Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 9 de 9
1.
Cell ; 172(4): 731-743.e12, 2018 02 08.
Article En | MEDLINE | ID: mdl-29425491

The noncanonical IKK family member TANK-binding kinase 1 (TBK1) is activated by pro-inflammatory cytokines, but its role in controlling metabolism remains unclear. Here, we report that the kinase uniquely controls energy metabolism. Tbk1 expression is increased in adipocytes of HFD-fed mice. Adipocyte-specific TBK1 knockout (ATKO) attenuates HFD-induced obesity by increasing energy expenditure; further studies show that TBK1 directly inhibits AMPK to repress respiration and increase energy storage. Conversely, activation of AMPK under catabolic conditions can increase TBK1 activity through phosphorylation, mediated by AMPK's downstream target ULK1. Surprisingly, ATKO also exaggerates adipose tissue inflammation and insulin resistance. TBK1 suppresses inflammation by phosphorylating and inducing the degradation of the IKK kinase NIK, thus attenuating NF-κB activity. Moreover, TBK1 mediates the negative impact of AMPK activity on NF-κB activation. These data implicate a unique role for TBK1 in mediating bidirectional crosstalk between energy sensing and inflammatory signaling pathways in both over- and undernutrition.


Adipocytes/metabolism , Adipose Tissue/metabolism , Energy Metabolism , Protein Serine-Threonine Kinases/metabolism , Signal Transduction , AMP-Activated Protein Kinases/genetics , AMP-Activated Protein Kinases/metabolism , Adipocytes/pathology , Adipose Tissue/pathology , Animals , Autophagy-Related Protein-1 Homolog/genetics , Autophagy-Related Protein-1 Homolog/metabolism , Cell Line, Transformed , Dietary Fats/adverse effects , Dietary Fats/pharmacology , Inflammation/chemically induced , Inflammation/genetics , Inflammation/metabolism , Inflammation/pathology , Mice , Mice, Knockout , NF-kappa B/genetics , NF-kappa B/metabolism , Oxygen Consumption/drug effects , Phosphorylation/drug effects , Phosphorylation/genetics , Protein Serine-Threonine Kinases/genetics , NF-kappaB-Inducing Kinase
2.
Sci Signal ; 10(471)2017 Mar 21.
Article En | MEDLINE | ID: mdl-28325821

Insulin stimulates glucose uptake through the translocation of the glucose transporter GLUT4 to the plasma membrane. The exocyst complex tethers GLUT4-containing vesicles to the plasma membrane, a process that requires the binding of the G protein (heterotrimeric guanine nucleotide-binding protein) RalA to the exocyst complex. We report that upon activation of RalA, the protein kinase TBK1 phosphorylated the exocyst subunit Exo84. Knockdown of TBK1 blocked insulin-stimulated glucose uptake and GLUT4 translocation; knockout of TBK1 in adipocytes blocked insulin-stimulated glucose uptake; and ectopic overexpression of a kinase-inactive mutant of TBK1 reduced insulin-stimulated glucose uptake in 3T3-L1 adipocytes. The phosphorylation of Exo84 by TBK1 reduced its affinity for RalA and enabled its release from the exocyst. Overexpression of a kinase-inactive mutant of TBK1 blocked the dissociation of the TBK1/RalA/exocyst complex, and treatment of 3T3-L1 adipocytes with specific inhibitors of TBK1 reduced the rate of complex dissociation. Introduction of phosphorylation-mimicking or nonphosphorylatable mutant forms of Exo84 blocked insulin-stimulated GLUT4 translocation. Thus, these data indicate that TBK1 controls GLUT4 vesicle engagement and disengagement from the exocyst, suggesting that exocyst components not only constitute a tethering complex for the GLUT4 vesicle but also act as "gatekeepers" controlling vesicle fusion at the plasma membrane.


Adipocytes/drug effects , Glucose Transporter Type 4/metabolism , Insulin/pharmacology , Protein Serine-Threonine Kinases/metabolism , Vesicular Transport Proteins/metabolism , 3T3-L1 Cells , Adipocytes/metabolism , Animals , Glucose Transporter Type 4/genetics , Hypoglycemic Agents/pharmacology , Immunoblotting , Mice , Mutation , Phosphorylation , Protein Binding , Protein Serine-Threonine Kinases/genetics , Protein Transport/drug effects , RNA Interference , Vesicular Transport Proteins/genetics , ral GTP-Binding Proteins/genetics , ral GTP-Binding Proteins/metabolism
3.
Nat Commun ; 6: 6047, 2015 Jan 12.
Article En | MEDLINE | ID: mdl-25581158

The search for effective treatments for obesity and its comorbidities is of prime importance. We previously identified IKK-ε and TBK1 as promising therapeutic targets for the treatment of obesity and associated insulin resistance. Here we show that acute inhibition of IKK-ε and TBK1 with amlexanox treatment increases cAMP levels in subcutaneous adipose depots of obese mice, promoting the synthesis and secretion of the cytokine IL-6 from adipocytes and preadipocytes, but not from macrophages. IL-6, in turn, stimulates the phosphorylation of hepatic Stat3 to suppress expression of genes involved in gluconeogenesis, in the process improving glucose handling in obese mice. Preliminary data in a small cohort of obese patients show a similar association. These data support an important role for a subcutaneous adipose tissue-liver axis in mediating the acute metabolic benefits of amlexanox on glucose metabolism, and point to a new therapeutic pathway for type 2 diabetes.


Gluconeogenesis , Liver/metabolism , Signal Transduction , Subcutaneous Fat/metabolism , 3T3-L1 Cells , Adipocytes/drug effects , Adipocytes/metabolism , Adult , Aged , Aminopyridines/pharmacology , Animals , Cyclic AMP/metabolism , Female , Gene Knockdown Techniques , Gluconeogenesis/drug effects , Glucose-6-Phosphatase/metabolism , Humans , Inflammation/pathology , Insulin Resistance , Interleukin-6/metabolism , Liver/drug effects , Male , Mice , Mice, Inbred C57BL , Middle Aged , Receptors, Adrenergic, beta/metabolism , STAT3 Transcription Factor/metabolism , Signal Transduction/drug effects , Subcutaneous Fat/drug effects , Young Adult , p38 Mitogen-Activated Protein Kinases/metabolism
4.
Immunity ; 42(1): 15-7, 2015 Jan 20.
Article En | MEDLINE | ID: mdl-25607455

The biogenesis of beige fat is poorly understood. In recent issues of Nature and Cell, Brestoff et al. (2014) and Lee et al. (2015) demonstrate that resident innate lymphoid cells in subcutaneous fat generate and activate beige adipocytes, producing thermogenesis.


Adipose Tissue, Brown/metabolism , Adipose Tissue, White/cytology , Adipose Tissue, White/immunology , Immunity, Innate/immunology , Lymphocytes/metabolism , Lymphocytes/physiology , Obesity/immunology , Animals , Female , Humans , Male
5.
Mol Biol Cell ; 25(19): 3059-69, 2014 Oct 01.
Article En | MEDLINE | ID: mdl-25103239

Insulin-stimulated glucose uptake in fat and muscle is mediated by the major facilitative glucose transporter Glut4. Insulin controls the trafficking of Glut4 to the plasma membrane via regulation of a series of small G proteins, including RalA and Rab10. We demonstrate here that Rab10 is a bona fide target of the GTPase-activating protein AS160, which is inhibited after phosphorylation by the protein kinase Akt. Once activated, Rab10 can increase the GTP binding of RalA by recruiting the Ral guanyl nucleotide exchange factor, Rlf/Rgl2. Rab10 and RalA reside in the same pool of Glut4-storage vesicles in untreated cells, and, together with Rlf, they ensure maximal glucose transport. Overexpression of membrane-tethered Rlf compensates for the loss of Rab10 in Glut4 translocation, suggesting that Rab10 recruits Rlf to membrane compartments for RalA activation and that RalA is downstream of Rab10. Together these studies identify a new G protein cascade in the regulation of insulin-stimulated Glut4 trafficking and glucose uptake.


Glucose Transporter Type 4/metabolism , Insulin/metabolism , Transcription Factors/metabolism , rab GTP-Binding Proteins/metabolism , ral GTP-Binding Proteins/metabolism , 3T3 Cells , Adipocytes/metabolism , Animals , Biological Transport , COS Cells , Cell Line , Chlorocebus aethiops , Enzyme Activation , GTPase-Activating Proteins/antagonists & inhibitors , GTPase-Activating Proteins/genetics , GTPase-Activating Proteins/metabolism , Glucose/metabolism , Guanine Nucleotide Exchange Factors , Guanosine Triphosphate/metabolism , HEK293 Cells , Humans , Mice , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , RNA Interference , RNA, Small Interfering , Transcription Factors/biosynthesis , Transcription Factors/genetics , rab GTP-Binding Proteins/agonists
6.
Diabetes ; 63(4): 1340-52, 2014 Apr.
Article En | MEDLINE | ID: mdl-24379350

Chronic low-grade inflammation is emerging as a pathogenic link between obesity and metabolic disease. Persistent immune activation in white adipose tissue (WAT) impairs insulin sensitivity and systemic metabolism, in part, through the actions of proinflammatory cytokines. Whether obesity engages an adaptive mechanism to counteract chronic inflammation in adipose tissues has not been elucidated. Here we identified otopetrin 1 (Otop1) as a component of a counterinflammatory pathway that is induced in WAT during obesity. Otop1 expression is markedly increased in obese mouse WAT and is stimulated by tumor necrosis factor-α in cultured adipocytes. Otop1 mutant mice respond to high-fat diet with pronounced insulin resistance and hepatic steatosis, accompanied by augmented adipose tissue inflammation. Otop1 attenuates interferon-γ (IFN-γ) signaling in adipocytes through selective downregulation of the transcription factor STAT1. Using a tagged vector, we found that Otop1 physically interacts with endogenous STAT1. Thus, Otop1 defines a unique target of cytokine signaling that attenuates obesity-induced adipose tissue inflammation and plays an adaptive role in maintaining metabolic homeostasis in obesity.


Adipose Tissue/pathology , Inflammation/prevention & control , Membrane Proteins/pharmacology , Obesity/metabolism , Adipose Tissue, White/metabolism , Animals , Diet, High-Fat , Homeostasis/drug effects , Inflammation/immunology , Insulin Resistance/physiology , Interferon-gamma/drug effects , Male , Membrane Proteins/metabolism , Mice , STAT1 Transcription Factor/metabolism
7.
Elife ; 2: e01119, 2013 Dec 24.
Article En | MEDLINE | ID: mdl-24368730

Obesity produces a chronic inflammatory state involving the NFκB pathway, resulting in persistent elevation of the noncanonical IκB kinases IKKε and TBK1. In this study, we report that these kinases attenuate ß-adrenergic signaling in white adipose tissue. Treatment of 3T3-L1 adipocytes with specific inhibitors of these kinases restored ß-adrenergic signaling and lipolysis attenuated by TNFα and Poly (I:C). Conversely, overexpression of the kinases reduced induction of Ucp1, lipolysis, cAMP levels, and phosphorylation of hormone sensitive lipase in response to isoproterenol or forskolin. Noncanonical IKKs reduce catecholamine sensitivity by phosphorylating and activating the major adipocyte phosphodiesterase PDE3B. In vivo inhibition of these kinases by treatment of obese mice with the drug amlexanox reversed obesity-induced catecholamine resistance, and restored PKA signaling in response to injection of a ß-3 adrenergic agonist. These studies suggest that by reducing production of cAMP in adipocytes, IKKε and TBK1 may contribute to the repression of energy expenditure during obesity. DOI: http://dx.doi.org/10.7554/eLife.01119.001.


Adipocytes/enzymology , Adipose Tissue, White/enzymology , Catecholamines/metabolism , Cyclic Nucleotide Phosphodiesterases, Type 3/metabolism , I-kappa B Kinase/metabolism , Inflammation/enzymology , Obesity/enzymology , Protein Serine-Threonine Kinases/metabolism , 3T3-L1 Cells , Adipocytes/drug effects , Adipose Tissue, White/drug effects , Adrenergic beta-3 Receptor Agonists/pharmacology , Aminopyridines/pharmacology , Animals , COS Cells , Chlorocebus aethiops , Cyclic AMP/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Cyclic Nucleotide Phosphodiesterases, Type 3/genetics , Dioxoles/pharmacology , Disease Models, Animal , Energy Metabolism , Enzyme Activation , HEK293 Cells , Humans , I-kappa B Kinase/antagonists & inhibitors , I-kappa B Kinase/genetics , Inflammation/genetics , Ion Channels/metabolism , Lipolysis , Male , Mice , Mice, Inbred C57BL , Mitochondrial Proteins/metabolism , Obesity/genetics , Phosphorylation , Poly I-C/pharmacology , Protein Kinase Inhibitors/pharmacology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/genetics , Receptors, Adrenergic, beta/metabolism , Signal Transduction , Sterol Esterase/metabolism , Time Factors , Transfection , Tumor Necrosis Factor-alpha/pharmacology , Uncoupling Protein 1
8.
J Biol Chem ; 288(13): 9272-83, 2013 Mar 29.
Article En | MEDLINE | ID: mdl-23386617

RGC1 and RGC2 comprise a functional RalGAP complex (RGC) that suppresses RalA activity. The PI3-kinase/Akt signaling pathway activates RalA through phosphorylation-mediated inhibition of the RGC. Here we identify a novel phosphorylation-dependent interaction between 14-3-3 and the RGC. 14-3-3 binds to the complex through an Akt-phosphorylated residue, threonine 715, on RGC2. Interaction with 14-3-3 does not alter in vitro activity of the GTPase-activating protein complex. However, blocking the interaction between 14-3-3 and RGC2 in cells increases suppression of RalA activity by the RGC, suggesting that 14-3-3 inhibits the complex through a non-catalytic mechanism. Together, these data show that 14-3-3 negatively regulates the RGC downstream of the PI3-kinase/Akt signaling pathway.


14-3-3 Proteins/metabolism , GTPase-Activating Proteins/metabolism , Gene Expression Regulation , Nerve Tissue Proteins/metabolism , ral GTP-Binding Proteins/metabolism , 3T3 Cells , Adipocytes/cytology , Amino Acid Motifs , Androstadienes/pharmacology , Animals , DNA/metabolism , Enzyme Inhibitors/pharmacology , GTP-Binding Proteins/metabolism , HEK293 Cells , Humans , Mice , Phosphatidylinositol 3-Kinases/metabolism , Phosphoric Monoester Hydrolases/metabolism , Phosphorylation , Protein Binding , Signal Transduction , Wortmannin
9.
Nat Med ; 19(3): 313-21, 2013 Mar.
Article En | MEDLINE | ID: mdl-23396211

Emerging evidence suggests that inflammation provides a link between obesity and insulin resistance. The noncanonical IκB kinases IKK-ɛ and TANK-binding kinase 1 (TBK1) are induced in liver and fat by NF-κB activation upon high-fat diet feeding and in turn initiate a program of counterinflammation that preserves energy storage. Here we report that amlexanox, an approved small-molecule therapeutic presently used in the clinic to treat aphthous ulcers and asthma, is an inhibitor of these kinases. Treatment of obese mice with amlexanox elevates energy expenditure through increased thermogenesis, producing weight loss, improved insulin sensitivity and decreased steatosis. Because of its record of safety in patients, amlexanox may be an interesting candidate for clinical evaluation in the treatment of obesity and related disorders.


Aminopyridines/pharmacology , Anti-Obesity Agents/pharmacology , Energy Metabolism/drug effects , I-kappa B Kinase/antagonists & inhibitors , Insulin Resistance , Obesity/metabolism , Protein Serine-Threonine Kinases/antagonists & inhibitors , Animals , Anti-Allergic Agents/pharmacology , Cell Line , Diet, High-Fat , Enzyme Activation , Fatty Liver/drug therapy , Glucose Metabolism Disorders/drug therapy , I-kappa B Kinase/metabolism , Insulin Resistance/immunology , Intra-Abdominal Fat/drug effects , Intra-Abdominal Fat/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Obese , NF-kappa B/metabolism , Obesity/drug therapy , Obesity/immunology , Oxygen Consumption/drug effects , Protein Serine-Threonine Kinases/metabolism , Weight Loss/drug effects
...