Skip to main content

Advertisement

Log in

A Decade of Genetic and Metabolomic Contributions to Type 2 Diabetes Risk Prediction

  • Genetics (AP Morris, Section Editor)
  • Published:
Current Diabetes Reports Aims and scope Submit manuscript

Abstract

Purpose of Review

The purpose of this review was to summarize and reflect on advances over the past decade in human genetic and metabolomic discovery with particular focus on their contributions to type 2 diabetes (T2D) risk prediction.

Recent Findings

In the past 10 years, a combination of advances in genotyping efficiency, metabolomic profiling, bioinformatics approaches, and international collaboration have moved T2D genetics and metabolomics from a state of frustration to an abundance of new knowledge.

Summary

Efforts to control and prevent T2D have failed to stop this global epidemic. New approaches are needed, and although neither genetic nor metabolomic profiling yet have a clear clinical role, the rapid pace of accumulating knowledge offers the possibility for “multi-omic” prediction to improve health.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Fig. 1
Fig. 2

Similar content being viewed by others

References

Papers of particular interest, published recently, have been highlighted as: • Of importance •• Of major importance

  1. Ashley EA. Towards precision medicine. Nat Rev Genet. 2016;17:507–22.

    Article  CAS  PubMed  Google Scholar 

  2. Florez JC. Mining the genome for therapeutic targets. Diabetes. 2017;66:1770–8.

    Article  PubMed  Google Scholar 

  3. Florez JC. Leveraging genetics to advance type 2 diabetes prevention. PLoS Med. 2016;13:e1002102.

    Article  PubMed  PubMed Central  Google Scholar 

  4. Visscher PM, Wray NR, Zhang Q, Sklar P, McCarthy MI, Brown MA, et al. 10 years of GWAS discovery: biology, function, and translation. Am J Hum Genet. 2017;101:5–22.

    Article  CAS  PubMed  Google Scholar 

  5. Almgren P, Lehtovirta M, Isomaa B, Sarelin L, Taskinen MR, Lyssenko V, et al. Heritability and familiality of type 2 diabetes and related quantitative traits in the Botnia Study. Diabetologia. 2011;54:2811–9.

    Article  CAS  PubMed  Google Scholar 

  6. Fajans SS, Bell GI, Polonsky KS. Molecular mechanisms and clinical pathophysiology of maturity-onset diabetes of the young. N Engl J Med. 2001;345:971–80.

    Article  CAS  PubMed  Google Scholar 

  7. Vaxillaire M, Froguel P. Genetic basis of maturity-onset diabetes of the young. Endocrinol Metab Clin North Am. 2006;35:371–84.

    Article  CAS  PubMed  Google Scholar 

  8. Gloyn AL, Pearson ER, Antcliff JF, Proks P, Bruining GJ, Slingerland AS, et al. Activating mutations in the gene encoding the ATP-sensitive potassium-channel subunit Kir6.2 and permanent neonatal diabetes. N Engl J Med. 2004;350:1838–49.

    Article  CAS  PubMed  Google Scholar 

  9. Babenko AP, Polak M, Cave H, Busiah K, Czernichow P, Scharfmann R, et al. Activating mutations in the ABCC8 gene in neonatal diabetes mellitus. N Engl J Med. 2006;355:456–66.

    Article  CAS  PubMed  Google Scholar 

  10. Sagen JV, Raeder H, Hathout E, Shehadeh N, Gudmundsson K, Baevre H, et al. Permanent neonatal diabetes due to mutations in KCNJ11 encoding Kir6.2: patient characteristics and initial response to sulfonylurea therapy. Diabetes. 2004;53:2713–8.

    Article  CAS  PubMed  Google Scholar 

  11. Altshuler D, Hirschhorn JN, Klannemark M, Lindgren CM, Vohl MC, Nemesh J, et al. The common PPARg Pro12Ala polymorphism is associated with decreased risk of type 2 diabetes. Nat Genet. 2000;26:76–80.

    Article  CAS  PubMed  Google Scholar 

  12. Gloyn AL, Weedon MN, Owen KR, Turner MJ, Knight BA, Hitman G, et al. Large-scale association studies of variants in genes encoding the pancreatic b-cell KATP channel subunits Kir6.2 (KCNJ11) and SUR1 (ABCC8) confirm that the KCNJ11 E23K variant is associated with type 2 diabetes. Diabetes. 2003;52:568–72.

    Article  CAS  PubMed  Google Scholar 

  13. Grant SFA, Thorleifsson G, Reynisdottir I, Benediktsson R, Manolescu A, Sainz J, et al. Variant of transcription factor 7-like 2 (TCF7L2) gene confers risk of type 2 diabetes. Nat Genet. 2006;38:320–3.

    Article  CAS  PubMed  Google Scholar 

  14. •• Mohlke KL, Boehnke M. Recent advances in understanding the genetic architecture of type 2 diabetes. Hum Mol Genet. 2015;24:R85–92. This review provides a helpful overview of genetic discoveries for type 2 diabetes and glycemic traits.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. • Scott RA, Scott LJ, Magi R, Marullo L, Gaulton KJ, Kaakinen M, et al. Replication DIG, meta-analysis C: an expanded genome-wide association study of type 2 diabetes in Europeans. Diabetes. 2017. This is the largest genome-wide study to date in predominantly European populations and includes imputation into the 1000 genomes multi-ethnic reference panel.

  16. Morris AP, Voight BF, Teslovich TM, Ferreira T, Segre AV, Steinthorsdottir V, et al. Replication DIG, meta-analysis C: large-scale association analysis provides insights into the genetic architecture and pathophysiology of type 2 diabetes. Nat Genet. 2012;44:981–90.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. •• Mahajan A, Go MJ, Zhang W, Below JE, Gaulton KJ, Ferreira T, et al. Genome-wide trans-ancestry meta-analysis provides insight into the genetic architecture of type 2 diabetes susceptibility. Nat Genet. 2014;46:234–44. This is the largest trans-ethnic genome-wide association study.

    Article  CAS  PubMed  Google Scholar 

  18. Ng MC, Shriner D, Chen BH, Li J, Chen WM, Guo X, et al. Consortium ME-aotDiAA: Meta-analysis of genome-wide association studies in African Americans provides insights into the genetic architecture of type 2 diabetes. PLoS Genet. 2014;10:e1004517.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  19. Cho YS, Chen CH, Hu C, Long J, Ong RT, Sim X, et al. Meta-analysis of genome-wide association studies identifies eight new loci for type 2 diabetes in east Asians. Nat Genet. 2011;44:67–72.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  20. Manolio TA, Collins FS, Cox NJ, Goldstein DB, Hindorff LA, Hunter DJ, et al. Finding the missing heritability of complex diseases. Nature. 2009;461:747–53.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Franks PW, McCarthy MI. Exposing the exposures responsible for type 2 diabetes and obesity. Science. 2016;354:69–73.

    Article  CAS  PubMed  Google Scholar 

  22. Dimas AS, Lagou V, Barker A, Knowles JW, Magi R, Hivert MF, et al. Impact of type 2 diabetes susceptibility variants on quantitative glycemic traits reveals mechanistic heterogeneity. Diabetes. 2014;63:2158–71.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Scott RA, Lagou V, Welch RP, Wheeler E, Montasser ME, Luan J, et al. Large-scale association analyses identify new loci influencing glycemic traits and provide insight into the underlying biological pathways. Nat Genet. 2012;44:991–1005.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. •• Fuchsberger C, Flannick J, Teslovich TM, Mahajan A, Agarwala V, Gaulton KJ, et al. The genetic architecture of type 2 diabetes. Nature. 2016;536:41–7. This paper includes analysis of the largest set of exome sequences from type 2 diabetes cases and controls and provides insight into the genetic architecture of the disease.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Huyghe JR, Jackson AU, Fogarty MP, Buchkovich ML, Stancakova A, Stringham HM, et al. Exome array analysis identifies new loci and low-frequency variants influencing insulin processing and secretion. Nat Genet. 2013;45:197–201.

    Article  CAS  PubMed  Google Scholar 

  26. Albrechtsen A, Grarup N, Li Y, Sparso T, Tian G, Cao H, et al. Exome sequencing-driven discovery of coding polymorphisms associated with common metabolic phenotypes. Diabetologia. 2013;56:298–310.

    Article  CAS  PubMed  Google Scholar 

  27. • Steinthorsdottir V, Thorleifsson G, Sulem P, Helgason H, Grarup N, Sigurdsson A, et al. Identification of low-frequency and rare sequence variants associated with elevated or reduced risk of type 2 diabetes. Nat Genet. 2014;46:294–8. The authors analyze whole-genome sequences of 2630 Icelanders and imputation into 11,114 Icelandic cases and 267,140 controls to identify low frequency and rare variants associated with T2D.

    Article  CAS  PubMed  Google Scholar 

  28. Schork NJ, Murray SS, Frazer KA, Topol EJ. Common vs. rare allele hypotheses for complex diseases. Curr Opin Genet Dev. 2009;19:212–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Moltke I, Grarup N, Jorgensen ME, Bjerregaard P, Treebak JT, Fumagalli M, et al. A common Greenlandic TBC1D4 variant confers muscle insulin resistance and type 2 diabetes. Nature. 2014;512:190–3.

    Article  CAS  PubMed  Google Scholar 

  30. Estrada K, Aukrust I, Bjorkhaug L, Burtt NP, Mercader JM, Garcia-Ortiz H, et al. Association of a low-frequency variant in HNF1A with type 2 diabetes in a Latino population. JAMA. 2014;311:2305–14.

    Article  PubMed  CAS  Google Scholar 

  31. Hindorff LA, Sethupathy P, Junkins HA, Ramos EM, Mehta JP, Collins FS, et al. Potential etiologic and functional implications of genome-wide association loci for human diseases and traits. Proc Natl Acad Sci U S A. 2009;106:9362–7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Bouatia-Naji N, Bonnefond A, Cavalcanti-Proenca C, Sparso T, Holmkvist J, Marchand M, et al. A variant near MTNR1B is associated with increased fasting plasma glucose levels and type 2 diabetes risk. Nat Genet. 2009;41:89–94.

    Article  CAS  PubMed  Google Scholar 

  33. Lyssenko V, Nagorny CL, Erdos MR, Wierup N, Jonsson A, Spegel P, et al. Common variant in MTNR1B associated with increased risk of type 2 diabetes and impaired early insulin secretion. Nat Genet. 2009;41:82–8.

    Article  CAS  PubMed  Google Scholar 

  34. Gaulton KJ, Ferreira T, Lee Y, Raimondo A, Magi R, Reschen ME, et al. Replication DIG, meta-analysis C: genetic fine mapping and genomic annotation defines causal mechanisms at type 2 diabetes susceptibility loci. Nat Genet. 2015;47:1415–25.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Lane JM, Chang AM, Bjonnes AC, Aeschbach D, Anderson C, Cade BE, et al. Impact of common diabetes risk variant in MTNR1B on sleep, circadian, and melatonin physiology. Diabetes. 2016;65:1741–51.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. McCarthy MI, Abecasis GR, Cardon LR, Goldstein DB, Little J, Ioannidis JP, et al. Genome-wide association studies for complex traits: consensus, uncertainty and challenges. Nat Rev Genet. 2008;9:356–69.

    Article  CAS  PubMed  Google Scholar 

  37. Manolio TA. Bringing genome-wide association findings into clinical use. Nat Rev Genet. 2013;14:549–58.

    Article  CAS  PubMed  Google Scholar 

  38. Meigs JB, Shrader P, Sullivan LM, McAteer JB, Fox CS, Dupuis J, et al. Genotype score in addition to common risk factors for prediction of type 2 diabetes. N Engl J Med. 2008;359:2208–19.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Vassy JL, Dasmahapatra P, Meigs JB, Schork NJ, Magnussen CG, Chen W, et al. Genotype prediction of adult type 2 diabetes from adolescence in a multiracial population. Pediatrics. 2012;130:e1235–42.

    Article  PubMed  PubMed Central  Google Scholar 

  40. Vassy JL, Durant NH, Kabagambe EK, Carnethon MR, Rasmussen-Torvik LJ, Fornage M, et al. A genotype risk score predicts type 2 diabetes from young adulthood: the CARDIA study. Diabetologia. 2012;55:2604–12.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. • Vassy JL, Hivert MF, Porneala B, Dauriz M, Florez JC, Dupuis J, et al. Polygenic type 2 diabetes prediction at the limit of common variant detection. Diabetes. 2014;63:2172–82. This is the most recent assessment of type 2 diabetes genetic risk score on prediction of new-onset type 2 diabetes.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Lyssenko V, Jonsson A, Almgren P, Pulizzi N, Isomaa B, Tuomi T, et al. Clinical risk factors, DNA variants, and the development of type 2 diabetes. N Engl J Med. 2008;359:2220–32.

    Article  CAS  PubMed  Google Scholar 

  43. Kooner JS, Saleheen D, Sim X, Sehmi J, Zhang W, Frossard P, et al. Genome-wide association study in individuals of South Asian ancestry identifies six new type 2 diabetes susceptibility loci. Nat Genet. 2011;43:984–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Palmer ND, Goodarzi MO, Langefeld CD, Wang N, Guo X, Taylor KD, et al. Genetic variants associated with quantitative glucose homeostasis traits translate to type 2 diabetes in Mexican Americans: the GUARDIAN (Genetics Underlying Diabetes in Hispanics) Consortium. Diabetes. 2015;64:1853–66.

    Article  CAS  PubMed  Google Scholar 

  45. Liu CT, Raghavan S, Maruthur N, Kabagambe EK, Hong J, Ng MC, et al. Trans-ethnic meta-analysis and functional annotation illuminates the genetic architecture of fasting glucose and insulin. Am J Hum Genet. 2016;99:56–75.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Consortium STD, Williams AL, Jacobs SB, Moreno-Macias H, Huerta-Chagoya A, Churchhouse C, et al. Sequence variants in SLC16A11 are a common risk factor for type 2 diabetes in Mexico. Nature. 2014;506:97–101.

    Google Scholar 

  47. Ma RC, Hu C, Tam CH, Zhang R, Kwan P, Leung TF, et al. Genome-wide association study in a Chinese population identifies a susceptibility locus for type 2 diabetes at 7q32 near PAX4. Diabetologia. 2013;56:1291–305.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Manning A, Highland HM, Gasser J, Sim X, Tukiainen T, Fontanillas P, et al. A low-frequency inactivating Akt2 variant enriched in the Finnish population is associated with fasting insulin levels and type 2 diabetes risk. Diabetes. 2017.

  49. • Lawlor DA, Harbord RM, Sterne JA, Timpson N, Davey Smith G. Mendelian randomization: using genes as instruments for making causal inferences in epidemiology. Stat Med. 2008;27:1133–63. This study describes the use of Mendelian randomization for causal inference in epidemiology.

    Article  PubMed  Google Scholar 

  50. Burgess S, Timpson NJ, Ebrahim S, Davey Smith G. Mendelian randomization: where are we now and where are we going? Int J Epidemiol. 2015;44:379–88.

    Article  PubMed  Google Scholar 

  51. Palmer TM, Lawlor DA, Harbord RM, Sheehan NA, Tobias JH, Timpson NJ, et al. Using multiple genetic variants as instrumental variables for modifiable risk factors. Stat Methods Med Res. 2012;21:223–42.

    Article  PubMed  PubMed Central  Google Scholar 

  52. Palmer TM, Sterne JA, Harbord RM, Lawlor DA, Sheehan NA, Meng S, et al. Instrumental variable estimation of causal risk ratios and causal odds ratios in Mendelian randomization analyses. Am J Epidemiol. 2011;173:1392–403.

    Article  PubMed  Google Scholar 

  53. Marathe PH, Gao HX, Close KL. American diabetes association standards of medical care in diabetes 2017. J Diabetes. 2017.

  54. Ahmad OS, Morris JA, Mujammami M, Forgetta V, Leong A, Li R, et al. A Mendelian randomization study of the effect of type-2 diabetes on coronary heart disease. Nat Commun. 2015;6:7060.

    Article  PubMed  PubMed Central  Google Scholar 

  55. Ross S, Gerstein HC, Eikelboom J, Anand SS, Yusuf S, Pare G. Mendelian randomization analysis supports the causal role of dysglycaemia and diabetes in the risk of coronary artery disease. Eur Heart J. 2015;36:1454–62.

    Article  PubMed  Google Scholar 

  56. Larsson SC, Scott RA, Traylor M, Langenberg CC, Hindy G, Melander O, et al. Network NSG: type 2 diabetes, glucose, insulin, BMI, and ischemic stroke subtypes: Mendelian randomization study. Neurology. 2017.

  57. Xu M, Huang Y, Xie L, Peng K, Ding L, Lin L, et al. Diabetes and risk of arterial stiffness: a Mendelian randomization analysis. Diabetes. 2016;65:1731–40.

    Article  CAS  PubMed  Google Scholar 

  58. Xu M, Bi Y, Huang Y, Xie L, Hao M, Zhao Z, et al. Type 2 diabetes, diabetes genetic score and risk of decreased renal function and albuminuria: a Mendelian randomization study. EBioMedicine. 2016;6:162–70.

    Article  PubMed  PubMed Central  Google Scholar 

  59. Merino J, Leong A, Posner DC, Porneala B, Masana L, Dupuis J, Florez JC. Genetically driven hyperglycemia increases risk of coronary artery disease separately from type 2 diabetes. Diabetes Care. 2017.

  60. Ahmad OS, Leong A, Miller JA, Morris JA, Forgetta V, Mujammami M, et al. A Mendelian randomization study of the effect of type-2 diabetes and glycemic traits on bone mineral density. J Bone Miner Res. 2017;32:1072–81.

    Article  CAS  PubMed  Google Scholar 

  61. Hagenaars SP, Gale CR, Deary IJ, Harris SE. Cognitive ability and physical health: a Mendelian randomization study. Sci Rep. 2017;7:2651.

    Article  PubMed  PubMed Central  Google Scholar 

  62. Ostergaard SD, Mukherjee S, Sharp SJ, Proitsi P, Lotta LA, Day F, et al. Alzheimer’s Disease Genetics C, Consortium G, Consortium EP-I, Larson EB, Powell JF, Langenberg C, Crane PK, Wareham NJ, Scott RA. Associations between potentially modifiable risk factors and Alzheimer disease: a Mendelian randomization study. PLoS Med 2015;12:e1001841; discussion e1001841.

  63. Stevens A, De Leonibus C, Hanson D, Dowsey AW, Whatmore A, Meyer S, et al. Network analysis: a new approach to study endocrine disorders. J Mol Endocrinol. 2013;52:R79–93.

    Article  PubMed  CAS  Google Scholar 

  64. •• Newgard CB. Metabolomics and metabolic diseases: where do we stand? Cell Metab. 2017;25:43–56. This is the most recent update on the application of metabolomics to defining predictive biomarkers for incident cardiometabolic diseases.

    Article  CAS  PubMed  Google Scholar 

  65. Peddinti G, Cobb J, Yengo L, Froguel P, Kravic J, Balkau B, et al. Early metabolic markers identify potential targets for the prevention of type 2 diabetes. Diabetologia. 2017.

  66. Fall T, Salihovic S, Brandmaier S, Nowak C, Ganna A, Gustafsson S, et al. Non-targeted metabolomics combined with genetic analyses identifies bile acid synthesis and phospholipid metabolism as being associated with incident type 2 diabetes. Diabetologia. 2016;59:2114–24.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  67. Sun L, Liang L, Gao X, Zhang H, Yao P, Hu Y, et al. Early prediction of developing type 2 diabetes by plasma acylcarnitines: a population-based study. Diabetes Care. 2016;39:1563–70.

    Article  PubMed  Google Scholar 

  68. Drogan D, Dunn WB, Lin W, Buijsse B, Schulze MB, Langenberg C, et al. Untargeted metabolic profiling identifies altered serum metabolites of type 2 diabetes mellitus in a prospective, nested case control study. Clin Chem. 2015;61:487–97.

    Article  CAS  PubMed  Google Scholar 

  69. Zhao J, Zhu Y, Hyun N, Zeng D, Uppal K, Tran VT, et al. Novel metabolic markers for the risk of diabetes development in American Indians. Diabetes Care. 2015;38:220–7.

    Article  CAS  PubMed  Google Scholar 

  70. • Floegel A, Stefan N, Yu Z, Muhlenbruch K, Drogan D, Joost H-G, et al. Identification of serum metabolites associated with risk of type 2 diabetes using a targeted metabolomic approach. Diabetes. 2013;62:639–48. The study investigated the association between serum metabolites measured by targeted metabolomics and risk of T2D in a large prospective case–cohort.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  71. Wang TJ, Ngo D, Psychogios N, Dejam A, Larson MG, Vasan RS, et al. 2-Aminoadipic acid is a biomarker for diabetes risk. J Clin Invest. 2013;123:4309–17.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  72. Ferrannini E, Natali A, Camastra S, Nannipieri M, Mari A, Adam K-P, et al. Early metabolic markers of the development of dysglycemia and type 2 diabetes and their physiological significance. Diabetes. 2013;62:1730–7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  73. Cheng S, Rhee EP, Larson MG, Lewis GD, McCabe EL, Shen D, et al. Metabolite profiling identifies pathways associated with metabolic risk in humans. Circulation. 2012;125:2222–31.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  74. Stancakova A, Civelek M, Saleem NK, Soininen P, Kangas AJ, Cederberg H, et al. Hyperglycemia and a common variant of GCKR are associated with the levels of eight amino acids in 9,369 Finnish men. Diabetes. 2012;61:1895–902.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  75. Wang-Sattler R, Yu Z, Herder C, Messias AC, Floegel A, He Y, et al. Novel biomarkers for pre-diabetes identified by metabolomics. Mol Syst Biol. 2012;8:615.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  76. Rhee EP, Cheng S, Larson MG, Walford GA, Lewis GD, McCabe E, et al. Lipid profiling identifies a triacylglycerol signature of insulin resistance and improves diabetes prediction in humans. J Clin Investig. 2011;121:1402–11.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  77. Wang TJ, Larson MG, Vasan RS, Cheng S, Rhee EP, McCabe E, et al. Metabolite profiles and the risk of developing diabetes. Nat Med. 2011;17:448–53.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  78. Felig P, Marliss E, Cahill GF. Plasma amino acid levels and insulin secretion in obesity. N Engl J Med. 1969;281:811–6.

    Article  CAS  PubMed  Google Scholar 

  79. Guasch-Ferr M, Hruby A, Toledo EA, Clish CB, Martinez-Gonzalez MA, Salas-Salvad J, et al. Metabolomics in prediabetes and diabetes: a systematic review and meta-analysis. Diabetes Care. 2016;39:833–46.

    Article  CAS  Google Scholar 

  80. Menni C, Fauman E, Erte I, Perry JR, Kastenmuller G, Shin SY, et al. Biomarkers for type 2 diabetes and impaired fasting glucose using a nontargeted metabolomics approach. Diabetes. 2013;62:4270–6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  81. Laferrère B, Reilly D, Arias S, Swerdlow N, Gorroochurn P, Bawa B, et al. Differential metabolic impact of gastric bypass surgery versus dietary intervention in obese diabetic subjects despite identical weight loss. Sci Transl Med. 2011;3:80re82.

    Article  CAS  Google Scholar 

  82. Magkos F, Bradley D, Schweitzer GG, Finck BN, Eagon JC, Ilkayeva O, et al. Effect of roux-en-Y gastric bypass and laparoscopic adjustable gastric banding on branched-chain amino acid metabolism. Diabetes. 2013;62:2757–61.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  83. Walford GA, Ma Y, Clish C, Florez JC, Wang TJ, Gerszten RE, et al. Metabolite profiles of diabetes incidence and intervention response in the Diabetes Prevention Program. Diabetes. 2016;65:1424–33.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  84. • Xie W, Wood AR, Lyssenko V, Weedon MN, Knowles JW, Alkayyali S, et al. Genetic variants associated with glycine metabolism and their role in insulin sensitivity and type 2 diabetes. Diabetes 2013;62:2141–2150. This study identified common genetic variants associated with metabolites in insulin sensitivity pathway and showed no robust evidence for association between these variants and insulin resistance or diabetes.

  85. Palmer ND, Stevens RD, Antinozzi PA, Anderson A, Bergman RN, Wagenknecht LE, et al. Metabolomic profile associated with insulin resistance and conversion to diabetes in the Insulin Resistance Atherosclerosis Study. J Clin Endocrinol Metab. 2015;100:E463–8.

    Article  CAS  PubMed  Google Scholar 

  86. Sekhar RV, McKay SV, Patel SG, Guthikonda AP, Reddy VT, Balasubramanyam A, et al. Glutathione synthesis is diminished in patients with uncontrolled diabetes and restored by dietary supplementation with cysteine and glycine. Diabetes Care. 2011;34:162–7.

    Article  CAS  PubMed  Google Scholar 

  87. Wurtz P, Soininen P, Kangas AJ, Ronnemaa T, Lehtimaki T, Kahonen M, et al. Branched-chain and aromatic amino acids are predictors of insulin resistance in young adults. Diabetes Care. 2013;36:648–55.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  88. International Consortium for Blood Pressure Genome-Wide Association Study, Ehret GB, Munroe PB, Rice KM, Bochud M, Johnson AD, et al. Genetic variants in novel pathways influence blood pressure and cardiovascular disease risk. Nature. 2011;478:103–9.

    Article  CAS  Google Scholar 

  89. Muoio DM, Neufer PD. Lipid-induced mitochondrial stress and insulin action in muscle. Cell Metab. 2012;15:595–605.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  90. Aichler M, Borgmann D, Krumsiek J, Buck A, MacDonald PE, Fox JEM, et al. N-acyl taurines and acylcarnitines cause an imbalance in insulin synthesis and secretion provoking β cell dysfunction in type 2 diabetes. Cell Metab. 2017;25:1334–47.e1334

    Article  CAS  PubMed  Google Scholar 

  91. Meikle PJ, Summers SA. Sphingolipids and phospholipids in insulin resistance and related metabolic disorders. Nat Rev Endocrinol. 2017;13:79–91.

    Article  CAS  PubMed  Google Scholar 

  92. Chavez JA, Summers SA. A ceramide-centric view of insulin resistance. Cell Metab. 2012;15:585–94.

    Article  CAS  PubMed  Google Scholar 

  93. Bikman BT, Summers SA. Ceramides as modulators of cellular and whole-body metabolism. J Clin Invest. 2011;121:4222–30.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  94. Turpin SM, Nicholls HT, Willmes DM, Mourier A, Brodesser S, Wunderlich CM, et al. Obesity-induced CerS6-dependent C16:0 ceramide production promotes weight gain and glucose intolerance. Cell Metab. 2014;20:678–86.

    Article  CAS  PubMed  Google Scholar 

  95. Raichur S, Wang ST, Chan PW, Li Y, Ching J, Chaurasia B, et al. CerS2 haploinsufficiency inhibits β-oxidation and confers susceptibility to diet-induced steatohepatitis and insulin resistance. Cell Metab. 2014;20:687–95.

    Article  CAS  PubMed  Google Scholar 

  96. Huang H, Kasumov T, Gatmaitan P, Heneghan HM, Kashyap SR, Schauer PR, et al. Gastric bypass surgery reduces plasma ceramide subspecies and improves insulin sensitivity in severely obese patients. Obesity (Silver Spring, Md). 2011;19:2235–40.

    Article  CAS  Google Scholar 

  97. Barber MN, Risis S, Yang C, Meikle PJ, Staples M, Febbraio MA, et al. Plasma lysophosphatidylcholine levels are reduced in obesity and type 2 diabetes. PLoS One. 2012;7:e41456.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  98. Yore MM, Syed I, Moraes-Vieira PM, Zhang T, Herman MA, Homan EA, et al. Discovery of a class of endogenous mammalian lipids with anti-diabetic and anti-inflammatory effects. Cell. 2014;159:318–32.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  99. Newgard CB, An J, Bain JR, Muehlbauer MJ, Stevens RD, Lien LF, et al. A branched-chain amino acid-related metabolic signature that differentiates obese and lean humans and contributes to insulin resistance. Cell Metab. 2009;9:311–26.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  100. • Newgard CB. Interplay between lipids and branched-chain amino acids in development of insulin resistance. Cell Metab. 2012;15:606–14. This perspective develops a model to explain how lipids and branched-chain amino acids may synergize to promote metabolic diseases.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  101. • Shin SY, Fauman EB, Petersen AK, Krumsiek J, Santos R, Huang J, et al. An atlas of genetic influences on human blood metabolites. Nat Genet. 2014;46:543–50. This study is the most comprehensive exploration of genetic loci influencing human metabolism comprising 7824 adult individuals from 2 European population studies.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  102. • Rhee EP, Ho JE, Chen M-H, Shen D, Cheng S, Larson MG, et al. A genome-wide association study of the human metabolome in a community-based cohort. Cell Metab. 2013;18:130–43. In this study, authors conducted a genome-wide association analysis of 217 plasma metabolites in 2076 participants of the Framingham Heart Study and identified 23 genetic loci for plasma metabolites that have not previously been reported.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  103. Long T, Hicks M, Yu H-C, Biggs WH, Kirkness EF, Menni C, et al. Whole-genome sequencing identifies common-to-rare variants associated with human blood metabolites. Nat Genet. 2017;49:568–78.

    Article  CAS  PubMed  Google Scholar 

  104. Lotta LA, Scott RA, Sharp SJ, Burgess S, Luan J, Tillin T, 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.

    Article  PubMed  PubMed Central  Google Scholar 

  105. Mahendran Y, Jonsson A, Have CT, Allin KH, Witte DR, Jorgensen ME, et al. Genetic evidence of a causal effect of insulin resistance on branched-chain amino acid levels. Diabetologia. 2017;60:873–8.

    Article  CAS  PubMed  Google Scholar 

  106. Kettunen J, Demirkan A, Wurtz P, Draisma HH, Haller T, Rawal R, et al. Genome-wide study for circulating metabolites identifies 62 loci and reveals novel systemic effects of LPA. Nat Commun. 2016;7:11122.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  107. • Walford GA, Porneala BC, Dauriz M, Vassy JL, Cheng S, Rhee EP, et al. Metabolite traits and genetic risk provide complementary information for the prediction of future type 2 diabetes. Diabetes Care. 2014;37:2508–14. This study is one of the first demonstrations that metabolite and genetic traits provide complementary information to each other for the prediction of future type 2 diabetes.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  108. Goff DC Jr, Lloyd-Jones DM, Bennett G, Coady S, D’Agostino RB Sr, Gibbons R, et al. American College of Cardiology/American Heart Association Task Force on Practice G: 2013 ACC/AHA guideline on the assessment of cardiovascular risk: a report of the American College of Cardiology/American Heart Association Task Force on Practice Guidelines. J Am Coll Cardiol. 2014;63:2935–59.

    Article  PubMed  Google Scholar 

  109. Griffin SJ, Borch-Johnsen K, Davies MJ, Khunti K, Rutten GE, Sandbaek A, et al. Effect of early intensive multifactorial therapy on 5-year cardiovascular outcomes in individuals with type 2 diabetes detected by screening (ADDITION-Europe): a cluster-randomised trial. Lancet. 2011;378:156–67.

    Article  PubMed  PubMed Central  Google Scholar 

Download references

Funding

Research is supported by NIDDK U01 DK078616. JBM is also supported by K24 DK080140, and MSU is supported by NIDDK 1K23DK114551-01.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Miriam S. Udler.

Ethics declarations

Conflict of Interest

Jordi Merino, Miriam Udler, Aaron Leong, and James Meigs declare no conflict of interest related to this manuscript.

Human and Animal Rights and Informed Consent

This article does not report any new studies with human or animal subjects performed by any of the authors.

Additional information

This article is part of the Topical Collection on Genetics

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Merino, J., Udler, M.S., Leong, A. et al. A Decade of Genetic and Metabolomic Contributions to Type 2 Diabetes Risk Prediction. Curr Diab Rep 17, 135 (2017). https://doi.org/10.1007/s11892-017-0958-0

Download citation

  • Published:

  • DOI: https://doi.org/10.1007/s11892-017-0958-0

Keywords

Navigation