Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
1.
Gastroenterology ; 161(3): 968-981.e12, 2021 09.
Article in English | MEDLINE | ID: mdl-34004161

ABSTRACT

BACKGROUND AND AIMS: Insulin resistance is a key factor in the pathogenesis of nonalcoholic fatty liver disease (NAFLD). We evaluated the importance of subcutaneous abdominal adipose tissue (SAAT) inflammation and both plasma and SAAT-derived exosomes in regulating insulin sensitivity in people with obesity and NAFLD. METHODS: Adipose tissue inflammation (macrophage and T-cell content and expression of proinflammatory cytokines), liver and whole-body insulin sensitivity (assessed using a hyperinsulinemic-euglycemic clamp and glucose tracer infusion), and 24-hour serial plasma cytokine concentrations were evaluated in 3 groups stratified by adiposity and intrahepatic triglyceride (IHTG) content: (1) lean with normal IHTG content (LEAN; N = 14); (2) obese with normal IHTG content (OB-NL; N = 28); and (3) obese with NAFLD (OB-NAFLD; N = 28). The effect of plasma and SAAT-derived exosomes on insulin-stimulated Akt phosphorylation in human skeletal muscle myotubes and mouse primary hepatocytes was assessed in a subset of participants. RESULTS: Proinflammatory macrophages, proinflammatory CD4 and CD8 T-cell populations, and gene expression of several cytokines in SAAT were greater in the OB-NAFLD than the OB-NL and LEAN groups. However, with the exception of PAI-1, which was greater in the OB-NAFLD than the LEAN and OB-NL groups, 24-hour plasma cytokine concentration areas-under-the-curve were not different between groups. The percentage of proinflammatory macrophages and plasma PAI-1 concentration areas-under-the-curve were inversely correlated with both hepatic and whole-body insulin sensitivity. Compared with exosomes from OB-NL participants, plasma and SAAT-derived exosomes from the OB-NAFLD group decreased insulin signaling in myotubes and hepatocytes. CONCLUSIONS: Systemic insulin resistance in people with obesity and NAFLD is associated with increased plasma PAI-1 concentrations and both plasma and SAAT-derived exosomes. ClinicalTrials.gov number: NCT02706262 (https://clinicaltrials.gov/ct2/show/NCT02706262).


Subject(s)
Cytokines/blood , Exosomes/metabolism , Insulin Resistance , Macrophages/metabolism , Memory T Cells/metabolism , Non-alcoholic Fatty Liver Disease/blood , Obesity/blood , Plasminogen Activator Inhibitor 1/blood , Subcutaneous Fat, Abdominal/metabolism , Adult , Animals , Biomarkers/blood , Blood Glucose/metabolism , Cells, Cultured , Exosomes/immunology , Female , Hepatocytes/metabolism , Humans , Insulin/blood , Liver/metabolism , Macrophages/immunology , Male , Memory T Cells/immunology , Mice , Mice, Inbred C57BL , Muscle Fibers, Skeletal/metabolism , Non-alcoholic Fatty Liver Disease/diagnosis , Non-alcoholic Fatty Liver Disease/immunology , Non-alcoholic Fatty Liver Disease/physiopathology , Obesity/diagnosis , Obesity/immunology , Obesity/physiopathology , Subcutaneous Fat, Abdominal/immunology , Tissue Culture Techniques
2.
Nat Immunol ; 22(5): 639-653, 2021 05.
Article in English | MEDLINE | ID: mdl-33907320

ABSTRACT

White adipose tissue (WAT) is an essential regulator of energy storage and systemic metabolic homeostasis. Regulatory networks consisting of immune and structural cells are necessary to maintain WAT metabolism, which can become impaired during obesity in mammals. Using single-cell transcriptomics and flow cytometry, we unveil a large-scale comprehensive cellular census of the stromal vascular fraction of healthy lean and obese human WAT. We report new subsets and developmental trajectories of adipose-resident innate lymphoid cells, dendritic cells and monocyte-derived macrophage populations that accumulate in obese WAT. Analysis of cell-cell ligand-receptor interactions and obesity-enriched signaling pathways revealed a switch from immunoregulatory mechanisms in lean WAT to inflammatory networks in obese WAT. These results provide a detailed and unbiased cellular landscape of homeostatic and inflammatory circuits in healthy human WAT.


Subject(s)
Immunity, Innate , Obesity/immunology , Subcutaneous Fat, Abdominal/immunology , Abdominoplasty , Adipocytes/immunology , Adipocytes/metabolism , Adult , Cell Communication/immunology , Cell Line , Dendritic Cells, Follicular/immunology , Dendritic Cells, Follicular/metabolism , Female , Humans , Inflammation/immunology , Inflammation/pathology , Lymphocytes/immunology , Lymphocytes/metabolism , Macrophages/immunology , Macrophages/metabolism , Obesity/pathology , Obesity/surgery , RNA-Seq , Signal Transduction/immunology , Single-Cell Analysis , Subcutaneous Fat, Abdominal/pathology , Subcutaneous Fat, Abdominal/surgery
3.
Int J Mol Sci ; 21(2)2020 Jan 12.
Article in English | MEDLINE | ID: mdl-31940889

ABSTRACT

Gestational diabetes (GDM) is among the most challenging diseases in westernized countries, affecting mother and child, immediately and in later life. Obesity is a major risk factor for GDM. However, the impact visceral obesity and related epigenetics play for GDM etiopathogenesis have hardly been considered so far. Our recent findings within the prospective 'EaCH' cohort study of women with GDM or normal glucose tolerance (NGT), showed the role, critical factors of insulin resistance (i.e., adiponectin, insulin receptor) may have for GDM pathophysiology with epigenetically modified expression in subcutaneous (SAT) and visceral (VAT) adipose tissues. Here we investigated the expression and promoter methylation of key inflammatory candidates, tumor necrosis factor-alpha (TNF-α) and suppressor of cytokine signaling 3 (SOCS3) in maternal adipose tissues collected during caesarian section (GDM, n = 19; NGT, n = 22). The mRNA expression of TNF-α and SOCS3 was significantly increased in VAT, but not in SAT, of GDM patients vs. NGT, accompanied by specific alterations of respective promoter methylation patterns. In conclusion, we propose a critical role of VAT and visceral obesity for the pathogenesis of GDM, with epigenetic alterations of the expression of inflammatory factors as a potential factor.


Subject(s)
DNA Methylation , Diabetes, Gestational/immunology , Intra-Abdominal Fat/immunology , Suppressor of Cytokine Signaling 3 Protein/genetics , Tumor Necrosis Factor-alpha/genetics , Adult , Case-Control Studies , Cesarean Section , Diabetes, Gestational/genetics , Epigenesis, Genetic , Female , Humans , Maternal Age , Organ Specificity , Pregnancy , Promoter Regions, Genetic , Subcutaneous Fat, Abdominal/immunology , Up-Regulation
4.
Nutrients ; 10(7)2018 Jul 18.
Article in English | MEDLINE | ID: mdl-30021962

ABSTRACT

Aging is the main factor involved in the onset of degenerative diseases. Dietary protein restriction has been shown to increase the lifespan of rodents and improve metabolic phenotype. Branched-chain amino acids (BCAA) can act as nutrient signals that increase the lifespan of mice after prolonged supplementation. It remains unclear whether the combination of protein restriction and BCAA supplementation improves metabolic and immunological profiles during aging. Here, we investigated how dietary protein levels and BCAA supplementation impact metabolism and immune profile during a 12-month intervention in adult male C57BL/6J mice. We found that protein restriction improved insulin tolerance and increased hepatic fibroblast growth factor 21 mRNA, circulating interleukin (IL)-5 concentration, and thermogenic uncoupling protein 1 in subcutaneous white fat. Surprisingly, BCAA supplementation conditionally increased body weight, lean mass, and fat mass, and deteriorated insulin intolerance during protein restriction, but not during protein sufficiency. BCAA also induced pro-inflammatory gene expression in visceral adipose tissue under both normal and low protein conditions. These results suggest that dietary protein levels and BCAA supplementation coordinate a complex regulation of metabolism and tissue inflammation during prolonged feeding.


Subject(s)
Aging , Amino Acids, Branched-Chain/therapeutic use , Diet, Protein-Restricted , Dietary Proteins/therapeutic use , Dietary Supplements , Gene Expression Regulation, Developmental , Sarcopenia/prevention & control , Adiposity , Amino Acids, Branched-Chain/adverse effects , Amino Acids, Branched-Chain/metabolism , Animals , Cytokines/blood , Diet, Protein-Restricted/adverse effects , Dietary Proteins/adverse effects , Dietary Proteins/metabolism , Dietary Supplements/adverse effects , Gene Expression Profiling , Insulin Resistance , Liver/growth & development , Liver/immunology , Liver/metabolism , Liver/pathology , Male , Mice, Inbred C57BL , Organ Size , Proteomics/methods , Random Allocation , Sarcopenia/immunology , Sarcopenia/metabolism , Sarcopenia/pathology , Spleen/growth & development , Spleen/immunology , Spleen/metabolism , Spleen/pathology , Subcutaneous Fat, Abdominal/growth & development , Subcutaneous Fat, Abdominal/immunology , Subcutaneous Fat, Abdominal/metabolism , Subcutaneous Fat, Abdominal/pathology , Thymus Gland/growth & development , Thymus Gland/immunology , Thymus Gland/metabolism , Thymus Gland/pathology , Weight Gain
5.
Diabetes ; 67(8): 1549-1560, 2018 08.
Article in English | MEDLINE | ID: mdl-29794241

ABSTRACT

Obesity increases the risk of vascular diseases, including aortic aneurysm (AA). Perivascular adipose tissue (PVAT) surrounding arteries are altered during obesity. However, the underlying mechanism of adipose tissue, especially PVAT, in the pathogenesis of AA is still unclear. Here we showed that angiotensin II (AngII) infusion increases the incidence of AA in leptin-deficient obese mice (ob/ob) and high-fat diet-induced obese mice with adventitial inflammation. Furthermore, transcriptome analysis revealed that platelet-derived growth factor-D (PDGF-D) was highly expressed in the PVAT of ob/ob mice. Therefore, we hypothesized that PDGF-D mediates adventitial inflammation, which provides a direct link between PVAT dysfunction and AA formation in AngII-infused obese mice. We found that PDGF-D promotes the proliferation, migration, and inflammatory factors expression in cultured adventitial fibroblasts. In addition, the inhibition of PDGF-D function significantly reduced the incidence of AA in AngII-infused obese mice. More importantly, adipocyte-specific PDGF-D transgenic mice are more susceptible to AA formation after AngII infusion accompanied by exaggerated adventitial inflammatory and fibrotic responses. Collectively, our findings reveal a notable role of PDGF-D in the AA formation during obesity, and modulation of this cytokine might be an exploitable treatment strategy for the condition.


Subject(s)
Aorta, Abdominal/metabolism , Aortic Aneurysm, Abdominal/etiology , Intra-Abdominal Fat/metabolism , Lymphokines/metabolism , Obesity/physiopathology , Platelet-Derived Growth Factor/metabolism , Adventitia/drug effects , Adventitia/immunology , Adventitia/metabolism , Adventitia/pathology , Angiotensin II/administration & dosage , Angiotensin II/adverse effects , Animals , Aorta, Abdominal/diagnostic imaging , Aorta, Abdominal/drug effects , Aorta, Abdominal/pathology , Aortic Aneurysm, Abdominal/diagnostic imaging , Aortic Aneurysm, Abdominal/metabolism , Aortic Aneurysm, Abdominal/pathology , Benzimidazoles/pharmacology , Cells, Cultured , Diet, High-Fat/adverse effects , Drug Implants , Gene Expression Regulation/drug effects , Inflammation Mediators/metabolism , Intra-Abdominal Fat/drug effects , Intra-Abdominal Fat/immunology , Intra-Abdominal Fat/pathology , Lymphokines/agonists , Lymphokines/antagonists & inhibitors , Lymphokines/genetics , Male , Mice , Mice, Mutant Strains , Mice, Transgenic , Obesity/etiology , Obesity/metabolism , Obesity/pathology , Organ Specificity , Platelet-Derived Growth Factor/agonists , Platelet-Derived Growth Factor/antagonists & inhibitors , Platelet-Derived Growth Factor/genetics , Quinolines/pharmacology , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Subcutaneous Fat, Abdominal/drug effects , Subcutaneous Fat, Abdominal/immunology , Subcutaneous Fat, Abdominal/metabolism , Subcutaneous Fat, Abdominal/pathology , Survival Analysis
6.
Endocrinology ; 159(1): 227-237, 2018 01 01.
Article in English | MEDLINE | ID: mdl-29059354

ABSTRACT

The prevalence of primary aldosteronism is much higher than previously thought. Recent studies have shown that primary aldosteronism is related to a higher risk of cardiovascular events. However, the underlying mechanism is not yet clear. Here we investigate the characteristics, including inflammation, fibrosis, and adipokine expression, of adipose tissues from different deposits in patients with aldosterone-producing adenoma (APA). Inflammation and fibrosis changes were evaluated in perirenal and subcutaneous adipose tissues obtained from patients with APA (n = 16), normotension (NT; n = 10), and essential hypertension (EH; n = 5) undergoing laparoscopic surgery. We also evaluated the effect of aldosterone in isolated human perirenal adipose tissue stromal vascular fraction (SVF) cells and investigated the effect of aldosterone in mouse 3T3-L1 and brown preadipocytes. Compared with the EH group, significantly higher levels of interleukin-6 (IL-6) and tumor necrosis factor-α messenger RNA (mRNA) and protein were observed in perirenal adipose tissue of patients with APA. Expression of genes related to fibrosis and adipogenesis in perirenal adipose tissue was notably higher in patients with APA than in patients with NT and EH. Aldosterone significantly induced IL-6 and fibrosis gene mRNA expression in differentiated SVF cells. Aldosterone treatment enhanced mRNA expression of genes associated with inflammation and fibrosis and stimulated differentiation of 3T3-L1 and brown preadipocytes. In conclusion, these data indicate that high aldosterone in patients with APA may induce perirenal adipose tissue dysfunction and lead to inflammation and fibrosis, which may be involved in the high risk of cardiovascular events observed in patients with primary aldosteronism.


Subject(s)
Adenoma/physiopathology , Aldosterone/metabolism , Essential Hypertension/complications , Hyperaldosteronism/etiology , Intra-Abdominal Fat/pathology , Panniculitis/etiology , 3T3-L1 Cells , Adenoma/complications , Adenoma/metabolism , Adenoma/surgery , Adipocytes, Brown/immunology , Adipocytes, Brown/metabolism , Adipocytes, Brown/pathology , Adipogenesis , Adipokines/metabolism , Adrenalectomy , Animals , Cells, Cultured , Endothelium, Vascular/immunology , Endothelium, Vascular/metabolism , Endothelium, Vascular/pathology , Female , Fibrosis , Humans , Intra-Abdominal Fat/immunology , Intra-Abdominal Fat/metabolism , Male , Mice , Middle Aged , Panniculitis/immunology , Panniculitis/metabolism , Panniculitis/pathology , Stromal Cells/immunology , Stromal Cells/metabolism , Stromal Cells/pathology , Subcutaneous Fat, Abdominal/immunology , Subcutaneous Fat, Abdominal/metabolism , Subcutaneous Fat, Abdominal/pathology
7.
Diabetes Obes Metab ; 19(3): 457-462, 2017 03.
Article in English | MEDLINE | ID: mdl-27868366

ABSTRACT

We aimed to investigate the effects of gemigliptin, a dipeptidyl peptidase-4 inhibitor, on postprandial lipoprotein levels and endotoxemia in a randomized, double-blind, placebo-controlled, crossover study. Ten people with type 2 diabetes mellitus (T2DM), inadequately controlled with oral antidiabetic medications and/or lifestyle modification, were randomized to gemigliptin or placebo for 4 weeks. At the end of each treatment phase, the study participants underwent a high-fat meal tolerance test and needle aspiration of abdominal subcutaneous adipose tissue. The median (range) fasting and total area under the curve of apolipoprotein B48 (ApoB48) were significantly lower with gemigliptin than with placebo (2.9 [1.5-15.8] µg/mL vs 4.2 [1.3-23.4] µg/mL; P = .020; 35.3 [14.4-87.4] µg/mL × hour vs 42.2 [17.5-109.0] µg/mL × hour; P = .020, respectively), whereas apolipoprotein B100 showed no significant difference. Serum endotoxin levels were undetectable in 70% of the samples, so we were not able to evaluate the effect of gemigliptin on endotoxemia. The gene expression of inflammatory cytokines in subcutaneous adipose tissue was not affected by gemigliptin. Gemigliptin reduced ApoB48 levels after a high-fat meal in participants with T2DM. Whether systemic endotoxin levels can be reduced by gemigliptin requires further investigation.


Subject(s)
Cytokines/immunology , Diabetes Mellitus, Type 2/drug therapy , Dipeptidyl-Peptidase IV Inhibitors/therapeutic use , Lipid Metabolism , Piperidones/therapeutic use , Pyrimidines/therapeutic use , Adiponectin/metabolism , Adult , Aged , Apolipoprotein B-100/metabolism , Apolipoprotein B-48/metabolism , Blood Glucose/metabolism , C-Reactive Protein/immunology , Cholesterol/metabolism , Cholesterol, HDL/metabolism , Cholesterol, LDL/metabolism , Cross-Over Studies , Cytokines/genetics , Diabetes Mellitus, Type 2/immunology , Diabetes Mellitus, Type 2/metabolism , Diet, High-Fat , Double-Blind Method , Endotoxemia , Endotoxins/blood , Female , Glucagon/metabolism , Glucagon-Like Peptide 1/metabolism , Humans , Insulin/metabolism , Insulin Resistance , Interleukin-6/immunology , Male , Middle Aged , Postprandial Period , RNA, Messenger/metabolism , Subcutaneous Fat, Abdominal/immunology , Subcutaneous Fat, Abdominal/metabolism , Transcriptome , Triglycerides/metabolism , Tumor Necrosis Factor-alpha/immunology
8.
Endocrine ; 57(3): 455-463, 2017 Sep.
Article in English | MEDLINE | ID: mdl-27844208

ABSTRACT

The sirtuin family comprises seven NAD+-dependent deacetylases which control the overall health of organisms through the regulation of pleiotropic metabolic pathways. Sirtuins are important modulators of adipose tissue metabolism and their expression is higher in lean than obese subjects. At present, the role of sirtuins in adipose-derived stem cells has not been investigated yet. Therefore, in this study, we evaluated the expression of the complete panel of sirtuins in adipose-derived stem cells isolated from both subcutaneous and visceral fat of non-obese and obese subjects. We aimed at investigating the influence of obesity on sirtuins' levels, their role in obesity-associated inflammation, and the relationship with the peroxisome proliferator-activated receptor delta, which also plays functions in adipose tissue metabolism. The mRNA levels in the four types of adipose-derived stem cells were evaluated by quantitative polymerase chain reaction, in untreated cells and also after 8 h of hypoxia exposure. Correlations among sirtuins' expression and clinical and molecular parameters were also analyzed. We found that sirtuin1-6 exhibited significant higher mRNA expression in visceral adipose-derived stem cells compared to subcutaneous adipose-derived stem cells of non-obese subjects. Sirtuin1-6 levels were markedly reduced in visceral adipose-derived stem cells of obese patients. Sirtuins' expression in visceral adipose-derived stem cells correlated negatively with body mass index and C-reactive protein and positively with peroxisome proliferator-activated receptor delta. Finally, only in the visceral adipose-derived stem cells of obese patients hypoxia-induced mRNA expression of all of the sirtuins. Our results highlight that sirtuins' levels in adipose-derived stem cells are consistent with protective effects against visceral obesity and inflammation, and suggest a transcriptional mechanism through which acute hypoxia up-regulates sirtuins in the visceral adipose-derived stem cells of obese patients.


Subject(s)
Adult Stem Cells/metabolism , Gene Expression Regulation, Enzymologic , Intra-Abdominal Fat/metabolism , Obesity/metabolism , Overweight/metabolism , Sirtuins/metabolism , Subcutaneous Fat, Abdominal/metabolism , Adult , Adult Stem Cells/immunology , Adult Stem Cells/pathology , Body Mass Index , C-Reactive Protein/analysis , C-Reactive Protein/metabolism , Cell Dedifferentiation , Cell Hypoxia , Cells, Cultured , Cohort Studies , Female , Humans , Intra-Abdominal Fat/immunology , Intra-Abdominal Fat/pathology , Isoenzymes/genetics , Isoenzymes/metabolism , Male , Middle Aged , Obesity/blood , Obesity/immunology , Obesity/pathology , Overweight/blood , Overweight/immunology , Overweight/pathology , PPAR delta/genetics , PPAR delta/metabolism , RNA, Messenger/metabolism , Reproducibility of Results , Sirtuins/genetics , Subcutaneous Fat, Abdominal/immunology , Subcutaneous Fat, Abdominal/pathology
9.
J Immunol ; 197(9): 3735-3745, 2016 11 01.
Article in English | MEDLINE | ID: mdl-27698011

ABSTRACT

Adipose tissue (AT) macrophages (ATMs) are key players for regulation of AT homeostasis and obesity-related metabolic disorders. However, the phenotypes of human ATMs and regulatory mechanisms of their polarization have not been clearly described. In this study, we investigated human ATMs in both abdominal visceral AT and s.c. AT and proposed an 11ß-hydroxysteroid dehydrogenase type 1 (11ß-HSD1)-glucocorticoid receptor regulatory axis that might dictate M1/M2 polarization in ATMs. The accumulation of CD11c+CD163+ ATMs in both visceral AT and s.c. AT of obese individuals was confirmed at the cellular level and was found to be clearly correlated with body mass index and production of reactive oxygen species. Using our in vitro system where human peripheral blood monocytes (hPBMs) were cocultured with Simpson-Golabi-Behmel syndrome adipocytes, M1/M2 polarization was found to be dependent on 11ß-HSD1, an intracellular glucocorticoid reactivating enzyme. Exposure of hPBMs to cortisol-induced expression of CD163 and RU-486, a glucocorticoid receptor antagonist, significantly abrogated CD163 expression through coculture of mature adipocytes with hPBMs. Moreover, 11ß-HSD1 was expressed in crown ATMs in obese AT. Importantly, conditioned medium from coculture of adipocytes with hPBMs enhanced proliferation of human breast cancer MCF7 and MDA-MB-231 cells. In summary, the phenotypic switch of ATMs from M2 to mixed M1/M2 phenotype occurred through differentiation of adipocytes in obese individuals, and upregulation of intracellular 11ß-HSD1 might play a role in the process.


Subject(s)
11-beta-Hydroxysteroid Dehydrogenase Type 1/metabolism , Arrhythmias, Cardiac/immunology , Breast Neoplasms/immunology , Genetic Diseases, X-Linked/immunology , Gigantism/immunology , Heart Defects, Congenital/immunology , Intellectual Disability/immunology , Intra-Abdominal Fat/immunology , Macrophages/immunology , Obesity/immunology , Subcutaneous Fat, Abdominal/immunology , 11-beta-Hydroxysteroid Dehydrogenase Type 1/genetics , Adipocytes/pathology , Adult , Aged , Antigens, CD/metabolism , Antigens, Differentiation, Myelomonocytic/metabolism , CD11c Antigen/metabolism , Cell Differentiation , Cell Proliferation , Coculture Techniques , Cytokines/metabolism , Female , Humans , MCF-7 Cells , Male , Middle Aged , Receptors, Cell Surface/metabolism , Up-Regulation , Young Adult
10.
J Clin Endocrinol Metab ; 101(11): 4021-4029, 2016 Nov.
Article in English | MEDLINE | ID: mdl-27459538

ABSTRACT

CONTEXT: Cardiometabolic complications in obesity may be linked to white adipose tissue (WAT) dysfunction. Transcriptomic studies of Sc WAT have reported that CCL18, encoding the CC chemokine ligand 18 (CCL18), is increased in obesity/insulin resistance but its functional role is unknown. OBJECTIVE: Our objectives were to determine if CCL18 is secreted from Sc WAT and if secreted and/or serum levels associate with metabolic phenotypes. We also planned to define the primary cellular source and if CCL18 exerts effects on adipocytes. DESIGN: This is a cohort study. SETTING: The study took place in an outpatient academic clinic. PARTICIPANTS: A total of 130 obese women scheduled for bariatric surgery and 35 nonobese controls were included. METHODS: Insulin sensitivity was assessed by hyperinsulinemic euglycemic clamp or homeostasis model assessment. CCL18 was analyzed in serum/WAT incubates by ELISA. Effects of recombinant CCL18 was determined in cultures of primary human adipocytes and the monocyte cell line THP-1 differentiated into M0/M1/M2 macrophages. MAIN OUTCOME MEASURE: Association with metabolic risk factors was measured. RESULTS: CCL18 was secreted from WAT and the levels correlated positively with insulin resistance, Adult Treatment Panel III risk score and plasma triglycerides, independent of body mass index and better than other established adipocytokines. In 80 obese women, S-CCL18 levels were significantly higher in insulin resistant compared with insulin sensitive subjects. In WAT CCL18 mRNA was expressed in macrophages and correlated positively with immune-related genes, particularly those enriched in M2 macrophages. While CCL18 increased cyto-/chemokine expression in M0/M2-THP-1 cells, human adipocytes showed no responses in vitro. CONCLUSIONS: Circulating and WAT-secreted CCL18 correlates with insulin resistance and metabolic risk score. Because CCL18 is macrophage-specific and associates with adipose immune gene expression, it may constitute a marker of WAT inflammation.


Subject(s)
Adiposity , Chemokines, CC/metabolism , Macrophages/metabolism , Metabolic Syndrome/etiology , Obesity, Morbid/metabolism , Panniculitis/etiology , Subcutaneous Fat, Abdominal/metabolism , Adult , Bariatric Surgery , Biomarkers/blood , Biomarkers/metabolism , Body Mass Index , Cell Line , Cells, Cultured , Chemokines, CC/blood , Chemokines, CC/genetics , Cohort Studies , Female , Gene Expression Regulation , Gene Ontology , Humans , Hypertriglyceridemia/etiology , Insulin Resistance , Macrophages/immunology , Macrophages/pathology , Metabolic Syndrome/epidemiology , Obesity, Morbid/immunology , Obesity, Morbid/pathology , Obesity, Morbid/physiopathology , Recombinant Proteins/metabolism , Risk Factors , Subcutaneous Fat, Abdominal/immunology , Subcutaneous Fat, Abdominal/pathology , Sweden/epidemiology
11.
Diabetes ; 65(5): 1380-6, 2016 05.
Article in English | MEDLINE | ID: mdl-26884439

ABSTRACT

Most often, diabetic ketoacidosis (DKA) in adults results from insufficient insulin administration and acute infection. DKA is assumed to release proinflammatory cytokines and stress hormones that stimulate lipolysis and ketogenesis. We tested whether this perception of DKA can be reproduced in an experimental human model by using combined insulin deficiency and acute inflammation and tested which intracellular mediators of lipolysis are affected in adipose tissue. Nine subjects with type 1 diabetes were studied twice: 1) insulin-controlled euglycemia and 2) insulin deprivation and endotoxin administration (KET). During KET, serum tumor necrosis factor-α, cortisol, glucagon, and growth hormone levels increased, and free fatty acids and 3-hydroxybutyrate concentrations and the rate of lipolysis rose markedly. Serum bicarbonate and pH decreased. Adipose tissue mRNA contents of comparative gene identification-58 (CGI-58) increased and G0/G1 switch 2 gene (G0S2) mRNA decreased robustly. Neither protein levels of adipose triglyceride lipase (ATGL) nor phosphorylations of hormone-sensitive lipase were altered. The clinical picture of incipient DKA in adults can be reproduced by combined insulin deficiency and endotoxin-induced acute inflammation. The precipitating steps involve the release of proinflammatory cytokines and stress hormones, increased lipolysis, and decreased G0S2 and increased CGI-58 mRNA contents in adipose tissue, compatible with latent ATGL stimulation.


Subject(s)
Diabetes Mellitus, Type 1/immunology , Diabetic Ketoacidosis/immunology , Lipolysis , Models, Immunological , Panniculitis/immunology , Signal Transduction , Subcutaneous Fat, Abdominal/immunology , 1-Acylglycerol-3-Phosphate O-Acyltransferase/genetics , 1-Acylglycerol-3-Phosphate O-Acyltransferase/metabolism , Adult , Biopsy , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cross-Over Studies , Diabetes Mellitus, Type 1/drug therapy , Diabetes Mellitus, Type 1/metabolism , Diabetes Mellitus, Type 1/pathology , Diabetic Ketoacidosis/metabolism , Diabetic Ketoacidosis/pathology , Diabetic Ketoacidosis/prevention & control , Endotoxins/toxicity , Gene Expression Regulation/drug effects , Humans , Hyperglycemia/chemically induced , Hyperglycemia/prevention & control , Hypoglycemic Agents/administration & dosage , Hypoglycemic Agents/therapeutic use , Insulin/administration & dosage , Insulin/therapeutic use , Insulin, Long-Acting/administration & dosage , Insulin, Long-Acting/therapeutic use , Insulin, Short-Acting/administration & dosage , Insulin, Short-Acting/therapeutic use , Lipolysis/drug effects , Male , Panniculitis/drug therapy , Panniculitis/metabolism , Panniculitis/pathology , Signal Transduction/drug effects , Subcutaneous Fat, Abdominal/drug effects , Subcutaneous Fat, Abdominal/metabolism , Subcutaneous Fat, Abdominal/pathology , Young Adult
12.
J Clin Endocrinol Metab ; 100(11): E1467-76, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26252355

ABSTRACT

CONTEXT: Molecular mechanisms associated with physiological variations in adipose tissue (AT) are not fully recognized. The most recent reports highlight the critical relevance of microRNAs (miRNAs) found in AT. OBJECTIVE: To identify changes in messenger RNA (mRNA) and miRNA expressions and their interaction in human AT before and after surgery-induced weight loss. Research Design and Setting: Genome-wide mRNA and miRNA expressions were assessed by microarrays in abdominal subcutaneous AT of 16 morbidly obese women before and 2 years after laparoscopic Roux-en-Y gastric bypass. The association of changes in miRNAs with their respective mRNA targets was studied. The results were replicated in publicly available microarray datasets. Validation was made by real-time polymerase chain reaction in additional fat samples from 26 age-matched lean women and in isolated human adipocytes. RESULTS: A total of 5018 different mRNA probe sets and 15 miRNAs were differentially expressed after surgery-induced weight loss. The clustering of similar expression patterns for gene products with related functions revealed molecular footprints that elucidate significant changes in cell cycle, development, lipid metabolism, and the inflammatory response. The participation of inflammation was demonstrated by results assessed in isolated adipocytes. Interestingly, when transcriptomes were analyzed taking into account the presence of miRNA target sites, miRNA target mRNAs were upregulated in obese AT (P value = 2 × 10(-181)) and inflamed adipocytes (P value = 4 × 10(-61)), according to the number of target sites harbored by each transcript. CONCLUSIONS: Current findings suggest impaired miRNA target gene expression in obese AT in close association with inflammation, both improving after weight loss.


Subject(s)
Down-Regulation , Gastric Bypass , MicroRNAs/metabolism , Obesity, Morbid/surgery , Subcutaneous Fat, Abdominal/metabolism , Adipocytes, White/cytology , Adipocytes, White/immunology , Adipocytes, White/metabolism , Adult , Body Mass Index , Cell Line , Cells, Cultured , Cohort Studies , Cross-Sectional Studies , Female , Gene Expression Profiling , Genome-Wide Association Study , Humans , Longitudinal Studies , Middle Aged , Monocytes/immunology , Monocytes/metabolism , Obesity, Morbid/genetics , Obesity, Morbid/immunology , Obesity, Morbid/metabolism , RNA, Messenger/metabolism , Subcutaneous Fat, Abdominal/immunology , Weight Loss
13.
Am J Clin Nutr ; 102(2): 433-43, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26156741

ABSTRACT

BACKGROUND: Metabolic and transcriptomic differences between visceral adipose tissue (VAT) and subcutaneous adipose tissue (SAT) compartments, particularly in the context of obesity, may play a role in colorectal carcinogenesis. We investigated the differential functions of their metabolic compositions. OBJECTIVES: Biochemical differences between adipose tissues (VAT compared with SAT) in patients with colorectal carcinoma (CRC) were investigated by using mass spectrometry metabolomics and gene expression profiling. Metabolite compositions were compared between VAT, SAT, and serum metabolites. The relation between patients' tumor stage and metabolic profiles was assessed. DESIGN: Presurgery blood and paired VAT and SAT samples during tumor surgery were obtained from 59 CRC patients (tumor stages I-IV) of the ColoCare cohort. Gas chromatography time-of-flight mass spectrometry and liquid chromatography quadrupole time-of-flight mass spectrometry were used to measure 1065 metabolites in adipose tissue (333 identified compounds) and 1810 metabolites in serum (467 identified compounds). Adipose tissue gene expression was measured by using Illumina's HumanHT-12 Expression BeadChips. RESULTS: Compared with SAT, VAT displayed elevated markers of inflammatory lipid metabolism, free arachidonic acid, phospholipases (PLA2G10), and prostaglandin synthesis-related enzymes (PTGD/PTGS2S). Plasmalogen concentrations were lower in VAT than in SAT, which was supported by lower gene expression of FAR1, the rate-limiting enzyme for ether-lipid synthesis in VAT. Serum sphingomyelin concentrations were inversely correlated (P = 0.0001) with SAT adipose triglycerides. Logistic regression identified lipids in patients' adipose tissues, which were associated with CRC tumor stage. CONCLUSIONS: As one of the first studies, we comprehensively assessed differences in metabolic, lipidomic, and transcriptomic profiles between paired human VAT and SAT and their association with CRC tumor stage. We identified markers of inflammation in VAT, which supports prior evidence regarding the role of visceral adiposity and cancer.


Subject(s)
Carcinoma/physiopathology , Colorectal Neoplasms/physiopathology , Gene Expression Regulation, Neoplastic , Intra-Abdominal Fat/metabolism , Panniculitis/etiology , Paraneoplastic Syndromes/etiology , Subcutaneous Fat, Abdominal/metabolism , Aged , Biomarkers/blood , Biomarkers/metabolism , Carcinoma/metabolism , Carcinoma/pathology , Carcinoma/surgery , Cohort Studies , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Colorectal Neoplasms/surgery , Female , Gene Expression Profiling , Humans , Intra-Abdominal Fat/immunology , Intra-Abdominal Fat/pathology , Lipid Metabolism , Male , Metabolomics/methods , Middle Aged , Neoplasm Staging , Oligonucleotide Array Sequence Analysis , Panniculitis/immunology , Panniculitis/metabolism , Panniculitis/pathology , Paraneoplastic Syndromes/immunology , Paraneoplastic Syndromes/metabolism , Paraneoplastic Syndromes/pathology , Prospective Studies , Subcutaneous Fat, Abdominal/immunology , Subcutaneous Fat, Abdominal/pathology
14.
PLoS One ; 10(3): e0122872, 2015.
Article in English | MEDLINE | ID: mdl-25894202

ABSTRACT

BACKGROUND/OBJECTIVES: Hyperglycemia represents one of possible mediators for activation of immune system and may contribute to worsening of inflammatory state associated with obesity. The aim of our study was to investigate the effect of a short-term hyperglycemia (HG) on the phenotype and relative content of immune cells in circulation and subcutaneous abdominal adipose tissue (SAAT) in obese women without metabolic complications. SUBJECTS/METHODS: Three hour HG clamp with infusion of octreotide and control investigations with infusion of octreotide or saline were performed in three groups of obese women (Group1: HG, Group 2: Octreotide, Group 3: Saline, n=10 per group). Before and at the end of the interventions, samples of SAAT and blood were obtained. The relative content of immune cells in blood and SAAT was determined by flow cytometry. Gene expression analysis of immunity-related markers in SAAT was performed by quantitative real-time PCR. RESULTS: In blood, no changes in analysed immune cell population were observed in response to HG. In SAAT, HG induced an increase in the content of CD206 negative monocytes/macrophages (p<0.05) and T lymphocytes (both T helper and T cytotoxic lymphocytes, p<0.01). Further, HG promoted an increase of mRNA levels of immune response markers (CCL2, TLR4, TNFα) and lymphocyte markers (CD3g, CD4, CD8a, TBX21, GATA3, FoxP3) in SAAT (p<0.05 and 0.01). Under both control infusions, none of these changes were observed. CONCLUSIONS: Acute HG significantly increased the content of monocytes and lymphocytes in SAAT of healthy obese women. This result suggests that the short-term HG can modulate an immune status of AT in obese subjects.


Subject(s)
Health , Hyperglycemia/chemically induced , Hyperglycemia/immunology , Monocytes/cytology , Obesity/complications , Subcutaneous Fat, Abdominal/immunology , T-Lymphocytes/cytology , Adult , Biomarkers/metabolism , Blood Glucose/metabolism , C-Peptide/blood , Cell Count , Female , Gene Expression Regulation/drug effects , Humans , Hyperglycemia/blood , Hyperglycemia/complications , Insulin/blood , Macrophages/cytology , Macrophages/drug effects , Monocytes/drug effects , Octreotide/pharmacology , Phenotype , RNA, Messenger/genetics , RNA, Messenger/metabolism , Subcutaneous Fat, Abdominal/drug effects , T-Lymphocytes/drug effects
15.
Trends Endocrinol Metab ; 26(4): 193-200, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25703677

ABSTRACT

The adipose tissue is crucial in regulating insulin sensitivity and risk for diabetes through its lipid storage capacity and thermogenic and endocrine functions. Subcutaneous adipose tissue (SAT) stores excess lipids through expansion of adipocytes (hypertrophic obesity) and/or recruitment of new precursor cells (hyperplastic obesity). Hypertrophic obesity in humans, a characteristic of genetic predisposition for diabetes, is associated with abdominal obesity, ectopic fat accumulation, and the metabolic syndrome (MS), while the ability to recruit new adipocytes prevents this. We review the regulation of adipogenesis, its relation to SAT expandability and the risks of ectopic fat accumulation, and insulin resistance. The actions of GLUT4 in SAT, including a novel family of lipids enhancing insulin sensitivity/secretion, and the function of bone morphogenetic proteins (BMPs) in white and beige/brown adipogenesis in humans are highlighted.


Subject(s)
Adipogenesis , Down-Regulation , Glucose Transporter Type 4/metabolism , Insulin Resistance , Metabolic Syndrome/metabolism , Models, Biological , Subcutaneous Fat, Abdominal/metabolism , Adipose Tissue, Brown/immunology , Adipose Tissue, Brown/metabolism , Adipose Tissue, Brown/pathology , Adiposity , Animals , Glucose Transporter Type 4/genetics , Humans , Hypertrophy , Lipid Metabolism , Metabolic Syndrome/genetics , Metabolic Syndrome/immunology , Metabolic Syndrome/pathology , Organ Specificity , Subcutaneous Fat, Abdominal/immunology , Subcutaneous Fat, Abdominal/pathology
16.
Am J Clin Nutr ; 101(1): 228-39, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25527767

ABSTRACT

BACKGROUND: Previously, a healthy Nordic diet (ND) has been shown to have beneficial health effects close to those of Mediterranean diets. OBJECTIVE: The objective was to explore whether the ND has an impact on gene expression in abdominal subcutaneous adipose tissue (SAT) and whether changes in gene expression are associated with clinical and biochemical effects. DESIGN: Obese adults with features of the metabolic syndrome underwent an 18- to 24-wk randomized intervention study comparing the ND with the control diet (CD) (the SYSDIET study, carried out within Nordic Centre of Excellence of the Systems Biology in Controlled Dietary Interventions and Cohort Studies). The present study included participants from 3 Nordic SYSDIET centers [Kuopio (n = 20), Lund (n = 18), and Oulu (n = 18)] with a maximum weight change of ±4 kg, highly sensitive C-reactive protein concentration <10 mg/L at the beginning and the end of the intervention, and baseline body mass index (in kg/m²) <38. SAT biopsy specimens were obtained before and after the intervention and subjected to global transcriptome analysis with Gene 1.1 ST Arrays (Affymetrix). RESULTS: Altogether, 128 genes were differentially expressed in SAT between the ND and CD (nominal P < 0.01; false discovery rate, 25%). These genes were overrepresented in pathways related to immune response (adjusted P = 0.0076), resulting mainly from slightly decreased expression in the ND and increased expression in the CD. Immune-related pathways included leukocyte trafficking and macrophage recruitment (e.g., interferon regulatory factor 1, CD97), adaptive immune response (interleukin32, interleukin 6 receptor), and reactive oxygen species (neutrophil cytosolic factor 1). Interestingly, the regulatory region of the 128 genes was overrepresented for binding sites for the nuclear transcription factor κB. CONCLUSION: A healthy Nordic diet reduces inflammatory gene expression in SAT compared with a control diet independently of body weight change in individuals with features of the metabolic syndrome.


Subject(s)
Diet , Down-Regulation , Gene Expression Regulation , Health Promotion , Metabolic Syndrome/diet therapy , Nutrition Policy , Subcutaneous Fat, Abdominal/metabolism , Adaptive Immunity , Biopsy , Body Mass Index , C-Reactive Protein/analysis , Diet/ethnology , Female , Finland , Gene Expression Profiling , Humans , Male , Metabolic Syndrome/complications , Metabolic Syndrome/immunology , Metabolic Syndrome/metabolism , Middle Aged , Obesity/complications , Oligonucleotide Array Sequence Analysis , Subcutaneous Fat, Abdominal/immunology , Subcutaneous Fat, Abdominal/pathology , Sweden
17.
Br J Nutr ; 111(3): 445-51, 2014 Feb.
Article in English | MEDLINE | ID: mdl-23947577

ABSTRACT

The objective of the present study was to investigate the optimal dietary n-6:n-3 PUFA ratios that regulate lipid metabolism and inflammation in pigs. A total of ninety-six cross-bred (Large White × Landrace) growing-finishing pigs (73·8 (SEM 1·6) kg) were chosen and fed one of the four isoenergetic diets with n-6:n-3 PUFA ratios of 1:1, 2·5:1, 5:1 and 10:1. The growth performance of pigs fed the diet with an n-6:n-3 PUFA ratio of 5:1 was the best, but the group fed the diet with an n-6:n-3 PUFA ratio of 1:1 had the highest muscle mass and the lowest adipose tissue mass (P< 0·05). The concentrations of IL-6 and IL-1ß of pigs fed the diet with an n-6:n-3 PUFA ratio of 1:1 were decreased compared with those of the other groups (P< 0·05). The concentration of adiponectin of pigs fed the diet with an n-6:n-3 PUFA ratio of 1:1 was also markedly decreased, but the concentration of leptin was increased compared with that of the groups fed the diets with n-6:n-3 PUFA ratios of 5:1 and 10:1 (P< 0·05). Additionally, the optimal dietary ratios of n-6:n-3 PUFA of 1:1 and 5:1 markedly suppressed the expression levels of lipid metabolism-related genes and proteins such as phosphoinositide-3-kinase-α, fatty acid transport protein-1 and PPARγ. They also significantly suppressed the expression levels of the inflammatory cytokines IL-1ß, TNF-α and IL-6. The results indicated that the optimal n-6:n-3 PUFA ratios of 1:1 and 5:1 exerted beneficial effects on lipid metabolism and inflammatory system, leading to the availability of more energy and nutrients for high performance and homeostatic pathways.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/metabolism , Dietary Fats/metabolism , Fatty Acids, Omega-3/metabolism , Fatty Acids, Omega-6/metabolism , Lipid Metabolism , Muscle, Skeletal/metabolism , Subcutaneous Fat, Abdominal/metabolism , Adipokines/agonists , Adipokines/antagonists & inhibitors , Adipokines/blood , Adipokines/metabolism , Adiposity , Animals , Anti-Inflammatory Agents, Non-Steroidal/adverse effects , Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , China , Crosses, Genetic , Cytokines/agonists , Cytokines/antagonists & inhibitors , Cytokines/blood , Cytokines/metabolism , Dietary Fats/adverse effects , Dietary Fats/therapeutic use , Energy Intake , Fatty Acids, Omega-3/adverse effects , Fatty Acids, Omega-3/therapeutic use , Fatty Acids, Omega-6/adverse effects , Fatty Acids, Omega-6/therapeutic use , Gene Expression Regulation, Developmental , Hypolipidemic Agents/adverse effects , Hypolipidemic Agents/metabolism , Hypolipidemic Agents/therapeutic use , Male , Muscle Development , Muscle, Skeletal/growth & development , Muscle, Skeletal/immunology , Random Allocation , Subcutaneous Fat, Abdominal/growth & development , Subcutaneous Fat, Abdominal/immunology , Sus scrofa , Weight Gain
18.
Obesity (Silver Spring) ; 22(2): 418-25, 2014 Feb.
Article in English | MEDLINE | ID: mdl-23804488

ABSTRACT

OBJECTIVE: Circulating macrophage migration inhibitory factor (MIF) levels have been shown to positively correlate with body mass index (BMI) in humans. Our objective in this study was to determine the effects of MIF deficiency in a model of high-fat diet-induced obesity. DESIGN AND METHODS: MIF wild type (MIF WT) and MIF deficient (MIF(-/-)) C57Bl/6J mice were fed a high-fat diet (HFD) for up to 15 weeks. Weight and metabolic responses were measured over the course of the disease. Immune cell infiltrates in visceral and subcutaneous adipose tissue were examined by flow cytometry. RESULTS: There was no difference in weight gain or adipose tissue mass in MIF(-/-) mice compared to MIF WT mice. Both groups fed HFD developed glucose intolerance at the same rate and had similar elevations in fasted blood insulin. MDSC abundance was evaluated and showed no MIF-dependent differences. Macrophages were elevated in the visceral adipose tissue of obese mice, but there was no difference between the two groups. CONCLUSIONS: While HFD feeding induced obesity with the expected perturbations in glucose homeostasis and adipose tissue inflammation, the presence or absence of MIF had no effect on any parameter examined.


Subject(s)
Glucose Intolerance/etiology , Intra-Abdominal Fat/metabolism , Macrophage Migration-Inhibitory Factors/metabolism , Macrophages/metabolism , Monocytes/metabolism , Obesity/metabolism , Subcutaneous Fat, Abdominal/metabolism , Adiposity , Animals , Diet, High-Fat/adverse effects , Disease Progression , Hyperinsulinism/etiology , Intra-Abdominal Fat/immunology , Intra-Abdominal Fat/pathology , Lipid Metabolism , Liver/immunology , Liver/metabolism , Liver/pathology , Macrophage Migration-Inhibitory Factors/deficiency , Macrophage Migration-Inhibitory Factors/genetics , Macrophages/immunology , Macrophages/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Monocytes/immunology , Monocytes/pathology , Obesity/immunology , Obesity/pathology , Obesity/physiopathology , Subcutaneous Fat, Abdominal/immunology , Subcutaneous Fat, Abdominal/pathology , Time Factors , Weight Gain
19.
Am J Clin Nutr ; 98(6): 1385-94, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24172304

ABSTRACT

BACKGROUND: The ability to identify obese subjects who will lose weight in response to energy restriction is an important strategy in obesity treatment. OBJECTIVE: We aimed to identify obese subjects who would lose weight and maintain weight loss through 6 wk of energy restriction and 6 wk of weight maintenance. DESIGN: Fifty obese or overweight subjects underwent a 6-wk energy-restricted, high-protein diet followed by another 6 wk of weight maintenance. Network modeling by using combined biological, gut microbiota, and environmental factors was performed to identify predictors of weight trajectories. RESULTS: On the basis of body weight trajectories, 3 subject clusters were identified. Clusters A and B lost more weight during energy restriction. During the stabilization phase, cluster A continued to lose weight, whereas cluster B remained stable. Cluster C lost less and rapidly regained weight during the stabilization period. At baseline, cluster C had the highest plasma insulin, interleukin (IL)-6, adipose tissue inflammation (HAM56+ cells), and Lactobacillus/Leuconostoc/Pediococcus numbers in fecal samples. Weight regain after energy restriction correlated positively with insulin resistance (homeostasis model assessment of insulin resistance: r = 0.5, P = 0.0002) and inflammatory markers (IL-6; r = 0.43, P = 0.002) at baseline. The Bayesian network identified plasma insulin, IL-6, leukocyte number, and adipose tissue (HAM56) at baseline as predictors that were sufficient to characterize the 3 clusters. The prediction accuracy reached 75.5%. CONCLUSION: The resistance to weight loss and proneness to weight regain could be predicted by the combination of high plasma insulin and inflammatory markers before dietary intervention.


Subject(s)
Caloric Restriction , Insulin Resistance , Leukocytes/immunology , Obesity/diet therapy , Overweight/diet therapy , Subcutaneous Fat, Abdominal/immunology , Adult , Antibodies, Monoclonal/metabolism , Bayes Theorem , Biomarkers/blood , Biomarkers/metabolism , Body Mass Index , Female , Humans , Insulin/blood , Interleukin-6/blood , Kinetics , Male , Middle Aged , Obesity/immunology , Obesity/metabolism , Obesity/prevention & control , Overweight/immunology , Overweight/metabolism , Overweight/prevention & control , ROC Curve , Secondary Prevention , Subcutaneous Fat, Abdominal/metabolism , Weight Gain , Weight Loss
20.
J Clin Endocrinol Metab ; 98(12): E1891-900, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24152681

ABSTRACT

CONTEXT: Recently cartonectin was reported as a novel adipokine, with lower levels in diet-induced obese mice, glucose-lowering effects, and antiinflammatory and cardioprotective properties. Polycystic ovary syndrome (PCOS) is a proinflammatory state associated with obesity, diabetes, dyslipidemia, and cardiovascular complications. OBJECTIVES: The objective of the study was to investigate cartonectin levels and regulation in sera and adipose tissue (AT) as well as the effects of metformin of women with PCOS and control subjects. DESIGN: This was a cross-sectional study [PCOS (n = 83) and control (n = 39) subjects]. Real-time PCR and Western blotting were used to assess mRNA and protein expression of cartonectin. Serum cartonectin was measured by an ELISA. RESULTS: Serum and omental adipose tissue cartonectin were significantly lower in women with PCOS compared with control subjects (P < .05 and P < .01, respectively). Furthermore, cartonectin showed a significant negative association with body mass index, waist to hip ratio, glucose, insulin, total cholesterol, low-density lipoprotein-cholesterol, triglycerides, High sensitivity C-reactive protein (hs-CRP) and intima-media thickness (P < .05 and P < .01, respectively); in multiple regression analyses, triglycerides (P =.040) and hs-CRP (P = .031) were predictive of cartonectin levels (P < .05). After 6 months of metformin treatment, there was an associated increase in serum cartonectin (P < .05). Importantly, changes in hs-CRP were significantly negatively correlated with changes in serum cartonectin (P = .033). Finally, cartonectin protein production and secretion into conditioned media were significantly increased by metformin in control human omental AT explants (P < .05). CONCLUSIONS: Serum and omental AT cartonectin are lower in women with PCOS. Metformin treatment increases serum cartonectin levels in these women and in omental AT explants.


Subject(s)
Adipokines/agonists , Hypoglycemic Agents/therapeutic use , Metformin/therapeutic use , Polycystic Ovary Syndrome/drug therapy , Subcutaneous Fat, Abdominal/drug effects , Tumor Necrosis Factors/agonists , Up-Regulation/drug effects , Adipokines/genetics , Adipokines/metabolism , Adult , Biomarkers/blood , Biomarkers/metabolism , Body Mass Index , C-Reactive Protein/analysis , Cardiovascular Diseases/complications , Cardiovascular Diseases/prevention & control , Carotid Intima-Media Thickness , Cross-Sectional Studies , Female , Humans , Hypertriglyceridemia/complications , Omentum , Overweight/complications , Polycystic Ovary Syndrome/complications , Polycystic Ovary Syndrome/immunology , Polycystic Ovary Syndrome/metabolism , Subcutaneous Fat, Abdominal/immunology , Subcutaneous Fat, Abdominal/metabolism , Tissue Culture Techniques , Tumor Necrosis Factors/genetics , Tumor Necrosis Factors/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...