Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Am J Physiol Endocrinol Metab ; 298(3): E419-28, 2010 Mar.
Article in English | MEDLINE | ID: mdl-19996381

ABSTRACT

Chronic inflammation is an important etiology underlying obesity-related disorders such as insulin resistance and type 2 diabetes, and recent findings indicate that the macrophage can be the initiating cell type responsible for this chronic inflammatory state. The mammalian silent information regulator 2 homolog SIRT1 modulates several physiological processes important for life span, and a potential role of SIRT1 in the regulation of insulin sensitivity has been shown. However, with respect to inflammation, the role of SIRT1 in regulating the proinflammatory pathway within macrophages is poorly understood. Here, we show that knockdown of SIRT1 in the mouse macrophage RAW264.7 cell line and in intraperitoneal macrophages broadly activates the JNK and IKK inflammatory pathways and increases LPS-stimulated TNFalpha secretion. Moreover, gene expression profiles reveal that SIRT1 knockdown leads to an increase in inflammatory gene expression. We also demonstrate that SIRT1 activators inhibit LPS-stimulated inflammatory pathways, as well as secretion of TNFalpha, in a SIRT1-dependent manner in RAW264.7 cells and in primary intraperitoneal macrophages. Treatment of Zucker fatty rats with a SIRT1 activator leads to greatly improved glucose tolerance, reduced hyperinsulinemia, and enhanced systemic insulin sensitivity during glucose clamp studies. These in vivo insulin-sensitizing effects were accompanied by a reduction in tissue inflammation markers and a decrease in the adipose tissue macrophage proinflammatory state, fully consistent with the in vitro effects of SIRT1 in macrophages. In conclusion, these results define a novel role for SIRT1 as an important regulator of macrophage inflammatory responses in the context of insulin resistance and raise the possibility that targeting of SIRT1 might be a useful strategy for treating the inflammatory component of metabolic diseases.


Subject(s)
Inflammation/metabolism , Insulin Resistance/genetics , Insulin/metabolism , Macrophage Activation/genetics , Macrophages/metabolism , Sirtuin 1/metabolism , Animals , Cells, Cultured , Gene Expression , Male , Mice , Rats , Rats, Zucker , Signal Transduction
2.
Mol Cell Biol ; 29(5): 1363-74, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19103747

ABSTRACT

SIRT1 is a prominent member of a family of NAD(+)-dependent enzymes and affects a variety of cellular functions ranging from gene silencing, regulation of the cell cycle and apoptosis, to energy homeostasis. In mature adipocytes, SIRT1 triggers lipolysis and loss of fat content. However, the potential effects of SIRT1 on insulin signaling pathways are poorly understood. To assess this, we used RNA interference to knock down SIRT1 in 3T3-L1 adipocytes. SIRT1 depletion inhibited insulin-stimulated glucose uptake and GLUT4 translocation. This was accompanied by increased phosphorylation of JNK and serine phosphorylation of insulin receptor substrate 1 (IRS-1), along with inhibition of insulin signaling steps, such as tyrosine phosphorylation of IRS-1, and phosphorylation of Akt and ERK. In contrast, treatment of cells with specific small molecule SIRT1 activators led to an increase in glucose uptake and insulin signaling as well as a decrease in serine phosphorylation of IRS-1. Moreover, gene expression profiles showed that SIRT1 expression was inversely related to inflammatory gene expression. Finally, we show that treatment of 3T3-L1 adipocytes with a SIRT1 activator attenuated tumor necrosis factor alpha-induced insulin resistance. Taken together, these data indicate that SIRT1 is a positive regulator of insulin signaling at least partially through the anti-inflammatory actions in 3T3-L1 adipocytes.


Subject(s)
Inflammation , Insulin Resistance , Insulin/physiology , Sirtuins/physiology , 3T3-L1 Cells , Adipocytes , Animals , Glucose/metabolism , Glucose Transporter Type 4/metabolism , Insulin/metabolism , Mice , RNA Interference , Signal Transduction , Sirtuin 1
3.
Proc Natl Acad Sci U S A ; 105(18): 6614-9, 2008 May 06.
Article in English | MEDLINE | ID: mdl-18445652

ABSTRACT

Glucagon-like peptide-1 (GLP-1) is a polypeptide hormone secreted from enteroendocrine L cells and potentiates glucose-dependent insulin secretion in pancreatic beta cells. Recently the GLP-1 receptor (GLP-1 R) has been a focus for new anti-diabetic therapy with the introduction of GLP-1 analogues and DPP-IV inhibitors, and this has stimulated additional interest in the mechanisms of GLP-1 signaling. Here we identify a mechanism for GLP-1 action, showing that the scaffold protein beta-arrestin-1 mediates the effects of GLP-1 to stimulate cAMP production and insulin secretion in beta cells. Using a coimmunoprecipitation technique, we also found a physical association between the GLP-1 R and beta-arrestin-1 in cultured INS-1 pancreatic beta cells. beta-Arrestin-1 knockdown broadly attenuated GLP-1 signaling, causing decreased ERK and CREB activation and IRS-2 expression as well as reduced cAMP levels and impaired insulin secretion. However, beta-arrestin-1 knockdown did not affect GLP-1 R surface expression and ligand-induced GLP-1 R internalization/desensitization. Taken together, these studies indicate that beta-arrestin-1 plays a role in GLP-1 signaling leading to insulin secretion, defining a previously undescribed mechanism for GLP-1 action.


Subject(s)
Arrestins/metabolism , Glucagon-Like Peptide 1/pharmacology , Insulin-Secreting Cells/drug effects , Insulin-Secreting Cells/metabolism , Insulin/metabolism , Signal Transduction/drug effects , Animals , Cells, Cultured , Cyclic AMP/metabolism , Endocytosis/drug effects , Glucagon-Like Peptide-1 Receptor , Insulin Receptor Substrate Proteins , Insulin Secretion , Intracellular Signaling Peptides and Proteins/metabolism , Phosphoproteins/metabolism , Protein Binding/drug effects , Rats , Receptors, Glucagon/metabolism , beta-Arrestin 1 , beta-Arrestins
4.
J Biol Chem ; 282(39): 28549-28556, 2007 Sep 28.
Article in English | MEDLINE | ID: mdl-17664271

ABSTRACT

Tumor necrosis factor-alpha (TNFalpha) is a proinflammatory cytokine secreted from macrophages and adipocytes. It is well known that chronic TNFalpha exposure can lead to insulin resistance both in vitro and in vivo and that elevated blood levels of TNFalpha are observed in obese and/or diabetic individuals. TNFalpha has many acute biologic effects, mediated by a complex intracellular signaling pathway. In these studies we have identified new G-protein signaling components to this pathway in 3T3-L1 adipocytes. We found that beta-arrestin-1 is associated with TRAF2 (TNF receptor-associated factor 2), an adaptor protein of TNF receptors, and that TNFalpha acutely stimulates tyrosine phosphorylation of G alpha(q/11) with an increase in G alpha(q/11) activity. Small interfering RNA-mediated knockdown of beta-arrestin-1 inhibits TNFalpha-induced tyrosine phosphorylation of G alpha(q/11) by interruption of Src kinase activation. TNFalpha stimulates lipolysis in 3T3-L1 adipocytes, and beta-arrestin-1 knockdown blocks the effects of TNFalpha to stimulate ERK activation and glycerol release. TNFalpha also led to activation of JNK with increased expression of the proinflammatory gene, monocyte chemoattractant protein-1 and matrix metalloproteinase 3, and beta-arrestin-1 knockdown inhibited both of these effects. Taken together these results reveal novel elements of TNFalpha action; 1) the trimeric G-protein component G alpha(q/11) and the adapter protein beta-arrestin-1 can function as signaling molecules in the TNFalpha action cascade; 2) beta-arrestin-1 can couple TNFalpha stimulation to ERK activation and lipolysis; 3) beta-arrestin-1 and G alpha(q/11) can mediate TNFalpha-induced phosphatidylinositol 3-kinase activation and inflammatory gene expression.


Subject(s)
Adipocytes/metabolism , Arrestins/metabolism , GTP-Binding Protein alpha Subunits, Gq-G11/metabolism , MAP Kinase Signaling System/drug effects , Multiprotein Complexes/metabolism , Receptors, Tumor Necrosis Factor, Type I/metabolism , Tumor Necrosis Factor-alpha/pharmacology , 3T3-L1 Cells , Animals , Diabetes Complications/metabolism , Enzyme Activation/drug effects , Extracellular Signal-Regulated MAP Kinases/metabolism , Glycerol/metabolism , Humans , Inflammation Mediators/metabolism , Insulin Resistance , Lipolysis/drug effects , Mice , Obesity/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation/drug effects , Tumor Necrosis Factor-alpha/metabolism , beta-Arrestin 1 , beta-Arrestins
5.
Mol Cell Biol ; 27(14): 5172-83, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17515613

ABSTRACT

Phosphatidylinositol 3-kinase activation of Akt signaling is critical to insulin-stimulated glucose transport and GLUT4 translocation. However, the downstream signaling events following Akt activation which mediate glucose transport stimulation remain relatively unknown. Here we identify an Akt consensus phosphorylation motif in the actin-based motor protein myosin 5a and show that insulin stimulation leads to phosphorylation of myosin 5a at serine 1650. This Akt-mediated phosphorylation event enhances the ability of myosin 5a to interact with the actin cytoskeleton. Small interfering RNA-induced inhibition of myosin 5a and expression of dominant-negative myosin 5a attenuate insulin-stimulated glucose transport and GLUT4 translocation. Furthermore, knockdown of Akt2 or expression of dominant-negative Akt (DN-Akt) abolished insulin-stimulated phosphorylation of myosin 5a, inhibited myosin 5a binding to actin, and blocked insulin-stimulated glucose transport. Taken together, these data indicate that myosin 5a is a newly identified direct substrate of Akt2 and, upon insulin stimulation, phosphorylated myosin 5a facilitates anterograde movement of GLUT4 vesicles along actin to the cell surface.


Subject(s)
Glucose Transporter Type 4/metabolism , Insulin/pharmacology , Myosin Heavy Chains/metabolism , Myosin Type V/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Transport Vesicles/drug effects , Transport Vesicles/metabolism , 3T3-L1 Cells , Actins/metabolism , Amino Acid Sequence , Animals , Biological Transport/drug effects , Cell Membrane/drug effects , Cell Membrane/metabolism , Genes, Dominant , Glucose/metabolism , Humans , Isoenzymes/metabolism , Mice , Molecular Sequence Data , Myosin Heavy Chains/chemistry , Myosin Type V/chemistry , Phosphorylation/drug effects , Protein Binding/drug effects , Protein Kinase C/metabolism , RNA Interference , Substrate Specificity/drug effects , rab4 GTP-Binding Proteins/metabolism
6.
RNA ; 12(5): 851-61, 2006 May.
Article in English | MEDLINE | ID: mdl-16540693

ABSTRACT

The scanning model of RNA translation proposes that highly stable secondary structures within mRNAs can inhibit translation, while structures of lower thermal stability also affect translation if close enough to the 5' methyl G cap. However, only fragmentary information is available about the dependence of translation efficiency in live mammalian cells on the thermodynamic stability, location, and GC content of RNA structures in the 5'-untranslated region. We devised a two-color fluorescence assay for translation efficiency in single live cells and compared a wide range of hairpins with predicted thermal stabilities ranging from -10 to -50 kcal/mol and 5' G cap-to-hairpin distances of 1-46 bases. Translation efficiency decreased abruptly as hairpin stabilities increased from deltaG = -25 to -35 kcal/mol. Shifting a hairpin as little as nine bases relative to the 5' cap could modulate translation more than 50-fold. Increasing GC content diminished translation efficiency when predicted thermal stability and cap-to-hairpin distances were held constant. We additionally found naturally occurring 5'-untranslated regions affected translation differently in live cells compared with translation in in vitro lysates. Our study will assist scientists in designing experiments that deliberately modulate mammalian translation with designed 5' UTRs.


Subject(s)
RNA Caps/metabolism , Animals , Base Sequence , CHO Cells , COS Cells , Carrier Proteins/pharmacology , Cells, Cultured , Chlorocebus aethiops , Cricetinae , Cytomegalovirus/genetics , DNA Repair Enzymes , DNA-Binding Proteins/genetics , Fibroblast Growth Factor 5/pharmacology , Fibroblasts/cytology , Fibroblasts/drug effects , In Vitro Techniques , Mice , Molecular Sequence Data , Nuclear Proteins/genetics , Promoter Regions, Genetic , RNA Caps/chemistry , RNA Caps/genetics , RNA, Viral/genetics , Ubiquitin-Protein Ligases
7.
J Biol Chem ; 280(51): 42300-6, 2005 Dec 23.
Article in English | MEDLINE | ID: mdl-16239226

ABSTRACT

Although the cytoskeletal network is important for insulin-induced glucose uptake, several studies have assessed the effects of microtubule disruption on glucose transport with divergent results. Here, we investigated the effects of microtubule-depolymerizing reagent, nocodazole and colchicine, on GLUT4 translocation in 3T3-L1 adipocytes. After nocodazole treatment to disrupt microtubules, GLUT4 vesicles were dispersed from the perinuclear region in the basal state, and insulin-induced GLUT4 translocation was partially inhibited by 20-30%, consistent with other reports. We found that platelet-derived growth factor (PDGF), which did not stimulate GLUT4 translocation in intact cells, was surprisingly able to enhance GLUT4 translocation to approximately 50% of the maximal insulin response, in nocodazole-treated cells with disrupted microtubules. This effect of PDGF was blocked by pretreatment with wortmannin and attenuated in cells pretreated with cytochalasin D. Using confocal microscopy, we found an increased co-localization of GLUT4 and F-actin in nocodazole-treated cells upon PDGF stimulation compared with control cells. Furthermore, microinjection of small interfering RNA targeting the actin-based motor Myo1c, but not the microtubule-based motor KIF3, significantly inhibited both insulin- and PDGF-stimulated GLUT4 translocation after nocodazole treatment. In summary, our data suggest that 1) proper perinuclear localization of GLUT4 vesicles is a requirement for insulin-specific stimulation of GLUT4 translocation, and 2) nocodazole treatment disperses GLUT4 vesicles from the perinuclear region allowing them to engage insulin and PDGF-sensitive actin filaments, which can participate in GLUT4 translocation in a phosphatidylinositol 3-kinase-dependent manner.


Subject(s)
Adipocytes/metabolism , Glucose Transporter Type 4/metabolism , Insulin/metabolism , Microtubules/metabolism , Signal Transduction , 3T3-L1 Cells , Actins/metabolism , Animals , Mice , Phosphatidylinositol 3-Kinases/metabolism , Platelet-Derived Growth Factor/pharmacology , Protein Transport , Proto-Oncogene Proteins c-akt/metabolism
8.
J Biol Chem ; 280(42): 35361-71, 2005 Oct 21.
Article in English | MEDLINE | ID: mdl-16085647

ABSTRACT

Lipid infusion and high fat feeding are established causes of systemic and adipose tissue insulin resistance. In this study, we treated 3T3-L1 adipocytes with a mixture of free fatty acids (FFAs) to investigate the molecular mechanisms underlying fat-induced insulin resistance. FFA treatment impaired insulin receptor-mediated signal transduction and decreased insulin-stimulated GLUT4 translocation and glucose transport. FFAs activated the stress/inflammatory kinases c-Jun N-terminal kinase (JNK) and IKKbeta, and the suppressor of cytokine signaling protein 3, increased secretion of the inflammatory cytokine tumor necrosis factor (TNF)-alpha, and decreased secretion of adiponectin into the medium. RNA interference-mediated down-regulation of JNK blocked JNK activation and prevented most of the FFA-induced defects in insulin action. Blockade of TNF-alpha signaling with neutralizing antibodies to TNF-alpha or its receptors or with a dominant negative TNF-alpha peptide had a partial effect to inhibit FFA-induced cellular insulin resistance. We found that JNK activation by FFAs was not inhibited by blocking TNF-alpha signaling, whereas the FFA-induced increase in TNF-alpha secretion was inhibited by RNA interference-mediated JNK knockdown. Together, these results indicate that 1) JNK can be activated by FFAs through TNF-alpha-independent mechanisms, 2) activated JNK is a major contributor to FFA-induced cellular insulin resistance, and 3) TNF-alpha is an autocrine/paracrine downstream effector of activated JNK that can also mediate insulin resistance.


Subject(s)
Fatty Acids, Nonesterified/metabolism , Insulin Resistance , MAP Kinase Kinase 4/physiology , Tumor Necrosis Factor-alpha/physiology , 3T3-L1 Cells , Adipocytes/metabolism , Adiponectin/metabolism , Animals , Biological Transport , Blotting, Western , Cell Differentiation , Deoxyglucose/metabolism , Down-Regulation , Enzyme Activation , Enzyme-Linked Immunosorbent Assay , Genes, Dominant , Glucose/metabolism , Glucose Transporter Type 4 , I-kappa B Kinase/metabolism , Inflammation , Insulin/metabolism , JNK Mitogen-Activated Protein Kinases/metabolism , Lipids/chemistry , MAP Kinase Kinase 4/metabolism , Mice , Protein Transport , RNA Interference , Signal Transduction , Suppressor of Cytokine Signaling 3 Protein , Suppressor of Cytokine Signaling Proteins/metabolism , Time Factors , Tumor Necrosis Factor-alpha/metabolism
9.
Mol Endocrinol ; 19(11): 2760-8, 2005 Nov.
Article in English | MEDLINE | ID: mdl-15994203

ABSTRACT

G protein-coupled receptor kinases (GRKs) regulate seven-transmembrane receptors (7TMRs) by phosphorylating agonist-activated 7TMRs. Recently, we have reported that GRK2 can function as a negative regulator of insulin action by interfering with G protein-q/11 alpha-subunit (Galphaq/11) signaling, causing decreased glucose transporter 4 (GLUT4) translocation. We have also reported that chronic endothelin-1 (ET-1) treatment leads to heterologous desensitization of insulin signaling with decreased tyrosine phosphorylation of insulin receptor substrate (IRS)-1 and Galphaq/11, and decreased insulin-stimulated glucose transport in 3T3-L1 adipocytes. In the current study, we have investigated the role of GRK2 in chronic ET-1-induced insulin resistance. Insulin-induced GLUT4 translocation was inhibited by pretreatment with ET-1 for 24 h, and we found that this inhibitory effect was rescued by microinjection of anti-GRK2 antibody or GRK2 short interfering RNA. We further found that GRK2 mediates the inhibitory effects of ET-1 by two distinct mechanisms. Firstly, adenovirus-mediated overexpression of either wild-type (WT)- or kinase-deficient (KD)-GRK2 inhibited Galphaq/11 signaling, including tyrosine phosphorylation of Galphaq/11 and cdc42-associated phosphatidylinositol 3-kinase activity. Secondly, ET-1 treatment caused Ser/Thr phosphorylation of IRS-1 and IRS-1 protein degradation. Overexpression of KD-GRK2, but not WT-GRK2, inhibited ET-1-induced serine 612 phosphorylation of IRS-1 and restored activation of this pathway. Taken together, these results suggest that GRK2 mediates ET-1-induced insulin resistance by 1) inhibition of Galphaq/11 activation, and this effect is independent of GRK2 kinase activity, and 2) GRK2 kinase activity-mediated IRS-1 serine phosphorylation and degradation.


Subject(s)
Adipocytes/metabolism , Endothelin-1/pharmacology , GTP-Binding Protein alpha Subunits, Gq-G11/antagonists & inhibitors , Insulin Resistance , Phosphoproteins/antagonists & inhibitors , Protein Serine-Threonine Kinases/metabolism , 3T3-L1 Cells , Adipocytes/drug effects , Animals , Antibodies/pharmacology , Endothelin-1/metabolism , Endothelin-1/toxicity , GTP-Binding Protein alpha Subunits, Gq-G11/metabolism , Glucose Transporter Type 4/metabolism , Insulin/pharmacology , Insulin Receptor Substrate Proteins , Mice , Mutation , Phosphoproteins/metabolism , Phosphorylation , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/genetics , Protein Transport , RNA, Small Interfering/pharmacology , Serine/metabolism , Transcriptional Activation
10.
Bioorg Med Chem Lett ; 15(17): 3917-20, 2005 Sep 01.
Article in English | MEDLINE | ID: mdl-15993597

ABSTRACT

We have developed two photolabile insulin prodrugs, insulin-2P and insulin-3P. These prodrugs were synthesized by protecting GlyA1 (N(alphaA1)), and one or both of the PheB1 (N(alphaB1)) and LysB29 (N(epsilonB29)) amino groups in insulin using 5'-(alpha-methyl-nitro-piperonyl)oxy-carbonyl as the protecting group. These insulin prodrugs were efficiently activated by exposure to longwave UV light to produce insulin quantitatively. Using 2-deoxyglucose uptake assays, both di- and tri-protected compounds were less active than native insulin in the protected state, and showed comparable activity to native insulin upon photoactivation.


Subject(s)
Insulin/analogs & derivatives , Prodrugs/chemical synthesis , 3T3-L1 Cells , Animals , Chromatography, High Pressure Liquid , Deoxyglucose/pharmacokinetics , Insulin/pharmacology , Mice , Photochemistry , Prodrugs/pharmacology , Prodrugs/radiation effects , Structure-Activity Relationship , Ultraviolet Rays
11.
EMBO J ; 23(14): 2821-9, 2004 Jul 21.
Article in English | MEDLINE | ID: mdl-15241473

ABSTRACT

G protein-coupled receptor kinases (GRKs) represent a class of proteins that classically phosphorylate agonist-activated G protein-coupled receptors, leading to uncoupling of the receptor from further G protein activation. Recently, we have reported that the heterotrimeric G protein alpha-subunit, Galphaq/11, can mediate insulin-stimulated glucose transport. GRK2 contains a regulator of G protein signaling (RGS) domain with specificity for Galphaq/11. Therefore, we postulated that GRK2 could be an inhibitor of the insulin signaling cascade leading to glucose transport in 3T3-L1 adipocytes. In this study, we demonstrate that microinjection of anti-GRK2 antibody or siRNA against GRK2 increased insulin-stimulated insulin-responsive glucose transporter 4 (GLUT4) translocation, while adenovirus-mediated overexpression of wild-type or kinase-deficient GRK2 inhibited insulin-stimulated GLUT4 translocation as well as 2-deoxyglucose uptake. Importantly, a mutant GRK2 lacking the RGS domain was without effect. Taken together, these results indicate that through its RGS domain endogenous GRK2 functions as a negative regulator of insulin-stimulated glucose transport by interfering with Galphaq/11 signaling to GLUT4 translocation. Furthermore, inhibitors of GRK2 can lead to enhanced insulin sensitivity.


Subject(s)
Cyclic AMP-Dependent Protein Kinases/antagonists & inhibitors , Glucose/metabolism , Insulin/metabolism , Signal Transduction , 3T3-L1 Cells , Adenoviridae/genetics , Adipocytes/drug effects , Adipocytes/metabolism , Animals , Biological Transport , Cell Differentiation , Cell Line , Cyclic AMP-Dependent Protein Kinases/chemistry , Cyclic AMP-Dependent Protein Kinases/genetics , Deoxyglucose/metabolism , GTP-Binding Protein alpha Subunits, Gq-G11/metabolism , Gene Deletion , Gene Expression Regulation/drug effects , Glucose Transporter Type 4 , Insulin/pharmacology , Mice , Monosaccharide Transport Proteins/drug effects , Monosaccharide Transport Proteins/metabolism , Muscle Proteins/drug effects , Muscle Proteins/metabolism , Phosphatidylinositol 3-Kinases/analysis , Phosphatidylinositol 3-Kinases/metabolism , Plasmids/metabolism , Protein Structure, Tertiary , RNA, Messenger/analysis , RNA, Messenger/metabolism , RNA, Small Interfering/metabolism , beta-Adrenergic Receptor Kinases
SELECTION OF CITATIONS
SEARCH DETAIL
...