Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
EBioMedicine ; 11: 101-117, 2016 Sep.
Article in English | MEDLINE | ID: mdl-27568222

ABSTRACT

RORα is a member of the nuclear receptor (NR) superfamily and analysis of the (global) RORα-deficient mouse model revealed this NR has a role in glycemic control and fat deposition. Therefore, we generated an adipose-specific RORα 'gain of function' mouse model under the control of the fatty acid binding protein 4 (FABP4) promoter to elucidate the function of RORα in adipose tissue. The Tg-FABP4-RORα4 mice demonstrated a shift in fat distribution to non-adipose tissues when challenged with a high fat diet (HFD). Specifically, we observed a subcutaneous lipodystrophy, accompanied by hepatomegaly (fatty liver/mild portal fibrosis) and splenomegaly; in a background of decreased weight gain and total body fat after HFD. Moreover, we observed significantly higher fasting blood glucose and impaired clearance of glucose in Tg-FABP4-RORα4 mice. Genome wide expression and qPCR profiling analysis identified: (i) subcutaneous adipose specific decreases in the expression of genes involved in fatty acid biosynthesis, lipid droplet expansion and glycemic control, and (ii) the fibrosis pathway as the most significant pathway [including dysregulation of the collagen/extracellular matrix (ECM) pathways] in subcutaneous adipose and liver. The pathology presented in the Tg-FABP4-RORα4 mice is reminiscent of human metabolic disease (associated with aberrant ECM expression) highlighting the therapeutic potential of this NR.


Subject(s)
Adipose Tissue/metabolism , Adiposity/genetics , Blood Glucose , Gene Expression , Nuclear Receptor Subfamily 1, Group F, Member 1/genetics , Adipose Tissue/immunology , Adipose Tissue/pathology , Adiposity/immunology , Animals , Biomarkers , Cluster Analysis , Extracellular Matrix/metabolism , Fibrosis , Gene Expression Profiling , Genotype , Glucose Tolerance Test , Hepatomegaly/genetics , Hepatomegaly/metabolism , Hepatomegaly/pathology , Humans , Insulin Resistance , Lipid Metabolism , Lipids/blood , Liver/metabolism , Liver/pathology , Mice , Mice, Transgenic , Organ Specificity , Phenotype , Splenomegaly/genetics , Splenomegaly/metabolism , Splenomegaly/pathology , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Transgenes , Weight Gain
2.
IUBMB Life ; 65(8): 657-64, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23846999

ABSTRACT

Glycogen is an energy storage depot for the mammalian species. This review focuses on recent developments that have identified the role of nuclear hormone receptor (NR) signaling and epigenomic control in the regulation of important genes that modulate glycogen metabolism. Specifically, new studies have revealed that the NR4A subgroup (of the NR superfamily) are strikingly sensitive to beta-adrenergic stimulation in skeletal muscle, and transgenic studies in mice have revealed the expression of these NRs affects endurance and glycogen levels in muscle. Furthermore, other studies have demonstrated that one of the NR coregulator class of enzymes that mediate chromatin remodeling, the histone methyltransferases (for example, protein arginine methyltransferase 4) regulates the expression of several genes involved in glycogen metabolism and glycogen storage diseases in skeletal muscle. Importantly, NRs and histone methyltransferases, have the potential to be pharmacologically exploited and may provide novel targets in the quest to treat disorders of glycogen storage.


Subject(s)
Epigenomics , Glycogen/metabolism , Muscle, Skeletal/metabolism , Receptors, Cytoplasmic and Nuclear/physiology , Animals , Glycogen Storage Disease/physiopathology , Mice , Nuclear Receptor Subfamily 4, Group A, Member 1/physiology , Protein-Arginine N-Methyltransferases/metabolism
3.
Biochem J ; 444(2): 323-31, 2012 Jun 01.
Article in English | MEDLINE | ID: mdl-22428544

ABSTRACT

CARM1 (co-activator-associated arginine methyltransferase 1)/PRMT4 (protein arginine methyltransferase 4), functions as a co-activator for transcription factors that are regulators of muscle fibre type and oxidative metabolism, including PGC (peroxisome-proliferator-activated receptor γ co-activator)-1α and MEF2 (myocyte enhancer factor 2). We observed significantly higher Prmt4 mRNA expression in comparison with Prmt1-Prmt6 mRNA expression in mouse muscle (in vitro and in vivo). Transfection of Prmt4 siRNA (small interfering RNA) into mouse skeletal muscle C2C12 cells attenuated PRMT4 mRNA and protein expression. We subsequently performed additional qPCR (quantitative PCR) analysis (in the context of metabolism) to examine the effect of Prmt4 siRNA expression on >200 critical genes that control (and are involved in) lipid, glucose and energy homoeostasis, and circadian rhythm. This analysis revealed a strikingly specific metabolic expression footprint, and revealed that PRMT4 is necessary for the expression of genes involved in glycogen metabolism in skeletal muscle cells. Prmt4 siRNA expression selectively suppressed the mRNAs encoding Gys1 (glycogen synthase 1), Pgam2 (muscle phosphoglycerate mutase 2) and Pygm (muscle glycogen phosphorylase). Significantly, PGAM, PYGM and GYS1 deficiency in humans causes glycogen storage diseases type X, type V/McArdle's disease and type 0 respectively. Attenuation of PRMT4 was also associated with decreased expression of the mRNAs encoding AMPK (AMP-activated protein kinase) α2/γ3 (Prkaa2 and Prkag3) and p38 MAPK (mitogen-activated protein kinase), previously implicated in Wolff-Parkinson-White syndrome and Pompe Disease (glycogen storage disease type II). Furthermore, stable transfection of two PRMT4-site-specific (methyltransferase deficient) mutants (CARM1/PRMT4 VLD and CARM1E267Q) significantly repressed the expression of Gys1, Pgam2 and AMPKγ3. Finally, in concordance, we observed increased and decreased glycogen levels in PRMT4 (native)- and VLD (methylation deficient mutant)-transfected skeletal muscle cells respectively. This demonstrated that PRMT4 expression and the associated methyltransferase activity is necessary for the gene expression programme involved in glycogen metabolism and human glycogen storage diseases.


Subject(s)
Gene Expression Regulation/genetics , Glycogen/biosynthesis , Glycogen/genetics , Muscle, Skeletal/cytology , Muscle, Skeletal/metabolism , Protein-Arginine N-Methyltransferases/physiology , Animals , Cells, Cultured , Humans , Mice , Mice, Inbred C57BL , Protein-Arginine N-Methyltransferases/biosynthesis , Protein-Arginine N-Methyltransferases/genetics
4.
Physiol Genomics ; 43(4): 213-27, 2011 Feb 24.
Article in English | MEDLINE | ID: mdl-21119012

ABSTRACT

We demonstrate that chicken ovalbumin upstream promoter-transcription factor II (COUP-TFII) mRNA is more abundantly expressed (than COUP-TFI mRNA) in skeletal muscle C2C12 cells and in (type I and II) skeletal muscle tissue from C57BL/10 mice. Consequently, we have utilized the ABI TaqMan Low Density Array (TLDA) platform to analyze gene expression changes specifically attributable to ectopic COUP-TFII (relative to vector only) expression in muscle cells. Utilizing a TLDA-based platform and 5 internal controls, we analyze the entire NR superfamily, 96 critical metabolic genes, and 48 important myogenic regulatory genes on the TLDA platform utilizing 5 internal controls. The low density arrays were analyzed by rigorous statistical analysis (with Genorm normalization, Bioconductor R, and the Empirical Bayes statistic) using the (integromics) statminer software. In addition, we validated the differentially expressed patho-physiologically relevant gene (identified on the TLDA platform) glucose transporter type 4 (Glut4). We demonstrated that COUP-TFII expression increased the steady state levels of Glut4 mRNA and protein, while ectopic expression of truncated COUP-TFII lacking helix 12 (COUP-TFΔH12) reduced Glut4 mRNA expression in C2C12 cells. Moreover, COUP-TFII expression trans-activated the Glut4 promoter (-997/+3), and ChIP analysis identified selective recruitment of COUP-TFII to a region encompassing a highly conserved SP1 binding site (in mouse, rat, and human) at nt positions -131/-118. Mutation of the SpI site ablated COUP-TFII mediated trans-activation of the Glut4 promoter. In conclusion, this study demonstrates that in skeletal muscle cells, COUP-TFII regulates several nuclear hormone receptors, and critical metabolic and muscle specific genes.


Subject(s)
COUP Transcription Factor II/metabolism , Gene Expression Regulation, Developmental , Muscle Cells/metabolism , Muscle Development/genetics , Muscle, Skeletal/cytology , Animals , COUP Transcription Factor II/genetics , Cell Line , Glucose Transporter Type 4/genetics , Glucose Transporter Type 4/metabolism , Humans , Male , Mice , Muscle Fibers, Skeletal/metabolism , Oligonucleotide Array Sequence Analysis , Promoter Regions, Genetic/genetics , Protein Binding , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Transcriptional Activation/genetics
5.
Mol Cell Endocrinol ; 315(1-2): 146-52, 2010 Feb 05.
Article in English | MEDLINE | ID: mdl-19631715

ABSTRACT

Estrogen-related receptors (ERRs) are constitutively active orphan nuclear receptors. Natural ligands have not been identified, however, recent reports have demonstrated the synthetic phenolic acyl hydrazone, GSK4716, functions as a selective ERRbeta/gamma agonist. We demonstrate that ERRbeta is transiently induced, and ERRgamma is dramatically induced (and accumulates) in a differentiation-dependent manner in skeletal muscle cells. Treatment of differentiated skeletal muscle cells with the ERRbeta/gamma agonist (GSK4716) produced a significant increase in the expression of GRalpha (isoform D) protein. Quantitative RT-PCR (Q-RT-PCR) analysis after treatment with GSK4716, revealed induction of the mRNAs encoding the glucocorticoid receptor (GR), 11beta-hydroxysteroid dehydrogenase type 1 (11beta-HSD1), the enzyme that converts inactive cortisone to cortisol and hexose-6-phosphate dehydrogenase expression (H6PDH) that stimulates oxoreduction by 11beta-HSD1. Candidate based expression profiling also demonstrated the mRNAs encoding characterized GR target genes, including C/EBP, ApoD and Monoamine oxidase-A (MAO-A) are induced in GSK4716 treated cells. In concordance with these observations, siRNA-mediated suppression of the mRNA encoding ERRgamma (but not ERRalpha and beta) attenuated the expression of mRNAs encoding GR, 11betaHSD1 and GR target gene(s). Similarly, treatment with the ERRgamma (and ERalpha) antagonist diethylstilbestrol (DES) suppressed glucocorticoid responsive gene expression in skeletal muscle cells. Interestingly, we observed that GSK4716 trans-activated GRE-TK-LUC in a GR-dependent manner. This study highlights the regulatory crosstalk between ERRgamma and GR signaling in skeletal muscle cells, and suggests the ERRgamma agonist modulates the expression of critical genes that control GR signaling and glucocorticoid sensitive gene expression.


Subject(s)
Gene Expression Regulation/drug effects , Glucocorticoids/metabolism , Hydrazines/pharmacology , Muscle, Skeletal , Receptors, Estrogen/agonists , Receptors, Estrogen/metabolism , Receptors, Glucocorticoid/metabolism , Animals , Cell Differentiation/physiology , Cell Line , Diethylstilbestrol/pharmacology , Estrogens, Non-Steroidal/pharmacology , Mice , Muscle, Skeletal/cytology , Muscle, Skeletal/drug effects , Muscle, Skeletal/physiology , RNA, Small Interfering/metabolism , Receptors, Estrogen/genetics , Receptors, Glucocorticoid/genetics , Signal Transduction/physiology
6.
Mol Cell Endocrinol ; 309(1-2): 101-8, 2009 Oct 15.
Article in English | MEDLINE | ID: mdl-19465082

ABSTRACT

The nuclear hormone receptor (NR) 4A subgroup of orphan nuclear receptors includes three members, Nur77 (NR4A1), Nurr1 (NR4A2) and Nor-1 (NR4A3). Previously we have identified the rapid and robust (in vitro and in vivo) induction of the NR4A subgroup following beta-adrenergic stimulation in mouse skeletal muscle. This was concomitant with changes in the expression of genes involved in the regulation of nutrient metabolism. We have isolated mouse tissue of cardiovascular, endocrine and gastrointestinal origin at 1, 4, 8 and 24h after a single intraperitoneal injection of the beta-adrenergic agonist, isoprenaline. We similarly identified the significant induction (between 1 and 4h) of the NR4A genes in many of these tissues. Moreover, we have utilized TaqMan((R)) Low Density Arrays to determine the beta-adrenergic-sensitive metabolic gene expression in liver, white adipose and heart. In summary, cross-talk between beta-adrenergic and NR4A signaling occurs in several tissues, and is accompanied by modulation of metabolic gene expression.


Subject(s)
Gene Expression Profiling , Gene Expression Regulation , Organ Specificity/genetics , Receptors, Adrenergic, beta/metabolism , Receptors, Cytoplasmic and Nuclear/genetics , Signal Transduction , Animals , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Gene Expression Regulation/drug effects , Glucose/metabolism , Isoproterenol/pharmacology , Lipid Metabolism/drug effects , Lipid Metabolism/genetics , Male , Mice , Mice, Inbred C57BL , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Nuclear Receptor Subfamily 4, Group A, Member 1 , Nuclear Receptor Subfamily 4, Group A, Member 2 , Organ Specificity/drug effects , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Receptors, Steroid/genetics , Receptors, Steroid/metabolism , Receptors, Thyroid Hormone/genetics , Receptors, Thyroid Hormone/metabolism , Signal Transduction/drug effects , Transcription Factors/genetics , Transcription Factors/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...