Time-resolved and tissue-specific systems analysis of the pathogenesis of insulin resistance

Robert Kleemann, Marjan van Erk, Lars Verschuren, Anita M van den Hoek, Maud Koek, Peter Y Wielinga, Annie Jie, Linette Pellis, Ivana Bobeldijk-Pastorova, Thomas Kelder, Karin Toet, Suzan Wopereis, Nicole Cnubben, Chris Evelo, Ben van Ommen, Teake Kooistra, Robert Kleemann, Marjan van Erk, Lars Verschuren, Anita M van den Hoek, Maud Koek, Peter Y Wielinga, Annie Jie, Linette Pellis, Ivana Bobeldijk-Pastorova, Thomas Kelder, Karin Toet, Suzan Wopereis, Nicole Cnubben, Chris Evelo, Ben van Ommen, Teake Kooistra

Abstract

Background: The sequence of events leading to the development of insulin resistance (IR) as well as the underlying pathophysiological mechanisms are incompletely understood. As reductionist approaches have been largely unsuccessful in providing an understanding of the pathogenesis of IR, there is a need for an integrative, time-resolved approach to elucidate the development of the disease.

Methodology/principal findings: Male ApoE3Leiden transgenic mice exhibiting a humanized lipid metabolism were fed a high-fat diet (HFD) for 0, 1, 6, 9, or 12 weeks. Development of IR was monitored in individual mice over time by performing glucose tolerance tests and measuring specific biomarkers in plasma, and hyperinsulinemic-euglycemic clamp analysis to assess IR in a tissue-specific manner. To elucidate the dynamics and tissue-specificity of metabolic and inflammatory processes key to IR development, a time-resolved systems analysis of gene expression and metabolite levels in liver, white adipose tissue (WAT), and muscle was performed. During HFD feeding, the mice became increasingly obese and showed a gradual increase in glucose intolerance. IR became first manifest in liver (week 6) and then in WAT (week 12), while skeletal muscle remained insulin-sensitive. Microarray analysis showed rapid upregulation of carbohydrate (only liver) and lipid metabolism genes (liver, WAT). Metabolomics revealed significant changes in the ratio of saturated to polyunsaturated fatty acids (liver, WAT, plasma) and in the concentrations of glucose, gluconeogenesis and Krebs cycle metabolites, and branched amino acids (liver). HFD evoked an early hepatic inflammatory response which then gradually declined to near baseline. By contrast, inflammation in WAT increased over time, reaching highest values in week 12. In skeletal muscle, carbohydrate metabolism, lipid metabolism, and inflammation was gradually suppressed with HFD.

Conclusions/significance: HFD-induced IR is a time- and tissue-dependent process that starts in liver and proceeds in WAT. IR development is paralleled by tissue-specific gene expression changes, metabolic adjustments, changes in lipid composition, and inflammatory responses in liver and WAT involving p65-NFkB and SOCS3. The alterations in skeletal muscle are largely opposite to those in liver and WAT.

Conflict of interest statement

Competing Interests: Authors employed by TNO (R.K., M.v.E., A.J., L.V., A.M.v.d.H., M.K, P.Y.W, L.P., I.B.–P., K.T., S.W., N.C., B.v.O., and T.K.) and the Top Institute Food and Nutrition (R.K., P.Y.W., and K.T.) have a potential conflict of interest as their organizations may benefit from a product or patent generated on the basis of the published data. In these cases, the authors will however not receive additional salary, additional personal income, or any form of financial support.

Figures

Figure 1. Fat mass over time.
Figure 1. Fat mass over time.
(A) Bars represent total fat mass subdivided into the epididymal, subcutaneous and visceral adipose tissue depots. Groups of n = 15 animals were sacrificed at the time points indicated to determine adipose mass of the various depots. Data are presented as means ± SEM. *P<0.05 indicates significance compared to t = 0 for each type of adipose tissue depot or the total fat mass. (B) Plasma leptin levels over time presented as means±SEM. *P<0.01 compared to t = 0.
Figure 2. HFD feeding alters the relative…
Figure 2. HFD feeding alters the relative amount of saturated, monounsaturated and polyunsaturated fatty acids.
Free fatty acid analysis of plasma. Representative fatty acids of the C18 category (stearic acid, oleic acid, alpha-linolenic acid and linoleic acid) are shown. Data presented are relative amounts and values are means ± SEM. *P

Figure 3. Effect of HFD feeding on…

Figure 3. Effect of HFD feeding on fasting plasma insulin and glucose levels and glucose…

Figure 3. Effect of HFD feeding on fasting plasma insulin and glucose levels and glucose tolerance.
Plasma insulin (A) and glucose (B) levels determined after a 5 hr fasting period. A glucose tolerance test (GTT) (C) was performed 2.5 weeks prior to inclusion into the study (not shown) and each animal underwent a second GTT, in week 0, 1, 6, 9, and 12 of HFD feeding (n = 15 each). After a 5 h fast, animals received 2 g glucose/kg body weight (i.p.). Glucose was recorded at the time points indicated and the area under the curve (AUC) (D) was calculated. Values are means ± SEM. **P<0.01; *P<0.05

Figure 4. Glucose tolerance test and hyperinsulinemic,…

Figure 4. Glucose tolerance test and hyperinsulinemic, euglycemic clamp analysis.

In a parallel, independent animal…

Figure 4. Glucose tolerance test and hyperinsulinemic, euglycemic clamp analysis.
In a parallel, independent animal experiment, HFD-fed mice were subjected to hyperinsulinemic, euglycemic clamp analysis. (A) shows the glucose infusion rate (GIR) under hyperinsulinemic conditions. Radioactive tracers allowed determining hepatic glucose production rate (HGP) (B) as well as the tissue-specific uptake of glucose into WAT (C) and skeletal muscle (D). ‘Basal’ indicates prior to hyperinsulinemic clamp conditions and ‘hyper’ indicates hyperinsulinemic clamp conditions. Values are means ± SEM. *P<0.05 vs. t = 0.

Figure 5. Time-resolved pathway analysis for liver,…

Figure 5. Time-resolved pathway analysis for liver, WAT and muscle.

Hierarchically clustered heat map of…

Figure 5. Time-resolved pathway analysis for liver, WAT and muscle.
Hierarchically clustered heat map of enriched functional groups of genes (Gene Ontology). Shown are functional groups of genes that are significantly enriched in at least one time point and tissue. Clustering analysis (Pearson correlation, complete linkage) was performed on average t-values for each time point. Red (blue) indicates up-regulation (down-regulation) of a functional group and a positive (negative) t-value. The color intensity reflects the magnitude of a change. Columns represent the different tissues over time and rows represent the functional clusters.

Figure 6. Analysis of biological processes in…

Figure 6. Analysis of biological processes in liver, WAT and muscle over time.

Response profiles…

Figure 6. Analysis of biological processes in liver, WAT and muscle over time.
Response profiles of specific biological processes are shown together with their average t-scores.

Figure 7. WAT is an important source…

Figure 7. WAT is an important source of inflammation.

Comparison of the expression of prototype…

Figure 7. WAT is an important source of inflammation.
Comparison of the expression of prototype inflammatory genes in WAT and liver. A, PAI-1 (Serpine1) and MCP-1 (Ccl2); B, IL-receptor antagonist (Ilm1) and C, Fibrinogen-alpha, -beta, -gamma in WAT (left) and liver (right). Data were confirmed in independent RT-PCR analyses.

Figure 8. Chronic inflammatory processes in liver…

Figure 8. Chronic inflammatory processes in liver modulated by HFD.

Transcriptomics data were analyzed by…

Figure 8. Chronic inflammatory processes in liver modulated by HFD.
Transcriptomics data were analyzed by PathVisio with criterion ANOVA qvalue

Figure 9. Chronic inflammatory processes in WAT…

Figure 9. Chronic inflammatory processes in WAT and muscle modulated by HFD.

Transcriptomics data were…

Figure 9. Chronic inflammatory processes in WAT and muscle modulated by HFD.
Transcriptomics data were analyzed by PathVisio with criterion ANOVA qvalue (A) as well as the major metabolic processes of skeletal muscle (B). Red bars indicate upregulated differentially expressed genes, green bars indicated downregulated genes. Blue nodes represent biological processes.

Figure 10. HFD feeding causes hepatosteatosis and…

Figure 10. HFD feeding causes hepatosteatosis and WAT hypertrophy.

( A ) Representative photomicrographs of…

Figure 10. HFD feeding causes hepatosteatosis and WAT hypertrophy.
(A) Representative photomicrographs of Oil Red O-stained liver cross-sections at t = 0 (i), week 6 (ii) and week 12 (iii). (B) Analysis of intrahepatic triglycerides. Values are mean ± SEM *P<0.01 vs t = 0, **P<0.001 vs t = 0. (C) HPS-stained epididymal adipose tissue at t = 0 (i) and week 12 (ii). Biopsies from tissues also analyzed by microarray were used. Bars indicate 100 µm. (D) Leptin expression over time.

Figure 11. Overview of development of IR…

Figure 11. Overview of development of IR in ApoE3Leiden mice fed a HFD.

Weeks of…

Figure 11. Overview of development of IR in ApoE3Leiden mice fed a HFD.
Weeks of HFD feeding are indicated together with the time point at which liver, and WAT developed IR. Changes in the lipid composition (ie. the quality and nature of fatty acids) occurred within the first 6 weeks and changes persisted until the end of the study. Extensive metabolic adjustments (indicating metabolic stress) and development of local inflammation in liver and WAT from week 1 onward. Hepatosteatosis developed after week 6.
All figures (11)
Similar articles
Cited by
References
    1. Kahn SE, Hull RL, Utzschneider KM. Mechanisms linking obesity to insulin resistance and type 2 diabetes. Nature. 2006;444:840–846. - PubMed
    1. Schenk S, Saberi M, Olefsky JM. Insulin sensitivity: modulation by nutrients and inflammation. J Clin Invest. 2008;118:2992–3002. - PMC - PubMed
    1. Hotamisligil GS. Inflammation and metabolic disorders. Nature. 2006;444:860–867. - PubMed
    1. Herder C, Kolb H, Koenig W, Haastert B, Muller-Scholze S, et al. Association of systemic concentrations of macrophage migration inhibitory factor with impaired glucose tolerance and type 2 diabetes: results from the Cooperative Health Research in the Region of Augsburg, Survey 4 (KORA S4). Diabetes Care. 2006;29:368–371. - PubMed
    1. Verschuren L, Kooistra T, Bernhagen J, Voshol PJ, Ouwens DM, et al. MIF Deficiency Reduces Chronic Inflammation in White Adipose Tissue and Impairs the Development of Insulin Resistance, Glucose Intolerance, and Associated Atherosclerotic Disease. Circ Res. 2009;105:99–107. - PMC - PubMed
Show all 59 references
Publication types
MeSH terms
Substances
[x]
Cite
Copy Download .nbib
Format: AMA APA MLA NLM
Figure 3. Effect of HFD feeding on…
Figure 3. Effect of HFD feeding on fasting plasma insulin and glucose levels and glucose tolerance.
Plasma insulin (A) and glucose (B) levels determined after a 5 hr fasting period. A glucose tolerance test (GTT) (C) was performed 2.5 weeks prior to inclusion into the study (not shown) and each animal underwent a second GTT, in week 0, 1, 6, 9, and 12 of HFD feeding (n = 15 each). After a 5 h fast, animals received 2 g glucose/kg body weight (i.p.). Glucose was recorded at the time points indicated and the area under the curve (AUC) (D) was calculated. Values are means ± SEM. **P<0.01; *P<0.05
Figure 4. Glucose tolerance test and hyperinsulinemic,…
Figure 4. Glucose tolerance test and hyperinsulinemic, euglycemic clamp analysis.
In a parallel, independent animal experiment, HFD-fed mice were subjected to hyperinsulinemic, euglycemic clamp analysis. (A) shows the glucose infusion rate (GIR) under hyperinsulinemic conditions. Radioactive tracers allowed determining hepatic glucose production rate (HGP) (B) as well as the tissue-specific uptake of glucose into WAT (C) and skeletal muscle (D). ‘Basal’ indicates prior to hyperinsulinemic clamp conditions and ‘hyper’ indicates hyperinsulinemic clamp conditions. Values are means ± SEM. *P<0.05 vs. t = 0.
Figure 5. Time-resolved pathway analysis for liver,…
Figure 5. Time-resolved pathway analysis for liver, WAT and muscle.
Hierarchically clustered heat map of enriched functional groups of genes (Gene Ontology). Shown are functional groups of genes that are significantly enriched in at least one time point and tissue. Clustering analysis (Pearson correlation, complete linkage) was performed on average t-values for each time point. Red (blue) indicates up-regulation (down-regulation) of a functional group and a positive (negative) t-value. The color intensity reflects the magnitude of a change. Columns represent the different tissues over time and rows represent the functional clusters.
Figure 6. Analysis of biological processes in…
Figure 6. Analysis of biological processes in liver, WAT and muscle over time.
Response profiles of specific biological processes are shown together with their average t-scores.
Figure 7. WAT is an important source…
Figure 7. WAT is an important source of inflammation.
Comparison of the expression of prototype inflammatory genes in WAT and liver. A, PAI-1 (Serpine1) and MCP-1 (Ccl2); B, IL-receptor antagonist (Ilm1) and C, Fibrinogen-alpha, -beta, -gamma in WAT (left) and liver (right). Data were confirmed in independent RT-PCR analyses.
Figure 8. Chronic inflammatory processes in liver…
Figure 8. Chronic inflammatory processes in liver modulated by HFD.
Transcriptomics data were analyzed by PathVisio with criterion ANOVA qvalue

Figure 9. Chronic inflammatory processes in WAT…

Figure 9. Chronic inflammatory processes in WAT and muscle modulated by HFD.

Transcriptomics data were…

Figure 9. Chronic inflammatory processes in WAT and muscle modulated by HFD.
Transcriptomics data were analyzed by PathVisio with criterion ANOVA qvalue (A) as well as the major metabolic processes of skeletal muscle (B). Red bars indicate upregulated differentially expressed genes, green bars indicated downregulated genes. Blue nodes represent biological processes.

Figure 10. HFD feeding causes hepatosteatosis and…

Figure 10. HFD feeding causes hepatosteatosis and WAT hypertrophy.

( A ) Representative photomicrographs of…

Figure 10. HFD feeding causes hepatosteatosis and WAT hypertrophy.
(A) Representative photomicrographs of Oil Red O-stained liver cross-sections at t = 0 (i), week 6 (ii) and week 12 (iii). (B) Analysis of intrahepatic triglycerides. Values are mean ± SEM *P<0.01 vs t = 0, **P<0.001 vs t = 0. (C) HPS-stained epididymal adipose tissue at t = 0 (i) and week 12 (ii). Biopsies from tissues also analyzed by microarray were used. Bars indicate 100 µm. (D) Leptin expression over time.

Figure 11. Overview of development of IR…

Figure 11. Overview of development of IR in ApoE3Leiden mice fed a HFD.

Weeks of…

Figure 11. Overview of development of IR in ApoE3Leiden mice fed a HFD.
Weeks of HFD feeding are indicated together with the time point at which liver, and WAT developed IR. Changes in the lipid composition (ie. the quality and nature of fatty acids) occurred within the first 6 weeks and changes persisted until the end of the study. Extensive metabolic adjustments (indicating metabolic stress) and development of local inflammation in liver and WAT from week 1 onward. Hepatosteatosis developed after week 6.
All figures (11)
Figure 9. Chronic inflammatory processes in WAT…
Figure 9. Chronic inflammatory processes in WAT and muscle modulated by HFD.
Transcriptomics data were analyzed by PathVisio with criterion ANOVA qvalue (A) as well as the major metabolic processes of skeletal muscle (B). Red bars indicate upregulated differentially expressed genes, green bars indicated downregulated genes. Blue nodes represent biological processes.
Figure 10. HFD feeding causes hepatosteatosis and…
Figure 10. HFD feeding causes hepatosteatosis and WAT hypertrophy.
(A) Representative photomicrographs of Oil Red O-stained liver cross-sections at t = 0 (i), week 6 (ii) and week 12 (iii). (B) Analysis of intrahepatic triglycerides. Values are mean ± SEM *P<0.01 vs t = 0, **P<0.001 vs t = 0. (C) HPS-stained epididymal adipose tissue at t = 0 (i) and week 12 (ii). Biopsies from tissues also analyzed by microarray were used. Bars indicate 100 µm. (D) Leptin expression over time.
Figure 11. Overview of development of IR…
Figure 11. Overview of development of IR in ApoE3Leiden mice fed a HFD.
Weeks of HFD feeding are indicated together with the time point at which liver, and WAT developed IR. Changes in the lipid composition (ie. the quality and nature of fatty acids) occurred within the first 6 weeks and changes persisted until the end of the study. Extensive metabolic adjustments (indicating metabolic stress) and development of local inflammation in liver and WAT from week 1 onward. Hepatosteatosis developed after week 6.

References

    1. Kahn SE, Hull RL, Utzschneider KM. Mechanisms linking obesity to insulin resistance and type 2 diabetes. Nature. 2006;444:840–846.
    1. Schenk S, Saberi M, Olefsky JM. Insulin sensitivity: modulation by nutrients and inflammation. J Clin Invest. 2008;118:2992–3002.
    1. Hotamisligil GS. Inflammation and metabolic disorders. Nature. 2006;444:860–867.
    1. Herder C, Kolb H, Koenig W, Haastert B, Muller-Scholze S, et al. Association of systemic concentrations of macrophage migration inhibitory factor with impaired glucose tolerance and type 2 diabetes: results from the Cooperative Health Research in the Region of Augsburg, Survey 4 (KORA S4). Diabetes Care. 2006;29:368–371.
    1. Verschuren L, Kooistra T, Bernhagen J, Voshol PJ, Ouwens DM, et al. MIF Deficiency Reduces Chronic Inflammation in White Adipose Tissue and Impairs the Development of Insulin Resistance, Glucose Intolerance, and Associated Atherosclerotic Disease. Circ Res. 2009;105:99–107.
    1. Kanda H, Tateya S, Tamori Y, Kotani K, Hiasa K, et al. MCP-1 contributes to macrophage infiltration into adipose tissue, insulin resistance, and hepatic steatosis in obesity. J Clin Invest. 2006;116:1494–1505.
    1. Hotamisligil GS, Shargill NS, Spiegelman BM. Adipose expression of tumor necrosis factor-alpha: direct role in obesity-linked insulin resistance. Science. 1993;259:87–91.
    1. Cai D, Yuan M, Frantz DF, Melendez PA, Hansen L, et al. Local and systemic insulin resistance resulting from hepatic activation of IKK-beta and NF-kappaB. Nat Med. 2005;11:183–190.
    1. Kaneto H, Nakatani Y, Miyatsuka T, Kawamori D, Matsuoka TA, et al. Possible novel therapy for diabetes with cell-permeable JNK-inhibitory peptide. Nat Med. 2004;10:1128–1132.
    1. Ueki K, Kondo T, Kahn CR. Suppressor of cytokine signaling 1 (SOCS-1) and SOCS-3 cause insulin resistance through inhibition of tyrosine phosphorylation of insulin receptor substrate proteins by discrete mechanisms. Mol Cell Biol. 2004;24:5434–5446.
    1. Zadelaar S, Kleemann R, Verschuren L, de Vries-van der Weij, van der HJ, et al. Mouse models for atherosclerosis and pharmaceutical modifiers. Arterioscler Thromb Vasc Biol. 2007;27:1706–1721.
    1. Verschuren L, Kleemann R, Offerman EH, Szalai AJ, Emeis SJ, et al. Effect of low dose atorvastatin versus diet-induced cholesterol lowering on atherosclerotic lesion progression and inflammation in apolipoprotein E*3-Leiden transgenic mice. Arterioscler Thromb Vasc Biol. 2005;25:161–167.
    1. Rein D, Schijlen E, Kooistra T, Herbers K, Verschuren L, et al. Transgenic flavonoid tomato intake reduces C-reactive protein in human C-reactive protein transgenic mice more than wild-type tomato. J Nutr. 2006;136:2331–2337.
    1. Haslinger B, Kleemann R, Toet KH, Kooistra T. Simvastatin suppresses tissue factor expression and increases fibrinolytic activity in tumor necrosis factor-alpha-activated human peritoneal mesothelial cells. Kidney Int. 2003;63:2065–2074.
    1. Kleemann R, Verschuren L, van Erk MJ, Nikolsky Y, Cnubben NH, et al. Atherosclerosis and liver inflammation induced by increased dietary cholesterol intake: a combined transcriptomics and metabolomics analysis. Genome Biol. 2007;8:R200.
    1. De Groot PJ, Reiff C, Mayer C, Muller M. NuGO contributions to GenePattern. Genes Nutr. 2008;3:143–146.
    1. Baccini M, Bachmaier EM, Biggeri A, Boekschoten MV, Bouwman FG, et al. The NuGO proof of principle study package: a collaborative research effort of the European Nutrigenomics Organisation. Genes Nutr. 2008;3:147–151.
    1. Boorsma A, Foat BC, Vis D, Klis F, Bussemaker HJ. T-profiler: scoring the activity of predefined groups of genes using gene expression data. Nucleic Acids Res. 2005;33:W592–W595.
    1. van Iersel MP, Kelder T, Pico AR, Hanspers K, Coort S, et al. Presenting and exploring biological pathways with PathVisio. BMC Bioinformatics. 2008;9:399.
    1. Pico AR, Kelder T, van Iersel MP, Hanspers K, Conklin BR, et al. WikiPathways: pathway editing for the people. PLoS Biol. 2008;6:e184.
    1. Cline MS, Smoot M, Cerami E, Kuchinsky A, Landys N, et al. Integration of biological networks and gene expression data using Cytoscape. Nat Protoc. 2007;2:2366–2382.
    1. Bobeldijk I, Hekman M, de Vries-van der Weij, Coulier L, Ramaker R, et al. Quantitative profiling of bile acids in biofluids and tissues based on accurate mass high resolution LC-FT-MS: compound class targeting in a metabolomics workflow. J Chromatogr B Analyt Technol Biomed Life Sci. 2008;871:306–313.
    1. Pettinella C, Lee SH, Cipollone F, Blair IA. Targeted quantitative analysis of fatty acids in atherosclerotic plaques by high sensitivity liquid chromatography/tandem mass spectrometry. J Chromatogr B Analyt Technol Biomed Life Sci. 2007;850:168–176.
    1. Koek MM, Muilwijk B, van Stee LL, Hankemeier T. Higher mass loadability in comprehensive two-dimensional gas chromatography-mass spectrometry for improved analytical performance in metabolomics analysis. J Chromatogr A. 2008;1186:420–429.
    1. Abdul-Hussien H, Soekhoe RG, Weber E, von der Thusen JH, Kleemann R, et al. Collagen degradation in the abdominal aneurysm: a conspiracy of matrix metalloproteinase and cysteine collagenases. Am J Pathol. 2007;170:809–817.
    1. Gervois P, Kleemann R, Pilon A, Percevault F, Koenig W, et al. Global suppression of IL-6-induced acute phase response gene expression after chronic in vivo treatment with the peroxisome proliferator-activated receptor-alpha activator fenofibrate. J Biol Chem. 2004;279:16154–16160.
    1. Wu Y, Ouyang JP, Wu K, Wang SS, Wen CY, et al. Rosiglitazone ameliorates abnormal expression and activity of protein tyrosine phosphatase 1B in the skeletal muscle of fat-fed, streptozotocin-treated diabetic rats. Br J Pharmacol. 2005;146:234–243.
    1. Lindeman JH, bdul-Hussien H, van Bockel JH, Wolterbeek R, Kleemann R. Clinical trial of doxycycline for matrix metalloproteinase-9 inhibition in patients with an abdominal aneurysm: doxycycline selectively depletes aortic wall neutrophils and cytotoxic T cells. Circulation. 2009;119:2209–2216.
    1. Guerra B, Fuentes T, gado-Guerra S, Guadalupe-Grau A, Olmedillas H, et al. Gender dimorphism in skeletal muscle leptin receptors, serum leptin and insulin sensitivity. PLoS One. 2008;3:e3466.
    1. Kleemann R, Gervois PP, Verschuren L, Staels B, Princen HM, et al. Fibrates down-regulate IL-1-stimulated C-reactive protein gene expression in hepatocytes by reducing nuclear p50-NFkappa B-C/EBP-beta complex formation. Blood. 2003;101:545–551.
    1. Bligh EG, Dyer WJ. A rapid method of total lipid extraction and purification. Can J Biochem Physiol. 1959;37:911–917.
    1. Zhou YT, Wang ZW, Higa M, Newgard CB, Unger RH. Reversing adipocyte differentiation: implications for treatment of obesity. Proc Natl Acad Sci U S A. 1999;96:2391–2395.
    1. Elchebly M, Payette P, Michaliszyn E, Cromlish W, Collins S, et al. Increased insulin sensitivity and obesity resistance in mice lacking the protein tyrosine phosphatase-1B gene. Science. 1999;283:1544–1548.
    1. Nilsson M, Holst JJ, Bjorck IM. Metabolic effects of amino acid mixtures and whey protein in healthy subjects: studies using glucose-equivalent drinks. Am J Clin Nutr. 2007;85:996–1004.
    1. Kawaguchi T, Nagao Y, Matsuoka H, Ide T, Sata M. Branched-chain amino acid-enriched supplementation improves insulin resistance in patients with chronic liver disease. Int J Mol Med. 2008;22:105–112.
    1. Burcelin R, Crivelli V, Dacosta A, Roy-Tirelli A, Thorens B. Heterogeneous metabolic adaptation of C57BL/6J mice to high-fat diet. Am J Physiol Endocrinol Metab. 2002;282:E834–E842.
    1. Kooistra T, Verschuren L, de Vries-van der Weij, Koenig W, Toet K, et al. Fenofibrate reduces atherogenesis in ApoE*3Leiden mice: evidence for multiple antiatherogenic effects besides lowering plasma cholesterol. Arterioscler Thromb Vasc Biol. 2006;26:2322–2330.
    1. Chisholm KW, O'Dea K. Effect of short-term consumption of a high fat diet on glucose tolerance and insulin sensitivity in the rat. J Nutr Sci Vitaminol (Tokyo) 1987;33:377–390.
    1. Zhou YE, Egeland GM, Meltzer SJ, Kubow S. The association of desaturase 9 and plasma fatty acid composition with insulin resistance-associated factors in female adolescents. Metabolism. 2009;58:158–166.
    1. Fedor D, Kelley DS. Prevention of insulin resistance by n-3 polyunsaturated fatty acids. Curr Opin Clin Nutr Metab Care. 2009;12:138–146.
    1. Warensjo E, Sundstrom J, Lind L, Vessby B. Factor analysis of fatty acids in serum lipids as a measure of dietary fat quality in relation to the metabolic syndrome in men. Am J Clin Nutr. 2006;84:442–448.
    1. Vassilopoulos S, Esk C, Hoshino S, Funke BH, Chen CY, et al. A role for the CHC22 clathrin heavy-chain isoform in human glucose metabolism. Science. 2009;324:1192–1196.
    1. Boden G. Role of fatty acids in the pathogenesis of insulin resistance and NIDDM. Diabetes. 1997;46:3–10.
    1. Singh U, Zhong S, Xiong M, Li TB, Sniderman A, et al. Increased plasma non-esterified fatty acids and platelet-activating factor acetylhydrolase are associated with susceptibility to atherosclerosis in mice. Clin Sci (Lond) 2004;106:421–432.
    1. Oosterveer MH, van Dijk TH, Tietge UJ, Boer T, Havinga R, et al. High fat feeding induces hepatic fatty acid elongation in mice. PLoS One. 2009;4:e6066.
    1. Postic C, Girard J. Contribution of de novo fatty acid synthesis to hepatic steatosis and insulin resistance: lessons from genetically engineered mice. J Clin Invest. 2008;118:829–838.
    1. Gentile CL, Pagliassotti MJ. The role of fatty acids in the development and progression of nonalcoholic fatty liver disease. J Nutr Biochem. 2008;19:567–576.
    1. Montecucco F, Steffens S, Mach F. Insulin resistance: a proinflammatory state mediated by lipid-induced signaling dysfunction and involved in atherosclerotic plaque instability. Mediators Inflamm. 2008: 767623.
    1. Herder C, Klopp N, Baumert J, Muller M, Khuseyinova N, et al. Effect of macrophage migration inhibitory factor (MIF) gene variants and MIF serum concentrations on the risk of type 2 diabetes: results from the MONICA/KORA Augsburg Case-Cohort Study, 1984-2002. Diabetologia. 2008;51:276–284.
    1. Perrier S, Darakhshan F, Hajduch E. IL-1 receptor antagonist in metabolic diseases: Dr Jekyll or Mr Hyde? FEBS Lett. 2006;580:6289–6294.
    1. Somm E, Cettour-Rose P, Asensio C, Charollais A, Klein M, et al. Interleukin-1 receptor antagonist is upregulated during diet-induced obesity and regulates insulin sensitivity in rodents. Diabetologia. 2006;49:387–393.
    1. Lagathu C, Yvan-Charvet L, Bastard JP, Maachi M, Quignard-Boulange A, et al. Long-term treatment with interleukin-1beta induces insulin resistance in murine and human adipocytes. Diabetologia. 2006;49:2162–2173.
    1. Shi H, Cave B, Inouye K, Bjorbaek C, Flier JS. Overexpression of suppressor of cytokine signaling 3 in adipose tissue causes local but not systemic insulin resistance. Diabetes. 2006;55:699–707.
    1. Steinberg GR, McAinch AJ, Chen MB, O'Brien PE, Dixon JB, et al. The suppressor of cytokine signaling 3 inhibits leptin activation of AMP-kinase in cultured skeletal muscle of obese humans. J Clin Endocrinol Metab. 2006;91:3592–3597.
    1. Somm E, Cettour-Rose P, Asensio C, Charollais A, Klein M, et al. Interleukin-1 receptor antagonist is upregulated during diet-induced obesity and regulates insulin sensitivity in rodents. Diabetologia. 2006;49:387–393.
    1. Kato K, Satoh H, Endo Y, Yamada D, Midorikawa S, et al. Thiazolidinediones down-regulate plasminogen activator inhibitor type 1 expression in human vascular endothelial cells: A possible role for PPARgamma in endothelial function. Biochem Biophys Res Commun. 1999;258:431–435.
    1. Schultz DR, Arnold PI. Properties of four acute phase proteins: C-reactive protein, serum amyloid A protein, alpha 1-acid glycoprotein, and fibrinogen. Semin Arthritis Rheum. 1990;20:129–147.
    1. Poitou C, Viguerie N, Cancello R, De MR, Cinti S, et al. Serum amyloid A: production by human white adipocyte and regulation by obesity and nutrition. Diabetologia. 2005;48:519–528.
    1. Carlsen H, Haugen F, Zadelaar S, Kleemann R, Kooistra T, et al. Diet-induced obesity increases NF-kappaB signaling in reporter mice. Genes Nutr. 2009;4:215–222.

Source: PubMed

3
Prenumerera