Skip to main content
Log in

Genome-wide association analysis identifies ancestry-specific genetic variation associated with acute response to metformin and glipizide in SUGAR-MGH

  • Article
  • Published:
Diabetologia Aims and scope Submit manuscript

Abstract

Aims/hypothesis

Characterisation of genetic variation that influences the response to glucose-lowering medications is instrumental to precision medicine for treatment of type 2 diabetes. The Study to Understand the Genetics of the Acute Response to Metformin and Glipizide in Humans (SUGAR-MGH) examined the acute response to metformin and glipizide in order to identify new pharmacogenetic associations for the response to common glucose-lowering medications in individuals at risk of type 2 diabetes.

Methods

One thousand participants at risk for type 2 diabetes from diverse ancestries underwent sequential glipizide and metformin challenges. A genome-wide association study was performed using the Illumina Multi-Ethnic Genotyping Array. Imputation was performed with the TOPMed reference panel. Multiple linear regression using an additive model tested for association between genetic variants and primary endpoints of drug response. In a more focused analysis, we evaluated the influence of 804 unique type 2 diabetes- and glycaemic trait-associated variants on SUGAR-MGH outcomes and performed colocalisation analyses to identify shared genetic signals.

Results

Five genome-wide significant variants were associated with metformin or glipizide response. The strongest association was between an African ancestry-specific variant (minor allele frequency [MAFAfr]=0.0283) at rs149403252 and lower fasting glucose at Visit 2 following metformin (p=1.9×10−9); carriers were found to have a 0.94 mmol/l larger decrease in fasting glucose. rs111770298, another African ancestry-specific variant (MAFAfr=0.0536), was associated with a reduced response to metformin (p=2.4×10−8), where carriers had a 0.29 mmol/l increase in fasting glucose compared with non-carriers, who experienced a 0.15 mmol/l decrease. This finding was validated in the Diabetes Prevention Program, where rs111770298 was associated with a worse glycaemic response to metformin: heterozygous carriers had an increase in HbA1c of 0.08% and non-carriers had an HbA1c increase of 0.01% after 1 year of treatment (p=3.3×10−3). We also identified associations between type 2 diabetes-associated variants and glycaemic response, including the type 2 diabetes-protective C allele of rs703972 near ZMIZ1 and increased levels of active glucagon-like peptide 1 (GLP-1) (p=1.6×10−5), supporting the role of alterations in incretin levels in type 2 diabetes pathophysiology.

Conclusions/interpretation

We present a well-phenotyped, densely genotyped, multi-ancestry resource to study gene–drug interactions, uncover novel variation associated with response to common glucose-lowering medications and provide insight into mechanisms of action of type 2 diabetes-related variation.

Data availability

The complete summary statistics from this study are available at the Common Metabolic Diseases Knowledge Portal (https://hugeamp.org) and the GWAS Catalog (www.ebi.ac.uk/gwas/, accession IDs: GCST90269867 to GCST90269899).

Graphical Abstract

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
Fig. 3

Similar content being viewed by others

Abbreviations

AOC:

Area over the curve

DPP:

Diabetes Prevention Program

EAF:

Effect allele frequency

gePS:

Global extended polygenic score

GLP-1:

Glucagon-like peptide 1

GWAS:

Genome-wide association study

LD:

Linkage disequilibrium

MAF:

Minor allele frequency

PC:

Principal component

PP:

Posterior probability

pPS:

Process-specific polygenic score

SUGAR-MGH:

Study to Understand the Genetics of the Acute Response to Metformin and Glipizide in Humans

V1:

Visit 1

V2:

Visit 2

References

  1. Davies MJ, D’Alessio DA, Fradkin J et al (2018) Management of hyperglycemia in type 2 diabetes, 2018. A consensus report by the American Diabetes Association (ADA) and the European Association for the Study of Diabetes (EASD). Diabetes Care 41(12):2669–2701. https://doi.org/10.2337/dci18-0033

    Article  PubMed  PubMed Central  Google Scholar 

  2. Chung WK, Erion K, Florez JC et al (2020) Precision medicine in diabetes: a consensus report from the American Diabetes Association (ADA) and the European Association for the Study of Diabetes (EASD). Diabetes Care 43(7):1617–1635. https://doi.org/10.2337/dci20-0022

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Zhou K, Yee SW, Seiser EL et al (2016) Variation in the glucose transporter gene SLC2A2 is associated with glycemic response to metformin. Nat Genet 48(9):1055–1059. https://doi.org/10.1038/ng.3632

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Zhou K, Bellenguez C, Spencer CC et al (2011) Common variants near ATM are associated with glycemic response to metformin in type 2 diabetes. Nat Genet 43(2):117–120. https://doi.org/10.1038/ng.735

    Article  CAS  PubMed  Google Scholar 

  5. Srinivasan S, Kaur V, Chamarthi B et al (2018) TCF7L2 Genetic variation augments incretin resistance and influences response to a sulfonylurea and metformin: the study to understand the genetics of the acute response to Metformin and Glipizide in Humans (SUGAR-MGH). Diabetes Care 41(3):554–561. https://doi.org/10.2337/dc17-1386

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Zhou K, Donnelly L, Burch L et al (2010) Loss-of-function CYP2C9 variants improve therapeutic response to sulfonylureas in type 2 diabetes: a Go-DARTS study. Clin Pharmacol Ther 87(1):52–56. https://doi.org/10.1038/clpt.2009.176

    Article  CAS  PubMed  Google Scholar 

  7. Dujic T, Zhou K, Donnelly LA, Leese G, Palmer CNA, Pearson ER (2018) Interaction between variants in the CYP2C9 and POR genes and the risk of sulfonylurea-induced hypoglycaemia: A GoDARTS Study. Diabetes Obes Metab 20(1):211–214. https://doi.org/10.1111/dom.13046

    Article  CAS  PubMed  Google Scholar 

  8. Chen L, Li JH, Kaur V et al (2020) The presence of two reduced function variants in CYP2C9 influences the acute response to glipizide. Diabet Med 37(12):2124–2130. https://doi.org/10.1111/dme.14176

    Article  CAS  PubMed  Google Scholar 

  9. Dawed AY, Yee SW, Zhou K et al (2021) Genome-wide meta-analysis identifies genetic variants associated with glycemic response to sulfonylureas. Diabetes Care 44(12):2673–2682. https://doi.org/10.2337/dc21-1152

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Xue A, Wu Y, Zhu Z et al (2018) Genome-wide association analyses identify 143 risk variants and putative regulatory mechanisms for type 2 diabetes. Nat Commun 9(1):2941. https://doi.org/10.1038/s41467-018-04951-w

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Florez JC, Jablonski KA, Bayley N et al (2006) TCF7L2 polymorphisms and progression to diabetes in the Diabetes Prevention Program. N Engl J Med 355(3):241–250. https://doi.org/10.1056/NEJMoa062418

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Lyssenko V, Lupi R, Marchetti P et al (2007) Mechanisms by which common variants in the TCF7L2 gene increase risk of type 2 diabetes. J Clin Invest 117(8):2155–2163. https://doi.org/10.1172/JCI30706

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Villareal DT, Robertson H, Bell GI et al (2010) TCF7L2 variant rs7903146 affects the risk of type 2 diabetes by modulating incretin action. Diabetes 59(2):479–485. https://doi.org/10.2337/db09-1169

    Article  CAS  PubMed  Google Scholar 

  14. Walford GA, Colomo N, Todd JN et al (2015) The study to understand the genetics of the acute response to metformin and glipizide in humans (SUGAR-MGH): design of a pharmacogenetic resource for type 2 diabetes. PloS One 10(3):e0121553. https://doi.org/10.1371/journal.pone.0121553

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Chang CC, Chow CC, Tellier LC, Vattikuti S, Purcell SM, Lee JJ (2015) Second-generation PLINK: rising to the challenge of larger and richer datasets. Gigascience 4:7. https://doi.org/10.1186/s13742-015-0047-8

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Delaneau O, Zagury J-F, Marchini J (2013) Improved whole-chromosome phasing for disease and population genetic studies. Nat Methods 10(1):5–6. https://doi.org/10.1038/nmeth.2307

    Article  CAS  PubMed  Google Scholar 

  17. Taliun D, Harris DN, Kessler MD et al (2021) Sequencing of 53,831 diverse genomes from the NHLBI TOPMed Program. Nature 590(7845):290–299. https://doi.org/10.1038/s41586-021-03205-y

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. R Core Team (2017) R: A language and environment for statistical computing. R Foundation for Statistical Computing. Vienna, Austria

  19. Pruim RJ, Welch RP, Sanna S et al (2010) LocusZoom: regional visualization of genome-wide association scan results. Bioinformatics (Oxford, England) 26(18):2336–2337. https://doi.org/10.1093/bioinformatics/btq419

    Article  CAS  PubMed  Google Scholar 

  20. Vujkovic M, Keaton JM, Lynch JA et al (2020) Discovery of 318 new risk loci for type 2 diabetes and related vascular outcomes among 1.4 million participants in a multi-ancestry meta-analysis. Nat Genet 52(7):680–691. https://doi.org/10.1038/s41588-020-0637-y

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Mahajan A, Spracklen CN, Zhang W et al (2022) Multi-ancestry genetic study of type 2 diabetes highlights the power of diverse populations for discovery and translation. Nat Genet 54(5):560–572. https://doi.org/10.1038/s41588-022-01058-3

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Chen J, Spracklen CN, Marenne G et al (2021) The trans-ancestral genomic architecture of glycemic traits. Nat Genet 53(6):840–860. https://doi.org/10.1038/s41588-021-00852-9

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Machiela MJ, Chanock SJ (2015) LDlink: a web-based application for exploring population-specific haplotype structure and linking correlated alleles of possible functional variants. Bioinformatics (Oxford, England) 31(21):3555–3557. https://doi.org/10.1093/bioinformatics/btv402

    Article  CAS  PubMed  Google Scholar 

  24. Giambartolomei C, Zhenli Liu J, Zhang W et al (2018) A Bayesian framework for multiple trait colocalization from summary association statistics. Bioinformatics (Oxford, England) 34(15):2538–2545. https://doi.org/10.1093/bioinformatics/bty147

    Article  CAS  PubMed  Google Scholar 

  25. Ge T, Chen C-Y, Ni Y, Feng Y-CA, Smoller JW (2019) Polygenic prediction via Bayesian regression and continuous shrinkage priors. Nat Commun 10(1):1776. https://doi.org/10.1038/s41467-019-09718-5

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Udler MS, Kim J, von Grotthuss M et al (2018) Type 2 diabetes genetic loci informed by multi-trait associations point to disease mechanisms and subtypes: A soft clustering analysis. PLoS Med 15(9):e1002654. https://doi.org/10.1371/journal.pmed.1002654

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. The Diabetes Prevention Program Research Group (2000) The Diabetes Prevention Program: baseline characteristics of the randomized cohort. Diabetes Care 23(11):1619–1629. https://doi.org/10.2337/diacare.23.11.1619

    Article  Google Scholar 

  28. The Diabetes Prevention Program (1999) Design and methods for a clinical trial in the prevention of type 2 diabetes. Diabetes Care 22(4):623–634. https://doi.org/10.2337/diacare.22.4.623

    Article  Google Scholar 

  29. Li JH, Perry JA, Jablonski KA et al (2022) Identification of genetic variation influencing metformin response in a multi-ancestry genome-wide association study in the Diabetes Prevention Program (DPP). Diabetes. https://doi.org/10.2337/db22-0702

    Article  PubMed  PubMed Central  Google Scholar 

  30. Sung K-C, Reaven GM, Kim SH (2009) Utility of homeostasis model assessment of β-cell function in predicting diabetes in 12,924 healthy Koreans. Diabetes Care 33(1):200–202. https://doi.org/10.2337/dc09-1070

    Article  PubMed  PubMed Central  Google Scholar 

  31. Song Y, Manson JE, Tinker L et al (2007) Insulin sensitivity and insulin secretion determined by homeostasis model assessment and risk of diabetes in a multiethnic cohort of women: the Women’s Health Initiative Observational Study. Diabetes Care 30(7):1747–1752. https://doi.org/10.2337/dc07-0358

    Article  PubMed  Google Scholar 

  32. Li JH, Szczerbinski L, Dawed AY et al (2021) A polygenic score for type 2 diabetes risk is associated with both the acute and sustained response to sulfonylureas. Diabetes 70(1):293–300. https://doi.org/10.2337/db20-0530

    Article  CAS  PubMed  Google Scholar 

  33. Ohara-Imaizumi M, Aoyagi K, Ohtsuka T (2019) Role of the active zone protein, ELKS, in insulin secretion from pancreatic β-cells. Mol Metab 27(Suppl):S81–S91. https://doi.org/10.1016/j.molmet.2019.06.017

    Article  CAS  PubMed Central  Google Scholar 

  34. Omar-Hmeadi M, Idevall-Hagren O (2021) Insulin granule biogenesis and exocytosis. Cell Mol Life Sci 78(5):1957–1970. https://doi.org/10.1007/s00018-020-03688-4

    Article  CAS  PubMed  Google Scholar 

  35. Wrann CD, Eguchi J, Bozec A et al (2012) FOSL2 promotes leptin gene expression in human and mouse adipocytes. J Clin Invest 122(3):1010–1021. https://doi.org/10.1172/jci58431

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Accelerating Medicines Partnership. T2D Knowledge Portal. Available from https://t2d.hugeamp.org/region.html?chr=2&end=28690184&phenotype=TG&start=28565315. Accessed 21 Sep 2022

  37. Thomsen SK, Ceroni A, van de Bunt M et al (2016) Systematic functional characterization of candidate causal genes for type 2 diabetes risk variants. Diabetes 65(12):3805–3811. https://doi.org/10.2337/db16-0361

    Article  CAS  PubMed  Google Scholar 

  38. Ruetten H, Gebauer M, Raymond RH et al (2018) Mixed meal and intravenous L-arginine tests both stimulate incretin release across glucose tolerance in man: lack of correlation with β cell function. Metab Syndr Relat Disord 16(8):406–415. https://doi.org/10.1089/met.2018.0022

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Dennis JM, Shields BM, Henley WE, Jones AG, Hattersley AT (2019) Disease progression and treatment response in data-driven subgroups of type 2 diabetes compared with models based on simple clinical features: an analysis using clinical trial data. Lancet Diabetes Endocrinol 7(6):442–451. https://doi.org/10.1016/s2213-8587(19)30087-7

    Article  PubMed  PubMed Central  Google Scholar 

  40. The GTEx Consortium (2020) The GTEx Consortium atlas of genetic regulatory effects across human tissues. Science (New York, NY) 369(6509):1318–1330. https://doi.org/10.1126/science.aaz1776

    Article  CAS  Google Scholar 

  41. Shu Y, Brown C, Castro RA et al (2008) Effect of genetic variation in the organic cation transporter 1, OCT1, on metformin pharmacokinetics. Clin Pharmacol Ther 83(2):273–280. https://doi.org/10.1038/sj.clpt.6100275

    Article  CAS  PubMed  Google Scholar 

  42. Zhou K, Donnelly LA, Kimber CH et al (2009) Reduced-function SLC22A1 polymorphisms encoding organic cation transporter 1 and glycemic response to metformin: a GoDARTS study. Diabetes 58(6):1434–1439. https://doi.org/10.2337/db08-0896

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Author information

Authors and Affiliations

Authors

Consortia

Corresponding author

Correspondence to Jose C. Florez.

Ethics declarations

Acknowledgements

We would like to thank J. Del Rio (freelance graphic designer, Cambridge, MA, USA) for his support with Fig. 1 generation. Portions of this study were previously presented as an oral presentation at the 81st Virtual Scientific Sessions of the American Diabetes Association, 25–29 June 2021.

Data availability

The complete summary statistics from this study will be deposited and made available at the Common Metabolic Diseases Knowledge Portal (https://hugeamp.org) and the GWAS Catalog (www.ebi.ac.uk/gwas/, accession IDs: GCST90269867 to GCST90269899) following article publication. Related study documents, including the original study protocol and informed consent forms, are available [14]. Additional data requests should be sent by email to the corresponding author.

Funding

This work was conducted with support from National Institutes of Health/NIDDK awards R01 GM117163, R01 DK088214, R03 DK077675 and P30 DK036836; from the Joslin Clinical Research Center from its philanthropic donors; and from the Harvard Catalyst: the Harvard Clinical and Translational Science Center (National Center for Research Resources and the National Center for Advancing Translational Sciences, NIH Awards M01-RR-01066, 1 UL1 RR025758-04 and 8UL1TR000170-05, and financial contributions from Harvard University and its affiliated academic healthcare centres). JHL received individual support from NIH T32DK007028 and NIDDK K23DK131345. LNB is supported by NIDDK K23DK125839. MSU is supported by NIDDK K23DK114551. AL is supported by grant 2020096 from the Doris Duke Charitable Foundation and the American Diabetes Association grant 7-22-ICTSPM-23. JMM is supported by American Diabetes Association Innovative and Clinical Translational Award 1-19-ICTS-068, American Diabetes Association grant #11-22-ICTSPM-16, and by NHGRI U01HG011723. JCF is supported by NHLBI K24HL157960.

Authors’ relationships and activities

The authors declare that there are no relationships or activities that might bias, or be perceived to bias, their work.

Contribution statement

All authors took part in designing the experiments presented in this manuscript. VK, LNB, MSU, AL and JCF recruited participants in SUGAR-MGH. VK supervised participant recruitment, data collection, and IRB review and approval, and performed DNA extractions and managed GWAS genotyping. Quality control, imputation of the genetic data and GWAS analyses were performed by JMM. JHL, LNB, VK, KF, PS, AH-C and JMM performed follow-up of GWAS data analysis. JHL, LNB, VK, JMM and JCF contributed to the interpretation of the results. JHL, LNB, VK and JMM wrote and prepared the manuscript. All authors revised and approved the final manuscript. JMM and JCF jointly supervised this study. JCF is the guarantor of this work.

Additional information

Publisher's note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Josep M. Mercader and Jose C. Florez jointly directed this work.

Members of the MAGIC Consortium and the DPP Research Group are included as collaborators and listed in the electronic supplementary material (ESM) text.

Supplementary Information

Below is the link to the electronic supplementary material.

Supplementary file1 (PDF 1209 KB)

Rights and permissions

Springer Nature or its licensor (e.g. a society or other partner) holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Li, J.H., Brenner, L.N., Kaur, V. et al. Genome-wide association analysis identifies ancestry-specific genetic variation associated with acute response to metformin and glipizide in SUGAR-MGH. Diabetologia 66, 1260–1272 (2023). https://doi.org/10.1007/s00125-023-05922-7

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s00125-023-05922-7

Keywords

Navigation