Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
J Steroid Biochem Mol Biol ; 118(4-5): 219-30, 2010 Feb 28.
Article in English | MEDLINE | ID: mdl-19815064

ABSTRACT

As breast cancer cells develop secondary resistance to estrogen deprivation therapy, they increase their utilization of non-genomic signaling pathways. Our prior work demonstrated that estradiol causes an association of ERalpha with Shc, Src and the IGF-1-R. In cells developing resistance to estrogen deprivation (surrogate for aromatase inhibition) and to the anti-estrogens tamoxifen, 4-OH-tamoxifen, and fulvestrant, an increased association of ERalpha with c-Src and the EGF-R occurs. At the same time, there is a translocation of ERalpha out of the nucleus and into the cytoplasm and cell membrane. Blockade of c-Src with the Src kinase inhibitor, PP-2 causes relocation of ERalpha into the nucleus. While these changes are not identical in response to each anti-estrogen, ERalpha binding to the EGF-R is increased in response to 4-OH-tamoxifen when compared with tamoxifen. The changes in EGF-R interactions with ERalpha impart an enhanced sensitivity of tamoxifen-resistant cells to the inhibitory properties of the specific EGF-R tyrosine kinase inhibitor, AG 1478. However, with long term exposure of tamoxifen-resistant cells to AG 1478, the cells begin to re-grow but can now be inhibited by the IGF-R tyrosine kinase inhibitor, AG 1024. These data suggest that the IGF-R system becomes the predominant signaling mechanism as an adaptive response to the EGF-R inhibitor. Taken together, this information suggests that both the EGF-R and IGF-R pathways can mediate ERalpha signaling. To further examine the effects of fulvestrant on ERalpha function, we examined the acute effects of fulvestrant, on non-genomic functionality. Fulvestrant enhanced ERalpha association with the membrane IGF-1-receptor (IGF-1-R). Using siRNA or expression vectors to knock-down or knock-in selective proteins, we further demonstrated that the ERalpha/IGF-1-R association is Src-dependent. Fulvestrant rapidly induced IGF-1-R and MAPK phosphorylation. The Src inhibitor PP2 and IGF-1-R inhibitor AG1024 greatly blocked fulvestrant-induced ERalpha/IGF-1-R interaction leading to a further depletion of total cellular ERalpha induced by fulvestrant and further enhanced fulvestrant-induced cell growth arrest. More dramatic was the translocation of ERalpha to the plasma membrane in combination with the IGF-1-R as shown by confocal microscopy. Taken in aggregate, these studies suggest that secondary resistance to hormonal therapy results in usage of both IGF-R and EGF-R for non-genomic signaling.


Subject(s)
Breast Neoplasms/metabolism , Drug Resistance, Neoplasm/drug effects , ErbB Receptors/metabolism , Estrogens/metabolism , Receptor, IGF Type 1/metabolism , Signal Transduction/drug effects , Antineoplastic Agents, Hormonal/pharmacology , Apoptosis , Breast Neoplasms/drug therapy , Cell Line, Tumor , Estradiol/analogs & derivatives , Estradiol/pharmacology , Estrogen Receptor alpha/genetics , Estrogen Receptor alpha/metabolism , Female , Fulvestrant , Gene Expression Regulation, Neoplastic/drug effects , Humans , Mitogen-Activated Protein Kinases/metabolism
2.
Steroids ; 74(7): 586-94, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19138696

ABSTRACT

Activation of IGF-1R can activate metalloproteinases which release heparin-binding EGF (Hb-EGF) and lead to EGFR-dependent MAPK activation in certain tissues. We postulated that this pathway is operative in E(2)-induced MAPK activation in breast cancer tissues. As evidence, we showed that E(2) rapidly induced the phosphorylation of both IGF-1R and EGFR and that siRNA knockdown or selective inhibitors against either growth factor receptor inhibited E(2)-induced MAPK activation. The selective inhibitors or knockdown of either IGF-1R or EGFR significantly inhibited cell growth and reversed cell death protection induced by E(2) in MCF-7 cells. Our data support the conclusion that the IGF-1R acts upstream of EGFR in a linear pathway which mediates E(2) action on MAPK activation, cell growth stimulation and anti-apoptosis in breast cancer cells. During the process of development of tamoxifen resistance this pathway is up-regulated with increased sensitivity to activate EGFR for cell growth and protection against apoptosis. Surprisingly, translocation of ERalpha out of the nucleus into the cytoplasm, mediated by c-Src, occurs during development of resistance. This effect can be abrogated by administration of the c-Src inhibitor, PP2 which also restores sensitivity to tamoxifen.


Subject(s)
Antineoplastic Agents, Hormonal/pharmacology , Breast Neoplasms/drug therapy , Cell Nucleus/metabolism , Drug Resistance, Neoplasm , ErbB Receptors/metabolism , Estrogens/metabolism , Receptor, IGF Type 1/metabolism , Tamoxifen/pharmacology , Apoptosis , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Estrogen Receptor alpha/metabolism , Humans , Signal Transduction
3.
Cancer Res ; 67(12): 5717-26, 2007 Jun 15.
Article in English | MEDLINE | ID: mdl-17575138

ABSTRACT

Secondary resistance to hormonal therapy for breast cancer commonly develops after an initial response to tamoxifen or aromatase inhibitors. Agents to abrogate these adaptive changes would substantially enhance the long-term benefits of hormonal therapy. Our studies with a stilbene derivative called TMS (2,3',4,5'-tetramethoxystilbene) identified unexpected effects with potential utility for treatment of breast tumors secondarily resistant to hormonal therapy. TMS was originally developed as an inhibitor of cytochrome P450 1B1 to block the conversion of estradiol to 4-OH-estradiol. While studying this agent in three models of hormone resistance, we detected direct antitumor effects not related to its role as an inhibitor of catecholestrogens. During examination of the mechanisms involved, we showed that treatment with 3 micromol/L TMS for 24 h inhibited tubulin polymerization and microtubule formation, caused a cell cycle block at the G2-M phase, and induced apoptosis. TMS also inhibited activated focal adhesion kinase (FAK), Akt, and mammalian target of rapamycin (mTOR) and stimulated c-jun-NH2-kinase and p38 mitogen-activated protein kinase activity. With respect to antitumor effects, TMS at a concentrations of 0.2 to 0.3 micromol/L inhibited the growth of long-term tamoxifen-treated MCF-7 cells by 80% and fulvestrant-treated MCF-7 cells by 70%. In vivo studies, involving 8 weeks of treatment with TMS via a 30-mg s.c. implant, reduced tumor volume of tamoxifen-resistant MCF-7 breast cancer xenografts by 53%. Our data suggest that TMS is a promising therapeutic agent because of its unique ability to block several pathways involved in the development of hormone resistance.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Cell Proliferation/drug effects , Drug Resistance, Neoplasm/physiology , Mammary Neoplasms, Experimental/drug therapy , Stilbenes/pharmacology , Animals , Aromatase Inhibitors/pharmacology , Cell Cycle/drug effects , Cell Line, Tumor , Enzyme-Linked Immunosorbent Assay , Estrogen Receptor Modulators/pharmacology , Estrogens/metabolism , Female , Fluorescent Antibody Technique , Humans , Immunoblotting , In Situ Nick-End Labeling , Mammary Neoplasms, Experimental/metabolism , Mice , Tamoxifen/pharmacology
4.
Endocrinology ; 148(8): 4091-101, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17525128

ABSTRACT

We present an integrated model of an extranuclear, estrogen receptor-alpha (ERalpha)-mediated, rapid MAPK activation pathway in breast cancer cells. In noncancer cells, IGF-I initiates a linear process involving activation of the IGF-I receptor (IGF-IR) and matrix metalloproteinases (MMP), release of heparin-binding epidermal growth factor (HB-EGF), and activation of EGF receptor (EGFR)-dependent MAPK. 17beta-Estradiol (E2) rapidly activates IGF-IR in breast cancer cells. We hypothesize that E2 induces a similar linear pathway involving IGF-IR, MMP, HB-EGF, EGFR, and MAPK. Using MCF-7 breast cancer cells, we for the first time demonstrated that a sequential activation of IGF-IR, MMP, and EGFR existed in E2 and IGF-I actions, which was supported by evidence that the selective inhibitors of IGF-IR and MMP or knockdown of IGF-IR all inhibited E2- or IGF-I-induced EGFR phosphorylation. Using the inhibitors and small inhibitory RNA strategies, we also demonstrated that the same sequential activation of the receptors occurred in E2-, IGF-I-, but not EGF-induced MAPK phosphorylation. Additionally, a HB-EGF neutralizing antibody significantly blocked E2-induced MAPK activation, further supporting our hypothesis. The biological effects of sequential activation of IGF-IR and EGFR on E2 stimulation of cell proliferation were also investigated. Knockdown or blockade of IGF-IR significantly inhibited E2- or IGF-I-stimulated but not EGF-induced cell growth. Knockdown or blockade of EGFR abrogated cell growth induced by E2, IGF-I, and EGF, indicating that EGFR is a downstream molecule of IGF-IR in E2 and IGF-I action. Together, our data support the novel view that E2 can activate a linear pathway involving the sequential activation of IGF-IR, MMP, HB-EGF, EGFR, and MAPK.


Subject(s)
Breast Neoplasms/metabolism , ErbB Receptors/metabolism , Estradiol/metabolism , Matrix Metalloproteinases/metabolism , Mitogen-Activated Protein Kinases/metabolism , Receptor, IGF Type 1/metabolism , Signal Transduction/physiology , Apoptosis/physiology , Breast Neoplasms/pathology , Cell Division/physiology , Cell Line, Tumor , Epidermal Growth Factor/metabolism , Epidermal Growth Factor/pharmacology , Estradiol/pharmacology , Estrogen Receptor alpha/metabolism , Heparin-binding EGF-like Growth Factor , Humans , Insulin-Like Growth Factor I/metabolism , Insulin-Like Growth Factor I/pharmacology , Intercellular Signaling Peptides and Proteins , Signal Transduction/drug effects
5.
Semin Reprod Med ; 25(3): 187-97, 2007 May.
Article in English | MEDLINE | ID: mdl-17447208

ABSTRACT

Although classical concepts had assigned priority to the nuclear-initiated steroid signaling pathway of estrogen receptor (ER), recent studies document that the ER also possesses the membrane-initiated steroid signaling (MISS) pathway. A small fraction of ER is associated with the cell membrane and mediates the rapid effects of estrogen. Unlike classical growth factor receptors, such as insulinlike growth factor 1 receptor and epidermal growth factor receptor, ER has no transmembrane and kinase domains. Instead, the initiating signals of MISS action of ER require a rapid formation of ER-centered protein complexes with many signaling molecules, leading to the activation of mitogen-activated protein kinase and Akt signaling pathways. In this review, we focus on the MISS action of ER and its role in the development of hormone resistance in breast cancer. A full understanding of the mechanisms, with the ultimate aim of abrogating specific steps, should lead to more targeted strategies for treatment of hormone-dependent breast cancer.


Subject(s)
Breast Neoplasms/metabolism , Estrogens/metabolism , Neoplasms, Hormone-Dependent/metabolism , Receptors, Cell Surface/metabolism , Receptors, Estrogen/metabolism , Signal Transduction , Animals , Antineoplastic Agents, Hormonal/therapeutic use , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Proliferation , Drug Resistance, Neoplasm , Female , Humans , Neoplasms, Hormone-Dependent/drug therapy , Neoplasms, Hormone-Dependent/genetics , Neoplasms, Hormone-Dependent/pathology , Transcription, Genetic
6.
J Cell Sci ; 119(Pt 22): 4623-33, 2006 Nov 15.
Article in English | MEDLINE | ID: mdl-17062641

ABSTRACT

Here we address the molecular mechanism of serum-independent survival and growth of human bladder carcinoma cell line 5637. Serum starvation promoted tyrosine phosphorylation of a 145-kDa protein and activation of the tyrosine kinase Src and the receptor for epidermal growth factor (EGFR) over a slow time course (>8 hours). The phosphorylated 145-kDa protein was identified as the beta-subunit of c-Met/hepatocyte growth factor (HGF) receptor, p145(met), in which tyrosine residues 1003, 1234, and 1235 were phosphorylated. Inhibitors of Src (PP2, SU6656) or EGFR (AG99), but not p145(met) (K252a), effectively blocked tyrosine phosphorylation of p145(met) and promoted cell death accompanied by activation of caspase-like proteases. Conditioned medium from the serum-starved 5637 cells or purified EGF readily promoted the activation of Src and EGFR, and tyrosine phosphorylation of p145(met) in normally grown 5637 cells, suggesting that autocrine signaling of EGFR ligands is responsible for signal transduction events in serum-starved cells. Consistent with this idea, a monoclonal antibody against EGFR that would interfere with the ligand binding to EGFR blocked tyrosine phosphorylation events and promoted the caspase activation and cell death in serum-free conditions. Such apoptotic cell death was also induced by pretreatment of cells with a high concentration of HGF that downregulated endogenous p145(met). Nevertheless, Cu2+ ions, competitive inhibitors for HGF-binding to p145(met), did not show any effect on cellular functions in serum-free conditions. These results suggest that the serum-independent growth of 5637 cells involves the transmembrane signaling cascade via EGFR ligand(s) (but not HGF), EGFR, Src and p145(met).


Subject(s)
ErbB Receptors/physiology , Proto-Oncogene Proteins c-met/metabolism , Tyrosine/metabolism , src-Family Kinases/physiology , Amino Acid Sequence , Apoptosis , Carbazoles/pharmacology , Cell Line, Tumor , Cell Survival , Culture Media, Serum-Free , Enzyme Activation , ErbB Receptors/antagonists & inhibitors , Hepatocyte Growth Factor/metabolism , Humans , Indole Alkaloids , Indoles/pharmacology , Molecular Sequence Data , Phosphorylation , Protein Subunits/metabolism , Signal Transduction , Sulfonamides/pharmacology , Tyrphostins/pharmacology , Urinary Bladder Neoplasms , src-Family Kinases/antagonists & inhibitors
7.
Biol Reprod ; 75(1): 9-16, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16571873

ABSTRACT

Recent research has focused on effects of the estrogen receptor acting at the level of the cell membrane in breast cancer. In this review we describe 17beta-estradiol (E2)-initiated membrane signaling pathways involving the activation of several kinases that contribute to the regulation of cell proliferation and prevention of apoptosis. Although classical concepts had assigned priority to the nuclear actions of estrogen receptor, recent studies document the additional importance of estrogen receptor residing in or near the plasma membrane. A small fraction of estrogen receptor is associated with the cell membrane and mediates the rapid effects of E2. Unlike classical growth factor receptors, such as insulin-like growth factor 1 receptor (IGF1R) and epidermal growth factor receptor (EGFR), estrogen receptor has no transmembrane and kinase domains and is known to initiate E2 rapid signals by forming a protein complex with many signaling molecules. The formation of the protein complex is a critical step, leading to the activation of the MAPK1/3 (also known as MAP kinase) and AKT1 (also known as Akt) pathways. A full understanding of the mechanisms underlying these relationships, with the ultimate aim of abrogating specific steps, should lead to more-targeted strategies for treatment of hormone dependent-breast cancer.


Subject(s)
Breast Neoplasms/metabolism , Estradiol/metabolism , Signal Transduction , Animals , CSK Tyrosine-Protein Kinase , ErbB Receptors/metabolism , Estrogen Receptor alpha/metabolism , Humans , Membrane Proteins/metabolism , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Protein-Tyrosine Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Receptors, Somatomedin/metabolism , src-Family Kinases
8.
Endocr Relat Cancer ; 13 Suppl 1: S3-13, 2006 Dec.
Article in English | MEDLINE | ID: mdl-17259556

ABSTRACT

Our recent studies have examined the role of various receptor complexes in the mediation of rapid, extranuclear effects of estradiol. This review describes 17beta-estradiol (E2)-initiated extranuclear signaling pathways, which involve the insulin-like growth factor 1 receptor (IGF-1R) and epidermal growth factor receptor (EGFR) and result in the activation of several kinase cascades. The biologic results of these effects are the enhancement of cell proliferation and diminution of programmed cell death (apoptosis). Until recently, most studies assigned priority to the nuclear transcriptional actions of estrogen receptor alpha (ER alpha). Present investigative emphasis focuses on the additional importance of ER alpha residing in or near the plasma membrane. A small fraction of ER alpha is associated with the cell membrane and mediates the rapid effects of E2. Unlike classical growth factor receptors, such as IGF-1R and EGFR, ER alpha has no transmembrane and kinase domains and is known to initiate E2 rapid signals by forming protein/protein complexes with many signaling molecules. Our recent studies demonstrate that the IGF-1R is involved in tethering ER alpha to the plasma membrane, in activating the EGFR, and in the initiation of mitogen-activated protein kinase and phosphoinositide 3-kinase signaling. The formation of a multi-protein complex containing these receptors as well as adaptor proteins is a critical step in this process. A full understanding of the mechanisms underlying these relationships with the ultimate aim of abrogating specific steps, should lead to more targeted strategies for treatment of hormone-dependent breast cancer.


Subject(s)
Breast Neoplasms/metabolism , Carcinoma/metabolism , ErbB Receptors/physiology , Estrogen Receptor alpha/metabolism , Receptor, IGF Type 1/physiology , Animals , Carrier Proteins/metabolism , Cell Membrane/metabolism , Cytosol/metabolism , Estradiol/metabolism , Humans , MAP Kinase Signaling System/physiology , Models, Biological , Oncogene Protein v-akt/physiology , Signal Transduction
9.
Trends Endocrinol Metab ; 16(8): 347-53, 2005 Oct.
Article in English | MEDLINE | ID: mdl-16126407

ABSTRACT

This review provides insight into biomolecular knowledge regarding the non-genomic actions of estrogen in hormone-dependent breast cancer, particularly its role in the rapid stimulation of pathways that transmit signals to increase cell division or decrease programmed cell death. Until recently, attention to estrogenic effects focused primarily on events in the nucleus, where most estrogen receptors (ERalpha and beta) reside. However, a fraction of ERalpha associated with the cell membrane also participates in rapid estrogen-induced cell membrane-mediated events via formation of a protein complex with many signaling molecules, leading to activation of the mitogen-activated protein kinase and Akt signaling pathways. Understanding the mechanisms underlying these relationships, with the aim of abrogating specific steps, should lead to more targeted strategies to treat hormone-dependent breast cancer.


Subject(s)
Estrogen Receptor alpha/physiology , Estrogens/physiology , src-Family Kinases/physiology , Adaptor Proteins, Signal Transducing/physiology , Animals , Breast Neoplasms/physiopathology , Caveolins/physiology , Co-Repressor Proteins , ErbB Receptors/physiology , Estrogen Receptor beta/physiology , GTP-Binding Protein alpha Subunits, Gi-Go/physiology , Humans , Mitogen-Activated Protein Kinases/metabolism , Phosphatidylinositol 3-Kinases/physiology , Protein Structure, Tertiary , Proto-Oncogene Proteins c-akt/physiology , Receptors, G-Protein-Coupled/physiology , Shc Signaling Adaptor Proteins , Src Homology 2 Domain-Containing, Transforming Protein 1 , Trans-Activators/physiology , Transcription Factors
10.
J Clin Endocrinol Metab ; 90(3): 1407-13, 2005 Mar.
Article in English | MEDLINE | ID: mdl-15572429

ABSTRACT

A recent analysis of data from nine studies provided convincing evidence that plasma estradiol measurements predict the risk of breast cancer in normal postmenopausal women. However, the median values detected by the various assays used in this study varied by 5-fold. These and other published data in normal postmenopausal women suggest that assays measuring low plasma estradiol concentrations suffer from problems of sensitivity, specificity, and precision. Availability of a practical, low-cost, specific, precise, and ultrasensitive estrogen assay might allow enhanced prediction of the risk of breast cancer and provide an objective means of selecting postmenopausal women for breast cancer prevention. A recombinant cell ultrasensitive bioassay (RCUB) for estrogen was recently validated for use in prepubertal children. We postulated that the RCUB might also prove useful for measurement of postmenopausal levels and designed the present study to examine this possibility. Thirty normal postmenopausal volunteers provided blood samples for measurement of estrogen by RCUB and, for comparison, by RIA. The estrogenic activity measured by RCUB [mean +/- sd, 11.9 +/- 10.9 pmol/liter (SI units, 3.23 +/- 2.96 pg/ml] was significantly lower than estradiol levels measured by RIA [43.7 +/- 44.0 pmol/liter (11.9 +/- 12.0 pg/ml)] in our volunteer subjects (P < 0.00001). Nonetheless, plasma estradiol levels measured by bioassay were significantly correlated with the estrogenic activity measured by RIA (r = 0.84) and by gas chromatography/tandem mass spectrometry (r = 0.85). To obtain biological evidence of the validity of the RCUB, we related plasma estrogen levels to body weight and body mass index and found highly significant correlations (r = 0.54 and r = 0.53, respectively). Surprisingly, 28 of 30 postmenopausal women were found to have estrogen levels in the prepubertal range with the RCUB. The levels detected by RCUB were similar to those previously reported using an ultrasensitive but less practical yeast bioassay. These results provide validation for the RCUB in postmenopausal women and suggest that it might prove useful for selection of women for drug therapy to prevent breast cancer.


Subject(s)
Biological Assay/methods , Estrogens/blood , Postmenopause , Aged , Body Mass Index , Breast Neoplasms/blood , Breast Neoplasms/epidemiology , DNA, Recombinant , Evaluation Studies as Topic , Female , Humans , Middle Aged , Predictive Value of Tests , Risk Factors , Sensitivity and Specificity , Yeasts
11.
Steroids ; 69(8-9): 523-9, 2004 Aug.
Article in English | MEDLINE | ID: mdl-15288764

ABSTRACT

Breast cancer is one of the most common malignancies in the United States. Seventy percent of breast cancers are hormone-responsive due to the presence of estrogen receptors ERalpha and ERbeta, which are important diagnostic and therapeutic targets in cancer treatment. Estrogen acts through its receptors, which reside on the cell membrane as demonstrated recently and in the nucleus, leading to cancer cell proliferation and protection from cell death. The membrane ERalpha has been reported in MCF-7 human breast cancer cells and is believed to mediate estrogen effects to activate mitogen-activated protein (MAP) kinase and phosphoinositide 3-kinase (PI3-kinase). Activation of many growth factor receptors require adapter proteins to delivery the upstream signals to downstream kinases, such as MAP kinase. Both Shc and the p85alpha subunit of PI3-kinase are adapter proteins. In addition to their roles in transducing signals from membrane growth factor receptors, they have been demonstrated to interact with ERalpha in an estrogen dependent manner. In this review, the role of Shc in mediating estrogen effects on MAP Kinase regulation, cell growth and anti-apoptosis will be discussed. The possible role of PI3-kinase in estrogen rapid action is also reviewed in brief.


Subject(s)
Adaptor Proteins, Signal Transducing/physiology , Estrogens/physiology , Animals , Apoptosis/physiology , Breast Neoplasms/enzymology , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Division/physiology , Cell Line, Tumor , Cell Membrane/metabolism , Enzyme Activation , Estradiol/physiology , Estrogen Receptor alpha/metabolism , Estrogen Receptor beta/metabolism , Humans , MAP Kinase Signaling System/physiology , Mitogen-Activated Protein Kinases/metabolism , Phosphatidylinositol 3-Kinases/chemistry , Phosphatidylinositol 3-Kinases/metabolism , Shc Signaling Adaptor Proteins , Src Homology 2 Domain-Containing, Transforming Protein 1
12.
Biochem Biophys Res Commun ; 294(5): 926-33, 2002 Jun 28.
Article in English | MEDLINE | ID: mdl-12074565

ABSTRACT

Estrogen rapidly activates MAPK in many cell types but the mechanisms have not been fully understood. We previously demonstrated that 17-beta-estradiol (estradiol) rapidly induced membrane translocation of estrogen receptor alpha (ERalpha) and activated MAPK in MCF-7 breast cancer cells. This study further determines the cause and effect relationship between the presence of membrane ERalpha and MAPK activation. ERalpha with a membrane localization signal (HE241G-mem) was expressed and compared with the ones in nucleus (HEGO) or cytosol (HE241G) localization. Confocal microscopy showed that HE241G-mem was expressed in the cell membrane as well as in the cytosol in COS-1 cells. HE241G localized in the cytosol and HEGO in the nucleus. Functional studies showed that only membrane ERalpha, not cytosol and nuclear ones, responded to estradiol by inducing MAPK phosphorylation. HE241G-mem neither increased basal nor estradiol-induced ERE promoter activation, indicating no transcriptional action involved. Our data support the view that membrane-associated ERalpha is critical in estrogen-initiated MAPK activation.


Subject(s)
Estradiol/pharmacology , Mitogen-Activated Protein Kinases/metabolism , Receptors, Estrogen/metabolism , Animals , COS Cells , Cell Membrane/metabolism , Enzyme Activation , Estrogen Receptor alpha , Phosphorylation , Promoter Regions, Genetic , Receptors, Estrogen/analysis , Response Elements
13.
Mol Endocrinol ; 16(1): 116-27, 2002 Jan.
Article in English | MEDLINE | ID: mdl-11773443

ABSTRACT

E2 rapidly activates MAPK in breast cancer cells, and the mechanism for this effect has not been fully identified. Since growth factor-induced MAPK activation involves signaling via the adapter protein Shc (Src-homology and collagen homology) and its association with membrane receptors, we hypothesized that breast cancer cells utilize similar signaling mechanisms in response to E2. In the present study, we demonstrated that E2 rapidly induced Shc phosphorylation and Shc-Grb2 (growth factor receptor binding protein 2)-Sos (son of sevenless) complex formation in MCF-7 cells. Overexpression of dominant negative Shc blocked the effect of E2 on MAPK, indicating a critical role of Shc in E2 action. Using selective inhibitors, we also demonstrated that ERalpha and Src are upstream regulators of Shc. A rapid physical association between ERalpha and Shc upon E2 stimulation further evidenced the role of ERalpha on Shc activation. Mutagenesis studies showed that the phosphotyrosine binding and SH2 domains of Shc are required to interact with the activation function 1, but not activation function 2, domain of ERalpha. Using a glutathione-S-transferase-Shc pull-down assay, we demonstrated that this ERalpha-Shc association was direct. Biological consequences of this pathway were further investigated at the genomic and nongenomic levels. E2 stimulated MAPK-mediated Elk-1 transcriptional activity. Confocal microscopy studies showed that E2 rapidly induced formation of membrane ruffles, pseudopodia, and ERalpha membrane translocation. The E2-induced morphological changes were prevented by antiestrogen. Together our results demonstrate that ERalpha can mediate the rapid effects of E2 on Shc, MAPK, Elk-1, and morphological changes in breast cancer cells


Subject(s)
Adaptor Proteins, Signal Transducing , Adaptor Proteins, Vesicular Transport , DNA-Binding Proteins , Mitogen-Activated Protein Kinases/metabolism , Proteins/metabolism , Receptors, Estrogen/metabolism , Signal Transduction , Transcription Factors , Animals , Binding Sites , Breast Neoplasms/metabolism , COS Cells , Enzyme Inhibitors/pharmacology , Estradiol/metabolism , Estradiol/pharmacology , Estrogen Receptor alpha , Flavonoids/pharmacology , Humans , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Mitogen-Activated Protein Kinases/drug effects , Mutation , Phosphorylation , Phosphotyrosine/metabolism , Proteins/genetics , Proto-Oncogene Proteins/drug effects , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Shc Signaling Adaptor Proteins , Src Homology 2 Domain-Containing, Transforming Protein 1 , Tumor Cells, Cultured , ets-Domain Protein Elk-1
SELECTION OF CITATIONS
SEARCH DETAIL
...