Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Nat Commun ; 12(1): 6951, 2021 11 29.
Article in English | MEDLINE | ID: mdl-34845204

ABSTRACT

To improve the power of mediation in high-throughput studies, here we introduce High-throughput mediation analysis (Hitman), which accounts for direction of mediation and applies empirical Bayesian linear modeling. We apply Hitman in a retrospective, exploratory analysis of the SLIMM-T2D clinical trial in which participants with type 2 diabetes were randomized to Roux-en-Y gastric bypass (RYGB) or nonsurgical diabetes/weight management, and fasting plasma proteome and metabolome were assayed up to 3 years. RYGB caused greater improvement in HbA1c, which was mediated by growth hormone receptor (GHR). GHR's mediation is more significant than clinical mediators, including BMI. GHR decreases at 3 months postoperatively alongside increased insulin-like growth factor binding proteins IGFBP1/BP2; plasma GH increased at 1 year. Experimental validation indicates (1) hepatic GHR expression decreases in post-bariatric rats; (2) GHR knockdown in primary hepatocytes decreases gluconeogenic gene expression and glucose production. Thus, RYGB may induce resistance to diabetogenic effects of GH signaling.Trial Registration: Clinicaltrials.gov NCT01073020.


Subject(s)
Diabetes Mellitus, Type 2/blood , Gastric Bypass , Liver/metabolism , Metabolome , Obesity/blood , Proteome , Animals , Biomarkers/blood , Blood Glucose/metabolism , Body Mass Index , Carrier Proteins/blood , Carrier Proteins/genetics , Diabetes Mellitus, Type 2/genetics , Diabetes Mellitus, Type 2/pathology , Diabetes Mellitus, Type 2/surgery , Dipeptidases/blood , Dipeptidases/genetics , Fasting/physiology , Gene Expression Regulation , Glycated Hemoglobin/genetics , Glycated Hemoglobin/metabolism , Hepatocytes/metabolism , Hepatocytes/pathology , Human Growth Hormone/blood , Human Growth Hormone/genetics , Humans , Insulin-Like Growth Factor Binding Protein 1/blood , Insulin-Like Growth Factor Binding Protein 1/genetics , Insulin-Like Growth Factor Binding Protein 2/blood , Insulin-Like Growth Factor Binding Protein 2/genetics , Liver/pathology , Obesity/genetics , Obesity/pathology , Obesity/surgery , Primary Cell Culture , Rats , Retrospective Studies
2.
J Lipid Res ; 62: 100108, 2021.
Article in English | MEDLINE | ID: mdl-34418413

ABSTRACT

Branched fatty acid esters of hydroxy fatty acids (FAHFAs) are endogenous lipids with antidiabetic and anti-inflammatory effects. Each FAHFA family consists of esters with different acyl chains and multiple isomers with branch points at different carbons. Some FAHFAs, including palmitic acid hydroxy stearic acids (PAHSAs), improve insulin sensitivity and glucose tolerance in mice by enhancing glucose-stimulated insulin secretion (GSIS), insulin-stimulated glucose transport, and insulin action to suppress hepatic glucose production and reducing adipose tissue inflammation. However, little is known about the biological effects of other FAHFAs. Here, we investigated whether PAHSAs, oleic acid hydroxy stearic acid, palmitoleic acid hydroxy stearic acid, and stearic acid hydroxy stearic acid potentiate GSIS in ß-cells and human islets, insulin-stimulated glucose uptake in adipocytes, and anti-inflammatory effects in immune cells. We also investigated whether they activate G protein-coupled receptor 40, which mediates the effects of PAHSAs on insulin secretion and sensitivity in vivo. We show that many FAHFAs potentiate GSIS, activate G protein-coupled receptor 40, and attenuate LPS-induced chemokine and cytokine expression and secretion and phagocytosis in immune cells. However, fewer FAHFAs augment insulin-stimulated glucose uptake in adipocytes. S-9-PAHSA, but not R-9-PAHSA, potentiated GSIS and glucose uptake, while both stereoisomers had anti-inflammatory effects. FAHFAs containing unsaturated acyl chains with higher branching from the carboxylate head group are more likely to potentiate GSIS, whereas FAHFAs with lower branching are more likely to be anti-inflammatory. This study provides insight into the specificity of the biological actions of different FAHFAs and could lead to the development of FAHFAs to treat metabolic and immune-mediated diseases.


Subject(s)
Esters/metabolism , Fatty Acids/metabolism , Adult , Esters/chemistry , Fatty Acids/chemistry , Female , Glucose/metabolism , Humans , Insulin Secretion , Male , Middle Aged , Molecular Structure , Stereoisomerism
3.
Proc Natl Acad Sci U S A ; 117(49): 31309-31318, 2020 12 08.
Article in English | MEDLINE | ID: mdl-33214151

ABSTRACT

Adipose tissue (AT) inflammation contributes to systemic insulin resistance. In obesity and type 2 diabetes (T2D), retinol binding protein 4 (RBP4), the major retinol carrier in serum, is elevated in AT and has proinflammatory effects which are mediated partially through Toll-like receptor 4 (TLR4). We now show that RBP4 primes the NLRP3 inflammasome for interleukin-1ß (IL1ß) release, in a glucose-dependent manner, through the TLR4/MD2 receptor complex and TLR2. This impairs insulin signaling in adipocytes. IL1ß is elevated in perigonadal white AT (PGWAT) of chow-fed RBP4-overexpressing mice and in serum and PGWAT of high-fat diet-fed RBP4-overexpressing mice vs. wild-type mice. Holo- or apo-RBP4 injection in wild-type mice causes insulin resistance and elevates PGWAT inflammatory markers, including IL1ß. TLR4 inhibition in RBP4-overexpressing mice reduces PGWAT inflammation, including IL1ß levels and improves insulin sensitivity. Thus, the proinflammatory effects of RBP4 require NLRP3-inflammasome priming. These studies may provide approaches to reduce AT inflammation and insulin resistance in obesity and diabetes.


Subject(s)
Inflammasomes/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Retinol-Binding Proteins, Plasma/metabolism , Signal Transduction , Toll-Like Receptor 2/metabolism , Toll-Like Receptor 4/metabolism , 3T3-L1 Cells , Adipocytes/metabolism , Adipose Tissue, White/pathology , Animals , Glucose/pharmacology , Glycolysis/drug effects , Humans , Inflammation/pathology , Insulin Resistance , Interleukin-1beta/metabolism , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Models, Biological , Tumor Necrosis Factor-alpha/metabolism
4.
Cell Metab ; 32(4): 665-675.e6, 2020 10 06.
Article in English | MEDLINE | ID: mdl-32941798

ABSTRACT

The thiazolidinediones (TZDs) are ligands of PPARγ that improve insulin sensitivity, but their use is limited by significant side effects. Recently, we demonstrated a mechanism wherein TZDs improve insulin sensitivity distinct from receptor agonism and adipogenesis: reversal of obesity-linked phosphorylation of PPARγ at serine 273. However, the role of this modification hasn't been tested genetically. Here we demonstrate that mice encoding an allele of PPARγ that cannot be phosphorylated at S273 are protected from insulin resistance, without exhibiting differences in body weight or TZD-associated side effects. Indeed, hyperinsulinemic-euglycemic clamp experiments confirm insulin sensitivity. RNA-seq in these mice reveals reduced expression of Gdf3, a BMP family member. Ectopic expression of Gdf3 is sufficient to induce insulin resistance in lean, healthy mice. We find Gdf3 inhibits BMP signaling and insulin signaling in vitro. Together, these results highlight the diabetogenic role of PPARγ S273 phosphorylation and focus attention on a putative target, Gdf3.


Subject(s)
Growth Differentiation Factor 3/metabolism , Obesity/drug therapy , PPAR gamma/metabolism , Thiazolidinediones/pharmacology , Alleles , Animals , Cells, Cultured , Growth Differentiation Factor 3/genetics , Humans , Insulin Resistance , Male , Mice , Mice, Inbred C57BL , Obesity/metabolism , PPAR gamma/genetics , Phosphorylation/drug effects
5.
FASEB J ; 34(5): 6099-6110, 2020 05.
Article in English | MEDLINE | ID: mdl-32167208

ABSTRACT

Retinol-binding protein-4 (RBP4) is elevated in serum and adipose tissue (AT) in obesity-induced insulin resistance and correlates inversely with insulin-stimulated glucose disposal. But its role in insulin-mediated suppression of lipolysis, free fatty acids (FFA), and endogenous glucose production (EGP) in humans is unknown. RBP4 mRNA or protein levels were higher in liver, subcutaneous adipose tissue (SAT), and visceral adipose tissue (VAT) in morbidly obese subjects undergoing Roux-en-Y gastric bypass surgery compared to lean controls undergoing elective laparoscopic cholecystectomy. RBP4 mRNA expression in SAT correlated with the expression of several macrophage and other inflammation markers. Serum RBP4 levels correlated inversely with glucose disposal and insulin-mediated suppression of lipolysis, FFA, and EGP. Mechanistically, RBP4 treatment of human adipocytes in vitro directly stimulated basal lipolysis. Treatment of adipocytes with conditioned media from RBP4-activated macrophages markedly increased basal lipolysis and impaired insulin-mediated lipolysis suppression. RBP4 treatment of macrophages increased TNFα production. These data suggest that elevated serum or adipose tissue RBP4 levels in morbidly obese subjects may cause hepatic and systemic insulin resistance by stimulating basal lipolysis and by activating macrophages in adipose tissue, resulting in release of pro-inflammatory cytokines that impair lipolysis suppression. While we have demonstrated this mechanism in human adipocytes in vitro, and correlations from our flux studies in humans strongly support this, further studies are needed to determine whether this mechanism explains RBP4-induced insulin resistance in humans.


Subject(s)
Adipose Tissue/pathology , Glucose Intolerance/pathology , Insulin Resistance , Lipolysis , Liver/pathology , Obesity, Morbid/complications , Retinol-Binding Proteins, Plasma/metabolism , Adipose Tissue/metabolism , Adult , Blood Glucose/analysis , Female , Glucose Intolerance/etiology , Glucose Intolerance/metabolism , Humans , Liver/metabolism , Middle Aged , Retinol-Binding Proteins, Plasma/genetics
6.
Cell Metab ; 27(2): 419-427.e4, 2018 02 06.
Article in English | MEDLINE | ID: mdl-29414687

ABSTRACT

Palmitic acid hydroxystearic acids (PAHSAs) are endogenous lipids with anti-diabetic and anti-inflammatory effects. PAHSA levels are reduced in serum and adipose tissue of insulin-resistant people and high-fat diet (HFD)-fed mice. Here, we investigated whether chronic PAHSA treatment enhances insulin sensitivity and which receptors mediate PAHSA effects. Chronic PAHSA administration in chow- and HFD-fed mice raises serum and tissue PAHSA levels ∼1.4- to 3-fold. This improves insulin sensitivity and glucose tolerance without altering body weight. PAHSA administration in chow-fed, but not HFD-fed, mice augments insulin and glucagon-like peptide (GLP-1) secretion. PAHSAs are selective agonists for GPR40, increasing Ca+2 flux, but not intracellular cyclic AMP. Blocking GPR40 reverses improvements in glucose tolerance and insulin sensitivity in PAHSA-treated chow- and HFD-fed mice and directly inhibits PAHSA augmentation of glucose-stimulated insulin secretion in human islets. In contrast, GLP-1 receptor blockade in PAHSA-treated chow-fed mice reduces PAHSA effects on glucose tolerance, but not on insulin sensitivity. Thus, PAHSAs activate GPR40, which is involved in their beneficial metabolic effects.


Subject(s)
Glucose/metabolism , Homeostasis , Palmitic Acid/pharmacology , Receptors, G-Protein-Coupled/metabolism , Stearic Acids/pharmacology , Adiposity/drug effects , Animals , Eating/drug effects , HEK293 Cells , Homeostasis/drug effects , Humans , Inflammation/pathology , Insulin Resistance , Mice, Inbred C57BL
7.
Cell Rep ; 21(4): 1021-1035, 2017 Oct 24.
Article in English | MEDLINE | ID: mdl-29069585

ABSTRACT

Lower adipose-ChREBP and de novo lipogenesis (DNL) are associated with insulin resistance in humans. Here, we generated adipose-specific ChREBP knockout (AdChREBP KO) mice with negligible sucrose-induced DNL in adipose tissue (AT). Chow-fed AdChREBP KO mice are insulin resistant with impaired insulin action in the liver, muscle, and AT and increased AT inflammation. HFD-fed AdChREBP KO mice are also more insulin resistant than controls. Surprisingly, adipocytes lacking ChREBP display a cell-autonomous reduction in insulin-stimulated glucose transport that is mediated by impaired Glut4 translocation and exocytosis, not lower Glut4 levels. AdChREBP KO mice have lower levels of palmitic acid esters of hydroxy stearic acids (PAHSAs) in serum, and AT. 9-PAHSA supplementation completely rescues their insulin resistance and AT inflammation. 9-PAHSA also normalizes impaired glucose transport and Glut4 exocytosis in ChREBP KO adipocytes. Thus, loss of adipose-ChREBP is sufficient to cause insulin resistance, potentially by regulating AT glucose transport and flux through specific lipogenic pathways.


Subject(s)
Adipocytes/metabolism , Glucose/metabolism , Insulin Resistance , Nuclear Proteins/metabolism , Transcription Factors/metabolism , 3T3 Cells , Animals , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors , Cells, Cultured , Glucose Transporter Type 4/genetics , Glucose Transporter Type 4/metabolism , Liver/metabolism , Male , Mice , Mice, Inbred C57BL , Muscle, Skeletal/metabolism , Nuclear Proteins/genetics , Palmitic Acids/blood , Stearic Acids/blood , Transcription Factors/genetics
8.
J Biol Chem ; 291(42): 22207-22217, 2016 Oct 14.
Article in English | MEDLINE | ID: mdl-27573241

ABSTRACT

We recently discovered a structurally novel class of endogenous lipids, branched palmitic acid esters of hydroxy stearic acids (PAHSAs), with beneficial metabolic and anti-inflammatory effects. We tested whether PAHSAs protect against colitis, which is a chronic inflammatory disease driven predominantly by defects in the innate mucosal barrier and adaptive immune system. There is an unmet clinical need for safe and well tolerated oral therapeutics with direct anti-inflammatory effects. Wild-type mice were pretreated orally with vehicle or 5-PAHSA (10 mg/kg) and 9-PAHSA (5 mg/kg) once daily for 3 days, followed by 10 days of either 0% or 2% dextran sulfate sodium water with continued vehicle or PAHSA treatment. The colon was collected for histopathology, gene expression, and flow cytometry. Intestinal crypt fractions were prepared for ex vivo bactericidal assays. Bone marrow-derived dendritic cells pretreated with vehicle or PAHSA and splenic CD4+ T cells from syngeneic mice were co-cultured to assess antigen presentation and T cell activation in response to LPS. PAHSA treatment prevented weight loss, improved colitis scores (stool consistency, hematochezia, and mouse appearance), and augmented intestinal crypt Paneth cell bactericidal potency via a mechanism that may involve GPR120. In vitro, PAHSAs attenuated dendritic cell activation and subsequent T cell proliferation and Th1 polarization. The anti-inflammatory effects of PAHSAs in vivo resulted in reduced colonic T cell activation and pro-inflammatory cytokine and chemokine expression. These anti-inflammatory effects appear to be partially GPR120-dependent. We conclude that PAHSA treatment regulates innate and adaptive immune responses to prevent mucosal damage and protect against colitis. Thus, PAHSAs may be a novel treatment for colitis and related inflammation-driven diseases.


Subject(s)
Adaptive Immunity/immunology , Colitis/drug therapy , Fatty Acids/pharmacology , Immunity, Innate/drug effects , Immunity, Mucosal/drug effects , Paneth Cells/immunology , Th1 Cells/immunology , Animals , Colitis/chemically induced , Colitis/immunology , Colitis/pathology , Dextran Sulfate/adverse effects , Dextran Sulfate/pharmacology , Male , Mice , Paneth Cells/pathology , Receptors, G-Protein-Coupled/immunology , Th1 Cells/pathology
9.
Neuron ; 91(1): 25-33, 2016 07 06.
Article in English | MEDLINE | ID: mdl-27321921

ABSTRACT

AMP-activated protein kinase (AMPK) plays an important role in regulating food intake. The downstream AMPK substrates and neurobiological mechanisms responsible for this, however, are ill defined. Agouti-related peptide (AgRP)-expressing neurons in the arcuate nucleus regulate hunger. Their firing increases with fasting, and once engaged they cause feeding. AgRP neuron activity is regulated by state-dependent synaptic plasticity: fasting increases dendritic spines and excitatory synaptic activity; feeding does the opposite. The signaling mechanisms underlying this, however, are also unknown. Using neuron-specific approaches to measure and manipulate kinase activity specifically within AgRP neurons, we establish that fasting increases AMPK activity in AgRP neurons, that increased AMPK activity in AgRP neurons is both necessary and sufficient for fasting-induced spinogenesis and excitatory synaptic activity, and that the AMPK phosphorylation target mediating this plasticity is p21-activated kinase. This provides a signaling and neurobiological basis for both AMPK regulation of energy balance and AgRP neuron state-dependent plasticity.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Fasting , Neuronal Plasticity/physiology , Neurons/physiology , Signal Transduction , p21-Activated Kinases/metabolism , Animals , Dendritic Spines/metabolism , Eating/drug effects , Energy Metabolism/physiology , Mice, Transgenic , Neuropeptide Y/metabolism
10.
Diabetes ; 65(5): 1317-27, 2016 05.
Article in English | MEDLINE | ID: mdl-26936962

ABSTRACT

Adipose tissue (AT) inflammation contributes to impaired insulin action, which is a major cause of type 2 diabetes. RBP4 is an adipocyte- and liver-derived protein with an important role in insulin resistance, metabolic syndrome, and AT inflammation. RBP4 elevation causes AT inflammation by activating innate immunity, which elicits an adaptive immune response. RBP4-overexpressing mice (RBP4-Ox) are insulin resistant and glucose intolerant and have increased AT macrophages and T-helper 1 cells. We show that high-fat diet-fed RBP4(-/-) mice have reduced AT inflammation and improved insulin sensitivity versus wild type. We also elucidate the mechanism for RBP4-induced macrophage antigen presentation and subsequent T-cell activation. In RBP4-Ox, AT macrophages display enhanced c-Jun N-terminal kinase, extracellular signal-related kinase, and p38 phosphorylation. Inhibition of these pathways and of NF-κB reduces activation of macrophages and CD4 T cells. MyD88 is an adaptor protein involved in proinflammatory signaling. In macrophages from MyD88(-/-) mice, RBP4 fails to stimulate secretion of tumor necrosis factor, IL-12, and IL-6 and CD4 T-cell activation. In vivo blockade of antigen presentation by treating RBP4-Ox mice with CTLA4-Ig, which blocks costimulation of T cells, is sufficient to reduce AT inflammation and improve insulin resistance. Thus, MyD88 and downstream mitogen-activated protein kinase and NF-κB pathways are necessary for RBP4-induced macrophage antigen presentation and subsequent T-cell activation. Also, blocking antigen presentation with CTLA4-Ig improves RBP4-induced insulin resistance and macrophage-induced T-cell activation.


Subject(s)
Adaptive Immunity , Antigen Presentation , CD4-Positive T-Lymphocytes/immunology , Insulin Resistance , Lymphocyte Activation , Obesity/immunology , Retinol-Binding Proteins, Plasma/metabolism , Adipose Tissue/immunology , Adipose Tissue/metabolism , Adipose Tissue/pathology , Animals , Antigen-Presenting Cells/immunology , Antigen-Presenting Cells/metabolism , Antigen-Presenting Cells/pathology , Bone Marrow Cells/immunology , Bone Marrow Cells/metabolism , Bone Marrow Cells/pathology , CD4-Positive T-Lymphocytes/metabolism , CD4-Positive T-Lymphocytes/pathology , Cells, Cultured , Coculture Techniques , Diet, High-Fat/adverse effects , Glucose Intolerance/etiology , Glucose Intolerance/immunology , Glucose Intolerance/metabolism , Glucose Intolerance/pathology , Heterozygote , Macrophages/immunology , Macrophages/metabolism , Macrophages/pathology , Male , Mice, Inbred C57BL , Mice, Knockout , Myeloid Differentiation Factor 88/genetics , Myeloid Differentiation Factor 88/metabolism , Obesity/etiology , Obesity/metabolism , Obesity/pathology , Retinol-Binding Proteins, Plasma/genetics , Spleen/immunology , Spleen/metabolism , Spleen/pathology
11.
Cell Metab ; 19(3): 512-26, 2014 Mar 04.
Article in English | MEDLINE | ID: mdl-24606904

ABSTRACT

Insulin resistance is a major cause of diabetes and is highly associated with adipose tissue (AT) inflammation in obesity. RBP4, a retinol transporter, is elevated in insulin resistance and contributes to increased diabetes risk. We aimed to determine the mechanisms for RBP4-induced insulin resistance. Here we show that RBP4 elevation causes AT inflammation by activating innate immunity that elicits an adaptive immune response. RBP4-overexpressing mice (RBP4-Ox) are insulin resistant and glucose intolerant and have increased AT macrophage and CD4 T cell infiltration. In RBP4-Ox, AT CD206(+) macrophages express proinflammatory markers and activate CD4 T cells while maintaining alternatively activated macrophage markers. These effects result from direct activation of AT antigen-presenting cells (APCs) by RBP4 through a JNK-dependent pathway. Transfer of RBP4-activated APCs into normal mice is sufficient to induce AT inflammation, insulin resistance, and glucose intolerance. Thus, RBP4 causes insulin resistance, at least partly, by activating AT APCs that induce CD4 T cell Th1 polarization and AT inflammation.


Subject(s)
Adipose Tissue/metabolism , Antigen-Presenting Cells/cytology , Retinol-Binding Proteins, Plasma/metabolism , Adipose Tissue/cytology , Animals , Antigen-Presenting Cells/metabolism , Bone Marrow Cells/cytology , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Cell Proliferation , Glucose Intolerance , Humans , Insulin Resistance , JNK Mitogen-Activated Protein Kinases/metabolism , Lectins, C-Type/metabolism , Liver/cytology , Macrophages/cytology , Macrophages/drug effects , Macrophages/metabolism , Male , Mannose Receptor , Mannose-Binding Lectins/metabolism , Mice , Mice, Inbred C57BL , Receptors, Cell Surface/metabolism , Recombinant Proteins/biosynthesis , Recombinant Proteins/genetics , Recombinant Proteins/pharmacology , Retinol-Binding Proteins, Plasma/genetics , Retinol-Binding Proteins, Plasma/pharmacology , Signal Transduction , Th1 Cells/cytology , Th1 Cells/metabolism , Transcriptome
12.
J Biol Chem ; 288(2): 1250-65, 2013 Jan 11.
Article in English | MEDLINE | ID: mdl-23105095

ABSTRACT

Vitamin A (retinol) is absorbed in the small intestine, stored in liver, and secreted into circulation bound to serum retinol-binding protein (RBP4). Circulating retinol may be taken up by extrahepatic tissues or recycled back to liver multiple times before it is finally metabolized or degraded. Liver exhibits high affinity binding sites for RBP4, but specific receptors have not been identified. The only known high affinity receptor for RBP4, Stra6, is not expressed in the liver. Here we report discovery of RBP4 receptor-2 (RBPR2), a novel retinol transporter expressed primarily in liver and intestine and induced in adipose tissue of obese mice. RBPR2 is structurally related to Stra6 and highly conserved in vertebrates, including humans. Expression of RBPR2 in cultured cells confers high affinity RBP4 binding and retinol transport, and RBPR2 knockdown reduces RBP4 binding/retinol transport. RBPR2 expression is suppressed by retinol and retinoic acid and correlates inversely with liver retinol stores in vivo. We conclude that RBPR2 is a novel retinol transporter that potentially regulates retinol homeostasis in liver and other tissues. In addition, expression of RBPR2 in liver and fat suggests a possible role in mediating established metabolic actions of RBP4 in those tissues.


Subject(s)
Carrier Proteins/metabolism , Liver/metabolism , Retinol-Binding Proteins, Plasma/metabolism , Amino Acid Sequence , Animals , Base Sequence , Cell Line , Cloning, Molecular , DNA Primers , Humans , Membrane Proteins/genetics , Mice , Mice, Inbred C57BL , Molecular Sequence Data , Polymerase Chain Reaction , Promoter Regions, Genetic , Sequence Homology, Amino Acid , Transcriptome
13.
Mol Cell Biol ; 32(10): 2010-9, 2012 May.
Article in English | MEDLINE | ID: mdl-22431523

ABSTRACT

Retinol-binding protein 4 (RBP4), the sole retinol transporter in blood, is secreted from adipocytes and liver. Serum RBP4 levels correlate highly with insulin resistance, other metabolic syndrome factors, and cardiovascular disease. Elevated serum RBP4 causes insulin resistance, but the molecular mechanisms are unknown. Here we show that RBP4 induces expression of proinflammatory cytokines in mouse and human macrophages and thereby indirectly inhibits insulin signaling in cocultured adipocytes. This occurs through activation of c-Jun N-terminal protein kinase (JNK) and Toll-like receptor 4 (TLR4) pathways independent of the RBP4 receptor, STRA6. RBP4 effects are markedly attenuated in JNK1-/- JNK2-/- macrophages and TLR4-/- macrophages. Because RBP4 is a retinol-binding protein, we investigated whether these effects are retinol dependent. Unexpectedly, retinol-free RBP4 (apo-RBP4) is as potent as retinol-bound RBP4 (holo-RBP4) in inducing proinflammatory cytokines in macrophages. Apo-RBP4 is likely to be physiologically significant since RBP4/retinol ratios are increased in serum of lean and obese insulin-resistant humans compared to ratios in insulin-sensitive humans, indicating that higher apo-RBP4 is associated with insulin resistance independent of obesity. Thus, RBP4 may cause insulin resistance by contributing to the development of an inflammatory state in adipose tissue through activation of proinflammatory cytokines in macrophages. This process reveals a novel JNK- and TLR4-dependent and retinol- and STRA6-independent mechanism of action for RBP4.


Subject(s)
Adipocytes/metabolism , Macrophages/metabolism , Retinol-Binding Proteins, Plasma/metabolism , 3T3 Cells , Animals , Cell Communication , Coculture Techniques , Cytokines/metabolism , Humans , Insulin/metabolism , Insulin Resistance , JNK Mitogen-Activated Protein Kinases/metabolism , Macrophage Activation , Membrane Proteins/metabolism , Mice , Signal Transduction , Toll-Like Receptor 4/metabolism , Vitamin A/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...