNA (Citosina-5-)-Metiltransferases/fisiologia , Humanos , Proteína do Retinoblastoma/metabolismo , Fator de Transcrição AP-2/antagonistas & inibidores
20.
Cancer Res ; 69(16): 6445-53, 2009 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-19654299

RESUMO

We previously showed that nicotine stimulates non-small cell lung carcinoma (NSCLC) cell proliferation through nicotinic acetylcholine receptor (nAChR)-mediated signals. Activation of peroxisome proliferator-activated receptor beta/delta (PPARbeta/delta) has also been shown to induce NSCLC cell growth. Here, we explore the potential link between nicotine and PPARbeta/delta and report that nicotine increases the expression of PPARbeta/delta protein; this effect was blocked by an alpha7 nAChR antagonist (alpha-bungarotoxin), by alpha7 nAChR short interfering RNA, and by inhibitors of phosphatidylinositol 3-kinase (PI3K; wortmannin and LY294002) and mammalian target of rapamycin (mTOR; rapamycin). In contrast, this effect was enhanced by PUN282987, an alpha7 nAChR agonist. Silencing of PPARbeta/delta attenuated the stimulatory effect of nicotine on cell growth, which was overcome by transfection of an exogenous PPARbeta/delta expression vector. Of note, nicotine induced complex formation between alpha7 nAChR and PPARbeta/delta protein and increased PPARbeta/delta gene promoter activity through inhibition of AP-2alpha as shown by reduced AP-2alpha binding using electrophoretic gel mobility shift and chromatin immunoprecipitation assays. In addition, silencing of Sp1 attenuated the effect of nicotine on PPARbeta/delta. Collectively, our results show that nicotine increases PPARbeta/delta gene expression through alpha7 nAChR-mediated activation of PI3K/mTOR signals that inhibit AP-2alpha protein expression and DNA binding activity to the PPARbeta/delta gene promoter. Sp1 seems to modulate this process. This study unveils a novel mechanism by which nicotine promotes human lung carcinoma cell growth.


Assuntos
Carcinoma Pulmonar de Células não Pequenas/metabolismo , Neoplasias Pulmonares/metabolismo , Nicotina/farmacologia , PPAR delta/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Quinases/metabolismo , Fator de Transcrição AP-2/antagonistas & inibidores , Bungarotoxinas/farmacologia , Carcinoma Pulmonar de Células não Pequenas/genética , Proliferação de Células/efeitos dos fármacos , Agonistas Colinérgicos/farmacologia , Ativação Enzimática/efeitos dos fármacos , Ativação Enzimática/fisiologia , Regulação Neoplásica da Expressão Gênica , Humanos , Neoplasias Pulmonares/genética , Modelos Biológicos , PPAR delta/genética , PPAR beta/genética , PPAR beta/metabolismo , Fosfatidilinositol 3-Quinases/fisiologia , Regiões Promotoras Genéticas/efeitos dos fármacos , Proteínas Quinases/fisiologia , Receptores Nicotínicos/fisiologia , Serina-Treonina Quinases TOR , Fator de Transcrição AP-2/metabolismo , Células Tumorais Cultivadas , Regulação para Cima/efeitos dos fármacos , Receptor Nicotínico de Acetilcolina alfa7
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA