Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 3 de 3
Filter
Add more filters










Database
Language
Publication year range
1.
J Biol Chem ; 284(39): 26456-65, 2009 Sep 25.
Article in English | MEDLINE | ID: mdl-19617623

ABSTRACT

Cyclic AMP is a fundamentally important second messenger for numerous peptide hormones and neurotransmitters that control gene expression, cell proliferation, and metabolic homeostasis. Here we show that cAMP works with the POU homeodomain protein Oct-1 to regulate gene expression in pancreatic and intestinal endocrine cells. This ubiquitously expressed transcription factor is known as a stress sensor. We found that it also functions as a repressor of Cdx-2, a proglucagon gene activator. Through a mechanism that involves the activation of exchange protein activated by cyclic AMP, elevation of cAMP leads to enhanced phosphorylation and nuclear exclusion of Oct-1 and reduced interactions between Oct-1 or nuclear co-repressors and the Cdx-2 gene promoter, detected by chromatin immunoprecipitation. In rat primary pancreatic islet cells, cAMP elevation also reduces nuclear Oct-1 content, which causes increased proglucagon and proinsulin mRNA expression. Our study therefore identifies a novel mechanism by which cAMP regulates hormone-gene expression and suggests that ubiquitously expressed Oct-1 may play a role in metabolic homeostasis by functioning as a sensor for cAMP.


Subject(s)
Cyclic AMP/metabolism , Homeodomain Proteins/physiology , Octamer Transcription Factor-1/physiology , 1-Methyl-3-isobutylxanthine/pharmacology , Animals , Base Sequence , Blotting, Western , CDX2 Transcription Factor , Caco-2 Cells , Cell Line , Cells, Cultured , Chromatin Immunoprecipitation , Colforsin/pharmacology , Gene Expression Regulation , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , Islets of Langerhans/cytology , Islets of Langerhans/drug effects , Islets of Langerhans/metabolism , Molecular Sequence Data , Nuclear Proteins/metabolism , Nuclear Receptor Co-Repressor 1 , Octamer Transcription Factor-1/genetics , Octamer Transcription Factor-1/metabolism , Promoter Regions, Genetic/genetics , Protein Binding , RNA Interference , Rats , Repressor Proteins/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Sequence Analysis, DNA , Sequence Homology, Nucleic Acid , Trans-Activators/genetics , Trans-Activators/metabolism
2.
Mol Endocrinol ; 19(7): 1904-17, 2005 Jul.
Article in English | MEDLINE | ID: mdl-15731170

ABSTRACT

Transcription of the prolactin gene is dynamically controlled by positive and negative hormone signals that target the regulatory promoter region. Based on the inducibility of prolactin gene expression by inhibitors of histone deacetylases (HDACs), we examined the role of histone acetylation at the genomic prolactin promoter as a late step in transcriptional regulation. Chromatin immunoprecipitation analysis of GH4 cells revealed elevated levels of acetylated histones in the promoter and enhancer regions of the gene, compared with downstream intron sequences. 17beta-Estradiol stimulated histone H4 acetylation in the promoter region by 2- to 3-fold within 30 min. Dopamine inhibited histone H4 acetylation by 2-fold in 30 min, an effect mimicked by the MAPK kinase (MEK1) inhibitor U0126. In contrast, the synthetic glucocorticoid dexamethasone, which inhibits prolactin transcription, failed to alter histone acetylation over the same time frame. Association of transcription activator Pit-1 with the prolactin promoter was unchanged by hormone treatment. However, in response to dopamine, histone deacetylase HDAC2 and corepressor mSin3A were rapidly recruited to the prolactin promoter, and association was sustained above basal levels over a 1-h period. Consistent with this corepressor function, depletion of endogenous mSin3A by small interfering RNA was sufficient to enhance prolactin gene expression by 70%, comparable to the induction by the HDAC inhibitor, trichostatin A. These studies demonstrate that dopamine D2 receptor activation and inhibition of MAPK (ERK1/2) signaling lead to rapid deacetylation of histones at the genomic prolactin promoter. Recruitment of specific HDAC/ corepressor complexes may be an important mechanism for repression of target gene transcription by Gi/o-coupled receptors.


Subject(s)
Epigenesis, Genetic , Histones/metabolism , Pituitary Gland/metabolism , Prolactin/genetics , Receptors, Dopamine D2/agonists , Acetylation/drug effects , Animals , Cells, Cultured , Chromatin Immunoprecipitation , DNA-Binding Proteins/metabolism , Dopamine/pharmacology , Down-Regulation/genetics , Enhancer Elements, Genetic/genetics , Estradiol/pharmacology , Histone Deacetylase 2 , Histone Deacetylase Inhibitors , Histone Deacetylases/metabolism , Humans , Mice , Mitogen-Activated Protein Kinase 1/antagonists & inhibitors , Mitogen-Activated Protein Kinase 3/antagonists & inhibitors , Promoter Regions, Genetic/genetics , Rats , Receptors, Dopamine D2/physiology , Repressor Proteins/antagonists & inhibitors , Repressor Proteins/genetics , Repressor Proteins/metabolism , Sin3 Histone Deacetylase and Corepressor Complex , Transcription Factor Pit-1 , Transcription Factors/metabolism , Transcription, Genetic
3.
J Biol Chem ; 277(39): 35819-25, 2002 Sep 27.
Article in English | MEDLINE | ID: mdl-12121979

ABSTRACT

In pituitary lactotrophs the prolactin gene is stimulated by neuropeptides and estrogen and is suppressed by dopamine via D2-type receptors. Stimulatory signals converge on activation of the mitogen-activated protein kinases ERK1/2, but dopamine regulation of this pathway is not well defined. Paradoxically, D2 agonists activate ERK1/2 in many cell types. Here we show that in prolactin-secreting GH4ZR7 cells and primary pituitary cells, dopamine treatment leads to a rapid, pronounced, and specific decrease in activated ERK1/2. The response is blocked by D2-specific antagonists and pertussis toxin. Interestingly, in stable lines expressing specific pertussis toxin-resistant Galpha subunits, toxin treatment blocks dopamine suppression of MAPK in Galpha(i2)- but not Galphao-expressing cells, demonstrating that G(o)-dependent pathways can effect the inhibitory MAPK response. At the nuclear level, the MEK1 inhibitor U0126 mimics the D2-agonist bromocryptine in suppressing levels of endogenous prolactin transcripts. Moreover, a good correlation is seen between the IC(50) values for inhibition of MEK1 and suppression of prolactin promoter function (PD184352 > U0126 > U0125). Both dopamine and U0126 enhance the nuclear localization of ERF, a MAPK-sensitive ETS repressor that inhibits prolactin promoter activity. In addition, U0126 suppression is transferred by tandem copies of the Pit-1-binding site, consistent with mapping experiments for dopamine responsiveness. Our data suggest that ERK1/2 suppression is an obligatory step in the dopaminergic control of prolactin gene transcription and that bidirectional control of ERK1/2 function in the pituitary may provide a key mechanism for endocrine gene control.


Subject(s)
Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinases/metabolism , Pituitary Gland/cytology , Prolactin/biosynthesis , Prolactin/genetics , Receptors, Dopamine D2/metabolism , Transcription, Genetic , Animals , Binding Sites , Bromocriptine/pharmacology , Butadienes/pharmacology , Cells, Cultured , Cloning, Molecular , Dopamine Agonists/pharmacology , Enzyme Activation , Enzyme Inhibitors/pharmacology , Gene Expression Regulation , Green Fluorescent Proteins , Immunoblotting , Inhibitory Concentration 50 , Luminescent Proteins/metabolism , MAP Kinase Signaling System , Microscopy, Confocal , Mitogen-Activated Protein Kinase 3 , Models, Chemical , Mutagenesis, Site-Directed , Nitriles/pharmacology , Pertussis Toxin/pharmacology , Phosphorylation , Plasmids/metabolism , Prolactin/metabolism , Promoter Regions, Genetic , RNA/metabolism , Rats , Rats, Sprague-Dawley , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...