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










Publication year range
2.
Steroids ; 73(9-10): 901-5, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18261753

ABSTRACT

Estrogens play a critical role in the regulation of cellular proliferation, differentiation, and apoptosis. Evidence indicates that this regulation is mediated by a complex interface of direct control of gene expression (so-called "genomic action") and by regulation of cell-signaling/phosphorylation cascades (referred to as the "non-genomic", or "extranuclear" action). However, the mechanisms of the non-genomic action of estrogens are not well defined. We have recently described the identification of a novel scaffold protein termed MNAR (modulator of non-genomic action of estrogen receptor), that couples conventional steroid receptors with extranuclear signal transduction pathways, thus potentially providing additional and tissue- or cell-specific level of steroid hormone regulation of cell functions. We have demonstrated that the MNAR is required for ER alpha (ERa) interaction with p60(src) (Src), which leads to activation of Src/MAPK pathway. Our new data also suggest that activation of cSrc in response to E2 leads to MNAR phosphorylation, interaction with p85, and activation of the PI3 and Akt kinases. These data therefore suggest that MNAR acts as an important scaffold that integrates ERa action in regulation of important signaling pathways. ERa non-genomic action has been suggested to play a key role in estrogen-induced cardio-, neuro-, and osteo-protection. Therefore, evaluation of the molecular crosstalk between MNAR and ERa may lead to development of functionally selective ER modulators that can separate between beneficial, prodifferentiative effects in bone, the cardiovascular system and the CNS and the "detrimental", proliferative effects in reproductive tissues and organs.


Subject(s)
Estrogen Receptor alpha/metabolism , Estrogens/metabolism , Gene Expression Regulation, Neoplastic , Phosphatidylinositol 3-Kinases/metabolism , Trans-Activators/physiology , src-Family Kinases/metabolism , Catalytic Domain , Cell Line, Tumor , Cell Proliferation , Co-Repressor Proteins , Enzyme Activation , Humans , Phosphorylation , Proto-Oncogene Proteins pp60(c-src)/metabolism , Signal Transduction , Structure-Activity Relationship , Trans-Activators/metabolism , Transcription Factors
3.
J Cell Physiol ; 213(3): 610-7, 2007 Dec.
Article in English | MEDLINE | ID: mdl-17886255

ABSTRACT

By regulating activities and expression levels of key signaling molecules, estrogens control mechanisms that are responsible for crucial cellular functions. Ligand binding to estrogen receptor (ER) leads to conformational changes that regulate the receptor activity, its interaction with other proteins and DNA. In the cytoplasm, receptor interactions with kinases and scaffolding molecules regulate cell signaling cascades (extranuclear/nongenomic action). In the nucleus, estrogens control a repertoire of coregulators and other auxiliary proteins that are associated with ER, which in turn determines the nature of regulated genes and level of their expression (genomic action). The combination of genomic and nongenomic actions of estrogens ultimately confers the cell-type and tissue-type selectivity. Recent studies have revealed some important new insights into the molecular mechanisms underlying ER action, which may help to explain the functional basis of existing selective ER modulators (SERMs) and provide evidence into how ER might be selectively targeted to achieve specific therapeutic goals. In this review, we will summarize some new molecular details that relate to estrogen signaling. We will also discuss some new strategies that may potentially lead to the development of functionally selective ER modulators that can separate between the beneficial, prodifferentiative effects in bone, the cardiovascular system and the CNS as well as the "detrimental," proliferative effects in reproductive tissues and organs.


Subject(s)
Estrogens/pharmacology , Signal Transduction/drug effects , Animals , Estrogen Antagonists/pharmacology , Female , Gene Expression Regulation/drug effects , Humans , Ligands , Models, Biological , Receptors, Estrogen/metabolism , Receptors, Estrogen/physiology , Selective Estrogen Receptor Modulators/pharmacology
4.
Mol Cell Biol ; 27(5): 1904-13, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17194752

ABSTRACT

Estrogen actions are mediated by a complex interface of direct control of gene expression (the so-called "genomic action") and by regulation of cell signaling/phosphorylation cascades, referred to as the "nongenomic," or extranuclear, action. We have previously described the identification of MNAR (modulator of nongenomic action of estrogen receptor) as a novel scaffold protein that regulates estrogen receptor alpha (ERalpha) activation of cSrc. In this study, we have investigated the role of MNAR in 17beta-estradiol (E2)-induced activation of the phosphatidylinositol 3-kinase (PI3K)/Akt pathway. Consistent with our previous results, a direct correlation was established between MNAR expression levels and E2-induced activation of PI3 and Akt kinases. Endogenous MNAR, ERalpha, cSrc, and p85, the regulatory subunit of PI3 kinase, interacted in MCF7 cells treated with E2. The interaction between p85 and MNAR required activation of cSrc and MNAR phosphorylation on Tyr 920. Consequently, the mutation of this tyrosine to alanine (Y920A) abrogated the interaction between MNAR and p85 and the E2-induced activation of the PI3K/Akt pathway, which was required for the E2-induced protection of MCF7 cells from apoptosis. Nonetheless, the Y920A mutant potentiated the E2-induced activation of the Src/MAPK pathway and MCF7 cell proliferation, as observed with the wild-type MNAR. These results provide new and important insights into the molecular mechanisms of E2-induced regulation of cell proliferation and apoptosis.


Subject(s)
Estrogens/pharmacology , Phosphatidylinositol 3-Kinases/metabolism , Receptors, Estrogen/metabolism , Alanine/metabolism , Amino Acid Substitution , Animals , Apoptosis/drug effects , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , COS Cells , Cell Line, Tumor , Chlorocebus aethiops , Enzyme Activation/drug effects , Estradiol/metabolism , Estradiol/pharmacology , Estrogen Receptor alpha/metabolism , Female , Green Fluorescent Proteins/metabolism , Humans , Immunoprecipitation , MAP Kinase Signaling System , Models, Biological , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , RNA, Small Interfering/metabolism , Transfection , Tyrosine/metabolism , src-Family Kinases/metabolism
5.
Mol Endocrinol ; 21(2): 359-75, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17138644

ABSTRACT

Human progesterone receptor (PR) contains a motif that interacts with the SH3 domain of Src and mediates rapid activation of Src and downstream MAPK (Erk-1/-2) without relying on the transcriptional activity of the receptor. Here we investigated the role and intracellular location of this nontranscriptional activity of PR. Progestin activation of Src/MAPK occurred outside the nucleus with the B isoform of PR that was distributed between the cytoplasm and nucleus, but not with PR-A that was predominantly nuclear. Breast cancer cells stably expressing wild-type PR-B or PR-B with disrupting point mutations in the SH3 domain binding motif (PR-BDeltaSH3) that do not affect the transcriptional activity of PR, were compared for effects of progestin on endogenous target gene expression and cell proliferation. Progestin induction of the cyclin D1 gene, which lacks a progesterone response element, was dependent on PR activation of the Src/MAPK pathway, whereas induction of the Sgk (serum and glucocorticoid regulated kinase) gene that contains a functional progesterone response element was unaffected by mutations that interfere with PR activation of Src. Progestin induction of cell cycle progression was also abrogated in cells expressing PR-BDeltaSH3, and no effect of progestin on cyclin D1 expression and cell cycle was observed in the presence of PR-A. These results highlight the importance of PR activation of the Src/MAPK signaling pathway for progesterone-induced transcription of select target genes and cell cycle progression.


Subject(s)
Cell Cycle , Gene Expression Regulation, Neoplastic , Progesterone/physiology , Receptors, Progesterone/physiology , Animals , Cell Line , Cell Nucleus/metabolism , Cytoplasm/metabolism , Enzyme Activation/drug effects , Female , Genes, bcl-1 , Humans , Immediate-Early Proteins/metabolism , MAP Kinase Signaling System/physiology , Progesterone Congeners/pharmacology , Protein Serine-Threonine Kinases/metabolism , Receptors, Progesterone/agonists , Receptors, Progesterone/genetics , Signal Transduction , src Homology Domains , src-Family Kinases/metabolism
6.
Curr Opin Investig Drugs ; 7(10): 906-11, 2006 Oct.
Article in English | MEDLINE | ID: mdl-17086935

ABSTRACT

1alpha,25-Dihydroxyvitamin D3 (1,25-(OH)2D3), the biologically active metabolite of vitamin D, mediates its actions via the vitamin D receptor (VDR), a member of the superfamily of steroid/thyroid hormone/retinoid receptors. 1,25-(OH)2D3 is required for calcium and phosphorus homeostasis, and for normal skeletal development as well as maintenance of skeletal architecture. Two VDR ligands, calcitriol (1,25-(OH)2D3) and its synthetic analog alfacalcidol (1alpha-hydroxyvitamin D3), have been approved for the treatment of osteoporosis. However, the use of calcitriol and alfacalcidol is limited by their major side effect, hypercalcemia, which is mediated mainly by VDR activity in the small intestine. In order to identify VDR ligands with less hypercalcemia liability, a number of pharmaceutical companies are pursuing efforts to develop synthetic vitamin D analogs. This review discusses the mechanism of action of vitamin D, and summarizes the currently approved anti-osteoporotic VDR agonists and compounds that are under development. The future directions of vitamin D research for the discovery of novel VDR agonists for osteoporosis are also discussed.


Subject(s)
Bone Density Conservation Agents/pharmacology , Bone Density Conservation Agents/therapeutic use , Osteoporosis/drug therapy , Receptors, Calcitriol/drug effects , Animals , Bone and Bones/physiology , Humans , Ligands , Receptors, Calcitriol/genetics , Vitamin D/analogs & derivatives , Vitamin D/pharmacology , Vitamin D/physiology
7.
Steroids ; 71(4): 317-22, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16297421

ABSTRACT

We have previously demonstrated that modulator of nongenomic action of estrogen receptor (MNAR) integrates action of estrogen receptor alpha (ERalpha), and potentially some other nuclear receptors (NRs), in regulation of Src/Ras/Raf/mitogen-activated protein kinase (MAPK) signaling pathway. MNAR is a scaffolding protein that contains 10 LXXLL type motifs that can interact with NRs and 3 PXXP type motifs that can bind to SH3 domains present in kinases and other signaling molecules. Formation of ER-MNAR-cSrc complex leads to activation of Src and downstream Ras/Raf/MAPK pathway. The goal for this study was to compare MNAR expression in various cell lines, to optimize methods that can be used to manipulate its expression and to evaluate MNAR cellular distribution. We found that MNAR is differentially expressed. The highest levels of its expression were found in fast proliferating cells, such as breast adenocarcinoma (MCF-7)-, T cell lymphoma (Jurkat)-, prostate carcinoma (LNCaP)- and osteosarcoma (SaOS2)-derived cell lines. MNAR was undetectable in African green monkey kidney cells (COS-7) and Chinese hamster ovary cells (CHO-K1). We established and optimized a protocol to knockdown MNAR using siRNA and to overexpress it in MCF-7 cells. Exogenously expressed MNAR was found in both cytoplasmic and nuclear fractions, the majority of MNAR, however, was found in the cytoplasmic fraction. Presence of MNAR in the cell nucleus indicates that it may play a role in regulation of gene expression.


Subject(s)
Gene Expression Regulation, Neoplastic/physiology , Trans-Activators/genetics , Animals , Breast Neoplasms/metabolism , CHO Cells , COS Cells , Cell Line, Tumor , Cell Nucleus/metabolism , Chlorocebus aethiops , Co-Repressor Proteins , Cricetinae , Cricetulus , Humans , Jurkat Cells , RNA, Small Interfering , Trans-Activators/biosynthesis , Transcription Factors
8.
J Cell Biochem ; 93(1): 20-7, 2004 Sep 01.
Article in English | MEDLINE | ID: mdl-15352158

ABSTRACT

Steroid hormones control a wide variety of cellular functions important for cell homeostasis, proliferation, differentiation, and apoptosis. Evidence collected in the last few years indicates that this regulation is mediated by a complex interface of direct control of gene expression (so-called "transcriptional" action, driven by receptors that are localized in cell nucleus) and by regulation of cell signaling/phosphorylation cascades ("extranuclear" action, mediated by receptors that are localized in close proximity to cellular membrane). Regulation of gene expression takes place via ligand-dependent receptor binding to target gene promoters as part of the preinitiation complex, which leads to chromatin remodeling and ultimately regulates the rate of gene expression. Steroid-mediated regulation of cell signaling does not depend on gene transcription or protein biosynthesis. The molecular mechanism of this phenomenon is not fully understood. This review will focus on recent developments related to our understanding of the molecular mechanism of the extranuclear action of steroid hormones.


Subject(s)
Gene Expression Regulation/drug effects , Signal Transduction/drug effects , Steroids/pharmacology , Animals , Cell Communication/physiology , Humans , Phosphorylation , Receptors, Steroid/metabolism
9.
Mol Endocrinol ; 18(5): 1096-108, 2004 May.
Article in English | MEDLINE | ID: mdl-14963108

ABSTRACT

In this study, we have evaluated the molecular mechanism of Src activation after its interaction with estrogen receptor alpha (ERalpha) and a newly identified scaffold protein, called MNAR (modulator of nongenomic activity of ER). Under basal condition, Src enzymatic activity is inhibited by intramolecular interactions. The enzyme can be activated by interaction between the SH2 domain of Src and phosphotyrosine-containing sequences and/or by interaction between the SH3 domain of Src and proteins containing PXXP motifs. Mutational analysis and functional evaluation of MNAR and the use of ERalpha and cSrc mutants revealed that MNAR interacts with Src's SH3 domain via its N-terminal PXXP motif. Mutation of this motif abolished both the MNAR-induced activation of Src and the stimulation of ER transcriptional activity. ER interacts with Src's SH2 domain using phosphotyrosine 537, and this complex was further stabilized by MNAR-ER interaction. Mapping studies reveal that both the A/B domain and Y537 of ERalpha are required for MNAR-induced activation of ER transcriptional activity. The region responsible for MNAR interaction with ER maps to two N-terminal LXXLL motifs of MNAR. Mutation of these motifs prevented ER-MNAR complex formation and eliminated activation of the Src/MAPK pathway. These data explicate how the coordinate interactions between MNAR, ER, and Src lead to Src activation. Our findings also demonstrate that MNAR is a scaffold protein that mediates ER-Src interaction and plays an important role in the integration of ER action in Src-mediated signaling.


Subject(s)
Estrogen Receptor alpha/metabolism , MAP Kinase Signaling System/physiology , Trans-Activators/metabolism , src Homology Domains/physiology , src-Family Kinases/metabolism , Co-Repressor Proteins , Humans , Mutation/genetics , Phosphorylation , Protein Binding , Protein Structure, Tertiary/physiology , Transcription Factors , Tumor Cells, Cultured
10.
J Biol Chem ; 278(15): 13271-7, 2003 Apr 11.
Article in English | MEDLINE | ID: mdl-12540843

ABSTRACT

Nuclear receptor (NR)-mediated transcription is driven by dynamic multiprotein coactivator complexes, the composition of which is thought to determine the biological activity of NRs at specific promoters. The extent to which NRs discriminate between a spectrum of potential binding partners is intuitively a function of the inherent affinities of these individual interactions. Using real time interaction analysis with BIAcore, we evaluated the affinities and kinetics of the interactions of full-length members of the SRC/p160 coactivator family with estrogen receptor alpha (ER alpha) and ER beta bound to a variety of ligands. We substantiate that 17beta-estradiol enhances the affinity of ER-SRC/p160 interactions, whereas 4(OH)-tamoxifen, raloxifene, and ICI-182,780 inhibit these interactions. We show that a well defined, ER isoform-specific hierarchy governs the association of liganded ERs with full length SRC/p160 family members. Moreover, our data indicate that the interaction affinities of the full-length SRC/p160s with ERs are significantly higher then those of the NR interaction domains of the same coactivators, indicating that portions of coactivator molecules distinct from NR interaction domains might participate in receptor-coactivator complex formation. Finally, the interaction kinetics of SRC/p160s with ERs are consistent with a bipartite model, involving initial rapid formation of an unstable intermediate complex, and a subsequent slower reaction leading to its stabilization. We interpret our results as evidence that hierarchical coactivator interaction affinities are an important source of diversity in NR-mediated signaling and that the complexity of receptor-coactivator cross-talk might be best understood in the context of full-length molecules.


Subject(s)
Receptors, Estrogen/physiology , Transcription Factors/metabolism , Animals , Estradiol/pharmacology , Estrogen Receptor alpha , Estrogen Receptor beta , Genetic Vectors , Histone Acetyltransferases , Kinetics , Nuclear Receptor Coactivator 1 , Oocytes/physiology , Receptors, Estrogen/drug effects , Receptors, Estrogen/genetics , Recombinant Proteins/metabolism , Sequence Deletion , Transcription Factors/drug effects , Xenopus
11.
Proc Natl Acad Sci U S A ; 99(23): 14783-8, 2002 Nov 12.
Article in English | MEDLINE | ID: mdl-12415108

ABSTRACT

Numerous studies have demonstrated that estrogens induce rapid and transient activation of the Src/Erk phosphorylation cascade. Activation of this cascade triggers vital cellular functions including cell proliferation and differentiation. However, the details of the molecular mechanism of this process remain to be elucidated. We have identified a previously uncharacterized nuclear receptor-interacting protein designated as modulator of nongenomic activity of estrogen receptor (MNAR). Here we show that MNAR modulates estrogen-receptor (ER) interaction with members of the Src family of tyrosine kinases, which leads to a stimulation of Src enzymatic activity and activation of Erk1 and Erk2 kinases. We also show that MNAR, through activation of the Src/Erk phosphorylation cascade, affects ER transcriptional activity and ultimately ER-mediated gene expression. These data reveal that MNAR mediates the crosstalk between two important classes of signal transducing molecules and suggest that ER "genomic" and "nongenomic" activities are interrelated.


Subject(s)
MAP Kinase Signaling System/physiology , Receptor Cross-Talk/physiology , Receptors, Estrogen/physiology , src-Family Kinases/metabolism , Amino Acid Sequence , Base Sequence , Breast Neoplasms , Cloning, Molecular , DNA Primers , Estrogen Receptor alpha , Female , Humans , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3 , Mitogen-Activated Protein Kinases/metabolism , Molecular Sequence Data , Oligonucleotide Probes , Peptide Fragments/chemistry , Peptide Fragments/metabolism , Phosphorylation , Protein Binding , Transcription, Genetic , Transfection , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...