Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 39
Filter
Add more filters

Publication year range
1.
J Hepatol ; 79(4): 898-909, 2023 10.
Article in English | MEDLINE | ID: mdl-37230231

ABSTRACT

BACKGROUND & AIMS: Roux-en-Y gastric bypass (RYGB), the most effective surgical procedure for weight loss, decreases obesity and ameliorates comorbidities, such as non-alcoholic fatty liver (NAFLD) and cardiovascular (CVD) diseases. Cholesterol is a major CVD risk factor and modulator of NAFLD development, and the liver tightly controls its metabolism. How RYGB surgery modulates systemic and hepatic cholesterol metabolism is still unclear. METHODS: We studied the hepatic transcriptome of 26 patients with obesity but not diabetes before and 1 year after undergoing RYGB. In parallel, we measured quantitative changes in plasma cholesterol metabolites and bile acids (BAs). RESULTS: RYGB surgery improved systemic cholesterol metabolism and increased plasma total and primary BA levels. Transcriptomic analysis revealed specific alterations in the liver after RYGB, with the downregulation of a module of genes implicated in inflammation and the upregulation of three modules, one associated with BA metabolism. A dedicated analysis of hepatic genes related to cholesterol homeostasis pointed towards increased biliary cholesterol elimination after RYGB, associated with enhancement of the alternate, but not the classical, BA synthesis pathway. In parallel, alterations in the expression of genes involved in cholesterol uptake and intracellular trafficking indicate improved hepatic free cholesterol handling. Finally, RYGB decreased plasma markers of cholesterol synthesis, which correlated with an improvement in liver disease status after surgery. CONCLUSIONS: Our results identify specific regulatory effects of RYGB on inflammation and cholesterol metabolism. RYGB alters the hepatic transcriptome signature, likely improving liver cholesterol homeostasis. These gene regulatory effects are reflected by systemic post-surgery changes of cholesterol-related metabolites, corroborating the beneficial effects of RYGB on both hepatic and systemic cholesterol homeostasis. IMPACT AND IMPLICATIONS: Roux-en-Y gastric bypass (RYGB) is a widely used bariatric surgery procedure with proven efficacy in body weight management, combatting cardiovascular disease (CVD) and non-alcoholic fatty liver disease (NAFLD). RYGB exerts many beneficial metabolic effects, by lowering plasma cholesterol and improving atherogenic dyslipidemia. Using a cohort of patients undergoing RYGB, studied before and 1 year after surgery, we analyzed how RYGB modulates hepatic and systemic cholesterol and bile acid metabolism. The results of our study provide important insights on the regulation of cholesterol homeostasis after RYGB and open avenues that could guide future monitoring and treatment strategies targeting CVD and NAFLD in obesity.


Subject(s)
Gastric Bypass , Non-alcoholic Fatty Liver Disease , Obesity, Morbid , Humans , Gastric Bypass/methods , Non-alcoholic Fatty Liver Disease/genetics , Non-alcoholic Fatty Liver Disease/surgery , Transcriptome , Obesity/complications , Cholesterol , Homeostasis , Inflammation/complications , Obesity, Morbid/complications
2.
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
3.
Nucleic Acids Res ; 44(22): 10539-10553, 2016 12 15.
Article in English | MEDLINE | ID: mdl-27576532

ABSTRACT

Adaptation to fasting involves both Glucocorticoid Receptor (GRα) and Peroxisome Proliferator-Activated Receptor α (PPARα) activation. Given both receptors can physically interact we investigated the possibility of a genome-wide cross-talk between activated GR and PPARα, using ChIP- and RNA-seq in primary hepatocytes. Our data reveal extensive chromatin co-localization of both factors with cooperative induction of genes controlling lipid/glucose metabolism. Key GR/PPAR co-controlled genes switched from transcriptional antagonism to cooperativity when moving from short to prolonged hepatocyte fasting, a phenomenon coinciding with gene promoter recruitment of phosphorylated AMP-activated protein kinase (AMPK) and blocked by its pharmacological inhibition. In vitro interaction studies support trimeric complex formation between GR, PPARα and phospho-AMPK. Long-term fasting in mice showed enhanced phosphorylation of liver AMPK and GRα Ser211. Phospho-AMPK chromatin recruitment at liver target genes, observed upon prolonged fasting in mice, is dampened by refeeding. Taken together, our results identify phospho-AMPK as a molecular switch able to cooperate with nuclear receptors at the chromatin level and reveal a novel adaptation mechanism to prolonged fasting.


Subject(s)
Adenylate Kinase/metabolism , Chromatin/metabolism , PPAR alpha/physiology , Receptors, Glucocorticoid/physiology , Animals , Base Sequence , Binding Sites , Cells, Cultured , Enhancer Elements, Genetic , Fasting , Hepatocytes/metabolism , Lipid Metabolism , Mice, Inbred C57BL , Mice, Knockout , Protein Transport , Sequence Analysis, DNA , Transcriptional Activation , Transcriptome
4.
J Hepatol ; 63(1): 164-73, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25703085

ABSTRACT

BACKGROUND & AIMS: Peroxisome proliferator-activated receptors (PPARs) have been implicated in non-alcoholic steatohepatitis (NASH) pathogenesis, mainly based on animal data. Gene expression data in NASH patients are scarce. We studied liver PPARα, ß/δ, and γ expression in a large cohort of obese patients assessed for presence of NAFLD at baseline and 1 year follow-up. METHODS: Patients presented to the obesity clinic underwent a hepatic work-up. If NAFLD was suspected, liver biopsy was performed. Gene expression was studied by mRNA quantification. Patients were reassessed after 1 year. RESULTS: 125 patients were consecutively included in the study, of which 85 patients had paired liver biopsy taken at 1 year of follow-up. Liver PPARα expression negatively correlated with the presence of NASH (p=0.001) and with severity of steatosis (p=0.003), ballooning (p=0.001), NASH activity score (p=0.008) and fibrosis (p=0.003). PPARα expression was positively correlated to adiponectin (R(2)=0.345, p=0.010) and inversely correlated to visceral fat (R(2)=-0.343, p<0.001), HOMA IR (R(2)=-0.411, p<0.001) and CK18 (R(2)=-0.233, p=0.012). Liver PPARß/δ and PPARγ expression did not correlate with any histological feature nor with glucose metabolism or serum lipids. At 1 year, correlation of PPARα expression with liver histology was confirmed. In longitudinal analysis, an increase in expression of PPARα and its target genes was significantly associated with histological improvement (p=0.008). CONCLUSION: Human liver PPARα gene expression negatively correlates with NASH severity, visceral adiposity and insulin resistance and positively with adiponectin. Histological improvement is associated with an increase in expression of PPARα and its target genes. These data might suggest that PPARα is a potential therapeutic target in NASH.


Subject(s)
Gene Expression Regulation , Liver/pathology , Non-alcoholic Fatty Liver Disease/genetics , PPAR alpha/genetics , RNA/genetics , Adolescent , Adult , Aged , Biopsy , Female , Follow-Up Studies , Humans , Liver/metabolism , Male , Middle Aged , Non-alcoholic Fatty Liver Disease/metabolism , Non-alcoholic Fatty Liver Disease/therapy , PPAR alpha/biosynthesis , Prospective Studies , Real-Time Polymerase Chain Reaction , Young Adult
5.
Hepatology ; 59(1): 121-9, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24375485

ABSTRACT

UNLABELLED: An independent role of nonalcoholic fatty liver disease (NAFLD) in the development of cardiovascular disease has been suggested, probably mediated through increased levels of prothrombotic factors. Therefore, we examined whether NAFLD is linked to a prothrombotic state, independently of metabolic risk factors in a large single-center cohort of overweight/obese patients. Patients presenting to the obesity clinic underwent a detailed metabolic and liver assessment, including an extensive panel of coagulation factors. If NAFLD was suspected, a liver biopsy was proposed. A series of 273 consecutive patients (65% female) with a liver biopsy were included (age, 44 ± 0.76 years; body mass index: 39.6 ± 0.40 kg/m(2)). Increase in fibrinogen, factor VIII, and von Willebrand factor and decrease in antithrombin III correlated with metabolic features, but not with liver histology. Levels of plasminogen activator inhibitor-1 (PAI-1) increased significantly with increasing severity of steatosis (P < 0.001), lobular inflammation (P < 0.001), ballooning (P = 0.002), and fibrosis (P < 0.001). Patients with nonalcoholic steatohepatitis had significantly higher PAI-1 values than those with normal liver (P < 0.001). In multiple regression, including anthropometric and metabolic parameters, steatosis remained an independent predictor of PAI-1 levels, explaining, together with fasting C-peptide and waist circumference, 21% of the variance in PAI-1. No consistent correlations with histology were found for the other coagulation factors. CONCLUSION: In obesity, NAFLD severity independently contributes to the increase in PAI-1 levels, whereas other coagulation factors are unaltered. This finding might, in part, explain the increased cardiovascular risk associated with NAFLD.


Subject(s)
Fatty Liver/complications , Obesity/complications , Plasminogen Activator Inhibitor 1/blood , Thrombosis/etiology , Adult , Anthropometry , Blood Coagulation , Fatty Liver/blood , Fatty Liver/pathology , Female , Gene Expression , Humans , Liver/pathology , Liver Function Tests , Male , Metabolic Syndrome/blood , Metabolic Syndrome/complications , Middle Aged , Prospective Studies , Regression Analysis , Statistics, Nonparametric
6.
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
7.
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
8.
Diabetologia ; 57(4): 785-96, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24463962

ABSTRACT

AIMS/HYPOTHESIS: Gene polymorphisms of TCF7L2 are associated with increased risk of type 2 diabetes and transcription factor 7-like 2 (TCF7L2) plays a role in hepatic glucose metabolism. We therefore addressed the impact of TCF7L2 isoforms on hepatocyte nuclear factor 4α (HNF4α) and the regulation of gluconeogenesis genes. METHODS: Liver TCF7L2 transcripts were analysed by quantitative PCR in 33 non-diabetic and 31 type 2 diabetic obese individuals genotyped for TCF7L2 rs7903146. To analyse transcriptional regulation by TCF7L2, small interfering RNA transfection, luciferase reporter and co-immunoprecipitation assays were performed in human hepatoma HepG2 cells. RESULTS: In livers of diabetic compared with normoglycaemic individuals, five C-terminal TCF7L2 transcripts showed increased expression. The type 2 diabetes risk allele of rs7903146 positively correlated with TCF7L2 expression in livers from normoglycaemic individuals only. In HepG2 cells, transcript and TCF7L2 protein levels were increased upon incubation in high glucose and insulin. Of the exon 13 transcripts, six were increased in a glucose dose-responsive manner. TCF7L2 transcriptionally regulated 29 genes related to glucose metabolism, including glucose-6-phosphatase. In cultured HepG2 cells, TCF7L2 did not regulate HNF4Α and FOXO1 transcription, but did affect HNF4α protein expression. The TCF7L2 isoforms T6 and T8 (without exon 13 and with exon 15/14, respectively) specifically interacted with HNF4α. CONCLUSIONS/INTERPRETATION: The different levels of expression of alternative C-terminal TCF7L2 transcripts in HepG2 cells, in livers of normoglycaemic individuals carrying the rs7901346 type 2 diabetes risk allele and in livers of diabetic individuals suggest that these transcripts play a role in the pathophysiology of type 2 diabetes. We also report for the first time a protein interaction in HepG2 cells between HNF4α and the T6 and T8 isoforms of TCF7L2, which suggests a distinct role for these specific alternative transcripts.


Subject(s)
Gluconeogenesis/physiology , Hepatocyte Nuclear Factor 4/metabolism , Liver/metabolism , Transcription Factor 7-Like 2 Protein/metabolism , Adult , Blotting, Western , Female , Gluconeogenesis/genetics , Hep G2 Cells , Hepatocyte Nuclear Factor 4/genetics , Humans , Immunoprecipitation , Male , Middle Aged , Protein Binding , Protein Isoforms/genetics , Protein Isoforms/metabolism , Transcription Factor 7-Like 2 Protein/genetics
9.
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
10.
Am J Physiol Endocrinol Metab ; 302(9): E1063-9, 2012 May 15.
Article in English | MEDLINE | ID: mdl-22338072

ABSTRACT

Liver fat is increased in carriers of the minor G allele in rs738409 (I148M amino acid substitution) in patatin-like phospholipase domain-containing 3 (PNPLA3)/adiponutrin. We studied transcriptional regulation of PNPLA3 in immortalized human hepatocytes (IHH) and human hepatoma cells (HuH7) and the impact of PNPLA3 I148M mutant on hepatocyte triglyceride metabolism. Studies in IHH showed that silencing of the carbohydrate response element-binding protein (ChREBP) abolished induction of PNPLA3 mRNA by glucose. Glucose-dependent binding of ChREBP to a newly identified carbohydrate response element in the PNPLA3 promoter was demonstrated by chromatin immunoprecipitation. Adenoviral overexpression of mouse ChREBP in IHH failed to induce PNPLA3 mRNA. [(3)H]acetate or [(3)H]oleate incorporation with 1-h pulse labeling or 18-h [(3)H]oleate labeling in HuH7 cells showed no effect of PNPLA3 I148M on triglyceride (TG) synthesis in the absence of free fatty acid (FFA) loading. Increased [(3)H]oleate accumulation into triglycerides in I148M-expressing cells was observed after 18 h of labeling in the presence of 200 µM FFA-albumin complexes. This was accompanied by increased PNPLA3 protein levels. The rate of hydrolysis of [(3)H]TG during lipid depletion was decreased significantly by PNPLA3 I148M. Our results suggest that PNPLA3 is regulated in human hepatocytes by glucose via ChREBP. PNPLA3 I148M enhances cellular accumulation of [(3)H]TG in the presence of excess FFA, which is known to stabilize PNPLA3 protein. These data do not exclude an effect of PNPLA3 I148M on hepatocyte lipogenesis but show that the mutant increases the stability of triglycerides.


Subject(s)
Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , Glucose/physiology , Hepatocytes/metabolism , Lipase/metabolism , Membrane Proteins/metabolism , Triglycerides/metabolism , Amino Acid Substitution , Cell Line , Humans , Lipogenesis/physiology , Mutation , Statistics, Nonparametric , Transcription, Genetic/physiology
11.
Cytokine ; 59(2): 442-9, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22658783

ABSTRACT

The liver is a major target of injury in obese patients. Non-alcoholic fatty liver disease (NAFLD) is present in 60-90% of obese Americans and can range from simple steatosis to the more severe non-alcoholic steatohepatitis (NASH). The onset of a chronic inflammatory reaction marks the progression from simple steatosis to NASH and the expansion of adipose tissue is strongly associated with angiogenesis. Therefore, we determined the serum concentration of inflammatory [tumor necrosis factor alpha (TNFα) and interleukin 6 (IL6)] and angiogenic [vascular endothelial growth factor A (VEGF)] cytokines and soluble VEGF receptors 1 and 2 (sVEGFR1, sVEGFR2) in the serum of an obese population with simple steatosis and NASH compared to healthy controls. Moreover, we determined the TNFα, IL6, VEGF, VEGFR1 and VEGFR2 gene expression in the liver of these simple steatosis and NASH patients. The population consisted of 30 obese patients, which were diagnosed with simple steatosis and 32 patients with NASH and compared to 30 age-and-sex matched healthy controls. Mean serum TNFα levels were elevated in the serum of simple steatosis and NASH patients compared to healthy controls, reaching significance in NASH patients. IL6 was significantly increased in simple steatosis and NASH patients compared to the healthy controls. VEGF levels were significantly elevated in patients with simple steatosis and borderline significantly elevated in NASH patients compared to the serum levels of healthy control subjects. The concentration of sVEGFR1 was significantly increased in serum of simple steatosis and NASH patients compared to controls. sVEGFR2 concentration was not significantly different in the three groups. TNFα mRNA expression was higher in NASH patients compared to simple steatosis patients. Hepatic gene expression of VEGF, VEGFR1 and VEGFR2 were slightly decreased in NASH patients compared to simple steatosis patients. These data indicate the involvement of inflammatory (TNFα and IL6), angiogenic (VEGF) cytokines and sVEGFR1 in the pathophysiology of NAFLD.


Subject(s)
Angiogenesis Inducing Agents/blood , Angiogenesis Inducing Agents/metabolism , Fatty Liver/blood , Fatty Liver/genetics , Inflammation/blood , Inflammation/genetics , Adult , Biopsy , Case-Control Studies , Fatty Liver/pathology , Female , Gene Expression Regulation , Health , Humans , Inflammation/pathology , Interleukin-6/blood , Linear Models , Liver/metabolism , Liver/pathology , Male , Middle Aged , Non-alcoholic Fatty Liver Disease , Tumor Necrosis Factor-alpha/blood , Vascular Endothelial Growth Factor A/blood , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor Receptor-1/blood , Vascular Endothelial Growth Factor Receptor-1/genetics , Vascular Endothelial Growth Factor Receptor-1/metabolism , Vascular Endothelial Growth Factor Receptor-2/blood , Vascular Endothelial Growth Factor Receptor-2/genetics , Vascular Endothelial Growth Factor Receptor-2/metabolism , Young Adult
12.
Arterioscler Thromb Vasc Biol ; 31(7): 1573-9, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21474829

ABSTRACT

OBJECTIVE: Peroxisome proliferator-activated receptor-α (PPARα) is a ligand-activated transcription factor that controls lipid metabolism and inflammation. PPARα is activated by fibrates, hypolipidemic drugs used in the treatment of dyslipidemia. Previous studies assessing the influence of PPARα agonists on atherosclerosis in mice yielded conflicting results, and the implication of PPARα therein has not been assessed. The human apolipoprotein E2 knock-in (apoE2-KI) mouse is a model of mixed dyslipidemia, atherosclerosis, and nonalcoholic steatohepatitis (NASH). The aim of this study was to analyze, using homo- and heterozygous PPARα-deficient mice, the consequences of quantitative variations of PPARα gene levels and their response to the synthetic PPARα agonist fenofibrate on NASH and atherosclerosis in apoE2-KI mice. METHODS AND RESULTS: Wild-type (+/+), heterozygous (+/-), and homozygous (-/-) PPARα-deficient mice in the apoE2-KI background were generated and subjected to a Western diet supplemented with fenofibrate or not supplemented. Western diet-fed PPARα-/- apoE2-KI mice displayed an aggravation of liver steatosis and inflammation compared with PPARα+/+ and PPARα+/- apoE2-KI mice, indicating a role of PPARα in liver protection. Moreover, PPARα expression was required for the fenofibrate-induced protection against NASH. Interestingly, fenofibrate treatment induced a similar response on hepatic lipid metabolism in PPARα+/+ and PPARα+/- apoE2-KI mice, whereas, for a maximal antiinflammatory response, both alleles of the PPARα gene were required. Surprisingly, atherosclerosis development was not significantly different among PPARα+/+, PPARα+/-, and PPARα-/- apoE2-KI mice. However, PPARα gene level determined both the antiatherosclerotic and vascular antiinflammatory responses to fenofibrate in a dose-dependent manner. CONCLUSIONS: These results demonstrate a necessary but quantitatively different role of PPARα in the modulation of liver metabolism, inflammation, and atherogenesis.


Subject(s)
Aorta/metabolism , Apolipoprotein E2/metabolism , Atherosclerosis/metabolism , Inflammation/metabolism , Lipid Metabolism , Liver/metabolism , PPAR alpha/metabolism , Analysis of Variance , Animals , Anti-Inflammatory Agents/pharmacology , Aorta/drug effects , Aorta/pathology , Apolipoprotein E2/genetics , Atherosclerosis/drug therapy , Atherosclerosis/genetics , Atherosclerosis/pathology , Disease Models, Animal , Fatty Liver/drug therapy , Fatty Liver/genetics , Fatty Liver/metabolism , Fatty Liver/pathology , Female , Fenofibrate/pharmacology , Gene Expression Regulation , Gene Knock-In Techniques , Heterozygote , Homozygote , Humans , Hypolipidemic Agents/pharmacology , Inflammation/drug therapy , Inflammation/genetics , Inflammation/pathology , Lipid Metabolism/drug effects , Lipid Metabolism/genetics , Lipids/blood , Liver/drug effects , Liver/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Non-alcoholic Fatty Liver Disease , PPAR alpha/agonists , PPAR alpha/genetics
13.
Arterioscler Thromb Vasc Biol ; 31(3): 513-9, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21183731

ABSTRACT

OBJECTIVE: Hypertriglyceridemia and fatty liver are common in patients with type 2 diabetes, but the factors connecting alterations in glucose metabolism with plasma and liver lipid metabolism remain unclear. Apolipoprotein CIII (apoCIII), a regulator of hepatic and plasma triglyceride metabolism, is elevated in type 2 diabetes. In this study, we analyzed whether apoCIII is affected by altered glucose metabolism. METHODS AND RESULTS: Liver-specific insulin receptor-deficient mice display lower hepatic apoCIII mRNA levels than controls, suggesting that factors other than insulin regulate apoCIII in vivo. Glucose induces apoCIII transcription in primary rat hepatocytes and immortalized human hepatocytes via a mechanism involving the transcription factors carbohydrate response element-binding protein and hepatocyte nuclear factor-4α. ApoCIII induction by glucose is blunted by treatment with agonists of farnesoid X receptor and peroxisome proliferator-activated receptor-α but not liver X receptor, ie, nuclear receptors controlling triglyceride metabolism. Moreover, in obese humans, plasma apoCIII protein correlates more closely with plasma fasting glucose and glucose excursion after oral glucose load than with insulin. CONCLUSIONS: Glucose induces apoCIII transcription, which may represent a mechanism linking hyperglycemia, hypertriglyceridemia, and cardiovascular disease in type 2 diabetes.


Subject(s)
Apolipoprotein C-III/genetics , Diabetes Complications/etiology , Diabetes Mellitus, Type 2/complications , Dyslipidemias/etiology , Glucose/metabolism , Hepatocytes/metabolism , Transcriptional Activation , Adult , Animals , Apolipoprotein C-III/blood , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/genetics , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , Blood Glucose/metabolism , Cells, Cultured , Diabetes Complications/genetics , Diabetes Complications/metabolism , Diabetes Mellitus, Type 2/genetics , Diabetes Mellitus, Type 2/metabolism , Dyslipidemias/genetics , Dyslipidemias/metabolism , Heat-Shock Proteins/agonists , Heat-Shock Proteins/metabolism , Hepatocyte Nuclear Factor 4/genetics , Hepatocyte Nuclear Factor 4/metabolism , Humans , Insulin/blood , Liver X Receptors , Male , Mice , Mice, Knockout , Middle Aged , Obesity/blood , Orphan Nuclear Receptors/genetics , Orphan Nuclear Receptors/metabolism , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha , Promoter Regions, Genetic , RNA Interference , RNA, Messenger/metabolism , RNA-Binding Proteins/agonists , RNA-Binding Proteins/metabolism , Rats , Receptor, Insulin/deficiency , Receptor, Insulin/genetics , Receptors, Cytoplasmic and Nuclear/agonists , Receptors, Cytoplasmic and Nuclear/metabolism , Time Factors , Transcription Factors/agonists , Transcription Factors/metabolism , Transfection , Up-Regulation
14.
Proc Natl Acad Sci U S A ; 106(18): 7397-402, 2009 May 05.
Article in English | MEDLINE | ID: mdl-19376972

ABSTRACT

Glucocorticoid receptor alpha (GRalpha) and peroxisome proliferator-activated receptor alpha (PPARalpha) are transcription factors with clinically important immune-modulating properties. Either receptor can inhibit cytokine gene expression, mainly through interference with nuclear factor kappaB (NF-kappaB)-driven gene expression. The present work aimed to investigate a functional cross-talk between PPARalpha- and GRalpha-mediated signaling pathways. Simultaneous activation of PPARalpha and GRalpha dose-dependently enhances transrepression of NF-kappaB-driven gene expression and additively represses cytokine production. In sharp contrast and quite unexpectedly, PPARalpha agonists inhibit the expression of classical glucocorticoid response element (GRE)-driven genes in a PPARalpha-dependent manner, as demonstrated by experiments using PPARalpha wild-type and knockout mice. The underlying mechanism for this transcriptional antagonism relies on a PPARalpha-mediated interference with the recruitment of GRalpha, and concomitantly of RNA polymerase II, to GRE-driven gene promoters. Finally, the biological relevance of this phenomenon is underscored by the observation that treatment with the PPARalpha agonist fenofibrate prevents glucocorticoid-induced hyperinsulinemia of mice fed a high-fat diet. Taken together, PPARalpha negatively interferes with GRE-mediated GRalpha activity while potentiating its antiinflammatory effects, thus providing a rationale for combination therapy in chronic inflammatory disorders.


Subject(s)
Glucocorticoids/pharmacology , NF-kappa B/metabolism , PPAR alpha/metabolism , Receptors, Glucocorticoid/metabolism , Transcriptional Activation , Animals , Cell Line, Tumor , Dietary Fats/administration & dosage , Fenofibrate/pharmacology , Gene Expression/drug effects , Hepatocytes/drug effects , Hepatocytes/metabolism , Humans , Hyperinsulinism/chemically induced , Hyperinsulinism/prevention & control , Mice , Mice, Knockout , PPAR alpha/agonists , PPAR alpha/antagonists & inhibitors , PPAR alpha/genetics , Promoter Regions, Genetic , Receptors, Glucocorticoid/agonists
15.
J Biol Chem ; 285(47): 36759-67, 2010 Nov 19.
Article in English | MEDLINE | ID: mdl-20851881

ABSTRACT

The bile acid receptor farnesoid X receptor (FXR) is expressed in adipose tissue, but its function remains poorly defined. Peroxisome proliferator-activated receptor-γ (PPARγ) is a master regulator of adipocyte differentiation and function. The aim of this study was to analyze the role of FXR in adipocyte function and to assess whether it modulates PPARγ action. Therefore, we tested the responsiveness of FXR-deficient mice (FXR(-/-)) and cells to the PPARγ activator rosiglitazone. Our results show that genetically obese FXR(-/-)/ob/ob mice displayed a resistance to rosiglitazone treatment. In vitro, rosiglitazone treatment did not induce normal adipocyte differentiation and lipid droplet formation in FXR(-/-) mouse embryonic fibroblasts (MEFs) and preadipocytes. Moreover, FXR(-/-) MEFs displayed both an increased lipolysis and a decreased de novo lipogenesis, resulting in reduced intracellular triglyceride content, even upon PPARγ activation. Retroviral-mediated FXR re-expression in FXR(-/-) MEFs restored the induction of adipogenic marker genes during rosiglitazone-forced adipocyte differentiation. The expression of Wnt/ß-catenin pathway and target genes was increased in FXR(-/-) adipose tissue and MEFs. Moreover, the expression of several endogenous inhibitors of this pathway was decreased early during the adipocyte differentiation of FXR(-/-) MEFs. These findings demonstrate that FXR regulates adipocyte differentiation and function by regulating two counteracting pathways of adipocyte differentiation, the PPARγ and Wnt/ß-catenin pathways.


Subject(s)
Adipocytes/cytology , Cell Differentiation , PPAR gamma/metabolism , Receptors, Cytoplasmic and Nuclear/physiology , Wnt Proteins/metabolism , beta Catenin/metabolism , Adipocytes/drug effects , Adipocytes/metabolism , Animals , Biomarkers/metabolism , Blotting, Western , Cells, Cultured , Drug Resistance , Embryo, Mammalian/cytology , Embryo, Mammalian/drug effects , Embryo, Mammalian/metabolism , Female , Fibroblasts/drug effects , Fibroblasts/metabolism , Fusion Regulatory Protein-1 , Gene Expression Profiling , Humans , Hypoglycemic Agents/pharmacology , Kidney/cytology , Kidney/drug effects , Kidney/metabolism , Lipolysis , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Obese , Oligonucleotide Array Sequence Analysis , PPAR gamma/genetics , RNA, Messenger/genetics , Reverse Transcriptase Polymerase Chain Reaction , Rosiglitazone , Signal Transduction , Thiazolidinediones/pharmacology , Wnt Proteins/genetics , beta Catenin/genetics
16.
Curr Diab Rep ; 11(3): 160-6, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21431855

ABSTRACT

Type 2 diabetes (T2D) is a growing health problem worldwide, but the currently available strategies for therapy and prevention are insufficient. Recent observations indicate that bile acid homeostasis is altered in T2D. Bile acids are metabolic regulators that act as signaling molecules through receptor-dependent and -independent pathways. The most prominent signaling molecules mediating bile acid signaling are the nuclear receptor farnesoid X receptor (FXR) and the membrane receptor TGR5. Both are implicated in the regulation of lipid, glucose, and energy metabolism. Dysregulation of these pathways might contribute to the development of T2D and associated complications. Interestingly, data from studies with bile acids or bile acid sequestrants indicate that the manipulation of bile acid homeostasis might be an attractive approach for T2D therapy. In this review, we summarize the mechanisms of bile acid-mediated metabolic control that might be relevant in the pathogenesis of T2D.


Subject(s)
Bile Acids and Salts/metabolism , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/physiopathology , Animals , Dyslipidemias/metabolism , Dyslipidemias/physiopathology , Humans , Obesity/metabolism , Obesity/physiopathology
17.
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
19.
FEBS Lett ; 582(6): 949-55, 2008 Mar 19.
Article in English | MEDLINE | ID: mdl-18298956

ABSTRACT

The purpose of this study was to determine whether bile acids (BAs) modulate hepatic pro-protein convertase subtilisin/kexin 9 (PCSK9) gene expression. Immortalized human hepatocytes were treated with various BAs. Chenodeoxycholic acid (CDCA) treatment specifically decreased both PCSK9 mRNA and protein contents. Moreover, activation of the BA-activated farnesoid X receptor (FXR) by its synthetic specific agonist GW4064 also decreased PCSK9 expression. Of functional relevance, coadministration of CDCA counteracted the statin-induced PCSK9 expression, leading to a potentiation of LDL receptor activity. This study suggests that a transcriptional repression of PCSK9 by CDCA or FXR agonists may potentiate the hypolipidemic effect of statins.


Subject(s)
Chenodeoxycholic Acid/pharmacology , DNA-Binding Proteins/agonists , Hepatocytes/drug effects , Receptors, Cytoplasmic and Nuclear/agonists , Serine Endopeptidases/genetics , Transcription Factors/agonists , Transcription, Genetic/drug effects , Hepatocytes/enzymology , Humans , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Isoxazoles/pharmacology , Pravastatin/pharmacology , Proprotein Convertase 9 , Proprotein Convertases , RNA, Messenger/antagonists & inhibitors , RNA, Messenger/metabolism , Receptors, LDL/agonists , Receptors, LDL/metabolism , Serine Endopeptidases/metabolism
20.
Biochem J ; 407(3): 461-9, 2007 Nov 01.
Article in English | MEDLINE | ID: mdl-17655523

ABSTRACT

FXR (farnesoid X receptor), a nuclear receptor activated by BAs (bile acids), is a key factor in the regulation of BA, lipid and carbohydrate metabolism. The recent development of synthetic FXR agonists and knockout mouse models has accelerated the discovery of FXR target genes. In the present study, we identify human fetuin-B as a novel FXR target gene. Treatment with FXR agonists increased fetuin-B expression in human primary hepatocytes and in the human hepatoma HepG2 cell line. In contrast, fetuin-B expression was not responsive to FXR agonist treatment in murine primary hepatocytes. Fetuin-B induction by FXR agonist was abolished upon FXR knockdown by siRNA (small interfering RNA). In addition to the previously described P1 promoter, we show that the human fetuin-B gene is also transcribed from an alternative promoter, termed P2. Transcription via the P2 promoter was induced by FXR agonist treatment, whereas P1 promoter activity was not sensitive to FXR agonist treatment. Two putative FXR-response elements [IR-1 (inverted repeat-1)] were identified in the region -1.6 kb upstream of the predicted P2 transcriptional start site. Both motifs bound FXR-RXR (retinoid X receptor) complexes in vitro and were activated by FXR in transient transfection reporter assays. Mutations in the IR-1 sites abolished FXR-RXR binding and activation. Taken together, these results identify human fetuin-B as a new FXR target gene in human hepatocytes.


Subject(s)
DNA-Binding Proteins/physiology , Hepatocytes/metabolism , Receptors, Cytoplasmic and Nuclear/physiology , Transcription Factors/physiology , Up-Regulation/physiology , alpha-Fetoproteins/biosynthesis , alpha-Fetoproteins/genetics , Alternative Splicing , Amino Acid Sequence , Animals , Base Sequence , Cell Line, Tumor , DNA-Binding Proteins/agonists , Fetuin-B , Gene Expression Profiling , Humans , Mice , Molecular Sequence Data , Promoter Regions, Genetic , Receptors, Cytoplasmic and Nuclear/agonists , Transcription Factors/agonists
SELECTION OF CITATIONS
SEARCH DETAIL