Concordance of changes in metabolic pathways based on plasma metabolomics and skeletal muscle transcriptomics in type 1 diabetes

Tumpa Dutta, High Seng Chai, Lawrence E Ward, Aditya Ghosh, Xuan-Mai T Persson, G Charles Ford, Yogish C Kudva, Zhifu Sun, Yan W Asmann, Jean-Pierre A Kocher, K Sreekumaran Nair, Tumpa Dutta, High Seng Chai, Lawrence E Ward, Aditya Ghosh, Xuan-Mai T Persson, G Charles Ford, Yogish C Kudva, Zhifu Sun, Yan W Asmann, Jean-Pierre A Kocher, K Sreekumaran Nair

Abstract

Insulin regulates many cellular processes, but the full impact of insulin deficiency on cellular functions remains to be defined. Applying a mass spectrometry-based nontargeted metabolomics approach, we report here alterations of 330 plasma metabolites representing 33 metabolic pathways during an 8-h insulin deprivation in type 1 diabetic individuals. These pathways included those known to be affected by insulin such as glucose, amino acid and lipid metabolism, Krebs cycle, and immune responses and those hitherto unknown to be altered including prostaglandin, arachidonic acid, leukotrienes, neurotransmitters, nucleotides, and anti-inflammatory responses. A significant concordance of metabolome and skeletal muscle transcriptome-based pathways supports an assumption that plasma metabolites are chemical fingerprints of cellular events. Although insulin treatment normalized plasma glucose and many other metabolites, there were 71 metabolites and 24 pathways that differed between nondiabetes and insulin-treated type 1 diabetes. Confirmation of many known pathways altered by insulin using a single blood test offers confidence in the current approach. Future research needs to be focused on newly discovered pathways affected by insulin deficiency and systemic insulin treatment to determine whether they contribute to the high morbidity and mortality in T1D despite insulin treatment.

Figures

FIG. 1.
FIG. 1.
Heat map analysis of plasma metabolites in T1D during insulin deficiency (I−) and insulin treatment (I+) and comparison with ND. Metabolite perturbations in plasma were calculated based on the median for each metabolite level of three independent biological replicates of plasma samples from each study participant. Each row represents a metabolite, and each column depicts a subject. The study groups are color coded as follows: insulin-deprived (I−) T1D is denoted in blue, insulin-treated (I+) T1D is denoted in red, and ND groups are denoted in maroon. The fold change in metabolite levels is color coded: red pixels, upregulation; blue, downregulation; yellow, no significant change. Metabolites such as acetate, lactate, acetoacetate, hydroxybutyrate, gluconate, hydroxy adipate, carnitines, glucosamine, and taurocholate including amino acid (e.g., leucine, isoleucine, valine, N-methyl histidine, keto glutarate, glutamate, alanine, phenylalanine) were all found to be elevated in I− T1D (Supplementary Table 4). A consistent decrease was observed in other metabolites, e.g., hydroxypyridine, nicotinamide, hydroxyl nicotinic acid, adipate, methylthioribose, uridine, xanthine, hypoxanthine, methylguanosine, N-acetyl tryptophan, pipecolate, homoserine, aldosterone, arachidonyl lysolecithin, phosphoethanolamine, etc.
FIG. 2.
FIG. 2.
A: The effect of differential regulation of metabolites on the canonical pathways during I− in T1D in comparison with I+ T1D and ND. The significance of the pathways was evaluated using P values and false discovery rate <0.05. B: Altered canonical pathways following insulin treatment in T1D in comparison with ND. *Metabolic pathways that were observed exclusively after systemic insulin treatment. The significance of the pathways was evaluated using P values and false discovery rate <0.05. nNOS, neuronal nitric oxide synthase; PDGF, platelet-derived growth factor; TCA, tricarboxylic acid; GPCR, G protein-coupled receptor; FM, function and metabolism.
FIG. 2.
FIG. 2.
A: The effect of differential regulation of metabolites on the canonical pathways during I− in T1D in comparison with I+ T1D and ND. The significance of the pathways was evaluated using P values and false discovery rate <0.05. B: Altered canonical pathways following insulin treatment in T1D in comparison with ND. *Metabolic pathways that were observed exclusively after systemic insulin treatment. The significance of the pathways was evaluated using P values and false discovery rate <0.05. nNOS, neuronal nitric oxide synthase; PDGF, platelet-derived growth factor; TCA, tricarboxylic acid; GPCR, G protein-coupled receptor; FM, function and metabolism.
FIG. 3.
FIG. 3.
Comparison of plasma metabolome with transcriptome of I− versus ND (A) and I+ versus ND (B). The coclustering between the metabolomic changes and the transcripts of the corresponding muscle genes showed similar directional changes on the canonical pathways, although the statistical significance was different. The microarray/transcriptome data and the metabolome data are marked with an orange bar and blue bar, respectively. *Pathways used to build metabolic networks, as shown in Fig 4. EGF, epidermal growth factor; ERK, extracellular signal–related kinase; nNOS, neuronal nitric oxide synthase; PDGF, platelet-derived growth factor; TGF, transforming growth factor; GPCR, G protein-coupled receptor; LTD4, leukotriene receptor D4; HGF, hepatocyte growth factor; EMT, epithelial mesenchymal transition; VDR, vitamin D3 receptor; fMLP, N-formylated peptides like fMLP.
FIG. 3.
FIG. 3.
Comparison of plasma metabolome with transcriptome of I− versus ND (A) and I+ versus ND (B). The coclustering between the metabolomic changes and the transcripts of the corresponding muscle genes showed similar directional changes on the canonical pathways, although the statistical significance was different. The microarray/transcriptome data and the metabolome data are marked with an orange bar and blue bar, respectively. *Pathways used to build metabolic networks, as shown in Fig 4. EGF, epidermal growth factor; ERK, extracellular signal–related kinase; nNOS, neuronal nitric oxide synthase; PDGF, platelet-derived growth factor; TGF, transforming growth factor; GPCR, G protein-coupled receptor; LTD4, leukotriene receptor D4; HGF, hepatocyte growth factor; EMT, epithelial mesenchymal transition; VDR, vitamin D3 receptor; fMLP, N-formylated peptides like fMLP.
FIG. 4.
FIG. 4.
Integration of the metabolomics with transcriptomics data and their superimposition to build metabolic networks. A: Metabolic network of PPAR transcription pathway, which is connected to other metabolic processes such as lipid homeostasis, glucose, fatty acid metabolism, and inflammatory response. B: Network model of downstream of insulin-signaling pathways. The metabolites and the gene names shown in red are upregulated, and the same shown in blue are downregulated during insulin deficiency. B, binding; C, cleavage; CoA, coenzyme A; Erk, extracellular signal–related kinase; HPODE, hydroperoxylinoleic acid; IE, influence on expression; MAP, mitogen-activated protein; MAPK, MAP kinase; PDGF, platelet-derived growth factor; PI3K, phosphatidylinositol 3-kinase; PKA, cAMP-dependent protein kinase; PKB, protein kinase B; P-, dephosphorylation; RXR, retinoid X receptor; SREBP1c, sterol regulatory element–binding protein 1c; T, transformation; TGF, transforming growth factor; TR, transcription regulation; +P, phosphorylation; Z, catalysis; GPCR, G protein-coupled receptor; 15d-PGJ2, deoxy-delta prostaglandin J2; PDK/PDK1, 3-phosphoinositide-dependent protein kinase -1; ACACA, acetyl-CoA carboxylase; ACSL, acyl-CoA synthetase long-chain family members; ACLY, ATP citrate lyase; BCAA, branch chain amino acid; CISY, citrate synthase; DAG, diacylglycerol; ELOVL, elongation-of-very-long-chain-fatty acids; EMT, epithelial-mesenchymal transition; BEH, ethylene-bridged hybrid; 4E-BP1, eukaryotic translation initiation factor 4E binding protein 1; FADS1, fatty acid desaturase 1; FASN, fatty acid synthase; GSK3β, glycogen synthase kinase 3; GNAS, G protein αs- dependent adenylate cyclase; GRB2, growth factor receptor-bound protein 2; H-Ras, Harvey rat sarcoma viral oncogene homolog; HGF, hepatocyte growth factor; HXK, hexokinase; HSS, high-strength silica; HODE, hydroxyoctadecadienoic acid; INSIG2, insulin-induced gene 2; IRS-1 and IRS-2, insulin receptor substrates-1 and -2; TRIP, mediator complex subunit 1; MEK/MAP1, mitogen-activated protein kinase kinase 1; NCOA1, nuclear receptor coactivator 1; NRC1/SRC1, nuclear receptor coactivator 1; N-CoR, nuclear receptor corepressor; SMRT, nuclear receptor corepressors; NUDT1, nudix (nucleoside diphosphate-linked moiety X)-type motif 1; PtdIns(3,4,5)P3, phosphatidylinositol 3,4,5-triphosphate; P13K, phospatidylinositol 3-kinase; PtdIns(4,5)P2, phosphatidylinositol 4,5-biphosphate; PGE, prostaglandin; PTGIS, prostaglandin I2 (prostacyclin) synthase; PDGHS, prostaglandin-endoperoxide synthase 2 prostaglandin G/H synthase; COX2, cyclooxygenase 2; PDHA, pyruvate dehydrogenase (lipoamide) α1; QCs, quality controls; RARs, retinoic acid receptors; RXRA, retinoid X nuclear receptor (α; SHC, Src homology 2 domain containing transforming protein 1; SHP, small heterodimer partner; SOS, son of sevenless protein homologs 1 and 2; c-Raf-1, gene homolog 1; XIAP, X-linked inhibitor of apoptosis.
FIG. 4.
FIG. 4.
Integration of the metabolomics with transcriptomics data and their superimposition to build metabolic networks. A: Metabolic network of PPAR transcription pathway, which is connected to other metabolic processes such as lipid homeostasis, glucose, fatty acid metabolism, and inflammatory response. B: Network model of downstream of insulin-signaling pathways. The metabolites and the gene names shown in red are upregulated, and the same shown in blue are downregulated during insulin deficiency. B, binding; C, cleavage; CoA, coenzyme A; Erk, extracellular signal–related kinase; HPODE, hydroperoxylinoleic acid; IE, influence on expression; MAP, mitogen-activated protein; MAPK, MAP kinase; PDGF, platelet-derived growth factor; PI3K, phosphatidylinositol 3-kinase; PKA, cAMP-dependent protein kinase; PKB, protein kinase B; P-, dephosphorylation; RXR, retinoid X receptor; SREBP1c, sterol regulatory element–binding protein 1c; T, transformation; TGF, transforming growth factor; TR, transcription regulation; +P, phosphorylation; Z, catalysis; GPCR, G protein-coupled receptor; 15d-PGJ2, deoxy-delta prostaglandin J2; PDK/PDK1, 3-phosphoinositide-dependent protein kinase -1; ACACA, acetyl-CoA carboxylase; ACSL, acyl-CoA synthetase long-chain family members; ACLY, ATP citrate lyase; BCAA, branch chain amino acid; CISY, citrate synthase; DAG, diacylglycerol; ELOVL, elongation-of-very-long-chain-fatty acids; EMT, epithelial-mesenchymal transition; BEH, ethylene-bridged hybrid; 4E-BP1, eukaryotic translation initiation factor 4E binding protein 1; FADS1, fatty acid desaturase 1; FASN, fatty acid synthase; GSK3β, glycogen synthase kinase 3; GNAS, G protein αs- dependent adenylate cyclase; GRB2, growth factor receptor-bound protein 2; H-Ras, Harvey rat sarcoma viral oncogene homolog; HGF, hepatocyte growth factor; HXK, hexokinase; HSS, high-strength silica; HODE, hydroxyoctadecadienoic acid; INSIG2, insulin-induced gene 2; IRS-1 and IRS-2, insulin receptor substrates-1 and -2; TRIP, mediator complex subunit 1; MEK/MAP1, mitogen-activated protein kinase kinase 1; NCOA1, nuclear receptor coactivator 1; NRC1/SRC1, nuclear receptor coactivator 1; N-CoR, nuclear receptor corepressor; SMRT, nuclear receptor corepressors; NUDT1, nudix (nucleoside diphosphate-linked moiety X)-type motif 1; PtdIns(3,4,5)P3, phosphatidylinositol 3,4,5-triphosphate; P13K, phospatidylinositol 3-kinase; PtdIns(4,5)P2, phosphatidylinositol 4,5-biphosphate; PGE, prostaglandin; PTGIS, prostaglandin I2 (prostacyclin) synthase; PDGHS, prostaglandin-endoperoxide synthase 2 prostaglandin G/H synthase; COX2, cyclooxygenase 2; PDHA, pyruvate dehydrogenase (lipoamide) α1; QCs, quality controls; RARs, retinoic acid receptors; RXRA, retinoid X nuclear receptor (α; SHC, Src homology 2 domain containing transforming protein 1; SHP, small heterodimer partner; SOS, son of sevenless protein homologs 1 and 2; c-Raf-1, gene homolog 1; XIAP, X-linked inhibitor of apoptosis.

References

    1. Tessari P, Nosadini R, Trevisan R, et al. Defective suppression by insulin of leucine-carbon appearance and oxidation in type 1, insulin-dependent diabetes mellitus. Evidence for insulin resistance involving glucose and amino acid metabolism. J Clin Invest 1986;77:1797–1804
    1. Rizza RA, Jensen MD, Nair KS. Type I diabetes mellitus (insulin-dependent diabetes mellitus). In Handbook of Physiology New York, Oxford University Press, 2001, p. 1093–1114
    1. Melloul D, Marshak S, Cerasi E. Regulation of insulin gene transcription. Diabetologia 2002;45:309–326
    1. Sreekumar R, Halvatsiotis P, Schimke JC, Nair KS. Gene expression profile in skeletal muscle of type 2 diabetes and the effect of insulin treatment. Diabetes 2002;51:1913–1920
    1. Kimball SR, Jefferson LS. Cellular mechanisms involved in the action of insulin on protein synthesis. Diabetes Metab Rev 1988;4:773–787
    1. Jaleel A, Henderson GC, Madden BJ, et al. Identification of de novo synthesized and relatively older proteins: accelerated oxidative damage to de novo synthesized apolipoprotein A-1 in type 1 diabetes. Diabetes 2010;59:2366–2374
    1. Kahn CR. Diabetes. Causes of insulin resistance. Nature 1995;373:384–385
    1. Bain JR, Stevens RD, Wenner BR, Ilkayeva O, Muoio DM, Newgard CB. Metabolomics applied to diabetes research: moving from information to knowledge. Diabetes 2009;58:2429–2443
    1. Wang X, Jia S, Geoffrey R, Alemzadeh R, Ghosh S, Hessner MJ. Identification of a molecular signature in human type 1 diabetes mellitus using serum and functional genomics. J Immunol 2008;180:1929–1937
    1. Gehlenborg N, O’Donoghue SI, Baliga NS, et al. Visualization of omics data for systems biology. Nat Methods 2010;7(Suppl.):S56–S68
    1. Connor SC, Hansen MK, Corner A, Smith RF, Ryan TE. Integration of metabolomics and transcriptomics data to aid biomarker discovery in type 2 diabetes. Mol Biosyst 2010;6:909–921
    1. Zhang X, Wang Y, Hao F, et al. Human serum metabonomic analysis reveals progression axes for glucose intolerance and insulin resistance statuses. J Proteome Res 2009;8:5188–5195
    1. Zhang S, Nagana Gowda GA, Asiago V, Shanaiah N, Barbas C, Raftery D. Correlative and quantitative 1H NMR-based metabolomics reveals specific metabolic pathway disturbances in diabetic rats. Anal Biochem 2008;383:76–84
    1. Wang TJ, Larson MG, Vasan RS, et al. Metabolite profiles and the risk of developing diabetes. Nat Med 2011;17:448–453
    1. Zhao X, Fritsche J, Wang J, et al. Metabonomic fingerprints of fasting plasma and spot urine reveal human pre-diabetic metabolic traits. Metabolomics 2010;6:362–374
    1. Nair KS, Halliday D, Garrow JS. Increased energy expenditure in poorly controlled type 1 (insulin-dependent) diabetic patients. Diabetologia 1984;27:13–16
    1. Nair KS, Ford GC, Ekberg K, Fernqvist-Forbes E, Wahren J. Protein dynamics in whole body and in splanchnic and leg tissues in type I diabetic patients. J Clin Invest 1995;95:2926–2937
    1. Felig P, Wahren J, Sherwin R, Hendler R. Insulin, glucagon, and somatostatin in normal physiology and diabetes mellitus. Diabetes 1976;25:1091–1099
    1. Polonsky KS, Rubenstein AH. C-peptide as a measure of the secretion and hepatic extraction of insulin. Pitfalls and limitations. Diabetes 1984;33:486–494
    1. Kubota T, Kubota N, Kumagai H, et al. Impaired insulin signaling in endothelial cells reduces insulin-induced glucose uptake by skeletal muscle. Cell Metab 2011;13:294–307
    1. DeFronzo RA, Tobin JD, Andres R. Glucose clamp technique: a method for quantifying insulin secretion and resistance. Am J Physiol 1979;237:E214–E223
    1. Jaleel A, Klaus KA, Morse DM, et al. Differential effects of insulin deprivation and systemic insulin treatment on plasma protein synthesis in type 1 diabetic people. Am J Physiol Endocrinol Metab 2009;297:E889–E897
    1. Lanza IR, Zhang S, Ward LE, Karakelides H, Raftery D, Nair KS. Quantitative metabolomics by H-NMR and LC-MS/MS confirms altered metabolic pathways in diabetes. PLoS ONE 2010;5:e10538.
    1. Karakelides H, Asmann YW, Bigelow ML, et al. Effect of insulin deprivation on muscle mitochondrial ATP production and gene transcript levels in type 1 diabetic subjects. Diabetes 2007;56:2683–2689
    1. Nair KS, Halliday D, Griggs RC. Leucine incorporation into mixed skeletal muscle protein in humans. Am J Physiol 1988;254:E208–E213
    1. Chen J, Zhao X, Fritsche J, et al. Practical approach for the identification and isomer elucidation of biomarkers detected in a metabonomic study for the discovery of individuals at risk for diabetes by integrating the chromatographic and mass spectrometric information. Anal Chem 2008;80:1280–1289
    1. Sana TR, Fischer S, Wohlgemuth G, et al. Metabolomic and transcriptomic analysis of the rice response to the bacterial blight pathogen Xanthomonas oryzae pv. oryzae. Metabolomics 2010;6:451–465
    1. Asmann YW, Stump CS, Short KR, et al. Skeletal muscle mitochondrial functions, mitochondrial DNA copy numbers, and gene transcript profiles in type 2 diabetic and nondiabetic subjects at equal levels of low or high insulin and euglycemia. Diabetes 2006;55:3309–3319
    1. Balagopal P, Schimke JC, Ades P, Adey D, Nair KS. Age effect on transcript levels and synthesis rate of muscle MHC and response to resistance exercise. Am J Physiol Endocrinol Metab 2001;280:E203–E208
    1. Rubin DB. (Ed.) Multiple Imputation for Nonresponse in Surveys. New York, John Wiley & Sons, 1987, p. 75–107
    1. Schuierer S, Tranchevent LC, Dengler U, Moreau Y. Large-scale benchmark of Endeavour using MetaCore maps. Bioinformatics 2010;26:1922–1923
    1. Shaham O, Wei R, Wang TJ, et al. Metabolic profiling of the human response to a glucose challenge reveals distinct axes of insulin sensitivity. Mol Syst Biol 2008;4:214.
    1. Arisaka M, Arisaka O, Fukuda Y, Yamashiro Y. Prostaglandin metabolism in children with diabetes mellitus. I. Plasma prostaglandin E2, F2 alpha, TXB2, and serum fatty acid levels. J Pediatr Gastroenterol Nutr 1986;5:878–882
    1. Robertson RP. Arachidonic acid metabolite regulation of insulin secretion. Diabetes Metab Rev 1986;2:261–296
    1. Persaud SJ, Muller D, Belin VD, et al. The role of arachidonic acid and its metabolites in insulin secretion from human islets of langerhans. Diabetes 2007;56:197–203
    1. Robertson RP, Chen M. A role for prostaglandin E in defective insulin secretion and carbohydrate intolerance in diabetes mellitus. J Clin Invest 1977;60:747–753
    1. Robertson RP. Prostaglandins, glucose homeostasis, and diabetes mellitus. Annu Rev Med 1983;34:1–12
    1. Xie Z, Li H, Wang K, et al. Analysis of transcriptome and metabolome profiles alterations in fatty liver induced by high-fat diet in rat. Metabolism 2010;59:554–560
    1. Ma K, Nunemaker CS, Wu R, Chakrabarti SK, Taylor-Fishwick DA, Nadler JL. 12-lipoxygenase products reduce insulin secretion and beta-cell viability in human islets. J Clin Endocrinol Metab 2010;95:887–893
    1. Li JH, Chou CL, Li B, et al. A selective EP4 PGE2 receptor agonist alleviates disease in a new mouse model of X-linked nephrogenic diabetes insipidus. J Clin Invest 2009;119:3115–3126
    1. Takahashi HK, Zhang J, Mori S, et al. Prostaglandin E2 inhibits advanced glycation end product-induced adhesion molecule expression on monocytes, cytokine production, and lymphocyte proliferation during human mixed lymphocyte reaction. J Pharmacol Exp Ther 2010;334:964–972
    1. Metz S, VanRollins M, Strife R, Fujimoto W, Robertson RP. Lipoxygenase pathway in islet endocrine cells. Oxidative metabolism of arachidonic acid promotes insulin release. J Clin Invest 1983;71:1191–1205
    1. Filep JG, Zouki C, Petasis NA, Hachicha M, Serhan CN. Anti-inflammatory actions of lipoxin A(4) stable analogs are demonstrable in human whole blood: modulation of leukocyte adhesion molecules and inhibition of neutrophil-endothelial interactions. Blood 1999;94:4132–4142
    1. Gambert SR, Garthwaite TL, Pontzer CH, et al. Running elevates plasma beta-endorphin immunoreactivity and ACTH in untrained human subjects. Proc Soc Exp Biol Med 1981;168:1–4
    1. Carr DB, Bullen BA, Skrinar GS, et al. Physical conditioning facilitates the exercise-induced secretion of beta-endorphin and beta-lipotropin in women. N Engl J Med 1981;305:560–563
    1. Li LO, Hu YF, Wang L, Mitchell M, Berger A, Coleman RA. Early hepatic insulin resistance in mice: a metabolomics analysis. Mol Endocrinol 2010;24:657–666
    1. Le Bouter S, Rodriguez M, Guigal-Stephan N, et al. Coordinate transcriptomic and metabolomic effects of the insulin sensitizer rosiglitazone on fundamental metabolic pathways in liver, soleus muscle, and adipose tissue in diabetic db/db mice. PPAR Res 10 October 2010 [Epub ahead of print]
    1. Freyse EJ, Fischer U, Knospe S, Ford GC, Nair KS. Differences in protein and energy metabolism following portal versus systemic administration of insulin in diabetic dogs. Diabetologia 2006;49:543–551
    1. Charlton MR, Adey DB, Nair KS. Evidence for a catabolic role of glucagon during an amino acid load. J Clin Invest 1996;98:90–99
    1. Charlton M, Nair KS. Protein metabolism in insulin-dependent diabetes mellitus. J Nutr 1998;128(Suppl.):323S–327S

Source: PubMed

3
Subscribe