Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
Oncol Rep ; 27(6): 2057-65, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22426783

ABSTRACT

It is well established that estrogen is a potent mitogen in cells expressing estrogen receptors (ER). However, a large body of evidence has demonstrated that the effects of mitogenic estrogen signaling exhibit a non-monotonic or biphasic, dose-response curve; estrogen at low concentrations, elicits a mitogenic signaling pathway to stimulate cell proliferation, while at high concentrations, estrogen inhibits cell growth. The molecular mechanism underlying this paradoxical effect of estrogen on cell proliferation remains largely unknown. Recently, we reported that ER-α36, a variant of ER-α, mediates mitogenic estrogen signaling in ER-negative breast cancer cells. Here, we investigated the molecular mechanisms underlying the biphasic estrogen signaling in MDA-MB-231 and MDA-MB-436 ER-negative breast cancer cells. We found that 17ß-estradiol (E2ß) at l nM induced the phosphorylation of Src-Y416, an event that activates Src, while at 5 µM failed to induce Src-Y416 phosphorylation but induced Src-Y527 phosphorylation an event that inactivates Src. E2ß at 1 nM, but not at 5 µM, also induced phosphorylation of MAPK/ERK and activated Cyclin D1 promoter activity through the Src/EGFR/STAT5 pathway. Knockdown of ER­α36 abrogated the biphasic estrogen signaling in these cells. Our results thus indicate that in ER-negative breast cancer cells Src functions as a switch in ER­α36-mediated biphasic estrogen signaling through the EGFR/STAT5 pathway.


Subject(s)
Breast Neoplasms/metabolism , ErbB Receptors/metabolism , Estrogen Receptor alpha/metabolism , STAT5 Transcription Factor/metabolism , src-Family Kinases/metabolism , Cell Line, Tumor , Cyclin D1/biosynthesis , Cyclin D1/genetics , Estradiol/metabolism , Estradiol/pharmacology , Estrogen Receptor alpha/genetics , Extracellular Signal-Regulated MAP Kinases/metabolism , Female , Gene Knockdown Techniques , Humans , Phosphorylation , Promoter Regions, Genetic , Signal Transduction
2.
PLoS One ; 7(1): e30174, 2012.
Article in English | MEDLINE | ID: mdl-22276155

ABSTRACT

It is prevailingly thought that the antiestrogens tamoxifen and ICI 182, 780 are competitive antagonists of the estrogen-binding site of the estrogen receptor-alpha (ER-α). However, a plethora of evidence demonstrated both antiestrogens exhibit agonist activities in different systems such as activation of the membrane-initiated signaling pathways. The mechanisms by which antiestrogens mediate estrogen-like activities have not been fully established. Previously, a variant of ER-α, EP-α36, has been cloned and showed to mediate membrane-initiated estrogen and antiestrogen signaling in cells only expressing ER-α36. Here, we investigated the molecular mechanisms underlying the antiestrogen signaling in ER-negative breast cancer MDA-MB-231 and MDA-MB-436 cells that express high levels of endogenous ER-α36. We found that the effects of both 4-hydoxytamoxifen (4-OHT) and ICI 182, 780 (ICI) exhibited a non-monotonic, or biphasic dose response curve; antiestrogens at low concentrations, elicited a mitogenic signaling pathway to stimulate cell proliferation while at high concentrations, antiestrogens inhibited cell growth. Antiestrogens at l nM induced the phosphorylation of the Src-Y416 residue, an event to activate Src, while at 5 µM induced Src-Y527 phosphorylation that inactivates Src. Antiestrogens at 1 nM also induced phosphorylation of the MAPK/ERK and activated the Cyclin D1 promoter activity through the Src/EGFR/STAT5 pathways but not at 5 µM. Knock-down of ER-α36 abrogated the biphasic antiestrogen signaling in these cells. Our results thus indicated that ER-α36 mediates biphasic antiestrogen signaling in the ER-negative breast cancer cells and Src functions as a switch of antiestrogen signaling dependent on concentrations of antiestrogens through the EGFR/STAT5 pathway.


Subject(s)
Breast Neoplasms/metabolism , Estrogen Receptor Modulators/pharmacology , Estrogen Receptor alpha/antagonists & inhibitors , Estrogen Receptor alpha/metabolism , Blotting, Western , Cell Line, Tumor , Cell Proliferation/drug effects , Chromones/pharmacology , Estradiol/analogs & derivatives , Estradiol/pharmacology , Female , Fulvestrant , Humans , Hydroxytestosterones/pharmacology , Immunoblotting , Immunoprecipitation , Morpholines/pharmacology , Phosphorylation/drug effects , Signal Transduction/drug effects
3.
J Steroid Biochem Mol Biol ; 127(3-5): 262-8, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21907803

ABSTRACT

Accumulating evidence supports the theory that breast cancer arises from a subpopulation of mammary stem/progenitor cell which posses the ability to self-renew. However, the involvement of estrogen signaling in regulation of breast cancer stem/progenitor cells has not been fully established, mainly because expression and function of ER-α in breast cancer stem cells remains controversial. Previously, our laboratory cloned a variant of ER-α, ER-α36, and found that ER-α36-mediated non-genomic estrogen signaling plays an important role in malignant growth of triple-negative breast cancer cells. In this study, we found that ER-α36 was highly expressed in ER-negative breast cancer SK-BR-3 cells and mediated non-genomic estrogen signaling such as activation of the MAPK/ERK signaling in these cells. Knock-down of ER-α36 expression in these cells using the shRNA method diminished their responsiveness to estrogen and significantly down-regulated HER2 expression. HER2 signaling activated ER-α36 transcription through an AP1 site in the ER-α36 promoter and ER-α36 physically interacted with HER2. We also found that ER-α36 is highly expressed in a subset of SK-BR-3 cells that was positive for ALDH1, a breast cancer stem cell marker, and knock-down of ER-α36 expression reduced the population of ALDH1 positive cells. Our results thus demonstrated that ER-α36 positively regulates HER2 expression and the population of ALDH1 positive breast cancer cells, and suggested that non-genomic estrogen signaling mediated by ER-α36 is involved in maintenance and regulation of breast cancer stem cells.


Subject(s)
Breast Neoplasms/pathology , Estrogen Receptor alpha/physiology , Isoenzymes/metabolism , Receptor, ErbB-2/physiology , Retinal Dehydrogenase/metabolism , Aldehyde Dehydrogenase 1 Family , Base Sequence , Breast Neoplasms/metabolism , Cell Line, Tumor , DNA Primers , Estrogens/metabolism , Female , Humans , MAP Kinase Signaling System , Polymerase Chain Reaction , Signal Transduction
4.
Oncol Lett ; 2(2): 337-341, 2011.
Article in English | MEDLINE | ID: mdl-22737185

ABSTRACT

The genomic and non-genomic signaling pathways are well-known estrogen signaling pathways. The 66-kDa estrogen receptor-α (ER-α66) is a typical ligand-inducible transcription factor that mainly mediates genomic estrogen signaling. Recently, we identified and cloned a 36-kDa variant of ER-α66, known as ER-α36. This variant lacks intrinsic transcription activity and predominantly mediates non-genomic estrogen signaling. Thus, the expression of ER-α66 and ER-α36 should be firmly regulated and carefully correlated to maintain a balance between genomic and non-genomic estrogen signaling. However, the molecular mechanisms underlying this correlation remain poorly understood. The Wilms' tumor suppressor gene, wt1, encodes a zinc-finger protein WT1 that functions as a dual transcription regulator to activate or suppress gene transcription. High levels of WT1 expression are associated with breast cancer malignancy. In the present study, high-passage ER-positive breast cancer MCF7 cells were found to express ER-α66 and WT1 at higher levels and ER-α36 at a very low level. Using the small hairpin RNA method, stable MCF7 cells were established that expressed knocked-down levels of WT1. The cells expressed a reduced level of ER-α66 but an increased level of ER-α36, suggesting that WT1 regulates the expression of ER-α66 and ER-α36 oppositely. Further co-transfection assays showed that all isoforms of WT1 directly activated the promoter activity of the ER-α66 gene while suppressing ER-α36 promoter activity. Our results therefore indicate that WT1 acts as a dual transcription factor that regulates the promoter activity of ER-α66 and ER-α36 oppositely, implicating WT1 as one of the coordinators that orchestrate genomic and non-genomic estrogen signaling.

5.
Mol Endocrinol ; 24(4): 709-21, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20197310

ABSTRACT

Accumulating evidence suggested that an orphan G protein-coupled receptor (GPR)30, mediates nongenomic responses to estrogen. The present study was performed to investigate the molecular mechanisms underlying GPR30 function. We found that knockdown of GPR30 expression in breast cancer SK-BR-3 cells down-regulated the expression levels of estrogen receptor (ER)-alpha36, a variant of ER-alpha. Introduction of a GPR30 expression vector into GPR30 nonexpressing cells induced endogenous ER-alpha36 expression, and cotransfection assay demonstrated that GPR30 activated the promoter activity of ER-alpha36 via an activator protein 1 binding site. Both 17beta-estradiol (E2) and G1, a compound reported to be a selective GPR30 agonist, increased the phosphorylation levels of the MAPK/ERK1/2 in SK-BR-3 cells, which could be blocked by an anti-ER-alpha36-specific antibody against its ligand-binding domain. G1 induced activities mediated by ER-alpha36, such as transcription activation activity of a VP16-ER-alpha36 fusion protein and activation of the MAPK/ERK1/2 in ER-alpha36-expressing cells. ER-alpha36-expressing cells, but not the nonexpressing cells, displayed high-affinity, specific E2 and G1 binding, and E2- and G1-induced intracellular Ca(2+) mobilization only in ER-alpha36 expressing cells. Taken together, our results demonstrated that previously reported activities of GPR30 in response to estrogen were through its ability to induce ER-alpha36 expression. The selective G protein-coupled receptor (GPR)30 agonist G1 actually interacts with ER-alpha36. Thus, the ER-alpha variant ER-alpha36, not GPR30, is involved in nongenomic estrogen signaling.


Subject(s)
Estrogen Receptor alpha/radiation effects , Receptors, G-Protein-Coupled/metabolism , Animals , Blotting, Western , COS Cells , Cell Line , Cell Line, Tumor , Chlorocebus aethiops , Estradiol/pharmacology , Estrogen Receptor alpha/genetics , Humans , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Mitogen-Activated Protein Kinases/metabolism , Phosphorylation/drug effects , Receptors, Estrogen , Receptors, G-Protein-Coupled/agonists , Receptors, G-Protein-Coupled/genetics , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/drug effects , Signal Transduction/genetics
6.
J Cell Mol Med ; 14(6B): 1485-93, 2010 Jun.
Article in English | MEDLINE | ID: mdl-19840189

ABSTRACT

The dietary isothiocyanates (ITCs) exhibit strong chemopreventive activities for a variety of neoplasms including breast cancer. However, the molecular mechanisms underlying ITC function in breast cancer cells have not been well established. Here, we found that phenethyl isothiocyanate (PEITC) acted more potently than the 'pure' anti-oestrogen ICI 182,780 to inhibit the growth of oestrogen receptor (ER)(+) breast cancer MCF7 and H3396 cells and ER(-) MDA-MB-231 and SK-BR-3 cells. PEITC reduced the steady state levels of ER-alpha and its novel variant, ER-alpha36 in a dose-and time-dependent manner and inhibited oestrogen-induced activation of the mitogen activated protein kinase/ERK 1/2 signaling pathway. However, ICI 182,780 that is potent in destabilization of ER-alpha protein, failed to down-regulate ER-alpha36. Our results thus demonstrated that PEITC functions as a more potent ER-alpha'disruptor' than the well-known ICI 182,780 to abrogate ER-mediated mitogenic oestrogen signaling in breast cancer cells, which provides a molecular explanation for the strong growth inhibitory activity of ITCs in breast cancer cells, and a rational for further exploration of ITCs as chemopreventive agents for human mammary carcinogenesis.


Subject(s)
Breast Neoplasms/pathology , Down-Regulation/drug effects , Estradiol/analogs & derivatives , Estrogen Receptor alpha/metabolism , Isothiocyanates/pharmacology , Breast Neoplasms/enzymology , Breast Neoplasms/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Estradiol/chemistry , Estradiol/pharmacology , Extracellular Signal-Regulated MAP Kinases/metabolism , Female , Fulvestrant , Genome, Human/genetics , Humans , Isothiocyanates/chemistry , Leupeptins/pharmacology , Phosphorylation/drug effects , Signal Transduction/drug effects
7.
Oncol Rep ; 21(1): 185-92, 2009 Jan.
Article in English | MEDLINE | ID: mdl-19082460

ABSTRACT

The isothiocyanates (ITCs) have long been known to possess chemopreventive activities for a variety of neoplasms including breast cancer, but the molecular mechanism by which ITCs prevent breast cancer development has not been established. In this study, we investigated the effects of benzyl and phenethyl isothiocyanate (BITC and PEITC) on the estrogen-stimulated growth of estrogen receptor alpha (ERalpha)-positive breast cancer MCF7 and T-47D cells. BITC and PEITC inhibited estrogen-stimulated cell growth and reduced the expression levels of ERalpha in MCF7 and T-47D cells in a dose- and time-dependent and reversible manner. In addition, BITC and PEITC also abrogated the transcriptional activity of ERalpha and hence inhibited estrogen-stimulated expression of the estrogen responsive gene, pS2. These results demonstrated that BITC and PEITC function as potent ERalpha disruptors to abrogate mitogenic estrogen signaling in ER-positive breast cancer cells, which provides a molecular explanation for the growth inhibitory function of ITCs in breast cancer development, and a rational for further exploration of ITCs as chemopreventive agents for human mammary carcinogenesis.


Subject(s)
Antineoplastic Agents/pharmacology , Breast Neoplasms/drug therapy , Estrogen Receptor alpha/drug effects , Isothiocyanates/pharmacology , Blotting, Western , Breast Neoplasms/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Estrogen Receptor alpha/metabolism , Female , Humans , Reverse Transcriptase Polymerase Chain Reaction , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL