当前位置: 首页 > 医学版 > 期刊论文 > 内科学 > 糖尿病学杂志 > 2005年 > 第10期 > 正文
编号:11256856
Tumor Necrosis Factor- Induces Skeletal Muscle Insulin Resistance in Healthy Human Subjects via Inhibition of Akt Substrate 160 Phosphorylat
     1 Department of Infectious Diseases and the Copenhagen Muscle Research Centre, The Centre of Inflammation and Metabolism, Rigshospitalet University of Copenhagen, Faculty of Health Sciences, Copenhagen, Denmark

    2 Department of Surgical Sciences, Section for Integrative Physiology, Karolinska Institutet, Stockholm, Sweden

    3 Washington University, School of Medicine, St. Louis, Missouri

    ABSTRACT

    Most lifestyle-related chronic diseases are characterized by low-grade systemic inflammation and insulin resistance. Excessive tumor necrosis factor- (TNF-) concentrations have been implicated in the development of insulin resistance, but direct evidence in humans is lacking. Here, we demonstrate that TNF- infusion in healthy humans induces insulin resistance in skeletal muscle, without effect on endogenous glucose production, as estimated by a combined euglycemic insulin clamp and stable isotope tracer method. TNF- directly impairs glucose uptake and metabolism by altering insulin signal transduction. TNF- infusion increases phosphorylation of p70 S6 kinase, extracellular signaleCregulated kinase-1/2, and c-Jun NH2-terminal kinase, concomitant with increased serine and reduced tyrosine phosphorylation of insulin receptor substrate-1. These signaling effects are associated with impaired phosphorylation of Akt substrate 160, the most proximal step identified in the canonical insulin signaling cascade regulating GLUT4 translocation and glucose uptake. Thus, excessive concentrations of TNF- negatively regulate insulin signaling and whole-body glucose uptake in humans. Our results provide a molecular link between low-grade systemic inflammation and the metabolic syndrome.

    Low-grade systemic inflammation is a feature of obesity and insulin resistance (1). There is now an increasing appreciation that type 2 diabetes and systemic inflammation are related and that elevated cytokine levels may contribute to insulin resistance (2eC4). Type 2 diabetes is associated with elevated levels of the proinflammatory cytokine tumor necrosis factor- (TNF-) in skeletal muscle (5), adipose tissue (6), and plasma (7eC9). However, the clinical consequence of elevated TNF- concentrations on hepatic insulin sensitivity and peripheral glucose uptake is unknown. In isolated rat skeletal muscle, TNF- had no effect on insulin-mediated glucose uptake, even at very high doses (10). Although this may imply the inhibitory effects of TNF- on insulin action are targeted exclusively at the vascular tissue, cell culture studies provide direct evidence that TNF- inhibits insulin signaling and glucose metabolism (11eC13).

    Excessive TNF- levels cause insulin resistance through modifications of insulin signal transduction. In cultured cells, TNF- induces insulin resistance through increased serine phosphorylation of insulin receptor substrate-1 (IRS-1), which subsequently converts IRS-1 to an inhibitor of insulin receptor tyrosine kinase activity (14). IRS-1 plays a critical role in mediating insulin signal transduction along pathways governing metabolic responses, such as GLUT4 translocation and glucose uptake in skeletal muscle (15). Direct evidence for a role of TNF- as a negative regulator of insulin action is revealed from functional studies in knockout mice (16). Ablation of TNF- improves insulin sensitivity in dietary-induced obese and ob/ob mice by preventing obesity-related reductions in insulin signaling in skeletal muscle and adipose tissue (16).

    Evidence implicating TNF- in the development of insulin resistance in humans is correlative (17). TNF- is produced locally in adipose tissue (6) and skeletal muscle (5) and may function in an autocrine manner to inhibit insulin signaling and glucose transport. Attempts to improve insulin sensitivity in obese type 2 diabetic patients by neutralizing TNF-, using antieCTNF- antibodies (18) or recombinant TNF receptors, have been ineffective (19). The lack of efficacy may be attributable to the treatment strategy because only a single injection of a neutralizing antibody toward TNF/TNF receptor was evaluated (18,19). Alternatively, the neutralizing antibodies may be ineffective in skeletal muscle. Although animal studies provide convincing evidence for a role of TNF- in promoting insulin resistance, results from human studies are inconclusive.

    We determined the effects of TNF- infusion on whole-body insulin-mediated glucose uptake and signal transduction in healthy humans. Furthermore, we assessed the effects of insulin and TNF- on phosphorylation of Akt substrate 160 (AS160). Phosphorylation of AS160, a protein containing a Rab GAP (GTPase activating protein) domain (20), is the most proximal step in the insulin signaling cascade identified to date, and it promotes GLUT4 exocytosis to the plasma membrane to facilitate glucose uptake and metabolism (21,22).

    RESEARCH DESIGN AND METHODS

    Nine healthy men participated in this study. Before the two experimental days, the subjects underwent a thorough clinical examination and a standard blood chemistry analysis. Volunteers were aged 26.3 ± 1.7 years (mean ± SE) and had a height of 181.7 ± 1.7 cm, weight of 78.8 ± 2.2 kg, and BMI of 23.9 ± 0.7 kg/cm2. Subjects received oral and written information regarding the potential hazards and discomfort of the experiment. The information was approved by the local ethical committee (no. KF 01-006/04) in accordance with the Helsinki Declaration.

    In vivo protocol.

    The study protocol is depicted in Fig. 1. Subjects underwent two separate experiments with 4-h saline or human recombinant TNF- (Beromun; Boeringer-Ingelheim) infusion. On each occasion, both arms were catheterized in the antecubital vein. One side was used for infusion and the other for blood sampling. A priming dose of the glucose tracer [6,6-2H2]glucose (22 e蘭ol/kg; Cambridge Isotope Laboratories) was given, and the infusion was started at a rate of 0.22 e蘭ol · kgeC1 · mineC1 for 2 h to achieve equilibrium in the glucose pool for subsequent determination of glucose turnover. During the insulin infusion, the tracer infusion rate was reduced to 0.11 e蘭ol · kgeC1 · mineC1. The placebo or TNF- (rate: 1,000 ng/h per m2) infusion was started at 60 min, and TNF- was administered in saline with 20% human albumin. Placebo consisted of saline infusion with 20% human albumin. After 2 h of tracer infusion and 1 h of TNF-/placebo, a hyperinsulinemic-euglycemic clamp was started. The infusion rate of insulin was 20 mU/min per m2, and the plasma glucose was kept at 5 mmol/l by infusion of 20% glucose enriched to 2.5% with the glucose tracer. Plasma glucose was measured frequently (EML 105 radiometer) every 5 min the 1st h and every 10 min thereafter to maintain the glucose concentration at fasting levels. Blood samples and biopsies from the vastus lateralis of musculus quadriceps were obtained at the designated time points (Fig. 1). Skeletal muscle biopsies were obtained at 0, 30, 60, and 180 min after insulin infusion. Biopsies were frozen in liquid nitrogen and stored at eC80°C until further analysis. Blood was separated into plasma and stored at eC80°C.

    Stable isotopes.

    Isotope enrichment was measured, using liquid chromatographyeCmass spectrometry (Finnegan aQa). First, the glucose was derivatized by benzoylation. Then, 20 e蘬 of plasma was vigorously mixed (5 min) together with 1 mol/l K2HPO4, 20 e蘬 of 8 mol/l NaOH, and 10 e蘬 benzoyl chloride. Thereafter, 10 e蘬 of 1.4 mol/l H3PO4 and 600 e蘬 ethylacetate was added, and the samples were vortexed. The ethylacetate phase (75 e蘬) containing the benzoylated glucose was transferred to a new tube and dried under a stream of nitrogen. The derivate was solubilized in the liquid chromatography buffer (80:20 acetonitril:water), filtrated, and transferred to a liquid chromatography vial. The derivate was separated by liquid chromatography using a column flow at 0.05 ml/min isocratic 80:20 acetonitril:water; the water contained 125 mmol/l NH4-acetate for optimal ionization. The ionization source was electro-spray ionization at 37 kV and a temperature of 250°C for soft fragmentation. The fragment of the derivate contained both deuterium atoms and had a mass at 231 m/z. The area under the curves for the non-enriched M-peak (231 m/z) and the enriched M+2-peak (233 m/z) were measured. The tracer-to-tracee ratio was calculated as the ratio of the M and M+2 area under the curve minus the natural background enrichment.

    Clinical chemistry.

    Hormone analysis was performed using commercially available kits. Insulin and C-peptide were measured using enzyme-linked immunosorbent assay kits from DAKO; the glucagon kit was from Linco Research, and enzyme immunosorbent assay cortisol kits were from Diagnostic Systems Laboratories. TNF- and interleukin (IL)-6 were measured using kits from R&D Systems. Free fatty acids were measured using a nonesterified fatty acid kit from Wako Chemicals, and glycerol was assessed using a method previously described (23). Both assays were automated and performed on a Cobas Fara robot (Roche).

    Calculations.

    The glucose release and uptake from the circulation was calculated using a single-pool noneCsteady-state model, modified after the Steele equation. The exogenous glucose infused during the clamp was enriched to 2.5%, thereby improving the sensitivity of the method. The volume of glucose distribution was set at 145 ml/kg. Endogenous glucose production (rate of appearance [Ra]) and glucose uptake (rate of disappearance [Rd]) was calculated using the following:

    and

    where F is the tracer infusion rate (e蘭ol/kg x min), pV is the distribution volume of the tracee (ml/kg), C is the concentration (e蘭ol/ml), E is the enrichment (tracer-to-tracee ratio), Eexo is the enrichment of exogenous glucose, Ginf is the glucose infusion rate (e蘭ol/min x kg), and t is time (min).

    Determination of protein phosphorylation.

    Muscles biopsies were homogenized at 4°C in 20 mmol/l Tris-HCl (pH 7.4), 140 mmol/l NaCl, 10 mmol/l EDTA, 4 mmol/l NaVO4, 100 mmol/l NaF, 10 mmol/l pyrophosphate, and 1% Nonidet P-40, supplemented with protease inhibitors. Lysates were centrifuged (12,000g for 15 min) in lysate buffer containing 1% Nonidet P-40, 0.5% sodium deoxycholate, and 0.1% SDS, supplemented with a freshly prepared cocktail of protease inhibitors (ICN Pharmaceuticals). Total protein was determined using the Bradford method (Bio-Rad). Aliquots of muscle lysate (50 e蘥 protein) were separated by SDS-PAGE, and proteins were transferred to polyvinylidene fluoride membranes. Membranes were incubated overnight with antieCphospho-p70S6 kinase, antieCphospho-extracellular signaleCregulated kinase (ERK)-1/2 (recognizing ERK-1/2 phosphorylation at Thr202 and Tyr204, antieCphospho-c-Jun NH2-terminal kinase (JNK; recognizing phosphorylation of the 46- and 54-kDa isoforms of JNK at Thr183 and Tyr185, or antieCphospho-(Ser/Thr) Akt substrate antibody (Cell Signaling Technology). For IRS-1 phosphorylation, aliquots of muscle lysate (500 e蘥 protein) were immunoprecipitated with antieCIRS-1 and subjected to SDS-PAGE. Proteins were transferred to polyvinylidene fluoride membranes and probed with polyclonal anti-phosphotyrosine, antieCphospho-Ser312, or antieCphospho-Ser639 (Cell Signaling Technology). Blots were stripped and reprobed with antieCIRS-1 to normalize for equal protein amount. Immunoreactive proteins were visualized by enhanced chemiluminescence (ECL Plus; Amersham) and quantified by densitometry, using Molecular Analyst software (Bio-Rad). Results are reported in arbitrary units, relative to each respective basal biopsy obtained during saline infusion.

    Statistics.

    When data were normally distributed, arithmetical means ± SE were presented. Otherwise, a logarithmic transformation was performed, and geometric means ± SE were presented. The statistical comparisons in the stable isotope experiments are based on the area under the curve measurements obtained during the basal (eC20 to 0 min) and insulin-stimulated (60eC180 min) conditions. Student’s paired t test was used to assess differences between saline and TNF- treatment when only one variable was compared. To evaluate the difference between groups, a two-way repeated-measurement ANOVA was performed and a two-sample t test was used as a post hoc test. To assess the effect of time, a one-way repeated-measurement ANOVA was performed. A paired t test was applied as post hoc analysis to compare change from the "pre" sample. Both tests were Bonferroni corrected. Systat version 8.0 for Windows (SPSS) was used for the statistical analysis. Differences were considered significant at P < 0.05.

    RESULTS

    Effect of TNF- infusion on whole-body glucose uptake and hepatic glucose production.

    To investigate the effects of TNF- on glucose homeostasis, nine healthy human volunteers received an infusion of either TNF- or placebo for 4 h (Fig. 1). Before TNF- infusion, [6,6-2H2]glucose was administered to allow for the determination of glucose turnover. A euglycemic insulin clamp was initiated after 1 h of TNF- infusion. TNF- infusion inhibited whole-body glucose uptake (Fig. 2). Ra decreased in response to the insulin treatment, without any effect of TNF- administration. Rd increased in response to insulin infusion in both trials (P < 0.001). During insulin infusion, the Rd largely reflects insulin-mediated glucose uptake in skeletal muscle. Importantly, hepatic glucose production was unaltered by TNF- infusion (Fig. 2), demonstrating a direct effect of TNF- on peripheral tissues.

    Clinical chemistry and subject characteristics during the in vivo study.

    Plasma TNF- concentration increased eightfold during the infusion (Table 1). Plasma IL-6 concentration was gradually elevated in response to TNF infusion, with a 12-fold increase noted at the end of the infusion (Table 1). TNF- infusion induced a minor increase in body temperature and heart rate (Table 1), but volunteers did not experience any adverse effects or major changes in metabolic hormones. The plasma insulin concentration during the euglycemic clamp corresponded to levels generally obtained after a light meal. Insulin infusion suppressed free fatty acids and glycerol concentrations (Table 1).

    Effect of TNF- infusion on signal transduction in skeletal muscle.

    To determine the mechanism underlying the TNF-eCinduced inhibition of insulin-stimulated peripheral glucose uptake, biopsies of vastus lateralis quadriceps muscle were obtained (Fig. 1). Here, we show that TNF- infusion for 1 h, before the administration of exogenous insulin, increased phosphorylation of p70 S6 kinase (S6K), ERK-1/2, and JNK in skeletal muscle (Fig. 3A, B, and C, respectively), concomitant with increased serine phosphorylation of IRS-1 at site 312 (Fig. 3D). These targets have been implicated in the negative regulation of insulin signal transduction (24). Given this profile, we hypothesized that insulin action on signal transduction would be blunted by TNF-.

    TNF- infusion altered insulin action on IRS-1 by interfering with tyrosine and serine phosphorylation. Insulin-stimulated tyrosine phosphorylation of IRS-1 was prevented by TNF- infusion (Fig. 4A), whereas IRS-1 serine phosphorylation was increased at two different sites (Fig. 4B and C). TNF- increased serine phosphorylation on site 312 and 636/639 with different kinetics. Under insulin-stimulated conditions, TNF- infusion further increased serine phosphorylation of IRS-1 on site 312, with the most dramatic response at 180 min (Fig. 4B). Phosphorylation of IRS-1 on site 636/639 was unaltered by TNF- infusion. Under insulin-stimulated conditions, TNF- infusion was associated with a dramatic increase in serine phosphorylation of IRS-1 on site 636/639 at 30 min (Fig. 4C). The latter findings were observed in parallel with impaired phosphorylation of IRS-1 on tyrosine residues (Fig. 4A). Moreover, using a polyclonal affinity-purified antibody that recognizes Akt phosphorylation motif peptide sequences [RXRXX(T/S)] (20,21), we show TNF- infusion impairs insulin action of AS160 (Fig. 4D). Reduced AS160 phosphorylation may provide a mechanism for TNF-eCinduced insulin resistance on glucose uptake because GLUT4 exocytosis is dependent on AS160 phosphorylation (20,21).

    DISCUSSION

    In healthy humans, infusion of physiologically relevant systemic concentrations of TNF- inhibits peripheral insulin-mediated glucose uptake, without influencing endogenous glucose production from the liver. This observation is consistent with the notion that inflammatory pathways are coupled to metabolic control in humans. Our results indicate that TNF- infusion primarily compromises skeletal muscle insulin action. We resolved this impairment to the level of insulin signaling. The TNF- concentrations achieved during the in vivo infusion were slightly higher than typically found in patients with low-grade systemic inflammation, such as type 2 diabetes and cardiovascular diseases, but considerably lower than in septic diseases (25). Our finding that a relatively minor elevation in the TNF- concentration has deleterious effects on peripheral insulin action is of clinical relevance and highlights fundamental differences between rodents and humans in sensitivity to cytokines (26).

    TNF- has been shown to have a direct (insulin-independent) effect on S6K (27) and ERK-1/2 (28) in cultured cells. Here, we provide evidence that TNF- infusion increases phosphorylation of S6K, ERK-1/2, and JNK, concomitant with increased serine phosphorylation of IRS-1. These direct effects of TNF- raise the question of whether a prolonged exposure of this cytokine would induce insulin resistance. However, blood glucose concentration was unaltered in fasting subjects after a 3-h TNF- infusion (29). Furthermore, here, we provide evidence that basal glucose uptake is unaltered after 1 h of TNF- infusion. Thus, under these acute in vivo conditions, TNF- has a direct insulin-independent effect on signal transduction in human skeletal muscle that does not appear to modify basal hepatic or peripheral glucose metabolism.

    TNF- impairs insulin-stimulated rates of glucose storage in cultured human muscle cells (30) and whole-body insulin-mediated glucose uptake in rats (31). Here, we provide evidence that TNF- infusion negatively regulates insulin signaling and whole-body glucose uptake in humans. In cultured cells (14,32) and rodents (33), TNF- impairs insulin signaling by increasing serine phosphorylation of IRS-1. Phosphorylation of IRS-1 at Ser 307 (site 312 in human IRS-1) is critical for the inhibitory effect of TNF-, which appears to be mediated by an association of activated JNK with IRS-1 and the insulin receptor, thereby providing a mechanism to partly explain the development of insulin resistance in response to excessive cytokine production (34,35). Under insulin-stimulated conditions, infusion of TNF- prevented tyrosine phosphorylation of IRS-1 and increased serine phosphorylation of IRS-1 at sites 312 and 636/639 in human skeletal muscle. The impairment in IRS-1 signaling was associated with reduced phosphorylation of AS160, a Rab GTPaseeCactivating protein (20) that plays a role in insulin-stimulated GLUT4 translocation/exocytosis to the plasma membrane in 3T3-L1 adipocytes (20,21). Insulin-stimulated AS160 phosphorylation is also impaired in skeletal muscle from type 2 diabetic patients (36). Collectively, these studies provide evidence that AS160 plays a role in the regulation of glucose metabolism and represent a target for insulin resistance in response to excessive TNF- exposure. Thus, TNF- infusion rapidly promotes skeletal muscle insulin resistance by negatively regulating insulin signal transduction to glucose uptake.

    In addition to effects on glucose metabolism, TNF- has been implicated in endothelial dysfunction. An acute exposure of cultured endothelial cells to TNF- inhibits insulin signaling and nitric oxide production, concomitant with impaired insulin-stimulated vasodilation (37). The effect of low-grade systemic TNF- concentration on blood flow in humans is unknown. TNF- and insulin oppose each other, such that insulin action is most vulnerable to TNF-eCmediated inhibition when insulin concentrations are low (as in the present study) and least vulnerable when insulin concentrations are high (38). In rats, an acute in vivo TNF- infusion completely blocked the hemodynamic effects of insulin on blood flow and capillary recruitment, and it inhibited skeletal muscle glucose uptake (31). Moreover, in a human perfused forearm model, a 200-fold "local" increase in plasma TNF- concentration was associated with impaired insulin-mediated endothelium-dependent vasodilation and skeletal muscle glucose uptake (39). Clearly, effects of TNF- on endothelial function may contribute to defects in glucose metabolism. However, our results provide evidence that TNF- also has a direct effect in skeletal muscle. Phosphorylation of S6K, ERK-1/2, JNK, and IRS-1 at site 312 was directly increased in response to the TNF- infusion, and this was associated with negative regulation along the canonical insulin signaling cascade and a subsequent impairment in glucose uptake.

    TNF- and IL-6 are early mediators of inflammation. These two cytokines are tightly linked, such that TNF- induces IL-6 production, which consequently inhibits TNF- gene expression (25). We observed a gradual rise in IL-6 concentrations during the TNF- infusion, which could conceivably contribute to the development of skeletal muscle insulin resistance. Elevations in IL-6 may lead to hepatic insulin resistance (40); however, effects on insulin signaling and glucose metabolism in skeletal muscle and adipose tissue are equivocal. IL-6 has been reported to enhance (41) and suppress (42) insulin-stimulated glucose transport in myotubes and adipocytes. In the present study, the deleterious effect of TNF- on insulin action was localized to skeletal muscle, rather than liver. Thus, we propose that TNF-, rather than IL-6, is the driver behind the observed impairment in insulin signaling and glucose uptake in skeletal muscle. This interpretation is strengthened by clinical observations in healthy humans stating that endogenous glucose production and peripheral glucose uptake are unaltered by acute infusion of recombinant human IL-6 (43).

    Our results provide a molecular link between low-grade systemic inflammation and the metabolic syndrome. Systemic inflammation and insulin resistance accompany a cluster of clinically related diseases that constitute the metabolic syndrome, as well as autoimmune and infectious diseases (1,44,45). The regulation of insulin-mediated glucose uptake in humans is extremely sensitive to cytokines. Excessive TNF- concentrations negatively regulate insulin signaling and whole-body glucose uptake in humans. The identification of the molecular targets that mediate the deleterious effects of TNF- on insulin signaling and glucose metabolism opens the possibility for new therapeutic strategies for treatment of a global clinical health problem.

    ACKNOWLEDGMENTS

    This study funded from the Commission of the European Communities (contract no. LSHM-CT-2004-005272 Exgenesis), the Swedish Medical Research Council, the Swedish Foundation for Strategic Research, the Novo Nordic Foundation, and the Swedish Diabetes Association. The Centre of Inflammation and Metabolism was supported by a grant from the Danish National Research Foundation (DG 02-512-555). The Copenhagen Muscle Research Centre was supported by grants from the University of Copenhagen Faculty of Health Sciences and the Copenhagen Hospital Corporation. This study also received support from the National Institutes of Health Clinical Nutrition Research Unit (DK 56341) and the Danish Diabetes Association.

    We thank Professor Christian Torp-Pedersen, University of Copenhagen, for donating rhTNF-.

    FOOTNOTES

    P.P. and K.B. contributed equally to this work.

    AS160, Akt substrate 160; ERK, extracellular signaleCregulated kinase; IL, interleukin; IRS, insulin receptor substrate; JNK, c-Jun NH2-terminal kinase; S6K, p70 S6 kinase; TNF-, tumor necrosis factor-

    REFERENCES

    Dandona P, Aljada A, Bandyopadhyay A: Inflammation: the link between insulin resistance, obesity and diabetes. Trends Immunol25 :4 eC7,2004

    Duncan BB, Schmidt MI, Pankow JS, Ballantyne CM, Couper D, Vigo A, Hoogeveen R, Folsom AR, Heiss G: Low-grade systemic inflammation and the development of type 2 diabetes: the Atherosclerosis Risk in Communities Study. Diabetes52 :1799 eC1805,2003

    Freeman DJ, Norrie J, Caslake MJ, Gaw A, Ford I, Lowe GD, O’Reilly DS, Packard CJ, Sattar N: C-reactive protein is an independent predictor of risk for the development of diabetes in the West of Scotland Coronary Prevention Study. Diabetes51 :1596 eC1600,2002

    Pradhan AD, Manson JE, Rifai N, Buring JE, Ridker PM: C-reactive protein, interleukin 6, and risk of developing type 2 diabetes mellitus. JAMA286 :327 eC334,2001

    Saghizadeh M, Ong JM, Garvey WT, Henry RR, Kern PA: The expression of TNF alpha by human muscle: relationship to insulin resistance. J Clin Invest97 :1111 eC1116,1996

    Hotamisligil GS, Arner P, Caro JF, Atkinson RL, Spiegelman BM: Increased adipose tissue expression of tumor necrosis factor-alpha in human obesity and insulin resistance. J Clin Invest95 :2409 eC2415,1995

    Feingold KR, Grunfeld C: Role of cytokines in inducing hyperlipidemia. Diabetes41 (Suppl. 2) :97 eC101,1992

    Mishima Y, Kuyama A, Tada A, Takahashi K, Ishioka T, Kibata M: Relationship between serum tumor necrosis factor-alpha and insulin resistance in obese men with type 2 diabetes mellitus. Diabetes Res Clin Pract52 :119 eC123,2001

    Winkler G, Lakatos P, Nagy Z, Speer G, Salamon F, Szekeres O, Kovacs M, Cseh K: Elevated serum tumor necrosis factor-alpha and endothelin 1 levels correlate with increased C-peptide concentration in android type obesity (Letter). Diabetes Care21 :1778 eC1779,1998

    Nolte LA, Hansen PA, Chen MM, Schluter JM, Gulve EA, Holloszy JO: Short-term exposure to tumor necrosis factor- does not affect insulin-stimulated glucose uptake in skeletal muscle. Diabetes47 :721 eC726,1998

    Ranganathan S, Davidson MB: Effect of tumor necrosis factor-alpha on basal and insulin-stimulated glucose transport in cultured muscle and fat cells. Metabolism45 :1089 eC1094,1996

    Patiag D, Gray S, Idris I, Donnelly R: Effects of tumour necrosis factor-alpha and inhibition of protein kinase C on glucose uptake in L6 myoblasts. Clin Sci (Lond )99 :303 eC307,2000

    de Alvaro C, Teruel T, Hernandez R, Lorenzo M: Tumor necrosis factor alpha produces insulin resistance in skeletal muscle by activation of inhibitor kappaB kinase in a p38 MAPK-dependent manner. J Biol Chem279 :17070 eC17078,2004

    Hotamisligil GS, Peraldi P, Budavari A, Ellis R, White MF, Spiegelman BM: IRS-1-mediated inhibition of insulin receptor tyrosine kinase activity in TNF-alpha- and obesity-induced insulin resistance. Science271 :665 eC668,1996

    Huang C, Thirone AC, Huang X, Klip A: Differential contribution of IRS-1 vs. IRS-2 to insulin signaling and glucose uptake in L6 myotubes. J Biol Chem280 :19426 eC19435,2005

    Uysal KT, Wiesbrock SM, Marino MW, Hotamisligil GS: Protection from obesity-induced insulin resistance in mice lacking TNF-alpha function. Nature389 :610 eC614,1997

    Lechleitner M, Herold M, Dzien-Bischinger C, Hoppichler F, Dzien A: Tumour necrosis factor-alpha plasma levels in elderly patients with type 2 diabetes mellitus-observations over 2 years. Diabet Med19 :949 eC953,2002

    Ofei F, Hurel S, Newkirk J, Sopwith M, Taylor R: Effects of an engineered human anti-TNF- antibody (CDP571) on insulin sensitivity and glycemic control in patients with NIDDM. Diabetes45 :881 eC885,1996

    Paquot N, Castillo MJ, Lefebvre PJ, Scheen AJ: No increased insulin sensitivity after a single intravenous administration of a recombinant human tumor necrosis factor receptor: Fc fusion protein in obese insulin-resistant patients. J Clin Endocrinol Metab85 :1316 eC1319,2000

    Kane S, Sano H, Liu SC, Asara JM, Lane WS, Garner CC, Lienhard GE: A method to identify serine kinase substrates: Akt phosphorylates a novel adipocyte protein with a Rab GTPase-activating protein (GAP) domain. J Biol Chem277 :22115 eC22118,2002

    Sano H, Kane S, Sano E, Miinea CP, Asara JM, Lane WS, Garner CW, Lienhard GE: Insulin-stimulated phosphorylation of a Rab GTPase-activating protein regulates GLUT4 translocation. J Biol Chem278 :14599 eC14602,2003

    Zeigerer A, McBrayer MK, McGraw TE: Insulin stimulation of GLUT4 exocytosis, but not its inhibition of endocytosis, is dependent on RabGAP AS160. Mol Biol Cell15 :4406 eC4415,2004

    van Hall G, Sacchetti M, Radegran G, Saltin B: Human skeletal muscle fatty acid and glycerol metabolism during rest, exercise and recovery. J Physiol543 :1047 eC1058,2002

    Um SH, Frigerio F, Watanabe M, Picard F, Joaquin M, Sticker M, Fumagalli S, Allegrini PR, Kozma SC, Auwerx J, Thomas G: Absence of S6K1 protects against age- and diet-induced obesity while enhancing insulin sensitivity. Nature431 :200 eC205,2004

    Petersen AM, Pedersen BK: The anti-inflammatory effects of exercise. J Appl Physiol98 :1154 eC1162,2005

    Burrell R: Human responses to bacterial endotoxin. Circ Shock43 :137 eC153,1994

    Glantschnig H, Fisher JE, Wesolowski G, Rodan GA, Reszka AA: M-CSF, TNFalpha and RANK ligand promote osteoclast survival by signaling through mTOR/S6 kinase. Cell Death Differ10 :1165 eC1177,2003

    Vary TC, Deiter G, Lang CH: Diminished ERK 1/2 and p38 MAPK phosphorylation in skeletal muscle during sepsis. Shock22 :548 eC554,2004

    Plomgaard P, Keller P, Keller C, Pedersen BK: TNF-alpha, but not IL-6, stimulates plasminogen activator inhibitor-1 expression in human subcutaneous adipose tissue. J Appl Physiol98 :2019 eC2023,2005

    Halse R, Pearson SL, McCormack JG, Yeaman SJ, Taylor R: Effects of tumor necrosis factor- on insulin action in cultured human muscle cells. Diabetes50 :1102 eC1109,2001

    Youd JM, Rattigan S, Clark MG: Acute impairment of insulin-mediated capillary recruitment and glucose uptake in rat skeletal muscle in vivo by TNF-. Diabetes49 :1904 eC1909,2000

    Paz K, Hemi R, LeRoith D, Karasik A, Elhanany E, Kanety H, Zick Y: A molecular basis for insulin resistance. Elevated serine/threonine phosphorylation of IRS-1 and IRS-2 inhibits their binding to the juxtamembrane region of the insulin receptor and impairs their ability to undergo insulin-induced tyrosine phosphorylation. J Biol Chem272 :29911 eC29918,1997

    Hotamisligil GS, Budavari A, Murray D, Spiegelman BM: Reduced tyrosine kinase activity of the insulin receptor in obesity-diabetes: central role of tumor necrosis factor-alpha. J Clin Invest94 :1543 eC1549,1994

    Aguirre V, Uchida T, Yenush L, Davis R, White MF: The c-Jun NH(2)-terminal kinase promotes insulin resistance during association with insulin receptor substrate-1 and phosphorylation of Ser(307). J Biol Chem275 :9047 eC9054,2000

    Aguirre V, Werner ED, Giraud J, Lee YH, Shoelson SE, White MF: Phosphorylation of Ser307 in insulin receptor substrate-1 blocks interactions with the insulin receptor and inhibits insulin action. J Biol Chem277 :1531 eC1537,2002

    Karlsson HK, Zierath JR, Kane S, Krook A, Lienhard GE, Wallberg-Henriksson H: Insulin-stimulated phosphorylation of the Akt substrate AS160 is impaired in skeletal muscle of type 2 diabetic subjects. Diabetes54 :1692 eC1697,2005

    Kim F, Gallis B, Corson MA: TNF-alpha inhibits flow and insulin signaling leading to NO production in aortic endothelial cells. Am J Physiol Cell Physiol280 :C1057 eCC1065,2001

    Zhang L, Wheatley CM, Richards SM, Barrett EJ, Clark MG, Rattigan S: TNF-alpha acutely inhibits vascular effects of physiological but not high insulin or contraction. Am J Physiol Endocrinol Metab285 :E654 eCE660,2003

    Rask-Madsen C, Dominguez H, Ihlemann N, Hermann T, Kober L, Torp-Pedersen C: Tumor necrosis factor-alpha inhibits insulin’s stimulating effect on glucose uptake and endothelium-dependent vasodilation in humans. Circulation108 :1815 eC1821,2003

    Senn JJ, Klover PJ, Nowak IA, Mooney RA: Interleukin-6 induces cellular insulin resistance in hepatocytes. Diabetes51 :3391 eC3399,2002

    Weigert C, Hennige AM, Brodbeck K, Haring HU, Schleicher ED: Interleukin-6 (IL-6) acts as insulin sensitizer on glycogen synthesis in human skeletal muscle cells by phosphorylation of Ser-473 of Akt. Am J Physiol Endocrinol Metab,2005

    Rotter V, Nagaev I, Smith U: Interleukin-6 (IL-6) induces insulin resistance in 3T3eCL1 adipocytes and is, like IL-8 and tumor necrosis factor-alpha, overexpressed in human fat cells from insulin-resistant subjects. J Biol Chem278 :45777 eC45784,2003

    Steensberg A, Fischer CP, Sacchetti M, Keller C, Osada T, Schjerling P, van Hall G, Febbraio MA, Pedersen BK: Acute interleukin-6 administration does not impair muscle glucose uptake or whole-body glucose disposal in healthy humans. J Physiol548 :631 eC638,2003

    Mackay F, Kalled SL: TNF ligands and receptors in autoimmunity: an update. Curr Opin Immunol14 :783 eC790,2002

    Dinarello CA, Gelfand JA, Wolff SM: Anticytokine strategies in the treatment of the systemic inflammatory response syndrome. JAMA269 :1829 eC1835,1993(Peter Plomgaard, Karim Bo)