Plasma metabolomic profiles reflective of glucose homeostasis in non-diabetic and type 2 diabetic obese African-American women

Oliver Fiehn, W Timothy Garvey, John W Newman, Kerry H Lok, Charles L Hoppel, Sean H Adams, Oliver Fiehn, W Timothy Garvey, John W Newman, Kerry H Lok, Charles L Hoppel, Sean H Adams

Abstract

Insulin resistance progressing to type 2 diabetes mellitus (T2DM) is marked by a broad perturbation of macronutrient intermediary metabolism. Understanding the biochemical networks that underlie metabolic homeostasis and how they associate with insulin action will help unravel diabetes etiology and should foster discovery of new biomarkers of disease risk and severity. We examined differences in plasma concentrations of >350 metabolites in fasted obese T2DM vs. obese non-diabetic African-American women, and utilized principal components analysis to identify 158 metabolite components that strongly correlated with fasting HbA1c over a broad range of the latter (r = -0.631; p<0.0001). In addition to many unidentified small molecules, specific metabolites that were increased significantly in T2DM subjects included certain amino acids and their derivatives (i.e., leucine, 2-ketoisocaproate, valine, cystine, histidine), 2-hydroxybutanoate, long-chain fatty acids, and carbohydrate derivatives. Leucine and valine concentrations rose with increasing HbA1c, and significantly correlated with plasma acetylcarnitine concentrations. It is hypothesized that this reflects a close link between abnormalities in glucose homeostasis, amino acid catabolism, and efficiency of fuel combustion in the tricarboxylic acid (TCA) cycle. It is speculated that a mechanism for potential TCA cycle inefficiency concurrent with insulin resistance is "anaplerotic stress" emanating from reduced amino acid-derived carbon flux to TCA cycle intermediates, which if coupled to perturbation in cataplerosis would lead to net reduction in TCA cycle capacity relative to fuel delivery.

Conflict of interest statement

Competing Interests: The authors have declared that no competing interests exist.

Figures

Figure 1. Relative plasma concentrations of leucine…
Figure 1. Relative plasma concentrations of leucine (A) and valine (B) are increased in type 2 diabetic African-American obese women.
Results are expressed as total quantifier peak height percent of all summed plasma amino acid (AA) peak heights, i.e., as % of total AA. Bars represent the mean ± SEM for n = 12 and n = 43 non-diabetic and diabetic subjects, respectively. **p

Figure 2. Plasma branched chain amino acid…

Figure 2. Plasma branched chain amino acid (BCAA) concentrations are correlated with fasting HbA1c% and…

Figure 2. Plasma branched chain amino acid (BCAA) concentrations are correlated with fasting HbA1c% and plasma acylcarnitine concentrations in non-obese (yellow circles) and type 2 diabetic (blue circles) obese women.
Shown are correlations between plasma leucine (A,C) and valine (B,D) enrichments (% of their concentrations relative to total measured amino acid concentrations; see Figure 1 legend) with fasting blood HbA1c (top panels) or plasma acetylcarnitine concentration (bottom panels). Pearson's r and p values for the correlations are given within the figures.

Figure 3. Relative plasma concentrations of propionylcarnitine…

Figure 3. Relative plasma concentrations of propionylcarnitine (green symbols) were reduced concurrent with increases in…

Figure 3. Relative plasma concentrations of propionylcarnitine (green symbols) were reduced concurrent with increases in the relative plasma concentrations of a precursor, valine, with increasing plasma acetylcarnitine concentration in obese African-American women.
Symbols represent individuals included in metabolomics studies described in the text.

Figure 4. Separation of non-diabetics (yellow circles)…

Figure 4. Separation of non-diabetics (yellow circles) from type 2 diabetics (blue circles) due to…

Figure 4. Separation of non-diabetics (yellow circles) from type 2 diabetics (blue circles) due to variance in plasma metabolite factors.
Principal components analysis (PCA) in dimensions 1 and 2 using 158 metabolites illustrates differential distribution of diabetic and non-diabetic subjects along the PC1 (X axis) and PC2 (Y axis) axes, with each symbol plotting PC1-PC2 scores for a given subject. Metabolite components whose variance-derived loadings values contributed most to the PC separations scores are listed in Supplemental Table S3. Summarizing the loading contributions from known compounds, elevated fatty acids and the enrichment of the amino acid pool with branched chain AAs segregated diabetics from controls in PC1, while elevations In various carbohydrates as well as a suite of amino acids separated diabetics from controls in PC2.

Figure 5. Correlation between PC1 scores and…

Figure 5. Correlation between PC1 scores and blood HbA1c% in non-diabetic (yellow circles) and type…

Figure 5. Correlation between PC1 scores and blood HbA1c% in non-diabetic (yellow circles) and type 2 diabetic (blue circles) obese women.
PC1 scores derived from PCA analysis (see text and Figure 4 legend) were used for correlation to a marker of blood sugar control. Pearson's r and p values for the correlation is given within the figure.
Figure 2. Plasma branched chain amino acid…
Figure 2. Plasma branched chain amino acid (BCAA) concentrations are correlated with fasting HbA1c% and plasma acylcarnitine concentrations in non-obese (yellow circles) and type 2 diabetic (blue circles) obese women.
Shown are correlations between plasma leucine (A,C) and valine (B,D) enrichments (% of their concentrations relative to total measured amino acid concentrations; see Figure 1 legend) with fasting blood HbA1c (top panels) or plasma acetylcarnitine concentration (bottom panels). Pearson's r and p values for the correlations are given within the figures.
Figure 3. Relative plasma concentrations of propionylcarnitine…
Figure 3. Relative plasma concentrations of propionylcarnitine (green symbols) were reduced concurrent with increases in the relative plasma concentrations of a precursor, valine, with increasing plasma acetylcarnitine concentration in obese African-American women.
Symbols represent individuals included in metabolomics studies described in the text.
Figure 4. Separation of non-diabetics (yellow circles)…
Figure 4. Separation of non-diabetics (yellow circles) from type 2 diabetics (blue circles) due to variance in plasma metabolite factors.
Principal components analysis (PCA) in dimensions 1 and 2 using 158 metabolites illustrates differential distribution of diabetic and non-diabetic subjects along the PC1 (X axis) and PC2 (Y axis) axes, with each symbol plotting PC1-PC2 scores for a given subject. Metabolite components whose variance-derived loadings values contributed most to the PC separations scores are listed in Supplemental Table S3. Summarizing the loading contributions from known compounds, elevated fatty acids and the enrichment of the amino acid pool with branched chain AAs segregated diabetics from controls in PC1, while elevations In various carbohydrates as well as a suite of amino acids separated diabetics from controls in PC2.
Figure 5. Correlation between PC1 scores and…
Figure 5. Correlation between PC1 scores and blood HbA1c% in non-diabetic (yellow circles) and type 2 diabetic (blue circles) obese women.
PC1 scores derived from PCA analysis (see text and Figure 4 legend) were used for correlation to a marker of blood sugar control. Pearson's r and p values for the correlation is given within the figure.

References

    1. Kendall DM, Cuddihy RM, Bergenstal RM. Clinical application of incretin-based therapy: therapeutic potential, patient selection and clinical use. Am J Med. 2009;122:S37–50.
    1. McGarry JD. What if Minkowski had been ageusic? An alternative angle on diabetes. Science. 1992;258:766–770.
    1. Kusminski CM, Shetty S, Orci L, Unger RH, Scherer PE. Diabetes and apoptosis: lipotoxicity. Apoptosis. 2009;14:1484–1495.
    1. Marchesini G, Bianchi GP, Vilstrup H, Capelli M, Zoli M, et al. Elimination of infused branched-chain amino-acids from plasma of patients with non-obese type 2 diabetes mellitus. Clin Nutr. 1991;10:105–113.
    1. Wijekoon EP, Skinner C, Brosnan ME, Brosnan JT. Amino acid metabolism in the Zucker diabetic fatty rat: effects of insulin resistance and of type 2 diabetes. Can J Physiol Pharmacol. 2004;82:506–514.
    1. She P, Van Horn C, Reid T, Hutson SM, Cooney RN, et al. Obesity-related elevations in plasma leucine are associated with alterations in enzymes involved in branched-chain amino acid metabolism. Am J Physiol Endocrinol Metab. 2007;293:E1552–E1563.
    1. Kuzuya T, Katano Y, Nakano I, Hirooka Y, Itoh A, et al. Regulation of branched-chain amino acid catabolism in rat models for spontaneous type 2 diabetes mellitus. Biochem Biophys Res Commun. 2008;373:94–98.
    1. Newgard CB, An J, Bain JR, Muehlbauer MJ, Stevens RD, et al. A branched-chain amino acid-related metabolic signature that differentiates obese and lean humans and contributes to insulin resistance. Cell Metab. 2009;9:311–326.
    1. Doisaki M, Katano Y, Nakano I, Hirooka Y, Itoh A, et al. Regulation of hepatic branched-chain alpha-keto acid dehydrogenase kinase in a rat model for type 2 diabetes mellitus at different stages of the disease. Biochem Biophys Res Commun. 2010;393:303–307.
    1. Jacob S, Machann J, Rett K, Brechtel K, Volk A, et al. Association of increased intramyocellular lipid content with insulin resistance in lean nondiabetic offspring of type 2 diabetic subjects. Diabetes. 1999;48:1113–1119.
    1. Krssak M, Falk Petersen K, Dresner A, DiPietro L, Vogel SM, et al. Intramyocellular lipid concentrations are correlated with insulin sensitivity in humans: a 1H NMR spectroscopy study. Diabetologia. 1999;42:113–116.
    1. Dobbins RL, Szczepaniak LS, Bentley B, Esser V, Myhill J, et al. Prolonged inhibition of muscle carnitine palmitoyltransferase-1 promotes intramyocellular lipid accumulation and insulin resistance in rats. Diabetes. 2001;50:123–130.
    1. McGarry JD. Banting lecture 2001: dysregulation of fatty acid metabolism in the etiology of type 2 diabetes. Diabetes. 2002;51:7–18.
    1. Mingrone G, Rosa G, Greco AV, Manco M, Vega N, et al. Intramyocitic lipid accumulation and SREBP-1c expression are related to insulin resistance and cardiovascular risk in morbid obesity. Atherosclerosis. 2003;170:155–161.
    1. He J, Goodpaster BH, Kelley DE. Effects of weight loss and physical activity on muscle lipid content and droplet size. Obes Res. 2004;12:761–769.
    1. Goodpaster BH, Brown NF. Skeletal muscle lipid and its association with insulin resistance: what is the role for exercise? Exerc Sport Sci Rev. 2005;33:150–154.
    1. Lara-Castro C, Newcomer BR, Rowell J, Wallace P, Shaughnessy SM, et al. Effects of short-term very low-calorie diet on intramyocellular lipid and insulin sensitivity in nondiabetic and type 2 diabetic subjects. Metabolism. 2008;57:1–8.
    1. Nagle CA, Klett EL, Coleman RA. Hepatic triacylglycerol accumulation and insulin resistance. J Lipid Res. 2009;50(Suppl):S74–79.
    1. Heilbronn L, Smith SR, Ravussin E. Failure of fat cell proliferation, mitochondrial function and fat oxidation results in ectopic fat storage, insulin resistance and type II diabetes mellitus. Int J Obes Relat Metab Disord. 2004;28(Suppl 4):S12–21.
    1. Lingvay I, Esser V, Legendre JL, Price AL, Wertz KM, et al. Noninvasive quantification of pancreatic fat in humans. J Clin Endocrinol Metab. 2009;94:4070–4076.
    1. Kelley DE. Skeletal muscle fat oxidation: timing and flexibility are everything. J Clin Invest. 2005;115:1699–1702.
    1. Koves TR, Ussher JR, Noland RC, Slentz D, Mosedale M, et al. Mitochondrial overload and incomplete fatty acid oxidation contribute to skeletal muscle insulin resistance. Cell Metab. 2008;7:45–56.
    1. Adams SH, Hoppel CL, Lok KH, Zhao L, Wong SW, et al. Plasma acylcarnitine profiles suggest incomplete long-chain fatty acid beta-oxidation and altered tricarboxylic acid cycle activity in type 2 diabetic African-American women. J Nutr. 2009;139:1073–1081.
    1. Shulman GI. Unraveling the cellular mechanism of insulin resistance in humans: new insights from magnetic resonance spectroscopy. Physiology (Bethesda) 2004;19:183–190.
    1. Muoio DM, Newgard CB. Mechanisms of disease: molecular and metabolic mechanisms of insulin resistance and beta-cell failure in type 2 diabetes. Nat Rev Mol Cell Biol. 2008;9:193–205.
    1. Summers SA. Sphingolipids and insulin resistance: the five Ws. Curr Opin Lipidol. 2010;21:128–135.
    1. Argyropoulos G, Brown AM, Willi SM, Zhu J, He Y, et al. Effects of mutations in the human uncoupling protein 3 gene on the respiratory quotient and fat oxidation in severe obesity and type 2 diabetes. J Clin Invest. 1998;102:1345–1351.
    1. McLean DC, Jr, Spruill I, Argyropoulos G, Page GP, Shriver MD, et al. Mitochondrial DNA (mtDNA) haplotypes reveal maternal population genetic affinities of Sea Island Gullah-speaking African Americans. Am J Phys Anthropol. 2005;127:427–438.
    1. Fiehn O Kind T Metabolite profiling in blood plasma. Methods in Molecular Biology. 2007;358:3–17.
    1. Sale MM, Lu L, Spruill IJ, Fernandes JK, Lok KH, et al. Genome-wide linkage scan in Gullah-speaking African American families with type 2 diabetes: the Sea Islands Genetic African American Registry (Project SuGAR). Diabetes. 2009;58:260–267.
    1. Divers J, Sale MM, Lu L, Chen WM, Lok KH, et al. The genetic architecture of lipoprotein subclasses in Gullah-speaking African American families enriched for type 2 diabetes: the Sea Islands Genetic African American Registry (Project SuGAR). J Lipid Res. 2010;51:586–597.
    1. Fiehn O, Wohlgemuth G, Scholz M. Setup and annotation of metabolomic experiments by integrating biological and mass spectrometric metadata. Lecture Notes in Computer Science. 2005;3615:224–239.
    1. Scholz M, Fiehn O. SetupX–a public study design database for metabolomic projects. Biocomputing 2007, Proceedings of the Pacific Symposium. 2007:169–180.
    1. Benjamini Y, Hochberg Y. Controlling false discovery rate: a practical and powerful approach to multiple testing. J Royal Statistical Soc., Series B (Methodological), 1995;57:289–300.
    1. Randle PJ, Garland PB, Hales CN, Newsholme EA. The glucose fatty-acid cycle. Its role in insulin sensitivity and the metabolic disturbances of diabetes mellitus. Lancet. 1963;1:785–789.
    1. Petersen KF, Dufour S, Befroy D, Garcia R, Shulman GI. Impaired mitochondrial activity in the insulin-resistant offspring of patients with type 2 diabetes. N Engl J Med. 2004;350:664–671.
    1. Morino K, Falk Petersen K, Dufour S, Befroy D, Frattini J, et al. Reduced mitochondrial density and increased IRS-1 serine phosphorylation in muscle of insulin-resistant offspring of type 2 diabetic parents. J Clin Invest. 2005;115:3587–3593.
    1. Befroy DE, Falk Petersen K, Dufour S, Mason GF, de Graaf RA, et al. Impaired mitochondrial substrate oxidation in muscle of insulin-resistant offspring of type 2 diabetic patients. Diabetes. 2007;56:1376–1381.
    1. Ritov VB, Menshikova EV, He J, Ferrell RE, Goodpaster BH, et al. Deficiency of subsarcolemmal mitochondria in obesity and type 2 diabetes. Diabetes. 2005;54:8–14.
    1. Mogensen M, Sahlin K, Fernstrom M, Glintborg D, Vind BF, et al. Mitochondrial respiration is decreased in skeletal muscle of patients with type 2 diabetes. Diabetes. 2007;56:1592–1599.
    1. Kawasaki T, Akanuma H, Yamanouchi T. Increased fructose concentrations in blood and urine in patients with diabetes. Diabetes Care. 2002;25:353–357.
    1. Salek RM, Maguire ML, Bentley E, Rubtsov DV, Hough T, et al. A metabolomic comparison of urinary changes in type 2 diabetes in mouse, rat, and human. Physiol Genomics. 2007;29:99–108.
    1. Li X, Xu Z, Lu X, Yang X, Yin P, et al. Comprehensive two-dimensional gas chromatography/time-of-flight mass spectrometry for metabonomics: Biomarker discovery for diabetes mellitus. Anal Chim Acta. 2009;633:257–262.
    1. Zhang X, Wang Y, Hao F, Zhou X, Han X, et al. Human serum metabonomic analysis reveals progression axes for glucose intolerance and insulin resistance statuses. J Proteome Res. 2009;8:5188–5195.
    1. Zhao X, Fritsche J, Wang J, Chen J, Rittig K, et al. Metabonomic fingerprints of fasting plasma and spot urine reveal human pre-diabetic metabolic traits. Metabolomics. 2010;6:362–374.
    1. Gall WE, Beebe K, Lawton KA, Adam KP, Mitchell MW, et al. alpha-hydroxybutyrate is an early biomarker of insulin resistance and glucose intolerance in a nondiabetic population. PLoS One. 2010;5:e10883.
    1. Elshorbagy AK, Refsum H, Smith AD, Graham IM. The association of plasma cysteine and gamma-glutamyltransferase with BMI and obesity. Obesity. 2009;17:1435–1440.
    1. Bajotto G, Murakami T, Nagasaki M, Sato Y, Shimomura Y. Decreased enzyme activity and contents of hepatic branched-chain alpha-keto acid dehydrogenase complex subunits in a rat model for type 2 diabetes mellitus. Metabolism. 2009;58:1489–1495.
    1. Huffman KM, Shah SH, Stevens RD, Bain JR, Muehlbauer M, et al. Relationships between circulating metabolic intermediates and insulin action in overweight to obese, inactive men and women. Diabetes Care. 2009;32:1678–1683.
    1. Zemel MB. The role of dairy foods in weight management. J Am Coll Nutr. 2005;24:537S–546S.
    1. Nairizi A, She P, Vary TC, Lynch CJ. Leucine supplementation of drinking water does not alter susceptibility to diet-induced obesity in mice. J Nutr. 2009;139:715–719.
    1. Zhang Y, Guo K, LeBlanc RE, Loh D, Schwartz GJ, et al. Increasing dietary leucine intake reduces diet-induced obesity and improves glucose and cholesterol metabolism in mice via multimechanisms. Diabetes. 2007;56:1647–1654.
    1. Layman DK, Clifton P, Gannon MC, Krauss RM, Nuttall FQ. Protein in optimal health: heart disease and type 2 diabetes. Am J Clin Nutr. 2008;87:1571S–1575S.
    1. Devkota S, Layman DK. Protein metabolic roles in treatment of obesity. Curr Opin Clin Nutr Metab Care. 2010;13:403–407.
    1. Pereira S, Marliss EB, Morais JA, Chevalier S, Gougeon R. Insulin resistance of protein metabolism in type 2 diabetes. Diabetes. 2008;57:56–63.
    1. Guillet C, Delcourt I, Rance M, Giraudet C, Walrand S, et al. Changes in basal and insulin and amino acid response of whole body and skeletal muscle proteins in obese men. J Clin Endocrinol Metab. 2009;94:3044–3050.
    1. Gougeon R, Pencharz PB, Marliss EB. Effect of NIDDM on the kinetics of whole-body protein metabolism. Diabetes. 1994;43:318–328.
    1. Staten MA, Matthews DE, Bier DM. Leucine metabolism in type II diabetes mellitus. Diabetes. 1986;35:1249–1253.
    1. Umpleby AM, Scobie IN, Boroujerdi MA, Carson ER, Sonksen PH. Diurnal variation in glucose and leucine metabolism in non-insulin-dependent diabetes. Diabetes Res Clin Pract. 1990;9:89–96.
    1. Hoffer LJ, Taveroff A, Hamadeh MJ. Dietary protein restriction alters glucose but not protein metabolism in non-insulin-dependent diabetes mellitus. Metabolism. 1998;47:1145–1151.
    1. Halvatsiotis PG, Turk D, Alzaid A, Dinneen S, Rizza RA, et al. Insulin effect on leucine kinetics in type 2 diabetes mellitus. Diabetes Nutr Metab. 2002;15:136–142.
    1. Halvatsiotis P, Short KR, Bigelow M, Nair KS. Synthesis rate of muscle proteins, muscle functions, and amino acid kinetics in type 2 diabetes. Diabetes. 2002;51:2395–2404.
    1. Gougeon R, Morais JA, Chevalier S, Pereira S, Lamarche M, et al. Determinants of whole-body protein metabolism in subjects with and without type 2 diabetes. Diabetes Care. 2008;31:128–133.
    1. Fois A, Cioni M, Balestri P, Bartalini G, Baumgartner R, et al. Biotinidase deficiency: metabolites in CSF. J Inherit Metab Dis. 1986;9:284–285.
    1. Baez-Saldana A, Zendejas-Ruiz I, Revilla-Monsalve C, Islas-Andrade S, Cardenas A, et al. Effects of biotin on pyruvate carboxylase, acetyl-CoA carboxylase, propionyl-CoA carboxylase, and markers for glucose and lipid homeostasis in type 2 diabetic patients and nondiabetic subjects. Am J Clin Nutr. 2004;79:238–243.
    1. He J, Watkins S, Kelley DE. Skeletal muscle lipid content and oxidative enzyme activity in relation to muscle fiber type in type 2 diabetes and obesity. Diabetes. 2001;50:817–823.
    1. Kelley DE, He J, Menshikova EV, Ritov VB. Dysfunction of mitochondria in human skeletal muscle in type 2 diabetes. Diabetes. 2002;51:2944–2950.
    1. Holloszy JO. Skeletal muscle “mitochondrial deficiency” does not mediate insulin resistance. Am J Clin Nutr. 2009;89:463S–466S.
    1. Holloway GP, Bonen A, Spriet LL. Regulation of skeletal muscle mitochondrial fatty acid metabolism in lean and obese individuals. Am J Clin Nutr. 2009;89:455S–462S.
    1. Zoppi CC, Silveira LR, Oliveira CA, Boschero AC, Curi R, et al. Insulin release, peripheral insulin resistance and muscle function in protein malnutrition: a role of tricarboxylic acid cycle anaplerosis. Br J Nutr. 2010;103:1237–1250.
    1. Seifert EL, Fiehn O, Bezaire V, Bickel DR, Wohlgemuth G, et al. Long-chain fatty acid combustion rate is associated with unique metabolite profiles in skeletal muscle mitochondria. PLoS One. 2010;5:e9834.
    1. Pecqueur C, Couplan E, Bouillaud F, Ricquier D. Genetic and physiological analysis of the role of uncoupling proteins in human energy homeostasis. J Mol Med. 2001;79:48–56.

Source: PubMed

3
Se inscrever