Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 13 de 13
1.
Nature ; 574(7776): 63-68, 2019 10.
Article En | MEDLINE | ID: mdl-31554967

The gp130 receptor cytokines IL-6 and CNTF improve metabolic homeostasis but have limited therapeutic use for the treatment of type 2 diabetes. Accordingly, we engineered the gp130 ligand IC7Fc, in which one gp130-binding site is removed from IL-6 and replaced with the LIF-receptor-binding site from CNTF, fused with the Fc domain of immunoglobulin G, creating a cytokine with CNTF-like, but IL-6-receptor-dependent, signalling. Here we show that IC7Fc improves glucose tolerance and hyperglycaemia and prevents weight gain and liver steatosis in mice. In addition, IC7Fc either increases, or prevents the loss of, skeletal muscle mass by activation of the transcriptional regulator YAP1. In human-cell-based assays, and in non-human primates, IC7Fc treatment results in no signs of inflammation or immunogenicity. Thus, IC7Fc is a realistic next-generation biological agent for the treatment of type 2 diabetes and muscle atrophy, disorders that are currently pandemic.


Cytokine Receptor gp130/metabolism , Cytokines/chemical synthesis , Cytokines/therapeutic use , Diabetes Mellitus, Type 2/drug therapy , Immunoglobulin G/therapeutic use , Recombinant Fusion Proteins/therapeutic use , Adaptor Proteins, Signal Transducing/metabolism , Animals , Binding, Competitive , Cytokines/chemistry , Diabetes Mellitus, Type 2/metabolism , Drug Design , Fatty Liver/prevention & control , Glucose Tolerance Test , Humans , Hyperglycemia/drug therapy , Hyperglycemia/metabolism , Incretins/metabolism , Interleukin-6/antagonists & inhibitors , Interleukin-6/metabolism , Male , Mice , Muscle, Skeletal/drug effects , Obesity/metabolism , Pancreas/metabolism , Phosphoproteins/metabolism , Protein Engineering , Receptors, Interleukin-6/metabolism , Signal Transduction , Transcription Factors , Weight Gain/drug effects , YAP-Signaling Proteins
2.
Basic Clin Pharmacol Toxicol ; 123(6): 732-738, 2018 Dec.
Article En | MEDLINE | ID: mdl-29956485

The use of anaesthetics severely influences substrate metabolism. This poses challenges for patients in clinical settings and for the use of animals in diabetes research. Sevoflurane can affect regulation of glucose homoeostasis at several steps, but the tissue-specific response remains to be determined. The aim of the study was to investigate the pharmacological effect of sevoflurane anaesthesia on glucose homoeostasis during hyperinsulinaemic clamp conditions, the gold standard method for assessment of whole-body insulin sensitivity. Conscious mice (n = 6) and mice under sevoflurane anaesthesia (n = 8) underwent a hyperinsulinaemic clamp where constant infusion of insulin and donor blood was administered during variable glucose infusion to maintain isoglycaemia. 2-[1-14 C]-deoxy-D-glucose was infused to determine tissue-specific uptake of glucose in adipose tissue, heart, brain and skeletal muscle. Sevoflurane anaesthesia severely impaired insulin-stimulated whole-body glucose uptake demonstrated by a 50% lower glucose infusion rate (GIR). This was associated with decreased glucose uptake in brain, soleus, triceps and gastrocnemius muscles in sevoflurane-anaesthetized mice compared to conscious mice. Plasma-free fatty acids (FFA), a potent inducer of insulin resistance, increased by 42% in mice during sevoflurane anaesthesia. In addition, insulin secretion from pancreatic ß-cell was lower in fasted, anaesthetized mice. Sevoflurane anaesthesia impairs insulin secretion, induces insulin resistance in mice and reduces glucose uptake in non-insulin-sensitive tissue like the brain. The underlying mechanisms may involve sevoflurane-induced mobilization of FFA.


Anesthetics, Inhalation/pharmacology , Glucose/antagonists & inhibitors , Insulin Antagonists/pharmacology , Insulin/metabolism , Sevoflurane/pharmacology , Animals , Blood Glucose/analysis , Fatty Acids, Nonesterified/blood , Glucose/metabolism , Glucose Clamp Technique , Hyperglycemia/chemically induced , Insulin/blood , Insulin Resistance , Male , Mice , Mice, Inbred C57BL
3.
Am J Physiol Endocrinol Metab ; 314(4): E377-E395, 2018 04 01.
Article En | MEDLINE | ID: mdl-29208611

Nicotinamide adenine dinucleotide (NAD+) can be synthesized by nicotinamide phosphoribosyltransferase (NAMPT). We aimed to determine the role of NAMPT in maintaining NAD+ levels, mitochondrial function, and metabolic homeostasis in skeletal muscle cells. We generated stable Nampt knockdown (sh Nampt KD) C2C12 cells using a shRNA lentiviral approach. Moreover, we applied gene electrotransfer to express Cre recombinase in tibialis anterior muscle of floxed Nampt mice. In sh Nampt KD C2C12 myoblasts, Nampt and NAD+ levels were reduced by 70% and 50%, respectively, and maximal respiratory capacity was reduced by 25%. Moreover, anaerobic glycolytic flux increased by 55%, and 2-deoxyglucose uptake increased by 25% in sh Nampt KD cells. Treatment with the NAD+ precursor nicotinamide riboside restored NAD+ levels in sh Nampt cells and increased maximal respiratory capacity by 18% and 32% in control and sh Nampt KD cells, respectively. Expression of Cre recombinase in muscle of floxed Nampt mice reduced NAMPT and NAD+ levels by 38% and 43%, respectively. Glucose uptake increased by 40%, and mitochondrial complex IV respiration was compromised by 20%. Hypoxia-inducible factor (HIF)-1α-regulated genes and histone H3 lysine 9 (H3K9) acetylation, a known sirtuin 6 (SIRT6) target, were increased in shNampt KD cells. Thus, we propose that the shift toward glycolytic metabolism observed, at least in part, is mediated by the SIRT6/HIF1α axis. Our findings suggest that NAMPT plays a key role for maintaining NAD+ levels in skeletal muscle and that NAMPT deficiency compromises oxidative phosphorylation capacity and alters energy homeostasis in this tissue.


Cytokines/genetics , Energy Metabolism/genetics , Mitochondria, Muscle/physiology , Muscle, Skeletal/metabolism , Myoblasts/metabolism , NAD/metabolism , Nicotinamide Phosphoribosyltransferase/genetics , Animals , Carbohydrate Metabolism/genetics , Cells, Cultured , Cytokines/metabolism , Homeostasis/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Nicotinamide Phosphoribosyltransferase/metabolism , Oxidative Phosphorylation , Signal Transduction/genetics
4.
Metabolism ; 65(12): 1706-1719, 2016 Dec.
Article En | MEDLINE | ID: mdl-27832859

BACKGROUND: The obesogenic potential of high-fat diets (HFD) in rodents is attenuated when the protein:carbohydrate ratio is increased. However, it is not known if intake of an HFD irrespective of the protein:carbohydrate ratio and in the absence of weight gain, affects glucose homeostasis and the gut microbiota. METHODS: We fed C57BL6/J mice 3 different HFDs with decreasing protein:carbohydrate ratios for 8weeks and compared the results to a LFD reference group. We analyzed the gut microbiota composition by 16S rDNA amplicon sequencing and the intestinal gene expression by real-time PCR. Whole body glucose homeostasis was evaluated by insulin and glucose tolerance tests as well as by a hyperinsulinemic euglycemic clamp experiment. RESULTS: Compared with LFD-fed reference mice, HFD-fed mice, irrespective of protein:carbohydrate ratio, exhibited impaired glucose tolerance, whereas no differences were observed during insulin tolerance tests. The hyperinsulinemic euglycemic clamp revealed tissue-specific effects on glucose homeostasis in all HFD-fed groups. HFD-fed mice exhibited decreased insulin-stimulated glucose uptake in white but not in brown adipose tissue, and sustained endogenous glucose production under insulin-stimulated conditions. We observed no impairment of insulin-stimulated glucose uptake in skeletal muscles of different fiber type composition. HFD-feeding altered the gut microbiota composition paralleled by increased expression of pro-inflammatory cytokines and genes involved in gluconeogenesis in intestinal epithelial cells of the jejunum. CONCLUSIONS: Intake of a HFD profoundly affected glucose homeostasis, gut inflammatory responses, and gut microbiota composition in the absence of fat mass accretion.


Dietary Fats/pharmacology , Inflammation/chemically induced , Insulin Resistance , Intestines/pathology , Weight Gain , Adipose Tissue, White/metabolism , Animals , Blood Glucose/metabolism , Blood Glucose/physiology , Glucose Intolerance , Homeostasis/drug effects , Intestines/microbiology , Male , Mice , Mice, Inbred C57BL , Microbiota/drug effects , Muscle, Skeletal/metabolism
5.
Biochem Biophys Res Commun ; 463(4): 818-24, 2015 Aug 07.
Article En | MEDLINE | ID: mdl-26086096

RATIONALE: Cardiac metabolism is thought to be altered in insulin resistance and type 2 diabetes (T2D). Our understanding of the regulation of cardiac substrate metabolism and insulin sensitivity has largely been derived from ex vivo preparations which are not subject to the same metabolic regulation as in the intact heart in vivo. Studies are therefore required to examine in vivo cardiac glucose metabolism under physiologically relevant conditions. OBJECTIVE: To determine the temporal pattern of the development of cardiac insulin resistance and to compare with dynamic approaches to interrogate cardiac glucose and intermediary metabolism in vivo. METHODS AND RESULTS: Studies were conducted to determine the evolution of cardiac insulin resistance in C57Bl/6 mice fed a high-fat diet (HFD) for between 1 and 16 weeks. Dynamic in vivo cardiac glucose metabolism was determined following oral administration of [U-(13)C] glucose. Hearts were collected after 15 and 60 min and flux profiling was determined by measuring (13)C mass isotopomers in glycolytic and tricarboxylic acid (TCA) cycle intermediates. Cardiac insulin resistance, determined by euglycemic-hyperinsulinemic clamp, was evident after 3 weeks of HFD. Despite the presence of insulin resistance, in vivo cardiac glucose metabolism following oral glucose administration was not compromised in HFD mice. This contrasts our recent findings in skeletal muscle, where TCA cycle activity was reduced in mice fed a HFD. Similar to our report in muscle, glucose derived pyruvate entry into the TCA cycle in the heart was almost exclusively via pyruvate dehydrogenase, with pyruvate carboxylase mediated anaplerosis being negligible after oral glucose administration. CONCLUSIONS: Under experimental conditions which closely mimic the postprandial state, the insulin resistant mouse heart retains the ability to stimulate glucose metabolism.


Diet, High-Fat , Glucose Clamp Technique , Glucose/metabolism , Hyperinsulinism/metabolism , Metabolomics , Myocardium/metabolism , Animals , Gas Chromatography-Mass Spectrometry , Insulin Resistance , Male , Mice , Mice, Inbred C57BL
6.
Cell Metab ; 21(5): 718-30, 2015 May 05.
Article En | MEDLINE | ID: mdl-25955207

Accumulation of diacylglycerol (DG) in muscle is thought to cause insulin resistance. DG is a precursor for phospholipids, thus phospholipid synthesis could be involved in regulating muscle DG. Little is known about the interaction between phospholipid and DG in muscle; therefore, we examined whether disrupting muscle phospholipid synthesis, specifically phosphatidylethanolamine (PtdEtn), would influence muscle DG content and insulin sensitivity. Muscle PtdEtn synthesis was disrupted by deleting CTP:phosphoethanolamine cytidylyltransferase (ECT), the rate-limiting enzyme in the CDP-ethanolamine pathway, a major route for PtdEtn production. While PtdEtn was reduced in muscle-specific ECT knockout mice, intramyocellular and membrane-associated DG was markedly increased. Importantly, however, this was not associated with insulin resistance. Unexpectedly, mitochondrial biogenesis and muscle oxidative capacity were increased in muscle-specific ECT knockout mice and were accompanied by enhanced exercise performance. These findings highlight the importance of the CDP-ethanolamine pathway in regulating muscle DG content and challenge the DG-induced insulin resistance hypothesis.


Cytidine Diphosphate/analogs & derivatives , Diglycerides/metabolism , Ethanolamines/metabolism , Insulin Resistance , Muscle, Skeletal/metabolism , Organelle Biogenesis , Animals , Cytidine Diphosphate/metabolism , Glucose/metabolism , Lipid Metabolism , Mice , Mice, Knockout , Obesity/genetics , Obesity/metabolism , RNA Nucleotidyltransferases/genetics , RNA Nucleotidyltransferases/metabolism
7.
Front Physiol ; 6: 85, 2015.
Article En | MEDLINE | ID: mdl-25852572

The mitochondrial protein deacetylase sirtuin (SIRT) 3 may mediate exercise training-induced increases in mitochondrial biogenesis and improvements in reactive oxygen species (ROS) handling. We determined the requirement of AMP-activated protein kinase (AMPK) for exercise training-induced increases in skeletal muscle abundance of SIRT3 and other mitochondrial proteins. Exercise training for 6.5 weeks increased SIRT3 (p < 0.01) and superoxide dismutase 2 (MnSOD; p < 0.05) protein abundance in quadriceps muscle of wild-type (WT; n = 13-15), but not AMPK α2 kinase dead (KD; n = 12-13) mice. We also observed a strong trend for increased MnSOD abundance in exercise-trained skeletal muscle of healthy humans (p = 0.051; n = 6). To further elucidate a role for AMPK in mediating these effects, we treated WT (n = 7-8) and AMPK α2 KD (n = 7-9) mice with 5-amino-1-ß-D-ribofuranosyl-imidazole-4-carboxamide (AICAR). Four weeks of daily AICAR injections (500 mg/kg) resulted in AMPK-dependent increases in SIRT3 (p < 0.05) and MnSOD (p < 0.01) in WT, but not AMPK α2 KD mice. We also tested the effect of repeated AICAR treatment on mitochondrial protein levels in mice lacking the transcriptional coactivator peroxisome proliferator-activated receptor γ-coactivator 1α (PGC-1α KO; n = 9-10). Skeletal muscle SIRT3 and MnSOD protein abundance was reduced in sedentary PGC-1α KO mice (p < 0.01) and AICAR-induced increases in SIRT3 and MnSOD protein abundance was only observed in WT mice (p < 0.05). Finally, the acetylation status of SIRT3 target lysine residues on MnSOD (K122) or oligomycin-sensitivity conferring protein (OSCP; K139) was not altered in either mouse or human skeletal muscle in response to acute exercise. We propose an important role for AMPK in regulating mitochondrial function and ROS handling in skeletal muscle in response to exercise training.

8.
Cell Metab ; 21(3): 403-16, 2015 Mar 03.
Article En | MEDLINE | ID: mdl-25738456

Interleukin-6 (IL-6) plays a paradoxical role in inflammation and metabolism. The pro-inflammatory effects of IL-6 are mediated via IL-6 "trans-signaling," a process where the soluble form of the IL-6 receptor (sIL-6R) binds IL-6 and activates signaling in inflammatory cells that express the gp130 but not the IL-6 receptor. Here we show that trans-signaling recruits macrophages into adipose tissue (ATM). Moreover, blocking trans-signaling with soluble gp130Fc protein prevents high-fat diet (HFD)-induced ATM accumulation, but does not improve insulin action. Importantly, however, blockade of IL-6 trans-signaling, unlike complete ablation of IL-6 signaling, does not exacerbate obesity-induced weight gain, liver steatosis, or insulin resistance. Our data identify the sIL-6R as a critical chemotactic signal for ATM recruitment and suggest that selectively blocking IL-6 trans-signaling may be a more favorable treatment option for inflammatory diseases, compared with current treatments that completely block the action of IL-6 and negatively impact upon metabolic homeostasis.


Adipose Tissue/metabolism , Diet, High-Fat/adverse effects , Insulin Resistance/physiology , Interleukin-6/metabolism , Macrophages/metabolism , Macrophages/physiology , Signal Transduction/physiology , Adipose Tissue/physiology , Animals , Cytokine Receptor gp130/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Receptors, Interleukin-6/metabolism
9.
J Physiol ; 591(20): 5207-20, 2013 Oct 15.
Article En | MEDLINE | ID: mdl-23918774

Deacetylases such as sirtuins (SIRTs) convert NAD to nicotinamide (NAM). Nicotinamide phosphoribosyl transferase (Nampt) is the rate-limiting enzyme in the NAD salvage pathway responsible for converting NAM to NAD to maintain cellular redox state. Activation of AMP-activated protein kinase (AMPK) increases SIRT activity by elevating NAD levels. As NAM directly inhibits SIRTs, increased Nampt activation or expression could be a metabolic stress response. Evidence suggests that AMPK regulates Nampt mRNA content, but whether repeated AMPK activation is necessary for increasing Nampt protein levels is unknown. To this end, we assessed whether exercise training- or 5-amino-1-ß-D-ribofuranosyl-imidazole-4-carboxamide (AICAR)-mediated increases in skeletal muscle Nampt abundance are AMPK dependent. One-legged knee-extensor exercise training in humans increased Nampt protein by 16% (P < 0.05) in the trained, but not the untrained leg. Moreover, increases in Nampt mRNA following acute exercise or AICAR treatment (P < 0.05 for both) were maintained in mouse skeletal muscle lacking a functional AMPK α2 subunit. Nampt protein was reduced in skeletal muscle of sedentary AMPK α2 kinase dead (KD), but 6.5 weeks of endurance exercise training increased skeletal muscle Nampt protein to a similar extent in both wild-type (WT) (24%) and AMPK α2 KD (18%) mice. In contrast, 4 weeks of daily AICAR treatment increased Nampt protein in skeletal muscle in WT mice (27%), but this effect did not occur in AMPK α2 KD mice. In conclusion, functional α2-containing AMPK heterotrimers are required for elevation of skeletal muscle Nampt protein, but not mRNA induction. These findings suggest AMPK plays a post-translational role in the regulation of skeletal muscle Nampt protein abundance, and further indicate that the regulation of cellular energy charge and nutrient sensing is mechanistically related.


Muscle, Skeletal/metabolism , Nicotinamide Phosphoribosyltransferase/metabolism , Adenylate Kinase/genetics , Adenylate Kinase/metabolism , Adult , Aminoimidazole Carboxamide/analogs & derivatives , Aminoimidazole Carboxamide/pharmacology , Animals , Exercise , HEK293 Cells , Humans , Male , Mice , Muscle, Skeletal/drug effects , Muscle, Skeletal/physiology , Nicotinamide Phosphoribosyltransferase/genetics , Physical Exertion , Ribonucleotides/pharmacology
10.
Endocrinology ; 154(1): 65-76, 2013 Jan.
Article En | MEDLINE | ID: mdl-23183172

FTY720 is a sphingosine-1-phosphate analog that has been shown to inhibit ceramide synthesis in vitro. Because ceramide accumulation in muscle is associated with insulin resistance, we aimed to examine whether FTY720 would prevent muscle ceramide accumulation in high fat-fed mice and subsequently improve glucose homeostasis. Male C57Bl/6 mice were fed either a chow or high fat-diet (HFD) for 6 wk, after which they were treated with vehicle or FTY720 (5 mg/kg) daily for a further 6 wk. The ceramide content of muscle was examined and insulin action was assessed. Whereas the HFD increased muscle ceramide, this was prevented by FTY720 treatment. This was not associated with alterations in the expression of genes involved in sphingolipid metabolism. Interestingly, the effects of FTY720 on lipid metabolism were not limited to ceramide because FTY720 also prevented the HFD-induced increase in diacylglycerol and triacylglycerol in muscle. Furthermore, the increase in CD36 mRNA expression induced by fat feeding was prevented in muscle of FTY720-treated mice. This was associated with an attenuation of the HFD-induced increase in palmitate uptake and esterification. In addition, FTY720 improved glucose homeostasis as demonstrated by a reduction in plasma insulin, an improvement in whole-body glucose tolerance, an increase in insulin-stimulated glucose uptake, and Akt phosphorylation in muscle. In conclusion, FTY720 exerts beneficial effects on muscle lipid metabolism that prevent lipid accumulation and improve glucose tolerance in high fat-fed mice. Thus, FTY720 and other compounds that target sphingosine-1-phosphate signaling may have therapeutic potential in treating insulin resistance.


Ceramides/metabolism , Dietary Fats/adverse effects , Glucose/metabolism , Muscle, Skeletal/drug effects , Muscle, Skeletal/metabolism , Propylene Glycols/therapeutic use , Sphingosine/analogs & derivatives , Animals , Fingolimod Hydrochloride , Insulin Resistance/physiology , Lipid Metabolism/drug effects , Male , Mice , Mice, Inbred C57BL , Obesity/drug therapy , Obesity/metabolism , Sphingosine/therapeutic use
11.
Diabetes ; 61(12): 3148-55, 2012 Dec.
Article En | MEDLINE | ID: mdl-22961081

The sphingolipids sphingosine-1-phosphate (S1P) and ceramide are important bioactive lipids with many cellular effects. Intracellular ceramide accumulation causes insulin resistance, but sphingosine kinase 1 (SphK1) prevents ceramide accumulation, in part, by promoting its metabolism into S1P. Despite this, the role of SphK1 in regulating insulin action has been largely overlooked. Transgenic (Tg) mice that overexpress SphK1 were fed a standard chow or high-fat diet (HFD) for 6 weeks before undergoing several metabolic analyses. SphK1 Tg mice fed an HFD displayed increased SphK activity in skeletal muscle, which was associated with an attenuated intramuscular ceramide accumulation compared with wild-type (WT) littermates. This was associated with a concomitant reduction in the phosphorylation of c-jun amino-terminal kinase, a serine threonine kinase associated with insulin resistance. Accordingly, skeletal muscle and whole-body insulin sensitivity were improved in SphK1 Tg, compared with WT mice, when fed an HFD. We have identified that the enzyme SphK1 is an important regulator of lipid partitioning and insulin action in skeletal muscle under conditions of increased lipid supply.


Ceramides/metabolism , Diet, High-Fat/adverse effects , Insulin Resistance/physiology , Muscle, Skeletal/enzymology , Muscle, Skeletal/metabolism , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Animals , Blotting, Western , Insulin Resistance/genetics , Mice , Phosphotransferases (Alcohol Group Acceptor)/genetics , Polymerase Chain Reaction
12.
PLoS One ; 7(7): e41456, 2012.
Article En | MEDLINE | ID: mdl-22848500

BACKGROUND: Obesity and type 2 diabetes (T2DM) are associated with increased circulating free fatty acids and triacylglycerols. However, very little is known about specific molecular lipid species associated with these diseases. In order to gain further insight into this, we performed plasma lipidomic analysis in a rodent model of obesity and insulin resistance as well as in lean, obese and obese individuals with T2DM. METHODOLOGY/PRINCIPAL FINDINGS: Lipidomic analysis using liquid chromatography coupled to mass spectrometry revealed marked changes in the plasma of 12 week high fat fed mice. Although a number of triacylglycerol and diacylglycerol species were elevated along with of a number of sphingolipids, a particularly interesting finding was the high fat diet (HFD)-induced reduction in lysophosphatidylcholine (LPC) levels. As liver, skeletal muscle and adipose tissue play an important role in metabolism, we next determined whether the HFD altered LPCs in these tissues. In contrast to our findings in plasma, only very modest changes in tissue LPCs were noted. To determine when the change in plasma LPCs occurred in response to the HFD, mice were studied after 1, 3 and 6 weeks of HFD. The HFD caused rapid alterations in plasma LPCs with most changes occurring within the first week. Consistent with our rodent model, data from our small human cohort showed a reduction in a number of LPC species in obese and obese individuals with T2DM. Interestingly, no differences were found between the obese otherwise healthy individuals and the obese T2DM patients. CONCLUSION: Irrespective of species, our lipidomic profiling revealed a generalized decrease in circulating LPC species in states of obesity. Moreover, our data indicate that diet and adiposity, rather than insulin resistance or diabetes per se, play an important role in altering the plasma LPC profile.


Diabetes Mellitus, Type 2/blood , Lysophosphatidylcholines/blood , Obesity/blood , Adipose Tissue/metabolism , Adiposity , Adult , Animals , Cohort Studies , Dietary Fats/pharmacology , Humans , Insulin Resistance , Liver/metabolism , Male , Mice , Muscle, Skeletal/metabolism , Time Factors
13.
Diabetes ; 60(4): 1100-10, 2011 Apr.
Article En | MEDLINE | ID: mdl-21378177

OBJECTIVE: The fatty acid translocase and scavenger receptor CD36 is important in the recognition and uptake of lipids. Accordingly, we hypothesized that it plays a role in saturated fatty acid-induced macrophage lipid accumulation and proinflammatory activation. RESEARCH DESIGN AND METHODS: In vitro, the effect of CD36 inhibition and deletion in lipid-induced macrophage inflammation was assessed using the putative CD36 inhibitor, sulfosuccinimidyl oleate (SSO), and bone marrow-derived macrophages from mice with (CD36KO) or without (wild-type) global deletion of CD36. To investigate whether deletion of macrophage CD36 would improve insulin sensitivity in vivo, wild-type mice were transplanted with bone marrow from CD36KO or wild-type mice and then fed a standard or high-fat diet (HFD) for 20 weeks. RESULTS: SSO treatment markedly reduced saturated fatty acid-induced lipid accumulation and inflammation in RAW264.7 macrophages. Mice harboring CD36-specific deletion in hematopoietic-derived cells (HSC CD36KO) fed an HFD displayed improved insulin signaling and reduced macrophage infiltration in adipose tissue compared with wild-type mice, but this did not translate into protection against HFD-induced whole-body insulin resistance. Contrary to our hypothesis and our results using SSO in RAW264.7 macrophages, neither saturated fatty acid-induced lipid accumulation nor inflammation was reduced when comparing CD36KO with wild-type bone marrow-derived macrophages. CONCLUSIONS: Although CD36 does not appear important in saturated fatty acid-induced macrophage lipid accumulation, our study uncovers a novel role for CD36 in the migration of proinflammatory phagocytes to adipose tissue in obesity, with a concomitant improvement in insulin action.


Adipose Tissue/metabolism , CD36 Antigens/metabolism , Dietary Fats/adverse effects , Insulin/pharmacology , Macrophages/drug effects , Adipose Tissue/drug effects , Animals , Blotting, Western , Bone Marrow Transplantation , CD36 Antigens/genetics , Cell Line , Cell Movement/drug effects , Cell Movement/genetics , Flow Cytometry , Immunohistochemistry , Macrophages/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Palmitic Acid/adverse effects , Polymerase Chain Reaction , Signal Transduction/drug effects , Signal Transduction/genetics , Stearic Acids/adverse effects
...