Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Nat Commun ; 13(1): 5324, 2022 09 10.
Article in English | MEDLINE | ID: mdl-36088459

ABSTRACT

Tissue injury triggers activation of mesenchymal lineage cells into wound-repairing myofibroblasts, whose unrestrained activity leads to fibrosis. Although this process is largely controlled at the transcriptional level, whether the main transcription factors involved have all been identified has remained elusive. Here, we report multi-omics analyses unraveling Basonuclin 2 (BNC2) as a myofibroblast identity transcription factor. Using liver fibrosis as a model for in-depth investigations, we first show that BNC2 expression is induced in both mouse and human fibrotic livers from different etiologies and decreases upon human liver fibrosis regression. Importantly, we found that BNC2 transcriptional induction is a specific feature of myofibroblastic activation in fibrotic tissues. Mechanistically, BNC2 expression and activities allow to integrate pro-fibrotic stimuli, including TGFß and Hippo/YAP1 signaling, towards induction of matrisome genes such as those encoding type I collagen. As a consequence, Bnc2 deficiency blunts collagen deposition in livers of mice fed a fibrogenic diet. Additionally, our work establishes BNC2 as potentially druggable since we identified the thalidomide derivative CC-885 as a BNC2 inhibitor. Altogether, we propose that BNC2 is a transcription factor involved in canonical pathways driving myofibroblastic activation in fibrosis.


Subject(s)
Liver Cirrhosis , Myofibroblasts , Animals , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Genomics , Humans , Liver Cirrhosis/genetics , Liver Cirrhosis/metabolism , Mice , Myofibroblasts/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism
2.
Mol Syst Biol ; 16(5): e9156, 2020 05.
Article in English | MEDLINE | ID: mdl-32407006

ABSTRACT

Liver injury triggers adaptive remodeling of the hepatic transcriptome for repair/regeneration. We demonstrate that this involves particularly profound transcriptomic alterations where acute induction of genes involved in handling of endoplasmic reticulum stress (ERS) is accompanied by partial hepatic dedifferentiation. Importantly, widespread hepatic gene downregulation could not simply be ascribed to cofactor squelching secondary to ERS gene induction, but rather involves a combination of active repressive mechanisms. ERS acts through inhibition of the liver-identity (LIVER-ID) transcription factor (TF) network, initiated by rapid LIVER-ID TF protein loss. In addition, induction of the transcriptional repressor NFIL3 further contributes to LIVER-ID gene repression. Alteration to the liver TF repertoire translates into compromised activity of regulatory regions characterized by the densest co-recruitment of LIVER-ID TFs and decommissioning of BRD4 super-enhancers driving hepatic identity. While transient repression of the hepatic molecular identity is an intrinsic part of liver repair, sustained disequilibrium between the ERS and LIVER-ID transcriptional programs is linked to liver dysfunction as shown using mouse models of acute liver injury and livers from deceased human septic patients.


Subject(s)
Chemical and Drug Induced Liver Injury/metabolism , Endoplasmic Reticulum Stress/genetics , Gene Expression Regulation/genetics , Liver Diseases/metabolism , Transcriptome/genetics , Animals , Basic-Leucine Zipper Transcription Factors/genetics , Basic-Leucine Zipper Transcription Factors/metabolism , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Line , Cells, Cultured , Chemical and Drug Induced Liver Injury/genetics , Chromatin Immunoprecipitation Sequencing , Down-Regulation , Endoplasmic Reticulum Stress/drug effects , Gene Expression Profiling , Gene Regulatory Networks , Hepatocytes/drug effects , Hepatocytes/metabolism , Humans , Liver Diseases/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Thapsigargin/toxicity , Transcription Factors/genetics , Transcription Factors/metabolism , Up-Regulation
3.
Proc Natl Acad Sci U S A ; 115(47): E11033-E11042, 2018 11 20.
Article in English | MEDLINE | ID: mdl-30397120

ABSTRACT

The nuclear receptor REV-ERBα integrates the circadian clock with hepatic glucose and lipid metabolism by nucleating transcriptional comodulators at genomic regulatory regions. An interactomic approach identified O-GlcNAc transferase (OGT) as a REV-ERBα-interacting protein. By shielding cytoplasmic OGT from proteasomal degradation and favoring OGT activity in the nucleus, REV-ERBα cyclically increased O-GlcNAcylation of multiple cytoplasmic and nuclear proteins as a function of its rhythmically regulated expression, while REV-ERBα ligands mostly affected cytoplasmic OGT activity. We illustrate this finding by showing that REV-ERBα controls OGT-dependent activities of the cytoplasmic protein kinase AKT, an essential relay in insulin signaling, and of ten-of-eleven translocation (TET) enzymes in the nucleus. AKT phosphorylation was inversely correlated to REV-ERBα expression. REV-ERBα enhanced TET activity and DNA hydroxymethylated cytosine (5hmC) levels in the vicinity of REV-ERBα genomic binding sites. As an example, we show that the REV-ERBα/OGT complex modulates SREBP-1c gene expression throughout the fasting/feeding periods by first repressing AKT phosphorylation and by epigenomically priming the Srebf1 promoter for a further rapid response to insulin. Conclusion: REV-ERBα regulates cytoplasmic and nuclear OGT-controlled processes that integrate at the hepatic SREBF1 locus to control basal and insulin-induced expression of the temporally and nutritionally regulated lipogenic SREBP-1c transcript.


Subject(s)
Insulin/metabolism , N-Acetylglucosaminyltransferases/metabolism , Nuclear Receptor Subfamily 1, Group D, Member 1/metabolism , Sterol Regulatory Element Binding Protein 1/biosynthesis , Animals , Cell Line, Tumor , Circadian Clocks/physiology , Gene Expression Regulation/genetics , Glucose/metabolism , HEK293 Cells , Hep G2 Cells , Humans , Lipid Metabolism/physiology , Liver/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , N-Acetylglucosaminyltransferases/genetics , Nuclear Receptor Subfamily 1, Group D, Member 1/genetics , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction , Sterol Regulatory Element Binding Protein 1/genetics
4.
J Hepatol ; 69(5): 1099-1109, 2018 Nov.
Article in English | MEDLINE | ID: mdl-29981427

ABSTRACT

BACKGROUND & AIMS: Embedded into a complex signaling network that coordinates glucose uptake, usage and production, the nuclear bile acid receptor FXR is expressed in several glucose-processing organs including the liver. Hepatic gluconeogenesis is controlled through allosteric regulation of gluconeogenic enzymes and by glucagon/cAMP-dependent transcriptional regulatory pathways. We aimed to elucidate the role of FXR in the regulation of fasting hepatic gluconeogenesis. METHODS: The role of FXR in hepatic gluconeogenesis was assessed in vivo and in mouse primary hepatocytes. Gene expression patterns in response to glucagon and FXR agonists were characterized by quantitative reverse transcription PCR and microarray analysis. FXR phosphorylation by protein kinase A was determined by mass spectrometry. The interaction of FOXA2 with FXR was identified by cistromic approaches and in vitro protein-protein interaction assays. The functional impact of the crosstalk between FXR, the PKA and FOXA2 signaling pathways was assessed by site-directed mutagenesis, transactivation assays and restoration of FXR expression in FXR-deficient hepatocytes in which gene expression and glucose production were assessed. RESULTS: FXR positively regulates hepatic glucose production through two regulatory arms, the first one involving protein kinase A-mediated phosphorylation of FXR, which allowed for the synergistic activation of gluconeogenic genes by glucagon, agonist-activated FXR and CREB. The second arm involves the inhibition of FXR's ability to induce the anti-gluconeogenic nuclear receptor SHP by the glucagon-activated FOXA2 transcription factor, which physically interacts with FXR. Additionally, knockdown of Foxa2 did not alter glucagon-induced and FXR agonist enhanced expression of gluconeogenic genes, suggesting that the PKA and FOXA2 pathways regulate distinct subsets of FXR responsive genes. CONCLUSIONS: Thus, hepatic glucose production is regulated during physiological fasting by FXR, which integrates the glucagon/cAMP signal and the FOXA2 signal, by being post-translationally modified, and by engaging in protein-protein interactions, respectively. LAY SUMMARY: Activation of the nuclear bile acid receptor FXR regulates gene expression networks, controlling lipid, cholesterol and glucose metabolism, which are mostly effective after eating. Whether FXR exerts critical functions during fasting is unknown. The results of this study show that FXR transcriptional activity is regulated by the glucagon/protein kinase A and the FOXA2 signaling pathways, which act on FXR through phosphorylation and protein-protein interactions, respectively, to increase hepatic glucose synthesis.


Subject(s)
Cyclic AMP-Dependent Protein Kinases/physiology , Fasting/metabolism , Gluconeogenesis , Hepatocyte Nuclear Factor 3-beta/physiology , Liver/metabolism , Receptors, Cytoplasmic and Nuclear/physiology , Animals , Gene Expression Regulation , Glucagon/physiology , Glucose/metabolism , Hepatocytes/metabolism , Male , Mice , Mice, Inbred C57BL , Phosphorylation
5.
Sci Rep ; 7(1): 14087, 2017 10 26.
Article in English | MEDLINE | ID: mdl-29075020

ABSTRACT

Adipocyte differentiation and function relies on a network of transcription factors, which is disrupted in obesity-associated low grade, chronic inflammation leading to adipose tissue dysfunction. In this context, there is a need for a thorough understanding of the transcriptional regulatory network involved in adipose tissue pathophysiology. Recent advances in the functional annotation of the genome has highlighted the role of non-coding RNAs in cellular differentiation processes in coordination with transcription factors. Using an unbiased genome-wide approach, we identified and characterized a novel long intergenic non-coding RNA (lincRNA) strongly induced during adipocyte differentiation. This lincRNA favors adipocyte differentiation and coactivates the master adipogenic regulator peroxisome proliferator-activated receptor gamma (PPARγ) through interaction with the paraspeckle component and hnRNP-like RNA binding protein 14 (RBM14/NCoAA), and was therefore called PPARγ-activator RBM14-associated lncRNA (Paral1). Paral1 expression is restricted to adipocytes and decreased in humans with increasing body mass index. A decreased expression was also observed in diet-induced or genetic mouse models of obesity and this down-regulation was mimicked in vitro by TNF treatment. In conclusion, we have identified a novel component of the adipogenic transcriptional regulatory network defining the lincRNA Paral1 as an obesity-sensitive regulator of adipocyte differentiation and function.


Subject(s)
Adipocytes/metabolism , Adipogenesis/physiology , Intracellular Signaling Peptides and Proteins/metabolism , PPAR gamma/metabolism , RNA, Long Noncoding/metabolism , Transcription Factors/metabolism , 3T3 Cells , Adult , Animals , Body Mass Index , Cell Nucleus/metabolism , Disease Models, Animal , Female , Humans , Inflammation , Mesenchymal Stem Cells/metabolism , Mice , Middle Aged , Obesity/metabolism , Transcription, Genetic
6.
ACS Chem Biol ; 12(3): 654-663, 2017 03 17.
Article in English | MEDLINE | ID: mdl-28059499

ABSTRACT

Chicken ovalbumin upstream promoter-transcription factor II (COUP-TFII/NR2F2) is an orphan member of the nuclear receptor family of transcription factors whose activities are modulated upon binding of small molecules into an hydrophobic ligand-binding pocket (LBP). Although the LBP of COUP-TFII is filled with aromatic amino-acid side chains, alternative modes of ligand binding could potentially lead to regulation of the orphan receptor. Here, we screened a synthetic and natural compound library in a yeast one-hybrid assay and identified 4-methoxynaphthol as an inhibitor of COUP-TFII. This synthetic inhibitor was able to counteract processes either positively or negatively regulated by COUP-TFII in different mammalian cell systems. Hence, we demonstrate that the true orphan receptor COUP-TFII can be targeted by small chemicals which could be used to study the physiological functions of COUP-TFII or to counteract detrimental COUP-TFII activities in various pathological conditions.


Subject(s)
COUP Transcription Factor II/antagonists & inhibitors , Small Molecule Libraries , 3T3-L1 Cells , Adipocytes/cytology , Adipocytes/drug effects , Animals , Binding Sites , COUP Transcription Factor II/metabolism , Cell Differentiation/drug effects , Hep G2 Cells , Humans , Mice
7.
J Clin Invest ; 124(3): 1037-51, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24531544

ABSTRACT

The nuclear bile acid receptor farnesoid X receptor (FXR) is an important transcriptional regulator of bile acid, lipid, and glucose metabolism. FXR is highly expressed in the liver and intestine and controls the synthesis and enterohepatic circulation of bile acids. However, little is known about FXR-associated proteins that contribute to metabolic regulation. Here, we performed a mass spectrometry-based search for FXR-interacting proteins in human hepatoma cells and identified AMPK as a coregulator of FXR. FXR interacted with the nutrient-sensitive kinase AMPK in the cytoplasm of target cells and was phosphorylated in its hinge domain. In cultured human and murine hepatocytes and enterocytes, pharmacological activation of AMPK inhibited FXR transcriptional activity and prevented FXR coactivator recruitment to promoters of FXR-regulated genes. Furthermore, treatment with AMPK activators, including the antidiabetic biguanide metformin, inhibited FXR agonist induction of FXR target genes in mouse liver and intestine. In a mouse model of intrahepatic cholestasis, metformin treatment induced FXR phosphorylation, perturbed bile acid homeostasis, and worsened liver injury. Together, our data indicate that AMPK directly phosphorylates and regulates FXR transcriptional activity to precipitate liver injury under conditions favoring cholestasis.


Subject(s)
Adenylate Kinase/metabolism , Bile Acids and Salts/biosynthesis , Homeostasis , Hypoglycemic Agents/pharmacology , Metformin/pharmacology , Receptors, Cytoplasmic and Nuclear/metabolism , Adenylate Kinase/antagonists & inhibitors , Amino Acid Sequence , Aminoimidazole Carboxamide/analogs & derivatives , Aminoimidazole Carboxamide/pharmacology , Animals , Biological Transport , Caco-2 Cells , Cholestasis, Intrahepatic/metabolism , Cholestasis, Intrahepatic/pathology , Hep G2 Cells , Humans , Intestinal Mucosa/metabolism , Intestines/drug effects , Liver/drug effects , Liver/metabolism , Liver/pathology , Male , Mice , Mice, Inbred C57BL , Molecular Sequence Data , Phosphorylation , Promoter Regions, Genetic , Protein Binding , Protein Processing, Post-Translational , Receptors, Cytoplasmic and Nuclear/chemistry , Ribonucleotides/pharmacology , Signal Transduction , Trans-Activators/metabolism , Transcription, Genetic , Transcriptional Activation/drug effects
8.
Hepatology ; 59(5): 2022-33, 2014 May.
Article in English | MEDLINE | ID: mdl-24037988

ABSTRACT

UNLABELLED: Bile acid metabolism is intimately linked to the control of energy homeostasis and glucose and lipid metabolism. The nuclear receptor farnesoid X receptor (FXR) plays a major role in the enterohepatic cycling of bile acids, but the impact of nutrients on bile acid homeostasis is poorly characterized. Metabolically active hepatocytes cope with increases in intracellular glucose concentrations by directing glucose into storage (glycogen) or oxidation (glycolysis) pathways, as well as to the pentose phosphate shunt and the hexosamine biosynthetic pathway. Here we studied whether the glucose nonoxidative hexosamine biosynthetic pathway modulates FXR activity. Our results show that FXR interacts with and is O-GlcNAcylated by O-GlcNAc transferase in its N-terminal AF1 domain. Increased FXR O-GlcNAcylation enhances FXR gene expression and protein stability in a cell type-specific manner. High glucose concentrations increased FXR O-GlcNAcylation, hence its protein stability and transcriptional activity by inactivating corepressor complexes, which associate in a ligand-dependent manner with FXR, and increased FXR binding to chromatin. Finally, in vivo fasting-refeeding experiments show that FXR undergoes O-GlcNAcylation in fed conditions associated with increased direct FXR target gene expression and decreased liver bile acid content. CONCLUSION: FXR activity is regulated by glucose fluxes in hepatocytes through a direct posttranslational modification catalyzed by the glucose-sensing hexosamine biosynthetic pathway.


Subject(s)
Bile Acids and Salts/metabolism , Glucose/metabolism , N-Acetylglucosaminyltransferases/physiology , Receptors, Cytoplasmic and Nuclear/physiology , Acylation , Animals , Gene Expression Regulation , Hep G2 Cells , Hepatocytes/metabolism , Hexosamines/biosynthesis , Humans , Male , Mice , Mice, Inbred C57BL , Pentose Phosphate Pathway , Receptors, Cytoplasmic and Nuclear/genetics , Signal Transduction
9.
PLoS One ; 8(6): e64880, 2013.
Article in English | MEDLINE | ID: mdl-23762261

ABSTRACT

Nuclear all-trans retinoic acid receptors (RARs) initiate early transcriptional events which engage pluripotent cells to differentiate into specific lineages. RAR-controlled transactivation depends mostly on agonist-induced structural transitions in RAR C-terminus (AF-2), thus bridging coactivators or corepressors to chromatin, hence controlling preinitiation complex assembly. However, the contribution of other domains of RAR to its overall transcriptional activity remains poorly defined. A proteomic characterization of nuclear proteins interacting with RAR regions distinct from the AF-2 revealed unsuspected functional properties of the RAR N-terminus. Indeed, mass spectrometry fingerprinting identified the Bromodomain-containing protein 4 (BRD4) and ALL1-fused gene from chromosome 9 (AF9/MLLT3), known to associate with and regulates the activity of Positive Transcription Elongation Factor b (P-TEFb), as novel RAR coactivators. In addition to promoter sequences, RAR binds to genomic, transcribed regions of retinoid-regulated genes, in association with RNA polymerase II and as a function of P-TEFb activity. Knockdown of either AF9 or BRD4 expression affected differentially the neural differentiation of stem cell-like P19 cells. Clusters of retinoid-regulated genes were selectively dependent on BRD4 and/or AF9 expression, which correlated with RAR association to transcribed regions. Thus RAR establishes physical and functional links with components of the elongation complex, enabling the rapid retinoid-induced induction of genes required for neuronal differentiation. Our data thereby extends the previously known RAR interactome from classical transcriptional modulators to components of the elongation machinery, and unravel a functional role of RAR in transcriptional elongation.


Subject(s)
Cell Differentiation , Nuclear Proteins/metabolism , Pluripotent Stem Cells/cytology , Positive Transcriptional Elongation Factor B/metabolism , Receptors, Retinoic Acid/genetics , Transcription Factors/metabolism , Transcriptional Activation , Antineoplastic Agents/pharmacology , Biomarkers/metabolism , Blotting, Western , Cell Cycle Proteins , Cell Proliferation , Chromatin Immunoprecipitation , Fluorescent Antibody Technique , Gene Expression Profiling , HeLa Cells , Humans , Immunoprecipitation , Nuclear Proteins/antagonists & inhibitors , Nuclear Proteins/genetics , Oligonucleotide Array Sequence Analysis , Pluripotent Stem Cells/drug effects , Pluripotent Stem Cells/metabolism , Positive Transcriptional Elongation Factor B/genetics , Promoter Regions, Genetic/genetics , RNA, Messenger/genetics , RNA, Small Interfering/genetics , Real-Time Polymerase Chain Reaction , Receptors, Retinoic Acid/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Transcription Factors/antagonists & inhibitors , Transcription Factors/genetics , Tretinoin/pharmacology
10.
Mol Cell Biol ; 33(11): 2202-11, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23530060

ABSTRACT

The glucose-activated transcription factor carbohydrate response element binding protein (ChREBP) induces the expression of hepatic glycolytic and lipogenic genes. The farnesoid X receptor (FXR) is a nuclear bile acid receptor controlling bile acid, lipid, and glucose homeostasis. FXR negatively regulates hepatic glycolysis and lipogenesis in mouse liver. The aim of this study was to determine whether FXR regulates the transcriptional activity of ChREBP in human hepatocytes and to unravel the underlying molecular mechanisms. Agonist-activated FXR inhibits glucose-induced transcription of several glycolytic genes, including the liver-type pyruvate kinase gene (L-PK), in the immortalized human hepatocyte (IHH) and HepaRG cell lines. This inhibition requires the L4L3 region of the L-PK promoter, known to bind the transcription factors ChREBP and hepatocyte nuclear factor 4α (HNF4α). FXR interacts directly with ChREBP and HNF4α proteins. Analysis of the protein complex bound to the L4L3 region reveals the presence of ChREBP, HNF4α, FXR, and the transcriptional coactivators p300 and CBP at high glucose concentrations. FXR activation does not affect either FXR or HNF4α binding to the L4L3 region but does result in the concomitant release of ChREBP, p300, and CBP and in the recruitment of the transcriptional corepressor SMRT. Thus, FXR transrepresses the expression of genes involved in glycolysis in human hepatocytes.


Subject(s)
Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/genetics , Hepatocytes/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Animals , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , Binding Sites , Cell Line , Gene Expression Regulation , Glucose/metabolism , Glucose/pharmacology , Glycolysis/genetics , Hepatocyte Nuclear Factor 4/genetics , Hepatocyte Nuclear Factor 4/metabolism , Hepatocytes/drug effects , Histones/metabolism , Humans , Liver/metabolism , Lysine/metabolism , Mice , Nuclear Receptor Co-Repressor 2/genetics , Nuclear Receptor Co-Repressor 2/metabolism , Peptide Fragments/genetics , Peptide Fragments/metabolism , Promoter Regions, Genetic , Protein Transport , Pyruvate Kinase/genetics , Pyruvate Kinase/metabolism , Receptors, Cytoplasmic and Nuclear/genetics , Sialoglycoproteins/genetics , Sialoglycoproteins/metabolism , p300-CBP Transcription Factors/genetics , p300-CBP Transcription Factors/metabolism
11.
FEBS Lett ; 587(23): 3883-90, 2013 Nov 29.
Article in English | MEDLINE | ID: mdl-24396871

ABSTRACT

Nur77 is a stress sensor in pancreatic ß-cells, which negatively regulates glucose-stimulated insulin secretion. We recently showed that a lipotoxic shock caused by exposure of ß-cells to the saturated fatty acid palmitate strongly increases Nur77 expression. Here, using dual luciferase reporter assays and Nur77 promoter deletion constructs, we identified a regulatory cassette between -1534 and -1512 bp upstream from the translational start site mediating Nur77 promoter activation in response to palmitate exposure. Chromatin immunoprecipitation, transient transfection and siRNA-mediated knockdown assays revealed that palmitate induced Nur77 promoter activation involves Sp1 recruitment and ZBP89 release from the gene promoter.


Subject(s)
DNA-Binding Proteins/metabolism , Insulin-Secreting Cells/metabolism , Nuclear Receptor Subfamily 4, Group A, Member 1/genetics , Palmitic Acid/pharmacology , Promoter Regions, Genetic , Sp1 Transcription Factor/metabolism , Transcription Factors/metabolism , Transcription, Genetic/drug effects , Animals , Cell Line, Tumor , DNA-Binding Proteins/genetics , Mice , Nuclear Receptor Subfamily 4, Group A, Member 1/metabolism , Protein Binding/drug effects , RNA, Small Interfering , Sp1 Transcription Factor/genetics , Transcription Factors/genetics
12.
FEBS Lett ; 2013 Oct 25.
Article in English | MEDLINE | ID: mdl-24512852

ABSTRACT

Nur77 is a stress sensor in pancreatic ß-cells, which negatively regulates glucose-stimulated insulin secretion. We recently showed that a lipotoxic shock caused by exposure of ß-cells to the saturated fatty acid palmitate strongly increases Nur77 expression. Here, using dual luciferase reporter assays and Nur77 promoter deletion constructs, we identified a regulatory cassette between -1534 and -1512 bp upstream from the translational start site mediating Nur77 promoter activation in response to palmitate exposure. Chromatin immunoprecipitation, transient transfection and siRNA-mediated knockdown assays revealed that palmitate induced Nur77 promoter activation involves Sp1 recruitment and ZBP89 release from the gene promoter.

13.
Mol Endocrinol ; 26(3): 399-413, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22301783

ABSTRACT

The NR4A orphan nuclear receptors Nur77, Nurr1, and Nor1 exert multiple cellular and metabolic functions. These transcriptional regulators are activated in response to extracellular stresses, including lipotoxic fatty acids (FA) and proinflammatory cytokines. The contribution of NR4As to ß-cell pathophysiology is, however, unknown. We have therefore examined the role of NR4As as downstream contributors to FA-induced ß-cell dysfunctions. Human pancreatic islets and insulinoma ß-cells were used to determine transcriptional programs elicited by NR4A, which were compared to those triggered by palmitate treatment. Functional studies evaluated the consequence of an increased NR4A expression on insulin biosynthesis and secretion and cell viability in insulinoma ß-cells. FA and cytokine treatment increased NR4A expression in pancreatic ß-cells, with Nur77 being most highly inducible in murine ß-cells. Nur77, Nurr1, or Nor1 modulated common and distinct clusters of genes involved notably in cation homeostasis and insulin gene transcription. By altering zinc homeostasis, insulin gene transcription, and secretion, Nur77 was found to be a major transcriptional mediator of part of FA-induced ß-cell dysfunctions. The repressive role of Nur77 in insulin gene regulation was tracked down to protein-protein interaction with FoxO1, a pivotal integrator of the insulin gene regulatory network. The present study identifies a member of the NR4A nuclear receptor subclass, Nur77/NR4A1, as a modulator of pancreatic ß-cell biology. Together with its previously documented role in liver and muscle, its role in ß-cells establishes Nur77 as an important integrator of glucose metabolism.


Subject(s)
Glucose/physiology , Insulin-Secreting Cells/metabolism , Insulin/metabolism , Nuclear Receptor Subfamily 4, Group A, Member 1/physiology , Animals , Cell Line , Chromogranin A/metabolism , Fatty Acids/physiology , Forkhead Box Protein O1 , Forkhead Transcription Factors/metabolism , Gene Expression , Gene Expression Profiling , Gene Expression Regulation , Glucose/metabolism , Humans , Insulin/genetics , Insulin Secretion , Maf Transcription Factors, Large/genetics , Maf Transcription Factors, Large/metabolism , Mice , Nuclear Receptor Subfamily 4, Group A, Member 1/genetics , Nuclear Receptor Subfamily 4, Group A, Member 1/metabolism , Pancreas/cytology , Pancreas/metabolism , Pancreas/physiology , Promoter Regions, Genetic , Protein Binding , Stress, Physiological
SELECTION OF CITATIONS
SEARCH DETAIL
...