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.
Mol Endocrinol ; 28(6): 965-75, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24725083

ABSTRACT

Persistent hypoxia caused by shallow trophoblast invasion and poor placental perfusion may underlie the pathophysiology of preeclampsia, a leading cause of maternal and neonatal morbidity and mortality. Previously, we found that estrogen-related receptor γ (ERRγ) serves a critical and O2-dependent role in differentiation of human trophoblasts in culture and expression of tissue kallikrein and voltage-gated K(+) channels. In this study, we surprisingly observed that ERRγ expression was significantly increased in placentas from preeclamptic women compared with that in gestation-matched normotensive women. To further investigate a functional role for ERRγ during pregnancy, we analyzed ERRγ-deficient mice. Maternal systolic blood pressure was significantly reduced in pregnant ERRγ(+/-) females bred to ERRγ(+/-) males compared with that in wild-type (WT) mice and was markedly up-regulated by treatment of WT pregnant mice with the ERRγ agonist DY131. Placentas of ERRγ(+/-) mice manifested increased vascular endothelial growth factor A expression compared with that in WT mice. Notably, circulating levels of the antiangiogenic factor, soluble fms-like tyrosine kinase-1, were significantly reduced in ERRγ(+/-) pregnant mice as was serum aldosterone. These effects were associated with a decrease in maternal adrenal Cyp11b1 (steroid 11ß-hydroxylase) and Cyp11b2 (aldosterone synthase) expression. In contrast, adrenal Cyp11b1 and Cyp11b2 mRNA were increased in pregnant WT mice treated with DY131. Moreover, chromatin immunoprecipitation and luciferase reporter assays identified Cyp11b2 as a transcriptional target of ERRγ. Collectively, these findings reveal a potential role of ERRγ in maternal blood pressure homeostasis during pregnancy and suggest that aberrant ERRγ expression may contribute to the pathogenesis of preeclampsia.


Subject(s)
Blood Pressure , Homeostasis , Receptors, Estrogen/metabolism , Animals , Cytochrome P-450 CYP11B2/genetics , Cytochrome P-450 CYP11B2/metabolism , Female , Gene Expression , Humans , Male , Mice, Inbred C57BL , Mice, Transgenic , Neovascularization, Physiologic , Placenta/blood supply , Placenta/metabolism , Pre-Eclampsia/metabolism , Pre-Eclampsia/physiopathology , Pregnancy , Promoter Regions, Genetic , Receptors, Estrogen/genetics , Steroid 11-beta-Hydroxylase/genetics , Steroid 11-beta-Hydroxylase/metabolism , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism
2.
Mol Endocrinol ; 27(6): 940-52, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23584901

ABSTRACT

Estrogen-related receptor γ (ERRγ) serves a critical O2-dependent regulatory role in the differentiation of human cytotrophoblasts to syncytiotrophoblast. In this study, we investigated expression of genes encoding tissue kallikrein (KLK1) and voltage-gated K(+) channels (KV7) during differentiation of human trophoblasts in culture and the roles of ERRγ and O2 tension in their regulation. Expression of KLK1 and the KV7 channel subunits, KCNQ1, KCNE1, KCNE3, and KCNE5, increased during differentiation of cultured human trophoblast cells in a 20% O2 environment. Notably, together with ERRγ, expression of KLK1, KCNQ1, KCNE1, KCNE3, and KCNE5 was markedly reduced when cells were cultured in a hypoxic environment (2% O2). Moreover, upon transduction of trophoblast cells with short hairpin RNAs for endogenous ERRγ, KLK1, KCNQ1, KCNE1, and KCNE3 expression was significantly decreased. Promoter and site-directed mutagenesis studies in transfected cells identified putative ERRγ response elements within the KLK1 and KCNE1 5'-flanking regions required for ERRγ-stimulated transcriptional activity. Binding of endogenous ERRγ to these ERRγ response elements increased during trophoblast differentiation in culture and was inhibited by hypoxia. The KV7 blocker linopirdine reduced human chorionic gonadotropin secretion and aggregation of cultured human trophoblasts, suggesting a possible role of KV7 channels in cell fusion and differentiation. Illumina gene expression arrays of cultured human trophoblast cells revealed several genes upregulated during syncytiotrophoblast differentiation and downregulated upon ERRγ knockdown involved in cell differentiation, adhesion, and synthesis of steroid and peptide hormones required for placental development and function. Collectively, these findings suggest that ERRγ mediates O2-dependent expression of genes involved in human trophoblast differentiation, function, and vascular homeostasis.


Subject(s)
Cell Differentiation , Oxygen/physiology , Potassium Channels, Voltage-Gated/metabolism , Receptors, Estrogen/physiology , Tissue Kallikreins/metabolism , Trophoblasts/physiology , Animals , COS Cells , Cell Differentiation/drug effects , Cell Hypoxia , Cells, Cultured , Chlorocebus aethiops , Gene Expression , Gene Expression Regulation , Humans , Potassium Channel Blockers/pharmacology , Potassium Channels, Voltage-Gated/genetics , Promoter Regions, Genetic , Protein Binding , Protein Subunits/genetics , Protein Subunits/metabolism , Response Elements , Tissue Kallikreins/genetics
3.
Mol Cell Biol ; 33(9): 1782-96, 2013 May.
Article in English | MEDLINE | ID: mdl-23438603

ABSTRACT

Mononuclear cytotrophoblasts of the human placenta proliferate rapidly, subsequently fuse, and differentiate to form multinucleated syncytiotrophoblast with induction of aromatase (hCYP19A1) and chorionic gonadotropin (hCGß) expression. Using microarray analysis, we identified members of the miR-17~92 cluster and its paralogs, miR-106a~363 and miR-106b~25, that are significantly downregulated upon syncytiotrophoblast differentiation. Interestingly, miR-19b and miR-106a directly targeted hCYP19A1 expression, while miR-19b also targeted human GCM1 (hGCM1), a transcription factor critical for mouse labyrinthine trophoblast development. Overexpression of these microRNAs (miRNAs) impaired syncytiotrophoblast differentiation. hGCM1 knockdown decreased hCYP19A1 and hCGß expression, substantiating its important role in human trophoblast differentiation. Expression of the c-Myc proto-oncogene was increased in proliferating cytotrophoblasts compared to that in differentiated syncytiotrophoblast. Moreover, c-Myc overexpression upregulated miR-17~92 and inhibited hCYP19A1 and hCGß expression. Binding of endogenous c-Myc to genomic regions upstream of the miR-17~92 and miR-106a~363 clusters in cytotrophoblasts dramatically decreased upon syncytiotrophoblast differentiation. Intriguingly, we observed higher levels of miR-106a and -19b and lower aromatase and hGCM1 expression in placentas from preeclamptic women than in placentas from gestation-matched normotensive women. Our findings reveal that c-Myc-regulated members of the miR-17~92 and miR-106a~363 clusters inhibit trophoblast differentiation by repressing hGCM1 and hCYP19A1 and suggest that aberrant regulation of these miRNAs may contribute to the pathogenesis of preeclampsia.


Subject(s)
Aromatase/genetics , MicroRNAs/genetics , Nuclear Proteins/genetics , Proto-Oncogene Proteins c-myc/metabolism , Transcription Factors/genetics , Trophoblasts/cytology , Aromatase/metabolism , Base Sequence , Cell Differentiation , Cells, Cultured , DNA-Binding Proteins , Female , Gene Expression Regulation, Developmental , Humans , MicroRNAs/metabolism , Nuclear Proteins/metabolism , Placenta/metabolism , Pre-Eclampsia/genetics , Pregnancy , Proto-Oncogene Mas , RNA, Long Noncoding , Transcription Factors/metabolism , Trophoblasts/metabolism
4.
Mol Endocrinol ; 25(9): 1513-26, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21757507

ABSTRACT

Differentiation of human cytotrophoblasts to syncytiotrophoblast and the associated induction of aromatase/hCYP19 gene expression are dependent upon a critical O(2) tension; however, the underlying molecular mechanisms remain undefined. In this study, we provide compelling evidence that expression of the orphan nuclear receptor, estrogen-related receptor γ (ERRγ), is also O(2) dependent, induced during human syncytiotrophoblast differentiation, and plays an obligatory role in the induction of placenta-specific hCYP19I.1 gene expression. Treatment with the selective ERRγ agonist, DY131, or overexpression of ERRγ, stimulated hCYP19 expression in syncytiotrophoblast. Overexpression of ERRγ prevented effects of hypoxia to repress hCYP19 gene expression in cultured trophoblasts. Conversely, small interfering RNA-mediated knockdown of endogenous ERRγ in primary trophoblasts markedly inhibited hCYP19 expression. Promoter and site-directed mutagenesis studies in transfected placental cells identified a nuclear receptor element within placenta-specific hCYP19 promoter I.1 required for ERRγ-stimulated activity. Recruitment of endogenous ERRγ to the nuclear receptor element region in hCYP19 promoter during trophoblast differentiation, assessed by chromatin immunoprecipitation, was prevented by hypoxia. Deferoxamine-induced hypoxia-inducible factor-1α (HIF-1α) levels decreased ERRγ expression, whereas knockdown of endogenous HIF-1α prevented ERRγ suppression by hypoxia. Chromatin immunoprecipitation analysis of trophoblasts cultured in hypoxia revealed recruitment of HIF-1α to one of two putative hypoxia response elements in the ERRγ promoter, providing in vivo evidence of a direct HIF-1α involvement in ERRγ expression. Collectively, these novel findings identify ERRγ as an O(2)-dependent transcription factor and HIF-1α target gene that serves a critical role in the induction of hCYP19 expression during human trophoblast differentiation.


Subject(s)
Aromatase/genetics , Cell Differentiation/drug effects , Gene Expression Regulation, Enzymologic/drug effects , Oxygen/pharmacology , Receptors, Estrogen/metabolism , Trophoblasts/cytology , Trophoblasts/enzymology , Aromatase/metabolism , Base Pairing/genetics , Cell Differentiation/genetics , Cell Hypoxia/drug effects , Cell Hypoxia/genetics , Cells, Cultured , Estrogen Receptor alpha/metabolism , Female , Gene Knockdown Techniques , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Models, Genetic , Protein Binding/drug effects , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, Estrogen/genetics , Response Elements/genetics , Transfection , Trophoblasts/drug effects
5.
Mol Endocrinol ; 23(6): 784-93, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19299445

ABSTRACT

A 246-bp region upstream of placenta-specific exon I.1 of the human aromatase (hCYP19) gene mediates placenta-specific, developmental, and O(2) regulation of expression. In this study, trophoblast differentiation and associated induction of CYP19 expression were prevented when cytotrophoblasts were cultured in phenol red-free medium containing charcoal-stripped serum or with the estrogen receptor (ER) antagonist, ICI 182,780, suggesting a stimulatory role of estrogen/ER. ERalpha protein was expressed in human trophoblasts and increased during syncytiotrophoblast differentiation, whereas ERbeta was undetectable. Mutational analysis revealed that an estrogen response element-like sequence (ERE-LS) at -208 bp is required for inductive effects of estradiol/ERalpha on hCYP19I.1 promoter activity in transfected COS-7 cells. Increased binding of syncytiotrophoblast compared with cytotrophoblast nuclear proteins to the ERE-LS was observed in vitro; however, ERalpha antibodies failed to supershift the complex and in vitro-transcribed/translated ERalpha did not bind. Nonetheless, chromatin immunoprecipitation assays in cultured trophoblasts revealed recruitment of endogenous ERalpha to the -255- to -155-bp region containing the ERE-LS before induction of hCYP19 expression; this was inhibited by ICI 182,780. Chromatin immunoprecipitation also revealed increased acetylated histone H3(K9/14) and decreased methylated histone H3(K9) associated with this region during trophoblast differentiation. These modifications were prevented when trophoblasts were incubated with ICI 182,780, suggesting that ERalpha recruitment to the -255- to -155-bp region promotes histone modifications leading to increased hCYP19 transcription. Thus, during trophoblast differentiation, estrogen/ERalpha exerts a positive feedback role, which promotes permissive histone modifications that are associated with induction of hCYP19 gene transcription.


Subject(s)
Aromatase/genetics , Estrogen Receptor alpha/metabolism , Estrogens/pharmacology , Gene Expression Regulation, Enzymologic/drug effects , Placenta/enzymology , Aromatase/metabolism , Base Pairing , Cell Differentiation/drug effects , Cells, Cultured , Charcoal , Chromatin/metabolism , Culture Media , Enzyme Induction/drug effects , Estradiol/analogs & derivatives , Estradiol/pharmacology , Estrogen Receptor alpha/genetics , Female , Fulvestrant , Histones/metabolism , Humans , Phenolsulfonphthalein/metabolism , Placenta/cytology , Placenta/drug effects , Pregnancy , Protein Binding/drug effects , RNA, Messenger/genetics , RNA, Messenger/metabolism , Response Elements/genetics , Serum Albumin, Bovine , Trophoblasts/cytology , Trophoblasts/drug effects , Trophoblasts/enzymology
6.
Mol Endocrinol ; 21(6): 1370-80, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17405905

ABSTRACT

Estrogen induces G protein-dependent nongenomic signaling in a variety of cell types via the activation of a plasma membrane-associated subpopulation of estrogen receptor alpha (ER alpha). Using pull-down experiments with purified recombinant proteins, we now demonstrate that ER alpha binds directly to G alpha i and G betagamma. Mutagenesis and the addition of blocking peptide reveals that this occurs via amino acids 251-260 and 271-595 of ER alpha, respectively. Studies of ER alpha complexed with heterotrimeric G proteins further show that estradiol causes the release of both G alpha i and G betagamma without stimulating GTP binding to G alpha i. Moreover, in COS-7 cells, the disruption of ER alpha-G alpha i interaction by deletion mutagenesis of ER alpha or expression of blocking peptide, as well as G betagamma sequestration with beta-adrenergic receptor kinase C terminus, prevents nongenomic responses to estradiol including src and erk activation. In endothelial cells, the disruption of ER alpha-G alpha i interaction prevents estradiol-induced nitric oxide synthase activation and the resulting attenuation of monocyte adhesion that contributes to estrogen-related cardiovascular protection. Thus, through direct interactions, ER alpha mediates a novel mechanism of G protein activation that provides greater diversity of function of both the steroid hormone receptor and G proteins.


Subject(s)
Estrogen Receptor alpha/metabolism , GTP-Binding Protein alpha Subunits, Gi-Go/metabolism , GTP-Binding Protein beta Subunits/metabolism , GTP-Binding Protein gamma Subunits/metabolism , Animals , COS Cells , Chlorocebus aethiops , Dimerization , Endothelial Cells/metabolism , Estradiol/pharmacology , Estrogen Receptor alpha/genetics , Extracellular Signal-Regulated MAP Kinases/metabolism , Humans , Mutagenesis , Protein Interaction Mapping , Protein Structure, Tertiary , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Signal Transduction , src-Family Kinases/metabolism
7.
J Biol Chem ; 277(43): 40799-809, 2002 Oct 25.
Article in English | MEDLINE | ID: mdl-12145316

ABSTRACT

Cyclophilin 40 (CyP40) is a tetratricopeptide repeat (TPR)-containing immunophilin and a modulator of steroid receptor function through its binding to heat shock protein 90 (Hsp90). Critical to this binding are the carboxyl-terminal MEEVD motif of Hsp90 and the TPR domain of CyP40. Two different models of the CyP40-MEEVD peptide interaction were used as the basis for a comprehensive mutational analysis of the Hsp90-interacting domain of CyP40. Using a carboxyl-terminal CyP40 construct as template, 24 amino acids from the TPR and flanking acidic and basic domains were individually mutated by site-directed mutagenesis, and the mutants were coexpressed in yeast with a carboxyl-terminal Hsp90beta construct and qualitatively assessed for binding using a beta-galactosidase filter assay. For quantitative assessment, mutants were expressed as glutathione S-transferase fusion proteins and assayed for binding to carboxyl-terminal Hsp90beta using conventional pulldown and enzyme-linked immunosorbent assay microtiter plate assays. Collectively, the models predict that the following TPR residues help define a binding groove for the MEEVD peptide: Lys-227, Asn-231, Phe-234, Ser-274, Asn-278, Lys-308, and Arg-312. Mutational analysis identified five of these residues (Lys-227, Asn-231, Asn-278, Lys-308, and Arg-312) as essential for Hsp90 binding. The other two residues (Phe-234 and Ser-274) and another three TPR domain residues not definitively associated with the binding groove (Leu-284, Lys-285, and Asp-329) are required for efficient Hsp90 binding. These data confirm the critical importance of the MEEVD binding groove in CyP40 for Hsp90 recognition and reveal that additional charged and hydrophobic residues within the CyP40 TPR domain are required for Hsp90 binding.


Subject(s)
Carrier Proteins/metabolism , Cyclophilins , HSP90 Heat-Shock Proteins/metabolism , Peptides/chemistry , Peptidylprolyl Isomerase/metabolism , Repetitive Sequences, Amino Acid , Amino Acid Motifs , Amino Acid Sequence , Base Sequence , Binding Sites , Carrier Proteins/chemistry , Carrier Proteins/genetics , Peptidyl-Prolyl Isomerase F , DNA Primers , Enzyme-Linked Immunosorbent Assay , Humans , Models, Molecular , Molecular Sequence Data , Peptidylprolyl Isomerase/chemistry , Peptidylprolyl Isomerase/genetics , Protein Binding , Protein Conformation , Sequence Homology, Amino Acid , Structure-Activity Relationship
SELECTION OF CITATIONS
SEARCH DETAIL
...