Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 35
Filter
Add more filters










Publication year range
1.
J Cardiothorac Surg ; 9: 2, 2014 Jan 03.
Article in English | MEDLINE | ID: mdl-24387639

ABSTRACT

BACKGROUND: In humans there is a positive association between epicardial adipose tissue (EAT) volume and coronary atherosclerosis (CAD) burden. We tested the hypothesis that EAT contributes locally to CAD in a pig model. METHODS: Ossabaw miniature swine (n=9) were fed an atherogenic diet for 6 months to produce CAD. A 15 mm length by 3-5 mm width coronary EAT (cEAT) resection was performed over the middle segment of the left anterior descending artery (LAD) 15 mm distal to the left main bifurcation. Pigs recovered for 3 months on atherogenic diet. Intravascular ultrasound (IVUS) was performed in the LAD to quantify atheroma immediately after adipectomy and was repeated after recovery before sacrifice. Coronary wall biopsies were stained immunohistochemically for atherosclerosis markers and cytokines and cEAT was assayed for atherosclerosis-related genes by RT-PCR. Total EAT volume was measured by non-contrast CT before each IVUS. RESULTS: Circumferential plaque length increased (p<0.05) in the proximal and distal LAD segments from baseline until sacrifice whereas plaque length in the middle LAD segment underneath the adipectomy site did not increase. T-cadherin, scavenger receptor A and adiponectin were reduced in the intramural middle LAD. Relative to control pigs without CAD, 11ß-hydroxysteroid dehydrogenase (11ßHSD-1), CCL19, CCL21, prostaglandin D2 synthase, gp91phox [NADPH oxidase], VEGF, VEGFGR1, and angiotensinogen mRNAs were up-regulated in cEAT. EAT volume increased over 3 months. CONCLUSION: In pigs used as their own controls, resection of cEAT decreased the progression of CAD, suggesting that cEAT may exacerbate coronary atherosclerosis.


Subject(s)
Adipose Tissue/surgery , Atherosclerosis/surgery , Cardiac Surgical Procedures/methods , Coronary Artery Disease/surgery , Pericardium/surgery , Animals , Atherosclerosis/diagnosis , Coronary Angiography , Coronary Artery Disease/diagnosis , Disease Models, Animal , Disease Progression , Male , Swine , Swine, Miniature , Ultrasonography, Interventional
2.
Metabolism ; 62(10): 1503-11, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23831442

ABSTRACT

BACKGROUND: Exercise training elevates circulating irisin and induces the expression of the FNDC5 gene in skeletal muscles of mice. Our objective was to determine whether exercise training also increases FNDC5 protein or mRNA expression in the skeletal muscles of pigs as well as plasma irisin. METHODS: Castrated male pigs of the Rapacz familial hypercholesterolemic (FHM) strain and normal (Yucatan miniature) pigs were sacrificed after 16-20 weeks of exercise training. Samples of cardiac muscle, deltoid and triceps brachii muscle, subcutaneous and epicardial fat were obtained and FNDC5 mRNA, along with that of 6 other genes, was measured in all tissues of FHM pigs by reverse transcription polymerase chain reaction. FNDC protein in deltoid and triceps brachii was determined by Western blotting in both FHM and normal pigs. Citrate synthase activity was measured in the muscle samples of all pigs as an index of exercise training. Irisin was measured by an ELISA assay. RESULTS: There was no statistically significant effect of exercise training on FNDC5 gene expression in epicardial or subcutaneous fat, deltoid muscle, triceps brachii muscle or heart muscle. Exercise-training elevated circulating levels of irisin in the FHM pigs and citrate synthase activity in deltoid and triceps brachii muscle. A similar increase in citrate synthase activity was seen in muscle extracts of exercise-trained normal pigs but there was no alteration in circulating irisin. CONCLUSION: Exercise training in pigs does not increase FNDC5 mRNA or protein in the deltoid or triceps brachii of FHM or normal pigs while increasing circulating irisin only in the FHM pigs. These data indicate that the response to exercise training in normal pigs is not comparable to that seen in mice.


Subject(s)
Fibronectins/genetics , Fibronectins/metabolism , Muscle, Skeletal/metabolism , Myocardium/metabolism , Physical Conditioning, Animal , RNA, Messenger/genetics , Swine/physiology , Animals , Citrate (si)-Synthase/metabolism , Gene Expression/genetics , Male , Muscle Proteins/genetics , Muscle Proteins/metabolism , Subcutaneous Fat/metabolism , Swine/genetics , Swine/metabolism
3.
J Clin Endocrinol Metab ; 98(9): E1448-55, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23824424

ABSTRACT

CONTEXT: Human epicardial fat has been designated previously as brown-like fat. The supraclavicular fat depot in man has been defined as beige coexistent with classical brown based on its gene expression profile. OBJECTIVE: The aim of the study was to establish the gene expression profile and morphology of human epicardial and visceral paracardial fat compared with sc fat. SETTING: The study was conducted at a tertiary care hospital cardiac center. PATIENTS: Epicardial, visceral paracardial, and sc fat samples had been taken from middle-aged patients with severe coronary atherosclerosis or valvular heart disease. INTERVENTIONS: Gene expression was determined by reverse transcription-quantitative PCR and relative abundance of the mitochondrial uncoupling protein-1 (UCP-1) by Western blotting. Epicardial tissue sections from patients were examined by light microscopy, UCP-1 immunohistochemistry, and cell morphometry. MAIN OUTCOME MEASURES: We hypothesized that epicardial fat has a mixed phenotype with a gene expression profile similar to that described for beige cell lineage. RESULTS: Immunoreactive UCP-1 was clearly measurable in each epicardial sample analyzed but was undetectable in each of the 4 other visceral and sc depots. Epicardial fat exhibited high expression of genes for UCP-1, PRDM16, PGC-1α, PPARγ, and the beige adipocyte-specific marker CD137, which were also expressed in visceral paracardial fat but only weakly in sternal, upper abdominal, and lower extremity sc fat. Histology of epicardial fat showed small unilocular adipocytes without UCP-1 immunostaining. CONCLUSION: UCP-1 is relatively abundant in epicardial fat, and this depot possesses molecular features characteristic of those found in vitro in beige lineage adipocytes.


Subject(s)
Adipose Tissue, Brown/metabolism , Intra-Abdominal Fat/metabolism , Ion Channels/metabolism , Mitochondrial Proteins/metabolism , Pericardium/metabolism , Aged , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Female , Gene Expression , Humans , Ion Channels/genetics , Male , Middle Aged , Mitochondrial Proteins/genetics , PPAR gamma/genetics , PPAR gamma/metabolism , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha , Transcription Factors/genetics , Transcription Factors/metabolism , Tumor Necrosis Factor Receptor Superfamily, Member 9/genetics , Tumor Necrosis Factor Receptor Superfamily, Member 9/metabolism , Uncoupling Protein 1
4.
Horm Mol Biol Clin Investig ; 14(1): 25-32, 2013 Aug.
Article in English | MEDLINE | ID: mdl-25436717

ABSTRACT

The glucocorticoid hormones alter the metabolism of the adipose tissue after an approximately 2-h lag period. The effects are mediated through the nuclear receptors that alter the expression of a wide variety of genes through the mechanisms that are similar to those seen in the other cells. There are many direct metabolic effects of the glucocorticoids on the adipose tissue metabolism, and every year, new effects are added to the list of proteins whose expression is influenced by the glucocorticoids. Furthermore, some enzymatic processes are affected by these hormones only in the presence of the other hormones such as growth hormone (GH) or insulin. Most of the effects of the glucocorticoids are on the gene transcription, and the effects on the mRNA are reflected in the altered levels of the target proteins. The glucocorticoids enhance the leptin release, while reducing that of the inflammatory adipokines and stimulating that of the lipoprotein lipase (LPL) in the presence of insulin. The activity of 11ß-hydroxysteroid dehydrogenase type 1 (HSD1) is enhanced by the glucocorticoids along with that of α1 glycoprotein 1 and serum amyloid A release by the adipose tissue. In contrast, the tumor necrosis factor α (TNF)-stimulated lipolysis in the adipose tissue is blocked by the glucocorticoids. It is still unclear which, if any, of these effects account for the insulin resistance due to the glucocorticoids in the adipose tissue. However, recent work suggests that, at least in mice, the reduction in the osteocalcin release by the osteoblasts in the presence of the glucocorticoids accounts for much of the in vivo insulin resistance. In summary, there are multiple direct effects of the glucocorticoids, both anti-inflammatory and proinflammatory, on the adipose tissue.

5.
Clin Exp Pharmacol Physiol ; 38(12): 879-87, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21895738

ABSTRACT

1. Putative physiological functions of human epicardial adipose tissue (EAT) include: (i) lipid storage for the energy needs of the myocardium; (ii) thermoregulation, whereby brown fat components of EAT generate heat by non-shivering thermogenesis in response to core cooling; (iii) neuroprotection of the cardiac autonomic ganglia and nerves; and (iv) regulation of vasomotion and luminal size of the coronary arteries. Under pathophysiological circumstances, EAT may play an adverse paracrine role in cardiac arrhythmias and in lipotoxic cardiomyopathy, but of major current interest is its hypothetical role as an immunological organ contributing to inflammation around coronary artery disease (CAD). 2. The amount of EAT measured either by echocardiographic thickness over the free wall of the right ventricle or as volume by computed tomography expands in patients with obesity both without and with CAD. The mechanisms other than obesity governing the increase in EAT volume in CAD are unknown, but EAT around CAD is infiltrated by chronic inflammatory cells and overexpresses genes for adipokines that have pro- or anti-inflammatory actions and regulate oxidative stress plus angiogenesis. 3. Many cross-sectional studies have shown positive associations between increased EAT mass and stable CAD burden. One prospective population-based epidemiological study suggested that EAT volume at baseline is a predictor of acute myocardial infarction, but was without significant incremental predictive value after adjustment for established cardiovascular risk factors. However, strategies are needed to obtain robust epidemiological, interventional and experimental evidence to prove or disprove the hypothesis that EAT is a cardiovascular risk factor locally contributing to CAD.


Subject(s)
Adipose Tissue/physiology , Pericardium/physiology , Adipokines/metabolism , Adipose Tissue/diagnostic imaging , Adipose Tissue/metabolism , Animals , Body Temperature Regulation/physiology , Coronary Artery Disease/diagnostic imaging , Coronary Artery Disease/immunology , Coronary Artery Disease/metabolism , Coronary Artery Disease/physiopathology , Coronary Vessels/immunology , Coronary Vessels/metabolism , Coronary Vessels/physiology , Female , Heart/diagnostic imaging , Heart/innervation , Heart/physiology , Humans , Male , Mice , Obesity/metabolism , Obesity/physiopathology , Pericardium/diagnostic imaging , Pericardium/metabolism , Radiography , Rats , Weight Loss/physiology
6.
Metab Syndr Relat Disord ; 9(6): 433-9, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21679057

ABSTRACT

BACKGROUND: Pro- and antiinflammatory genes are expressed in epicardial adipose tissue (EAT). Our objectives were to characterize genes in EAT that may contribute specifically to coronary atherogenesis and to measure circulating adipokines matched to their messenger RNAs (mRNAs) in EAT. We hypothesized that severe coronary atherosclerosis (CAD) would preferentially affect gene expression in EAT as compared to substernal fat or subcutaneous thoracic adipose tissue (SAT), as well as circulating levels of adipokines. METHODS: Fat mRNA was quantified using reverse transcription polymerase chain reaction (RT-PCR), and circulating adipokines were measured by enzyme-linked immunosorbent assays (ELISAs) in patients with severe stable CAD and controls without severe CAD undergoing open heart surgery. RESULTS: A total of 39 of 70 mRNAs in EAT were significantly increased in CAD. Only 4 and 3 of these mRNAs were increased in substernal fat and SAT, respectively. Of the mRNAs increased in EAT, 17 were either inflammatory adipokines or proteins known to be involved in inflammatory processes, 7 were involved in oxidative stress and or oxygen species regulation, whereas 15 were proteins involved in metabolism and regulation of gene transcription or proteins unique to fat cells. The largest increases, over three-fold, were seen in GPX3, gp91 phox, p47phox, heme oxygenase, and interleukin-8 (IL-8). Tpl2 mRNA was uniquely elevated in all three fat depots from CAD patients, and its expression in SAT, but not in EAT or substernal fat, was directly correlated with homeostasis model assessment of insulin resistance (HOMA-IR) values. Compared to controls, there were no associations between circulating levels of IL-8, lipocalin-2, nerve growth factor (NGF), RANTES, CD-163, GPX-3, monocyte chemotactic protein-1 (MCP-1)/CCL2, leptin, soluble vascular endothelial growth factor receptor-1 (sFLT1), fatty acid binding protein-4 (FABP-4), and plasminogen activator inhibitor-1 (PAI-1) and increases in their gene expression in EAT adjacent to CAD. CONCLUSIONS: Expression of proinflammatory, redox, endothelial cell, and angiogenic genes in EAT is depot specific and supports the hypothesis that pathophysiologically EAT contributes locally to CAD. CAD links with these fat depots might involve Tpl2 as a primary response indicator.


Subject(s)
Adipose Tissue/metabolism , Coronary Artery Disease/genetics , Endothelial Cells/metabolism , Inflammation/genetics , Neovascularization, Physiologic/genetics , Pericardium/metabolism , Adipose Tissue/pathology , Aged , Angiogenesis Inducing Agents/metabolism , Case-Control Studies , Coronary Artery Disease/metabolism , Coronary Artery Disease/pathology , Disease Progression , Endothelial Cells/pathology , Female , Gene Expression Regulation , Humans , Inflammation/metabolism , Inflammation Mediators/metabolism , Male , Middle Aged , Oxidation-Reduction , Severity of Illness Index , Up-Regulation/genetics
7.
Hypertension ; 58(1): 37-42, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21555675

ABSTRACT

Adipose tissue growth depends on angiogenesis. We tested the hypothesis that adipose tissue produces factors relevant to angiogenesis. We obtained fat biopsies in 2 different patient cohorts, cultured adipose-derived stem cells and studied mature adipocytes. We performed microarray, RT-PCR, and Western blotting; studied a rat obesity/metabolic syndrome model; and conducted viral gene transfer experiments in leptin-deficient mice. The microarray identified the splice variant of the vascular endothelial growth factor receptor, the soluble fms-like tyrosine kinase 1 (sFlt-1), as an antiangiogenesis candidate. We verified the expression findings and found that sFlt-1 was secreted by isolated mature human adipocytes. Tumor necrosis factor-α decreased sFlt-1 expression in mature adipocytes, whereas hypoxia had no effect. Separating cells from adipose tissue showed that the highest sFlt-1 expression was present in adipose-tissue nonfat cells rather than in the adipocytes themselves. We also found that sFlt-1 expression and sFlt-1 release by adipose-tissue explants were inversely correlated with body mass index of the corresponding patients but was directly correlated with adiponectin expression. In the obesity/metabolic syndrome rat model, we observed that circulating sFlt-1 levels and sFlt-1 expression in adipose tissue were also inversely correlated with body weight. To model our putative antiangiogenic factor further, we next overexpressed sFlt-1 by viral transfer in a mouse genetic model of leptin deficiency and observed that the transfected mice gained less weight than controls. We suggest that sFlt-1 could act as a paracrine factor inhibiting adipose tissue growth. Local sFlt-1 may regulate angiogenic potential and thereby influence adipose tissue mass.


Subject(s)
Adipocytes/metabolism , Adipokines/metabolism , Gene Expression Regulation , Neovascularization, Pathologic/genetics , Obesity/genetics , RNA/genetics , Vascular Endothelial Growth Factor Receptor-1/genetics , Adipocytes/pathology , Animals , Blotting, Western , Body Mass Index , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Female , Humans , Immunohistochemistry , Male , Mice , Mice, Obese , Neovascularization, Pathologic/etiology , Neovascularization, Pathologic/metabolism , Obesity/enzymology , Obesity/pathology , Rats , Rats, Transgenic , Reverse Transcriptase Polymerase Chain Reaction , Vascular Endothelial Growth Factor Receptor-1/biosynthesis
8.
Diabetes Care ; 34(3): 730-3, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21289232

ABSTRACT

OBJECTIVE: To determine changes in gene expression in epicardial adipose tissue (EAT) associated with coronary atherosclerosis (CAD) and effects of pioglitazone therapy. RESEARCH DESIGN AND METHODS: Genes were quantified by RT-PCR in EAT and thoracic subcutaneous adipose tissue (SAT) obtained during surgery in CAD patients with metabolic syndrome (MS) or type 2 diabetes and control subjects with minimal or no CAD and no MS or type 2 diabetes. RESULTS: Increased expression of interleukin-1 receptor antagonist (IL-1Ra) and IL-10, a trend for higher IL-1ß, and no change in peroxisome proliferator-activated receptor-γ (PPARγ) was found in EAT from MS or type 2 diabetes. Only PPARγ mRNA was reduced in SAT. Pioglitazone therapy in type 2 diabetes was associated with decreased expression of IL-1ß, IL-1Ra, and IL-10 in EAT; decreased IL-10 in SAT; and increased PPARγ in SAT. CONCLUSIONS: In MS and type 2 diabetes with CAD, proinflammatory and anti-inflammatory genes were differentially increased in EAT and selectively reduced in association with pioglitazone treatment.


Subject(s)
Coronary Artery Disease/drug therapy , Diabetes Mellitus, Type 2/drug therapy , Hypoglycemic Agents/therapeutic use , Metabolic Syndrome/drug therapy , Pericardium/metabolism , Thiazolidinediones/therapeutic use , Coronary Artery Disease/genetics , Diabetes Mellitus, Type 2/genetics , Humans , Interleukin 1 Receptor Antagonist Protein/genetics , Interleukin-10/genetics , Interleukin-1beta/genetics , Metabolic Syndrome/genetics , Pioglitazone , Reverse Transcriptase Polymerase Chain Reaction
9.
J Appl Physiol (1985) ; 109(6): 1904-12, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20947714

ABSTRACT

Epicardial adipose tissue (EAT) is contiguous with coronary arteries and myocardium and potentially may play a role in coronary atherosclerosis (CAD). Exercise is known to improve cardiovascular disease risk factors. The purpose of this study was to investigate the effect of aerobic exercise training on the expression of 18 genes, measured by RT-PCR and selected for their role in chronic inflammation, oxidative stress, and adipocyte metabolism, in peri-coronary epicardial (cEAT), peri-myocardial epicardial (mEAT), visceral abdominal (VAT), and subcutaneous (SAT) adipose tissues from a castrate male pig model of familial hypercholesterolemia with CAD. We tested the hypothesis that aerobic exercise training for 16 wk would reduce the inflammatory profile of mRNAs in both components of EAT and VAT but would have little effect on SAT. Exercise increased mEAT and total heart weights. EAT and heart weights were directly correlated. Compared with sedentary pigs matched for body weight to exercised animals, aerobic exercise training reduced the inflammatory response in mEAT but not cEAT, had no effect on inflammatory genes but preferentially decreased expression of adiponectin and other adipocyte-specific genes in VAT, and had no effect in SAT except that IL-6 mRNA went down and VEGFa mRNA went up. We conclude that 1) EAT is not homogeneous in its inflammatory response to aerobic exercise training, 2) cEAT around CAD remains proinflammatory after chronic exercise, 3) cEAT and VAT share similar inflammatory expression profiles but different metabolic mRNA responses to exercise, and 4) gene expression in SAT cannot be extrapolated to VAT and heart adipose tissues in exercise intervention studies.


Subject(s)
Adiposity/genetics , Coronary Artery Disease/genetics , Hyperlipoproteinemia Type II/genetics , Inflammation/genetics , Intra-Abdominal Fat/physiopathology , Physical Exertion , Subcutaneous Fat/physiopathology , Adipokines/genetics , Animals , Castration , Coronary Artery Disease/pathology , Coronary Artery Disease/physiopathology , Disease Models, Animal , Gene Expression Regulation , Hyperlipoproteinemia Type II/pathology , Hyperlipoproteinemia Type II/physiopathology , Inflammation/pathology , Inflammation/physiopathology , Inflammation/prevention & control , Inflammation Mediators/metabolism , Intra-Abdominal Fat/metabolism , Male , Oxidative Stress/genetics , Pericardium , Polymerase Chain Reaction , RNA, Messenger/metabolism , Subcutaneous Fat/metabolism , Swine
10.
Mediators Inflamm ; 2010: 513948, 2010.
Article in English | MEDLINE | ID: mdl-20508843

ABSTRACT

This paper considers the role of putative adipokines that might be involved in the enhanced inflammatory response of human adipose tissue seen in obesity. Inflammatory adipokines [IL-6, IL-10, ACE, TGFbeta1, TNFalpha, IL-1beta, PAI-1, and IL-8] plus one anti-inflammatory [IL-10] adipokine were identified whose circulating levels as well as in vitro release by fat are enhanced in obesity and are primarily released by the nonfat cells of human adipose tissue. In contrast, the circulating levels of leptin and FABP-4 are also enhanced in obesity and they are primarily released by fat cells of human adipose tissue. The relative expression of adipokines and other proteins in human omental as compared to subcutaneous adipose tissue as well as their expression in the nonfat as compared to the fat cells of human omental adipose tissue is also reviewed. The conclusion is that the release of many inflammatory adipokines by adipose tissue is enhanced in obese humans.


Subject(s)
Adipokines/metabolism , Adipose Tissue , Inflammation Mediators/metabolism , Inflammation/metabolism , Obesity/metabolism , Adipose Tissue/cytology , Adipose Tissue/metabolism , Adipose Tissue/physiology , Gene Expression , Humans , Hypoxia/metabolism , Omentum/anatomy & histology , Omentum/metabolism , Omentum/physiology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Subcutaneous Fat/metabolism , Subcutaneous Fat/physiology , Toll-Like Receptor 4/metabolism
11.
Metabolism ; 59(9): 1379-86, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20116810

ABSTRACT

We compared the gene expression of inflammatory and other proteins by real-time quantitative polymerase chain reaction in epicardial, substernal (mediastinal) and subcutaneous sternal, upper abdominal, and leg fat from coronary bypass patients and omental (visceral) fat from extremely obese women undergoing bariatric surgery. We hypothesized that (1) epicardial fat would exhibit higher expression of inflammatory messenger RNAs (mRNAs) than substernal and subcutaneous fat and (2) epicardial mRNAs would be similar to those in omental fat. Epicardial fat was clearly different from substernal fat because there was a far higher expression of haptoglobin, prostaglandin D(2) synthase, nerve growth factor beta, the soluble vascular endothelial growth factor receptor (FLT1), and alpha1 glycoprotein but not of inflammatory adipokines such as monocyte chemoattractant protein-1, interleukin (IL)-8, IL-1beta, tumor necrosis factor alpha, serum amyloid A, plasminogen activator inhibitor-1, or adiponectin despite underlying coronary atherosclerosis. However, the latter inflammatory adipokines as well as most other mRNAs were overexpressed in epicardial fat as compared with the subcutaneous depots except for IL-8, fatty acid binding protein 4, the angiotensin II receptor 1, IL-6, and superoxide dismutase-2. Relative to omental fat, about one third of the genes were expressed at the same levels, whereas monocyte chemoattractant protein-1, cyclooxygenase-2, plasminogen activator inhibitor-1, IL-1beta, and IL-6 were expressed at far lower levels in epicardial fat. In conclusion, epicardial fat does not appear to be a potentially more important source of inflammatory adipokines than substernal mediastinal fat. Furthermore, the expression of inflammatory cytokines such as IL-6 and IL-1beta is actually higher in omental fat from obese women without coronary atherosclerosis. The data do not support the hypothesis that most of the inflammatory adipokines are expressed at high levels in epicardial fat of humans.


Subject(s)
Adipokines/metabolism , Adipose Tissue/metabolism , Gene Expression , Obesity, Morbid/metabolism , Adipokines/genetics , Adipose Tissue/chemistry , Adult , Female , Humans , Obesity, Morbid/genetics , RNA, Messenger/analysis , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Tissue Distribution
12.
J Inflamm (Lond) ; 7: 4, 2010 Jan 21.
Article in English | MEDLINE | ID: mdl-20145729

ABSTRACT

BACKGROUND: The present studies were designed to investigate the changes in gene expression during in vitro incubation of human visceral omental adipose tissue explants as well as fat cells and nonfat cells derived from omental fat. METHODS: Adipose tissue was obtained from extremely obese women undergoing bariatric surgery. Explants of the tissue as well as fat cells and the nonfat cells derived by digestion with collagenase were incubated for 20 minutes to 48 h. The expression of interleukin 1beta [IL-1beta], tumor necrosis factor alpha [TNFalpha], interleukin 8 [IL-8], NFkappaB(1)p50 subunit, hypoxia-inducible factor 1alpha [HIF1alpha], omentin/intelectin, and 11beta-hydroxysteroid dehydrogenase 1 [11beta-HSD1] mRNA were measured by qPCR as well as the release of IL-8 and TNFalpha. RESULTS: There was an inflammatory response at 2 h in explants of omental adipose tissue that was reduced but not abolished in the absence of albumin from the incubation buffer for IL-8, IL-1beta and TNFalpha. There was also an inflammatory response with regard to upregulation of HIF1alpha and NFkappaB1 gene expression that was unaffected whether albumin was present or absent from the medium. In the nonfat cells derived by a 2 h collagenase digestion of omental fat there was an inflammatory response comparable but not greater than that seen in tissue. The exception was HIF1alpha where the marked increase in gene expression was primarily seen in intact tissue. The inflammatory response was not seen with respect to omentin/intelectin. Over a subsequent 48 h incubation there was a marked increase in IL-8 mRNA expression and IL-8 release in adipose tissue explants that was also seen to the same extent in the nonfat cells incubated in the absence of fat cells. CONCLUSION: The marked inflammatory response seen when human omental adipose tissue is incubated in vitro is reduced but not abolished in the presence of albumin with respect to IL-1beta, TNFalpha, IL-8, and is primarily in the nonfat cells of adipose tissue.

13.
Obesity (Silver Spring) ; 18(5): 890-6, 2010 May.
Article in English | MEDLINE | ID: mdl-19834460

ABSTRACT

The relative release in vitro of endothelin-1, zinc-alpha2-glycoprotein (ZAG), lipocalin-2, CD14, RANTES (regulated on activation, normal T cell expressed and secreted protein), lipoprotein lipase (LPL), osteoprotegerin (OPG), fatty acid-binding protein 4 (FABP-4), visfatin/PBEF/Nampt, glutathione peroxidase-3 (GPX-3), intracellular cell adhesion molecule 1 (ICAM-1), and amyloid A was examined using explants of human adipose tissue as well as the nonfat cell fractions and adipocytes from obese women. Over a 48-h incubation the majority of the release of LPL was by fat cells whereas that of lipocalin-2, RANTES, and ICAM-1 was by the nonfat cells present in human adipose tissue. In contrast appreciable amounts of OPG, amyloid A, ZAG, FABP-4, GPX-3, CD14, and visfatin/PBEF/Nampt were released by both fat cells and nonfat cells. There was an excellent correlation (r = 0.75) between the ratios of adipokine release by fat cells to nonfat cells over 48 h and the ratio of their mRNAs in fat cells to nonfat cells at the start of the incubation. The total release of ZAG, OPG, RANTES, and amyloid A by incubated adipose tissue explants from women with a fat mass of 65 kg was not different from that by women with a fat mass of 29 kg. In contrast that of ICAM-1, FABP-4, GPX-3, visfatin/PBEF/Nampt, CD14, lipocalin-2, LP, and endothelin-1 was significantly greater in tissue from women with a total fat mass of 65 kg.


Subject(s)
Adipocytes/metabolism , Adipokines/metabolism , Adipose Tissue/metabolism , Obesity/physiopathology , Adult , Female , Humans , Middle Aged , Reverse Transcriptase Polymerase Chain Reaction
14.
Mol Cell Endocrinol ; 315(1-2): 292-8, 2010 Feb 05.
Article in English | MEDLINE | ID: mdl-19853017

ABSTRACT

Dexamethasone is a synthetic glucocorticoid that is a potent anti-inflammatory agent. The present studies examined the changes in gene expression of 64 proteins in human omental adipose tissue explants incubated for 48h both in the absence and presence of dexamethasone as well as the release of 8 of these proteins that are putative adipokines. The proteins were chosen because they are inflammatory response proteins in other cells, are key regulatory proteins or are proteins with known functions. About 50% were significantly up-regulated while about 10% were unchanged and the remaining 40% were down-regulated. Dexamethasone significantly up-regulated the expression of about 33% of the proteins but down-regulated the expression of about 12% of the proteins. We conclude that dexamethasone is a selective anti-inflammatory agent since it inhibits only about one-fourth of the proteins up-regulated during in vitro incubation of human omental adipose tissue.


Subject(s)
Adipose Tissue , Anti-Inflammatory Agents/therapeutic use , Dexamethasone , Inflammation/drug therapy , Omentum , Adipose Tissue/drug effects , Adipose Tissue/pathology , Adult , Anti-Inflammatory Agents/pharmacology , Dexamethasone/pharmacology , Dexamethasone/therapeutic use , Female , Gene Expression Profiling , Humans , Interleukin-8/genetics , Interleukin-8/metabolism , Omentum/anatomy & histology , Omentum/drug effects , Omentum/pathology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rotenone/pharmacology , Tissue Culture Techniques , Uncoupling Agents/pharmacology , Up-Regulation
15.
J Clin Endocrinol Metab ; 94(9): 3611-5, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19567523

ABSTRACT

CONTEXT: Uncoupling protein-1 (UCP-1) is the inner mitochondrial membrane protein that is a specific marker for and mediator of nonshivering thermogenesis in brown adipocytes. OBJECTIVE: This study was performed to better understand the putative thermogenic function of human epicardial fat. DESIGN: We measured the expression of UCP-1 and brown adipocyte differentiation transcription factors PR-domain-missing 16 (PRDM16) and peroxisome-proliferator-activated receptor gamma co-activator-1 alpha (PGC-1 alpha) in epicardial, substernal, and sc thoracic, abdominal, and leg fat. SETTING: The study was conducted at a tertiary care hospital cardiac center. PATIENTS: Forty-four patients had coronary artery bypass surgery, and six had heart valve replacement. INTERVENTIONS: Fat samples were taken at open heart surgery. RESULTS: UCP-1 expression was 5-fold higher in epicardial fat than substernal fat and barely detectable in sc fat. Epicardial fat UCP-1 expression decreased with age, increased with body mass index, was similar in women and men and patients on and not on statin therapy, and showed no relationship to epicardial fat volume or waist circumference. UCP-1 expression was similar in patients without and with severe coronary atherosclerosis and metabolic syndrome or type 2 diabetes. PRDM16 and PGC-1 alpha expression was 2-fold greater in epicardial than sc fat. Epicardial fat UCP-1, PRDM16, and PGC1-alpha mRNAs were similar in diabetics treated with thiazolidinediones compared to diabetics not treated with thiazolidinediones. CONCLUSION: Because UCP-1 is expressed at high levels in epicardial fat as compared to other fat depots, the possibility should be considered that epicardial fat functions like brown fat to defend the myocardium and coronary vessels against hypothermia. This process could be blunted in the elderly.


Subject(s)
Adipose Tissue, Brown/metabolism , Adipose Tissue/metabolism , DNA-Binding Proteins/genetics , Heat-Shock Proteins/genetics , Ion Channels/genetics , Mitochondrial Proteins/genetics , Pericardium/metabolism , Transcription Factors/genetics , Aged , Coronary Artery Disease/metabolism , Diabetes Mellitus/drug therapy , Diabetes Mellitus/metabolism , Female , Humans , Male , Middle Aged , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha , RNA, Messenger/analysis , Uncoupling Protein 1
16.
Mol Cell Endocrinol ; 295(1-2): 101-5, 2008 Nov 25.
Article in English | MEDLINE | ID: mdl-18640775

ABSTRACT

Growth hormone [GH] administration results in a reduction in adiposity of humans that is attributed to stimulation of lipolysis. We examined the effect of direct addition of human GH, in both the absence and presence of dexamethasone [Dex], as well as that of interferon beta on lipolysis by omental adipose tissue explants from obese women incubated for 48h in primary culture. There was a significant stimulation of lipolysis by GH in the presence of Dex but not by Dex or GH alone. There was also a significant further stimulation by GH in the presence of Dex of hormone-sensitive lipase, perilipin, lipoprotein lipase and beta1 adrenergic receptor mRNA. We conclude that the direct lipolytic effect of GH is accompanied by an increase in HSL mRNA in the presence of DEX, but GH also increased the mRNAs for other proteins that could explain all or part of its lipolytic action.


Subject(s)
Dexamethasone/pharmacology , Glucocorticoids/pharmacology , Human Growth Hormone/metabolism , Intra-Abdominal Fat/drug effects , Lipolysis/drug effects , Obesity, Morbid/metabolism , Carrier Proteins , Female , Humans , Interferon-beta/metabolism , Intra-Abdominal Fat/enzymology , Intra-Abdominal Fat/metabolism , Lipase/genetics , Lipase/metabolism , Obesity, Morbid/enzymology , Omentum , Perilipin-1 , Phosphoproteins/genetics , Phosphoproteins/metabolism , RNA, Messenger/metabolism , Receptors, Adrenergic, beta-1/genetics , Receptors, Adrenergic, beta-1/metabolism , Sterol Esterase/genetics , Sterol Esterase/metabolism , Time Factors , Tissue Culture Techniques
17.
Metabolism ; 57(7): 1005-15, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18555844

ABSTRACT

The messenger RNA (mRNA) distribution of 60 proteins was examined in the 3 fractions obtained by collagenase digestion (fat cells and the nonfat cells comprising the tissue remaining after collagenase digestion [matrix] and the stromovascular cells) of omental adipose tissue obtained from morbidly obese women undergoing bariatric surgery. Fat cells were enriched by at least 3-fold as compared with nonfat cells in the mRNAs for retinol binding protein 4, angiotensinogen, adipsin, glutathione peroxidase 3, uncoupling protein 2, peroxisome proliferator-activated receptor gamma, cell death-inducing DFFA-like effector A, fat-specific protein 27, 11beta-hydroxysteroid dehydrogenase 1, glycerol channel aquaporin 7, NADPH:quinone oxidoreductase 1, cyclic adenosine monophosphate phosphodiesterase 3B, glyceraldehyde-3-phosphate dehydrogenase, insulin receptor, and amyloid A1. Fat cells were also enriched by at least 26-fold in the mRNAs for proteins involved in lipolysis such as hormone-sensitive lipase, lipoprotein lipase, adipose tissue triglyceride lipase, and FAT/CD36. The relative distribution of mRNAs in cultured preadipocytes was also compared with that of in vitro differentiated adipocytes derived from human omental adipose tissue. Cultured preadipocytes had far lower levels of the mRNAs for inflammatory proteins than the nonfat cells of omental adipose tissue. The nonfat cells were enriched by at least 5-fold in the mRNAs for proteins involved in the inflammatory response such as tumor necrosis factor alpha, interleukin lbeta, cyclooxygenase 2, interleukin 24, interleukin 6, and monocyte chemoattractant protein 1 plus the mRNAs for osteopontin, vaspin, endothelin, angiotensin II receptor 1, butyrylcholinesterase, lipocalin 2, and plasminogen activator inhibitor 1. The cells in the adipose tissue matrix were enriched at least 3-fold as compared with the isolated stromovascular cells in the mRNAs for proteins related to the inflammatory response, as well as osteopontin and endothelial nitric oxide synthase. We conclude that the mRNAs for inflammatory proteins are primarily present in the nonfat cells of human omental adipose tissue.


Subject(s)
Adipocytes/metabolism , Adipose Tissue/metabolism , Omentum/metabolism , RNA, Messenger/biosynthesis , Adipokines/biosynthesis , Adipokines/genetics , Adult , Body Mass Index , Cell Differentiation/physiology , Cell Separation , Cytokines/biosynthesis , Cytokines/genetics , Female , Humans , Obesity, Morbid/metabolism , Omentum/cytology , Signal Transduction/physiology
18.
Metabolism ; 56(11): 1583-90, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17950111

ABSTRACT

The relative release in vitro of adipsin, vascular cell adhesion molecule (VCAM) 1, angiotensin 1-converting enzyme (ACE), and soluble tumor necrosis factor alpha receptor 2 (sTNFR2) by explants of human omental and subcutaneous adipose tissue as well as the nonfat cell fractions and adipocytes from morbidly obese gastric bypass women was compared with that by tissue from obese abdominoplasty patients. Release of VCAM-1 and ACE by omental adipose tissue explants was 220% and 80% greater, respectively, over 48 hours of incubation than that by subcutaneous adipose tissue explants. However, this difference was not seen when release by adipocytes derived from omental fat was compared with that by adipocytes from subcutaneous fat. The release of adipsin and sTNFR2 by omental adipose tissue explants did not differ from that by subcutaneous tissue adipose tissue. The release of adipsin by tissue explants over 48 hours was 100-fold greater than that of VCAM-1, ACE, or sTNFR2. Most of the release of all 4 adipokines was due to the nonfat cells because adipsin release by adipocytes was only 13% of that by the nonfat cells derived from the same amount of adipose tissue, whereas ACE release by adipocytes was 7% and release of VCAM-1 as well as sTNFR2 by adipocytes was 4% or less of that by nonfat cells. The total release in vitro of ACE and sTNFR2, but not that of adipsin or VCAM-1, was enhanced in adipose tissue explants from morbidly obese women as compared with those by explants derived from obese women. We conclude that although human adipose tissue explants release appreciable amounts of adipsin and far smaller amounts of VCAM-1, ACE, and sTNFR2 in vitro, more than 87% of the release is due to the nonfat cells present in adipose tissue. Furthermore, the enhanced release of VCAM-1 and ACE seen in omental adipose tissue explants as compared with explants of subcutaneous adipose tissue is due to release by nonfat cells.


Subject(s)
Adipocytes/metabolism , Adipose Tissue/metabolism , Complement Factor D/metabolism , Immunoglobulin G/metabolism , Omentum/metabolism , Peptidyl-Dipeptidase A/metabolism , Receptors, Tumor Necrosis Factor/metabolism , Vascular Cell Adhesion Molecule-1/metabolism , Adipose Tissue/pathology , Etanercept , Female , Humans , Obesity/metabolism , Omentum/pathology , RNA, Messenger/genetics , Vascular Cell Adhesion Molecule-1/genetics
19.
Int J Pediatr Obes ; 2(3): 130-7, 2007.
Article in English | MEDLINE | ID: mdl-17852540

ABSTRACT

OBJECTIVE: Circulating leptin levels positively correlate with adult BMI and size at birth. Previous studies found gender-specific associations between polymorphisms in the leptin gene and postnatal obesity-related traits or circulating leptin levels. We examined the relationships among leptin gene polymorphisms, size for gestational age, umbilical cord leptin, and gender. METHODS: Six single nucleotide polymorphisms (SNPs) were genotyped in the leptin gene in 261 newborns (72 low birth weight Caucasians, 189 randomly-selected African-Americans). In African-Americans, umbilical cord leptin and free testosterone levels were measured. Linear regression was used to identify significant predictors of size for gestational age or cord leptin levels and gender x genotype interaction effects. RESULTS: There is a significant interaction between gender and genotype at site -2548 (A/G). Among low birth weight Caucasians, the A allele was associated with an increase in female size for gestational age, while the A allele was associated with decreased male birth size. Among African-Americans, the A allele was associated with a decrease in umbilical cord leptin in females and with an increase in cord leptin in males. Cord testosterone levels were not a significant predictor of cord leptin levels either among all African-American newborns or among strata of -2548 genotypes and gender. CONCLUSION: In male and female fetuses, site -2548 of the leptin gene may differently affect the expression level of the leptin gene or the rate of fetal growth. This gender-specific effect does not appear to be mediated by the level of free testosterone at delivery.


Subject(s)
Birth Weight , Body Size/genetics , Fetal Blood/chemistry , Leptin/blood , Adult , Black People , Body Mass Index , Female , Gestational Age , Humans , Infant, Newborn , Male , Maternal Age , Parity , Pregnancy , Sex Characteristics , United States , Weight Gain , White People
20.
Pediatr Res ; 62(4): 451-5, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17667841

ABSTRACT

The insulin (INS) and IGF 2 (IGF2) genes are in close proximity to each other and undergo maternal imprinting during fetal growth. We investigated the association between maternal and umbilical cord IGF 2 protein (IGF-II) levels and single nucleotide polymorphisms (SNPs) in the INS and IGF2 genes in 207 healthy African-American mother-newborn pairs. No association was found between maternal IGF-II levels and polymorphism in the INS-IGF2 locus. A significant association was found between newborn IGF-II levels and two SNPs (rs3842738 and rs689) at the 5' end of the INS-IGF2 locus. Analyses of haplotypes inferred from these two SNPs demonstrate a significant relationship between paternally transmitted haplotypes and newborn IGF-II levels, but no association with maternally transmitted haplotypes.


Subject(s)
Black or African American/genetics , Fetal Blood/chemistry , Genomic Imprinting , Insulin/genetics , Polymorphism, Single Nucleotide , Proteins/genetics , Adult , Female , Fetal Development/genetics , Gene Frequency , Gestational Age , Haplotypes , Humans , Infant, Newborn , Insulin-Like Growth Factor II , Linkage Disequilibrium , Male , Pedigree , Phenotype , Proteins/analysis , United States
SELECTION OF CITATIONS
SEARCH DETAIL
...