Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 4 de 4
Filter
1.
Mol Metab ; 79: 101861, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38142970

ABSTRACT

OBJECTIVE: The dorsal vagal complex (DVC) of the hindbrain is a major point of integration for central and peripheral signals that regulate a wide variety of metabolic functions to maintain energy balance. The REV-ERB nuclear receptors are important modulators of molecular metabolism, but their role in the DVC has yet to be established. METHODS: Male REV-ERBα/ß floxed mice received stereotaxic injections of a Cre expressing virus to the DVC to create the DVC REV-ERBα/ß double knockout (DVC RDKO). Control littermates received stereotaxic injections to the DVC of a green fluorescent protein expressing virus. Animals were maintained on a normal chow diet or a 60% high-fat diet to observe the metabolic phenotype arising from DVC RDKO under healthy and metabolically stressed conditions. RESULTS: DVC RDKO animals on high-fat diet exhibited increased weight gain compared to control animals maintained on the same diet. Increased weight gain in DVC RDKO animals was associated with decreased basal metabolic rate and dampened signature of brown adipose tissue activity. RDKO decreased gene expression of calcitonin receptor in the DVC and tyrosine hydroxylase in the brown adipose tissue. CONCLUSIONS: These results suggest a previously unappreciated role of REV-ERB nuclear receptors in the DVC for maintaining energy balance and metabolic rate potentially through indirect sympathetic outflow to the brown adipose tissue.


Subject(s)
Adipose Tissue, Brown , Rhombencephalon , Animals , Male , Mice , Adipose Tissue, Brown/metabolism , Diet, High-Fat/adverse effects , Obesity/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Rhombencephalon/metabolism , Weight Gain
2.
Nat Commun ; 15(1): 5394, 2024 Jun 25.
Article in English | MEDLINE | ID: mdl-38918428

ABSTRACT

Adipose tissue macrophages (ATMs) influence obesity-associated metabolic dysfunction, but the mechanisms by which they do so are not well understood. We show that miR-6236 is a bona fide miRNA that is secreted by ATMs during obesity. Global or myeloid cell-specific deletion of miR-6236 aggravates obesity-associated adipose tissue insulin resistance, hyperglycemia, hyperinsulinemia, and hyperlipidemia. miR-6236 augments adipocyte insulin sensitivity by inhibiting translation of negative regulators of insulin signaling, including PTEN. The human genome harbors a miR-6236 homolog that is highly expressed in the serum and adipose tissue of obese people. hsa-MIR-6236 expression negatively correlates with hyperglycemia and glucose intolerance, and positively correlates with insulin sensitivity. Together, our findings establish miR-6236 as an ATM-secreted miRNA that potentiates adipocyte insulin signaling and protects against metabolic dysfunction during obesity.


Subject(s)
Adipocytes , Hyperglycemia , Insulin Resistance , Insulin , MicroRNAs , Obesity , PTEN Phosphohydrolase , Signal Transduction , MicroRNAs/metabolism , MicroRNAs/genetics , Obesity/metabolism , Obesity/genetics , Animals , Adipocytes/metabolism , Hyperglycemia/metabolism , Hyperglycemia/genetics , Humans , Insulin/metabolism , Insulin Resistance/genetics , Mice , Male , PTEN Phosphohydrolase/metabolism , PTEN Phosphohydrolase/genetics , Mice, Inbred C57BL , Macrophages/metabolism , Adipose Tissue/metabolism , Myeloid Cells/metabolism , Mice, Knockout , Hyperinsulinism/metabolism , Hyperinsulinism/genetics
3.
Nat Metab ; 6(5): 825-836, 2024 May.
Article in English | MEDLINE | ID: mdl-38622413

ABSTRACT

Nuclear receptor corepressors (NCoRs) function in multiprotein complexes containing histone deacetylase 3 (HDAC3) to alter transcriptional output primarily through repressive chromatin remodelling at target loci1-5. In the liver, loss of HDAC3 causes a marked hepatosteatosis largely because of de-repression of genes involved in lipid metabolism6,7; however, the individual roles and contribution of other complex members to hepatic and systemic metabolic regulation are unclear. Here we show that adult loss of both NCoR1 and NCoR2 (double knockout (KO)) in hepatocytes phenocopied the hepatomegalic fatty liver phenotype of HDAC3 KO. In addition, double KO livers exhibited a dramatic reduction in glycogen storage and gluconeogenic gene expression that was not observed with hepatic KO of individual NCoRs or HDAC3, resulting in profound fasting hypoglycaemia. This surprising HDAC3-independent activation function of NCoR1 and NCoR2 is due to an unexpected loss of chromatin accessibility on deletion of NCoRs that prevented glucocorticoid receptor binding and stimulatory effect on gluconeogenic genes. These studies reveal an unanticipated, non-canonical activation function of NCoRs that is required for metabolic health.


Subject(s)
Gluconeogenesis , Histone Deacetylases , Liver , Mice, Knockout , Nuclear Receptor Co-Repressor 1 , Nuclear Receptor Co-Repressor 2 , Receptors, Glucocorticoid , Gluconeogenesis/genetics , Animals , Receptors, Glucocorticoid/metabolism , Receptors, Glucocorticoid/genetics , Nuclear Receptor Co-Repressor 1/metabolism , Nuclear Receptor Co-Repressor 1/genetics , Mice , Histone Deacetylases/metabolism , Histone Deacetylases/genetics , Nuclear Receptor Co-Repressor 2/metabolism , Nuclear Receptor Co-Repressor 2/genetics , Liver/metabolism , Hepatocytes/metabolism , Nuclear Receptor Coactivator 2/metabolism , Nuclear Receptor Coactivator 2/genetics
4.
Cell Metab ; 36(8): 1764-1778.e9, 2024 Aug 06.
Article in English | MEDLINE | ID: mdl-38889724

ABSTRACT

Deficiency of the epigenome modulator histone deacetylase 3 (HDAC3) in brown adipose tissue (BAT) impairs the ability of mice to survive in near-freezing temperatures. Here, we report that short-term exposure to mild cold temperature (STEMCT: 15°C for 24 h) averted lethal hypothermia of mice lacking HDAC3 in BAT (HDAC3 BAT KO) exposed to 4°C. STEMCT restored the induction of the thermogenic coactivator PGC-1α along with UCP1 at 22°C, which is greatly impaired in HDAC3-deficient BAT, and deletion of either UCP1 or PGC-1α prevented the protective effect of STEMCT. Remarkably, this protection lasted for up to 7 days. Transcriptional activator C/EBPß was induced by short-term cold exposure in mouse and human BAT and, uniquely, remained high for 7 days following STEMCT. Adeno-associated virus-mediated knockdown of BAT C/EBPß in HDAC3 BAT KO mice erased the persistent memory of STEMCT, revealing the existence of a C/EBPß-dependent and HDAC3-independent cold-adaptive epigenomic memory.


Subject(s)
Adipose Tissue, Brown , Cold Temperature , Histone Deacetylases , Mice, Knockout , Animals , Adipose Tissue, Brown/metabolism , Histone Deacetylases/metabolism , Mice , Humans , Thermogenesis/genetics , Mice, Inbred C57BL , CCAAT-Enhancer-Binding Protein-beta/metabolism , Uncoupling Protein 1/metabolism , Uncoupling Protein 1/genetics , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/genetics , Male , Epigenomics , Epigenesis, Genetic
SELECTION OF CITATIONS
SEARCH DETAIL