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










Publication year range
2.
Int J Obes (Lond) ; 41(3): 372-380, 2017 03.
Article in English | MEDLINE | ID: mdl-28008171

ABSTRACT

BACKGROUND/OBJECTIVE: Futile substrate cycling based on lipolytic release of fatty acids (FA) from intracellular triacylglycerols (TAG) and their re-esterification (TAG/FA cycling), as well as de novo FA synthesis (de novo lipogenesis (DNL)), represent the core energy-consuming biochemical activities of white adipose tissue (WAT). We aimed to characterize their roles in cold-induced thermogenesis and energy homeostasis. METHODS: Male obesity-resistant A/J and obesity-prone C57BL/6J mice maintained at 30 °C were exposed to 6 °C for 2 or 7 days. In epididymal WAT (eWAT), TAG synthesis and DNL were determined using in vivo 2H incorporation from 2H2O into tissue TAG and nuclear magnetic resonance spectroscopy. Quantitative real-time-PCR and/or immunohistochemistry and western blotting were used to determine the expression of selected genes and proteins in WAT and liver. RESULTS: The mass of WAT depots declined during cold exposure (CE). Plasma levels of TAG and non-esterified FA were decreased by day 2 but tended to normalize by day 7 of CE. TAG synthesis (reflecting TAG/FA cycle activity) gradually increased during CE. DNL decreased by day 2 of CE but increased several fold over the control values by day 7. Expression of genes involved in lipolysis, glyceroneogenesis, FA re-esterification, FA oxidation and mitochondrial biogenesis in eWAT was induced during CE. All these changes were more pronounced in obesity-resistant A/J than in B6 mice and occurred in the absence of uncoupling protein 1 in eWAT. Expression of markers of glyceroneogenesis in eWAT correlated negatively with hepatic FA synthesis by day 7 in both strains. Leptin and fibroblast growth factor 21 plasma levels were differentially affected by CE in the two mouse strains. CONCLUSIONS: Our results indicate integrated involvement of (i) TAG/FA cycling and DNL in WAT, and (ii) hepatic very-low-density lipoprotein-TAG synthesis in the control of blood lipid levels and provision of FA fuels for thermogenesis in cold. They suggest that lipogenesis in WAT contributes to a lean phenotype.


Subject(s)
Adipose Tissue, White/metabolism , Cold Temperature , Lipogenesis/physiology , Thermogenesis/physiology , Thinness/metabolism , Animals , Disease Models, Animal , Lipid Metabolism , Lipogenesis/genetics , Lipoproteins, VLDL/metabolism , Male , Mice , Mice, Inbred C57BL , Obesity/genetics , Obesity/metabolism , Phenotype , Thermogenesis/genetics , Thinness/genetics
3.
Physiol Res ; 63(Suppl 1): S93-118, 2014.
Article in English | MEDLINE | ID: mdl-24564669

ABSTRACT

Type 2 diabetes (T2D) as well as cardiovascular disease (CVD) represent major complications of obesity and associated metabolic disorders (metabolic syndrome). This review focuses on the effects of long-chain n-3 polyunsaturated fatty acids (omega-3) on insulin sensitivity and glucose homeostasis, which are improved by omega-3 in many animal models of metabolic syndrome, but remain frequently unaffected in humans. Here we focus on: (i) mechanistic aspects of omega-3 action, reflecting also our experiments in dietary obese mice; and (ii) recent studies analysing omega-3's effects in various categories of human subjects. Most animal experiments document beneficial effects of omega-3 on insulin sensitivity and glucose metabolism even under conditions of established obesity and insulin resistance. Besides positive results obtained in both cross-sectional and prospective cohort studies on healthy human populations, also some intervention studies in prediabetic subjects document amelioration of impaired glucose homeostasis by omega-3. However, the use of omega-3 to reduce a risk of new-onset diabetes in prediabetic subjects still remains to be further characterized. The results of a majority of clinical trials performed in T2D patients suggest that omega-3 have none or marginal effects on metabolic control, while effectively reducing hypertriglyceridemia in these patients. Despite most of the recent randomized clinical trials do not support the role of omega-3 in secondary prevention of CVD, this issue remains still controversial. Combined interventions using omega-3 and antidiabetic or hypolipidemic drugs should be further explored and considered for treatment of patients with T2D and other diseases.


Subject(s)
Blood Glucose/metabolism , Diabetes Mellitus, Type 2/drug therapy , Diabetes Mellitus, Type 2/metabolism , Fatty Acids, Omega-3/administration & dosage , Homeostasis/drug effects , Insulin Resistance , Insulin/blood , Animals , Evidence-Based Medicine , Fatty Acids, Omega-3/pharmacokinetics , Humans , Mice , Treatment Outcome
4.
Int J Obes (Lond) ; 38(2): 216-23, 2014 Feb.
Article in English | MEDLINE | ID: mdl-23756677

ABSTRACT

OBJECTIVE: Resolution of low-grade inflammation of white adipose tissue (WAT) is one of the keys for amelioration of obesity-associated metabolic dysfunctions. We focused on the identification of adipokines, which could be involved at the early stages of resolution of WAT inflammation. METHODS AND PROCEDURE: Male C57BL/6J mice with obesity induced in response to a 22-week feeding corn oil-based high-fat (cHF) diet were divided into four groups and were fed with, for 2 weeks, control cHF diet or cHF-based diets supplemented with: (i) concentrate of n-3 long-chain polyunsaturated fatty acids, mainly eicosapentaenoic and docosahexaenoic acids (cHF+F); (ii) thiazolidinedione drug rosiglitazone (cHF+TZD); and (iii) both compounds (cHF+F+TZD). RESULTS: The short-term combined intervention exerted additive effect in the amelioration of WAT inflammation in obese mice, namely in the epididymal fat, even in the absence of any changes in either adipocyte volume or fat mass. The combined intervention elicited hypolipidaemic effect and induced adiponectin, whereas the responses to single interventions (cHF+F, cHF+TZD) were less pronounced. In addition, analysis in WAT lysates using protein arrays revealed that the levels of a small set of adipose tissue-related proteins, namely macrophage inflammatory protein 1γ, endoglin, vascular cell adhesion molecule 1 and interleukin 1 receptor antagonist, changed in response to the anti-inflammatory interventions and were strongly reduced in the cHF+F+TZD mice. These results were verified using both the analysis of gene expression and enzyme-linked immunosorbent analysis in WAT lysates. In contrast with adiponectin, which showed changing plasma levels in response to dietary interventions, the levels of the above proteins were affected only in WAT. CONCLUSIONS: We identified several adipose tissue-related proteins, which are locally involved in resolution of low-grade inflammation and remodelling of WAT.


Subject(s)
Adipose Tissue, White/metabolism , Adipose Tissue, White/pathology , Docosahexaenoic Acids/pharmacology , Fatty Acids, Omega-3/pharmacology , Inflammation/pathology , Obesity/pathology , Thiazolidinediones/pharmacology , Adipocytes/metabolism , Adipokines/metabolism , Animals , Diet, High-Fat , Dietary Fats , Energy Metabolism , Enzyme-Linked Immunosorbent Assay , Immunohistochemistry , Male , Mice , Mice, Inbred C57BL , Mice, Obese , Obesity/immunology , Real-Time Polymerase Chain Reaction , Rosiglitazone
5.
Int J Obes (Lond) ; 36(2): 262-72, 2012 Feb.
Article in English | MEDLINE | ID: mdl-21540832

ABSTRACT

OBJECTIVE: Adverse effects of obesity on glucose homeostasis are linked to low-grade adipose tissue inflammation and accumulation of lipids in non-adipose tissues. The goal of this study was to evaluate the role of adipose tissue plasticity in a less severe deterioration of glucose homeostasis in females compared with males during the course of high-fat (HF) feeding in mice. DESIGN: Mice of the C57BL/6N strain were fed either a chow or obesogenic HF diet for up to 35 weeks after weaning. Metabolic markers and hormones in plasma, glucose homeostasis, adipocyte size and inflammatory status of gonadal (gWAT) and subcutaneous (scWAT) adipose depots and liver steatosis were evaluated at 15 and 35 weeks of HF feeding. RESULTS: HF-fed males were heavier than females until week ∼20, after which the body weights stabilized at a similar level (55-58 g) in both sexes. Greater weight gain and fat accumulation in females were associated with larger adipocytes in gWAT and scWAT at week 35. Although adipose tissue macrophage infiltration was in general less frequent in scWAT, it was reduced in both fat depots of female as compared with male mice; however, the expression of inflammatory markers in gWAT was similar in both sexes at week 35. In females, later onset of the impairment of glucose homeostasis and better insulin sensitivity were associated with higher plasma levels of adiponectin (weeks 0, 15 and 35) and reduced hepatosteatosis (weeks 15 and 35). CONCLUSIONS: Compared with males, female mice demonstrate increased capacity for adipocyte enlargement in response to a long-term HF feeding, which is associated with reduced adipose tissue macrophage infiltration and lower fat deposition in the liver, and with better insulin sensitivity. Our data suggest that adipose tissue expandability linked to adiponectin secretion might have a role in the sex differences observed in obesity-associated metabolic disorders.


Subject(s)
Adipose Tissue/pathology , Blood Glucose/metabolism , Elasticity , Lipids/blood , Obesity/metabolism , Obesity/pathology , Animals , Biomarkers/metabolism , Diet, High-Fat , Female , Glucose Tolerance Test , Immunohistochemistry , Male , Mice , Mice, Inbred C57BL , Obesity/blood , Obesity/etiology , Sex Factors , Weight Gain
6.
Diabetologia ; 54(10): 2626-38, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21779874

ABSTRACT

AIMS/HYPOTHESIS: Calorie restriction is an essential component in the treatment of obesity and associated diseases. Long-chain n-3 polyunsaturated fatty acids (LC n-3 PUFA) act as natural hypolipidaemics, reduce the risk of cardiovascular disease and could prevent the development of obesity and insulin resistance. We aimed to characterise the effectiveness and underlying mechanisms of the combination treatment with LC n-3 PUFA and 10% calorie restriction in the prevention of obesity and associated disorders in mice. METHODS: Male mice (C57BL/6J) were habituated to a corn-oil-based high-fat diet (cHF) for 2 weeks and then randomly assigned to various dietary treatments for 5 weeks or 15 weeks: (1) cHF, ad libitum; (2) cHF with LC n-3 PUFA concentrate replacing 15% (wt/wt) of dietary lipids (cHF + F), ad libitum; (3) cHF with calorie restriction (CR; cHF + CR); and (4) cHF + F + CR. Mice fed a chow diet were also studied. RESULTS: We show that white adipose tissue plays an active role in the amelioration of obesity and the improvement of glucose homeostasis by combining LC n-3 PUFA intake and calorie restriction in cHF-fed mice. Specifically in the epididymal fat in the abdomen, but not in other fat depots, synergistic induction of mitochondrial oxidative capacity and lipid catabolism was observed, resulting in increased oxidation of metabolic fuels in the absence of mitochondrial uncoupling, while low-grade inflammation was suppressed, reflecting changes in tissue levels of anti-inflammatory lipid mediators, namely 15-deoxy-Δ(12,15)-prostaglandin J(2) and protectin D1. CONCLUSIONS/INTERPRETATION: White adipose tissue metabolism linked to its inflammatory status in obesity could be modulated by combination treatment using calorie restriction and dietary LC n-3 PUFA to improve therapeutic strategies for metabolic syndrome.


Subject(s)
Adipose Tissue, White/metabolism , Caloric Restriction , Fatty Acids, Omega-3/pharmacology , Lipid Metabolism/drug effects , Adipose Tissue, White/drug effects , Animals , Diet, High-Fat , Dietary Fats/pharmacology , Docosahexaenoic Acids/metabolism , Energy Metabolism/drug effects , Immunohistochemistry , Male , Mice , Mice, Obese , Prostaglandin D2/analogs & derivatives , Prostaglandin D2/metabolism , Real-Time Polymerase Chain Reaction
7.
Diabetologia ; 52(5): 941-51, 2009 May.
Article in English | MEDLINE | ID: mdl-19277604

ABSTRACT

AIMS/HYPOTHESIS: Fatty acids of marine origin, i.e. docosahexaenoic acid (DHA) and eicosapentaenoic acid (EPA) act as hypolipidaemics, but they do not improve glycaemic control in obese and diabetic patients. Thiazolidinediones like rosiglitazone are specific activators of peroxisome proliferator-activated receptor gamma, which improve whole-body insulin sensitivity. We hypothesised that a combined treatment with a DHA and EPA concentrate (DHA/EPA) and rosiglitazone would correct, by complementary additive mechanisms, impairments of lipid and glucose homeostasis in obesity. METHODS: Male C57BL/6 mice were fed a corn oil-based high-fat diet. The effects of DHA/EPA (replacing 15% dietary lipids), rosiglitazone (10 mg/kg diet) or a combination of both on body weight, adiposity, metabolic markers and adiponectin in plasma, as well as on liver and muscle gene expression and metabolism were analysed. Euglycaemic-hyperinsulinaemic clamps were used to characterise the changes in insulin sensitivity. The effects of the treatments were also analysed in dietary obese mice with impaired glucose tolerance (IGT). RESULTS: DHA/EPA and rosiglitazone exerted additive effects in prevention of obesity, adipocyte hypertrophy, low-grade adipose tissue inflammation, dyslipidaemia and insulin resistance, while inducing adiponectin, suppressing hepatic lipogenesis and decreasing muscle ceramide concentration. The improvement in glucose tolerance reflected a synergistic stimulatory effect of the combined treatment on muscle glycogen synthesis and its sensitivity to insulin. The combination treatment also reversed dietary obesity, dyslipidaemia and IGT. CONCLUSIONS/INTERPRETATION: DHA/EPA and rosiglitazone can be used as complementary therapies to counteract dyslipidaemia and insulin resistance. The combination treatment may reduce dose requirements and hence the incidence of adverse side effects of thiazolidinedione therapy.


Subject(s)
Dietary Fats/pharmacology , Fatty Acids, Omega-3/pharmacology , Glycogen/biosynthesis , Insulin/physiology , Muscle, Skeletal/metabolism , Thiazolidinediones/pharmacology , Animals , Corn Oil/pharmacology , Docosahexaenoic Acids/pharmacology , Eicosapentaenoic Acid/pharmacology , Glucose Intolerance/metabolism , Hypoglycemic Agents/pharmacology , Male , Mice , Mice, Inbred C57BL , Muscle, Skeletal/drug effects , Rosiglitazone
8.
J Physiol Pharmacol ; 60(4): 135-40, 2009 Dec.
Article in English | MEDLINE | ID: mdl-20065507

ABSTRACT

UNLABELLED: In humans, antidiabetics thiazolidinediones (TZDs) upregulate stearoyl-CoA desaturase 1 (SCD1) gene in adipose tissue and increase plasma levels of SCD1 product palmitoleate, known to enhance muscle insulin sensitivity. Involvement of other tissues in the beneficial effects of TZDs on plasma lipid profile is unclear. In our previous study in mice, in which lipogenesis was suppressed by corn oil-based high-fat (cHF) diet, TZD rosiglitazone induced hepatic Scd1 expression, while liver triacylglycerol content increased, VLDL-triacylglycerol production decreased and plasma lipid profile and whole-body glycemic control improved. Aim of this study was to characterise contribution of liver to changes of plasma lipid profile in response to a 8-week-treatment by rosiglitazone in the cHF diet-fed mice. Rosiglitazone (10 mg/kg diet) upregulated expression of Scd1 in various tissues, with a stronger effect in liver as compared with adipose tissue or skeletal muscle. Rosiglitazone increased content of monounsaturated fatty acids in liver, adipose tissue and plasma, with palmitoleate being the most up-regulated fatty acid. In the liver, enhancement of SCD1 activity and specific enrichment of cholesteryl esters and phosphatidyl cholines with palmitoleate and vaccenate was found, while strong correlations between changes of various liver lipid fractions and total plasma lipids were observed (r=0.74-0.88). Insulin-stimulated glycogen synthesis was increased by rosiglitazone, with a stronger effect in muscle than in liver. CONCLUSIONS: changes in plasma lipid profile favouring monounsaturated fatty acids, mainly palmitoleate, due to the upregulation of Scd1 and enhancement of SCD1 activity in the liver, could be involved in the insulin-sensitizing effects of TZDs.


Subject(s)
Dietary Fats/administration & dosage , Fatty Acids, Monounsaturated/blood , Hypoglycemic Agents/pharmacology , Liver/drug effects , Thiazolidinediones/pharmacology , Adipose Tissue, White/chemistry , Adipose Tissue, White/metabolism , Animals , Corn Oil/administration & dosage , Fatty Acids/analysis , Fatty Acids/blood , Fatty Acids, Monounsaturated/analysis , Glucose Clamp Technique , Glycogen/metabolism , Insulin Resistance , Lipids/blood , Lipids/chemistry , Liver/chemistry , Liver/physiology , Mice , Muscle, Skeletal/metabolism , Oleic Acids/analysis , Oleic Acids/blood , Organ Specificity , Random Allocation , Rosiglitazone , Stearoyl-CoA Desaturase/genetics , Stearoyl-CoA Desaturase/metabolism , Up-Regulation
9.
Diabetologia ; 49(2): 394-7, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16397791

ABSTRACT

AIMS/HYPOTHESIS: Diets rich in n-3 polyunsaturated fatty acids, namely eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA), protect against insulin resistance and obesity in rodents and increase insulin sensitivity in healthy humans. We tested whether the anti-diabetic effects of EPA and DHA involve enhanced production of the endogenous insulin sensitiser, adiponectin. METHODS: We studied the effects, in an obesity-promoting high-fat diet, of partial replacement of vegetable oils by EPA/DHA concentrate (6% EPA, 51% DHA) over a 5-week period in adult male C57BL/6J mice that either had free access to food or had their food intake restricted by 30%. At the end of the treatment, systemic markers of lipid and glucose metabolism and full-length adiponectin and leptin were measured. Adiponectin (Adipoq) and leptin (Lep) gene expression in dorsolumbar and epididymal white adipose tissue (WAT) and isolated adipocytes was quantified and adipokine production from WAT explants evaluated. RESULTS: In mice with free access to food, plasma triacylglycerols, NEFA, and insulin levels were lower in the presence of EPA/DHA, while glucose and leptin levels were not significantly altered. Food restriction decreased plasma triacylglycerols, glucose, insulin and leptin, but not adiponectin. EPA/DHA increased plasma adiponectin levels, independent of food intake, reflecting the stimulation of Adipoq expression in adipocytes and the release of adiponectin from WAT, particularly from epididymal fat. Expression of Lep and the release of leptin from WAT, while being extremely sensitive to caloric restriction, was unaltered by EPA/DHA. CONCLUSIONS/INTERPRETATION: Intake of diets rich in EPA and DHA leads to elevated systemic concentrations of adiponectin, largely independent of food intake or adiposity and explain, to some extent, their anti-diabetic effects.


Subject(s)
Adiponectin/biosynthesis , Adiponectin/genetics , Dietary Fats/pharmacology , Docosahexaenoic Acids/pharmacology , Eicosapentaenoic Acid/pharmacology , AMP-Activated Protein Kinase Kinases , Adipocytes/chemistry , Adipocytes/metabolism , Adiponectin/blood , Adipose Tissue/chemistry , Adipose Tissue/metabolism , Animals , Body Composition , Caloric Restriction , Dietary Fats/administration & dosage , Docosahexaenoic Acids/administration & dosage , Eating , Eicosapentaenoic Acid/administration & dosage , Enzyme Activation , Gene Expression Regulation , Glucose/metabolism , Insulin/blood , Insulin/physiology , Insulin Resistance , Leptin/analysis , Leptin/blood , Leptin/genetics , Leptin/physiology , Male , Mice , Mice, Inbred C57BL , Obesity/physiopathology , Obesity/prevention & control , Protein Kinases/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Triglycerides/blood
10.
Diabetologia ; 48(11): 2365-75, 2005 Nov.
Article in English | MEDLINE | ID: mdl-16205884

ABSTRACT

AIMS/HYPOTHESIS: Intake of n-3 polyunsaturated fatty acids reduces adipose tissue mass, preferentially in the abdomen. The more pronounced effect of marine-derived eicosapentaenoic (EPA) and docosahexaenoic (DHA) acids on adiposity, compared with their precursor alpha-linolenic acid, may be mediated by changes in gene expression and metabolism in white fat. METHODS: The effects of EPA/DHA concentrate (6% EPA, 51% DHA) admixed to form two types of high-fat diet were studied in C57BL/6J mice. Oligonucleotide microarrays, cDNA PCR subtraction and quantitative real-time RT-PCR were used to characterise gene expression. Mitochondrial proteins were quantified using immunoblots. Fatty acid oxidation and synthesis were measured in adipose tissue fragments. RESULTS: Expression screens revealed upregulation of genes for mitochondrial proteins, predominantly in epididymal fat when EPA/DHA concentrate was admixed to a semisynthetic high-fat diet rich in alpha-linolenic acid. This was associated with a three-fold stimulation of the expression of genes encoding regulatory factors for mitochondrial biogenesis and oxidative metabolism (peroxisome proliferator-activated receptor gamma coactivator 1 alpha [Ppargc1a, also known as Pgc1alpha] and nuclear respiratory factor-1 [Nrf1] respectively). Expression of genes for carnitine palmitoyltransferase 1A and fatty acid oxidation was increased in epididymal but not subcutaneous fat. In the former depot, lipogenesis was depressed. Similar changes in adipose gene expression were detected after replacement of as little as 15% of lipids in the composite high-fat diet with EPA/DHA concentrate, while the development of obesity was reduced. The expression of Ppargc1a and Nrf1 was also stimulated by n-3 polyunsaturated fatty acids in 3T3-L1 cells. CONCLUSIONS/INTERPRETATION: The anti-adipogenic effect of EPA/DHA may involve a metabolic switch in adipocytes that includes enhancement of beta-oxidation and upregulation of mitochondrial biogenesis.


Subject(s)
Adipose Tissue/metabolism , Fatty Acids, Unsaturated/pharmacology , Mitochondria/drug effects , Adipocytes/drug effects , Adipocytes/metabolism , Adipose Tissue/drug effects , Animals , Carnitine O-Palmitoyltransferase/drug effects , Carnitine O-Palmitoyltransferase/genetics , Cells, Cultured , Docosahexaenoic Acids/pharmacology , Eicosapentaenoic Acid/pharmacology , Epididymis/drug effects , Epididymis/metabolism , Fatty Acids, Unsaturated/isolation & purification , Fatty Acids, Unsaturated/metabolism , Fish Oils/chemistry , Gene Expression Regulation/drug effects , Lipogenesis/drug effects , Male , Mice , Mice, Inbred C57BL , Mitochondria/metabolism , Mitochondrial Proteins/drug effects , Mitochondrial Proteins/metabolism , NF-E2-Related Factor 1/drug effects , NF-E2-Related Factor 1/genetics , Obesity/prevention & control , Oxidation-Reduction , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha , Subcutaneous Fat/drug effects , Subcutaneous Fat/metabolism , Trans-Activators/drug effects , Trans-Activators/genetics , Transcription Factors , alpha-Linolenic Acid/pharmacology
11.
Int J Obes Relat Metab Disord ; 28 Suppl 4: S38-44, 2004 Dec.
Article in English | MEDLINE | ID: mdl-15592485

ABSTRACT

As indicated by in vitro studies, both lipogenesis and lipolysis in adipocytes depend on the cellular ATP levels. Ectopic expression of mitochondrial uncoupling protein 1 (UCP1) in the white adipose tissue of the aP2-Ucp1 transgenic mice reduced obesity induced by genetic or dietary manipulations. Furthermore, respiratory uncoupling lowered the cellular energy charge in adipocytes, while the synthesis of fatty acids (FA) was inhibited and their oxidation increased. Importantly, the complex metabolic changes triggered by ectopic UCP1 were associated with the activation of AMP-activated protein kinase (AMPK), a metabolic master switch, in adipocytes. Effects of several typical treatments that reduce adiposity, such as administration of leptin, beta-adrenoceptor agonists, bezafibrate, dietary n-3 polyunsaturated FA or fasting, can be compared with a phenotype of the aP2-Ucp1 mice. These situations generally lead to the upregulation of mitochondrial UCPs and suppression of the cellular energy charge and FA synthesis in adipocytes. On the other hand, FA oxidation is increased. Moreover, it has been shown that AMPK in adipocytes can be activated by adipocyte-derived hormones leptin and adiponectin, and also by insulin-sensitizes thiazolidinediones. Thus, it is evident that metabolism of adipose tissue itself is important for the control of body fat content and that the cellular energy charge and AMPK are involved in the control of lipid metabolism in adipocytes. The reciprocal link between synthesis and oxidation of FA in adipocytes represents a prospective target for the new treatment strategies aimed at reducing obesity.


Subject(s)
Adipocytes/metabolism , Adipose Tissue/metabolism , Multienzyme Complexes/metabolism , Protein Serine-Threonine Kinases/metabolism , AMP-Activated Protein Kinases , Animals , Carrier Proteins/metabolism , Choristoma/metabolism , Energy Metabolism/physiology , Humans , Ion Channels , Membrane Proteins/metabolism , Mice , Mice, Transgenic , Mitochondria/metabolism , Mitochondrial Proteins , Models, Biological , Obesity/metabolism , Uncoupling Protein 1
12.
Physiol Res ; 53 Suppl 1: S225-32, 2004.
Article in English | MEDLINE | ID: mdl-15119952

ABSTRACT

Body fat content is controlled, at least in part, by energy charge of adipocytes. In vitro studies indicated that lipogenesis as well as lipolysis depend on cellular ATP levels. Respiratory uncoupling may, through the depression of ATP synthesis, control lipid metabolism of adipose cells. Expression of some uncoupling proteins (UCP2 and UCP5) as well as other protonophoric transporters can be detected in the adipose tissue. Expression of other UCPs (UCP1 and UCP3) can be induced by pharmacological treatments that reduce adiposity. A negative correlation between the accumulation of fat and the expression of UCP2 in adipocytes was also found. Ectopic expression of UCP1 in the white fat of aP2-Ucp1 transgenic mice mitigated obesity induced by genetic or dietary factors. In these mice, changes in lipid metabolism of adipocytes were associated with the depression of intracellular energy charge. Recent data show that AMP-activated protein kinase may be involved in the complex changes elicited by respiratory uncoupling in adipocytes. Changes in energy metabolism of adipose tissue may mediate effects of treatments directed against adiposity, dyslipidemia, and insulin resistance.


Subject(s)
Adipocytes/metabolism , Adipose Tissue/metabolism , Energy Metabolism , Lipid Metabolism , Obesity/physiopathology , AMP-Activated Protein Kinases , Animals , Carrier Proteins/metabolism , Ion Channels , Membrane Proteins/metabolism , Metabolic Syndrome/metabolism , Metabolic Syndrome/physiopathology , Metabolic Syndrome/prevention & control , Mice , Mice, Transgenic , Mitochondrial Proteins , Multienzyme Complexes/metabolism , Obesity/metabolism , Obesity/prevention & control , Protein Serine-Threonine Kinases/metabolism , Uncoupling Protein 1
13.
Cell Mol Life Sci ; 59(11): 1934-44, 2002 Nov.
Article in English | MEDLINE | ID: mdl-12530524

ABSTRACT

The differentiation of brown adipocytes during late fetal development or in cell culture is associated with enhanced mitochondrial biogenesis and increased gene expression for components of the respiratory chain/oxidative phosphorylation system. We have shown that this is due to a rise in mitochondrial DNA abundance and the corresponding increase in mitochondrial genome transcripts and gene products, as well as to the coordinate induction of nuclear-encoded genes for mitochondrial proteins. We studied how the expression of key components of the transcriptional regulation of mitochondrial biogenesis is regulated during this process. Changes in the expression of nuclear respiratory factor-2/GA-binding protein a and peroxisome proliferator-activated-receptor gamma coactivator-1 (increase) were opposite to those of nuclear respiratory factor-1 and Sp1 (decrease) during the developmental and differentiation-dependent induction of mitochondrial biogenesis in brown fat. These results indicate that the relative roles of transcription factors and coactivators in mediating mitochondrial biogenesis 'in vivo' are highly specific according to the cell type and stimulus that mediate the mitochondriogenic process.


Subject(s)
Adipose Tissue, Brown/embryology , Adipose Tissue, Brown/physiology , Mitochondria/physiology , Transcription Factors/genetics , Transcription Factors/physiology , Adipocytes/cytology , Adipocytes/physiology , Adipose Tissue, Brown/cytology , Animals , Cell Differentiation/physiology , Embryo, Mammalian/physiology , Embryo, Mammalian/ultrastructure , Embryonic and Fetal Development , Female , Male , Mice
14.
Biochem Soc Trans ; 29(Pt 6): 791-7, 2001 11.
Article in English | MEDLINE | ID: mdl-11709076

ABSTRACT

Metabolism of white adipose tissue is involved in the control of body fat content. In vitro experiments indicated a dependence of lipogenesis on mitochondrial ATP production, as well as a reciprocal link between hormonal effects on metabolism and energetics of adipocytes. Therefore, mitochondrial uncoupling in adipocytes that results in stimulation of energy dissipation and depression of ATP synthesis may contribute to control of lipid metabolism and adiposity. This is supported by the expression of protonophoric proteins in adipocytes, e.g. uncoupling proteins (UCPs) 2 and 5, and some anion transporters, and induction of UCP1 and UCP3 in white fat by pharmacological treatments that reduce adiposity. Negative correlation between expression of UCPs in adipocytes and accumulation of white fat was also found. Expression of UCP1 from the adipose-specific promoter in aP2-Ucp1 transgenic mice mitigated obesity induced by genetic or dietary factors. The obesity resistance, accompanied by mitochondrial uncoupling in adipocytes and increased energy expenditure, resulted from ectopic expression of UCP1 in white but not in brown fat. Probably due to depression of ATP/ADP ratio in white fat of transgenic mice, both fatty acid synthesis and lipolytic action of noradrenaline in adipocytes were relatively low. These results support the role of protonophoric proteins in adipocytes in the control of adiposity. The main function of these proteins in white fat may be modulation of lipogenesis and intracellular hormone signalling. Augmentation of energy expenditure may be of relatively small importance, in accordance with the low oxidative capacity of white adipocytes.


Subject(s)
Adipocytes/metabolism , Carrier Proteins/metabolism , Lipid Metabolism , Membrane Proteins/metabolism , Mitochondria/metabolism , Adipose Tissue/metabolism , Animals , Carrier Proteins/genetics , Humans , Ion Channels , Membrane Proteins/genetics , Mice , Mitochondrial Proteins , Models, Biological , Uncoupling Protein 1
15.
FASEB J ; 14(12): 1793-800, 2000 Sep.
Article in English | MEDLINE | ID: mdl-10973929

ABSTRACT

Synthesis of fatty acid (FA) in adipose tissue requires cooperation of mitochondrial and cytoplasmic enzymes. Mitochondria are required for the production of ATP and they also support the formation of acetyl-CoA and NADPH in cytoplasm. Since cellular levels of all these metabolites depend on the efficiency of mitochondrial energy conversion, mitochondrial proton leak via uncoupling proteins (UCPs) could modulate FA synthesis. In 3T3-L1 adipocytes, 2,4-dinitrophenol depressed the synthesis of FA 4-fold while increasing FA oxidation 1. 5-fold and the production of lactate 14-fold. Inhibition of FA synthesis in 3T3-L1 adipocytes was proportional to the decrease in mitochondrial membrane potential. FA synthesis from D-[U-(14)C] glucose was reduced up to fourfold by ectopic UCP1 in the white fat of transgenic aP2-Ucp1 mice, reflecting the magnitude of UCP1 expression in different fat depots and the reduction of adiposity. Transcript levels for lipogenic enzymes were lower in the white fat of the transgenic mice than in the control animals. Our results show that uncoupling of oxidative phosphorylation depresses FA synthesis in white fat. Reduction of adiposity via mitochondrial uncoupling in white fat not only reflects increased energy expenditure, but also decreased in situ lipogenesis.


Subject(s)
Adipose Tissue/metabolism , Fatty Acids/biosynthesis , Mitochondria/metabolism , 3T3 Cells , Adipose Tissue/cytology , Animals , Carrier Proteins/metabolism , Energy Metabolism , Gene Expression , Ion Channels , Male , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Mitochondria/physiology , Mitochondrial Proteins , Uncoupling Protein 1
16.
J Biol Chem ; 275(44): 34486-92, 2000 Nov 03.
Article in English | MEDLINE | ID: mdl-10931824

ABSTRACT

Among a selected group of mouse strains susceptible to dietary obesity, those with an enhanced capacity for Ucp1 and brown adipocyte induction in white fat preferentially lost body weight following adrenergic stimulation. Based on the generality of this mechanism for reducing obesity, a genetic analysis was initiated to identify genes that control brown adipocyte induction in white fat depots in mice. Quantitative trait locus (QTL) analysis was performed using the variations of retroperitoneal fat Ucp1 mRNA expression in progeny of genetic crosses between the A/J and C57BL/6J parental strains and selected AXB recombinant inbred strains. Three A/J-derived loci on chromosomes 2, 3, and 8 and one C57BL/6J locus on chromosome 19 were linked to Ucp1 induction in retroperitoneal fat. Although A/J-derived alleles seemed to contribute to elevated Ucp1 expression, the C57BL/6J allele on chromosome 19 increased Ucp1 mRNA to levels higher than parental values. Thus, novel patterns of C57BL/6J and A/J recombinant genotypes among the four mapped loci resulted in a transgressive variation of Ucp1 phenotypes. Although the extent of the interchromosomal interactions have not been fully explored, strong synergistic interactions occur between a C57BL/6J allele on chromosome 19 and an A/J allele on chromosome 8. In addition to selective synergistic interactions between loci, variations in recessive and dominant effects also contribute to the final levels of Ucp1 expression.


Subject(s)
Adipose Tissue/metabolism , Carrier Proteins/genetics , Gene Expression Regulation , Membrane Proteins/genetics , Animals , Base Sequence , Chromosome Mapping , Crosses, Genetic , DNA Primers , Genetic Variation , Ion Channels , Male , Mice , Mice, Inbred C57BL , Mitochondrial Proteins , Quantitative Trait, Heritable , Uncoupling Protein 1
17.
Am J Physiol ; 274(3): E527-33, 1998 03.
Article in English | MEDLINE | ID: mdl-9530137

ABSTRACT

The role of brown adipose tissue in total energy balance and cold-induced thermogenesis was studied. Mice expressing mitochondrial uncoupling protein 1 (UCP-1) from the fat-specific aP2 gene promoter (heterozygous and homozygous aP2-Ucp transgenic mice) and their nontransgenic C57BL6/J littermates were used. The transgenic animals are resistant to obesity induced by a high-fat diet, presumably due to ectopic synthesis of UCP-1 in white fat. These animals exhibited atrophy of brown adipose tissue, as indicated by smaller size of brown fat and reduction of its total UCP-1 and DNA contents. Norepinephrine-induced respiration (measured in pentobarbital sodium-anesthetized animals) was decreased proportionally to the dosage of the transgene, and the homozygous (but not heterozygous) transgenic mice exhibited a reduction in their capacity to maintain body temperature in the cold. Our results indicate that the role of brown fat in cold-induced thermogenesis cannot be substituted by increased energy expenditure in other tissues.


Subject(s)
Adipose Tissue, Brown/physiology , Body Temperature Regulation/physiology , Carrier Proteins/physiology , Cold Temperature , Membrane Proteins/physiology , Membrane Transport Proteins , Mitochondrial Proteins , Obesity/physiopathology , Adipose Tissue/physiology , Animals , Body Temperature Regulation/genetics , Body Weight , Carrier Proteins/genetics , Energy Metabolism , Homozygote , Immunity, Innate/genetics , Ion Channels , Male , Membrane Proteins/genetics , Mice , Mice, Inbred C57BL , Mice, Transgenic , Obesity/genetics , Promoter Regions, Genetic , Proteins/genetics , Proteins/physiology , Thyroid Gland/physiology , Transgenes , Uncoupling Protein 1 , Uncoupling Protein 2
18.
FEBS Lett ; 425(2): 185-90, 1998 Mar 27.
Article in English | MEDLINE | ID: mdl-9559644

ABSTRACT

To assess the putative role of mitochondrial uncoupling protein 2 (UCP2) during perinatal development, its expression was analysed in mice and rats. Expression was detected in a large range of foetal tissues. A unique developmental pattern of UCP2 expression was found in liver, where the level of UCP2 mRNA was about 30-fold higher in foetuses than in adults (mice data), and started to decline immediately after birth. Neither UCP1 nor UCP3 mRNA was expressed in foetal liver. As in adult liver, immunohistochemical analysis suggested exclusive localisation of UCP2 in the monocyte/macrophage cells. Our results indicate a role of UCP2 in haematopoietic system development.


Subject(s)
Gene Expression Regulation, Developmental , Liver/metabolism , Membrane Transport Proteins , Mitochondrial Proteins , Proteins/genetics , Animals , Electron Transport Complex IV/genetics , Enzyme Precursors/genetics , Female , Immunoenzyme Techniques , Ion Channels , Liver/embryology , Mice , Mice, Inbred C57BL , Protein Sorting Signals/genetics , Rats , Rats, Wistar , Uncoupling Protein 2
19.
Am J Physiol ; 270(5 Pt 1): E768-75, 1996 May.
Article in English | MEDLINE | ID: mdl-8967464

ABSTRACT

We seek to determine whether increased energy dissipation in adipose tissue can prevent obesity. Transgenic mice with C57BL6/J background and the adipocyte lipid-binding protein (aP2) gene promoter directing expression of the mitochondrial uncoupling protein (UCP) gene in white and brown fat were used. Physiologically, UCP is essential for nonshivering thermogenesis in brown fat. Mice were assigned to a chow or a high-fat (HF) diet at 3 mo of age. Over the next 25 wk, gains of body weight were similar in corresponding subgroups (n = 6-8) of female and male mice: 4-5 g in chow nontransgenic and transgenic, 20 g in HF nontransgenic, and 9-11 g in HF transgenic mice. The lower body weight gain in the HF transgenic vs. nontransgenic mice corresponded to a twofold lower feed efficiency. Gonadal fat was enlarged, but subcutaneous white fat was decreased in the transgenic vs. nontransgenic mice in both dietary conditions. The results suggest that UCP synthesized from the aP2 gene promoter is capable of reducing dietary obesity.


Subject(s)
Body Weight , Carrier Proteins/genetics , Dietary Fats/pharmacology , Membrane Proteins/genetics , Mice, Transgenic/genetics , Obesity/pathology , Adaptor Protein Complex 2 , Adaptor Protein Complex alpha Subunits , Adaptor Proteins, Vesicular Transport , Adipose Tissue/pathology , Animals , Body Composition , Eating , Female , Glucose/metabolism , Homeostasis , Ion Channels , Lipid Metabolism , Male , Mice , Mice, Inbred C57BL , Mitochondrial Proteins , Obesity/genetics , Obesity/physiopathology , Uncoupling Protein 1
20.
Am J Physiol ; 270(5 Pt 1): E776-86, 1996 May.
Article in English | MEDLINE | ID: mdl-8967465

ABSTRACT

C57BL6/J mice with the expression of the mitochondrial uncoupling protein (UCP) gene from the fat-specific aP2 gene promoter were used to study the mechanism by which the aP2-Ucp transgene affects adiposity and reduces high-fat diet induced obesity. In the transgenic mice, UCP synthesized in white fat was inserted into mitochondria, and oxygen uptake by epididymal fat fragments indicated UCP-induced thermogenesis. The respirometry data, UCP content, cytochrome oxidase activity, and tissue morphology suggested functional involution of brown fat. Despite 25- to 50-fold lower mitochondrial cytochrome oxidase activity in white than in brown fat cells, total oxidative capacity in white and brown adipose tissue is comparable. Appearance of novel small cells in the gonadal fat of the transgenic mice was associated with a higher DNA content than that of the nontransgenic mice. The results prove a potential of transgenically altered mitochondria in white fat to modulate adiposity and energy expenditure and suggest the existence of a yet unidentified site-specific link between energy metabolism in adipocytes and cellularity.


Subject(s)
Adipose Tissue/pathology , Carrier Proteins/genetics , Membrane Proteins/genetics , Mice, Transgenic/genetics , Obesity/pathology , Obesity/physiopathology , Adaptor Protein Complex 2 , Adaptor Protein Complex alpha Subunits , Adaptor Proteins, Vesicular Transport , Adipose Tissue/enzymology , Adipose Tissue, Brown/enzymology , Animals , Antigens/analysis , Carrier Proteins/immunology , Carrier Proteins/metabolism , Dietary Fats , Electron Transport Complex IV/immunology , Electron Transport Complex IV/metabolism , Ion Channels , Lipoprotein Lipase/metabolism , Male , Membrane Proteins/immunology , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Mitochondrial Proteins , Obesity/genetics , RNA, Messenger/metabolism , Respiration , Uncoupling Protein 1
SELECTION OF CITATIONS
SEARCH DETAIL
...