Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Cells ; 10(7)2021 07 03.
Article in English | MEDLINE | ID: mdl-34359849

ABSTRACT

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.


Subject(s)
Autophagy , Carcinoma, Non-Small-Cell Lung/pathology , Erlotinib Hydrochloride/pharmacology , Lung Neoplasms/pathology , Phosphofructokinase-2/antagonists & inhibitors , Adenylate Kinase/metabolism , Autophagosomes/drug effects , Autophagosomes/metabolism , Autophagy/drug effects , Carcinoma, Non-Small-Cell Lung/metabolism , Cell Death/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Chloroquine/pharmacology , Enzyme Activation/drug effects , ErbB Receptors/metabolism , Green Fluorescent Proteins/metabolism , Humans , Lung Neoplasms/metabolism , Microtubule-Associated Proteins/metabolism , Mutant Proteins/metabolism , Phosphofructokinase-2/metabolism , Protein Kinase Inhibitors/pharmacology , Sequestosome-1 Protein/metabolism
2.
J Neurotrauma ; 38(9): 1306-1326, 2021 05 01.
Article in English | MEDLINE | ID: mdl-33499736

ABSTRACT

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.


Subject(s)
Disease Models, Animal , Spinal Cord Injuries/physiopathology , Thoracic Vertebrae/injuries , Urinary Tract/physiopathology , Urodynamics/physiology , Animals , Female , Spinal Cord Injuries/pathology , Swine , Swine, Miniature , Urinary Bladder/innervation , Urinary Bladder/pathology , Urinary Bladder/physiopathology , Urinary Tract/pathology
3.
PLoS One ; 11(11): e0166363, 2016.
Article in English | MEDLINE | ID: mdl-27832204

ABSTRACT

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.


Subject(s)
Adenocarcinoma/genetics , Gene Expression Regulation, Neoplastic , Lung Neoplasms/genetics , Lung/pathology , RNA, Long Noncoding/genetics , Retinoblastoma Protein/metabolism , Signal Transduction , A549 Cells , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Adenocarcinoma of Lung , Animals , Cell Line , Cell Line, Tumor , Cell Proliferation , DNA (Cytosine-5-)-Methyltransferase 1 , DNA (Cytosine-5-)-Methyltransferases/genetics , DNA (Cytosine-5-)-Methyltransferases/metabolism , Gene Knockout Techniques , Humans , Lung/metabolism , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Mice , Retinoblastoma Protein/genetics
4.
Mol Cancer Ther ; 9(3): 594-605, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20197399

ABSTRACT

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.


Subject(s)
Anacardic Acids/pharmacology , Breast Neoplasms/pathology , Cell Proliferation/drug effects , DNA/metabolism , Estrogen Receptor alpha/antagonists & inhibitors , Transcription, Genetic/drug effects , Antineoplastic Agents, Phytogenic/pharmacology , Apoptosis/drug effects , Apoptosis/genetics , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , DNA/drug effects , Down-Regulation/drug effects , Down-Regulation/genetics , Estrogen Receptor alpha/metabolism , Female , Gene Expression Regulation, Neoplastic/drug effects , Genes, Reporter/drug effects , Humans , Protein Binding/drug effects , Transcriptional Activation/drug effects , Transfection , Tumor Cells, Cultured
5.
Am J Respir Cell Mol Biol ; 42(3): 320-30, 2010 Mar.
Article in English | MEDLINE | ID: mdl-19556604

ABSTRACT

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.


Subject(s)
Adenocarcinoma/metabolism , Estrogen Receptor alpha/metabolism , Estrogen Receptor beta/metabolism , Lung Neoplasms/metabolism , Sex Characteristics , Adenocarcinoma/enzymology , Adenocarcinoma/pathology , Cell Line, Tumor , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Cyclin D1/metabolism , Enzyme Activation/drug effects , Estradiol/analogs & derivatives , Estradiol/pharmacology , Female , Fluorescence , Fulvestrant , Genome, Human/genetics , Humans , Lung Neoplasms/enzymology , Lung Neoplasms/pathology , Male , Microscopy, Confocal , Mitogen-Activated Protein Kinases/metabolism , Mutant Proteins/metabolism , Phosphoserine/metabolism , Protein Transport/drug effects , Signal Transduction/drug effects , Subcellular Fractions/drug effects , Subcellular Fractions/metabolism , Tamoxifen/analogs & derivatives , Tamoxifen/pharmacology
6.
Mol Cell Endocrinol ; 305(1-2): 12-21, 2009 Jun 16.
Article in English | MEDLINE | ID: mdl-19433257

ABSTRACT

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.


Subject(s)
Adenocarcinoma/metabolism , Bronchi/metabolism , Estrogen Receptor alpha/metabolism , Estrogen Receptor beta/metabolism , Lung Neoplasms/metabolism , Adenocarcinoma/pathology , Bronchi/cytology , Bronchi/drug effects , Cell Nucleus/metabolism , Cells, Cultured , Cytoplasm/metabolism , Epithelial Cells/cytology , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Estradiol/pharmacology , Estrogen Receptor alpha/analysis , Estrogen Receptor beta/analysis , Female , Gene Expression Regulation, Neoplastic , Genes, Reporter , Humans , Hydroxytestosterones/pharmacology , Lung Neoplasms/pathology , Male , Mitochondria/metabolism , Sex Factors
7.
Nucleic Acids Res ; 37(8): 2584-95, 2009 May.
Article in English | MEDLINE | ID: mdl-19264808

ABSTRACT

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.


Subject(s)
Breast Neoplasms/genetics , Estradiol/pharmacology , Gene Expression Regulation, Neoplastic , MicroRNAs/metabolism , 3' Untranslated Regions/chemistry , Apoptosis Regulatory Proteins/genetics , Breast Neoplasms/metabolism , Cell Line, Tumor , Cycloheximide/pharmacology , Dactinomycin/pharmacology , Down-Regulation , Estradiol/analogs & derivatives , Estrogen Antagonists/pharmacology , Estrogen Receptor alpha/agonists , Estrogen Receptor alpha/antagonists & inhibitors , Estrogen Receptor alpha/genetics , Estrogen Receptor beta/agonists , Estrogen Receptor beta/antagonists & inhibitors , Estrogen Receptor beta/genetics , Female , Fulvestrant , Genes, Reporter , Humans , MicroRNAs/antagonists & inhibitors , MicroRNAs/genetics , Nucleic Acid Synthesis Inhibitors/pharmacology , Promoter Regions, Genetic , Protein Synthesis Inhibitors/pharmacology , RNA, Antisense/metabolism , RNA-Binding Proteins/genetics , Tamoxifen/analogs & derivatives , Tamoxifen/pharmacology , ras GTPase-Activating Proteins/genetics
8.
FASEB J ; 22(7): 2185-97, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18296501

ABSTRACT

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.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Caveolin 1/physiology , Endothelium, Vascular/physiology , Estrogen Receptor alpha/physiology , Stilbenes/pharmacology , Umbilical Veins/physiology , src-Family Kinases/physiology , Caveolin 1/drug effects , Endothelium, Vascular/drug effects , Estradiol/pharmacology , Estrogen Receptor alpha/drug effects , Ethanol/pharmacology , Humans , Kinetics , Phosphorylation , Resveratrol , Umbilical Veins/drug effects , src-Family Kinases/drug effects
9.
Toxicol Lett ; 174(1-3): 61-73, 2007 Nov 01.
Article in English | MEDLINE | ID: mdl-17920790

ABSTRACT

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.


Subject(s)
Endothelial Cells/drug effects , Particulate Matter/toxicity , Vehicle Emissions/toxicity , Cell Line , Endothelial Cells/metabolism , Estradiol/toxicity , Humans , Nitric Oxide/metabolism , Nitric Oxide Synthase Type III/metabolism , Particulate Matter/chemistry , Polycyclic Aromatic Hydrocarbons/analysis , Polycyclic Aromatic Hydrocarbons/toxicity , Protein Serine-Threonine Kinases/metabolism , Signal Transduction/drug effects
10.
Am J Physiol Cell Physiol ; 293(2): C566-73, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17360813

ABSTRACT

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.


Subject(s)
Capillary Permeability , Dihydrotestosterone/metabolism , Endothelial Cells/metabolism , Estradiol/metabolism , Hypergravity , Mitogen-Activated Protein Kinases/metabolism , Tight Junctions/metabolism , Capillary Permeability/drug effects , Cells, Cultured , Dihydrotestosterone/pharmacology , Electric Impedance , Electron Transport Complex IV/metabolism , Endothelial Cells/drug effects , Endothelial Cells/enzymology , Enzyme Activation , Estradiol/pharmacology , Female , Humans , Hypotension, Orthostatic/metabolism , Male , Membrane Proteins/metabolism , Occludin , Phosphorylation , Prostaglandin-Endoperoxide Synthases/metabolism , Sex Factors , Space Flight , Tight Junctions/drug effects , Time Factors , Tyrosine/metabolism
11.
Endocr Relat Cancer ; 13(1): 113-34, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16601283

ABSTRACT

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.


Subject(s)
Adenocarcinoma/metabolism , Estrogen Receptor alpha/metabolism , Estrogen Receptor beta/metabolism , Lung Neoplasms/metabolism , Adenocarcinoma/pathology , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Bronchi/cytology , Bronchi/drug effects , Bronchi/metabolism , Cadherins/metabolism , Cell Proliferation/drug effects , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Estradiol/pharmacology , Estrogen Receptor Modulators/pharmacology , Fibroblasts/cytology , Fibroblasts/drug effects , Fibroblasts/metabolism , Humans , Lung/cytology , Lung/drug effects , Lung/metabolism , Lung Neoplasms/pathology , Mediator Complex Subunit 1 , Peroxisome Proliferator-Activated Receptors/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, Progesterone/metabolism , Response Elements , Sex , Transcription Factors/metabolism , Transcription, Genetic , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...