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










Publication year range
1.
Nat Commun ; 12(1): 7045, 2021 12 02.
Article in English | MEDLINE | ID: mdl-34857762

ABSTRACT

Enhancer activation is essential for cell-type specific gene expression during cellular differentiation, however, how enhancers transition from a hypoacetylated "primed" state to a hyperacetylated-active state is incompletely understood. Here, we show SET domain-containing 5 (SETD5) forms a complex with NCoR-HDAC3 co-repressor that prevents histone acetylation of enhancers for two master adipogenic regulatory genes Cebpa and Pparg early during adipogenesis. The loss of SETD5 from the complex is followed by enhancer hyperacetylation. SETD5 protein levels were transiently increased and rapidly degraded prior to enhancer activation providing a mechanism for the loss of SETD5 during the transition. We show that induction of the CDC20 co-activator of the ubiquitin ligase leads to APC/C mediated degradation of SETD5 during the transition and this operates as a molecular switch that facilitates adipogenesis.


Subject(s)
Adipogenesis/genetics , CCAAT-Enhancer-Binding Proteins/genetics , Histone Deacetylases/genetics , Methyltransferases/genetics , Nuclear Receptor Co-Repressor 1/genetics , PPAR gamma/genetics , 3T3-L1 Cells , Acetylation , Anaphase-Promoting Complex-Cyclosome/genetics , Anaphase-Promoting Complex-Cyclosome/metabolism , Animals , CCAAT-Enhancer-Binding Proteins/metabolism , Cdc20 Proteins/genetics , Cdc20 Proteins/metabolism , Enhancer Elements, Genetic , Gene Expression Regulation , HEK293 Cells , Histone Deacetylases/metabolism , Histones/genetics , Histones/metabolism , Humans , Methyltransferases/metabolism , Mice , Mice, Nude , Nuclear Receptor Co-Repressor 1/metabolism , PPAR gamma/metabolism , Protein Binding , Proteolysis , Sf9 Cells , Signal Transduction
2.
PLoS One ; 11(11): e0164716, 2016.
Article in English | MEDLINE | ID: mdl-27835635

ABSTRACT

How the hypothalamus transmits hunger information to other brain regions to govern whole brain function to orchestrate feeding behavior has remained largely unknown. Our present study suggests the importance of a recently found lateral hypothalamic neuropeptide, QRFP, in this signaling. Qrfp-/- mice were hypophagic and lean, and exhibited increased anxiety-like behavior, and were hypoactive in novel circumstances as compared with wild type littermates. They also showed decreased wakefulness time in the early hours of the dark period. Histological studies suggested that QRFP neurons receive rich innervations from neurons in the arcuate nucleus which is a primary region for sensing the body's metabolic state by detecting levels of leptin, ghrelin and glucose. These observations suggest that QRFP is an important mediator that acts as a downstream mediator of the arcuate nucleus and regulates feeding behavior, mood, wakefulness and activity.


Subject(s)
Anxiety/genetics , Arcuate Nucleus of Hypothalamus/metabolism , Feeding Behavior , Neurons/metabolism , Peptides/genetics , Wakefulness/physiology , Animals , Anxiety/metabolism , Anxiety/physiopathology , Arcuate Nucleus of Hypothalamus/physiopathology , Eating/physiology , Gene Expression , Ghrelin/genetics , Ghrelin/metabolism , Glucose/metabolism , Intercellular Signaling Peptides and Proteins , Leptin/genetics , Leptin/metabolism , Locomotion , Male , Mice , Mice, Knockout , Neurons/pathology , Peptides/deficiency , Signal Transduction
3.
Nat Commun ; 6: 7052, 2015 May 07.
Article in English | MEDLINE | ID: mdl-25948511

ABSTRACT

Histone 3 lysine 9 (H3K9) demethylase JMJD1A regulates ß-adrenergic-induced systemic metabolism and body weight control. Here we show that JMJD1A is phosphorylated at S265 by protein kinase A (PKA), and this is pivotal to activate the ß1-adrenergic receptor gene (Adrb1) and downstream targets including Ucp1 in brown adipocytes (BATs). Phosphorylation of JMJD1A by PKA increases its interaction with the SWI/SNF nucleosome remodelling complex and DNA-bound PPARγ. This complex confers ß-adrenergic-induced rapid JMJD1A recruitment to target sites and facilitates long-range chromatin interactions and target gene activation. This rapid gene induction is dependent on S265 phosphorylation but not on demethylation activity. Our results show that JMJD1A has two important roles in regulating hormone-stimulated chromatin dynamics that modulate thermogenesis in BATs. In one role, JMJD1A is recruited to target sites and functions as a cAMP-responsive scaffold that facilitates long-range chromatin interactions, and in the second role, JMJD1A demethylates H3K9 di-methylation.


Subject(s)
Chromatin/metabolism , Chromosomal Proteins, Non-Histone/metabolism , Jumonji Domain-Containing Histone Demethylases/metabolism , Thermogenesis , Transcription Factors/metabolism , 3T3-L1 Cells , Adipose Tissue, Brown/metabolism , Amino Acid Sequence , Animals , Cyclic AMP-Dependent Protein Kinases/metabolism , Enhancer Elements, Genetic/genetics , Gene Expression Regulation , Genome , HeLa Cells , Humans , Jumonji Domain-Containing Histone Demethylases/chemistry , Jumonji Domain-Containing Histone Demethylases/genetics , Mice , Molecular Sequence Data , PPAR gamma/metabolism , Phosphorylation , Phosphoserine/metabolism , Promoter Regions, Genetic , Receptors, Adrenergic, beta/metabolism , Receptors, Adrenergic, beta-1/genetics , Receptors, Adrenergic, beta-1/metabolism , Thermogenesis/genetics , Transcription, Genetic
4.
J Biol Chem ; 290(7): 4163-77, 2015 Feb 13.
Article in English | MEDLINE | ID: mdl-25533466

ABSTRACT

Polycomb repressive complex 1 (PRC1) plays an essential role in the epigenetic repression of gene expression during development and cellular differentiation via multiple effector mechanisms, including ubiquitination of H2A and chromatin compaction. However, whether it regulates the stepwise progression of adipogenesis is unknown. Here, we show that FBXL10/KDM2B is an anti-adipogenic factor that is up-regulated during the early phase of 3T3-L1 preadipocyte differentiation and in adipose tissue in a diet-induced model of obesity. Interestingly, inhibition of adipogenesis does not require the JmjC demethylase domain of FBXL10, but it does require the F-box and leucine-rich repeat domains, which we show recruit a noncanonical polycomb repressive complex 1 (PRC1) containing RING1B, SKP1, PCGF1, and BCOR. Knockdown of either RING1B or SKP1 prevented FBXL10-mediated repression of 3T3-L1 preadipocyte differentiation indicating that PRC1 formation mediates the inhibitory effect of FBXL10 on adipogenesis. Using ChIP-seq, we show that FBXL10 recruits RING1B to key specific genomic loci surrounding the key cell cycle and the adipogenic genes Cdk1, Uhrf1, Pparg1, and Pparg2 to repress adipogenesis. These results suggest that FBXL10 represses adipogenesis by targeting a noncanonical PRC1 complex to repress key genes (e.g. Pparg) that control conversion of pluripotent cells into the adipogenic lineage.


Subject(s)
Adipocytes/metabolism , Adipogenesis/physiology , F-Box Proteins/metabolism , Jumonji Domain-Containing Histone Demethylases/metabolism , Polycomb Repressive Complex 1/metabolism , 3T3-L1 Cells , Adipocytes/cytology , Animals , Biomarkers/metabolism , Blotting, Western , Cell Cycle , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Proliferation , Chromatin Immunoprecipitation , F-Box Proteins/antagonists & inhibitors , F-Box Proteins/genetics , Gene Expression Profiling , Histones/metabolism , Immunoenzyme Techniques , Immunoprecipitation , Jumonji Domain-Containing Histone Demethylases/antagonists & inhibitors , Jumonji Domain-Containing Histone Demethylases/genetics , Leucine-Rich Repeat Proteins , Mice , Mice, Inbred C57BL , Oligonucleotide Array Sequence Analysis , PPAR gamma/genetics , PPAR gamma/metabolism , Polycomb Repressive Complex 1/genetics , Protein Isoforms , Protein Structure, Tertiary , Proteins/genetics , Proteins/metabolism , RNA, Messenger/genetics , RNA, Small Interfering/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Ubiquitination
5.
Sci Rep ; 4: 5412, 2014 Jun 24.
Article in English | MEDLINE | ID: mdl-24958459

ABSTRACT

Macrophages are important for maintaining intestinal immune homeostasis. Here, we show that PPARß/δ (peroxisome proliferator-activated receptor ß/δ) directly regulates CD300a in macrophages that express the immunoreceptor tyrosine based-inhibitory motif (ITIM)-containing receptor. In mice lacking CD300a, high-fat diet (HFD) causes chronic intestinal inflammation with low numbers of intestinal lymph capillaries and dramatically expanded mesenteric lymph nodes. As a result, these mice exhibit triglyceride malabsorption and reduced body weight gain on HFD. Peritoneal macrophages from Cd300a-/- mice on HFD are classically M1 activated. Activation of toll-like receptor 4 (TLR4)/MyD88 signaling by lipopolysaccharide (LPS) results in prolonged IL-6 secretion in Cd300a-/- macrophages. Bone marrow transplantation confirmed that the phenotype originates from CD300a deficiency in leucocytes. These results identify CD300a-mediated inhibitory signaling in macrophages as a critical regulator of intestinal immune homeostasis.


Subject(s)
Intestines/immunology , PPAR delta/immunology , PPAR-beta/immunology , Receptors, Immunologic/immunology , Animals , Antigens, CD/genetics , Antigens, CD/immunology , Antigens, CD/metabolism , Cell Line, Tumor , Diet, High-Fat/adverse effects , HEK293 Cells , Humans , Inflammation/etiology , Inflammation/genetics , Inflammation/immunology , Interleukin-6/immunology , Interleukin-6/metabolism , Intestinal Mucosa/metabolism , Lipopolysaccharides/immunology , Lipopolysaccharides/pharmacology , Macrophages/immunology , Macrophages/metabolism , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Oligonucleotide Array Sequence Analysis , PPAR delta/genetics , PPAR delta/metabolism , PPAR-beta/genetics , PPAR-beta/metabolism , Receptors, Immunologic/genetics , Receptors, Immunologic/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/drug effects , Signal Transduction/genetics , Signal Transduction/immunology , Toll-Like Receptor 4/immunology , Toll-Like Receptor 4/metabolism , Transcriptome/genetics , Transcriptome/immunology , Weight Gain/genetics , Weight Gain/immunology
6.
Genes Cells ; 14(8): 991-1001, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19624751

ABSTRACT

Histone H3 lysine 9 (H3K9) methylation is a crucial epigenetic mark of heterochromatin formation and transcriptional silencing. Recent studies demonstrated that most covalent histone lysine modifications are reversible and the jumonji C (JmjC)-domain-containing proteins have been shown to possess such demethylase activities. However, there is little information available on the biological roles of histone lysine demethylation in intact animal model systems. JHDM2A (JmjC-domain-containing histone demethylase 2A, also known as JMJD1A) catalyses removal of H3K9 mono- and dimethylation through iron and alpha-ketoglutarate dependent oxidative reactions. Here, we demonstrate that JHDM2a also regulates metabolic genes related to energy homeostasis including anti-adipogenesis, regulation of fat storage, glucose transport and type 2 diabetes. Mice deficient in JHDM2a (JHDM2a-/-) develop adult onset obesity, hypertriglyceridemia, hypercholesterolemia, hyperinsulinemia and hyperleptinemia, which are hallmarks of metabolic syndrome. JHDM2a-/- mice furthermore exhibit fasted induced hypothermia indicating reduced energy expenditure and also have a higher respiratory quotient indicating less fat utilization for energy production. These observations may explain the obesity phenotype in these mice. Thus, H3K9 demethylase JHDM2a is a crucial regulator of genes involved in energy expenditure and fat storage, which suggests it is a previously unrecognized key regulator of obesity and metabolic syndrome.


Subject(s)
Gene Expression Regulation , Histones/metabolism , Jumonji Domain-Containing Histone Demethylases/genetics , Metabolic Syndrome/genetics , Obesity/genetics , 3T3 Cells , Adipocytes , Animals , Cell Differentiation , Cells, Cultured , Energy Metabolism , Female , Fibroblasts/cytology , Homeostasis , Jumonji Domain-Containing Histone Demethylases/deficiency , Jumonji Domain-Containing Histone Demethylases/metabolism , Male , Metabolic Syndrome/etiology , Metabolic Syndrome/physiopathology , Methylation , Mice , Mice, Knockout , Obesity/etiology , Oxidoreductases, N-Demethylating/genetics , Oxidoreductases, N-Demethylating/metabolism , Phenotype
7.
Mol Cell Biol ; 28(12): 3917-31, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18426912

ABSTRACT

Type 1 iodothyronine deiodinase (Dio1), a selenoenzyme catalyzing the bioactivation of thyroid hormone, is highly expressed in the liver. Dio1 mRNA and enzyme activity levels are markedly reduced in the livers of hepatocyte nuclear factor 4alpha (HNF4alpha)-null mice, thus accounting for its liver-specific expression. Consistent with this deficiency, serum T4 and rT3 concentrations are elevated in these mice compared with those in HNF4alpha-floxed control littermates; however, serum T3 levels are unchanged. Promoter analysis of the mouse Dio1 gene demonstrated that HNF4alpha plays a key role in the transactivation of the mouse Dio1 gene. Deletion and substitution mutation analyses demonstrated that a proximal HNF4alpha site (direct repeat 1 [TGGACAAAGGTGC]; HNF4alpha-RE) is crucial for transactivation of the mouse Dio1 gene by HNF4alpha. Mouse Dio1 is also stimulated by thyroid hormone signaling, but a direct role for thyroid hormone receptor action has not been reported. We also showed that thyroid hormone-inducible Krüppel-like factor 9 (KLF9) stimulates the mouse Dio1 promoter very efficiently through two CACCC sequences that are located on either side of HNF4alpha-RE. Furthermore, KLF9 functions together with HNF4alpha and GATA4 to synergistically activate the mouse Dio1 promoter, suggesting that Dio1 is regulated by thyroid hormone in the mouse through an indirect mechanism requiring prior KLF9 induction. In addition, we showed that physical interactions between the C-terminal zinc finger domain (Cf) of GATA4 and activation function 2 of HNF4alpha and between the basic domain adjacent to Cf of GATA4 and a C-terminal domain of KLF9 are both required for this synergistic response. Taken together, these results suggest that HNF4alpha regulates thyroid hormone homeostasis through transcriptional regulation of the mouse Dio1 gene with GATA4 and KLF9.


Subject(s)
GATA4 Transcription Factor/metabolism , Gene Expression Regulation , Hepatocyte Nuclear Factor 4/physiology , Homeostasis , Iodide Peroxidase/biosynthesis , Kruppel-Like Transcription Factors/metabolism , Thyroid Hormones/metabolism , Animals , Hepatocyte Nuclear Factor 4/metabolism , Humans , Iodide Peroxidase/genetics , Mice , Mice, Transgenic , Models, Biological , Transcriptional Activation
8.
Mol Cell Biol ; 27(12): 4248-60, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17403900

ABSTRACT

Cholesterol homeostasis is maintained by coordinate regulation of cholesterol synthesis and its conversion to bile acids in the liver. The excretion of cholesterol from liver and intestine is regulated by ATP-binding cassette half-transporters ABCG5 and ABCG8. The genes for these two proteins are closely linked and divergently transcribed from a common intergenic promoter region. Here, we identified a binding site for hepatocyte nuclear factor 4alpha (HNF4alpha) in the ABCG5/ABCG8 intergenic promoter, through which HNF4alpha strongly activated the expression of a reporter gene in both directions. The HNF4alpha-responsive element is flanked by two conserved GATA boxes that were also required for stimulation by HNF4alpha. GATA4 and GATA6 bind to the GATA boxes, coexpression of GATA4 and HNF4alpha leads to a striking synergistic activation of both the ABCG5 and the ABCG8 promoters, and binding sites for HNF4alpha and GATA were essential for maximal synergism. We also show that HNF4alpha, GATA4, and GATA6 colocalize in the nuclei of HepG2 cells and that a physical interaction between HNF4alpha and GATA4 is critical for the synergistic response. This is the first demonstration that HNF4alpha acts synergistically with GATA factors to activate gene expression in a bidirectional fashion.


Subject(s)
ATP-Binding Cassette Transporters/metabolism , GATA4 Transcription Factor/metabolism , GATA6 Transcription Factor/metabolism , Hepatocyte Nuclear Factor 4/metabolism , Lipoproteins/metabolism , ATP Binding Cassette Transporter, Subfamily G, Member 5 , ATP Binding Cassette Transporter, Subfamily G, Member 8 , ATP-Binding Cassette Transporters/genetics , Adenoviridae/genetics , Amino Acid Motifs , Amino Acid Sequence , Base Sequence , Binding Sites , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Cell Line , Cell Line, Tumor , Consensus Sequence , Conserved Sequence , GATA4 Transcription Factor/genetics , GATA6 Transcription Factor/genetics , Gene Deletion , Genes, Reporter , Hepatocyte Nuclear Factor 4/chemistry , Humans , Lipoproteins/genetics , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Luciferases/metabolism , Molecular Sequence Data , Oligonucleotide Array Sequence Analysis , Promoter Regions, Genetic , Protein Binding , Protein Structure, Tertiary , RNA Interference , Sequence Homology, Amino Acid , Sequence Homology, Nucleic Acid
9.
J Biol Chem ; 280(45): 37669-80, 2005 Nov 11.
Article in English | MEDLINE | ID: mdl-16148004

ABSTRACT

In obesity-related insulin resistance, pancreatic islets compensate for insulin resistance by increasing secretory capacity. Here, we report the identification of sex-determining region Y-box 6 (SOX6), a member of the high mobility group box superfamily of transcription factors, as a co-repressor for pancreatic-duodenal homeobox factor-1 (PDX1). SOX6 mRNA levels were profoundly reduced by both a long term high fat feeding protocol in normal mice and in genetically obese ob/ob mice on a normal chow diet. Interestingly, we show that SOX6 is expressed in adult pancreatic insulin-producing beta-cells and that overexpression of SOX6 decreased glucose-stimulated insulin secretion, which was accompanied by decreased ATP/ADP ratio, Ca(2+) mobilization, proinsulin content, and insulin gene expression. In a complementary fashion, depletion of SOX6 by small interfering RNAs augmented glucose-stimulated insulin secretion in insulinoma mouse MIN6 and rat INS-1E cells. These effects can be explained by our mechanistic studies that show SOX6 acts to suppress PDX1 stimulation of the insulin II promoter through a direct protein/protein interaction. Furthermore, SOX6 retroviral expression decreased acetylation of histones H3 and H4 in chromatin from the promoter for the insulin II gene, suggesting that SOX6 may decrease PDX1 stimulation through changes in chromatin structure at specific promoters. These results suggest that perturbations in transcriptional regulation that are coordinated through SOX6 and PDX1 in beta-cells may contribute to the beta-cell adaptation in obesity-related insulin resistance.


Subject(s)
DNA-Binding Proteins/metabolism , Down-Regulation , Glucose/pharmacology , High Mobility Group Proteins/metabolism , Homeodomain Proteins/antagonists & inhibitors , Hyperinsulinism/metabolism , Insulin/metabolism , Obesity/metabolism , Trans-Activators/antagonists & inhibitors , Transcription Factors/metabolism , Acetylation , Adenosine Triphosphate/metabolism , Animals , Cell Movement , Chromatin/metabolism , Diet , Dietary Fats/administration & dosage , Dietary Fats/pharmacology , Glucose/metabolism , Histones/metabolism , Homeodomain Proteins/metabolism , Hyperinsulinism/genetics , Insulin/genetics , Insulin Secretion , Islets of Langerhans/metabolism , Mice , Mice, Obese , Mitochondria/metabolism , Obesity/genetics , Protein Structure, Tertiary , RNA, Messenger/metabolism , Repressor Proteins/metabolism , SOXD Transcription Factors , Trans-Activators/metabolism , Transcription, Genetic
10.
Biochem Biophys Res Commun ; 333(3): 778-86, 2005 Aug 05.
Article in English | MEDLINE | ID: mdl-15967412

ABSTRACT

The glucose-induced insulin secretion is fine-tuned by numerous factors. To systematically identify insulinotropic factors, we optimized a primary beta-cell-based functional assay to monitor intracellular Ca2+ flux ([Ca2+]i). By this assay system, we successfully identified several insulinotropic peptides including cholecystokinin, gastrin releasing peptide, vasopressin, and oxytocin from tissue extracts. Screening of an assortment of chemical compounds, we determined three novel insulin secretagogues: N-arachidonylglycine (NAGly), 3beta-(2-diethylamino-ethoxy) androstenone hydrochloride (U18666A), and 4-androstene-3,17-dione. The NAGly increased [Ca2+]i through stimulation of the voltage-dependent Ca2+ channels and it was dependent on extracellular glucose level. On the other hand, U18666A and 4-androstene-3,17-dione increased [Ca2+]i in the presence of K ATP channel opener diazoxide while it was inhibited by the presence of Ca2+ channel blocker nitrendipine, suggesting that their effects are independent of K ATP channel. These unique features will be useful for further development of insulinotropic factors and drugs for treating type 2 diabetes.


Subject(s)
Androstenedione/pharmacology , Androstenes/pharmacology , Arachidonic Acids/pharmacology , Glycine/analogs & derivatives , Insulin/metabolism , Animals , Calcium/metabolism , Glycine/pharmacology , Insulin Secretion , Male , Mice , Mice, Inbred ICR , Rats , Rats, Wistar
11.
J Biol Chem ; 279(17): 16954-62, 2004 Apr 23.
Article in English | MEDLINE | ID: mdl-14960588

ABSTRACT

Acetyl-CoA synthetase 2 (AceCS2) produces acetyl-CoA for oxidation through the citric acid cycle in the mitochondrial matrix. AceCS2 is highly expressed in the skeletal muscle and is robustly induced by fasting. Quantification of AceCS2 transcripts both in C2C12 and human myotubes indicated that fasting-induced AceCS2 gene expression appears to be independent on insulin action. Characterization of 5'-flanking region of the mouse AceCS2 gene demonstrates that Krüppel-like factor 15 (KLF15) plays a key role in the trans-activation of the AceCS2 gene. Deletion and mutation analyses of AceCS2 promoter region revealed that the most proximal KLF site is a curtail site for the trans-activation of the AceCS2 gene by KLF15. Using Sp-null Drosophila SL2 cells, we showed that the combination of KLF15 and Sp1 resulted in a synergistic activation of the AceCS2 promoter. Mutation analyses of three GC-boxes in the AceCS2 promoter indicated that the GC-box, located 8 bases downstream of the most proximal KLF15 site, is the most important GC-box in the synergistic trans-activation of the AceCS2 gene by KLF15 and Sp1. GST pull-down assays showed that KLF15 interacts with Sp1 in vitro. Quantification of various KLF transcripts revealed that 48 h fasting robustly induced the KLF15 transcripts in the skeletal muscle. Together with the trans-activation of the AceCS2 promoter, it is suggested that fasting-induced AceCS2 expression is largely contributed by KLF15. Furthermore, KLF15 overexpression induced the levels of AceCS2 transcripts both in myoblasts and in myotubes, indicating that AceCS2 gene expression in vivo is indeed induced by KLF15.


Subject(s)
Acetate-CoA Ligase/genetics , Nuclear Proteins/physiology , Transcription Factors/physiology , Transcriptional Activation , Acetate-CoA Ligase/metabolism , Amino Acid Motifs , Animals , Base Sequence , Cell Line , Citric Acid Cycle , Cloning, Molecular , DNA Mutational Analysis , DNA, Complementary/metabolism , DNA-Binding Proteins , Drosophila , Gene Deletion , Genes, Reporter , Glutathione Transferase/metabolism , Humans , Insulin/metabolism , Kruppel-Like Transcription Factors , Male , Mice , Mice, Inbred ICR , Models, Genetic , Molecular Sequence Data , Muscle Cells/metabolism , Muscle, Skeletal/metabolism , Nuclear Proteins/genetics , Plasmids/metabolism , Promoter Regions, Genetic , Protein Binding , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Sequence Homology, Amino Acid , Sp1 Transcription Factor/metabolism , Time Factors , Transcription Factors/genetics , Transfection
12.
Proc Natl Acad Sci U S A ; 100(26): 15924-9, 2003 Dec 23.
Article in English | MEDLINE | ID: mdl-14676330

ABSTRACT

In this study, we defined the role of peroxisome proliferator-activated receptor beta/delta (PPARdelta) in metabolic homeostasis by using subtype selective agonists. Analysis of rat L6 myotubes treated with the PPARdelta subtype-selective agonist, GW501516, by the Affymetrix oligonucleotide microarrays revealed that PPARdelta controls fatty acid oxidation by regulating genes involved in fatty acid transport, beta-oxidation, and mitochondrial respiration. Similar PPARdelta-mediated gene activation was observed in the skeletal muscle of GW501516-treated mice. Accordingly, GW501516 treatment induced fatty acid beta-oxidation in L6 myotubes as well as in mouse skeletal muscles. Administration of GW501516 to mice fed a high-fat diet ameliorated diet-induced obesity and insulin resistance, an effect accompanied by enhanced metabolic rate and fatty acid beta-oxidation, proliferation of mitochondria, and a marked reduction of lipid droplets in skeletal muscles. Despite a modest body weight change relative to vehicle-treated mice, GW501516 treatment also markedly improved diabetes as revealed by the decrease in plasma glucose and blood insulin levels in genetically obese ob/ob mice. These data suggest that PPARdelta is pivotal to control the program for fatty acid oxidation in the skeletal muscle, thereby ameliorating obesity and insulin resistance through its activation in obese animals.


Subject(s)
Fatty Acids, Nonesterified/metabolism , Insulin Resistance/physiology , Metabolic Syndrome/prevention & control , Muscle, Skeletal/physiology , Receptors, Cytoplasmic and Nuclear/agonists , Transcription Factors/agonists , Animals , Dimethyl Sulfoxide/pharmacology , Enzymes/genetics , Lipid Metabolism , Liver/physiology , Male , Mice , Mice, Inbred C57BL , Mice, Obese , Muscle, Skeletal/drug effects , Oxidation-Reduction , Rats , Receptors, Cytoplasmic and Nuclear/drug effects , Thiazoles/pharmacology , Transcription Factors/drug effects
13.
J Biol Chem ; 278(13): 11331-6, 2003 Mar 28.
Article in English | MEDLINE | ID: mdl-12509421

ABSTRACT

LDL receptor-related protein 5 (LRP5) plays multiple roles, including embryonic development and bone accrual development. Recently, we demonstrated that LRP5 is also required for normal cholesterol metabolism and glucose-induced insulin secretion. To further define the role of LRP5 in the lipoprotein metabolism, we compared plasma lipoproteins in mice lacking LRP5, apolipoprotein E (apoE), or both (apoE;LRP5 double knockout). On a normal chow diet, the apoE;LRP5 double knockout mice (older than 4 months of age) had approximately 60% higher plasma cholesterol levels compared with the age-matched apoE knockout mice. In contrast, LRP5 deficiency alone had no significant effects on the plasma cholesterol levels. High performance liquid chromatography analysis of plasma lipoproteins revealed that cholesterol levels in the very low density lipoprotein and low density lipoprotein fractions were markedly increased in the apoE;LRP5 double knockout mice. There were no apparent differences in the pattern of apoproteins between the apoE knockout mice and the apoE;LRP5 double knockout mice. The plasma clearance of intragastrically loaded triglyceride was markedly impaired by LRP5 deficiency. The atherosclerotic lesions of the apoE;LRP5 double knockout mice aged 6 months were approximately 3-fold greater than those in the age-matched apoE-knockout mice. Furthermore, histological examination revealed highly advanced atherosclerosis, with remarkable accumulation of foam cells and destruction of the internal elastic lamina in the apoE;LRP5 double knockout mice. These data suggest that LRP5 mediates both apoE-dependent and apoE-independent catabolism of plasma lipoproteins.


Subject(s)
Apolipoproteins E/physiology , Arteriosclerosis/genetics , Dietary Fats/metabolism , Hypercholesterolemia/genetics , Receptors, LDL/physiology , Animals , Apolipoproteins E/genetics , Chromatography, High Pressure Liquid , Electrophoresis, Polyacrylamide Gel , LDL-Receptor Related Proteins , Low Density Lipoprotein Receptor-Related Protein-5 , Mice , Mice, Knockout , Receptors, LDL/genetics
14.
Proc Natl Acad Sci U S A ; 100(1): 229-34, 2003 Jan 07.
Article in English | MEDLINE | ID: mdl-12509515

ABSTRACT

A Wnt coreceptor low-density lipoprotein receptor-related protein 5 (LRP5) plays an essential role in bone accrual and eye development. Here, we show that LRP5 is also required for normal cholesterol and glucose metabolism. The production of mice lacking LRP5 revealed that LRP5 deficiency led to increased plasma cholesterol levels in mice fed a high-fat diet, because of the decreased hepatic clearance of chylomicron remnants. In addition, when fed a normal diet, LRP5-deficient mice showed a markedly impaired glucose tolerance. The LRP5-deficient islets had a marked reduction in the levels of intracellular ATP and Ca(2+) in response to glucose, and thereby glucose-induced insulin secretion was decreased. The intracellular inositol 1,4,5-trisphosphate (IP3) production in response to glucose was also reduced in LRP5-- islets. Real-time PCR analysis revealed a marked reduction of various transcripts for genes involved in glucose sensing in LRP5-- islets. Furthermore, exposure of LRP5++ islets to Wnt-3a and Wnt-5a stimulates glucose-induced insulin secretion and this stimulation was blocked by the addition of a soluble form of Wnt receptor, secreted Frizzled-related protein-1. In contrast, LRP5-deficient islets lacked the Wnt-3a-stimulated insulin secretion. These data suggest that WntLRP5 signaling contributes to the glucose-induced insulin secretion in the islets.


Subject(s)
Cholesterol/blood , Glucose/pharmacology , Hypercholesterolemia/genetics , Insulin/metabolism , Islets of Langerhans/metabolism , Receptors, LDL/genetics , Animals , Blood Glucose/metabolism , Calcium/metabolism , Chylomicrons/metabolism , Dietary Fats , Genes, Essential , Glucose Intolerance/blood , Glucose Intolerance/genetics , Hypercholesterolemia/blood , Inositol 1,4,5-Trisphosphate/metabolism , Insulin/blood , Insulin Secretion , Islets of Langerhans/drug effects , LDL-Receptor Related Proteins , Liver/metabolism , Low Density Lipoprotein Receptor-Related Protein-5 , Mice , Mice, Knockout , Polymerase Chain Reaction , Receptors, LDL/deficiency , Transcription, Genetic
15.
J Biol Chem ; 278(9): 7344-9, 2003 Feb 28.
Article in English | MEDLINE | ID: mdl-12496272

ABSTRACT

By expression cloning using fluorescent-labeled high density lipoprotein (HDL), we isolated two clones that conferred the cell surface binding of HDL. Nucleotide sequence of the two clones revealed that one corresponds to scavenger receptor class B, type 1 (SRBI) and the other encoded a novel protein with 228 amino acids. The primary structure of the newly identified HDL-binding protein resembles GPI-anchored proteins consisting of an N-terminal signal sequence, an acidic region with a cluster of aspartate and glutamate residues, an Ly-6 motif highly conserved among the lymphocyte antigen family, and a C-terminal hydrophobic region. This newly identified HDL-binding protein designated GPI-anchored HDL-binding protein 1 (GPI-HBP1), was susceptible to phosphatidylinositol-specific phospholipase C treatment and binds HDL with high affinity (calculated K(d) = 2-3 microg/ml). Similar to SRBI, GPI-HBP1 mediates selective lipid uptake but not the protein component of HDL. Among various ligands for SRBI, HDL was most preferentially bound to GPI-HBP1. In contrast to SRBI, GPI-HBP1 lacked HDL-dependent cholesterol efflux. The GPI-HBP1 transcripts were detected with the highest levels in heart and, to a much lesser extent, in lung and liver. In situ hybridization revealed the accumulation of GPI-HBP1 transcripts in cardiac muscle cells, hepatic Kupffer cells and sinusoidal endothelium, and bronchial epithelium and alveolar macrophages in the lung.


Subject(s)
Glycosylphosphatidylinositols/metabolism , Lipoproteins, HDL/metabolism , Receptors, Lipoprotein/chemistry , Receptors, Lipoprotein/genetics , Amino Acid Motifs , Amino Acid Sequence , Animals , Blotting, Northern , CHO Cells , Cholesterol/metabolism , Cloning, Molecular , Cricetinae , DNA, Complementary/metabolism , Dose-Response Relationship, Drug , In Situ Hybridization , Kinetics , Kupffer Cells , Ligands , Liver/metabolism , Lung/metabolism , Mice , Molecular Sequence Data , Myocardium/metabolism , Protein Binding , Protein Structure, Tertiary , RNA, Messenger/metabolism , Time Factors , Tissue Distribution , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...