Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
FEBS Lett ; 594(17): 2923-2930, 2020 09.
Article in English | MEDLINE | ID: mdl-32767856

ABSTRACT

We previously reported the involvement of protein arginine methyltransferase 1 (PRMT1) in adipocyte thermogenesis. Here, we investigate the effects of PRMT1 inhibitors on thermogenesis. Unexpectedly, we find that the PRMT1 inhibitor TC-E 5003 (TC-E) induces the thermogenic properties of primary murine and human subcutaneous adipocytes. TC-E treatment upregulates the expression of Ucp1 and Fgf21 significantly and activates protein kinase A signaling and lipolysis in primary subcutaneous adipocytes from both mouse and humans. We further find that the thermogenic effects of TC-E are independent of PRMT1 and beta-adrenergic receptors. Our data indicate that TC-E exerts strong effects on murine and human subcutaneous adipocytes by activating beige adipocytes via PKA signaling.


Subject(s)
Benzeneacetamides/pharmacology , Cyclic AMP-Dependent Protein Kinases/genetics , Enzyme Inhibitors/pharmacology , Protein-Arginine N-Methyltransferases/genetics , Repressor Proteins/genetics , Signal Transduction/drug effects , Thermogenesis/drug effects , Adipocytes, Beige/cytology , Adipocytes, Beige/drug effects , Adipocytes, Beige/enzymology , Adipocytes, Brown/cytology , Adipocytes, Brown/drug effects , Adipocytes, Brown/enzymology , Adipocytes, White/cytology , Adipocytes, White/drug effects , Adipocytes, White/enzymology , Animals , Cyclic AMP-Dependent Protein Kinases/metabolism , Fibroblast Growth Factors/genetics , Fibroblast Growth Factors/metabolism , Gene Expression Regulation , Humans , Lipolysis/drug effects , Lipolysis/genetics , Mice , Primary Cell Culture , Protein-Arginine N-Methyltransferases/antagonists & inhibitors , Protein-Arginine N-Methyltransferases/metabolism , Receptors, Adrenergic, beta/genetics , Receptors, Adrenergic, beta/metabolism , Repressor Proteins/antagonists & inhibitors , Repressor Proteins/metabolism , Thermogenesis/genetics , Uncoupling Protein 1/genetics , Uncoupling Protein 1/metabolism
2.
Endocrinology ; 160(12): 2773-2786, 2019 12 01.
Article in English | MEDLINE | ID: mdl-31555811

ABSTRACT

Protein arginine methyltransferases (PRMTs) are enzymes that regulate the evolutionarily conserved process of arginine methylation. It has been reported that PRMTs are involved in many metabolic regulatory pathways. However, until now, their roles in adipocyte function, especially browning and thermogenesis, have not been evaluated. Even though Prmt1 adipocyte-specific-deleted mice (Prmt1fl/flAQcre) appeared normal at basal level, following cold exposure or ß-adrenergic stimulation, impaired induction of the thermogenic program was observed in both the interscapular brown adipose tissue and inguinal white adipose tissue of Prmt1fl/flAQcre mice compared with littermate controls. Different splicing variants of Prmt1 have been reported. Among them, PRMT1 variant 1 and PRMT1 variant 2 (PRMT1V2) are well conserved between humans and mice. Both variants contribute to the activation of thermogenic fat, with PRMT1V2 playing a more dominant role. Mechanistic studies using cultured murine and human adipocytes revealed that PRMT1V2 mediates thermogenic fat activation through PGC1α, a transcriptional coactivator that has been shown to play a key role in mitochondrial biogenesis. To our knowledge, our data are the first to demonstrate that PRMT1 plays a regulatory role in thermogenic fat function. These findings suggest that modulating PRMT1 activity may represent new avenues to regulate thermogenic fat and mediate energy homeostasis. This function is conserved in human primary adipocytes, suggesting that further investigation of this pathway may ultimately lead to therapeutic strategies against human obesity and associated metabolic disorders.


Subject(s)
Adipocytes, Beige/enzymology , Adipocytes, Brown/enzymology , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism , Protein-Arginine N-Methyltransferases/metabolism , Thermogenesis , Acclimatization , Animals , Female , Gene Expression Regulation , Humans , Male , Mice , Primary Cell Culture
3.
Mol Cell ; 76(3): 500-515.e8, 2019 11 07.
Article in English | MEDLINE | ID: mdl-31422874

ABSTRACT

Diet-induced obesity can be caused by impaired thermogenesis of beige adipocytes, the brown-like adipocytes in white adipose tissue (WAT). Promoting brown-like features in WAT has been an attractive therapeutic approach for obesity. However, the mechanism underlying beige adipocyte formation is largely unknown. N-α-acetyltransferase 10 protein (Naa10p) catalyzes N-α-acetylation of nascent proteins, and overexpression of human Naa10p is linked to cancer development. Here, we report that both conventional and adipose-specific Naa10p deletions in mice result in increased energy expenditure, thermogenesis, and beige adipocyte differentiation. Mechanistically, Naa10p acetylates the N terminus of Pgc1α, which prevents Pgc1α from interacting with Pparγ to activate key genes, such as Ucp1, involved in beige adipocyte function. Consistently, fat tissues of obese human individuals show higher NAA10 expression. Thus, Naa10p-mediated N-terminal acetylation of Pgc1α downregulates thermogenic gene expression, making inhibition of Naa10p enzymatic activity a potential strategy for treating obesity.


Subject(s)
Adipocytes, Beige/enzymology , Adipose Tissue, Beige/enzymology , N-Terminal Acetyltransferase A/metabolism , N-Terminal Acetyltransferase E/metabolism , Obesity/enzymology , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism , Protein Processing, Post-Translational , Thermogenesis , Acetylation , Adipose Tissue, Beige/physiopathology , Adiposity , Adolescent , Adult , Aged , Animals , Case-Control Studies , Diet, High-Fat , Disease Models, Animal , Energy Metabolism , Female , HEK293 Cells , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Middle Aged , N-Terminal Acetyltransferase A/deficiency , N-Terminal Acetyltransferase A/genetics , N-Terminal Acetyltransferase E/deficiency , N-Terminal Acetyltransferase E/genetics , NIH 3T3 Cells , Obesity/genetics , Obesity/physiopathology , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/genetics , Phenotype , Signal Transduction , Young Adult
4.
J Endocrinol ; 239(2): 153-165, 2018 11 01.
Article in English | MEDLINE | ID: mdl-30121575

ABSTRACT

Numerous studies have suggested that beige adipocyte abundance is correlated with improved metabolic performance, but direct evidence showing that beige adipocyte expansion protects animals from the development of obesity is missing. Previously, we have described that the liver kinase b1 (LKB1) regulates beige adipocyte renaissance in subcutaneous inguinal white adipose tissue (iWAT) through a class IIa histone deacetylase 4 (HDAC4)-dependent mechanism. This study investigates the physiological impact of persistent beige adipocyte renaissance in energy homeostasis in mice. Here we present that the transgenic mice H4-TG, overexpressing constitutively active HDAC4 in adipocytes, showed beige adipocyte expansion in iWAT at room temperature. H4-TG mice exhibited increased energy expenditure due to beige adipocyte expansion. They also exhibited reduced adiposity under both normal chow and high-fat diet (HFD) feeding conditions. Specific ablation of beige adipocytes reversed the protection against HFD-induced obesity in H4-TG mice. Taken together, our results directly demonstrate that beige adipocyte expansion regulates adiposity in mice and targeting beige adipocyte renaissance may present a novel strategy to tackle obesity in humans.


Subject(s)
Adipocytes, Beige/enzymology , Adipose Tissue, White/cytology , Adiposity , Energy Metabolism , Histone Deacetylases/metabolism , Animals , Male , Mice , Mice, Transgenic
5.
Biochem Biophys Res Commun ; 501(4): 851-857, 2018 07 02.
Article in English | MEDLINE | ID: mdl-29750960

ABSTRACT

Adipocyte differentiation is a tightly regulated process which requires the sequential and organized expression of numerous genes and proteins. Phosphorylation of cytoplasmic proteins and key transcription factors represents a critical regulatory mechanism of the process leading to adipocyte maturation and modulation of associated metabolic pathways. Despite the recognition of the importance of protein phosphorylation in adipocyte biology, relatively little is known about the role of specific kinases in thermogenic (brown or beige) adipocyte differentiation and function. In this study, we demonstrate that the non-receptor protein tyrosine kinase 2 beta (PTK2B) plays a critical role in murine beige adipocyte differentiation. We observed that PTK2B protein expression is associated with beige adipocyte differentiation in cultured, immortalized, inguinal stromal vascular fraction cells. CRISPR/Cas9-mediated knock-out of Ptk2b results in non-differentiating white adipocytes, and differentiated beige adipocytes with significantly reduced thermogenic gene and protein expression, enlarged lipid droplet size, and altered mitochondrial respiration. Together, our data in a cell culture system provides evidence for a role of PTK2B in the differentiation of murine beige adipocytes.


Subject(s)
Adipocytes, Beige/cytology , Adipocytes, Beige/enzymology , Cell Differentiation , Focal Adhesion Kinase 2/metabolism , Adipocytes, White/cytology , Adipocytes, White/metabolism , Adipogenesis/genetics , Animals , Cell Respiration , Cells, Cultured , Male , Mice, Inbred C57BL , Mice, Knockout , Mitochondria/metabolism , Thermogenesis/genetics
6.
Cell Rep ; 19(12): 2451-2461, 2017 06 20.
Article in English | MEDLINE | ID: mdl-28636934

ABSTRACT

Emerging evidence suggests that microbes resident in the human intestine represent a key environmental factor contributing to obesity-associated disorders. Here, we demonstrate that the gut microbiota-initiated trimethylamine N-oxide (TMAO)-generating pathway is linked to obesity and energy metabolism. In multiple clinical cohorts, systemic levels of TMAO were observed to strongly associate with type 2 diabetes. In addition, circulating TMAO levels were associated with obesity traits in the different inbred strains represented in the Hybrid Mouse Diversity Panel. Further, antisense oligonucleotide-mediated knockdown or genetic deletion of the TMAO-producing enzyme flavin-containing monooxygenase 3 (FMO3) conferred protection against obesity in mice. Complimentary mouse and human studies indicate a negative regulatory role for FMO3 in the beiging of white adipose tissue. Collectively, our studies reveal a link between the TMAO-producing enzyme FMO3 and obesity and the beiging of white adipose tissue.


Subject(s)
Methylamines/blood , Obesity/enzymology , Oxygenases/physiology , Subcutaneous Fat/enzymology , Adipocytes, Beige/enzymology , Animals , Diabetes Mellitus, Type 2/blood , Female , Gene Expression , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Obesity/blood , Obesity/pathology , Subcutaneous Fat/pathology , Subcutaneous Fat/physiopathology
7.
J Nutr Biochem ; 37: 76-82, 2016 11.
Article in English | MEDLINE | ID: mdl-27637001

ABSTRACT

Eicosapentaenoic acid (EPA), a n-3 long-chain polyunsaturated fatty acid, has been reported to have beneficial effects in obesity-associated metabolic disorders. The objective of the present study was to determine the effects of EPA on the regulation of genes involved in lipid metabolism, and the ability of EPA to induce mitochondrial biogenesis and beiging in subcutaneous adipocytes from overweight subjects. Fully differentiated human subcutaneous adipocytes from overweight females (BMI: 28.1-29.8kg/m2) were treated with EPA (100-200 µM) for 24 h. Changes in mRNA expression levels of genes involved in lipogenesis, fatty acid oxidation and mitochondrial biogenesis were determined by qRT-PCR. Mitochondrial content was evaluated using MitoTracker® Green stain. The effects on peroxisome proliferator-activated receptor gamma, co-activator 1 alpha (PGC-1α) and AMP-activated protein kinase (AMPK) were also characterized. EPA down-regulated lipogenic genes expression while up-regulated genes involved in fatty acid oxidation. Moreover, EPA-treated adipocytes showed increased mitochondrial content, accompanied by an up-regulation of nuclear respiratory factor-1, mitochondrial transcription factor A and cytochrome c oxidase IV mRNA expression. EPA also promoted the activation of master regulators of mitochondrial biogenesis such as sirtuin 1, PGC1-α and AMPK. In parallel, EPA induced the expression of genes that typify beige adipocytes such as fat determination factor PR domain containing 16, uncoupling protein 1 and cell death-inducing DFFA-like effector A, T-Box protein 1 and CD137. Our results suggest that EPA induces a remodeling of adipocyte metabolism preventing fat storage and promoting fatty acid oxidation, mitochondrial biogenesis and beige-like markers in human subcutaneous adipocytes from overweight subjects.


Subject(s)
Adipocytes, Beige/metabolism , Adipocytes, White/metabolism , Eicosapentaenoic Acid/metabolism , Gene Expression Regulation, Enzymologic , Mitochondrial Dynamics , Organelle Biogenesis , Subcutaneous Fat, Abdominal/metabolism , Acyl-CoA Oxidase/chemistry , Acyl-CoA Oxidase/genetics , Acyl-CoA Oxidase/metabolism , Adipocytes, Beige/enzymology , Adipocytes, Beige/pathology , Adipocytes, White/enzymology , Adipocytes, White/pathology , Adipogenesis , Biomarkers/metabolism , Body Mass Index , Carnitine O-Palmitoyltransferase/chemistry , Carnitine O-Palmitoyltransferase/genetics , Carnitine O-Palmitoyltransferase/metabolism , Cells, Cultured , Diacylglycerol O-Acyltransferase/antagonists & inhibitors , Diacylglycerol O-Acyltransferase/genetics , Diacylglycerol O-Acyltransferase/metabolism , Energy Metabolism , Fatty Acid Synthases/antagonists & inhibitors , Fatty Acid Synthases/genetics , Fatty Acid Synthases/metabolism , Female , Humans , Lipid Metabolism , Osmolar Concentration , Stearoyl-CoA Desaturase/antagonists & inhibitors , Stearoyl-CoA Desaturase/genetics , Stearoyl-CoA Desaturase/metabolism , Subcutaneous Fat, Abdominal/enzymology , Subcutaneous Fat, Abdominal/pathology
8.
PLoS One ; 11(6): e0157644, 2016.
Article in English | MEDLINE | ID: mdl-27322180

ABSTRACT

Beige adipocytes are special cells situated in the white adipose tissue. Beige adipocytes, lacking thermogenic cues, morphologically look quite similar to regular white adipocytes, but with a markedly different response to adrenalin. White adipocytes respond to adrenergic stimuli by enhancing lipolysis, while in beige adipocytes adrenalin induces mitochondrial biogenesis too. A key step in the differentiation and function of beige adipocytes is the deacetylation of peroxisome proliferator-activated receptor (PPARγ) by SIRT1 and the consequent mitochondrial biogenesis. AMP-activated protein kinase (AMPK) is an upstream activator of SIRT1, therefore we set out to investigate the role of AMPK in beige adipocyte differentiation using human adipose-derived mesenchymal stem cells (hADMSCs) from pericardial adipose tissue. hADMSCs were differentiated to white and beige adipocytes and the differentiation medium of the white adipocytes was supplemented with 100 µM [(2R,3S,4R,5R)-5-(4-Carbamoyl-5-aminoimidazol-1-yl)-3,4-dihydroxyoxolan-2-yl]methyl dihydrogen phosphate (AICAR), a known activator of AMPK. The activation of AMPK with AICAR led to the appearance of beige-like morphological properties in differentiated white adipocytes. Namely, smaller lipid droplets appeared in AICAR-treated white adipocytes in a similar fashion as in beige cells. Moreover, in AICAR-treated white adipocytes the mitochondrial network was more fused than in white adipocytes; a fused mitochondrial system was characteristic to beige adipocytes. Despite the morphological similarities between AICAR-treated white adipocytes and beige cells, functionally AICAR-treated white adipocytes were similar to white adipocytes. We were unable to detect increases in basal or cAMP-induced oxygen consumption rate (a marker of mitochondrial biogenesis) when comparing control and AICAR-treated white adipocytes. Similarly, markers of beige adipocytes such as TBX1, UCP1, CIDEA, PRDM16 and TMEM26 remained the same when comparing control and AICAR-treated white adipocytes. Our data point out that in human pericardial hADMSCs the role of AMPK activation in controlling beige differentiation is restricted to morphological features, but not to actual metabolic changes.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Adipocytes, Beige/cytology , Adipocytes, White/enzymology , Adipose Tissue, White/cytology , Aminoimidazole Carboxamide/analogs & derivatives , Pericardium/cytology , Ribonucleotides/pharmacology , Stem Cells/enzymology , Adipocytes, Beige/drug effects , Adipocytes, Beige/enzymology , Aminoimidazole Carboxamide/pharmacology , Cell Shape/drug effects , Enzyme Activation/drug effects , Humans , Mitochondria/drug effects , Mitochondria/metabolism , Mitochondrial Dynamics/drug effects , Phenotype , Stem Cells/cytology , Stem Cells/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...