Metabolomic Patterns in Type 2 Diabetes Mellitus: Comparison
Please note this is a comparison between Version 2 by Dean Liu and Version 1 by Mina Shahisavandi.

The spectrum of information related to precision medicine in diabetes generally includes clinical data, genetics, and omics-based biomarkers that can guide personalized decisions on diabetes care. Given the remarkable progress in patient risk characterization, there is particular interest in using molecular biomarkers to guide diabetes management. Metabolomics is an emerging molecular approach that helps better understand the etiology and promises the identification of novel biomarkers for complex diseases. Both targeted or untargeted metabolites extracted from cells, biofluids, or tissues can be investigated by established high-throughput platforms, like nuclear magnetic resonance (NMR) and mass spectrometry (MS) techniques. Metabolomics is proposed as a valuable tool in precision diabetes medicine to discover biomarkers for diagnosis, prognosis, and management of the progress of diabetes through personalized phenotyping and individualized drug-response monitoring.

  • type 2 diabetes mellitus
  • metabolomics
  • metabolites
  • glucose-lowering medications

1. Lipids

Previous epidemiological studies have consistently demonstrated a long-standing association between lipids and diabetes. However, due to the lack of specificity for traditional clinical measurements, the lipid profile was the only available target for prior metabolomics studies for a long time. Thus, most prior studies investigating dyslipidemia among diabetes mainly focused on triglycerides and high and low-density lipoprotein cholesterol. These findings mainly supported the notion that low levels of high-density lipoprotein cholesterol (HDL-C) and high levels of triglyceride (TG) are significant predictors of the occurrence of T2D development in adults [14,15][1][2]. The low levels of HDL-C also greatly enhanced the tyrosine serum level in patients with T2D [38][3].
Based on an updated systematic review and meta-analysis of prospective cohorts encompassing a vast participant pool of 71,196 individuals, among whom 11,771 were diagnosed with T2D, demonstrated that 123 metabolites are significantly associated with T2D risk using high throughput metabolomics data. They concluded that several glycerolipids, (lyso)phosphatidylethanolamines, dihydroceramide, and ceramides are linked to an elevated risk of developing T2D [16][4].
In addition, a recent study among the Chinese population included 5731 people, of whom 529 participants developed T2D. TResearchis study ers investigated a panel of novel sphingolipids, including ceramides, saturated sphingomyelins, unsaturated sphingomyelins, hydroxyl-sphingomyelins, and hexosyl ceramide and their association with T2D incidence in a six-year follow-up study. The result indicated a positive association of these metabolites with incident T2D and β-cell dysfunction. According to this study, sSphingolipids incorporate in developing impaired glucose homeostasis by inducing insulin resistance, impairing β-cell function, and inflammation [39][5].
Due to advances in high-throughput metabolomics technology and subtler lipid species or lipidomics analysis, wresearche rs can now study the total acyl chain carbon number and degree of unsaturation of plasma lipids. Therefore, odd-chain saturated fatty acids (OCFA)-containing lipids were found to exhibit a sex-specific association with the risk of developing T2D. At the same time, specific OCFA-containing Phospholipids, such as Phosphatidylcholine C15:0, were only negatively correlated with the risk of developing T2D in women but not men [18][6].
The results of a case-control study involving 107 men with T2D and 216 controls sourced from the longitudinal METSIM study indicated higher levels of triacylglycerols, di-acyl-phospholipids, lysoalkylphosphatidylcholine, and lysophosphatidylcholine acyl and that lower levels of alkyl-acyl phosphatidylcholines are linked to the higher risk of T2D [19][7]. The models of lipids remained reliable for the development of T2D within the fasting plasma glucose-matched subset even in the validation phase of the study. ThIt is study demonstratesd that a characteristic lipid molecule of T2D exists many years before diagnosis and enhances the likelihood of progression to T2D. Besides that, other endeavours reported that diacyl-phosphatidylcholines (C32:1, C36:0, C36:1, C38:3, and C40:5) were significantly altered in T2D compared to non-T2D subjects [20,40][8][9]. By repeatedly measuring plasma lipid metabolites at the baseline of 250 incident T2D cases and 692 participants without T2D at the baseline after one year of follow-up, the PREDIMED trial found that the plasma lipid profiles composed of triacylglycerols, diacylglycerols, and phosphatidylethanolamines were associated with a higher risk of T2D [17][10]. These fatty acids (FAs) are mainly derived from dietary triglycerides and phospholipids. Thus, they may be targeted as new interventions in diabetes dietary prevention.

2. Amino Acids

Among various amino acid metabolites, branch-chain amino acids (BCAAs) (i.e., leucine, isoleucine, and valine) and their related metabolites have been reported to be strongly associated with early diagnosis and predicting the occurrence of T2D [11,16,41,42,43][4][11][12][13][14]. BCAAs have an impact on various cellular signaling pathways and their association with insulin resistance. BCAAs are known to enhance protein synthesis and increase mitochondrial content in muscle and adipocytes. However, elevated circulating BCAA levels have been linked to insulin resistance, potentially due to dysregulated BCAA degradation [44][15]. BCAAs—particularly leucine—activate the rapamycin complex1 mTORC1, which is known to regulate cell growth and metabolism, glucose metabolism, and several more essential physiological processes [45][16]. When incorporated into a dietary pattern that includes high-fat consumption, BCAAs contribute to the development of obesity, insulin resistance, and diabetes. While BCAAs have been linked to anti-obesity effects, higher circulating levels of BCAAs are observed in individuals with obesity. They are associated with poorer metabolic health and increased risk of insulin resistance and T2D. Insulin resistance may also contribute to elevated levels of amino acids by promoting protein degradation and impairing BCAA oxidative metabolism in certain tissues [46,47][17][18]. Furthermore, results from the Framingham Heart Study (FHS) found that participants with higher serum levels of BCAAs had a higher risk of developing T2D even after adjusting for the body mass index (BMI) [48][19]. This might be explained by altering cellular insulin signaling due to increased serum BCAAs levels and involving the mammalian rapamycin pathway, pancreatic islet β-cells, and adipocytes, leading to cytotoxic metabolite build-up [49,50][20][21].
Besides that, imbalances in amino acid homeostasis are also linked to insulin resistance among people with low BMI and higher levels of the aromatic amino acids (AAAs) tyrosine,2-hydroxybutyrate, methionine, phenylalanine, lysine, histidine, 2-aminoadipate, alanine, and glutamate have also been associated with an increased risk of T2D [12,37,42,51,52,53,54][13][22][23][24][25][26][27]. On the other hand, a negative association has been found in serum concentration of Glycine, Glutamine, and the risk of T2D development [11]. However, the results are less consistent for specific amino acids such as Glycine, which was inversely associated with incident T2D in Europeans [40][9] but with a positive association in the Chinese population [13][28]. Moreover, results from the Mendelian Randomization (MR) analysis embedded in the FHS Offspring cohort reported a negative association between glycine and T2D risk [12][22]. This lack of consistency may contribute to mainly including white participants in mentioned studies, and evidence from other ethnicities is needed to achieve generalizability [43,55,56][14][29][30].
In a similar study, the metabolite signatures of obese children with T2D, obese children without diabetes (OB), and healthy normal weight controls (NW) were compared. Using targeted LC-MS/MS, 22 urine metabolites were identified that were uniquely associated with T2D. The results included metabolites related to the betaine pathway, nucleic acid metabolism, and BCAAs. Moreover, urine levels of succinylaminoimidazole carboxamide riboside (SAICA-riboside) were found to be increased in diabetic youth, suggesting its potential as a biomarker for T2D [57][31].

3. Carbohydrates

According to an updated meta-analysis of the case-cohort studies (71,196 participants and 11,771 T2D cases/events), higher carbohydrate metabolites, including mannose and trehalose, are also associated with an increased risk of T2D. Furthermore, higher levels of glycine, glutamine, betaine, indolepropionate, and (lyso)phosphatidylcholines were associated with lower T2D risk (hazard ratio 0.69–0.90) [6][32].
There is study showed a significant association between glycolysis/gluconeogenesis metabolite (i.e., pyruvate) and higher T2D risk [16][4]. Another systematic review involving 27 cross-sectional and 19 prospective cohort studies revealed that metabolites embedded in sugar, including glucose, hexose, mannose, arabinose, fructose, and glycolipids, were positively associated with the prevalence of T2D [58][33]. Additionally, using a non-targeted metabolomics platform in a case-control study (with 115 cases diagnosed with T2D, 192 individuals with impaired fasting glucose, and 1897 control subjects), results showed that subjects with lower plasma levels of 1,5-anhydroglucitol, and higher plasma concentrations of glucose, mannose, and fructose had more risk of impaired fasting glucose and T2D [22][34].
Complex interactions between genetic and environmental factors influence the metabolite profile of an individual. GWAS have been used to explore the impact of genetic variation on plasma metabolites. The identified genetic variants associated with metabolite levels, particularly enzymes and carriers involved in processes like β-oxidation, fatty acid and phospholipid biosynthesis, as well as amino acid metabolism. Notably, these genetic loci explain a significant portion of the variance in metabolites, highlighting the role of genetics. Furthermore, specific genetic variants have been linked to glycine, serine, and betaine levels, although their connection to diabetes-related traits remains unclear [59][35].
Environmental factors such as one’s diet, activity, medication, and the microbiome contribute to the complexity of the metabolome. Diet, physical activity, gender, and age influence metabolomic profiles and should be considered in study design and interpretation. The gut microbiome plays a significant role in host metabolism and metabolomics profiles, with obesity and T2D associated with altered microbial profiles and reduced diversity. Microbial populations can impact host metabolism, intestinal development, and insulin secretion and contribute to metabolic disorders. Microbiota transplantation studies in mice and limited human data suggest the potential for improving metabolic health through modulation of the microbiome [60,61,62][36][37][38].

References

  1. Chen, Z.; Hu, H.; Chen, M.; Luo, X.; Yao, W.; Liang, Q.; Yang, F.; Wang, X. Association of Triglyceride to high-density lipoprotein cholesterol ratio and incident of diabetes mellitus: A secondary retrospective analysis based on a Chinese cohort study. Lipids Health Dis. 2020, 19, 33.
  2. Liu, H.; Yan, S.; Chen, G.; Li, B.; Zhao, L.; Wang, Y.; Hu, X.; Jia, X.; Dou, J.; Mu, Y.; et al. Association of the Ratio of Triglycerides to High-Density Lipoprotein Cholesterol Levels with the Risk of Type 2 Diabetes: A Retrospective Cohort Study in Beijing. J. Diabetes Res. 2021, 2021, 1–8.
  3. Li, J.; Cao, Y.F.; Sun, X.Y.; Han, L.; Li, S.N.; Gu, W.Q.; Song, M.; Jiang, C.T.; Yang, X.; Fang, Z.Z. Plasma tyrosine and its interaction with low high-density lipoprotein cholesterol and the risk of type 2 diabetes mellitus in Chinese. J. Diabetes Investig. 2019, 10, 491–498.
  4. Morze, J.; Wittenbecher, C.; Schwingshackl, L.; Danielewicz, A.; Rynkiewicz, A.; Hu, F.B.; Guasch-Ferré, M. Metabolomics and Type 2 Diabetes Risk: An Updated Systematic Review and Meta-analysis of Prospective Cohort Studies. Diabetes Care 2022, 45, 1013–1024.
  5. Yun, H.; Sun, L.; Wu, Q.; Zong, G.; Qi, Q.; Li, H.; Zheng, H.; Zeng, R.; Liang, L.; Lin, X. Associations among circulating sphingolipids, β-cell function, and risk of developing type 2 diabetes: A population-based cohort study in China. PLoS Med. 2020, 17, e1003451.
  6. Prada, M.; Wittenbecher, C.; Eichelmann, F.; Wernitz, A.; Drouin-Chartier, J.P.; Schulze, M.B. Association of the odd-chain fatty acid content in lipid groups with type 2 diabetes risk: A targeted analysis of lipidomics data in the EPIC-Potsdam cohort. Clin. Nutr. 2021, 40, 4988–4999.
  7. Suvitaival, T.; Bondia-Pons, I.; Yetukuri, L.; Pöhö, P.; Nolan, J.J.; Hyötyläinen, T.; Kuusisto, J.; Orešič, M. Lipidome as a predictive tool in progression to type 2 diabetes in Finnish men. Metabolism 2018, 78, 1–12.
  8. Lee, H.S.; Xu, T.; Lee, Y.; Kim, N.H.; Kim, Y.J.; Kim, J.M.; Cho, S.Y.; Kim, K.Y.; Nam, M.; Adamski, J.; et al. Identification of putative biomarkers for type 2 diabetes using metabolomics in the Korea Association REsource (KARE) cohort. Metabolomics 2016, 12, 1–12.
  9. Floegel, A.; Stefan, N.; Yu, Z.; Muhlenbruch, K.; Drogan, D.; Joost, H.G.; Fritsche, A.; Haring, H.U.; Hrabe de Angelis, M.; Peters, A.; et al. Identification of serum metabolites associated with risk of type 2 diabetes using a targeted metabolomic approach. Diabetes 2013, 62, 639–648.
  10. Razquin, C.; Toledo, E.; Clish, C.B.; Ruiz-Canela, M.; Dennis, C.; Corella, D.; Papandreou, C.; Ros, E.; Estruch, R.; Guasch-Ferré, M.; et al. Plasma Lipidomic Profiling and Risk of Type 2 Diabetes in the PREDIMED Trial. Diabetes Care 2018, 41, 2617–2624.
  11. Mitro, S.D.; Liu, J.; Jaacks, L.M.; Fleisch, A.F.; Williams, P.L.; Knowler, W.C.; Laferrère, B.; Perng, W.; Bray, G.A.; Wallia, A.; et al. Per- and polyfluoroalkyl substance plasma concentrations and metabolomic markers of type 2 diabetes in the Diabetes Prevention Program trial. Int. J. Hyg. Environ. Health 2021, 232, 113680.
  12. Gall, W.E.; Beebe, K.; Lawton, K.A.; Adam, K.-P.; Mitchell, M.W.; Nakhle, P.J.; Ryals, J.A.; Milburn, M.V.; Nannipieri, M.; Camastra, S.; et al. α-Hydroxybutyrate Is an Early Biomarker of Insulin Resistance and Glucose Intolerance in a Nondiabetic Population. PLoS ONE 2010, 5, e10883.
  13. Alqudah, A.; Wedyan, M.; Qnais, E.; Jawarneh, H.; McClements, L. Plasma Amino Acids Metabolomics’ Important in Glucose Management in Type 2 Diabetes. Front. Pharmacol. 2021, 12, 695418.
  14. Lotta, L.A.; Scott, R.A.; Sharp, S.J.; Burgess, S.; Luan, J.; Tillin, T.; Schmidt, A.F.; Imamura, F.; Stewart, I.D.; Perry, J.R.B.; et al. Genetic Predisposition to an Impaired Metabolism of the Branched-Chain Amino Acids and Risk of Type 2 Diabetes: A Mendelian Randomisation Analysis. PLoS Med. 2016, 13, e1002179.
  15. Gannon, N.P.; Schnuck, J.K.; Vaughan, R.A. BCAA Metabolism and Insulin Sensitivity–Dysregulated by Metabolic Status? Mol. Nutr. Food Res. 2018, 62, e1700756.
  16. Cuomo, P.; Capparelli, R.; Iannelli, A.; Iannelli, D. Role of Branched-Chain Amino Acid Metabolism in Type 2 Diabetes, Obesity, Cardiovascular Disease and Non-Alcoholic Fatty Liver Disease. Int. J. Mol. Sci. 2022, 23, 4325.
  17. Newgard, C.B.; An, J.; Bain, J.R.; Muehlbauer, M.J.; Stevens, R.D.; Lien, L.F.; Haqq, A.M.; Shah, S.H.; Arlotto, M.; Slentz, C.A.; 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.
  18. Pan, X.F.; Chen, Z.Z.; Wang, T.J.; Shu, X.; Cai, H.; Cai, Q.; Clish, C.B.; Shi, X.; Zheng, W.; Gerszten, R.E.; et al. Plasma metabolomic signatures of obesity and risk of type 2 diabetes. Obesity 2022, 30, 2294–2306.
  19. Wang, T.J.; Larson, M.G.; Vasan, R.S.; Cheng, S.; Rhee, E.P.; McCabe, E.; Lewis, G.D.; Fox, C.S.; Jacques, P.F.; Fernandez, C.; et al. Metabolite profiles and the risk of developing diabetes. Nat. Med. 2011, 17, 448–453.
  20. Ardestani, A.; Lupse, B.; Kido, Y.; Leibowitz, G.; Maedler, K. mTORC1 Signaling: A Double-Edged Sword in Diabetic β Cells. Cell Metab. 2018, 27, 314–331.
  21. Yoon, M.S. The Role of Mammalian Target of Rapamycin (mTOR) in Insulin Signaling. Nutrients 2017, 9, 1176.
  22. Merino, J.; Leong, A.; Liu, C.T.; Porneala, B.; Walford, G.A.; von Grotthuss, M.; Wang, T.J.; Flannick, J.; Dupuis, J.; Levy, D.; et al. Metabolomics insights into early type 2 diabetes pathogenesis and detection in individuals with normal fasting glucose. Diabetologia 2018, 61, 1315–1324.
  23. Tai, E.S.; Tan, M.L.; Stevens, R.D.; Low, Y.L.; Muehlbauer, M.J.; Goh, D.L.; Ilkayeva, O.R.; Wenner, B.R.; Bain, J.R.; Lee, J.J.; et al. Insulin resistance is associated with a metabolic profile of altered protein metabolism in Chinese and Asian-Indian men. Diabetologia 2010, 53, 757–767.
  24. Zhang, X.; Wang, Y.; Hao, F.; Zhou, X.; Han, X.; Tang, H.; Ji, L. Human serum metabonomic analysis reveals progression axes for glucose intolerance and insulin resistance statuses. J. Proteome Res. 2009, 8, 5188–5195.
  25. Würtz, P.; Tiainen, M.; Mäkinen, V.-P.; Kangas, A.J.; Soininen, P.; Saltevo, J.; Keinänen-Kiukaanniemi, S.; Mäntyselkä, P.; Lehtimäki, T.; Laakso, M.; et al. Circulating Metabolite Predictors of Glycemia in Middle-Aged Men and Women. Diabetes Care 2012, 35, 1749–1756.
  26. Geidenstam, N.; Spégel, P.; Mulder, H.; Filipsson, K.; Ridderstråle, M.; Danielsson, A.P. Metabolite profile deviations in an oral glucose tolerance test-a comparison between lean and obese individuals. Obesity 2014, 22, 2388–2395.
  27. Gu, X.; Al Dubayee, M.; Alshahrani, A.; Masood, A.; Benabdelkamel, H.; Zahra, M.; Li, L.; Abdel Rahman, A.M.; Aljada, A. Distinctive Metabolomics Patterns Associated with Insulin Resistance and Type 2 Diabetes Mellitus. Front. Mol. Biosci. 2020, 7, 609806.
  28. Lu, Y.; Wang, Y.; Ong, C.N.; Subramaniam, T.; Choi, H.W.; Yuan, J.M.; Koh, W.P.; Pan, A. Metabolic signatures and risk of type 2 diabetes in a Chinese population: An untargeted metabolomics study using both LC-MS and GC-MS. Diabetologia 2016, 59, 2349–2359.
  29. Wang, Q.; Holmes, M.V.; Davey Smith, G.; Ala-Korpela, M. Genetic Support for a Causal Role of Insulin Resistance on Circulating Branched-Chain Amino Acids and Inflammation. Diabetes Care 2017, 40, 1779–1786.
  30. Wittemans, L.B.L.; Lotta, L.A.; Oliver-Williams, C.; Stewart, I.D.; Surendran, P.; Karthikeyan, S.; Day, F.R.; Koulman, A.; Imamura, F.; Zeng, L.; et al. Assessing the causal association of glycine with risk of cardio-metabolic diseases. Nat. Commun. 2019, 10, 1060.
  31. Concepcion, J.; Chen, K.; Saito, R.; Gangoiti, J.; Mendez, E.; Nikita, M.E.; Barshop, B.A.; Natarajan, L.; Sharma, K.; Kim, J.J. Identification of pathognomonic purine synthesis biomarkers by metabolomic profiling of adolescents with obesity and type 2 diabetes. PLoS ONE 2020, 15, e0234970.
  32. Li, B.; He, X.; Jia, W.; Li, H. Novel Applications of Metabolomics in Personalized Medicine: A Mini-Review. Molecules 2017, 22, 1173.
  33. Guasch-Ferré, M.; Hruby, A.; Toledo, E.; Clish, C.B.; Martínez-González, M.A.; Salas-Salvadó, J.; Hu, F.B. Metabolomics in Prediabetes and Diabetes: A Systematic Review and Meta-analysis. Diabetes Care 2016, 39, 833–846.
  34. Menni, C.; Fauman, E.; Erte, I.; Perry, J.R.; Kastenmüller, G.; Shin, S.Y.; Petersen, A.K.; Hyde, C.; Psatha, M.; Ward, K.J.; et al. Biomarkers for type 2 diabetes and impaired fasting glucose using a nontargeted metabolomics approach. Diabetes 2013, 62, 4270–4276.
  35. Gonzalez-Franquesa, A.; Burkart, A.M.; Isganaitis, E.; Patti, M.E. What Have Metabolomics Approaches Taught Us About Type 2 Diabetes? Curr. Diab. Rep. 2016, 16, 74.
  36. Vrieze, A.; Van Nood, E.; Holleman, F.; Salojärvi, J.; Kootte, R.S.; Bartelsman, J.F.; Dallinga-Thie, G.M.; Ackermans, M.T.; Serlie, M.J.; Oozeer, R.; et al. Transfer of intestinal microbiota from lean donors increases insulin sensitivity in individuals with metabolic syndrome. Gastroenterology 2012, 143, 913–916.e7.
  37. Wang, Z.; Klipfell, E.; Bennett, B.J.; Koeth, R.; Levison, B.S.; Dugar, B.; Feldstein, A.E.; Britt, E.B.; Fu, X.; Chung, Y.M.; et al. Gut flora metabolism of phosphatidylcholine promotes cardiovascular disease. Nature 2011, 472, 57–63.
  38. Koeth, R.A.; Wang, Z.; Levison, B.S.; Buffa, J.A.; Org, E.; Sheehy, B.T.; Britt, E.B.; Fu, X.; Wu, Y.; Li, L.; et al. Intestinal microbiota metabolism of L-carnitine, a nutrient in red meat, promotes atherosclerosis. Nat. Med. 2013, 19, 576–585.
More
Video Production Service