Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Acta Biochim Biophys Sin (Shanghai) ; 51(12): 1258-1266, 2019 Dec 13.
Artigo em Inglês | MEDLINE | ID: mdl-31769473

RESUMO

SRC3 plays critical roles in various biological processes of diseases, including proliferation, apoptosis, migration, and cell cycle arrest. However, the effect of SRC3 expression in mesenchymal stem cells (MSCs) on multiple myeloma (MM) is not clear yet. In our study, MSCs (MSC-SRC3, MSC-SRC3-/-) and MM cells were co-cultured in a direct or indirect way. The proliferation of MM cells was studied by CCK-8 and colony formation assays. The apoptosis and cell cycle of MM cells were detected by flow cytometry. In addition, the expressions of proteins in MM cells were detected by western blot analysis and the secretions of cytokines were measured by ELISA. Our data showed that the expression of SRC3 in bone marrow mesenchymal stem cells (BM-MSCs) could promote cell proliferation and colony formation of MM cells through accelerating the transformation of the G1/S phase, no matter what kind of culture method was adopted. Meanwhile, SRC3 expressed in BM-MSCs could inhibit the apoptosis of MM cells through the caspase apoptosis pathway and mitochondrial apoptosis pathway. Moreover, SRC3 could enhance the adhesion ability of MM cells through up-regulating the expression of adhesion molecules including CXCL4, ICAM1, VLA4, and syndecan-1. SRC3 also played a regulatory role in the progress of MM through the NF-κB and PI-3K/Akt pathways. SRC3 expressed in MSCs was found to promote the growth and survival of MM cells, while SRC3 silencing in MSCs could inhibit the development of MM. These results would be useful for developing a more effective new strategy for MM treatment.


Assuntos
Células-Tronco Mesenquimais/metabolismo , Mieloma Múltiplo/metabolismo , Coativador 3 de Receptor Nuclear/fisiologia , Animais , Apoptose , Adesão Celular , Ciclo Celular , Pontos de Checagem do Ciclo Celular , Proliferação de Células , Células Cultivadas , Técnicas de Cocultura , Citocinas/metabolismo , Células-Tronco Mesenquimais/patologia , Camundongos , Camundongos Endogâmicos C57BL
2.
Cell Mol Biol (Noisy-le-grand) ; 64(6): 65-70, 2018 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-29808803

RESUMO

AIB1 was involved in the development and progression of breast cancer. Although it was found that AIB1 could be phosphorylated by some kinases including PI3K, the function of AIB1 and AKT interaction in breast cancer is not well defined. MCF-7 cells were transfected with pERE-Luc AKT and/or AIB1 plasmids, and then ERE luciferase activity in presence or absence of estrogen (E2) were measured. Plasmids containing PTEN and an PI3K inhibitor LY294002 were transfected into or treated cells to identify the interaction of PI3K/AKT and activation of AIB1, and examine their roles in cell cycle regulation. The AKT phosphorylation activity was evaluated by kinase assay using H2B as a substrate. The association between A1B1 and pS2 promoter was detected by the Chromatin Immunoprecipitation (ChIP) assay. AIB1 and AKT in the same complex were detected by Pull-down assay. IGF-1 can increase AIB1 recruitment to PS2 and enhance the ER-dependent transcription activity through the PI3K/AKT pathway. AIB1 associate with AKT to regulate cell cycle. The special relations concerning the AIB1 and AKT may arouse some new viewpoints for potential therapeutic targets in breast cancer.


Assuntos
Neoplasias da Mama/patologia , Estradiol/farmacologia , Regulação Neoplásica da Expressão Gênica , Proteínas de Neoplasias/fisiologia , Neoplasias Hormônio-Dependentes/patologia , Coativador 3 de Receptor Nuclear/fisiologia , Fosfatidilinositol 3-Quinases/fisiologia , Proteínas Proto-Oncogênicas c-akt/fisiologia , Transdução de Sinais/fisiologia , Animais , Ciclo Celular/fisiologia , Feminino , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica/fisiologia , Humanos , Imunoprecipitação , Fator de Crescimento Insulin-Like I/farmacologia , Células MCF-7 , Camundongos , Proteínas de Neoplasias/genética , Coativador 3 de Receptor Nuclear/genética , Fosforilação , Presenilina-2/biossíntese , Presenilina-2/genética , Regiões Promotoras Genéticas , Processamento de Proteína Pós-Traducional , Proteínas Proto-Oncogênicas c-akt/genética , Receptores de Estrogênio/efeitos dos fármacos , Receptores de Estrogênio/fisiologia , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Transfecção
3.
Mol Cell ; 37(3): 321-32, 2010 Feb 12.
Artigo em Inglês | MEDLINE | ID: mdl-20159552

RESUMO

EGF induces signal transduction between EGFR and FAK, and FAK is required for EGF-induced cell migration. It is unknown, however, what factor mediates the interaction between EGFR and FAK and leads to EGF-induced FAK phosphorylation. Here, we identify SRC-3Delta4, a splicing isoform of the SRC-3 oncogene, as a signaling adaptor that links EGFR and FAK and promotes EGF-induced phosphorylations of FAK and c-Src. We identify three PAK1-mediated phosphorylations in SRC-3Delta4 that promote the localization of SRC-3Delta4 to the plasma membrane and mediate the interactions with EGFR and FAK. Importantly, overexpression of SRC-3Delta4 promotes MDA-MB231-induced breast tumor metastasis. Our findings identify phosphorylated SRC-3Delta4 as a missing adaptor between EGFR and its downstream signaling molecule FAK to coordinately regulate EGF-induced cell migration. Our study also reveals that a nuclear receptor coactivator can act in the periphery of a cell to directly mediate activation of an enzyme.


Assuntos
Movimento Celular/fisiologia , Receptores ErbB/metabolismo , Quinase 1 de Adesão Focal/metabolismo , Coativador 3 de Receptor Nuclear/fisiologia , Animais , Linhagem Celular Tumoral , Feminino , Quinase 1 de Adesão Focal/análise , Humanos , Neoplasias Pulmonares/secundário , Linfonodos/patologia , Metástase Linfática , Camundongos , Metástase Neoplásica , Coativador 3 de Receptor Nuclear/análise , Coativador 3 de Receptor Nuclear/genética , Fosforilação , Isoformas de Proteínas/genética , Isoformas de Proteínas/fisiologia , Transdução de Sinais , Quinases Ativadas por p21/metabolismo , Quinases da Família src/metabolismo , Quinases da Família src/fisiologia
4.
Nucleic Acids Res ; 41(18): 8515-25, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23887939

RESUMO

The glucocorticoid receptor (GR) is a ligand activated transcription factor, serving to regulate both energy metabolism and immune functions. Factors that influence cellular sensitivity to glucocorticoids (GC) are therefore of great interest. The N-terminal of the GR contains numerous potential proline-directed phosphorylation sites, some of which can regulate GR transactivation. Unrestricted proline isomerisation can be inhibited by adjacent serine phosphorylation and requires a prolyl isomerise, Pin1. Pin1 therefore determines the functional outcome of proline-directed kinases acting on the GR, as cis/trans isomers are distinct pools with different interacting proteins. We show that Pin1 mediates GR transactivation, but not GR trans-repression. Two N-terminal GR serines, S203 and S211, are targets for Pin1 potentiation of GR transactivation, establishing a direct link between Pin1 and the GR. We also demonstrate GC-activated co-recruitment of GR and Pin1 to the GILZ gene promoter. The Pin1 effect required both its WW and catalytic domains, and GR recruitment to its GRE was Pin1-dependent. Therefore, Pin1 is a selective regulator of GR transactivation, acting through N-terminal phospho-serine residues to regulate GR recruitment to its target sites in the genome. As Pin1 is dysregulated in disease states, this interaction may contribute to altered GC action in inflammatory conditions.


Assuntos
Peptidilprolil Isomerase/fisiologia , Receptores de Glucocorticoides/metabolismo , Ativação Transcricional , Linhagem Celular , Dexametasona/farmacologia , Humanos , Peptidilprolil Isomerase de Interação com NIMA , Coativador 3 de Receptor Nuclear/fisiologia , Peptidilprolil Isomerase/antagonistas & inibidores , Fosforilação , Regiões Promotoras Genéticas , Estabilidade Proteica , Receptores de Glucocorticoides/química , Proteínas Repressoras/metabolismo
5.
Tumour Biol ; 35(4): 3087-94, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24282087

RESUMO

Increasing evidence suggests that the three homologous members of steroid receptor co-activator (SRC) family (SRC-1, SRC-2, and SRC-3) play key roles in enhancing cell proliferation in various human cancers, such as breast, prostate, and hepatocellular carcinoma. However, the function of SRC-3 in osteosarcoma remains largely unexplored. In the current study, we found that SRC-3, but not SRC-1 and SRC-2, was dramatically up-regulated in human osteosarcoma tissues, compared with adjacent normal tissues. To explore the functions of SRC-3 in osteosarcoma, in vitro studies were performed in MG63 and U2OS cells. SRC-3 overexpression promoted osteosarcoma cell proliferation, whereas knockdown of SRC-3 inhibits its proliferation. In support of these findings, we further demonstrated that SRC-3 up-regulated FoxM1 expression through co-activation of C/EBPγ. Together our results show that SRC-3 drives osteosarcoma progression and imply it as a therapeutic target to abrogate osteosarcoma.


Assuntos
Neoplasias Ósseas/patologia , Fatores de Transcrição Forkhead/genética , Coativador 3 de Receptor Nuclear/fisiologia , Osteossarcoma/patologia , Sítios de Ligação , Neoplasias Ósseas/metabolismo , Proteínas Estimuladoras de Ligação a CCAAT/metabolismo , Linhagem Celular Tumoral , Progressão da Doença , Proteína Forkhead Box M1 , Regulação Neoplásica da Expressão Gênica , Humanos , Coativador 3 de Receptor Nuclear/antagonistas & inibidores , Osteossarcoma/metabolismo , Regiões Promotoras Genéticas , Regulação para Cima
6.
Br J Cancer ; 108(10): 2039-44, 2013 May 28.
Artigo em Inglês | MEDLINE | ID: mdl-23652306

RESUMO

BACKGROUND: Steroid receptor coactivator 3 (SRC3) is an important coactivator of a number of transcription factors and is associated with a poor outcome in numerous tumours. Steroid receptor coactivator 3 is amplified in 25% of epithelial ovarian cancers (EOCs) and its expression is higher in EOCs compared with non-malignant tissue. No data is currently available with regard to the expression of SRC-3 in EOC and its influence on outcome or the efficacy of treatment. METHODS: Immunohistochemistry was performed for SRC3, oestrogen receptor-α, HER2, PAX2 and PAR6, and protein expression was quantified using automated quantitative immunofluorescence (AQUA) in 471 EOCs treated between 1991 and 2006 with cytoreductive surgery followed by first-line treatment platinum-based therapy, with or without a taxane. RESULTS: Steroid receptor coactivator 3 expression was significantly associated with advanced stage and was an independent prognostic marker. High expression of SRC3 identified patients who have a significantly poorer survival with single-agent carboplatin chemotherapy, while with carboplatin/paclitaxel treatment such a difference was not seen. CONCLUSION: Steroid receptor coactivator 3 is a poor prognostic factor in EOCs and appears to identify a population of patients who would benefit from the addition of taxanes to their chemotherapy regimen, due to intrinsic resistance to platinum therapy.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Biomarcadores Farmacológicos , Resistencia a Medicamentos Antineoplásicos , Neoplasias Epiteliais e Glandulares/diagnóstico , Neoplasias Epiteliais e Glandulares/tratamento farmacológico , Coativador 3 de Receptor Nuclear/fisiologia , Neoplasias Ovarianas/diagnóstico , Neoplasias Ovarianas/tratamento farmacológico , Compostos de Platina/uso terapêutico , Adulto , Idoso , Idoso de 80 Anos ou mais , Biomarcadores Farmacológicos/metabolismo , Biomarcadores Tumorais/metabolismo , Biomarcadores Tumorais/fisiologia , Carcinoma Epitelial do Ovário , Estudos de Coortes , Resistencia a Medicamentos Antineoplásicos/fisiologia , Feminino , Humanos , Pessoa de Meia-Idade , Neoplasias Epiteliais e Glandulares/metabolismo , Neoplasias Epiteliais e Glandulares/mortalidade , Coativador 3 de Receptor Nuclear/metabolismo , Neoplasias Ovarianas/metabolismo , Neoplasias Ovarianas/mortalidade , Compostos de Platina/administração & dosagem , Prognóstico , Estudos Retrospectivos , Taxoides/administração & dosagem
7.
Am J Pathol ; 180(4): 1474-84, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22342158

RESUMO

The nuclear receptor coactivator amplified in breast cancer 1 (AIB1/SRC-3) has a well-defined role in steroid and growth factor signaling in cancer and normal epithelial cells. Less is known about its function in stromal cells, although AIB1/SRC-3 is up-regulated in tumor stroma and may, thus, contribute to tumor angiogenesis. Herein, we show that AIB1/SRC-3 depletion from cultured endothelial cells reduces their proliferation and motility in response to growth factors and prevents the formation of intact monolayers with tight junctions and of endothelial tubes. In AIB1/SRC-3(+/-) and (-/-) mice, the angiogenic responses to subcutaneous Matrigel implants was reduced by two-thirds, and exogenously added fibroblast growth factor (FGF) 2 did not overcome this deficiency. Furthermore, AIB1/SRC-3(+/-) and (-/-) mice showed similarly delayed healing of full-thickness excisional skin wounds, indicating that both alleles were required for proper tissue repair. Analysis of this defective wound healing showed reduced recruitment of inflammatory cells and macrophages, cytokine induction, and metalloprotease activity. Skin grafts from animals with different AIB1 genotypes and subsequent wounding of the grafts revealed that the defective healing was attributable to local factors and not to defective bone marrow responses. Indeed, wounds in AIB1(+/-) mice showed reduced expression of FGF10, FGFBP3, FGFR1, FGFR2b, and FGFR3, major local drivers of angiogenesis. We conclude that AIB1/SRC-3 modulates stromal cell responses via cross-talk with the FGF signaling pathway.


Assuntos
Neovascularização Fisiológica/fisiologia , Coativador 3 de Receptor Nuclear/fisiologia , Pele/lesões , Cicatrização/fisiologia , Animais , Células Cultivadas , Colágeno , Combinação de Medicamentos , Fatores de Crescimento de Fibroblastos/fisiologia , Células Endoteliais da Veia Umbilical Humana/citologia , Células Endoteliais da Veia Umbilical Humana/fisiologia , Humanos , Inflamação/fisiopatologia , Laminina , Masculino , Camundongos , Camundongos Knockout , Coativador 3 de Receptor Nuclear/deficiência , Proteoglicanas , Reação em Cadeia da Polimerase em Tempo Real/métodos , Transdução de Sinais/fisiologia , Pele/irrigação sanguínea , Pele/metabolismo , Fenômenos Fisiológicos da Pele , Transplante de Pele/métodos , Células Estromais/fisiologia
8.
Hepatology ; 55(6): 1820-9, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22213475

RESUMO

UNLABELLED: Transcriptional coactivator amplified in breast cancer 1 (AIB1) plays important roles in the progression of several cancers such as prostate cancer, breast cancer, and hepatocellular carcinoma. However, its role in cholangiocarcinoma (CCA), a chemoresistant bile duct carcinoma with a poor prognosis, remains unclear. In this study we found that AIB1 protein was frequently overexpressed in human CCA specimens and CCA cell lines. Down-regulation of AIB1 induced the G2/M arrest and decreased the expression of mitosis-promoting factors including Cyclin A, Cyclin B, and Cdk1 through suppressing the Akt pathway, which resulted in inhibiting CCA cell proliferation. In addition, AIB1 enhanced the chemoresistance of CCA cells at least in part through up-regulating the expression of antiapoptotic protein Bcl-2. AIB1 regulated the expression of Bcl-2 in CCA cells through activating the Akt pathway as well as suppressing intracellular reactive oxygen species (ROS). AIB1 suppressed ROS by up-regulating antioxidants such as glutathione synthetase and glutathione peroxidase, which are targets of the NF-E2-related factor 2 (Nrf2), a critical transcription factor that regulates antioxidants, detoxification enzymes, and drug efflux proteins. AIB1 also increased the expression of another two Nrf2 targets, ABCC2 and ABCG2, to enhance drug efflux. AIB1 served as an essential coactivator for Nrf2 activation by physically interacting with Nrf2 to enhance its transcriptional activity. CONCLUSION: AIB1 plays an important role in proliferation and chemoresistance of CCA through simultaneous activation of Akt and Nrf2 pathways, suggesting that AIB1 is a potential molecular target for CCA treatment.


Assuntos
Neoplasias dos Ductos Biliares/patologia , Ductos Biliares Intra-Hepáticos , Colangiocarcinoma/patologia , Resistencia a Medicamentos Antineoplásicos , Fator 2 Relacionado a NF-E2/fisiologia , Coativador 3 de Receptor Nuclear/fisiologia , Proteínas Proto-Oncogênicas c-akt/fisiologia , Membro 2 da Subfamília G de Transportadores de Cassetes de Ligação de ATP , Transportadores de Cassetes de Ligação de ATP/análise , Neoplasias dos Ductos Biliares/tratamento farmacológico , Pontos de Checagem do Ciclo Celular , Linhagem Celular Tumoral , Colangiocarcinoma/tratamento farmacológico , Humanos , Proteína 2 Associada à Farmacorresistência Múltipla , Proteínas Associadas à Resistência a Múltiplos Medicamentos/análise , Proteínas de Neoplasias/análise , Proteínas Proto-Oncogênicas c-bcl-2/análise , Espécies Reativas de Oxigênio/metabolismo , Transdução de Sinais , Ativação Transcricional
9.
BMC Cancer ; 13: 570, 2013 Dec 04.
Artigo em Inglês | MEDLINE | ID: mdl-24304549

RESUMO

BACKGROUND: The placenta-specific 1 (PLAC1) gene encodes a membrane-associated protein which is selectively expressed in the placental syncytiotrophoblast and in murine fetal tissues during embryonic development. In contrast to its transcriptional repression in all other adult normal tissues, PLAC1 is frequently activated and highly expressed in a variety of human cancers, in particular breast cancer, where it associates with estrogen receptor α (ERα) positivity. In a previous study, we showed that ERα-signaling in breast cancer cells transactivates PLAC1 expression in a non-classical pathway. As the members of the p160/nuclear receptor co-activator (NCOA) family, NCOA1, NCOA2 and NCOA3 are known to be overexpressed in breast cancer and essentially involved in estrogen-mediated cancer cell proliferation we asked if these proteins are involved in the ERα-mediated transactivation of PLAC1 in breast cancer cells. METHODS: Applying quantitative real-time RT-PCR (qRT-PCR), Western Blot analysis and chromatin immunoprecipitation, we analyzed the involvement of NCOA1, NCOA2, NCOA3 in the ERα-mediated transactivation of PLAC1 in the breast cancer cell lines MCF-7 and SK-BR-3. RNAi-mediated silencing of NCOA3, qRT-PCR, Western blot analysis and ERα activation assays were used to examine the role of NCOA3 in the ERα-mediated regulation of PLAC1 in further detail. Transcript expression of NCOA3 and PLAC1 in 48 human breast cancer samples was examined by qRT-PCR and statistical analysis was performed using Student's t-test. RESULTS: We detected selective recruitment of NCOA3 but not NCOA1 or NCOA2 to the PLAC1 promoter only in ERα-positive MCF-7 cells but not in ERα-negative SK-BR-3 breast cancer cells. In addition, we demonstrate that silencing of NCOA3 results in a remarkable decrease of PLAC1 expression levels in MCF-7 cells which cannot be restored by treatment with estradiol (E2). Moreover, significant higher transcript levels of PLAC1 were found only in ERα-positive human breast cancer samples which also show a NCOA3 overexpression. CONCLUSIONS: In this study, we identified NCOA3 as a selective co-activator of ERα-mediated transactivation of PLAC1 in MCF-7 breast cancer cells. Our data introduce PLAC1 as novel target gene of NCOA3 in breast cancer, supporting the important role of both factors in breast cancer biology.


Assuntos
Receptor alfa de Estrogênio/fisiologia , Coativador 3 de Receptor Nuclear/fisiologia , Proteínas da Gravidez/genética , Neoplasias da Mama , Estradiol/fisiologia , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Células MCF-7 , Coativador 1 de Receptor Nuclear/fisiologia , Coativador 2 de Receptor Nuclear/fisiologia , Proteínas da Gravidez/metabolismo , Regiões Promotoras Genéticas , Ligação Proteica , Transcrição Gênica , Ativação Transcricional
10.
Endocrinology ; 162(3)2021 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-33340403

RESUMO

Multicellular organisms have evolved sophisticated mechanisms to recover and maintain original tissue functions following injury. Injury responses require a robust transcriptomic response associated with cellular reprogramming involving complex gene expression programs critical for effective tissue repair following injury. Steroid receptor coactivators (SRCs) are master transcriptional regulators of cell-cell signaling that is integral for embryogenesis, reproduction, normal physiological function, and tissue repair following injury. Effective therapeutic approaches for facilitating improved tissue regeneration and repair will likely involve temporal and combinatorial manipulation of cell-intrinsic and cell-extrinsic factors. Pleiotropic actions of SRCs that are critical for wound healing range from immune regulation and angiogenesis to maintenance of metabolic regulation in diverse organ systems. Recent evidence derived from studies of model organisms during different developmental stages indicates the importance of the interplay of immune cells and stromal cells to wound healing. With SRCs being the master regulators of cell-cell signaling integral to physiologic changes necessary for wound repair, it is becoming clear that therapeutic targeting of SRCs provides a unique opportunity for drug development in wound healing. This review will provide an overview of wound healing-related functions of SRCs with a special focus on cellular and molecular interactions important for limiting tissue damage after injury. Finally, we review recent findings showing stimulation of SRCs following cardiac injury with the SRC small molecule stimulator MCB-613 can promote cardiac protection and inhibit pathologic remodeling after myocardial infarction.


Assuntos
Coativadores de Receptor Nuclear/fisiologia , Cicatrização/genética , Animais , Regulação da Expressão Gênica , Humanos , Família Multigênica/fisiologia , Neovascularização Fisiológica/genética , Coativador 1 de Receptor Nuclear/fisiologia , Coativador 2 de Receptor Nuclear/fisiologia , Coativador 3 de Receptor Nuclear/fisiologia , Transdução de Sinais/genética
11.
Oxid Med Cell Longev ; 2021: 6372430, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34777690

RESUMO

This study investigated the effects of chronic intermittent hypoxia (CIH), a model of sleep apnea syndrome (SAS), on cardiac function. SRC-3 was extremely lowly expressed in the adult mouse heart tissue, while SRC-3 was highly expressed in the adult mouse heart tissue after CIH, suggesting that SRC-3 is involved in CIH model. We further studied the role of SRC-3 in CIH-induced myocardial injury in mice. Twenty-four healthy Balb/c male mice (n = 16, wild type; n = 8, SRC-3 knockout (SRC3-KO)) were randomly divided into three groups: air control (Ctrl), CIH, and CIH+SRC3-KO. Mice were exposed to CIH for 12 weeks. qRT-PCR was used to evaluate cardiac expression of the following genes: 11HSD1, 11HSD2, GR, MR, COX-2, OPN, NOX2, HIF-1-α, IL-1ß, IL-6, iNOS, TNF-α, PC-1, and TGF-ß. Enzymatic levels of SOD, CAT, MDA, NOS, and NO in the mouse hearts were determined using commercially available kits. Immunohistochemistry (IHC) was used to evaluate NF-κB expression in cardiac tissues. A transmission electron microscope (TEM) was used to evaluate myocardial ultrastructure. TUNEL staining was used to assess myocardial cell apoptosis. CIH induced cardiac damage, which was ameliorated in the SRC-3 KO mice. CIH significantly increased the heart-to-body weight ratio, expression of all aforementioned genes except 11HSD1, GR, and MR, and increased the levels of MDA, NOS, NO, and NF-κB, which were attenuated in the SRC-3 KO mice. The CIH group had the lowest SOD and CAT levels, which were partially recovered in the CIH+SRC3-KO group. 11HSD2 gene expression was elevated in both the CIH and CIH+SRC3-KO groups compared to the Ctrl group. The CIH group had severe myocardial cell apoptosis and mitochondrial dysfunction, which were alleviated in the CIH+SRC3-KO group. CIH causes cardiac damage through inducing oxidative stress and inflammation. Knockout of SRC-3 ameliorates CIH-induced cardiac damage through antagonizing CIH-triggered molecular changes in cardiac tissue.


Assuntos
Modelos Animais de Doenças , Traumatismos Cardíacos/prevenção & controle , Hipóxia/complicações , Miocárdio/metabolismo , Coativador 3 de Receptor Nuclear/fisiologia , Estresse Oxidativo , Animais , Apoptose , Traumatismos Cardíacos/etiologia , Traumatismos Cardíacos/metabolismo , Traumatismos Cardíacos/patologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout , Miocárdio/patologia
12.
Ann Oncol ; 21(2): 238-244, 2010 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19628566

RESUMO

BACKGROUND: Clinical trials implicate the estrogen receptor (ER) coactivator amplified in breast cancer 1 (AIB1) to be a prognostic and a treatment-predictive factor, although results are not unanimous. We have further investigated this using a controlled randomised trial of tamoxifen versus control. MATERIALS AND METHODS: A total of 564 premenopausal women were entered into a randomised study independent of ER status. Using a tissue microarray, AIB1 and ER were analysed by immunohistochemistry. RESULTS: AIB1 scores were obtained from 349 women. High AIB1 correlated to factors of worse prognosis (human epidermal growth factor receptor 2, Nottingham histological grade 3, and lymph node metastases) and to ER negativity. In the control arm, high AIB1 was a negative prognostic factor for recurrence-free survival (RFS) (P = 0.02). However, ER-positive patients with high AIB1 responded significantly to tamoxifen treatment (P = 0.002), increasing RFS to the same level as for systemically untreated patients with low AIB1. Although ER-positive patients with low AIB1 had a better RFS from the beginning, this was not further improved by tamoxifen (P = 0.8). CONCLUSIONS: In the control group, high AIB1 was a negative prognostic factor. However, ER-positive patients with high AIB1 responded significantly to tamoxifen. This implicates high AIB1 to be an independent predictive factor of improved response to tamoxifen and not, as has previously been discussed, a factor predicting tamoxifen resistance.


Assuntos
Neoplasias da Mama/diagnóstico , Neoplasias da Mama/tratamento farmacológico , Coativador 3 de Receptor Nuclear/fisiologia , Pré-Menopausa/metabolismo , Tamoxifeno/uso terapêutico , Adulto , Algoritmos , Antineoplásicos/uso terapêutico , Biomarcadores Farmacológicos/metabolismo , Neoplasias da Mama/metabolismo , Neoplasias da Mama/mortalidade , Feminino , Seguimentos , Humanos , Pessoa de Meia-Idade , Coativador 3 de Receptor Nuclear/metabolismo , Pré-Menopausa/efeitos dos fármacos , Prognóstico , Análise de Sobrevida
13.
Mol Cell Biochem ; 333(1-2): 221-32, 2010 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19652917

RESUMO

The T:G mismatch specific DNA glycosylase (TDG) is known as an important enzyme in repairing damaged DNA. Recent studies also showed that TDG interacts with a p160 protein, steroid receptor coactivator 1 or nuclear receptor coactivator 1 (SRC1), and is involved in transcriptional activation of the estrogen receptor. However, whether other members of the p160 family are also involved in TDG-interaction and signal transduction regulation remains to be seen. In this study, we employed the mammalian two-hybrid system to investigate the interaction between TDG and another member of the p160 family, nuclear receptor coactivator 3 (NCoA-3). We found that a DXXD motif from aa 294-297 within TDG was responsible for the TDG-NCoA-3 interaction, we also found that a LLXXXL motif (X means any amino acid) from aa 1029-1037 (LLRNSL) and a merged LLXXL motif (LLDQLHTLL) from aa 1053-1061 in NCoA-3 were important for the TDG-NCoA-3 interactions. Mutation of the two aspartic acids (aa 294 and 297) into two alanines in TDG significantly affected the interaction and subsequent transcriptional activation of several steroid hormone receptors including, estrogen-, androgen- and progesterone- receptors in Huh7 cells. We also identified that mutations of NCoA-3 at either leucines 1029-1030 or 1053-1054 (replaced by alanines) also reduced the interaction activity between TDG and NCoA1. These data indicated that the TDG-NCoA-3 interaction is important for broad range activation of steroid hormone nuclear receptors, and may also contribute significantly to further understanding of TDG-related nuclear receptor regulation.


Assuntos
Coativador 3 de Receptor Nuclear/metabolismo , Receptores Citoplasmáticos e Nucleares/genética , Timina DNA Glicosilase/metabolismo , Ativação Transcricional , Motivos de Aminoácidos , Linhagem Celular Tumoral , Humanos , Mutagênese Sítio-Dirigida , Coativador 3 de Receptor Nuclear/fisiologia , Mapeamento de Interação de Proteínas , Timina DNA Glicosilase/fisiologia , Técnicas do Sistema de Duplo-Híbrido
14.
Endocrinology ; 160(8): 1811-1820, 2019 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-31157867

RESUMO

Cancer recurrence and metastasis involves many biological interactions, such as genetic, transcription, environmental, endocrine signaling, and metabolism. These interactions add a complex understanding of cancer recurrence and metastatic progression, delaying the advancement in therapeutic opportunities. We highlight the recent advances on the molecular complexities of endocrine-related cancers, focusing on breast and prostate cancer, and briefly review how endocrine signaling and metabolic programs can influence transcriptional complexes for metastasis competence. Nuclear receptors and transcriptional coregulators function as molecular nodes for the crosstalk between endocrine signaling and metabolism that alter downstream gene expression important for tumor progression and metastasis. This exciting regulatory axis may provide insights to the development of cancer therapeutics important for these desensitized endocrine-dependent cancers.


Assuntos
Neoplasias da Mama/tratamento farmacológico , Neoplasias da Próstata/tratamento farmacológico , Antineoplásicos Hormonais/uso terapêutico , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Castração , Resistencia a Medicamentos Antineoplásicos , Feminino , Humanos , Masculino , Metástase Neoplásica , Recidiva Local de Neoplasia , Coativador 3 de Receptor Nuclear/fisiologia , Neoplasias da Próstata/metabolismo , Neoplasias da Próstata/patologia , Receptores Citoplasmáticos e Nucleares/fisiologia
15.
Sci Rep ; 9(1): 10349, 2019 07 17.
Artigo em Inglês | MEDLINE | ID: mdl-31316078

RESUMO

Preeclampsia (PE) is characterized by poor placentation, consequent on aberrant extravillous trophoblast (EVT) cell function during placental development. The SRC family of proteins is important during pregnancy, especially SRC-3, which regulates placental morphogenesis and embryo survival. Although SRC-3 expression in mouse trophoblast giant cells has been documented, its role in the functional regulation of extravillous trophoblasts and the development of PE remains unknown. This study found that SRC-3 expression was significantly lower in placentas from PE pregnancies as compared to uncomplicated pregnancies. Additionally, both CoCl2-mimicked hypoxia and suppression of endogenous SRC-3 expression by lentivirus short hairpin RNA attenuated the migration and invasion abilities of HTR-8/SVneo cells. Moreover, we demonstrated that SRC-3 physically interacts with AKT to regulate the migration and invasion of HTR-8 cells, via the AKT/mTOR pathway. We also found that the inhibition of HTR-8 cell migration and invasion by CoCl2-mimicked hypoxia was through the SRC-3/AKT/mTOR axis. Our findings indicate that, in early gestation, accumulation of HIF-1α inhibits the expression of SRC-3, which impairs extravillous trophoblastic invasion and migration by directly interacting with AKT. This potentially leads to insufficient uterine spiral artery remodeling and placental hypoperfusion, and thus the development of PE.


Assuntos
Hipóxia/fisiopatologia , Coativador 3 de Receptor Nuclear/fisiologia , Placenta/metabolismo , Pré-Eclâmpsia/etiologia , Proteínas Proto-Oncogênicas c-akt/fisiologia , Transdução de Sinais/fisiologia , Serina-Treonina Quinases TOR/fisiologia , Trofoblastos/fisiologia , Acetatos/farmacologia , Adulto , Benzopiranos/farmacologia , Linhagem Celular , Movimento Celular , Regulação para Baixo , Feminino , Humanos , Hipóxia/genética , Subunidade alfa do Fator 1 Induzível por Hipóxia/fisiologia , Coativador 3 de Receptor Nuclear/biossíntese , Coativador 3 de Receptor Nuclear/genética , Pré-Eclâmpsia/genética , Pré-Eclâmpsia/fisiopatologia , Gravidez , Interferência de RNA , RNA Interferente Pequeno/genética , Artéria Uterina/fisiopatologia , Remodelação Vascular
16.
Eur Rev Med Pharmacol Sci ; 21(24): 5610-5617, 2017 12.
Artigo em Inglês | MEDLINE | ID: mdl-29271993

RESUMO

OBJECTIVE: To explore the mechanism of amplified in breast cancer 1 (AIB1) to promote ovarian cancer progress. MATERIALS AND METHODS: Cor correlation analysis was performed to obtain the top 100 lncRNAs that were positively correlated with AIB1. The relationship of taurine upregulated gene 1 (TUG1) and clinicopathological characteristics. Moreover, Gene Ontology (GO) and Gene Set Enrichment Analysis (GSEA) were performed to predict the biological process where TUG1 may be involved in. At last, Cell Counting Kit-8 (CCK-8), colon formation and flow cytometry were conducted to explore the biological process that TUG1 may influence. Meanwhile, Western blot was performed to explore the mechanism of TUG1. RESULTS: In this study, it was found that P73 antisense RNA 1T (TP73-AS1), LINC00654 and TUG1 had the tumor-promoting effect in the top 100 lncRNAs that were positively correlated with AIB1. The expression level of TUG1 was significantly decreased after intervention of AIB1. Then, the clinical data were analyzed and the results showed that TUG1 was related to the tumor residue, tumor staging, tumor grade and lymph node metastasis. Moreover, the bioinformatics analysis revealed that TUG1 was mainly involved in the regulation of cell cycle. After intervention in TUG1, it was found that the cell proliferation capacity was significantly decreased, and the cell cycle was arrested in G1 phase. Finally, Western blot revealed that the expressions of G1 phase-related proteins were significantly changed. This study indicated that AIB1 regulates the cycle of ovarian cancer cells through TUG1. CONCLUSIONS: This study proved that AIB1 can regulate the cell cycle through regulating TUG1.


Assuntos
Coativador 3 de Receptor Nuclear/fisiologia , Neoplasias Ovarianas/patologia , RNA Longo não Codificante/fisiologia , Ciclo Celular , Linhagem Celular Tumoral , Proliferação de Células/genética , Feminino , Humanos , Estadiamento de Neoplasias
17.
Cancer Cell ; 31(3): 436-451, 2017 03 13.
Artigo em Inglês | MEDLINE | ID: mdl-28292441

RESUMO

Recurrent point mutations in SPOP define a distinct molecular subclass of prostate cancer. Here, we describe a mouse model showing that mutant SPOP drives prostate tumorigenesis in vivo. Conditional expression of mutant SPOP in the prostate dramatically altered phenotypes in the setting of Pten loss, with early neoplastic lesions (high-grade prostatic intraepithelial neoplasia) with striking nuclear atypia and invasive, poorly differentiated carcinoma. In mouse prostate organoids, mutant SPOP drove increased proliferation and a transcriptional signature consistent with human prostate cancer. Using these models and human prostate cancer samples, we show that SPOP mutation activates both PI3K/mTOR and androgen receptor signaling, effectively uncoupling the normal negative feedback between these two pathways.


Assuntos
Mutação , Proteínas Nucleares/genética , Fosfatidilinositol 3-Quinases/fisiologia , Neoplasias da Próstata/etiologia , Receptores Androgênicos/fisiologia , Proteínas Repressoras/genética , Transdução de Sinais/fisiologia , Serina-Treonina Quinases TOR/fisiologia , Animais , Proliferação de Células , Humanos , Masculino , Camundongos , Coativador 3 de Receptor Nuclear/fisiologia , PTEN Fosfo-Hidrolase/genética , Neoplasias da Próstata/genética , Neoplasias da Próstata/patologia , Proteínas Proto-Oncogênicas c-ets/fisiologia
18.
Oncogene ; 34(30): 3935-3945, 2015 Jul 23.
Artigo em Inglês | MEDLINE | ID: mdl-25263446

RESUMO

Aberrant activation of Notch signaling has an essential role in colorectal cancer (CRC) progression. Amplified in breast cancer 1 (AIB1), also known as steroid receptor coactivator 3 or NCOA3, is a transcriptional coactivator that promotes cancer cell proliferation and invasiveness. However, AIB1 implication in CRC progression through enhancing Notch signaling is unknown. In this study, we found that several CRC cell lines expressed high levels of AIB1, and knockdown of AIB1 decreased cell proliferation, colony formation and tumorigenesis of these CRC cells. Specifically, knockdown of AIB1 inhibited cell cycle progression at G1 phase by decreasing the mRNA levels of cyclin A2, cyclin B1, cyclin E2 and hairy and enhancer of split (Hes) 1. Furthermore, AIB1 interacted with Notch intracellular domain and Mastermind-like 1 and was recruited to the Hes1 promoter to enhance Notch signaling. Downregulation of AIB1 also decreased CRC cell invasiveness in vitro and lung metastasis in vivo. Besides that, knockout of AIB1 in mice inhibited colon carcinogenesis induced by azoxymethane/dextran sodium sulfate treatment. The mRNA levels of cyclin B1 and Hes5 were downregulated, but p27, ATOH1 and MUC2 were upregulated in the colon tumors from AIB1-deficient mice compared with those from wild-type mice. Thus, our results signify the importance of AIB1 in CRC and demonstrate that AIB1 promotes CRC progression at least in part through enhancing Notch signaling, suggesting that AIB1 is a potential molecular target for CRC treatment.


Assuntos
Neoplasias Colorretais/metabolismo , Neoplasias Pulmonares/metabolismo , Coativador 3 de Receptor Nuclear/fisiologia , Receptores Notch/metabolismo , Pontos de Checagem do Ciclo Celular , Linhagem Celular Tumoral , Proliferação de Células , Sobrevivência Celular , Neoplasias Colorretais/patologia , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Progressão da Doença , Técnicas de Silenciamento de Genes , Células HEK293 , Humanos , Neoplasias Pulmonares/secundário , Transplante de Neoplasias , Ligação Proteica , Transdução de Sinais , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo
19.
Oncogene ; 34(37): 4879-89, 2015 Sep 10.
Artigo em Inglês | MEDLINE | ID: mdl-25531332

RESUMO

Pancreatic cancer (PC) is characterized by aberrant overexpression of mucins that contribute to its pathogenesis. Although the inflammatory cytokines contribute to mucin overexpression, the mucin profile of PC is markedly distinct from that of normal or inflamed pancreas. We postulated that de novo expression of various mucins in PC involves chromatin modifications. Analysis of chromatin modifying enzymes by PCR array identified differential expression of NCOA3 in MUC4-expressing PC cell lines. Immunohistochemistry analysis in tumor tissues from patients and spontaneous mouse models, and microarray analysis following the knockdown of NCOA3 were performed to elucidate its role in mucin regulation and overall impact on PC. Silencing of NCOA3 in PC cell lines resulted in significant downregulation of two most differentially expressed mucins in PC, MUC4 and MUC1 (P<0.01). Immunohistochemistry analysis in PC tissues and metastatic lesions established an association between NCOA3 and mucin (MUC1 and MUC4) expression. Spontaneous mouse model of PC (K-ras(G12D); Pdx-1cre) showed early expression of Ncoa3 during pre-neoplastic lesions. Mechanistically, NCOA3 knockdown abrogated retinoic acid-mediated MUC4 upregulation by restricting MUC4 promoter accessibility as demonstrated by micrococcus nuclease digestion (P<0.05) and chromatin immuno-precipitation analysis. NCOA3 also created pro-inflammatory conditions by upregulating chemokines like CXCL1, 2, 5 and CCL20 (P<0.001). AKT, ubiquitin C, ERK1/2 and NF-κB occupied dominant nodes in the networks significantly modulated after NCOA3 silencing. In addition, NCOA3 stabilized mucins post translationally through fucosylation by FUT8, as the knockdown of FUT8 resulted in the downregulation of MUC4 and MUC1 at protein levels.


Assuntos
Mucina-1 , Mucina-4 , Coativador 3 de Receptor Nuclear/fisiologia , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/metabolismo , Processamento de Proteína Pós-Traducional/genética , Animais , Transformação Celular Neoplásica/genética , Fucose/metabolismo , Fucosiltransferases/metabolismo , Perfilação da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Humanos , Camundongos , Análise em Microsséries , Mucina-1/genética , Mucina-1/metabolismo , Mucina-4/genética , Mucina-4/metabolismo , Neoplasias Pancreáticas/patologia , Ativação Transcricional , Células Tumorais Cultivadas , Regulação para Cima/genética
20.
Cancer Res ; 74(19): 5631-43, 2014 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-25274033

RESUMO

Somatic missense mutations in the substrate-binding pocket of the E3 ubiquitin ligase adaptor SPOP are present in up to 15% of human prostate adenocarcinomas, but are rare in other malignancies, suggesting a prostate-specific mechanism of action. SPOP promotes ubiquitination and degradation of several protein substrates, including the androgen receptor (AR) coactivator SRC-3. However, the relative contributions that SPOP substrates may make to the pathophysiology of SPOP-mutant (mt) prostate adenocarcinomas are unknown. Using an unbiased bioinformatics approach, we determined that the gene expression profile of prostate adenocarcinoma cells engineered to express mt-SPOP overlaps greatly with the gene signature of both SRC-3 and AR transcriptional output, with a stronger similarity to AR than SRC-3. This finding suggests that in addition to its SRC-3-mediated effects, SPOP also exerts SRC-3-independent effects that are AR-mediated. Indeed, we found that wild-type (wt) but not prostate adenocarcinoma-associated mutants of SPOP promoted AR ubiquitination and degradation, acting directly through a SPOP-binding motif in the hinge region of AR. In support of these results, tumor xenografts composed of prostate adenocarcinoma cells expressing mt-SPOP exhibited higher AR protein levels and grew faster than tumors composed of prostate adenocarcinoma cells expressing wt-SPOP. Furthermore, genetic ablation of SPOP was sufficient to increase AR protein levels in mouse prostate. Examination of public human prostate adenocarcinoma datasets confirmed a strong link between transcriptomic profiles of mt-SPOP and AR. Overall, our studies highlight the AR axis as the key transcriptional output of SPOP in prostate adenocarcinoma and provide an explanation for the prostate-specific tumor suppressor role of wt-SPOP.


Assuntos
Adenocarcinoma/fisiopatologia , Genes Supressores de Tumor , Proteínas Nucleares/genética , Neoplasias da Próstata/fisiopatologia , Receptores Androgênicos/fisiologia , Proteínas Repressoras/genética , Transcrição Gênica/fisiologia , Adenocarcinoma/genética , Androgênios/fisiologia , Perfilação da Expressão Gênica , Humanos , Masculino , Mutação , Coativador 3 de Receptor Nuclear/fisiologia , Neoplasias da Próstata/genética
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa