Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Cells ; 10(7)2021 07 03.
Artigo em Inglês | MEDLINE | ID: mdl-34359849

RESUMO

Tyrosine kinase inhibitors (TKIs) targeting the kinase domain of the epidermal growth factor receptor (EGFR), such as erlotinib, have dramatically improved clinical outcomes of patients with EGFR-driven non-small cell lung carcinomas (NSCLCs). However, intrinsic or acquired resistance remains a clinical barrier to the success of FDA-approved EGFR TKIs. Multiple mechanisms of resistance have been identified, including the activation of prosurvival autophagy. We have previously shown that the expression and activity of PFKFB3-a known driver of glycolysis-is associated with resistance to erlotinib and that PFKFB3 inhibition improves the response of NSCLC cells to erlotinib. This study focuses on investigating the role of PFKFB3 in regulating erlotinib-driven autophagy to escape resistance to erlotinib. We evaluated the consequence of pharmacological inhibition of PFKFB3 on erlotinib-driven autophagy in NSCLC cells with different mutation statuses. Here, we identify PFKFB3 as a mediator of erlotinib-induced autophagy in NSCLCs. We demonstrate that PFKFB3 inhibition sensitizes NCSLCs to erlotinib via impairing autophagy flux. In summary, our studies uncovered a novel crosstalk between PFKFB3 and EGFR that regulates erlotinib-induced autophagy, thus contributing to erlotinib sensitivity in NSCLCs.


Assuntos
Autofagia , Carcinoma Pulmonar de Células não Pequenas/patologia , Cloridrato de Erlotinib/farmacologia , Neoplasias Pulmonares/patologia , Fosfofrutoquinase-2/antagonistas & inibidores , Adenilato Quinase/metabolismo , Autofagossomos/efeitos dos fármacos , Autofagossomos/metabolismo , Autofagia/efeitos dos fármacos , Carcinoma Pulmonar de Células não Pequenas/metabolismo , Morte Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Cloroquina/farmacologia , Ativação Enzimática/efeitos dos fármacos , Receptores ErbB/metabolismo , Proteínas de Fluorescência Verde/metabolismo , Humanos , Neoplasias Pulmonares/metabolismo , Proteínas Associadas aos Microtúbulos/metabolismo , Proteínas Mutantes/metabolismo , Fosfofrutoquinase-2/metabolismo , Inibidores de Proteínas Quinases/farmacologia , Proteína Sequestossoma-1/metabolismo
2.
J Neurotrauma ; 38(9): 1306-1326, 2021 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-33499736

RESUMO

There is an increasing need to develop approaches that will not only improve the clinical management of neurogenic lower urinary tract dysfunction (NLUTD) after spinal cord injury (SCI), but also advance therapeutic interventions aimed at recovering bladder function. Although pre-clinical research frequently employs rodent SCI models, large animals such as the pig may play an important translational role in facilitating the development of devices or treatments. Therefore, the objective of this study was to develop a urodynamics protocol to characterize NLUTD in a porcine model of SCI. An iterative process to develop the protocol to perform urodynamics in female Yucatan minipigs began with a group of spinally intact, anesthetized pigs. Subsequently, urodynamic studies were performed in a group of awake, lightly restrained pigs, before and after a contusion-compression SCI at the T2 or T9-T11 spinal cord level. Bladder tissue was obtained for histological analysis at the end of the study. All anesthetized pigs had bladders that were acontractile, which resulted in overflow incontinence once capacity was reached. Uninjured, conscious pigs demonstrated appropriate relaxation and contraction of the external urethral sphincter during the voiding phase. SCI pigs demonstrated neurogenic detrusor overactivity and a significantly elevated post-void residual volume. Relative to the control, SCI bladders were heavier and thicker. The developed urodynamics protocol allows for repetitive evaluation of lower urinary tract function in pigs at different time points post-SCI. This technique manifests the potential for using the pig as an intermediary, large animal model for translational studies in NLUTD.


Assuntos
Modelos Animais de Doenças , Traumatismos da Medula Espinal/fisiopatologia , Vértebras Torácicas/lesões , Sistema Urinário/fisiopatologia , Urodinâmica/fisiologia , Animais , Feminino , Traumatismos da Medula Espinal/patologia , Suínos , Porco Miniatura , Bexiga Urinária/inervação , Bexiga Urinária/patologia , Bexiga Urinária/fisiopatologia , Sistema Urinário/patologia
3.
PLoS One ; 11(11): e0166363, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27832204

RESUMO

Maternally expressed gene 3 (MEG3, mouse homolog Gtl2) encodes a long noncoding RNA (lncRNA) that is expressed in many normal tissues, but is suppressed in various cancer cell lines and tumors, suggesting it plays a functional role as a tumor suppressor. Hypermethylation has been shown to contribute to this loss of expression. We now demonstrate that MEG3 expression is regulated by the retinoblastoma protein (Rb) pathway and correlates with a change in cell proliferation. Microarray analysis of mouse embryonic fibroblasts (MEFs) isolated from mice with genetic deletion of all three Rb family members (TKO) revealed a significant silencing of Gtl2/MEG3 expression compared to WT MEFs, and re-expression of Gtl2/MEG3 caused decrease in cell proliferation and increased apoptosis. MEG3 levels also were suppressed in A549 lung cancer cells compared with normal human bronchial epithelial (NHBE) cells, and, similar to the TKO cells, re-constitution of MEG3 led to a decrease in cell proliferation and elevated apoptosis. Activation of pRb by treatment of A549 and SK-MES-1 cells with palbociclib, a CDK4/6 inhibitor, increased the expression of MEG3 in a dose-dependent manner, while knockdown of pRb/p107 attenuated this effect. In addition, expression of phosphorylation-deficient mutant of pRb increased MEG3 levels in both lung cancer cell types. Treatment of these cells with palbociclib also decreased the expression of pRb-regulated DNA methyltransferase 1 (DNMT1), while conversely, knockdown of DNMT1 resulted in increased expression of MEG3. As gene methylation has been suggested for MEG3 regulation, we found that palbociclib resulted in decreased methylation of the MEG3 locus similar to that observed with 5-aza-deoxycytidine. Anti-sense oligonucleotide silencing of drug-induced MEG3 expression in A549 and SK-MES-1 cells partially rescued the palbociclib-mediated decrease in cell proliferation, while analysis of the TCGA database revealed decreased MEG3 expression in human lung tumors harboring a disrupted RB pathway. Together, these data suggest that disruption of the pRb-DNMT1 pathway leads to a decrease in MEG3 expression, thereby contributing to the pro-proliferative state of certain cancer cells.


Assuntos
Adenocarcinoma/genética , Regulação Neoplásica da Expressão Gênica , Neoplasias Pulmonares/genética , Pulmão/patologia , RNA Longo não Codificante/genética , Proteína do Retinoblastoma/metabolismo , Transdução de Sinais , Células A549 , Adenocarcinoma/metabolismo , Adenocarcinoma/patologia , Adenocarcinoma de Pulmão , Animais , Linhagem Celular , Linhagem Celular Tumoral , Proliferação de Células , DNA (Citosina-5-)-Metiltransferase 1 , DNA (Citosina-5-)-Metiltransferases/genética , DNA (Citosina-5-)-Metiltransferases/metabolismo , Técnicas de Inativação de Genes , Humanos , Pulmão/metabolismo , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patologia , Camundongos , Proteína do Retinoblastoma/genética
4.
Mol Cancer Ther ; 9(3): 594-605, 2010 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-20197399

RESUMO

Anacardic acid (AnAc; 2-hydroxy-6-alkylbenzoic acid) is a dietary and medicinal phytochemical with established anticancer activity in cell and animal models. The mechanisms by which AnAc inhibits cancer cell proliferation remain undefined. AnAc 24:1(omega5) was purified from geranium (Pelargonium x hortorum) and shown to inhibit the proliferation of estrogen receptor alpha (ERalpha)-positive MCF-7 and endocrine-resistant LCC9 and LY2 breast cancer cells with greater efficacy than ERalpha-negative primary human breast epithelial cells, MCF-10A normal breast epithelial cells, and MDA-MB-231 basal-like breast cancer cells. AnAc 24:1(omega5) inhibited cell cycle progression and induced apoptosis in a cell-specific manner. AnAc 24:1(omega5) inhibited estradiol (E(2))-induced estrogen response element (ERE) reporter activity and transcription of the endogenous E(2) target genes pS2, cyclin D1, and cathepsin D in MCF-7 cells. AnAc 24:1(omega5) did not compete with E(2) for ERalpha or ERbeta binding, nor did AnAc 24:1(omega5) reduce ERalpha or ERbeta steady-state protein levels in MCF-7 cells; rather, AnAc 24:1(omega5) inhibited ER-ERE binding in vitro. Virtual screening with the molecular docking software Surflex evaluated AnAc 24:1(omega5) interaction with ERalpha ligand binding (LBD) and DNA binding (DBD) domains in conjunction with experimental validation. Molecular modeling revealed AnAc 24:1(omega5) interaction with the ERalpha DBD but not the LBD. Chromatin immunoprecipitation experiments revealed that AnAc 24:1(omega5) inhibited E(2)-ERalpha interaction with the endogenous pS2 gene promoter region containing an ERE. These data indicate that AnAc 24:1(omega5) inhibits cell proliferation, cell cycle progression, and apoptosis in an ER-dependent manner by reducing ER-DNA interaction and inhibiting ER-mediated transcriptional responses.


Assuntos
Ácidos Anacárdicos/farmacologia , Neoplasias da Mama/patologia , Proliferação de Células/efeitos dos fármacos , DNA/metabolismo , Receptor alfa de Estrogênio/antagonistas & inibidores , Transcrição Gênica/efeitos dos fármacos , Antineoplásicos Fitogênicos/farmacologia , Apoptose/efeitos dos fármacos , Apoptose/genética , Neoplasias da Mama/genética , Neoplasias da Mama/metabolismo , DNA/efeitos dos fármacos , Regulação para Baixo/efeitos dos fármacos , Regulação para Baixo/genética , Receptor alfa de Estrogênio/metabolismo , Feminino , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Genes Reporter/efeitos dos fármacos , Humanos , Ligação Proteica/efeitos dos fármacos , Ativação Transcricional/efeitos dos fármacos , Transfecção , Células Tumorais Cultivadas
5.
Am J Respir Cell Mol Biol ; 42(3): 320-30, 2010 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-19556604

RESUMO

The role of estrogens in the increased risk of lung adenocarcinoma in women remains uncertain. We reported that lung adenocarcinoma cell lines from female, but not male, patients with non-small cell lung cancer respond proliferatively and transcriptionally to estradiol (E(2)), despite equal protein expression of estrogen receptors (ER) alpha and beta. To test the hypothesis that nuclear localization of ER alpha corresponds to genomic E(2) activity in lung adenocarcinoma cells from females, cell fractionation, immunoblot, and confocal immunohistochemical microscopy were performed. We report for the first time that E(2) increases phospho-serine-118-ER alpha (P-ser118-ER alpha) and cyclin D1 (CCND1) nuclear colocalization in H1793, but not A549 lung adenocarcinoma cells, derived from a female and male patient, respectively. ER beta was primarily in the cytoplasm and mitochondria, independent of E(2) treatment, and showed no difference between H1793 and A549 cells. E(2) induced higher transcription of endogenous ER alpha-regulated CCND1 in H1793 than in A549 cells. Likewise, higher rapid, non-genomic E(2)-induced extracellular signal-regulated kinase 1/2 activation was detected in H1793 compared with A549 cells, linking extracellular signal-regulated kinase activation to increased P-ser118-ER alpha. Furthermore, E(2) increased cyclin D1 and P-ser118-ER alpha nuclear localization in H1793, but not A549 cells. Together, our results indicate that nuclear localization of P-ser118-ER alpha provides one explanation for sex-dependent differences in E(2)-genomic responses in lung adenocarcinoma cell lines.


Assuntos
Adenocarcinoma/metabolismo , Receptor alfa de Estrogênio/metabolismo , Receptor beta de Estrogênio/metabolismo , Neoplasias Pulmonares/metabolismo , Caracteres Sexuais , Adenocarcinoma/enzimologia , Adenocarcinoma/patologia , Linhagem Celular Tumoral , Núcleo Celular/efeitos dos fármacos , Núcleo Celular/metabolismo , Ciclina D1/metabolismo , Ativação Enzimática/efeitos dos fármacos , Estradiol/análogos & derivados , Estradiol/farmacologia , Feminino , Fluorescência , Fulvestranto , Genoma Humano/genética , Humanos , Neoplasias Pulmonares/enzimologia , Neoplasias Pulmonares/patologia , Masculino , Microscopia Confocal , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Proteínas Mutantes/metabolismo , Fosfosserina/metabolismo , Transporte Proteico/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Frações Subcelulares/efeitos dos fármacos , Frações Subcelulares/metabolismo , Tamoxifeno/análogos & derivados , Tamoxifeno/farmacologia
6.
Mol Cell Endocrinol ; 305(1-2): 12-21, 2009 Jun 16.
Artigo em Inglês | MEDLINE | ID: mdl-19433257

RESUMO

Gender differences in lung disease and cancer are well-established. We reported estrogenic transcriptional responses in lung adenocarcinoma cells from females but not males despite similar estrogen receptor (ER) expression. Here we tested the hypothesis that normal human bronchial epithelial cells (HBECs) show gender-independent estrogenic responses. We report that a small sample of HBECs express approximately twice as much ERbeta as ERalpha. ERalpha and ERbeta were located in the cytoplasm, nucleus, and mitochondria. In contrast to lung adenocarcinoma cells, estradiol (E2) induced estrogen response element (ERE)-mediated luciferase reporter activity in transiently transfected HBECs regardless of donor gender. Overexpression of ERalpha-VP16 increased ERE-mediated transcriptional activity in all HBECs. E2 increased and 4-hydroxytamoxifen and ICI 182,780 inhibited HBEC proliferation and cyclin D1 expression in a cell line-specific manner. In conclusion, the response of HBECs to ER ligands is gender-independent suggesting that estrogenic sensitivity may be acquired during lung carcinogenesis.


Assuntos
Adenocarcinoma/metabolismo , Brônquios/metabolismo , Receptor alfa de Estrogênio/metabolismo , Receptor beta de Estrogênio/metabolismo , Neoplasias Pulmonares/metabolismo , Adenocarcinoma/patologia , Brônquios/citologia , Brônquios/efeitos dos fármacos , Núcleo Celular/metabolismo , Células Cultivadas , Citoplasma/metabolismo , Células Epiteliais/citologia , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/metabolismo , Estradiol/farmacologia , Receptor alfa de Estrogênio/análise , Receptor beta de Estrogênio/análise , Feminino , Regulação Neoplásica da Expressão Gênica , Genes Reporter , Humanos , Hidroxitestosteronas/farmacologia , Neoplasias Pulmonares/patologia , Masculino , Mitocôndrias/metabolismo , Fatores Sexuais
7.
Nucleic Acids Res ; 37(8): 2584-95, 2009 May.
Artigo em Inglês | MEDLINE | ID: mdl-19264808

RESUMO

Select changes in microRNA (miRNA) expression correlate with estrogen receptor alpha (ER alpha) expression in breast tumors. miR-21 is higher in ER alpha positive than negative tumors, but no one has examined how estradiol (E(2)) regulates miR-21 in breast cancer cells. Here we report that E(2) inhibits miR-21 expression in MCF-7 human breast cancer cells. The E(2)-induced reduction in miR-21 was inhibited by 4-hydroxytamoxifen (4-OHT), ICI 182 780 (Faslodex), and siRNA ER alpha indicating that the suppression is ER alpha-mediated. ER alpha and ER beta agonists PPT and DPN inhibited and 4-OHT increased miR-21 expression. E(2) increased luciferase activity from reporters containing the miR-21 recognition elements from the 3'-UTRs of miR-21 target genes, corroborating that E(2) represses miR-21 expression resulting in a loss of target gene suppression. The E(2)-mediated decrease in miR-21 correlated with increased protein expression of endogenous miR-21-targets Pdcd4, PTEN and Bcl-2. siRNA knockdown of ER alpha blocked the E(2)-induced increase in Pdcd4, PTEN and Bcl-2. Transfection of MCF-7 cells with antisense (AS) to miR-21 mimicked the E(2)-induced increase in Pdcd4, PTEN and Bcl-2. These results are the first to demonstrate that E(2) represses the expression of an oncogenic miRNA, miR-21, by activating estrogen receptor in MCF-7 cells.


Assuntos
Neoplasias da Mama/genética , Estradiol/farmacologia , Regulação Neoplásica da Expressão Gênica , MicroRNAs/metabolismo , Regiões 3' não Traduzidas/química , Proteínas Reguladoras de Apoptose/genética , Neoplasias da Mama/metabolismo , Linhagem Celular Tumoral , Cicloeximida/farmacologia , Dactinomicina/farmacologia , Regulação para Baixo , Estradiol/análogos & derivados , Antagonistas de Estrogênios/farmacologia , Receptor alfa de Estrogênio/agonistas , Receptor alfa de Estrogênio/antagonistas & inibidores , Receptor alfa de Estrogênio/genética , Receptor beta de Estrogênio/agonistas , Receptor beta de Estrogênio/antagonistas & inibidores , Receptor beta de Estrogênio/genética , Feminino , Fulvestranto , Genes Reporter , Humanos , MicroRNAs/antagonistas & inibidores , MicroRNAs/genética , Inibidores da Síntese de Ácido Nucleico/farmacologia , Regiões Promotoras Genéticas , Inibidores da Síntese de Proteínas/farmacologia , RNA Antissenso/metabolismo , Proteínas de Ligação a RNA/genética , Tamoxifeno/análogos & derivados , Tamoxifeno/farmacologia , Proteínas Ativadoras de ras GTPase/genética
8.
FASEB J ; 22(7): 2185-97, 2008 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-18296501

RESUMO

Epidemiological studies correlate moderate red wine consumption to reduced incidence of cardiovascular disease. Resveratrol is a polyphenolic compound in red wine that has cardioprotective effects in rodents. Although endothelial cell (EC) studies indicate that micromolar resveratrol has diverse biological activities, these concentrations are not physiologically relevant because human oral ingestion provides only brief exposure to nanomolar plasma levels. Previously, we reported that nanomolar resveratrol activated ERK1/2 signaling in bovine aortic ECs (BAECs). The goal of this study was to determine the mechanisms by which nanomolar resveratrol rapidly activates endothelial nitric oxide synthase (eNOS) in human umbilical vein ECs (HUVECs). We report for the first time that resveratrol increased interaction between estrogen receptor alpha (ER alpha), caveolin-1 (Cav-1) and c-Src, and increased phosphorylation of Cav-1, c-Src, and eNOS. Pretreatment with the lipid raft disruptor beta-methyl cyclodextrin or G alpha inhibitor pertussis toxin blocked resveratrol- and E(2)-induced eNOS activation and NO production. Depletion of endogenous ER alpha, not ERbeta, by siRNA attenuated resveratrol- and E(2)-induced ERK1/2, Src, and eNOS phosphorylation. Our data demonstrate that nanomolar resveratrol induces ER alpha-Cav-1-c-SRC interaction, resulting in NO production through a G alpha-protein-coupled mechanism. This study provides important new insights into mechanisms for the beneficial effects of resveratrol in ECs.


Assuntos
Inibidores da Angiogênese/farmacologia , Caveolina 1/fisiologia , Endotélio Vascular/fisiologia , Receptor alfa de Estrogênio/fisiologia , Estilbenos/farmacologia , Veias Umbilicais/fisiologia , Quinases da Família src/fisiologia , Caveolina 1/efeitos dos fármacos , Endotélio Vascular/efeitos dos fármacos , Estradiol/farmacologia , Receptor alfa de Estrogênio/efeitos dos fármacos , Etanol/farmacologia , Humanos , Cinética , Fosforilação , Resveratrol , Veias Umbilicais/efeitos dos fármacos , Quinases da Família src/efeitos dos fármacos
9.
Toxicol Lett ; 174(1-3): 61-73, 2007 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-17920790

RESUMO

Inhalation of ultrafine particulate matter (PM) in air pollution increases cardiovascular mortality by passing into systemic circulation and possibly affecting endothelial cell (EC) function. This study identified the chemical constituents, including polycyclic aromatic hydrocarbons (PAHs), in diesel exhaust particulate extracts (DEPEs) prepared from a truck run at different speeds and engine loads. The short-term effects of DEPEs alone or in combination with estradiol (E(2)) on MAPK (ERK1/2), AKT, and eNOS activation and nitric oxide (NO) production in human umbilical vein EC (HUVEC) were evaluated. Notably, DEPE from a truck run under increasing loads (L) stimulated phosphorylation of MAPK, AKT, and eNOS whereas DEPE from the truck run at increasing speeds (S) did not affect MAPK alone, but inhibited E(2)-induced MAPK and eNOS phosphorylation. Higher PAH concentrations in the DEPE L versus DEPE S samples correlate with the observed differences in cellular activities. Like E(2), DEPEs rapidly increased NO with the DEPE L sample acting additively with E(2) and then inhibiting E(2)-induced NO with longer treatment time. Like E(2), DEPEs increased trans-endothelial electrical resistance (TEER) across a monolayer of HUVEC. These data are the first characterization of rapid effects of DEPE in human EC and may indicate mechanisms for diesel exhaust in vascular function.


Assuntos
Células Endoteliais/efeitos dos fármacos , Material Particulado/toxicidade , Emissões de Veículos/toxicidade , Linhagem Celular , Células Endoteliais/metabolismo , Estradiol/toxicidade , Humanos , Óxido Nítrico/metabolismo , Óxido Nítrico Sintase Tipo III/metabolismo , Material Particulado/química , Hidrocarbonetos Policíclicos Aromáticos/análise , Hidrocarbonetos Policíclicos Aromáticos/toxicidade , Proteínas Serina-Treonina Quinases/metabolismo , Transdução de Sinais/efeitos dos fármacos
10.
Am J Physiol Cell Physiol ; 293(2): C566-73, 2007 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-17360813

RESUMO

Postflight orthostatic intolerance (POI) was reported to be higher in female than male astronauts and may result from sex-dependent differences in endothelial cell (EC) barrier permeability. Here the effect of 17beta-estradiol (E(2)) and dihydrotestosterone (DHT) on the expression of the tight junction protein occludin, EC barrier function, and MAPK activation over time was tested after subjecting human umbilical vein EC (HUVEC) to brief hypergravity identical to that experienced by astronauts during liftoff (LO) into space. After LO hypergravity, HUVEC showed a time-dependent decrease in occludin correlating with an increase in paracellular permeability and a decrease in transendothelial electrical resistance, indicating a decrease in EC barrier function. LO hypergravity inhibited MAPK activation, which remained suppressed 4 h after LO. Inhibition of MAPK activation correlated with decreased phosphotyrosine occludin, decreased cytochrome-c oxidase activity, and increased paracellular permeability, suggesting a mechanism by which LO hypergravity decreased EC barrier function. Time-dependent differences in MAPK activation, decreased occludin, and EC barrier function between HUVEC treated with E(2) vs. DHT were observed. HUVEC showed delayed activation of MAPK with DHT, i.e., 4 h rather than 2 h for E(2), which correlated with decreased paracellular permeability and the observed sex differences in POI in astronauts. These data temporally separate E(2) and DHT effects in HUVEC and provide evidence for the possible protective roles of sex steroids on EC function after brief exposure to low hypergravity.


Assuntos
Permeabilidade Capilar , Di-Hidrotestosterona/metabolismo , Células Endoteliais/metabolismo , Estradiol/metabolismo , Hipergravidade , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Junções Íntimas/metabolismo , Permeabilidade Capilar/efeitos dos fármacos , Células Cultivadas , Di-Hidrotestosterona/farmacologia , Impedância Elétrica , Complexo IV da Cadeia de Transporte de Elétrons/metabolismo , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/enzimologia , Ativação Enzimática , Estradiol/farmacologia , Feminino , Humanos , Hipotensão Ortostática/metabolismo , Masculino , Proteínas de Membrana/metabolismo , Ocludina , Fosforilação , Prostaglandina-Endoperóxido Sintases/metabolismo , Fatores Sexuais , Voo Espacial , Junções Íntimas/efeitos dos fármacos , Fatores de Tempo , Tirosina/metabolismo
11.
Endocr Relat Cancer ; 13(1): 113-34, 2006 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-16601283

RESUMO

The higher frequency of lung adenocarcinoma in women smokers than in men smokers suggests a role for gender-dependent factors in the etiology of lung cancer. We evaluated estrogen receptor (ER) alpha and beta expression and activity in human lung adenocarcinoma cell lines and normal lung fibroblasts. Full-length ERalpha and ERbeta proteins were expressed in all cell lines with higher ERbeta than ERalpha. Although estradiol (E(2)) binding was similar, E(2) stimulated proliferation only in cells from females, and this response was inhibited by anti-estrogens 4-hydroxytamoxifen (4-OHT) and ICI 182,780. In contrast, E(2) did not stimulate replication of lung adenocarcinoma cells from males and 4-OHT or ICI did not block cell proliferation. Similarly, transcription of an estrogen response element-driven reporter gene was stimulated by E(2) in lung adenocarcinoma cells from females, but not males. Progesterone receptor (PR) expression was increased by E(2) in two out of five adenocarcinoma cell lines from females, but none from males. E(2) decreased E-cadherin protein expression in some of the cell lines from females, as it did in MCF-7 breast cancer cells, but not in the cell lines from males. Thus, ERalpha and ERbeta expression does not correlate with the effect of ER ligands on cellular activities in lung adenocarcinoma cells. On the other hand, coactivator DRIP205 expression was higher in lung adenocarcinoma cells from females versus males and higher in adenocarcinoma cells than in normal human bronchial epithelial cells. DRIP205 and other ER coregulators may contribute to differences in estrogen responsiveness between lung adenocarcinoma cells in females and males.


Assuntos
Adenocarcinoma/metabolismo , Receptor alfa de Estrogênio/metabolismo , Receptor beta de Estrogênio/metabolismo , Neoplasias Pulmonares/metabolismo , Adenocarcinoma/patologia , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Brônquios/citologia , Brônquios/efeitos dos fármacos , Brônquios/metabolismo , Caderinas/metabolismo , Proliferação de Células/efeitos dos fármacos , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/metabolismo , Estradiol/farmacologia , Moduladores de Receptor Estrogênico/farmacologia , Fibroblastos/citologia , Fibroblastos/efeitos dos fármacos , Fibroblastos/metabolismo , Humanos , Pulmão/citologia , Pulmão/efeitos dos fármacos , Pulmão/metabolismo , Neoplasias Pulmonares/patologia , Subunidade 1 do Complexo Mediador , Receptores Ativados por Proliferador de Peroxissomo/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Receptores de Progesterona/metabolismo , Elementos de Resposta , Sexo , Fatores de Transcrição/metabolismo , Transcrição Gênica , Células Tumorais Cultivadas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...