Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
2.
Metabolism ; 151: 155720, 2024 Feb.
Article in English | MEDLINE | ID: mdl-37926201

ABSTRACT

BACKGROUND AND AIMS: Peroxisome Proliferator-Activated Receptor α (PPARα) is a key regulator of hepatic lipid metabolism and therefore a promising therapeutic target against Metabolic-dysfunction Associated Steatotic Liver Diseases (MASLD). However, its expression and activity decrease during disease progression and several of its agonists did not achieve sufficient efficiency in clinical trials with, surprisingly, a lack of steatosis improvement. Here, we identified the Human leukocyte antigen-F Adjacent Transcript 10 (FAT10) as an inhibitor of PPARα lipid metabolic activity during MASLD progression. APPROACH AND RESULTS: In vivo, the expression of FAT10 is upregulated in human and murine MASLD livers upon disease progression and correlates negatively with PPARα expression. The increase of FAT10 occurs in hepatocytes in which both proteins interact. FAT10 silencing in vitro in hepatocytes increases PPARα target gene expression, promotes fatty acid oxidation and decreases intra-cellular lipid droplet content. In line, FAT10 overexpression in hepatocytes in vivo inhibits the lipid regulatory activity of PPARα in response to fasting and agonist treatment in conditions of physiological and pathological hepatic lipid overload. CONCLUSIONS: FAT10 is induced during MASLD development and interacts with PPARα resulting in a decreased lipid metabolic response of PPARα to fasting or agonist treatment. Inhibition of the FAT10-PPARα interaction may provide a means to design potential therapeutic strategies against MASLD.


Subject(s)
Fatty Liver , Metabolic Diseases , Animals , Humans , Mice , Disease Progression , Fatty Acids/metabolism , Fatty Liver/metabolism , Lipid Metabolism/genetics , Liver/metabolism , Metabolic Diseases/metabolism , PPAR alpha/metabolism , Ubiquitin/metabolism , Ubiquitins/metabolism
3.
J Med Chem ; 66(17): 11732-11760, 2023 09 14.
Article in English | MEDLINE | ID: mdl-37639383

ABSTRACT

A novel series of potent agonists of the bile acid receptor TGR5 bearing a dihydropyridone scaffold was developed from a high-throughput screen. Starting from a micromolar hit compound, we implemented an extensive structure-activity-relationship (SAR) study with the synthesis and biological evaluation of 83 analogues. The project culminated with the identification of the potent nanomolar TGR5 agonist 77A. We report the GLP-1 secretagogue effect of our lead compound ex vivo in mouse colonoids and in vivo. In addition, to identify specific features favorable for TGR5 activation, we generated and optimized a three-dimensional quantitative SAR model that contributed to our understanding of our activity profile and could guide further development of this dihydropyridone series.


Subject(s)
Quantitative Structure-Activity Relationship , Transcription Factors , Animals , Mice , Glucagon-Like Peptide 1 , Bile Acids and Salts
4.
Mol Metab ; 69: 101686, 2023 03.
Article in English | MEDLINE | ID: mdl-36746333

ABSTRACT

OBJECTIVE: Obesity is associated with metabolic dysfunction of white adipose tissue (WAT). Activated adipocytes secrete pro-inflammatory cytokines resulting in the recruitment of pro-inflammatory macrophages, which contribute to WAT insulin resistance. The bile acid (BA)-activated nuclear Farnesoid X Receptor (FXR) controls systemic glucose and lipid metabolism. Here, we studied the role of FXR in adipose tissue function. METHODS: We first investigated the immune phenotype of epididymal WAT (eWAT) from high fat diet (HFD)-fed whole-body FXR-deficient (FXR-/-) mice by flow cytometry and gene expression analysis. We then generated adipocyte-specific FXR-deficient (Ad-FXR-/-) mice and analyzed systemic and eWAT metabolism and immune phenotype upon HFD feeding. Transcriptomic analysis was done on mature eWAT adipocytes from HFD-fed Ad-FXR-/- mice. RESULTS: eWAT from HFD-fed whole-body FXR-/- and Ad-FXR-/- mice displayed decreased pro-inflammatory macrophage infiltration and inflammation. Ad-FXR-/- mice showed lower blood glucose concentrations, improved systemic glucose tolerance and WAT insulin sensitivity and oxidative stress. Transcriptomic analysis identified Gsta4, a modulator of oxidative stress in WAT, as the most upregulated gene in Ad-FXR-/- mouse adipocytes. Finally, chromatin immunoprecipitation analysis showed that FXR binds the Gsta4 gene promoter. CONCLUSIONS: These results indicate a role for the adipocyte FXR-GSTA4 axis in controlling HFD-induced inflammation and systemic glucose homeostasis.


Subject(s)
Insulin Resistance , Animals , Mice , Adipocytes/metabolism , Adipose Tissue/metabolism , Glucose/metabolism , Homeostasis , Inflammation/metabolism , Insulin Resistance/physiology , Oxidative Stress , Receptors, Cytoplasmic and Nuclear/metabolism
5.
Front Mol Neurosci ; 14: 808603, 2021.
Article in English | MEDLINE | ID: mdl-35058750

ABSTRACT

The nuclear bile acid (BA) receptor farnesoid X receptor (FXR) is a major regulator of metabolic/energy homeostasis in peripheral organs. Indeed, enterohepatic-expressed FXR controls metabolic processes (BA, glucose and lipid metabolism, fat mass, body weight). The central nervous system (CNS) regulates energy homeostasis in close interaction with peripheral organs. While FXR has been reported to be expressed in the brain, its function has not been studied so far. We studied the role of FXR in brain control of energy homeostasis by treating wild-type and FXR-deficient mice by intracerebroventricular (ICV) injection with the reference FXR agonist GW4064. Here we show that pharmacological activation of brain FXR modifies energy homeostasis by affecting brown adipose tissue (BAT) function. Brain FXR activation decreases the rate-limiting enzyme in catecholamine synthesis, tyrosine hydroxylase (TH), and consequently the sympathetic tone. FXR activation acts by inhibiting hypothalamic PKA-CREB induction of TH expression. These findings identify a function of brain FXR in the control of energy homeostasis and shed new light on the complex control of energy homeostasis by BA through FXR.

6.
J Biol Chem ; 295(50): 17310-17322, 2020 12 11.
Article in English | MEDLINE | ID: mdl-33037071

ABSTRACT

In addition to their well-known role in the control of cellular proliferation and cancer, cell cycle regulators are increasingly identified as important metabolic modulators. Several GWAS have identified SNPs near CDKN2A, the locus encoding for p16INK4a (p16), associated with elevated risk for cardiovascular diseases and type-2 diabetes development, two pathologies associated with impaired hepatic lipid metabolism. Although p16 was recently shown to control hepatic glucose homeostasis, it is unknown whether p16 also controls hepatic lipid metabolism. Using a combination of in vivo and in vitro approaches, we found that p16 modulates fasting-induced hepatic fatty acid oxidation (FAO) and lipid droplet accumulation. In primary hepatocytes, p16-deficiency was associated with elevated expression of genes involved in fatty acid catabolism. These transcriptional changes led to increased FAO and were associated with enhanced activation of PPARα through a mechanism requiring the catalytic AMPKα2 subunit and SIRT1, two known activators of PPARα. By contrast, p16 overexpression was associated with triglyceride accumulation and increased lipid droplet numbers in vitro, and decreased ketogenesis and hepatic mitochondrial activity in vivo Finally, gene expression analysis of liver samples from obese patients revealed a negative correlation between CDKN2A expression and PPARA and its target genes. Our findings demonstrate that p16 represses hepatic lipid catabolism during fasting and may thus participate in the preservation of metabolic flexibility.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Cyclin-Dependent Kinase Inhibitor p16/metabolism , Fatty Acids/metabolism , Liver/metabolism , Mitochondria, Liver/metabolism , PPAR alpha/metabolism , Signal Transduction , Sirtuin 1/metabolism , AMP-Activated Protein Kinases/genetics , Animals , Cyclin-Dependent Kinase Inhibitor p16/genetics , Fatty Acids/genetics , Genome-Wide Association Study , Humans , Lipid Droplets/metabolism , Mice , Mice, Knockout , Mitochondria, Liver/genetics , Obesity/genetics , Obesity/metabolism , Oxidation-Reduction , PPAR alpha/genetics , Sirtuin 1/genetics
7.
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
8.
Sci Rep ; 10(1): 174, 2020 01 13.
Article in English | MEDLINE | ID: mdl-31932631

ABSTRACT

The gut microbiota participates in the control of energy homeostasis partly through fermentation of dietary fibers hence producing short-chain fatty acids (SCFAs), which in turn promote the secretion of the incretin Glucagon-Like Peptide-1 (GLP-1) by binding to the SCFA receptors FFAR2 and FFAR3 on enteroendocrine L-cells. We have previously shown that activation of the nuclear Farnesoid X Receptor (FXR) decreases the L-cell response to glucose. Here, we investigated whether FXR also regulates the SCFA-induced GLP-1 secretion. GLP-1 secretion in response to SCFAs was evaluated ex vivo in murine colonic biopsies and in colonoids of wild-type (WT) and FXR knock-out (KO) mice, in vitro in GLUTag and NCI-H716 L-cells activated with the synthetic FXR agonist GW4064 and in vivo in WT and FXR KO mice after prebiotic supplementation. SCFA-induced GLP-1 secretion was blunted in colonic biopsies from GW4064-treated mice and enhanced in FXR KO colonoids. In vitro FXR activation inhibited GLP-1 secretion in response to SCFAs and FFAR2 synthetic ligands, mainly by decreasing FFAR2 expression and downstream Gαq-signaling. FXR KO mice displayed elevated colonic FFAR2 mRNA levels and increased plasma GLP-1 levels upon local supply of SCFAs with prebiotic supplementation. Our results demonstrate that FXR activation decreases L-cell GLP-1 secretion in response to inulin-derived SCFA by reducing FFAR2 expression and signaling. Inactivation of intestinal FXR using bile acid sequestrants or synthetic antagonists in combination with prebiotic supplementation may be a promising therapeutic approach to boost the incretin axis in type 2 diabetes.


Subject(s)
Colon/metabolism , Fatty Acids, Volatile/pharmacology , Glucagon-Like Peptide 1/antagonists & inhibitors , Microbiota , Receptors, Cytoplasmic and Nuclear/physiology , Animals , Colon/drug effects , Glucagon-Like Peptide 1/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism
9.
JCI Insight ; 3(9)2018 05 03.
Article in English | MEDLINE | ID: mdl-29720572

ABSTRACT

Metabolic stresses such as dietary energy restriction or physical activity exert beneficial metabolic effects. In the liver, endospanin-1 and endospanin-2 cooperatively modulate calorie restriction-mediated (CR-mediated) liver adaptations by controlling growth hormone sensitivity. Since we found CR to induce endospanin protein expression in skeletal muscle, we investigated their role in this tissue. In vivo and in vitro endospanin-2 triggers ERK phosphorylation in skeletal muscle through an autophagy-dependent pathway. Furthermore, endospanin-2, but not endospanin-1, overexpression decreases muscle mitochondrial ROS production, induces fast-to-slow fiber-type switch, increases skeletal muscle glycogen content, and improves glucose homeostasis, ultimately promoting running endurance capacity. In line, endospanin-2-/- mice display higher lipid peroxidation levels, increased mitochondrial ROS production under mitochondrial stress, decreased ERK phosphorylation, and reduced endurance capacity. In conclusion, our results identify endospanin-2 as a potentially novel player in skeletal muscle metabolism, plasticity, and function.


Subject(s)
Energy Metabolism , Membrane Proteins/physiology , Muscle, Skeletal/metabolism , Physical Endurance/physiology , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/physiology , Animals , Autophagy , Caloric Restriction , Cell Plasticity/genetics , Cells, Cultured , Extracellular Signal-Regulated MAP Kinases/metabolism , Female , Humans , Intracellular Signaling Peptides and Proteins , MAP Kinase Signaling System , Male , Membrane Proteins/genetics , Mice , Mitochondria/metabolism , Muscle Fibers, Fast-Twitch/physiology , Muscle Fibers, Slow-Twitch/physiology , Muscle, Skeletal/cytology , Muscle, Skeletal/physiology , Oxidative Stress , Phenotype , Phosphorylation , Physical Exertion , RNA, Messenger/metabolism
10.
J Med Chem ; 60(10): 4185-4211, 2017 05 25.
Article in English | MEDLINE | ID: mdl-28414465

ABSTRACT

The role of the G-protein-coupled bile acid receptor TGR5 in various organs, tissues, and cell types, specifically in intestinal endocrine L-cells and brown adipose tissue, has made it a promising therapeutical target in several diseases, especially type-2 diabetes and metabolic syndrome. However, recent studies have shown deleterious on-target effects of systemic TGR5 agonists. To avoid these systemic effects while stimulating glucagon-like peptide-1 (GLP-1) secreting enteroendocrine L-cells, we have designed TGR5 agonists with low intestinal permeability. In this article, we describe their synthesis, characterization, and biological evaluation. Among them, compound 24 is a potent GLP-1 secretagogue, has low effect on gallbladder volume, and improves glucose homeostasis in a preclinical murine model of diet-induced obesity and insulin resistance, making the proof of concept of the potential of topical intestinal TGR5 agonists as therapeutic agents in type-2 diabetes.


Subject(s)
Glucagon-Like Peptide 1/metabolism , Glucose/metabolism , Hypoglycemic Agents/chemistry , Hypoglycemic Agents/pharmacology , Imidazoles/chemistry , Imidazoles/pharmacology , Receptors, G-Protein-Coupled/agonists , Amination , Animals , Caco-2 Cells , Diabetes Mellitus, Type 2/drug therapy , Diabetes Mellitus, Type 2/metabolism , Glucose Tolerance Test , Humans , Hypoglycemic Agents/pharmacokinetics , Imidazoles/pharmacokinetics , Intestinal Absorption , Intestinal Mucosa/metabolism , Mice, Inbred C57BL , Receptors, G-Protein-Coupled/metabolism
11.
Arterioscler Thromb Vasc Biol ; 36(12): 2324-2333, 2016 12.
Article in English | MEDLINE | ID: mdl-27758768

ABSTRACT

OBJECTIVE: Although initially seemingly paradoxical because of the lack of nucleus, platelets possess many transcription factors that regulate their function through DNA-independent mechanisms. These include the farnesoid X receptor (FXR), a member of the superfamily of ligand-activated transcription factors, that has been identified as a bile acid receptor. In this study, we show that FXR is present in human platelets and FXR ligands, GW4064 and 6α-ethyl-chenodeoxycholic acid, modulate platelet activation nongenomically. APPROACH AND RESULTS: FXR ligands inhibited the activation of platelets in response to stimulation of collagen or thrombin receptors, resulting in diminished intracellular calcium mobilization, secretion, fibrinogen binding, and aggregation. Exposure to FXR ligands also reduced integrin αIIbß3 outside-in signaling and thereby reduced the ability of platelets to spread and to stimulate clot retraction. FXR function in platelets was found to be associated with the modulation of cyclic guanosine monophosphate levels in platelets and associated downstream inhibitory signaling. Platelets from FXR-deficient mice were refractory to the actions of FXR agonists on platelet function and cyclic nucleotide signaling, firmly linking the nongenomic actions of these ligands to the FXR. CONCLUSIONS: This study provides support for the ability of FXR ligands to modulate platelet activation. The atheroprotective effects of GW4064, with its novel antiplatelet effects, indicate FXR as a potential target for the prevention of atherothrombotic disease.


Subject(s)
Blood Platelets/drug effects , Chenodeoxycholic Acid/analogs & derivatives , Hemostasis/drug effects , Isoxazoles/pharmacology , Platelet Activation/drug effects , Platelet Aggregation Inhibitors/pharmacology , Receptors, Cytoplasmic and Nuclear/agonists , Thrombosis/prevention & control , Animals , Blood Platelets/metabolism , Calcium Signaling/drug effects , Chenodeoxycholic Acid/pharmacology , Cyclic GMP/blood , Disease Models, Animal , Dose-Response Relationship, Drug , Fibrinogen/metabolism , Genotype , Humans , Ligands , Mice, Inbred C57BL , Mice, Knockout , Phenotype , Platelet Aggregation/drug effects , Platelet Glycoprotein GPIIb-IIIa Complex/metabolism , Receptors, Cytoplasmic and Nuclear/blood , Receptors, Cytoplasmic and Nuclear/deficiency , Receptors, Cytoplasmic and Nuclear/genetics , Thrombosis/blood , Time Factors
12.
Nat Commun ; 6: 7629, 2015 Jul 02.
Article in English | MEDLINE | ID: mdl-26134028

ABSTRACT

Bile acids are signalling molecules, which activate the transmembrane receptor TGR5 and the nuclear receptor FXR. BA sequestrants (BAS) complex bile acids in the intestinal lumen and decrease intestinal FXR activity. The BAS-BA complex also induces glucagon-like peptide-1 (GLP-1) production by L cells which potentiates ß-cell glucose-induced insulin secretion. Whether FXR is expressed in L cells and controls GLP-1 production is unknown. Here, we show that FXR activation in L cells decreases proglucagon expression by interfering with the glucose-responsive factor Carbohydrate-Responsive Element Binding Protein (ChREBP) and GLP-1 secretion by inhibiting glycolysis. In vivo, FXR deficiency increases GLP-1 gene expression and secretion in response to glucose hence improving glucose metabolism. Moreover, treatment of ob/ob mice with the BAS colesevelam increases intestinal proglucagon gene expression and improves glycaemia in a FXR-dependent manner. These findings identify the FXR/GLP-1 pathway as a new mechanism of BA control of glucose metabolism and a pharmacological target for type 2 diabetes.


Subject(s)
Enteroendocrine Cells/metabolism , Glucagon-Like Peptide 1/genetics , Intestinal Mucosa/metabolism , RNA, Messenger/metabolism , Receptors, Cytoplasmic and Nuclear/genetics , Animals , Anticholesteremic Agents/pharmacology , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , Bile Acids and Salts/metabolism , Blood Glucose/metabolism , Colesevelam Hydrochloride/pharmacology , Colon/cytology , Colon/metabolism , Diet, High-Fat , Glucagon-Like Peptide 1/metabolism , Glycolysis , Humans , Ileum/cytology , Ileum/metabolism , Insulin/metabolism , Insulin Secretion , Insulin-Secreting Cells/metabolism , Intestines/cytology , Jejunum/cytology , Jejunum/metabolism , Mice , Mice, Knockout , Mice, Obese , Nuclear Proteins/metabolism , Obesity/genetics , Obesity/metabolism , Proglucagon/drug effects , Proglucagon/genetics , Proglucagon/metabolism , Receptors, G-Protein-Coupled/genetics , Sequestering Agents/pharmacology , Signal Transduction , Transcription Factors/metabolism
13.
Transgenic Res ; 24(5): 803-12, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26091792

ABSTRACT

Conditional gene knockout technology is a powerful tool to study the function of a gene in a specific tissue, organ or cell lineage. The most commonly used procedure applies the Cre-LoxP strategy, where the choice of the Cre driver promoter is critical to determine the efficiency and specificity of the system. However, a considered choice of an appropriate promoter does not always protect against the risk of unwanted recombination and the consequent deletion of the gene in other tissues than the desired one(s), due to phenomena of non-specific activation of the Cre transgene. Furthermore, the causes of these phenomena are not completely understood and this can potentially affect every strain of Cre-mice. In our study on the deletion of a same gene in two different tissues, we show that the incidence rate of non-specific recombination in unwanted tissues depends on the Cre driver strain, ranging from 100%, rendering it useless (aP2-Cre strain), to ~5%, which is still compatible with their use (RIP-Cre strain). The use of a simple PCR strategy conceived to detect this occurrence is indispensable when producing a tissue-specific knockout mouse. Therefore, when choosing the Cre-driver promoter, researchers not only have to be careful about its tissue-specificity and timing of activation, but should also include a systematical screening in order to exclude mice in which atypical recombination has occurred and to limit the unnecessary use of laboratory animals in uninterpretable experiments.


Subject(s)
Integrases/genetics , Recombination, Genetic , Alleles , Animals , Germ Cells , Islets of Langerhans/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Polymerase Chain Reaction
14.
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
15.
Diabetes ; 60(7): 1861-71, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21593203

ABSTRACT

OBJECTIVE: Bile acids (BA) participate in the maintenance of metabolic homeostasis acting through different signaling pathways. The nuclear BA receptor farnesoid X receptor (FXR) regulates pathways in BA, lipid, glucose, and energy metabolism, which become dysregulated in obesity. However, the role of FXR in obesity and associated complications, such as dyslipidemia and insulin resistance, has not been directly assessed. RESEARCH DESIGN AND METHODS: Here, we evaluate the consequences of FXR deficiency on body weight development, lipid metabolism, and insulin resistance in murine models of genetic and diet-induced obesity. RESULTS: FXR deficiency attenuated body weight gain and reduced adipose tissue mass in both models. Surprisingly, glucose homeostasis improved as a result of an enhanced glucose clearance and adipose tissue insulin sensitivity. In contrast, hepatic insulin sensitivity did not change, and liver steatosis aggravated as a result of the repression of ß-oxidation genes. In agreement, liver-specific FXR deficiency did not protect from diet-induced obesity and insulin resistance, indicating a role for nonhepatic FXR in the control of glucose homeostasis in obesity. Decreasing elevated plasma BA concentrations in obese FXR-deficient mice by administration of the BA sequestrant colesevelam improved glucose homeostasis in a FXR-dependent manner, indicating that the observed improvements by FXR deficiency are not a result of indirect effects of altered BA metabolism. CONCLUSIONS: Overall, FXR deficiency in obesity beneficially affects body weight development and glucose homeostasis.


Subject(s)
Glucose/metabolism , Obesity/metabolism , Receptors, Cytoplasmic and Nuclear/deficiency , Adipose Tissue/drug effects , Adipose Tissue/metabolism , Animals , Bile Acids and Salts/blood , Disease Models, Animal , Homeostasis , Hypertriglyceridemia/etiology , Insulin/pharmacology , Insulin Resistance , Lipid Metabolism , Male , Mice , Mice, Obese , Weight Gain/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...