Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
FEBS Lett ; 585(5): 744-8, 2011 Mar 09.
Article in English | MEDLINE | ID: mdl-21315073

ABSTRACT

Cell death-inducing DNA fragmentation factor alpha-like effector A (CIDEA) is endogenously expressed in human but not rodent white adipocytes. We performed a bioinformatic analysis of the human CIDEA sequence and found conserved amino-acid motifs involved in binding to nuclear receptors. Protein-protein binding experiments and transactivation assays confirmed that CIDEA binds to liver X receptors and regulates their activity in vitro. Cell fractionation demonstrated that CIDEA localizes to both the cytoplasm and the nucleus in human white adipocytes. The interaction between CIDEA and nuclear receptors could therefore be of importance for the regulation of metabolic processes in human adipose tissue.


Subject(s)
Adipocytes, White/metabolism , Apoptosis Regulatory Proteins/metabolism , Orphan Nuclear Receptors/metabolism , Adipocytes, White/cytology , Amino Acid Motifs , Amino Acid Sequence , Animals , Apoptosis Regulatory Proteins/chemistry , Cell Line , Cell Nucleus/metabolism , Humans , Liver X Receptors , Mice , Molecular Sequence Data , Protein Binding
2.
J Biol Chem ; 286(1): 370-9, 2011 Jan 07.
Article in English | MEDLINE | ID: mdl-21030586

ABSTRACT

The Liver X receptor (LXR) is an important regulator of carbohydrate and lipid metabolism in humans and mice. We have recently shown that activation of LXR regulates cellular fuel utilization in adipocytes. In contrast, the role of LXR in human adipocyte lipolysis, the major function of human white fat cells, is not clear. In the present study, we stimulated in vitro differentiated human and murine adipocytes with the LXR agonist GW3965 and observed an increase in basal lipolysis. Microarray analysis of human adipocyte mRNA following LXR activation revealed an altered gene expression of several lipolysis-regulating proteins, which was also confirmed by quantitative real-time PCR. We show that expression and intracellular localization of perilipin1 (PLIN1) and hormone-sensitive lipase (HSL) are affected by GW3965. Although LXR activation does not influence phosphorylation status of HSL, HSL activity is required for the lipolytic effect of GW3965. This effect is abolished by PLIN1 knockdown. In addition, we demonstrate that upon activation, LXR binds to the proximal regions of the PLIN1 and HSL promoters. By selective knock-down of either LXR isoform, we show that LXRα is the major isoform mediating the lipolysis-related effects of LXR. In conclusion, the present study demonstrates that activation of LXRα up-regulates basal human adipocyte lipolysis. This is at least partially mediated through LXR binding to the PLIN1 promoter and down-regulation of PLIN1 expression.


Subject(s)
Adipocytes/metabolism , Lipolysis , Orphan Nuclear Receptors/metabolism , 3T3-L1 Cells , Adipocytes/cytology , Adipocytes/drug effects , Adipocytes, White/cytology , Adipocytes, White/drug effects , Adipocytes, White/metabolism , Animals , Carrier Proteins , Down-Regulation/drug effects , Humans , Insulin Resistance , Lipolysis/drug effects , Liver X Receptors , Mice , Orphan Nuclear Receptors/agonists , Perilipin-1 , Phosphoproteins/genetics , Phosphoproteins/metabolism , Phosphorylation/drug effects , Promoter Regions, Genetic/genetics , Protein Isoforms/agonists , Protein Isoforms/metabolism , Protein Transport/drug effects , Retinoid X Receptors/metabolism , Signal Transduction/drug effects , Sterol Esterase/genetics , Sterol Esterase/metabolism , Up-Regulation/drug effects
3.
BMC Endocr Disord ; 10: 1, 2010 Jan 29.
Article in English | MEDLINE | ID: mdl-20205832

ABSTRACT

BACKGROUND: Mice lacking Receptor-interacting protein 140 (RIP140) have reduced body fat which at least partly is mediated through increased lipid and glucose metabolism in adipose tissue. In humans, RIP140 is lower expressed in visceral white adipose tissue (WAT) of obese versus lean subjects. We investigated the role of RIP140 in human subcutaneous WAT, which is the major fat depot of the body. METHODS: Messenger RNA levels of RIP140 were measured in samples of subcutaneous WAT from women with a wide variation in BMI and in different human WAT preparations. RIP140 mRNA was knocked down with siRNA in in vitro differentiated adipocytes and the impact on glucose transport and mRNA levels of target genes determined. RESULTS: RIP140 mRNA levels in subcutaneous WAT were decreased among obese compared to lean women and increased by weight-loss, but did not associate with mitochondrial DNA copy number. RIP140 expression increased during adipocyte differentiation in vitro and was higher in isolated adipocytes compared to corresponding pieces of WAT. Knock down of RIP140 increased basal glucose transport and mRNA levels of glucose transporter 4 and uncoupling protein-1. CONCLUSIONS: Human RIP140 inhibits glucose uptake and the expression of genes promoting energy expenditure in the same fashion as the murine orthologue. Increased levels of human RIP140 in subcutaneous WAT of lean subjects may contribute to economize on energy stores. By contrast, the function and expression pattern does not support that RIP140 regulate human obesity.

4.
Endocrinology ; 150(9): 4104-13, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19556420

ABSTRACT

Liver X receptors (LXRs) are nuclear receptors with established roles in cholesterol, lipid, and carbohydrate metabolism, although their function in adipocytes is not well characterized. Increased adipose tissue mass in obesity is associated with increased adipocyte lipolysis. Fatty acids (FA) generated by lipolysis can be oxidized by mitochondrial beta-oxidation, reesterified, or released from the adipocyte. The latter results in higher circulating levels of free FAs, in turn causing obesity-related metabolic complications. However, mitochondrial beta-oxidation can at least in part counteract an increased output of FA into circulation. In this study, we provide evidence that activation of LXRs up-regulates mitochondrial beta-oxidation in both human and murine white adipocytes. We also show that the expression of a kinase regulating the cellular fuel switch, pyruvate dehydrogenase kinase 4 (PDK4), is up-regulated by the LXR agonist GW3965 in both in vitro differentiated human primary adipocytes and differentiated murine 3T3-L1 cells. Moreover, activation of LXR causes PDK4-dependent phosphorylation of the pyruvate dehydrogenase complex, thereby decreasing its activity and attenuating glucose oxidation. The specificity of the GW3965 effect on oxidation was confirmed by RNA interference targeting LXRs. We propose that LXR has an important role in the regulation of substrate oxidation and the switch between lipids and carbohydrates as cellular fuel in both human and murine white adipocytes.


Subject(s)
Adipocytes, White/metabolism , DNA-Binding Proteins/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , 3T3-L1 Cells , Animals , Benzoates/pharmacology , Benzylamines/pharmacology , Cell Differentiation , Humans , Liver X Receptors , Mice , Orphan Nuclear Receptors , Oxidation-Reduction , Palmitic Acid/metabolism , Phosphorylation , Protein Kinases/metabolism , Pyruvate Dehydrogenase Complex/metabolism , Up-Regulation
5.
J Biol Chem ; 284(27): 18282-91, 2009 Jul 03.
Article in English | MEDLINE | ID: mdl-19433586

ABSTRACT

Lipolysis is the catabolic pathway by which triglycerides are hydrolyzed into fatty acids. Adipose triglyceride lipase (ATGL) and hormone-sensitive lipase (HSL) have the capacity to hydrolyze in vitro the first ester bond of triglycerides, but their respective contributions to whole cell lipolysis in human adipocytes is unclear. Here, we have investigated the roles of HSL, ATGL, and its coactivator CGI-58 in basal and forskolin-stimulated lipolysis in a human white adipocyte model, the hMADS cells. The hMADS adipocytes express the various components of fatty acid metabolism and show lipolytic capacity similar to primary cultured adipocytes. We show that lipolysis and fatty acid esterification are tightly coupled except in conditions of stimulated lipolysis. Immunocytochemistry experiments revealed that acute forskolin treatment promotes HSL translocation from the cytosol to small lipid droplets and redistribution of ATGL from the cytosol and large lipid droplets to small lipid droplets, resulting in enriched colocalization of the two lipases. HSL or ATGL overexpression resulted in increased triglyceride-specific hydrolase capacity, but only ATGL overexpression increased whole cell lipolysis. HSL silencing had no effect on basal lipolysis and only partially reduced forskolin-stimulated lipolysis. Conversely, silencing of ATGL or CGI-58 significantly reduced basal lipolysis and essentially abolished forskolin-stimulated lipolysis. Altogether, these results suggest that ATGL/CGI-58 acts independently of HSL and precedes its action in the sequential hydrolysis of triglycerides in human hMADS adipocytes.


Subject(s)
Adipocytes/enzymology , Energy Metabolism/physiology , Lipase/metabolism , Lipolysis/physiology , Sterol Esterase/metabolism , 1-Acylglycerol-3-Phosphate O-Acyltransferase , Adipocytes/cytology , Adipocytes/drug effects , Cells, Cultured , Colforsin/pharmacology , Cytosol/enzymology , Esterification/physiology , Fatty Acids/metabolism , Green Fluorescent Proteins/genetics , Humans , Hydrolysis , Lipase/genetics , RNA, Small Interfering , Sterol Esterase/genetics
6.
Cancer Res ; 68(22): 9247-54, 2008 Nov 15.
Article in English | MEDLINE | ID: mdl-19010897

ABSTRACT

Loss of fat mass in cancer cachexia is linked to increased adipocyte lipolysis; however, the fate of the excess fatty acids (FA) generated by lipolysis is not known. We investigated if the adipocyte-specific gene cell death-inducing DNA fragmentation factor-alpha-like effector A (CIDEA) could be involved. CIDEA mRNA expression was assessed in s.c. white adipose tissue from 23 cancer cachexia patients, 17 weight-stable cancer patients, and 8 noncancer patients. CIDEA was also overexpressed in adipocytes in vitro. CIDEA expression was increased in cancer cachexia (P < 0.05) and correlated with elevated levels of FAs and reported weight loss (P < 0.001). CIDEA overexpression in vitro increased FA oxidation 2- to 4-fold (P < 0.01), decreased glucose oxidation by 40% (P < 0.01), increased the expression of pyruvate dehydrogenase kinase (PDK) 1 and PDK4 (P < 0.01), and enhanced the phosphorylation (inactivation) of the pyruvate dehydrogenase complex (PDC). Inactivation of PDC facilitates FA oxidation by favoring the metabolism of FAs over glucose to acetyl-CoA. In accordance with the in vitro data, PDK1 and PDK4 expression correlated strongly with CIDEA expression in white adipose tissue (P < 0.001). We conclude that CIDEA is involved in adipose tissue loss in cancer cachexia and this may, at least in part, be due to its ability to inactivate PDC, thereby switching substrate oxidation in human fat cells from glucose to FAs.


Subject(s)
Apoptosis Regulatory Proteins/physiology , Cachexia/etiology , Neoplasms/complications , 3T3-L1 Cells , Adipocytes/metabolism , Adipose Tissue, White/metabolism , Adult , Aged , Animals , Apoptosis Regulatory Proteins/genetics , Body Mass Index , Energy Metabolism , Fatty Acids/metabolism , Female , Glucose/metabolism , Humans , Male , Mice , Middle Aged , Phosphorylation , Protein Serine-Threonine Kinases/genetics , Pyruvate Dehydrogenase Acetyl-Transferring Kinase , Pyruvate Dehydrogenase Complex/metabolism , RNA, Messenger/analysis
7.
J Clin Endocrinol Metab ; 93(6): 2249-54, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18381574

ABSTRACT

CONTEXT: Six transmembrane protein of prostate 2 (STAMP2) is a counterregulator of adipose inflammation and insulin resistance in mice. Our hypothesis was that STAMP2 could be involved in human obesity and insulin resistance. OBJECTIVE: The objective of the study was to elucidate the role of adipose STAMP2 expression in human obesity and insulin resistance. DESIGN: The design was to quantify STAMP2 in human abdominal sc and omental white adipose tissue (WAT), isolated adipocytes, and stroma and in vitro differentiated preadipocytes and relate levels of STAMP2 in sc WAT to clinical and adipocyte phenotypes involved in insulin resistance. PARTICIPANTS: Nonobese and obese women and men (n = 236) recruited from an obesity clinic or through local advertisement. MAIN OUTCOME MEASUREMENT: Clinical measures included body mass index, body fat, total adiponectin, and homeostasis model assessment as measure of overall insulin resistance. In adipocytes we determined cell size, sensitivity of lipolysis and lipogenesis to insulin, adiponectin secretion, and inflammatory gene expression. RESULTS: STAMP2 levels in sc and visceral WAT and adipocytes were increased in obesity (P = 0.0008-0.05) but not influenced by weight loss. Increased WAT STAMP2 levels associated with a high amount of body fat (P = 0.04), high homeostasis model assessment (P = 0.01), and large adipocytes (P = 0.02). Subjects with high STAMP2 levels displayed reduced sensitivity of adipocyte lipogenesis (P = 0.04) and lipolysis (P = 0.03) to insulin but had normal adiponectin levels. WAT STAMP2 levels correlated with expression of the macrophage marker CD68 (P = 0.0006). CONCLUSION: Human WAT STAMP2 associates with obesity and insulin resistance independently of adiponectin, but the role of STAMP2 in obesity and its complications seems different from that in mice.


Subject(s)
Adipose Tissue/metabolism , Adiposity/genetics , Insulin Resistance/genetics , Membrane Proteins/genetics , Obesity/genetics , Oxidoreductases/genetics , Adipocytes/drug effects , Adipocytes/metabolism , Adiponectin/genetics , Adiponectin/metabolism , Adult , Cells, Cultured , Female , Gene Expression Regulation/drug effects , Genetic Predisposition to Disease , Humans , Insulin/pharmacology , Male , Membrane Proteins/metabolism , Middle Aged , Obesity/metabolism , Oxidoreductases/metabolism , RNA, Messenger/metabolism , Tumor Necrosis Factor-alpha/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...