Skip to content
Licensed Unlicensed Requires Authentication Published by De Gruyter July 30, 2021

Urine organic acids may be useful biomarkers for metabolic syndrome and its components in Korean adults

  • Ji-Hee Haam , Yun Kyong Lee , Eunkyung Suh , Sang-Woon Choi , Hyejin Chun and Young-Sang Kim ORCID logo EMAIL logo

Abstract

Objectives

Although metabolic syndrome (MetS) and its components are defined clinically, those with MetS may have various derangements in metabolic pathways. Thus, this study aimed to evaluate the traits of urine organic acid metabolites indicating the metabolic intermediates of the pathways in the subjects with MetS.

Methods

This cross-sectional study included 246 men and 283 women in a hospital health check-up setting. Urine organic acid metabolites were assayed via high-performance liquid chromatography–mass spectrometry analyses. A high level of each metabolite was defined as the fifth quintile of the distribution.

Results

The subjects with MetS had high levels of pyruvate, α-ketoglutarate, α-ketoisovalerate, α-ketoisocaproate, formiminoglutamate, and quinolinate (odds ratios from 1.915 to 2.809 in logistic models adjusted for age and sex). Among the metabolites, pyruvate, formiminoglutamate, and quinolinate were not independent of homeostatic model assessment of insulin resistance (HOMA2-IR). Several metabolites were associated with one or more components of MetS and HOMA2-IR.

Conclusions

Urine organic acid metabolites in MetS are characterized in altered carbohydrate and amino acid metabolism. MetS shared some traits in insulin resistance. These findings may promote the understanding of the pathophysiology of MetS.


Corresponding author: Young-Sang Kim, MD, PhD, Department of Family Medicine, CHA Bundang Medical Center, CHA University, 59 Yatap-ro, Bundang-gu, Seongnam-si, Gyeonggi-do, 13496, Korea. Phone: +82317805360, Fax: +82317805944, E-mail:

Award Identifier / Grant number: 2020-02-020

Acknowledgments

The authors thank to all staffs to help recruiting the subjects, conducting the measurements, and documenting the data in Chaum Life Center. The authors would like to thank Enago for the English language review.

  1. Research funding: This study was supported by Yuhan Co, Ltd. [grant number 2020-02-020] and Academic Fund from CHA university.

  2. Author contributions: Study concept and design: JH Haam, YS Kim; acquisition of data: JH Haam, YK Lee, E Suh; analysis and interpretation of data: YS Kim, JH Haam, SW Choi; drafting of the manuscript: YS Kim, JH Haam; critical revision of the manuscript for important intellectual content: YK Lee, E Suh, SW Choi, H Chun; statistical analysis: YS Kim, JH Haam, YK Lee, E Suh, SW Choi; obtained funding: YS Kim; administrative, technical, or material support: H Chun, E Suh; study supervision: YS Kim, SW Choi. All authors have accepted responsibility for the entire content of this manuscript and approved its submission.

  3. Competing interests: Authors state no conflict of interest.

  4. Informed consent: Patient consent was waived due to the retrospective design.

  5. Ethical approval: The study was conducted according to the guidelines of the Declaration of Helsinki, and approved by the Institutional Review Board of CHA Bundang Medical Center (2018-07-026).

References

1. Wilson, PW, D’Agostino, RB, Parise, H, Sullivan, L, Meigs, JB. Metabolic syndrome as a precursor of cardiovascular disease and type 2 diabetes mellitus. Circulation 2005;112:3066–72. https://doi.org/10.1161/circulationaha.105.539528.Search in Google Scholar

2. Pollex, RL, Hegele, RA. Genetic determinants of the metabolic syndrome. Nat Clin Pract Cardiovasc Med 2006;3:482–9. https://doi.org/10.1038/ncpcardio0638.Search in Google Scholar PubMed

3. Edwardson, CL, Gorely, T, Davies, MJ, Gray, LJ, Khunti, K, Wilmot, EG, et al.. Association of sedentary behaviour with metabolic syndrome: a meta-analysis. PloS One 2012;7:e34916. https://doi.org/10.1371/journal.pone.0034916.Search in Google Scholar PubMed PubMed Central

4. Gluvic, Z, Zaric, B, Resanovic, I, Obradovic, M, Mitrovic, A, Radak, D, et al.. Link between metabolic syndrome and insulin resistance. Curr Vasc Pharmacol 2017;15:30–9. https://doi.org/10.2174/1570161114666161007164510.Search in Google Scholar PubMed

5. Sonnenberg, GE, Krakower, GR, Kissebah, AH. A novel pathway to the manifestations of metabolic syndrome. Obes Res 2004;12:180–6. https://doi.org/10.1038/oby.2004.24.Search in Google Scholar PubMed

6. Sypniewska, G. Pro-inflammatory and prothrombotic factors and metabolic syndrome. EJIFCC 2007;18:39–46.Search in Google Scholar

7. Brownlee, M. Biochemistry and molecular cell biology of diabetic complications. Nature 2001;414:813–20. https://doi.org/10.1038/414813a.Search in Google Scholar PubMed

8. Maddux, BA, See, W, Lawrence, JCJr., Goldfine, AL, Goldfine, ID, Evans, JL. Protection against oxidative stress-induced insulin resistance in rat L6 muscle cells by mircomolar concentrations of alpha-lipoic acid. Diabetes 2001;50:404–10. https://doi.org/10.2337/diabetes.50.2.404.Search in Google Scholar PubMed

9. Rudich, A, Tirosh, A, Potashnik, R, Hemi, R, Kanety, H, Bashan, N. Prolonged oxidative stress impairs insulin-induced GLUT4 translocation in 3T3-L1 adipocytes. Diabetes 1998;47:1562–9. https://doi.org/10.2337/diabetes.47.10.1562.Search in Google Scholar PubMed

10. Matsuoka, T, Kajimoto, Y, Watada, H, Kaneto, H, Kishimoto, M, Umayahara, Y, et al.. Glycation-dependent, reactive oxygen species-mediated suppression of the insulin gene promoter activity in HIT cells. J Clin Invest 1997;99:144–50. https://doi.org/10.1172/jci119126.Search in Google Scholar

11. Nakazono, K, Watanabe, N, Matsuno, K, Sasaki, J, Sato, T, Inoue, M. Does superoxide underlie the pathogenesis of hypertension? Proc Natl Acad Sci U S A 1991;88:10045–8. https://doi.org/10.1073/pnas.88.22.10045.Search in Google Scholar PubMed PubMed Central

12. Ohara, Y, Peterson, TE, Harrison, DG. Hypercholesterolemia increases endothelial superoxide anion production. J Clin Invest 1993;91:2546–51. https://doi.org/10.1172/jci116491.Search in Google Scholar

13. Ntzouvani, A, Nomikos, T, Panagiotakos, D, Fragopoulou, E, Pitsavos, C, McCann, A, et al.. Amino acid profile and metabolic syndrome in a male Mediterranean population: a cross-sectional study. Nutr Metabol Cardiovasc Dis 2017;27:1021–30. https://doi.org/10.1016/j.numecd.2017.07.006.Search in Google Scholar PubMed

14. Sun, J, Beger, RD, Schnackenberg, LK. Metabolomics as a tool for personalizing medicine: 2012 update. Personalized Med 2013;10:149–61. https://doi.org/10.2217/pme.13.8.Search in Google Scholar PubMed

15. Ramakrishanan, N, Denna, T, Devaraj, S, Adams-Huet, B, Jialal, I. Exploratory lipidomics in patients with nascent Metabolic Syndrome. J Diabet Complicat 2018;32:791–4. https://doi.org/10.1016/j.jdiacomp.2018.05.014.Search in Google Scholar PubMed

16. Reddy, P, Leong, J, Jialal, I. Amino acid levels in nascent metabolic syndrome: a contributor to the pro-inflammatory burden. J Diabet Complicat 2018;32:465–9. https://doi.org/10.1016/j.jdiacomp.2018.02.005.Search in Google Scholar PubMed

17. Gallagher, RC, Pollard, L, Scott, AI, Huguenin, S, Goodman, S, Sun, Q, et al.. Laboratory analysis of organic acids, 2018 update: a technical standard of the American College of Medical Genetics and Genomics (ACMG). Genet Med 2018;20:683–91. https://doi.org/10.1038/gim.2018.45.Search in Google Scholar PubMed

18. Scolamiero, E, Cozzolino, C, Albano, L, Ansalone, A, Caterino, M, Corbo, G, et al.. Targeted metabolomics in the expanded newborn screening for inborn errors of metabolism. Mol Biosyst 2015;11:1525–35. https://doi.org/10.1039/c4mb00729h.Search in Google Scholar PubMed

19. Tai, ES, Tan, ML, Stevens, RD, Low, YL, Muehlbauer, MJ, Goh, DL, et al.. Insulin resistance is associated with a metabolic profile of altered protein metabolism in Chinese and Asian-Indian men. Diabetologia 2010;53:757–67. https://doi.org/10.1007/s00125-009-1637-8.Search in Google Scholar PubMed PubMed Central

20. Qiu, C, Enquobahrie, DA, Frederick, IO, Sorensen, TK, Fernandez, MA, David, RM, et al.. Early pregnancy urinary biomarkers of fatty acid and carbohydrate metabolism in pregnancies complicated by gestational diabetes. Diabetes Res Clin Pract 2014;104:393–400. https://doi.org/10.1016/j.diabres.2014.03.001.Search in Google Scholar PubMed PubMed Central

21. Lteif, AA, Han, K, Mather, KJ. Obesity, insulin resistance, and the metabolic syndrome: determinants of endothelial dysfunction in whites and blacks. Circulation 2005;112:32–8. https://doi.org/10.1161/circulationaha.104.520130.Search in Google Scholar PubMed

22. Murphy, M, Srivastava, R, Deans, K. Clinical Biochemistry E-Book: An Illustrated Colour Text, 6th ed.. Philadelphia: Elsevier Health Sciences; 2018.Search in Google Scholar

23. Rahimi, Y, Camporez, JP, Petersen, MC, Pesta, D, Perry, RJ, Jurczak, MJ, et al.. Genetic activation of pyruvate dehydrogenase alters oxidative substrate selection to induce skeletal muscle insulin resistance. Proc Natl Acad Sci U S A 2014;111:16508–13. https://doi.org/10.1073/pnas.1419104111.Search in Google Scholar PubMed PubMed Central

24. Jackson, S, Bartlett, K, Land, J, Moxon, ER, Pollitt, RJ, Leonard, JV, et al.. Long-chain 3-hydroxyacyl-CoA dehydrogenase deficiency. Pediatr Res 1991;29:406–11. https://doi.org/10.1203/00006450-199104000-00016.Search in Google Scholar PubMed

25. Chakraborty, S, Galla, S, Cheng, X, Yeo, JY, Mell, B, Singh, V, et al.. Salt-responsive metabolite, beta-hydroxybutyrate, attenuates hypertension. Cell Rep 2018;25:677–89. https://doi.org/10.1016/j.celrep.2018.09.058.e4.Search in Google Scholar PubMed PubMed Central

26. Youm, YH, Nguyen, KY, Grant, RW, Goldberg, EL, Bodogai, M, Kim, D, et al.. The ketone metabolite beta-hydroxybutyrate blocks NLRP3 inflammasome-mediated inflammatory disease. Nat Med 2015;21:263–9. https://doi.org/10.1038/nm.3804.Search in Google Scholar PubMed PubMed Central

27. Wu, N, Yang, M, Gaur, U, Xu, H, Yao, Y, Li, D. Alpha-ketoglutarate: physiological functions and applications. Biomol Ther 2016;24:1–8. https://doi.org/10.4062/biomolther.2015.078.Search in Google Scholar PubMed PubMed Central

28. van den Berg, EH, Flores-Guerrero, JL, Gruppen, EG, de Borst, MH, Wolak-Dinsmore, J, Connelly, MA, et al.. Non-alcoholic fatty liver disease and risk of incident type 2 diabetes: role of circulating branched-chain amino acids. Nutrients 2019;11. https://doi.org/10.3390/nu11030705.Search in Google Scholar PubMed PubMed Central

29. Adibi, SA. Influence of dietary deprivations on plasma concentration of free amino acids of man. J Appl Physiol 1968;25:52–7. https://doi.org/10.1152/jappl.1968.25.1.52.Search in Google Scholar PubMed

30. Holecek, M. Why are branched-chain amino acids increased in starvation and diabetes? Nutrients 2020;12. https://doi.org/10.3390/nu12103087.Search in Google Scholar PubMed PubMed Central

31. Nie, C, He, T, Zhang, W, Zhang, G, Ma, X. Branched chain amino acids: beyond nutrition metabolism. Int J Mol Sci 2018;19. https://doi.org/10.3390/ijms19040954.Search in Google Scholar PubMed PubMed Central

32. Maida, A, Chan, JSK, Sjoberg, KA, Zota, A, Schmoll, D, Kiens, B, et al.. Repletion of branched chain amino acids reverses mTORC1 signaling but not improved metabolism during dietary protein dilution. Mol Metab 2017;6:873–81. https://doi.org/10.1016/j.molmet.2017.06.009.Search in Google Scholar PubMed PubMed Central

33. Morgan, TM, Koreckij, TD, Corey, E. Targeted therapy for advanced prostate cancer: inhibition of the PI3K/Akt/mTOR pathway. Curr Cancer Drug Targets 2009;9:237–49. https://doi.org/10.2174/156800909787580999.Search in Google Scholar PubMed PubMed Central

34. Bloomgarden, Z. Diabetes and branched-chain amino acids: what is the link? J Diabetes 2018;10:350–2. https://doi.org/10.1111/1753-0407.12645.Search in Google Scholar PubMed

35. Holecek, M. Histidine in health and disease: metabolism, physiological importance, and use as a supplement. Nutrients 2020;12. https://doi.org/10.3390/nu12030848.Search in Google Scholar PubMed PubMed Central

36. Luhby, AL, Cooperman, JM, Teller, DN. Urinary excretion of formiminoglutamic acid: application in diagnosis of clinical folic acid deficiency. Am J Clin Nutr 1959;7:397–406. https://doi.org/10.1093/ajcn/7.4.397.Search in Google Scholar PubMed

37. Moro, J, Tome, D, Schmidely, P, Demersay, TC, Azzout-Marniche, D. Histidine: a systematic Review on metabolism and physiological effects in human and different animal species. Nutrients 2020;12. https://doi.org/10.3390/nu12051414.Search in Google Scholar PubMed PubMed Central

38. Sun, X, Feng, R, Li, Y, Lin, S, Zhang, W, Li, Y, et al.. Histidine supplementation alleviates inflammation in the adipose tissue of high-fat diet-induced obese rats via the NF-kappaB- and PPARgamma-involved pathways. Br J Nutr 2014;112:477–85. https://doi.org/10.1017/s0007114514001056.Search in Google Scholar PubMed

39. Lee, YT, Hsu, CC, Lin, MH, Liu, KS, Yin, MC. Histidine and carnosine delay diabetic deterioration in mice and protect human low density lipoprotein against oxidation and glycation. Eur J Pharmacol 2005;513:145–50. https://doi.org/10.1016/j.ejphar.2005.02.010.Search in Google Scholar PubMed

40. Li, Y, Zhang, H, Jiang, C, Xu, M, Pang, Y, Feng, J, et al.. Hyperhomocysteinemia promotes insulin resistance by inducing endoplasmic reticulum stress in adipose tissue. J Biol Chem 2013;288:9583–92. https://doi.org/10.1074/jbc.m112.431627.Search in Google Scholar

41. Li, J, Goh, CE, Demmer, RT, Whitcomb, BW, Du, P, Liu, Z. Association between serum folate and insulin resistance among U.S. Nondiabetic adults. Sci Rep 2017;7:9187. https://doi.org/10.1038/s41598-017-09522-5.Search in Google Scholar PubMed PubMed Central

42. Setola, E, Monti, LD, Galluccio, E, Palloshi, A, Fragasso, G, Paroni, R, et al.. Insulin resistance and endothelial function are improved after folate and vitamin B12 therapy in patients with metabolic syndrome: relationship between homocysteine levels and hyperinsulinemia. Eur J Endocrinol 2004;151:483–9. https://doi.org/10.1530/eje.0.1510483.Search in Google Scholar PubMed

43. Campbell, BM, Charych, E, Lee, AW, Moller, T. Kynurenines in CNS disease: regulation by inflammatory cytokines. Front Neurosci 2014;8:12. https://doi.org/10.3389/fnins.2014.00012.Search in Google Scholar

44. Lugo-Huitron, R, Ugalde Muniz, P, Pineda, B, Pedraza-Chaverri, J, Rios, C, Perez-de la Cruz, V. Quinolinic acid: an endogenous neurotoxin with multiple targets. Oxid Med Cell Longev 2013;2013: 104024. https://doi.org/10.1155/2013/104024.Search in Google Scholar

45. Claria, J, Moreau, R, Fenaille, F, Amoros, A, Junot, C, Gronbaek, H, et al.. Orchestration of tryptophan-kynurenine pathway, acute decompensation, and acute-on-chronic liver failure in cirrhosis. Hepatology 2019;69:1686–701. https://doi.org/10.1002/hep.30363.Search in Google Scholar

46. Favennec, M, Hennart, B, Caiazzo, R, Leloire, A, Yengo, L, Verbanck, M, et al.. The kynurenine pathway is activated in human obesity and shifted toward kynurenine monooxygenase activation. Obesity 2015;23:2066–74. https://doi.org/10.1002/oby.21199.Search in Google Scholar

47. Yu, E, Papandreou, C, Ruiz-Canela, M, Guasch-Ferre, M, Clish, CB, Dennis, C, et al.. Association of tryptophan metabolites with incident type 2 diabetes in the PREDIMED trial: a case-cohort study. Clin Chem 2018;64:1211–20. https://doi.org/10.1373/clinchem.2018.288720.Search in Google Scholar

48. Markaverich, BM, Gregory, RR, Alejandro, MA, Clark, JH, Johnson, GA, Middleditch, BS. Methyl p-hydroxyphenyllactate. An inhibitor of cell growth and proliferation and an endogenous ligand for nuclear type-II binding sites. J Biol Chem 1988;263:7203–10. https://doi.org/10.1016/s0021-9258(18)68628-4.Search in Google Scholar

49. Muting, D, Wuzel, H, Bucsis, L, Flasshoff, HJ. Urinary p-hydroxyphenyllactic acid as indicator of hepatic encephalopathy in patients with hepatic cirrhosis. Lancet 1985;2:1365–6. https://doi.org/10.1016/s0140-6736(85)92662-5.Search in Google Scholar

50. Levy, G. Sulfate conjugation in drug metabolism: role of inorganic sulfate. Fed Proc 1986;45:2235–40.Search in Google Scholar


Supplementary Material

The online version of this article offers supplementary material (https://doi.org/10.1515/cclm-2021-0598).


Received: 2021-05-19
Accepted: 2021-07-20
Published Online: 2021-07-30
Published in Print: 2021-10-26

© 2021 Walter de Gruyter GmbH, Berlin/Boston

Downloaded on 26.4.2024 from https://www.degruyter.com/document/doi/10.1515/cclm-2021-0598/html
Scroll to top button