Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Am J Cancer Res ; 2(4): 446-58, 2012.
Article in English | MEDLINE | ID: mdl-22860234

ABSTRACT

Endocrine therapy resistance is a primary cause of clinical breast cancer treatment failure. The p38 mitogen activated protein kinase (MAPK) signaling pathway is known to promote ligand independent tumor growth and resistance to endocrine therapy. In this study, we investigated the therapeutic potential of the p38 inhibitor RWJ67657 in the treatment of tamoxifen resistant MDA-MB-361 cells. RWJ67657 dose-dependently decreased both basal and stimulated activation of p38 MAPK signaling in this drug resistant cell system. Decreased activation of p38 by RWJ67657 resulted in inhibition of the downstream p38 targets hsp27 and MAPKAPK. Diminished p38 signaling resulted in inhibition of p38-medated gene transcription. Furthermore, pharmacological inhibition of p38 by RWJ67657 decreased biological effects of p38, including ER-mediated gene expression and clonogenic survival in a dose-dependent manner. Animal studies revealed significantly decreased p38 signaling in vivo following exposure to RWJ67657. Treatment with the inhibitor markedly decreased phosphorylation of p38 in MDA-MB-361 tumors, leading to decreased transcription of both Fra-1 and progesterone receptor. Utilizing well-established xenograft tumor models, we demonstrated that RWJ67657 exhibits potent anti-tumor properties. Treatment with RWJ67657 markedly decreased tamoxifen resistant tumor growth, both in the presence and absence of estrogen. Taken together, our findings demonstrate the therapeutic potential of targeting the p38-MAPK signaling cascade in the treatment of endocrine resistant breast cancer.

2.
Sci Rep ; 2: 539, 2012.
Article in English | MEDLINE | ID: mdl-22844580

ABSTRACT

Altered death receptor signaling and resistance to subsequent apoptosis is an important clinical resistance mechanism. Here, we investigated the role of death receptor resistance in breast cancer progression. Resistance of the estrogen receptor alpha (ER)-positive, chemosensitive MCF7 breast cancer cell line to tumor necrosis factor (TNF) was associated with loss of ER expression and a multi-drug resistant phenotype. Changes in three major pathways were involved in this transition to a multidrug resistance phenotype: ER, Death Receptor and epithelial to mesenchymal transition (EMT). Resistant cells exhibited altered ER signaling, resulting in decreased ER target gene expression. The death receptor pathway was significantly altered, blocking extrinsic apoptosis and increasing NF-kappaB survival signaling. TNF resistance promoted EMT changes, resulting in a more aggressive phenotype. This first report identifying specific mechanisms underlying acquired resistance to TNF could lead to a better understanding of the progression of breast cancer in response to chemotherapy treatment.


Subject(s)
Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Drug Resistance, Neoplasm , Epithelial-Mesenchymal Transition/genetics , Receptors, Tumor Necrosis Factor/metabolism , Signal Transduction , Animals , Apoptosis , Cell Line, Tumor , Cell Survival/genetics , Cell Transformation, Neoplastic/genetics , Cluster Analysis , Disease Models, Animal , Estrogen Receptor alpha/metabolism , Female , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Humans , Mice , Mice, Nude , NF-kappa B/metabolism , Receptors, Death Domain , Tumor Necrosis Factor-alpha/pharmacology , Tumor Necrosis Factor-alpha/toxicity , Xenograft Model Antitumor Assays
3.
Cancer Biol Ther ; 13(11): 1026-33, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22825349

ABSTRACT

The p38 mitogen activated protein kinase pathway (MAPK) is known to promote cell survival, endocrine therapy resistance and hormone independent breast cancer cell proliferation. Therefore, we utilized the novel p38 inhibitor RWJ67657 to investigate the relevance of targeting this pathway in the ER (+) breast cancer cell line MCF-7. Our results show that RWJ67657 inhibits both basal and estrogen stimulated phosphorylation of p38α, resulting in decreased activation of the downstream p38α targets hsp27 and MAPAPK. Furthermore, inhibition of p38α by RWJ67657 blocks clonogenic survival of MCF-7 cells with little effect on non-cancerous breast epithelial cells. Even though p38α is known to phosphorylate ERα at residue within ER's hinge region at Thr311, resulting in increased ERα transcriptional activation, our results suggest RWJ67657 inhibits the p38α-induced activation of ER by targeting both the AF-1 and AF-2 activation domains within ERα. We further show that RWJ67657 decreases the transcriptional activity of the ER coactivators SRC-1, SRC-2 and SRC-3. Taken together, our results strongly suggest that in addition to phosphorylating Thr311 within ERα, p38α indirectly activates the ER by phosphorylation and stimulation of the known ERα coactivators, SRC-1, -2 and-3. Overall, our data underscore the therapeutic potential of targeting the p38 MAPK pathway in the treatment of ER (+) breast cancer.


Subject(s)
Breast Neoplasms/metabolism , Estrogen Receptor alpha/metabolism , Mitogen-Activated Protein Kinase 14/antagonists & inhibitors , Mitogen-Activated Protein Kinases/antagonists & inhibitors , src-Family Kinases/metabolism , Breast Neoplasms/genetics , Cell Line, Tumor , Enzyme Activation , Estrogen Receptor alpha/genetics , Female , HEK293 Cells , Humans , Imidazoles/pharmacology , MAP Kinase Signaling System/drug effects , MCF-7 Cells , Mitogen-Activated Protein Kinase 14/genetics , Mitogen-Activated Protein Kinase 14/metabolism , Mitogen-Activated Protein Kinases/metabolism , Phosphorylation , Protein Kinase Inhibitors/pharmacology , Pyridines/pharmacology , Signal Transduction , Transcriptional Activation , Transfection
4.
Exp Cell Res ; 317(18): 2573-81, 2011 Nov 01.
Article in English | MEDLINE | ID: mdl-21906588

ABSTRACT

The majority of breast cancer cases ultimately become unresponsive to endocrine therapies, and this progression of breast cancer from hormone-responsive to hormone-independent represents an area in need of further research. Additionally, hormone-independent carcinomas are characterized as being more aggressive and metastatic, key features of more advanced disease. Having previously shown the ability of the stromal-cell derived factor-1 (SDF-1)-CXCR4 signaling axis to promote primary tumorigenesis and hormone independence by overexpressing CXCR4 in MCF-7 cells, in this study we further examined the role of SDF-1/CXCR4 in the endogenously CXCR4-positive, estrogen receptor α (ER-α)-positive breast carcinoma cell line, MDA-MB-361. In addition to regulating estrogen-induced and hormone-independent tumor growth, CXCR4 signaling stimulated the epithelial-to-mesenchymal transition, evidenced by decreased CDH1 expression following SDF-1 treatment. Furthermore, inhibition of CXCR4 with the small molecule inhibitor AMD3100 induced CDH1 gene expression and inhibited CDH2 gene expression in MDA-MB-361 cells. Further, exogenous SDF-1 treatment induced ER-α-phosphorylation in both MDA-MB-361 and MCF-7-CXCR4 cells, demonstrating ligand-independent activation of ER-α through CXCR4 crosstalk. qPCR microRNA array analyses of the MDA-MB-361 and MCF-7-CXCR4 cell lines revealed changes in microRNA expression profiles induced by SDF-1, consistent with a more advanced disease phenotype and further supporting our hypothesis that the SDF-1/CXCR4 signaling axis drives ER-α-positive breast cancer cells to a hormone independent and more aggressive phenotype. In this first demonstration of SDF-1-CXCR4-induced microRNAs in breast cancer, we suggest that this signaling axis may promote tumorigenesis via microRNA regulation. These findings represent future potential therapeutic targets for the treatment of hormone-independent and endocrine-resistant breast cancer.


Subject(s)
Breast Neoplasms/metabolism , Chemokine CXCL12/metabolism , Estrogen Receptor alpha/metabolism , Mammary Neoplasms, Experimental/metabolism , MicroRNAs/genetics , Receptors, CXCR4/metabolism , Signal Transduction , Animals , Benzylamines , Breast Neoplasms/pathology , Cell Line, Tumor , Cyclams , Female , Gene Expression Profiling , Heterocyclic Compounds/pharmacology , Humans , Mice , Mice, Nude , Polymerase Chain Reaction , Receptors, CXCR4/antagonists & inhibitors
5.
Cancer Res ; 71(2): 603-13, 2011 Jan 15.
Article in English | MEDLINE | ID: mdl-21123450

ABSTRACT

Estrogen independence and progression to a metastatic phenotype are hallmarks of therapeutic resistance and mortality in breast cancer patients. Metastasis has been associated with chemokine signaling through the SDF-1-CXCR4 axis. Thus, the development of estrogen independence and endocrine therapy resistance in breast cancer patients may be driven by SDF-1-CXCR4 signaling. Here we report that CXCR4 overexpression is indeed correlated with worse prognosis and decreased patient survival irrespective of the status of the estrogen receptor (ER). Constitutive activation of CXCR4 in poorly metastatic MCF-7 cells led to enhanced tumor growth and metastases that could be reversed by CXCR4 inhibition. CXCR4 overexpression in MCF-7 cells promoted estrogen independence in vivo, whereas exogenous SDF-1 treatment negated the inhibitory effects of treatment with the anti-estrogen ICI 182,780 on CXCR4-mediated tumor growth. The effects of CXCR4 overexpression were correlated with SDF-1-mediated activation of downstream signaling via ERK1/2 and p38 MAPK (mitogen activated protein kinase) and with an enhancement of ER-mediated gene expression. Together, these results show that enhanced CXCR4 signaling is sufficient to drive ER-positive breast cancers to a metastatic and endocrine therapy-resistant phenotype via increased MAPK signaling. Our findings highlight CXCR4 signaling as a rational therapeutic target for the treatment of ER-positive, estrogen-independent breast carcinomas needing improved clinical management.


Subject(s)
Antineoplastic Agents, Hormonal/pharmacology , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Estradiol/analogs & derivatives , Receptors, CXCR4/biosynthesis , Animals , Breast Neoplasms/pathology , Cell Line, Tumor , Drug Resistance, Neoplasm , Estradiol/pharmacology , Estrogen Antagonists/pharmacology , Female , Fulvestrant , Humans , MAP Kinase Signaling System , Mice , Mice, SCID , Neoplasm Metastasis , Neoplasms, Hormone-Dependent/metabolism , Neoplasms, Hormone-Dependent/pathology , Receptors, CXCR4/metabolism , Receptors, Estrogen/antagonists & inhibitors , Receptors, Estrogen/biosynthesis
6.
Carcinogenesis ; 31(12): 2049-57, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20876285

ABSTRACT

Tamoxifen is the most commonly prescribed therapy for patients with estrogen receptor (ER)α-positive breast tumors. Tumor resistance to tamoxifen remains a serious clinical problem especially in patients with tumors that also overexpress human epidermal growth factor receptor 2 (HER2). Current preclinical models of HER2 overexpression fail to recapitulate the clinical spectrum of endocrine resistance associated with HER2/ER-positive tumors. Here, we show that ectopic expression of a clinically important oncogenic isoform of HER2, HER2Δ16, which is expressed in >30% of ER-positive breast tumors, promotes tamoxifen resistance and estrogen independence of MCF-7 xenografts. MCF-7/HER2Δ16 cells evade tamoxifen through upregulation of BCL-2, whereas mediated suppression of BCL-2 expression or treatment of MCF-7/HER2Δ16 cells with the BCL-2 family pharmacological inhibitor ABT-737 restores tamoxifen sensitivity. Tamoxifen-resistant MCF-7/HER2Δ16 cells upregulate BCL-2 protein levels in response to suppressed ERα signaling mediated by estrogen withdrawal, tamoxifen treatment or fulvestrant treatment. In addition, HER2Δ16 expression results in suppression of BCL-2-targeting microRNAs miR-15a and miR-16. Reintroduction of miR-15a/16 reduced tamoxifen-induced BCL-2 expression and sensitized MCF-7/HER2Δ16 to tamoxifen. Conversely, inhibition of miR-15a/16 in tamoxifen-sensitive cells activated BCL-2 expression and promoted tamoxifen resistance. Our results suggest that HER2Δ16 expression promotes endocrine-resistant HER2/ERα-positive breast tumors and in contrast to wild-type HER2, preclinical models of HER2Δ16 overexpression recapitulate multiple phenotypes of endocrine-resistant human breast tumors. The mechanism of HER2Δ16 therapeutic evasion, involving tamoxifen-induced upregulation of BCL-2 and suppression of miR-15a/16, provides a template for unique therapeutic interventions combining tamoxifen with modulation of microRNAs and/or ABT-737-mediated BCL-2 inhibition and apoptosis.


Subject(s)
Breast Neoplasms/drug therapy , Estrogen Antagonists/therapeutic use , Genes, bcl-2 , MicroRNAs/antagonists & inhibitors , Receptor, ErbB-2/physiology , Tamoxifen/therapeutic use , Animals , Breast Neoplasms/pathology , Cell Line, Tumor , Drug Resistance, Neoplasm , Female , Gene Expression Regulation, Neoplastic , Humans , Mice , Mice, Nude , MicroRNAs/physiology
7.
Reprod Sci ; 17(10): 894-903, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20720261

ABSTRACT

Endometriosis is associated with activation of local and systemic inflammatory mechanisms, including increased levels of chemokines and other proinflammatory cytokines. We have previously reported increased gene expression of chemokine receptor 4 (CXCR4), the receptor for CXCL12, in lesions of the rat model of endometriosis. The CXCR4-CXCL12 axis has been shown to have both immune (HIV infection, lymphocyte chemotaxis) and nonimmune functions, including roles in tissue repair, angiogenesis, invasion, and migration. There is evidence indicating that these mechanisms are also at play in endometriosis; therefore, we hypothesized that activation of the CXCR4-CXCL12 axis could be responsible, at least in part, for the survival and establishment of endometrial cells ectopically. Immunohistochemistry (IHC) showed that CXCR4 protein levels were significantly higher in endometriotic lesions compared to the endometrium of controls. Next, we determined basal gene and protein expression of CXCR4 and CXCL12 and regulation by estradiol (E2) and/or progesterone (P4) in endometrial cell lines using quantitative polymerase chain reaction (qPCR), and Western blots. Basal CXCR4 gene expression levels were higher in epithelial versus stromal cells; conversely, CXCL12 was expressed at higher levels in stromal vs epithelial cells. CXCR4 gene expression was significantly downregulated by ovarian steroid hormones in endometrial epithelial. These data suggest that steroid modulation of CXCR4 is defective in endometriosis, although the specific mechanism involved remains to be elucidated. These findings have implications for future therapeutic strategies specifically targeting the inflammatory component in endometriosis.


Subject(s)
Chemokine CXCL12/immunology , Endometriosis/immunology , Estradiol/pharmacology , Gene Expression Regulation/immunology , Progesterone/pharmacology , Receptors, CXCR4/immunology , Adult , Blotting, Western , Cell Line , Chemokine CXCL12/genetics , DNA/chemistry , DNA/genetics , Female , Humans , Middle Aged , Polymerase Chain Reaction , Receptors, CXCR4/genetics , Young Adult
8.
J Pharmacol Exp Ther ; 332(1): 35-45, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19797619

ABSTRACT

Glyceollins, a group of novel phytoalexins isolated from activated soy, have recently been demonstrated to be novel antiestrogens that bind to the estrogen receptor (ER) and inhibit estrogen-induced tumor progression. Our previous publications have focused specifically on inhibition of tumor formation and growth by the glyceollin mixture, which contains three glyceollin isomers (I, II, and III). Here, we show the glyceollin mixture is also effective as a potential antiestrogenic, therapeutic agent that prevents estrogen-stimulated tumorigenesis and displays a differential pattern of gene expression from tamoxifen. By isolating the individual glyceollin isomers (I, II, and III), we have identified the active antiestrogenic component by using competition binding assays with human ERalpha and in an estrogen-responsive element-based luciferase reporter assay. We identified glyceollin I as the active component of the combined glyceollin mixture. Ligand-receptor modeling (docking) of glyceollin I, II, and III within the ERalpha ligand binding cavity demonstrates a unique type II antiestrogenic confirmation adopted by glyceollin I but not isomers II and III. We further compared the effects of glyceollin I to the antiestrogens, 4-hydroxytamoxifen and ICI 182,780 (fulvestrant), in MCF-7 breast cancer cells and BG-1 ovarian cancer cells on 17beta-estradiol-stimulated expression of progesterone receptor and stromal derived factor-1alpha. Our results establish a novel inhibition of ER-mediated gene expression and cell proliferation/survival. Glyceollin I may represent an important component of a phytoalexin-enriched food (activated) diet in terms of chemoprevention as well as a novel therapeutic agent for hormone-dependent tumors.


Subject(s)
Anticarcinogenic Agents/pharmacology , Estrogen Receptor Modulators/pharmacology , Glycine max/chemistry , Pterocarpans/pharmacology , Terpenes/pharmacology , Animals , Anticarcinogenic Agents/chemistry , Anticarcinogenic Agents/isolation & purification , Anticarcinogenic Agents/therapeutic use , Binding Sites , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Estrogen Receptor Modulators/chemistry , Estrogen Receptor Modulators/isolation & purification , Estrogen Receptor Modulators/therapeutic use , Estrogen Receptor alpha/antagonists & inhibitors , Estrogen Receptor alpha/biosynthesis , Estrogen Receptor alpha/genetics , Female , Humans , Mice , Mice, Nude , Molecular Structure , Neoplasm Transplantation , Pterocarpans/chemistry , Pterocarpans/isolation & purification , Pterocarpans/therapeutic use , Sesquiterpenes , Stereoisomerism , Tamoxifen/pharmacology , Terpenes/chemistry , Terpenes/isolation & purification , Terpenes/therapeutic use , Transcription, Genetic/drug effects , Xenograft Model Antitumor Assays , Phytoalexins
9.
Breast Cancer Res Treat ; 121(2): 293-300, 2010 Jun.
Article in English | MEDLINE | ID: mdl-19597705

ABSTRACT

Adult human mesenchymal stem cells (hMSCs) have been shown to home to sites of breast cancer and integrate into the tumor stroma. We demonstrate here the effect of hMSCs on primary breast tumor growth and the progression of these tumors to hormone independence. Co-injection of bone marrow-derived hMSCs enhances primary tumor growth of the estrogen receptor-positive, hormone-dependent breast carcinoma cell line MCF-7 in the presence or absence of estrogen in SCID/beige mice. We also show hormone-independent growth of MCF-7 cells when co-injected with hMSCs. These effects were found in conjunction with increased immunohistochemical staining of the progesterone receptor in the MCF-7/hMSC tumors as compared to MCF-7 control tumors. This increase in PgR expression indicates a link between MCF-7 cells and MSCs through ER-mediated signaling. Taken together, our data reveal the relationship between tumor microenvironment and tumor growth and the progression to hormone independence. This tumor stroma-cell interaction may provide a novel target for the treatment of estrogen receptor-positive, hormone-independent, and endocrine-resistant breast carcinoma.


Subject(s)
Drug Resistance, Neoplasm/physiology , Mammary Neoplasms, Experimental/metabolism , Mammary Neoplasms, Experimental/pathology , Mesenchymal Stem Cells/pathology , Animals , Antineoplastic Agents, Hormonal/pharmacology , Cell Line, Tumor , Disease Progression , Female , Humans , Immunohistochemistry , Mesenchymal Stem Cells/metabolism , Mice , Mice, SCID , Receptors, Estrogen/metabolism , Receptors, Progesterone/metabolism , Xenograft Model Antitumor Assays
10.
Breast Cancer Res Treat ; 118(1): 33-43, 2009 Nov.
Article in English | MEDLINE | ID: mdl-18855134

ABSTRACT

To investigate the therapeutic effect of methylselenocysteine (MSC) combined with tamoxifen in MCF-7 breast cancer xenograft and the underlying mechanisms. MCF-7 breast cancer xenograft was established in ovariectomized female athymic nude mice and treated with tamoxifen and/or MSC. Tumor size was measured twice a week. Immunohistochemistry and TUNEL assays were used to measure ERalpha expression, ERalpha target genes (progesterone receptor (PR) and cyclin D1 expression), Ki-67 index, apoptosis and microvessel density. Combined treatment with tamoxifen and MSC synergistically inhibited tumor growth compared to MSC alone and tamoxifen alone. MSC alone or MSC + tamoxifen significantly reduced ERalpha, PR and cyclin D1, Ki67 index and microvessel density while increasing apoptosis in tumor tissues. These findings demonstrate synergistic growth inhibition of ERalpha positive breast cancer xenografts by combination of tamoxifen with organic selenium compounds. Organic selenium may provide added benefit when combined with tamoxifen in adjuvant therapy or prevention.


Subject(s)
Adenocarcinoma/drug therapy , Angiogenesis Inhibitors/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Apoptosis/drug effects , Breast Neoplasms/drug therapy , Cysteine/analogs & derivatives , Estradiol , Neoplasms, Hormone-Dependent/drug therapy , Neovascularization, Pathologic/drug therapy , Organoselenium Compounds/pharmacology , Adenocarcinoma/blood supply , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Animals , Breast Neoplasms/blood supply , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Division/drug effects , Cyclin D1/biosynthesis , Cyclin D1/genetics , Cysteine/administration & dosage , Cysteine/pharmacology , Drug Synergism , Estradiol/toxicity , Estrogen Receptor alpha/biosynthesis , Estrogen Receptor alpha/genetics , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Mice , Mice, Nude , Neoplasm Proteins/biosynthesis , Neoplasm Proteins/genetics , Neoplasms, Hormone-Dependent/blood supply , Neoplasms, Hormone-Dependent/metabolism , Neoplasms, Hormone-Dependent/pathology , Organoselenium Compounds/administration & dosage , Random Allocation , Receptors, Progesterone/biosynthesis , Receptors, Progesterone/genetics , Selenocysteine/analogs & derivatives , Specific Pathogen-Free Organisms , Tamoxifen/administration & dosage , Xenograft Model Antitumor Assays
11.
FEMS Microbiol Lett ; 256(1): 132-6, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16487330

ABSTRACT

It is known that certain yeast strains, so called 'killers', can produce and excrete proteinaceous toxins that can induce death of other sensitive strains. We obtained a stable fungicidal factor (SKF) through concentration and stabilization of the excretion product of certain killer strains of Saccharomyces cerevisiae (K1 and K2). The isolated proteinaceous complex exhibited activity at broad ranges of pH (4-7.5) and temperatures (20-37.5 degrees C). It was significantly lethal against Candida albicans and Tricophyton mentagrophytes. SKF showed stability and activity after storage, with a mean half-life of 6 months at 4 degrees C or at -20 degrees C.


Subject(s)
Antifungal Agents/pharmacology , Candida albicans/drug effects , Saccharomyces cerevisiae Proteins/pharmacology , Saccharomyces cerevisiae , Trichophyton/drug effects , Antifungal Agents/isolation & purification , Arthrodermataceae/drug effects , Drug Storage/standards , Hydrogen-Ion Concentration , Microbial Sensitivity Tests/methods , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae Proteins/physiology , Survival Analysis , Time Factors
12.
Mol Endocrinol ; 20(5): 971-83, 2006 May.
Article in English | MEDLINE | ID: mdl-16410316

ABSTRACT

Nuclear hormone receptors, such as the estrogen receptors (ERs), are regulated by specific kinase signaling pathways. Here, we demonstrate that the p38 MAPK stimulates both ERalpha- and ERbeta-mediated transcription in MCF-7 breast carcinoma, Ishikawa endometrial adenocarcinoma, and human embryonic kidney 293 cells. Inhibition of this potentiation using the p38 inhibitor, RWJ67657, blocked estrogen-mediated transcription and proliferation. Activated ERs promote gene expression in part through the recruitment of the p160 class of coactivators. Because no direct p38 phosphorylation sites have been determined on either ERalpha or beta, we hypothesized that p38 could target the p160 class of coactivators. We show for the first time using pharmacological and molecular techniques that the p160 coactivator glucocorticoid receptor-interacting protein 1 (GRIP1) is phosphorylated and potentiated by the p38 MAPK signaling cascade in vitro and in vivo. S736 was identified as a necessary site for p38 induction of GRIP1 transcriptional activation. The C terminus of GRIP1 was also demonstrated to contain a p38-responsive region. Taken together, these results indicate that p38 stimulates ER-mediated transcription by targeting the GRIP1 coactivator.


Subject(s)
Estrogen Receptor alpha/metabolism , Estrogen Receptor beta/metabolism , Nuclear Receptor Coactivator 2/metabolism , Transcription, Genetic , p38 Mitogen-Activated Protein Kinases/metabolism , Cell Line, Tumor , Cell Proliferation , Estrogens/pharmacology , Humans , Imidazoles/pharmacology , MAP Kinase Kinase 6/metabolism , Nuclear Receptor Coactivator 2/genetics , Phosphorylation , Protein Kinase Inhibitors/pharmacology , Pyridines/pharmacology , Transcription, Genetic/drug effects , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors
SELECTION OF CITATIONS
SEARCH DETAIL
...