Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
Nat Med ; 23(5): 623-630, 2017 May.
Article in English | MEDLINE | ID: mdl-28414329

ABSTRACT

Adaptive thermogenesis is the process of heat generation in response to cold stimulation. It is under the control of the sympathetic nervous system, whose chief effector is the catecholamine norepinephrine (NE). NE enhances thermogenesis through ß3-adrenergic receptors to activate brown adipose tissue and by 'browning' white adipose tissue. Recent studies have reported that alternative activation of macrophages in response to interleukin (IL)-4 stimulation induces the expression of tyrosine hydroxylase (TH), a key enzyme in the catecholamine synthesis pathway, and that this activation provides an alternative source of locally produced catecholamines during the thermogenic process. Here we report that the deletion of Th in hematopoietic cells of adult mice neither alters energy expenditure upon cold exposure nor reduces browning in inguinal adipose tissue. Bone marrow-derived macrophages did not release NE in response to stimulation with IL-4, and conditioned media from IL-4-stimulated macrophages failed to induce expression of thermogenic genes, such as uncoupling protein 1 (Ucp1), in adipocytes cultured with the conditioned media. Furthermore, chronic treatment with IL-4 failed to increase energy expenditure in wild-type, Ucp1-/- and interleukin-4 receptor-α double-negative (Il4ra-/-) mice. In agreement with these findings, adipose-tissue-resident macrophages did not express TH. Thus, we conclude that alternatively activated macrophages do not synthesize relevant amounts of catecholamines, and hence, are not likely to have a direct role in adipocyte metabolism or adaptive thermogenesis.


Subject(s)
Adipocytes/metabolism , Adipose Tissue/metabolism , Macrophages/immunology , Norepinephrine/metabolism , Receptors, Adrenergic, beta-3/metabolism , Thermogenesis/immunology , Tyrosine 3-Monooxygenase/genetics , Adaptation, Physiological , Adipocytes/drug effects , Adipose Tissue/drug effects , Adipose Tissue, Brown/drug effects , Adipose Tissue, Brown/metabolism , Adipose Tissue, White/drug effects , Adipose Tissue, White/metabolism , Animals , Blotting, Western , Body Composition/immunology , Catecholamines/metabolism , Cell Differentiation , Culture Media, Conditioned , Energy Metabolism/genetics , Flow Cytometry , Fluorescent Antibody Technique , Gene Expression Profiling , Interleukin-4/immunology , Interleukin-4/pharmacology , Macrophages/drug effects , Mice , Mice, Knockout , Receptors, Cell Surface/genetics , Thermogenesis/genetics , Uncoupling Protein 1/genetics
2.
Nat Med ; 22(10): 1120-1130, 2016 10.
Article in English | MEDLINE | ID: mdl-27571348

ABSTRACT

Cachexia represents a fatal energy-wasting syndrome in a large number of patients with cancer that mostly results in a pathological loss of skeletal muscle and adipose tissue. Here we show that tumor cell exposure and tumor growth in mice triggered a futile energy-wasting cycle in cultured white adipocytes and white adipose tissue (WAT), respectively. Although uncoupling protein 1 (Ucp1)-dependent thermogenesis was dispensable for tumor-induced body wasting, WAT from cachectic mice and tumor-cell-supernatant-treated adipocytes were consistently characterized by the simultaneous induction of both lipolytic and lipogenic pathways. Paradoxically, this was accompanied by an inactivated AMP-activated protein kinase (Ampk), which is normally activated in peripheral tissues during states of low cellular energy. Ampk inactivation correlated with its degradation and with upregulation of the Ampk-interacting protein Cidea. Therefore, we developed an Ampk-stabilizing peptide, ACIP, which was able to ameliorate WAT wasting in vitro and in vivo by shielding the Cidea-targeted interaction surface on Ampk. Thus, our data establish the Ucp1-independent remodeling of adipocyte lipid homeostasis as a key event in tumor-induced WAT wasting, and we propose the ACIP-dependent preservation of Ampk integrity in the WAT as a concept in future therapies for cachexia.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Adipocytes, White/drug effects , Adipose Tissue, White/drug effects , Apoptosis Regulatory Proteins/drug effects , Cachexia/metabolism , Lipid Metabolism/drug effects , Neoplasms/metabolism , Peptide Fragments/pharmacology , AMP-Activated Protein Kinases/pharmacology , Adipocytes, White/metabolism , Adipose Tissue, White/metabolism , Animals , Apoptosis Regulatory Proteins/metabolism , Cachexia/etiology , Cells, Cultured , In Vitro Techniques , Lipogenesis/drug effects , Lipolysis/drug effects , Mice , Neoplasms/complications , Thermogenesis/drug effects , Uncoupling Protein 1/drug effects , Uncoupling Protein 1/metabolism
3.
PLoS One ; 10(2): e0118534, 2015.
Article in English | MEDLINE | ID: mdl-25714366

ABSTRACT

Regulatory T (Treg) cells are critical determinants of both immune responses and metabolic control. Here we show that systemic ablation of Treg cells compromised the adaptation of whole-body energy expenditure to cold exposure, correlating with impairment in thermogenic marker gene expression and massive invasion of pro-inflammatory macrophages in brown adipose tissue (BAT). Indeed, BAT harbored a unique sub-set of Treg cells characterized by a unique gene signature. As these Treg cells respond to BAT activation upon cold exposure, this study defines a BAT-specific Treg sub-set with direct implications for the regulation of energy homeostasis in response to environmental stress.


Subject(s)
Adipose Tissue, Brown/immunology , T-Lymphocyte Subsets/immunology , T-Lymphocytes, Regulatory/immunology , Adipose Tissue, Brown/metabolism , Adipose Tissue, Brown/pathology , Animals , Female , Gene Expression Profiling , Gene Expression Regulation , Inflammation/genetics , Inflammation/immunology , Inflammation/metabolism , Metabolic Networks and Pathways , Metabolome , Metabolomics/methods , Mice , Phenotype , Spleen/cytology , Spleen/immunology , Spleen/metabolism , T-Lymphocyte Subsets/metabolism , T-Lymphocytes, Regulatory/metabolism
4.
Lipids ; 49(11): 1081-9, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25204579

ABSTRACT

Long-chain n-3 polyunsaturated fatty acids (LC n-3 PUFA) in the diet protect against insulin resistance and obesity. Fibroblast growth factor-21 (Fgf21) is a hormonal factor released mainly by the liver that has powerful anti-diabetic effects. Here, we tested whether the beneficial metabolic effects of LC n-3 PUFA involve the induction of Fgf21. C57BL/6 J mice were exposed to an obesogenic, corn-oil-based, high-fat diet (cHF), or a diet in which corn oil was replaced with a fish-derived LC n-3 PUFA concentrate (cHF + F) using two experimental settings: short-term (3 weeks) and long-term treatment (8 weeks). CHF + F reduced body weight gain, insulinemia, and triglyceridemia compared to cHF. cHF increased plasma Fgf21 levels and hepatic Fgf21 gene expression compared with controls, but these effects were less pronounced or absent in cHF + F-fed mice. In contrast, hepatic expression of peroxisome proliferator-activated receptor (PPAR)-α target genes were more strongly induced by cHF + F than cHF, especially in the short-term treatment setting. The expression of genes encoding Fgf21, its receptors, and Fgf21 targets was unaltered by short-term LC n-3 PUFA treatment, with the exception of Ucp1 (uncoupling protein 1) and adiponectin genes, which were specifically up-regulated in white fat. In the long-term treatment setting, the expression of Fgf21 target genes and receptors was not differentially affected by LC n-3 PUFA. Collectively, our findings indicate that increased Fgf21 levels do not appear to be a major mechanism through which LC n-3 PUFA ameliorates high-fat-diet-associated metabolic disorders.


Subject(s)
Fatty Acids, Omega-3/pharmacology , Fibroblast Growth Factors/blood , Fibroblast Growth Factors/genetics , Gene Expression/drug effects , Adiponectin/genetics , Adipose Tissue/drug effects , Adipose Tissue/metabolism , Animals , Body Weight/drug effects , Corn Oil/administration & dosage , Diet, High-Fat , Fatty Acids, Omega-3/administration & dosage , Insulin/blood , Ion Channels/genetics , Liver/drug effects , Liver/metabolism , Male , Mice, Inbred C57BL , Mitochondrial Proteins/genetics , PPAR alpha/genetics , Reverse Transcriptase Polymerase Chain Reaction , Time Factors , Triglycerides/blood , Uncoupling Protein 1 , Up-Regulation/drug effects , Weight Gain/drug effects
5.
Semin Immunopathol ; 36(1): 13-25, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24212254

ABSTRACT

Throughout evolution, effective nutrient sensing and control of systemic energy homeostasis have relied on a close physical and functional interaction between immune and metabolically active cells. However, in today's obesogenic environment, this fine-tuned immunometabolic interface is perturbed. As a consequence, chronic inflammatory conditions and aberrant activation of immune cells have emerged as key features of obesity-related metabolic disorders, including insulin resistance, cardiovascular complications, and type 2 diabetes, whereas a major research focus has been placed on the adipocyte-macrophage interaction in the context of metabolic dysfunction; recent studies have not only expanded the scope of relevant immune cells in this setting but also highlight the impact of distinct metabolic organs, including the liver, on immunometabolic control, metabolic disease development, and potential anti-inflammatory therapeutic options in obesity-driven pathologies. This review will thus summarize recent progress in this emerging area of metabolic research.


Subject(s)
Metabolic Syndrome/immunology , Metabolic Syndrome/metabolism , Adipose Tissue/immunology , Adipose Tissue/metabolism , Animals , Humans , Immunity, Innate/physiology , Inflammation/immunology , Inflammation/metabolism , Insulin Resistance , Liver/cytology , Liver/immunology , Liver/metabolism , Obesity/immunology , Obesity/metabolism
6.
Biochim Biophys Acta ; 1841(2): 267-78, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24295779

ABSTRACT

Non-alcoholic fatty liver disease (NAFLD) accompanies obesity and insulin resistance. Recent meta-analysis suggested omega-3 polyunsaturated fatty acids DHA and EPA to decrease liver fat in NAFLD patients. Antiinflammatory, hypolipidemic, and insulin-sensitizing effects ofDHA/EPA depend on their lipid form, with marine phospholipids showing better efficacy than fish oils. We characterized the mechanisms underlying beneficial effects of DHA/EPA phospholipids, alone or combined with an antidiabetic drug, on hepatosteatosis. C57BL/6N mice were fed for 7 weeks an obesogenic high-fat diet (cHF) or cHF-based interventions: (i) cHF supplemented with phosphatidylcholine-rich concentrate from herring (replacing 10% of dietary lipids; PC), (ii) cHF containing rosiglitazone (10 mg/kg diet; R), or (iii) PC + R. Metabolic analyses, hepatic gene expression and lipidome profiling were performed. Results showed that PC and PC + R prevented cHlF-induced weight gain and glucose intolerance, while all interventions reduced abdominal fat and plasma triacylglycerols. PC and PC + R also lowered hepatic and plasma cholesterol and reduced hepatosteatosis. Microarray analysis revealed integrated downregulation of hepatic lipogenic and cholesterol biosynthesis pathways by PC, while R-induced lipogenesis was fully counteracted in PC + R Gene expression changes in PC and PC + R were associated with preferential enrichment of hepatic phosphatidylcholine and phosphatidylethanolamine fractions by DHA/EPA. The complex downregulation of hepatic lipogenic and cholesterol biosynthesis genes and the antisteatotic effects were unique to DHA/EPA-containing phospholipids, since they were absent in mice fed soy-derived phosphatidylcholine. Thus, inhibition of lipid and cholesterol biosynthesis associated with potent antisteatotic effects in the liver in response to DHA/EPA-containing phospholipids support their use in NAFLD prevention and treatment.


Subject(s)
Fatty Acids, Omega-3/pharmacology , Fatty Liver/prevention & control , Phospholipids/pharmacology , Animals , Biosynthetic Pathways/drug effects , Cholesterol/biosynthesis , Liver/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Obese , Non-alcoholic Fatty Liver Disease , Triglycerides/biosynthesis
7.
Biochim Biophys Acta ; 1831(5): 896-904, 2013 May.
Article in English | MEDLINE | ID: mdl-23051608

ABSTRACT

Epidemiological studies estimate that by the year 2030, 2.16 billion people worldwide will be overweight and 1.12 billion will be obese [1]. Besides its now established function as an endocrine organ, adipose tissue plays a fundamental role as an energy storage compartment. As such, adipose tissue is capable of extensive expansion or retraction depending on the energy balance or disease state of the host, a plasticity that is unparalleled in other organs and - under conditions of excessive energy intake - significantly contributes to the afore mentioned obesity pandemic. Expansion of adipose tissue is driven by both hypertrophy and hyperplasia of adipocytes, which can renew frequently to compensate for cell death. This underlines the importance of adipocyte progenitor cells within the distinct adipose tissue depots to control both energy storage and endocrine functions of adipose tissue. Here we summarize recent findings on the identity and plasticity of adipose stem cells, the involved signaling cascades, and potential clinical implications of these cells for the treatment of metabolic dysfunction in obesity. This article is part of a Special Issue entitled Brown and White Fat: From Signaling to Disease.


Subject(s)
Adipose Tissue, Brown/cytology , Adipose Tissue, White/cytology , Obesity/physiopathology , Stem Cells/cytology , Animals , Humans , Signal Transduction
8.
PLoS One ; 7(8): e43764, 2012.
Article in English | MEDLINE | ID: mdl-22952760

ABSTRACT

Insulin resistance, the key defect in type 2 diabetes (T2D), is associated with a low capacity to adapt fuel oxidation to fuel availability, i.e., metabolic inflexibility. This, in turn, contributes to a further damage of insulin signaling. Effectiveness of T2D treatment depends in large part on the improvement of insulin sensitivity and metabolic adaptability of the muscle, the main site of whole-body glucose utilization. We have shown previously in mice fed an obesogenic high-fat diet that a combined use of n-3 long-chain polyunsaturated fatty acids (n-3 LC-PUFA) and thiazolidinediones (TZDs), anti-diabetic drugs, preserved metabolic health and synergistically improved muscle insulin sensitivity. We investigated here whether n-3 LC-PUFA could elicit additive beneficial effects on metabolic flexibility when combined with a TZD drug rosiglitazone. Adult male C57BL/6N mice were fed an obesogenic corn oil-based high-fat diet (cHF) for 8 weeks, or randomly assigned to various interventions: cHF with n-3 LC-PUFA concentrate replacing 15% of dietary lipids (cHF+F), cHF with 10 mg rosiglitazone/kg diet (cHF+ROSI), cHF+F+ROSI, or chow-fed. Indirect calorimetry demonstrated superior preservation of metabolic flexibility to carbohydrates in response to the combined intervention. Metabolomic and gene expression analyses in the muscle suggested distinct and complementary effects of the interventions, with n-3 LC-PUFA supporting complete oxidation of fatty acids in mitochondria and the combination with n-3 LC-PUFA and rosiglitazone augmenting insulin sensitivity by the modulation of branched-chain amino acid metabolism. These beneficial metabolic effects were associated with the activation of the switch between glycolytic and oxidative muscle fibers, especially in the cHF+F+ROSI mice. Our results further support the idea that the combined use of n-3 LC-PUFA and TZDs could improve the efficacy of the therapy of obese and diabetic patients.


Subject(s)
Diet, High-Fat/adverse effects , Fatty Acids, Omega-3/pharmacology , Muscle, Skeletal/drug effects , Muscle, Skeletal/metabolism , Obesity/etiology , Obesity/metabolism , Thiazolidinediones/pharmacology , Animals , Drug Synergism , Gene Expression Regulation/drug effects , Glycolysis/drug effects , Male , Metabolomics , Mice , Mice, Inbred C57BL , Muscle Fibers, Skeletal/drug effects , Muscle Fibers, Skeletal/metabolism , Oxidation-Reduction/drug effects , Rosiglitazone
9.
PLoS One ; 7(6): e38834, 2012.
Article in English | MEDLINE | ID: mdl-22701720

ABSTRACT

BACKGROUND: n-3 polyunsaturated fatty acids, namely docosahexaenoic acid (DHA) and eicosapentaenoic acid (EPA), reduce the risk of cardiovascular disease and can ameliorate many of obesity-associated disorders. We hypothesised that the latter effect will be more pronounced when DHA/EPA is supplemented as phospholipids rather than as triglycerides. METHODOLOGY/PRINCIPAL FINDINGS: In a 'prevention study', C57BL/6J mice were fed for 9 weeks on either a corn oil-based high-fat obesogenic diet (cHF; lipids ∼35% wt/wt), or cHF-based diets in which corn oil was partially replaced by DHA/EPA, admixed either as phospholipids or triglycerides from marine fish. The reversal of obesity was studied in mice subjected to the preceding cHF-feeding for 4 months. DHA/EPA administered as phospholipids prevented glucose intolerance and tended to reduce obesity better than triglycerides. Lipemia and hepatosteatosis were suppressed more in response to dietary phospholipids, in correlation with better bioavailability of DHA and EPA, and a higher DHA accumulation in the liver, white adipose tissue (WAT), and muscle phospholipids. In dietary obese mice, both DHA/EPA concentrates prevented a further weight gain, reduced plasma lipid levels to a similar extent, and tended to improve glucose tolerance. Importantly, only the phospholipid form reduced plasma insulin and adipocyte hypertrophy, while being more effective in reducing hepatic steatosis and low-grade inflammation of WAT. These beneficial effects were correlated with changes of endocannabinoid metabolome in WAT, where phospholipids reduced 2-arachidonoylglycerol, and were more effective in increasing anti-inflammatory lipids such as N-docosahexaenoylethanolamine. CONCLUSIONS/SIGNIFICANCE: Compared with triglycerides, dietary DHA/EPA administered as phospholipids are superior in preserving a healthy metabolic profile under obesogenic conditions, possibly reflecting better bioavalability and improved modulation of the endocannabinoid system activity in WAT.


Subject(s)
Cannabinoid Receptor Modulators/metabolism , Diet, High-Fat , Endocannabinoids , Fatty Acids, Omega-3/metabolism , Obesity/diet therapy , Phospholipids/metabolism , Adipose Tissue, White/metabolism , Analysis of Variance , Animals , Biological Availability , Body Weight , Docosahexaenoic Acids/metabolism , Eicosapentaenoic Acid/metabolism , Fatty Acids, Omega-3/administration & dosage , Fatty Acids, Omega-3/pharmacology , Immunohistochemistry , Liver/drug effects , Liver/metabolism , Male , Metabolomics , Mice , Mice, Inbred C57BL , Microscopy , Muscle, Skeletal/metabolism , Obesity/prevention & control , Real-Time Polymerase Chain Reaction , Triglycerides/metabolism
10.
Clin Sci (Lond) ; 121(1): 29-41, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21275906

ABSTRACT

Advanced HF (heart failure) is associated with altered substrate metabolism. Whether modification of substrate use improves the course of HF remains unknown. The antihyperglycaemic drug MET (metformin) affects substrate metabolism, and its use might be associated with improved outcome in diabetic HF. The aim of the present study was to examine whether MET would improve cardiac function and survival also in non-diabetic HF. Volume-overload HF was induced in male Wistar rats by creating ACF (aortocaval fistula). Animals were randomized to placebo/MET (300 mg·kg(-1) of body weight·day(-1), 0.5% in food) groups and underwent assessment of metabolism, cardiovascular and mitochondrial functions (n=6-12/group) in advanced HF stage (week 21). A separate cohort served for survival analysis (n=10-90/group). The ACF group had marked cardiac hypertrophy, increased LVEDP (left ventricular end-diastolic pressure) and lung weight confirming decompensated HF, increased circulating NEFAs (non-esterified 'free' fatty acids), intra-abdominal fat depletion, lower glycogen synthesis in the skeletal muscle (diaphragm), lower myocardial triacylglycerol (triglyceride) content and attenuated myocardial (14)C-glucose and (14)C-palmitate oxidation, but preserved mitochondrial respiratory function, glucose tolerance and insulin sensitivity. MET therapy normalized serum NEFAs, decreased myocardial glucose oxidation, increased myocardial palmitate oxidation, but it had no effect on myocardial gene expression, AMPK (AMP-activated protein kinase) signalling, ATP level, mitochondrial respiration, cardiac morphology, function and long-term survival, despite reaching therapeutic serum levels (2.2±0.7 µg/ml). In conclusion, MET-induced enhancement of myocardial fatty acid oxidation had a neutral effect on cardiac function and survival. Recently reported cardioprotective effects of MET may not be universal to all forms of HF and may require AMPK activation or ATP depletion. No increase in mortality on MET supports its safe use in diabetic HF.


Subject(s)
Heart Failure/drug therapy , Hypoglycemic Agents/therapeutic use , Metformin/therapeutic use , AMP-Activated Protein Kinase Kinases , Animals , Blood Glucose/metabolism , Body Weight/drug effects , Disease Models, Animal , Drug Evaluation, Preclinical , Glycogen/metabolism , Heart Failure/diagnostic imaging , Heart Failure/physiopathology , Hemodynamics/drug effects , Hypoglycemic Agents/blood , Lipid Metabolism/drug effects , Lung/pathology , Male , Metformin/blood , Mitochondria, Heart/physiology , Myocardium/metabolism , Myocardium/pathology , Organ Size/drug effects , Protein Kinases/metabolism , Rats , Rats, Wistar , Survival Analysis , Ultrasonography
11.
Diabetes ; 59(11): 2737-46, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20693347

ABSTRACT

OBJECTIVE: The induction of obesity, dyslipidemia, and insulin resistance by high-fat diet in rodents can be prevented by n-3 long-chain polyunsaturated fatty acids (LC-PUFAs). We tested a hypothesis whether AMP-activated protein kinase (AMPK) has a role in the beneficial effects of n-3 LC-PUFAs. RESEARCH DESIGN AND METHODS: Mice with a whole-body deletion of the α2 catalytic subunit of AMPK (AMPKα2(-/-)) and their wild-type littermates were fed on either a low-fat chow, or a corn oil-based high-fat diet (cHF), or a cHF diet with 15% lipids replaced by n-3 LC-PUFA concentrate (cHF+F). RESULTS: Feeding a cHF diet induced obesity, dyslipidemia, hepatic steatosis, and whole-body insulin resistance in mice of both genotypes. Although cHF+F feeding increased hepatic AMPKα2 activity, the body weight gain, dyslipidemia, and the accumulation of hepatic triglycerides were prevented by the cHF+F diet to a similar degree in both AMPKα2(-/-) and wild-type mice in ad libitum-fed state. However, preservation of hepatic insulin sensitivity by n-3 LC-PUFAs required functional AMPKα2 and correlated with the induction of adiponectin and reduction in liver diacylglycerol content. Under hyperinsulinemic-euglycemic conditions, AMPKα2 was essential for preserving low levels of both hepatic and plasma triglycerides, as well as plasma free fatty acids, in response to the n-3 LC-PUFA treatment. CONCLUSIONS: Our results show that n-3 LC-PUFAs prevent hepatic insulin resistance in an AMPKα2-dependent manner and support the role of adiponectin and hepatic diacylglycerols in the regulation of insulin sensitivity. AMPKα2 is also essential for hypolipidemic and antisteatotic effects of n-3 LC-PUFA under insulin-stimulated conditions.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Fatty Acids, Omega-3/metabolism , Fatty Acids, Unsaturated/metabolism , Liver/physiology , AMP-Activated Protein Kinases/deficiency , Animals , Cell Culture Techniques , Diet, Fat-Restricted , Dietary Fats/pharmacology , Fatty Acids, Omega-3/therapeutic use , Fatty Acids, Unsaturated/pharmacology , Glucose Clamp Technique , Hepatocytes/cytology , Hepatocytes/physiology , Hyperinsulinism , Insulin Resistance , Liver/drug effects , Liver/enzymology , Metabolic Syndrome/prevention & control , Mice , Mice, Knockout , Protein Subunits/metabolism
12.
Obesity (Silver Spring) ; 17(5): 1023-31, 2009 May.
Article in English | MEDLINE | ID: mdl-19148125

ABSTRACT

The n-3 polyunsaturated fatty acids, especially eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA), exert hypolipidemic effects and prevent development of obesity and insulin resistance in animals fed high-fat diets. We sought to determine the efficacy of alpha-substituted DHA derivatives as lipid-lowering, antiobesity, and antidiabetic agents. C57BL/6 mice were given a corn oil-based high-fat (35% weight/weight) diet (cHF), or cHF with 1.5% of lipids replaced with alpha-methyl DHA ethyl ester (Substance 1), alpha-ethyl DHA ethyl ester (Substance 2), alpha,alpha-di-methyl DHA ethyl ester (Substance 3), or alpha-thioethyl DHA ethyl ester (Substance 4) for 4 months. Plasma markers of glucose and lipid metabolism, glucose tolerance, morphology, tissue lipid content, and gene regulation were characterized. The cHF induced obesity, hyperlipidemia, impairment of glucose homeostasis, and adipose tissue inflammation. Except for Substance 3, all other substances prevented weight gain and Substance 2 exerted the strongest effect (63% of cHF-controls). Glucose intolerance was significantly prevented (~67% of cHF) by both Substance 1 and Substance 2. Moreover, Substance 2 lowered fasting glycemia, plasma insulin, triacylglycerols, and nonesterified fatty acids (73, 9, 47, and 81% of cHF-controls, respectively). Substance 2 reduced accumulation of lipids in liver and skeletal muscle, as well as adipose tissue inflammation associated with obesity. Substance 2 also induced weight loss in dietary obese mice. In contrast to DHA administered either alone or as a component of the EPA/DHA concentrate (replacing 15% of dietary lipids), Substance 2 also reversed established glucose intolerance in obese mice. Thus, Substance 2 represents a novel compound with a promising potential in the treatment of obesity and associated metabolic disturbances.


Subject(s)
Docosahexaenoic Acids/therapeutic use , Glucose Intolerance/drug therapy , Obesity/drug therapy , Animals , Dietary Fats , Disease Models, Animal , Energy Intake , Glucose/metabolism , Glucose Intolerance/prevention & control , Glucose Tolerance Test , Hypolipidemic Agents/therapeutic use , Male , Mice , Mice, Inbred C57BL , Obesity/prevention & control , Polymerase Chain Reaction , RNA/genetics , RNA/isolation & purification , Triglycerides/metabolism , Weight Gain
13.
Am J Physiol Endocrinol Metab ; 295(2): E356-67, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18492779

ABSTRACT

The obesogenic effect of a high-fat (HF) diet is counterbalanced by stimulation of energy expenditure and lipid oxidation in response to a meal. The aim of this study was to reveal whether muscle nonshivering thermogenesis could be stimulated by a HF diet, especially in obesity-resistant A/J compared with obesity-prone C57BL/6J (B/6J) mice. Experiments were performed on male mice born and maintained at 30 degrees C. Four-week-old mice were randomly weaned onto a low-fat (LF) or HF diet for 2 wk. In the A/J LF mice, cold exposure (4 degrees C) resulted in hypothermia, whereas the A/J HF, B/6J LF, and B/6J HF mice were cold tolerant. Cold sensitivity of the A/J LF mice was associated with a relatively low whole body energy expenditure under resting conditions, which was normalized by the HF diet. In both strains, the HF diet induced uncoupling protein-1-mediated thermogenesis, with a stronger induction in A/J mice. Only in A/J mice: 1) the HF diet augmented activation of whole body lipid oxidation by cold; and 2) at 30 degrees C, oxygen consumption, total content, and phosphorylation of AMP-activated protein kinase (AMPK), and AICAR-stimulated palmitate oxidation in soleus muscle was increased by the HF diet in parallel with significantly increased leptinemia. Gene expression data in soleus muscle of the A/J HF mice indicated a shift from carbohydrate to fatty acid oxidation. Our results suggest a role for muscle nonshivering thermogenesis and lipid oxidation in the obesity-resistant phenotype of A/J mice and indicate that a HF diet could induce thermogenesis in oxidative muscle, possibly via the leptin-AMPK axis.


Subject(s)
Dietary Fats/administration & dosage , Muscle, Skeletal/physiology , Thermogenesis/physiology , AMP-Activated Protein Kinases , Aminoimidazole Carboxamide/analogs & derivatives , Aminoimidazole Carboxamide/metabolism , Animals , Animals, Newborn , Basal Metabolism , Body Temperature/physiology , Body Weight/physiology , Calorimetry, Indirect , Dietary Fats/metabolism , Fatty Acids, Nonesterified/blood , Male , Mice , Mice, Inbred C57BL , Multienzyme Complexes/metabolism , Muscle, Skeletal/metabolism , Obesity/etiology , Obesity/metabolism , Oxygen Consumption/physiology , Protein Serine-Threonine Kinases/metabolism , Random Allocation , Ribonucleotides/metabolism , Triglycerides/blood
14.
FEBS Lett ; 581(6): 1093-7, 2007 Mar 20.
Article in English | MEDLINE | ID: mdl-17316620

ABSTRACT

Mitochondrial uncoupling protein 2 (UCP2) is abundant in developing monocyte/macrophage cells and may affect hematopoiesis by reducing formation of reactive oxygen species. The aims of this study were to further characterize the involvement of UCP2 in hematopoiesis. In situ hybridization in mouse embryos identified UCP2-positive cells in liver and inside primitive blood vessels from 10.5 days of prenatal development. High UCP2 transcript levels were detected in reticulocytes and other maturating erythroid cells in peripheral blood of mice exposed to hypoxia, and in umbilical cord blood of human neonates and peripheral blood of adults. Our results suggest involvement of UCP2 in erythropoiesis.


Subject(s)
Cellular Senescence/genetics , Erythroid Cells/cytology , Proteins/genetics , RNA, Messenger/analysis , Animals , Embryo, Mammalian , Erythroid Cells/chemistry , Erythropoiesis/genetics , Fetal Blood , Humans , Liver/cytology , Mice , Proteins/analysis , Proteins/physiology , Reticulocytes/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...