Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 38
Filter
1.
Exp Clin Endocrinol Diabetes ; 130(1): 55-60, 2022 Jan.
Article in English | MEDLINE | ID: mdl-32767285

ABSTRACT

BACKGROUND AND STUDY AIMS: In patients with obesity and type-2 diabetes, short-time very low-calorie diet may ameliorate hyperglycemia and hepatic steatosis. Whether this also implies the glucose-regulating hormone glucagon remains to be elucidated. This study investigated the effects of a very low-calorie diet on plasma levels of glucagon and liver fat in obese patients with type-2 diabetes. PATIENTS AND METHODS: Ten obese patients with type-2 diabetes, 6 men and 4 women, were included. At baseline, fasting plasma glucagon, insulin and glucose were determined, and liver fat and stiffness evaluated by transient elastography. The subjects were then prescribed a very low-calorie diet of maximum 800 kcal/day for 7 weeks and reexamined after 7 weeks and 12 months. RESULTS: At baseline, BMI was 42±4 kg/m2 and fasting glucose 10.6±3.4 mmol/l. All patients had hepatic steatosis. Plasma glucagon was strongly related to liver fat (r2=0.52, p=0.018). After 7 weeks of very low-calorie diet, plasma glucagon was significantly decreased by nearly 30% (p=0.004) along with reductions of BMI (p<0.0001), glucose (p=0.02), insulin (p=0.03), liver fat (p=0.007) and liver stiffness (p=0.05). At 12 months follow-up, both glucagon and liver fat increased and were not different to basal levels, despite persistent reductions of BMI (p<0.002) and glucose (p=0.008). CONCLUSION: In obese type-2 diabetic subjects, plasma glucagon and liver fat are correlated and similarly affected by a very low-calorie diet, supporting a role of hepatic steatosis in glucagon metabolism.


Subject(s)
Caloric Restriction , Diabetes Mellitus, Type 2/diet therapy , Fatty Liver/diet therapy , Glucagon/blood , Obesity/drug therapy , Blood Glucose , Body Mass Index , Female , Humans , Insulin/blood , Male , Middle Aged , Outcome Assessment, Health Care
2.
Eur J Gastroenterol Hepatol ; 33(11): 1420-1426, 2021 11 01.
Article in English | MEDLINE | ID: mdl-32796353

ABSTRACT

BACKGROUND AND AIMS: It is unclear if improving glycemic control in persons with type 2 diabetes (T2D) also has liver-related effects. We aimed to evaluate if a personalized treatment program associates with improvement of liver-related parameters in persons with advanced T2D in a real-life setting. METHODS: Persons with advanced T2D underwent a 4-day personalized treatment program, with the aim of improving glycemic control by dietary advice, instructions on how to achieve optimal glucose control and individualized dosage of medications. Transient elastography was used to estimate liver steatosis and fibrosis. Persons with liver diseases other than non-alcoholic fatty liver disease (NAFLD) were excluded. After 3 months, study participants were offered re-examination. RESULTS: Ninety-one persons were included. Of these, 75 persons (82%) had controlled attenuation parameter (CAP) measurements of acceptable quality at baseline. Of these, 57 (76%) had NAFLD (defined as >268 dB/m). Twenty-two persons (24%) had elevated liver stiffness (>7.9 kPa), and eight (9%) had liver stiffness above 13.9 kPa, indicating advanced fibrosis. Over a median follow-up of 101 days, mean CAP in persons with NAFLD was reduced by 18.33 dB/m (P = 0.035). In persons with elevated liver stiffness, mean stiffness was reduced by 2.6 kPa (P = 0.047). In linear regression, one-unit improvement in fasting glucose (mg/dl) was associated with a decrease in hepatic steatosis with 0.48 dB/m (adjusted R2 = 0.35, P < 0.01). CONCLUSION: The prevalence of NAFLD with advanced fibrosis is high in persons with advanced T2D. Improving glycemic control through a personalized treatment program is associated with a reduction in liver steatosis and stiffness in this cohort.


Subject(s)
Diabetes Mellitus, Type 2 , Elasticity Imaging Techniques , Non-alcoholic Fatty Liver Disease , Diabetes Mellitus, Type 2/diagnosis , Diabetes Mellitus, Type 2/drug therapy , Diabetes Mellitus, Type 2/epidemiology , Humans , Liver/diagnostic imaging , Liver/pathology , Liver Cirrhosis/diagnosis , Liver Cirrhosis/pathology , Non-alcoholic Fatty Liver Disease/diagnosis , Non-alcoholic Fatty Liver Disease/epidemiology , Non-alcoholic Fatty Liver Disease/pathology , Precision Medicine
3.
Lakartidningen ; 1142017 02 17.
Article in Swedish | MEDLINE | ID: mdl-28221405

ABSTRACT

New guidelines for NAFLD - a Swedish perspective Non-alcoholic fatty liver disease (NAFLD) is the most common liver disease globally, with an estimated prevalence of 25%. It is clinically challenging to identify which persons with known or suspected NAFLD that have the highest risk for development of severe liver disease. New guidelines from several European organizations were recently presented. The suggested changes to healthcare practice include screening of high-risk groups in order to identify cases with cirrhosis. These guidelines and suggestions for adjustments to Swedish healthcare are discussed.


Subject(s)
Liver Cirrhosis/diagnosis , Non-alcoholic Fatty Liver Disease/diagnosis , Practice Guidelines as Topic , Risk Assessment/methods , Early Diagnosis , Humans , Liver Cirrhosis/etiology , Non-alcoholic Fatty Liver Disease/complications , Sweden
4.
Obes Surg ; 27(4): 1115-1118, 2017 04.
Article in English | MEDLINE | ID: mdl-28155057

ABSTRACT

AIM: Omentectomy in addition to bariatric surgery has been suggested to improve metabolic outcome but short-term (6-24 months) studies have refuted this notion. We investigated whether there was any long-term impact of omentectomy. METHODS: Forty-nine obese women underwent gastric bypass surgery and were randomly assigned to omentectomy (n = 26) or not (n = 23). They were re-examined after 5 years including dual-energy X-ray absorptiometry for body composition, blood pressure and blood sampling. RESULTS: There were no significant differences between the two groups at baseline (p = 0.07-0.93) or 5 years post-operatively (p = 0.15-0.93) regarding weight, BMI, body composition, HOMA-IR, plasma cholesterol, HDL cholesterol, or triglycerides. CONCLUSION: In agreement with previous shorter studies, removal of the greater omentum in addition to GBP is not associated with metabolic benefits after long-term follow-up.


Subject(s)
Bariatric Surgery/methods , Obesity, Morbid/surgery , Omentum/surgery , Absorptiometry, Photon , Adult , Blood Glucose/metabolism , Blood Pressure/physiology , Body Composition/physiology , Body Mass Index , Cholesterol, HDL/metabolism , Female , Follow-Up Studies , Humans , Middle Aged , Obesity, Morbid/blood , Obesity, Morbid/physiopathology , Triglycerides/blood
5.
Diabetes Care ; 40(1): 77-84, 2017 Jan.
Article in English | MEDLINE | ID: mdl-27852664

ABSTRACT

OBJECTIVE: Although long-term weight regain may occur after bariatric surgery, many patients are protected against relapse or development of type 2 diabetes. The study objective was to investigate whether this involves beneficial changes in adipose function. RESEARCH DESIGN AND METHODS: Forty-nine obese women were investigated before and 2 and 5 years after Roux-en-Y gastric bypass (RYGB). At the 5-year follow-up, 30 subjects were pairwise matched for BMI and age to 30 control women. Clinical parameters and fine-needle biopsies from subcutaneous abdominal adipose tissue were obtained; fat cell size and number, lipolysis, adiponectin, and proinflammatory protein secretion were determined. RESULTS: After 2 years, BMI decreased from 43 to 29 kg/m2, which was accompanied by improvements in insulin sensitivity (HOMA of insulin resistance [HOMA-IR]), increased circulating and adipose secreted adiponectin, and decreased adipose lipolysis and fat cell size but no change in adipocyte number. Between 2 and 5 years after surgery, BMI had increased to 31 kg/m2. This was associated with slightly increased HOMA-IR and unaltered circulating or adipose secreted adiponectin but higher secretion of tumor necrosis factor-α and increased lipolysis and number of fat cells but no change in adipocyte size. All these parameters, except lipolysis, were significantly more favorable compared with those in matched control subjects. Furthermore, the relationship between HOMA-IR and circulating adiponectin was less steep than in control subjects. CONCLUSIONS: RYGB improves long-term insulin sensitivity and adipose phenotypes beyond the control state despite weight regain. Postoperative beneficial alterations in adipose function may be involved in the diabetes-protective effect of bariatric surgery.


Subject(s)
Adipose Tissue/physiopathology , Diabetes Mellitus, Type 2/physiopathology , Gastric Bypass/methods , Obesity/physiopathology , Adipocytes/pathology , Adiponectin/metabolism , Adipose Tissue/surgery , Adiposity/physiology , Adult , Case-Control Studies , Diabetes Mellitus, Type 2/surgery , Female , Follow-Up Studies , Humans , Insulin Resistance , Lipolysis/physiology , Middle Aged , Obesity/surgery , Postoperative Period , Time , Tumor Necrosis Factor-alpha/metabolism
6.
Arterioscler Thromb Vasc Biol ; 36(5): 817-24, 2016 05.
Article in English | MEDLINE | ID: mdl-26966278

ABSTRACT

OBJECTIVE: The dyslipidemia of type 2 diabetes mellitus has multiple etiologies and impairs lipoprotein functionality, thereby increasing risk for cardiovascular disease. High-density lipoproteins (HDLs) have several beneficial effects, notably protecting the heart from myocardial ischemia. We hypothesized that glycation of HDL could compromise this cardioprotective effect. APPROACH AND RESULTS: We used in vitro (cardiomyocytes) and ex vivo (whole heart) models subjected to oxidative stress together with HDL isolated from diabetic patients and nondiabetic HDL glycated in vitro (methylglyoxal). Diabetic and in vitro glycated HDL were less effective (P<0.05) than control HDL in protecting from oxidative stress. Protection was significantly, inversely correlated with the degree of in vitro glycation (P<0.001) and the levels of hemoglobin A1c in diabetic patients (P<0.007). The ability to activate protective, intracellular survival pathways involving Akt, Stat3, and Erk1/2 was significantly reduced (P<0.05) using glycated HDL. Glycation reduced the sphingosine-1-phosphate (S1P) content of HDL, whereas the S1P concentrations of diabetic HDL were inversely correlated with hemoglobin A1c (P<0.005). The S1P contents of in vitro glycated and diabetic HDL were significantly, positively correlated (both <0.01) with cardiomyocyte survival during oxidative stress. Adding S1P to diabetic HDL increased its S1P content and restored its cardioprotective function. CONCLUSIONS: Our data demonstrate that glycation can reduce the S1P content of HDL, leading to increased cardiomyocyte cell death because of less effective activation of intracellular survival pathways. It has important implications for the functionality of HDL in diabetes mellitus because HDL-S1P has several beneficial effects on the vasculature.


Subject(s)
Diabetes Mellitus, Type 2/blood , Diabetic Cardiomyopathies/blood , Dyslipidemias/blood , Lipoproteins, HDL/blood , Lysophospholipids/blood , Myocytes, Cardiac/metabolism , Sphingosine/analogs & derivatives , Animals , Animals, Newborn , Case-Control Studies , Cell Survival , Cells, Cultured , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/diagnosis , Diabetic Cardiomyopathies/diagnosis , Diabetic Cardiomyopathies/etiology , Diabetic Cardiomyopathies/prevention & control , Dyslipidemias/diagnosis , Dyslipidemias/etiology , Genotype , Glycated Hemoglobin/metabolism , Glycosylation , Humans , Isolated Heart Preparation , Male , Mice, Inbred C57BL , Mice, Knockout , Myocardial Reperfusion Injury/metabolism , Myocardial Reperfusion Injury/pathology , Myocardial Reperfusion Injury/prevention & control , Myocytes, Cardiac/pathology , Oxidative Stress , Phenotype , RNA Interference , Rats, Wistar , Scavenger Receptors, Class B/deficiency , Scavenger Receptors, Class B/genetics , Scavenger Receptors, Class B/metabolism , Sphingosine/blood , Time Factors , Transfection
7.
Diabetes Care ; 37(7): 1831-6, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24760260

ABSTRACT

OBJECTIVE: Large subcutaneous fat cells associate with insulin resistance and high risk of developing type 2 diabetes. We investigated if changes in fat cell volume and fat mass correlate with improvements in the metabolic risk profile after bariatric surgery in obese patients. RESEARCH DESIGN AND METHODS: Fat cell volume and number were measured in abdominal subcutaneous adipose tissue in 62 obese women before and 2 years after Roux-en-Y gastric bypass (RYGB). Regional body fat mass by dual-energy X-ray absorptiometry; insulin sensitivity by hyperinsulinemic-euglycemic clamp; and plasma glucose, insulin, and lipid profile were assessed. RESULTS: RYGB decreased body weight by 33%, which was accompanied by decreased adipocyte volume but not number. Fat mass in the measured regions decreased and all metabolic parameters were improved after RYGB (P < 0.0001). Whereas reduced subcutaneous fat cell size correlated strongly with improved insulin sensitivity (P = 0.0057), regional changes in fat mass did not, except for a weak correlation between changes in visceral fat mass and insulin sensitivity and triglycerides. The curve-linear relationship between fat cell size and fat mass was altered after weight loss (P = 0.03). CONCLUSIONS: After bariatric surgery in obese women, a reduction in subcutaneous fat cell volume associates more strongly with improvement of insulin sensitivity than fat mass reduction per se. An altered relationship between adipocyte size and fat mass may be important for improving insulin sensitivity after weight loss. Fat cell size reduction could constitute a target to improve insulin sensitivity.


Subject(s)
Gastric Bypass/methods , Insulin Resistance , Obesity/surgery , Subcutaneous Fat/cytology , Absorptiometry, Photon , Adult , Blood Glucose/metabolism , Cell Size , Female , Glucose Clamp Technique , Humans , Insulin/blood , Intra-Abdominal Fat/metabolism , Lipids/blood , Middle Aged , Prospective Studies , Triglycerides/blood , Triglycerides/physiology , Weight Loss/physiology
8.
Clin Nutr ; 33(6): 991-6, 2014 Dec.
Article in English | MEDLINE | ID: mdl-24485000

ABSTRACT

BACKGROUND & AIMS: Accumulation of visceral adipose tissue is associated with insulin resistance and cardio-vascular disease. The aim of this study was to elucidate whether removal of a large amount of visceral fat by omentectomy in conjunction with Roux en-Y gastric bypass operation (RYGB) results in enhanced improvement of insulin sensitivity compared to gastric bypass surgery alone. METHODS: Eighty-one obese women scheduled for RYGB were included in the study. They were randomized to RYGB or RYGB in conjunction with omentectomy. Insulin sensitivity was measured by hyperinsulinemic euglycemic clamp before operation and sixty-two women were also reexamined 2 years post-operatively. The primary outcome measure was insulin sensitivity and secondary outcome measures included cardio-metabolic risk factors. RESULTS: Two-year weight loss was profound but unaffected by omentectomy. Before intervention, there were no clinical or metabolic differences between the two groups. The difference in primary outcome measure, insulin sensitivity, was not significant between the non-omentectomy (6.7 ± 1.6 mg/kg body weight/minute) and omentectomy groups (6.6 ± 1.5 mg/kg body weight/minute) after 2 years. Nor did any of the cardio-metabolic risk factors that were secondary outcome measures differ significantly. CONCLUSION: Addition of omentectomy to gastric bypass operation does not give an incremental effect on long term insulin sensitivity or cardio-metabolic risk factors. The clinical usefulness of omentectomy in addition to gastric bypass operation is highly questionable. CLINICAL TRIAL REGISTRATION NUMBER: NCT01785134.


Subject(s)
Gastric Bypass/methods , Insulin Resistance , Intra-Abdominal Fat/surgery , Obesity/surgery , Omentum/surgery , Adult , Blood Glucose/metabolism , Body Mass Index , Double-Blind Method , Female , Glucose Clamp Technique/methods , Humans , Middle Aged , Risk Factors , Treatment Outcome , Weight Loss , Young Adult
9.
J Clin Endocrinol Metab ; 98(5): E897-901, 2013 May.
Article in English | MEDLINE | ID: mdl-23543656

ABSTRACT

OBJECTIVE: Accumulation of visceral adipose tissue (VAT) is strongly linked to insulin resistance. Variations in the size of any adipose depot are determined by alterations in adipocyte volume and/or number. The individual contribution of each of the latter factors was determined in the major omentum, a fully resectable VAT depot. SUBJECTS: Total removal of the major omentum (omentectomy) was performed in conjunction with bariatric surgery in 55 obese patients. Tissue weight as well as mean adipocyte size and number in the omentum were determined. In subgroups, total VAT was estimated by computerized tomography (n = 17) or dual-energy x-ray absorptiometry (n = 34). RESULTS: The weight of the major omentum (on average 0.6 kg) correlated significantly with total VAT mass estimated by computerized tomography or dual-energy x-ray absorptiometry (r = 0.48-0.7; P < .01). Omental weight in relation to total body fat correlated with several features of the metabolic syndrome and inversely with serum-leptin (P < .001). Mean adipocyte size and total adipocyte number correlated strongly with omental weight (r = 0.6-0.8; P < .0001), irrespective of body mass index and total body fat mass, and accounted almost in total for interindividual variations in omental size. However, stepwise regression analysis demonstrated that adipocyte number was significantly (P < .0001) more important (62%) than adipocyte size (35%). CONCLUSION: The size of the major omentum is representative for VAT mass and correlates with a pernicious metabolic profile. Variations in omental weight are primarily determined by adipocyte number and to a lesser degree by adipocyte size, suggesting that increased VAT mass in obesity is predominantly dependent on adipocyte proliferation.


Subject(s)
Adiposity , Intra-Abdominal Fat/pathology , Obesity, Morbid/pathology , Omentum/pathology , Adult , Bariatric Surgery , Body Mass Index , Cell Count , Cell Size , Cohort Studies , Female , Humans , Intra-Abdominal Fat/diagnostic imaging , Leptin/blood , Male , Metabolic Syndrome/complications , Middle Aged , Obesity, Morbid/blood , Obesity, Morbid/complications , Obesity, Morbid/surgery , Omentum/diagnostic imaging , Omentum/surgery , Organ Size , Radiography , Subcutaneous Fat, Abdominal/diagnostic imaging , Subcutaneous Fat, Abdominal/pathology , Young Adult
10.
BMC Med Genomics ; 4: 51, 2011 Jun 28.
Article in English | MEDLINE | ID: mdl-21708048

ABSTRACT

BACKGROUND: Recent genome-wide association (GWA) analyses have identified common single nucleotide polymorphisms (SNPs) that are associated with obesity. However, the reported genetic variation in obesity explains only a minor fraction of the total genetic variation expected to be present in the population. Thus many genetic variants controlling obesity remain to be identified. The aim of this study was to use GWA followed by multiple stepwise validations to identify additional genes associated with obesity. METHODS: We performed a GWA analysis in 164 morbidly obese subjects (BMI:body mass index>40 kg/m2) and 163 Swedish subjects (>45 years) who had always been lean. The 700 SNPs displaying the strongest association with obesity in the GWA were analyzed in a second cohort comprising 460 morbidly obese subjects and 247 consistently lean Swedish adults. 23 SNPs remained significantly associated with obesity (nominal P<0.05) and were in a step-wise manner followed up in five additional cohorts from Sweden, France, and Germany together comprising 4214 obese and 5417 lean or population-based control individuals. Three samples, n=4133, were used to investigate the population-based associations with BMI. Gene expression in abdominal subcutaneous adipose tissue in relation to obesity was investigated for14 adults. RESULTS: Potassium channel, calcium activated, large conductance, subfamily M, alpha member (KCNMA1) rs2116830*G and BDNF rs988712*G were associated with obesity in five of six investigated case-control cohorts. In meta-analysis of 4838 obese and 5827 control subjects we obtained genome-wide significant allelic association with obesity for KCNMA1 rs2116830*G with P=2.82×10(-10) and an odds ratio (OR) based on cases vs controls of 1.26 [95% C.I. 1.12-1.41] and for BDNF rs988712*G with P=5.2×10(-17) and an OR of 1.36 [95% C.I. 1.20-1.55]. KCNMA1 rs2116830*G was not associated with BMI in the population-based samples. Adipose tissue (P=0.0001) and fat cell (P=0.04) expression of KCNMA1 was increased in obesity. CONCLUSIONS: We have identified KCNMA1 as a new susceptibility locus for obesity, and confirmed the association of the BDNF locus at the genome-wide significant level.


Subject(s)
Genetic Predisposition to Disease/genetics , Genome, Human , Large-Conductance Calcium-Activated Potassium Channel alpha Subunits/genetics , Obesity, Morbid/genetics , Alleles , Body Mass Index , Case-Control Studies , Cohort Studies , Female , France , Genome-Wide Association Study , Germany , Humans , Male , Middle Aged , Polymorphism, Single Nucleotide , Sweden
11.
J Clin Endocrinol Metab ; 95(10): E209-13, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20660030

ABSTRACT

CONTEXT: Obesity is a major risk factor for nonalcoholic fatty liver disease, but the primary pathophysiological mechanisms remain unclear. OBJECTIVE: The aim was to study the relative role of visceral and sc in vitro lipolysis for liver fat accumulation. PATIENTS: Eighteen morbidly obese women participated in the study. DESIGN: Hepatic triglyceride accumulation and abdominal visceral and sc fat area were assessed by computer tomography. Biopsies were taken from abdominal sc and visceral fat tissue. Basal and maximal lipolysis was measured using various lipolytic drugs acting at different steps in the lipolytic cascade. RESULTS: No difference in total body, visceral, or sc fat mass was found between patients with high, intermediate, or low amounts of liver fat. In patients with high liver fat content, there was an approximately 2-fold increase in visceral adipocyte maximal glycerol release induced by the different lipolytic agents (P = 0.002 to 0.01). Noradrenaline-mediated free fatty acid release from visceral adipocytes was also about twice as high in patients with high liver fat (P = 0.004). In contrast, in sc adipocytes, no relationship between liver fat content and either glycerol or free fatty acid release was found. CONCLUSIONS: We suggest a pathogenic role of visceral, but not sc, adipocyte lipolytic function in nonalcoholic fatty liver disease, independent of total body fat as well as abdominal fat distribution.


Subject(s)
Adipocytes/metabolism , Fatty Liver/etiology , Intra-Abdominal Fat/metabolism , Lipolysis/physiology , Adipocytes/drug effects , Adipocytes/physiology , Adult , Biopsy, Fine-Needle , Body Fat Distribution , Fatty Liver/metabolism , Female , Humans , In Vitro Techniques , Intra-Abdominal Fat/drug effects , Intra-Abdominal Fat/pathology , Intra-Abdominal Fat/physiology , Lipolysis/drug effects , Middle Aged , Norepinephrine/administration & dosage , Norepinephrine/pharmacology , Obesity, Morbid/complications , Obesity, Morbid/metabolism , Obesity, Morbid/pathology , Subcutaneous Fat/drug effects , Subcutaneous Fat/metabolism , Subcutaneous Fat/physiology , Up-Regulation , Young Adult
12.
Biochem Biophys Res Commun ; 383(1): 63-7, 2009 May 22.
Article in English | MEDLINE | ID: mdl-19332024

ABSTRACT

Carboxylesterase 1 (CES1) has recently been suggested to play a role in lipolysis. Our aim was to study the regulation of CES1 expression in human adipose tissue. In the SOS Sib Pair Study, CES1 expression was higher in obese compared with lean sisters (n=78 pairs, P=8.7x10(-18)) and brothers (n=12 pairs, P=0.048). CES1 expression was higher in subcutaneous compared with omental adipose tissue in lean (P=0.027) and obese subjects (P=0.00036), and reduced during diet-induced weight loss (n=24, weeks 8, 16, and 18 compared to baseline, P<0.0001 for all time points). CES1 expression was higher in isolated adipocytes compared with intact adipose tissue (P=0.0018) and higher in large compared with small adipocytes (P=4.1x10(-6)). Basal and stimulated lipolysis was not different in individuals with high, intermediate, and low expression of CES1. Thus, CES1 expression was linked to body fat and adipocyte fat content but not to lipolytic activity.


Subject(s)
Adipocytes/enzymology , Adipose Tissue/enzymology , Carboxylic Ester Hydrolases/biosynthesis , Obesity/enzymology , Adipocytes/cytology , Adipose Tissue/cytology , Adult , Female , Humans , Lipolysis , Male , Middle Aged , Weight Loss , Young Adult
13.
Obes Facts ; 2(5): 294-301, 2009.
Article in English | MEDLINE | ID: mdl-20057196

ABSTRACT

AIM: To examine the relation between adiposity assessment methods (percentage body fat (%BF), BMI, and waist circumference (WC)) and individual metabolic risk factors (f-insulin, HDL cholesterol, triglycerides) and a combined measure of metabolic risk. METHODS: Crosssectional study of 300 males (BMI 20.8 +/- 3.0 kg/m(2)) and females (BMI 21.3 +/- 2.9 kg/m(2)) 17 years of age. F-insulin and components of the metabolic syndrome defined by the International Diabetes Federation (IDF) were used as metabolic risk indicators, with samples stratified into BMI, %BF, and WC groups, respectively. Diagnostic accuracy was expressed as the area under the ROC curve (AUC). RESULTS: In males, diagnostic accuracy for HDL and f-insulin was poor to fair for BMI (AUC 0.70, p = 0.001; 0.60, p = 0.22), WC (0.68, p = 0.003; 0.63, p = 0.11), and %BF (0.65, p = 0.009; 0.66, p = 0.04). The diagnostic accuracy for triglycerides was greater for all three measures (BMI 0.92, WC 0.95, %BF 0.87; all p < 0.001). For females, neither test performed better than chance for f-insulin and HDL, and only %BF performed better than chance for triglycerides (0.65, p = 0.08). All three measures exhibited higher accuracy for presence of > or =2 metabolic risk factors (AUCs 0.76-0.91, p < 0.001) in both sexes. CONCLUSION: %BF was not superior to BMI and WC for detecting metabolic risk in the general adolescent population.


Subject(s)
Body Composition , Metabolic Syndrome/epidemiology , Metabolic Syndrome/metabolism , Obesity/epidemiology , Obesity/metabolism , Adolescent , Blood Glucose/metabolism , Cholesterol, HDL/blood , Cross-Sectional Studies , Female , Humans , Insulin/blood , Male , Prevalence , ROC Curve , Risk Factors , Sex Distribution , Sweden/epidemiology , Triglycerides/blood , Waist Circumference
14.
Nature ; 453(7196): 783-7, 2008 Jun 05.
Article in English | MEDLINE | ID: mdl-18454136

ABSTRACT

Obesity is increasing in an epidemic manner in most countries and constitutes a public health problem by enhancing the risk for cardiovascular disease and metabolic disorders such as type 2 diabetes. Owing to the increase in obesity, life expectancy may start to decrease in developed countries for the first time in recent history. The factors determining fat mass in adult humans are not fully understood, but increased lipid storage in already developed fat cells (adipocytes) is thought to be most important. Here we show that adipocyte number is a major determinant for the fat mass in adults. However, the number of fat cells stays constant in adulthood in lean and obese individuals, even after marked weight loss, indicating that the number of adipocytes is set during childhood and adolescence. To establish the dynamics within the stable population of adipocytes in adults, we have measured adipocyte turnover by analysing the integration of 14C derived from nuclear bomb tests in genomic DNA. Approximately 10% of fat cells are renewed annually at all adult ages and levels of body mass index. Neither adipocyte death nor generation rate is altered in early onset obesity, suggesting a tight regulation of fat cell number in this condition during adulthood. The high turnover of adipocytes establishes a new therapeutic target for pharmacological intervention in obesity.


Subject(s)
Adipocytes/cytology , Adipose Tissue/cytology , Stem Cells/cytology , Adipose Tissue/anatomy & histology , Adult , Body Mass Index , Carbon Radioisotopes , Cell Count , Cell Death , Cell Size , Humans , Obesity/pathology , Weight Loss
15.
J Clin Endocrinol Metab ; 93(6): 2249-54, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18381574

ABSTRACT

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


Subject(s)
Adipose Tissue/metabolism , Adiposity/genetics , Insulin Resistance/genetics , Membrane Proteins/genetics , Obesity/genetics , Oxidoreductases/genetics , Adipocytes/drug effects , Adipocytes/metabolism , Adiponectin/genetics , Adiponectin/metabolism , Adult , Cells, Cultured , Female , Gene Expression Regulation/drug effects , Genetic Predisposition to Disease , Humans , Insulin/pharmacology , Male , Membrane Proteins/metabolism , Middle Aged , Obesity/metabolism , Oxidoreductases/metabolism , RNA, Messenger/metabolism , Tumor Necrosis Factor-alpha/pharmacology
16.
Horm Res ; 69(1): 31-6, 2008.
Article in English | MEDLINE | ID: mdl-18059082

ABSTRACT

BACKGROUND: The uncoupling proteins (UCPs) and beta-adrenoceptors (ADRBs) are important for energy balance and may be involved in the pathogenesis of insulin resistance. Obesity is strongly hunted by insulin resistance and susceptibility genes for the two conditions could be separate or common. Variations within the UCPs and ADRBs genes may give important clues to their involvement in disease. METHODS: A total of four single nucleotide polymorphisms (SNPs) in the genes UCP1, UCP2, UCP3, and ADRB1 were examined for association with obesity and insulin sensitivity (HOMA(IR)) in obese (n = 292) and healthy non-obese (n = 481) females. RESULTS: None of the SNPs was associated with obesity status or body mass index. However, ADRB1 (rs1801253) was nominally associated with serum insulin (nominal p = 0.034) and HOMA(IR) (nominal p = 0.022). UCP3 (rs1800006) was in post-hoc analysis nominally associated with serum insulin and HOMA(IR) (nominal p = 0.013 and 0.048, respectively). UCP1 and UCP2 showed no association with insulin sensitivity. CONCLUSION: Polymorphisms in ADRB1 and UCP3 may contribute to insulin resistance rather than obesity among Swedish women.


Subject(s)
Insulin Resistance/genetics , Ion Channels/genetics , Mitochondrial Proteins/genetics , Obesity/genetics , Polymorphism, Single Nucleotide , Receptors, Adrenergic, beta-1/genetics , Body Mass Index , Female , Genetic Predisposition to Disease , Genotype , Humans , Phenotype , Uncoupling Protein 1 , Uncoupling Protein 2 , Uncoupling Protein 3
17.
J Lipid Res ; 49(3): 607-11, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18048838

ABSTRACT

We investigated the rs9939609 single nucleotide polymorphism of the FTO gene in relation to fat cell function and adipose tissue gene expression in 306 healthy women with a wide range in body mass index (18-53 kg/m(2)). Subcutaneous adipose tissue biopsies were taken for fat cell metabolism studies and in a subgroup (n = 90) for gene expression analyses. In homozygous carriers of the T-allele, the in vitro basal (spontaneous) adipocyte glycerol release was increased by 22% (P = 0.007) and the in vivo plasma glycerol level was increased by approximately 30% (P = 0.037) compared with carriers of the A allele. In contrast, there were no genotype effects on catecholamine-stimulated lipolysis or basal or insulin-induced lipogenesis. We found no difference between genotypes for adipose tissue mRNA levels of FTO, hormone-sensitive lipase, adipose triglyceride lipase, perilipin, or CGI-58. Finally, the adipose tissue level of FTO mRNA was increased in obesity (P = 0.002), was similar in subcutaneous and omental adipose tissue, was higher in fat cells than in fat tissue (P = 0.0007), and was induced at an early stage in the differentiation process (P = 0.004). These data suggest a role of the FTO gene in fat cell lipolysis, which may be important in explaining why the gene is implicated in body weight regulation.


Subject(s)
Adipocytes/metabolism , Lipolysis/genetics , Polymorphism, Single Nucleotide , Proteins/genetics , Alpha-Ketoglutarate-Dependent Dioxygenase FTO , Body Mass Index , Female , Genotype , Glycerol/analysis , Humans , Obesity/genetics , RNA, Messenger/analysis
18.
Cancer Res ; 67(11): 5531-7, 2007 Jun 01.
Article in English | MEDLINE | ID: mdl-17545636

ABSTRACT

Loss of fat mass is a key feature of cancer cachexia and has been attributed to increased adipocyte lipolysis. The mechanism behind this alteration is unknown and was presently investigated. We studied mature s.c. fat cells and differentiated preadipocytes from 26 cancer patients with and without cachexia. Hormone-induced lipolysis and expression of lipolysis-regulating genes were determined together with body composition and in vivo lipolytic activity (fasting plasma glycerol or fatty acids related to body fat). Body fat was reduced by 40% and in vivo lipolytic activity was 2-fold increased in cachexia (P = 0.001). In mature adipocytes, the lipolytic effects of catecholamines and natriuretic peptide were 2- to 3-fold increased in cachexia (P < 0.001). This was completely counteracted by inhibiting the rate-limiting lipolysis enzyme hormone-sensitive lipase (HSL). In cachexia, the expression levels of HSL mRNA and protein were increased by 50% and 100%, respectively (P = 0.005-0.03), which strongly correlated with in vitro lipolytic stimulation (r = 0.7-0.9). The antilipolytic effect of insulin in mature fat cells and the stimulated lipolytic effect in differentiated preadipocytes were unaltered in cachexia. Patients who lost weight due to other factors than cancer cachexia had no change in adipocyte lipolysis. In conclusion, adipocyte lipolysis is increased in cancer cachexia not due to nonepigenic factors or to weight loss per se, but most probably because of enhanced expression and function of adipocyte HSL. The selective inhibition of this enzyme may prevent fat loss in cancer patients.


Subject(s)
Cachexia/etiology , Cachexia/metabolism , Gastrointestinal Neoplasms/complications , Gastrointestinal Neoplasms/metabolism , Adipocytes/metabolism , Atrial Natriuretic Factor/pharmacology , Cachexia/genetics , Female , Gastrointestinal Neoplasms/genetics , Gene Expression , Humans , Insulin/pharmacology , Lipolysis/drug effects , Male , Norepinephrine/pharmacology , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Sterol Esterase/biosynthesis , Sterol Esterase/genetics , Weight Loss
19.
Am J Hum Genet ; 80(6): 1115-24, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17503329

ABSTRACT

The G-protein-coupled receptor GPR74 is a novel candidate gene for body weight regulation. In humans, it is predominantly expressed in brain, heart, and adipose tissue. We report a haplotype in the GPR74 gene, ATAG, with allele frequency ~4% in Scandinavian cohorts, which was associated with protection against obesity in two samples selected for obese and lean phenotypes (odds ratio for obesity 0.48 and 0.62; nominal P=.0014 and .014; n=1,013 and 1,423, respectively). In a population-based sample, it was associated with lower waist (P=.02) among 3,937 men and with obesity protection (odds ratio 0.36; P=.036) among those selected for obese or lean phenotypes. The ATAG haplotype was associated with increased adipocyte lipid mobilization (lipolysis) in vivo and in vitro. In human fat cells, GPR74 receptor stimulation and inhibition caused a significant and marked decrease and increase, respectively, of lipolysis, which could be linked to catecholamine stimulation of adipocytes through beta -adrenergic receptors. These findings suggest that a common haplotype in the GPR74 gene protects against obesity, which, at least in part, is caused by a relief of inhibition of lipid mobilization from adipose tissue. The latter involves a cross-talk between GPR74 and beta -adrenoceptor signaling to lipolysis in fat cells.


Subject(s)
Haplotypes , Lipolysis/genetics , Receptors, Neuropeptide/genetics , Thinness/genetics , Adipocytes/cytology , Adipocytes/drug effects , Adipocytes/physiology , Adrenergic alpha-Agonists/pharmacology , Adrenergic alpha-Antagonists/pharmacology , Adult , Alleles , Bucladesine/pharmacology , Cell Differentiation , Cohort Studies , Dose-Response Relationship, Drug , Drug Interactions , Female , Gene Frequency , Humans , Linkage Disequilibrium , Male , Middle Aged , Norepinephrine/pharmacology , Odds Ratio , Polymorphism, Single Nucleotide , RNA, Small Interfering/pharmacology , Yohimbine/pharmacology
20.
Am J Physiol Endocrinol Metab ; 292(6): E1847-55, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17327373

ABSTRACT

Hormone-sensitive lipase (HSL) and adipose triglyceride lipase (ATGL) regulate adipocyte lipolysis in rodents. The purpose of this study was to compare the roles of these lipases for lipolysis in human adipocytes. Subcutaneous adipose tissue was investigated. HSL and ATGL protein expression were related to lipolysis in isolated mature fat cells. ATGL or HSL were knocked down by RNA interference (RNAi) or selectively inhibited, and effects on lipolysis were studied in differentiated preadipocytes or adipocytes derived from human mesenchymal stem cells (hMSC). Subjects were all women. There were 12 lean controls, 8 lean with polycystic ovary syndrome (PCOS), and 27 otherwise healthy obese subjects. We found that norepinephrine-induced lipolysis was positively correlated with HSL protein levels (P < 0.0001) but not with ATGL protein. Women with PCOS or obesity had significantly decreased norepinephrine-induced lipolysis and HSL protein expression but no change in ATGL protein expression. HSL knock down by RNAi reduced basal and catecholamine-induced lipolysis. Knock down of ATGL decreased basal lipolysis but did not change catecholamine-stimulated lipolysis. Treatment of hMSC with a selective HSL inhibitor during and/or after differentiation in adipocytes reduced basal lipolysis by 50%, but stimulated lipolysis was inhibited completely. In contrast to findings in rodents, ATGL is of less importance than HSL in regulating catecholamine-induced lipolysis and cannot replace HSL when this enzyme is continuously inhibited. However, both lipases regulate basal lipolysis in human adipocytes. ATGL expression, unlike HSL, is not influenced by obesity or PCOS.


Subject(s)
Adipocytes/metabolism , Lipolysis/physiology , Obesity/metabolism , Phospholipases A/physiology , Polycystic Ovary Syndrome/metabolism , Sterol Esterase/physiology , Adipocytes/drug effects , Adult , Cohort Studies , Down-Regulation , Female , Glycerol/metabolism , Humans , Lipase , Mesenchymal Stem Cells/metabolism , Norepinephrine/pharmacology , Phospholipases A/antagonists & inhibitors , Phospholipases A/genetics , RNA Interference , Sterol Esterase/antagonists & inhibitors , Sterol Esterase/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...