Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Article
  • Published:

Variant of the lactase LCT gene explains association between milk intake and incident type 2 diabetes

Abstract

Cow’s milk is frequently included in the human diet, but the relationship between milk intake and type 2 diabetes (T2D) remains controversial. Here, using data from the Hispanic Community Health Study/Study of Latinos, we show that in both sexes, higher milk intake is associated with lower risk of T2D in lactase non-persistent (LNP) individuals (determined by a variant of the lactase LCT gene, single nucleotide polymorphism rs4988235) but not in lactase persistent individuals. We validate this finding in the UK Biobank. Further analyses reveal that among LNP individuals, higher milk intake is associated with alterations in gut microbiota (for example, enriched Bifidobacterium and reduced Prevotella) and circulating metabolites (for example, increased indolepropionate and reduced branched-chain amino acid metabolites). Many of these metabolites are related to the identified milk-associated bacteria and partially mediate the association between milk intake and T2D in LNP individuals. Our study demonstrates a protective association between milk intake and T2D among LNP individuals and a potential involvement of gut microbiota and blood metabolites in this association.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Fig. 1: An overview of study design and main analyses in the HCHS/SOL.
Fig. 2: Milk intake, LCT genotype and incident T2D.
Fig. 3: Milk intake, LCT genotype, gut microbial species and T2D-related metabolic traits.
Fig. 4: Milk intake, LCT genotype, circulating metabolites and risk of T2D.
Fig. 5: LCT genotype-specific milk-associated metabolites explain the milk–LCT interaction on risk of T2D.
Fig. 6: Relationship between LCT genotype-specific milk-associated gut bacterial species and metabolites.

Similar content being viewed by others

Data availability

The genetics data of HCHS/SOL used in this paper are archived at the NIH repositories dbGap (accession number phs000810.v1.p1) and BioLINCC (accession number HLB01141418a), and gut microbiome sequence data in this study are deposited in QIITA (study ID 11666). HCHS/SOL has established a process for the scientific community to apply for access to participant data and materials, with such requests reviewed by the project’s Steering Committee. These policies are described at https://sites.cscc.unc.edu/hchs/ (accessed December 2022). Original data from the UKB and the ARIC used in the present study are available through appropriate application as instructed. Details on data application from the UKB and the ARIC can be found at https://www.ukbiobank.ac.uk/ and https://aric.cscc.unc.edu/aric9/, respectively. The summary statistics on the relationship between gut microbiome and blood metabolites from SCAPIS are publicly available at https://gutsyatlas.serve.scilifelab.se/. Source data are provided with this paper.

Code availability

We used R 4.2 (https://www.r-project.org/), kinship2 v.1.1.3 (https://cran.r-project.org/web/packages/kinship2/), optmatch v.0.10.7 (https://cran.r-project.org/web/packages/optmatch/index.html), SHAPEIT2 v.2.r644 (https://mathgen.stats.ox.ac.uk/genetics_software/shapeit/shapeit.html), IMPUTE2 v.2.3.0 (https://mathgen.stats.ox.ac.uk/impute/impute_v2.html), SHOGUN v.1 (https://github.com/knights-lab/SHOGUN/), ancombc v1.4.0 (https://github.com/FrederickHuangLin/ANCOMBC/), metacoder v.0.3.5 (https://github.com/grunwaldlab/metacoder/), mediation v.4.5.0 (https://cran.r-project.org/web/packages/mediation/), survey v.4.1.1 (https://cran.r-project.org/web/packages/survey/), ppcor v.1.1 (https://cran.r-project.org/web/packages/ppcor/), TwoSampleMR v.0.5.7 (https://mrcieu.github.io/TwoSampleMR/) and mr.raps v.0.4.1 (https://github.com/qingyuanzhao/mr.raps/). No custom codes or functions were generated in this study.

References

  1. Pereira, P. C. Milk nutritional composition and its role in human health. Nutrition 30, 619–627 (2014).

    Article  PubMed  CAS  Google Scholar 

  2. Gijsbers, L. et al. Consumption of dairy foods and diabetes incidence: a dose-response meta-analysis of observational studies. Am. J. Clin. Nutr. 103, 1111–1124 (2016).

    Article  PubMed  CAS  Google Scholar 

  3. Alvarez-Bueno, C. et al. Effects of milk and dairy product consumption on type 2 diabetes: overview of systematic reviews and meta-analyses. Adv. Nutr. 10, S154–S163 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  4. Segurel, L. & Bon, C. On the evolution of lactase persistence in humans. Annu Rev. Genomics Hum. Genet. 18, 297–319 (2017).

    Article  PubMed  CAS  Google Scholar 

  5. Storhaug, C. L., Fosse, S. K. & Fadnes, L. T. Country, regional, and global estimates for lactose malabsorption in adults: a systematic review and meta-analysis. Lancet Gastroenterol. Hepatol. 2, 738–746 (2017).

    Article  PubMed  Google Scholar 

  6. Anguita-Ruiz, A., Aguilera, C. M. & Gil, A. Genetics of lactose intolerance: an updated review and online interactive world maps of phenotype and genotype frequencies. Nutrients 12, 2689 (2020).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  7. Bonder, M. J. et al. The effect of host genetics on the gut microbiome. Nat. Genet. 48, 1407–1412 (2016).

    Article  PubMed  CAS  Google Scholar 

  8. Kurilshikov, A. et al. Large-scale association analyses identify host factors influencing human gut microbiome composition. Nat. Genet. 53, 156–165 (2021).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  9. Qin, Y. et al. Combined effects of host genetics and diet on human gut microbiota and incident disease in a single population cohort. Nat. Genet. 54, 134–142 (2022).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  10. Lopera-Maya, E. A. et al. Effect of host genetics on the gut microbiome in 7,738 participants of the Dutch Microbiome Project. Nat. Genet. 54, 143–151 (2022).

    Article  PubMed  CAS  Google Scholar 

  11. Kitaoka, M. Bifidobacterial enzymes involved in the metabolism of human milk oligosaccharides. Adv. Nutr. 3, 422S–429S (2012).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  12. Chen, J., Chen, X. & Ho, C. L. Recent development of probiotic bifidobacteria for treating human diseases. Front. Bioeng. Biotechnol. 9, 770248 (2021).

    Article  PubMed  PubMed Central  Google Scholar 

  13. Qi, Q. et al. Host and gut microbial tryptophan metabolism and type 2 diabetes: an integrative analysis of host genetics, diet, gut microbiome and circulating metabolites in cohort studies. Gut 71, 1095–1105 (2022).

    Article  PubMed  CAS  Google Scholar 

  14. Roager, H. M. & Licht, T. R. Microbial tryptophan catabolites in health and disease. Nat. Commun. 9, 3294 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  15. Guenther, P. M. et al. The Healthy Eating Index-2010 is a valid and reliable measure of diet quality according to the 2010 Dietary Guidelines for Americans. J. Nutr. 144, 399–407 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  16. Caspi, R. et al. The MetaCyc database of metabolic pathways and enzymes—a 2019 update. Nucleic Acids Res. 48, D445–D453 (2020).

    Article  PubMed  CAS  Google Scholar 

  17. Parks, D. H. et al. A complete domain-to-species taxonomy for bacteria and archaea. Nat. Biotechnol. 38, 1079–1086 (2020).

  18. Cantarel, B. L. et al. The Carbohydrate-Active EnZymes database (CAZy): an expert resource for glycogenomics. Nucleic Acids Res. 37, D233–D238 (2009).

    Article  PubMed  CAS  Google Scholar 

  19. Gacesa, R. et al. Environmental factors shaping the gut microbiome in a Dutch population. Nature 604, 732–739 (2022).

    Article  PubMed  CAS  Google Scholar 

  20. Rothschild, D. et al. Environment dominates over host genetics in shaping human gut microbiota. Nature 555, 210–215 (2018).

    Article  PubMed  CAS  Google Scholar 

  21. de Leeuw, C., Savage, J., Bucur, I. G., Heskes, T. & Posthuma, D. Understanding the assumptions underlying Mendelian randomization. Eur. J. Hum. Genet. 30, 653–660 (2022).

    Article  PubMed  PubMed Central  Google Scholar 

  22. Diener, C. et al. Genome-microbiome interplay provides insight into the determinants of the human blood metabolome. Nat. Metab. 4, 1560–1572 (2022).

    Article  PubMed  PubMed Central  Google Scholar 

  23. Bar, N. et al. A reference map of potential determinants for the human serum metabolome. Nature 588, 135–140 (2020).

    Article  PubMed  Google Scholar 

  24. Chen, L. et al. Influence of the microbiome, diet and genetics on inter-individual variation in the human plasma metabolome. Nat. Med. 28, 2333–2343 (2022).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  25. Gojda, J. & Cahova, M. Gut microbiota as the link between elevated BCAA serum levels and insulin resistance. Biomolecules 11, 1414 (2021).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  26. Guzior, D. V. & Quinn, R. A. Review: microbial transformations of human bile acids. Microbiome 9, 140 (2021).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  27. Guha, S. & Majumder, K. Comprehensive review of gamma-glutamyl peptides (gamma-GPs) and their effect on inflammation concerning cardiovascular health. J. Agric. Food Chem. 70, 7851–7870 (2022).

    Article  PubMed  CAS  Google Scholar 

  28. Agus, A., Planchais, J. & Sokol, H. Gut microbiota regulation of tryptophan metabolism in health and disease. Cell Host Microbe 23, 716–724 (2018).

    Article  PubMed  CAS  Google Scholar 

  29. Koh, A. et al. Microbially produced imidazole propionate impairs insulin signaling through mTORC1. Cell 175, 947–961 (2018).

    Article  PubMed  CAS  Google Scholar 

  30. Dekkers, K. F. et al. An online atlas of human plasma metabolite signatures of gut microbiome composition. Nat. Commun. 13, 5370 (2022).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  31. Mitri, J. et al. Dairy intake and type 2 diabetes risk factors: a narrative review. Diabetes Metab. Syndr. 13, 2879–2887 (2019).

    Article  PubMed  Google Scholar 

  32. Jensen, C. F., Timofeeva, M. & Berg-Beckhoff, G. Milk consumption and the risk of type 2 diabetes: a systematic review of Mendelian randomization studies. Nutr. Metab. Cardiovasc. Dis. 33, 1316–1322 (2023).

    Article  PubMed  CAS  Google Scholar 

  33. Reitmeier, S. et al. Arrhythmic gut microbiome signatures predict risk of type 2 diabetes. Cell Host Microbe 28, 258–272 (2020).

    Article  PubMed  CAS  Google Scholar 

  34. Li, Q. et al. Implication of the gut microbiome composition of type 2 diabetic patients from northern China. Sci. Rep. 10, 5450 (2020).

    Article  PubMed  PubMed Central  Google Scholar 

  35. He, Y. et al. Linking gut microbiota, metabolic syndrome and economic status based on a population-level analysis. Microbiome 6, 172 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  36. Marcobal, A. et al. Bacteroides in the infant gut consume milk oligosaccharides via mucus-utilization pathways. Cell Host Microbe 10, 507–514 (2011).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  37. Sun, F. et al. A potential species of next-generation probiotics? The dark and light sides of Bacteroides fragilis in health. Food Res. Int. 126, 108590 (2019).

    Article  PubMed  CAS  Google Scholar 

  38. Chung, W. S. F. et al. Relative abundance of the Prevotella genus within the human gut microbiota of elderly volunteers determines the inter-individual responses to dietary supplementation with wheat bran arabinoxylan-oligosaccharides. BMC Microbiol. 20, 283 (2020).

    Article  PubMed  PubMed Central  Google Scholar 

  39. Natividad, J. M. et al. Bilophila wadsworthia aggravates high fat diet induced metabolic dysfunctions in mice. Nat. Commun. 9, 2802 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  40. Aslam, H. et al. The effects of dairy and dairy derivatives on the gut microbiota: a systematic literature review. Gut Microbes 12, 1799533 (2020).

    Article  PubMed  PubMed Central  Google Scholar 

  41. Fernandez-Raudales, D. et al. Consumption of different soymilk formulations differentially affects the gut microbiomes of overweight and obese men. Gut Microbes 3, 490–500 (2012).

    Article  PubMed  PubMed Central  Google Scholar 

  42. Precup, G. & Vodnar, D.-C. Gut Prevotella as a possible biomarker of diet and its eubiotic versus dysbiotic roles: a comprehensive literature review. Br. J. Nutr. 122, 131–140 (2019).

  43. Pedersen, H. K. et al. Human gut microbes impact host serum metabolome and insulin sensitivity. Nature 535, 376–381 (2016).

    Article  PubMed  CAS  Google Scholar 

  44. Smith, E. A. & Macfarlane, G. T. Enumeration of human colonic bacteria producing phenolic and indolic compounds: effects of pH, carbohydrate availability and retention time on dissimilatory aromatic amino acid metabolism. J. Appl. Bacteriol. 81, 288–302 (1996).

    Article  PubMed  CAS  Google Scholar 

  45. Crovesy, L., El-Bacha, T. & Rosado, E. L. Modulation of the gut microbiota by probiotics and symbiotics is associated with changes in serum metabolite profile related to a decrease in inflammation and overall benefits to metabolic health: a double-blind randomized controlled clinical trial in women with obesity. Food Funct. 12, 2161–2170 (2021).

    Article  PubMed  CAS  Google Scholar 

  46. Lotta, L. A. 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. 13, e1002179 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  47. Ferrell, J. M. & Chiang, J. Y. L. Understanding bile acid signaling in diabetes: from pathophysiology to therapeutic targets. Diabetes Metab. J. 43, 257–272 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  48. Prawitt, J., Caron, S. & Staels, B. Bile acid metabolism and the pathogenesis of type 2 diabetes. Curr. Diab. Rep. 11, 160–166 (2011).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  49. Wahlstrom, A., Sayin, S. I., Marschall, H. U. & Backhed, F. Intestinal crosstalk between bile acids and microbiota and its impact on host metabolism. Cell Metab. 24, 41–50 (2016).

    Article  PubMed  Google Scholar 

  50. Kaul, A., Mandal, S., Davidov, O. & Peddada, S. D. Analysis of microbiome data in the presence of excess zeros. Front. Microbiol. 8, 2114 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  51. Patti, M. E. et al. Serum bile acids are higher in humans with prior gastric bypass: potential contribution to improved glucose and lipid metabolism. Obes. 17, 1671–1677 (2009).

    Article  CAS  Google Scholar 

  52. Montonen, J., Knekt, P., Jarvinen, R. & Reunanen, A. Dietary antioxidant intake and risk of type 2 diabetes. Diabetes Care 27, 362–366 (2004).

    Article  PubMed  CAS  Google Scholar 

  53. Molinaro, A. et al. Imidazole propionate is increased in diabetes and associated with dietary patterns and altered microbial ecology. Nat. Commun. 11, 5881 (2020).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  54. Chai, J. C. et al. Serum metabolomics of incident diabetes and glycemic changes in a population with high diabetes burden: the Hispanic Community Health Study/Study of Latinos. Diabetes 71, 1338–1349 (2022).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  55. Lavange, L. M. et al. Sample design and cohort selection in the Hispanic Community Health Study/Study of Latinos. Ann. Epidemiol. 20, 642–649 (2010).

    Article  PubMed  PubMed Central  Google Scholar 

  56. Sorlie, P. D. et al. Design and implementation of the Hispanic Community Health Study/Study of Latinos. Ann. Epidemiol. 20, 629–641 (2010).

    Article  PubMed  PubMed Central  Google Scholar 

  57. Qi, Q. et al. Objectively measured sedentary time and cardiometabolic biomarkers in US Hispanic/Latino adults: the Hispanic Community Health Study/Study of Latinos (HCHS/SOL). Circulation 132, 1560–1569 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  58. Siega-Riz, A. M. et al. Food-group and nutrient-density intakes by Hispanic and Latino backgrounds in the Hispanic Community Health Study/Study of Latinos. Am. J. Clin. Nutr. 99, 1487–1498 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  59. Conomos, M. P. et al. Genetic diversity and association studies in US Hispanic/Latino Populations: applications in the Hispanic Community Health Study/Study of Latinos. Am. J. Hum. Genet. 98, 165–184 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  60. Feofanova, E. V. et al. A genome-wide association study discovers 46 loci of the human metabolome in the Hispanic Community Health Study/Study of Latinos. Am. J. Hum. Genet. 107, 849–863 (2020).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  61. Wang, Z. et al. Microbial co-occurrence complicates associations of gut microbiome with US immigration, dietary intake and obesity. Genome Biol. 22, 336 (2021).

    Article  PubMed  PubMed Central  Google Scholar 

  62. Hillmann, B. et al. SHOGUN: a modular, accurate and scalable framework for microbiome quantification. Bioinformatics 36, 4088–4090 (2020).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  63. Zhu, Q. et al. Phylogeny-aware analysis of metagenome community ecology based on matched reference genomes while bypassing taxonomy. mSystems 7, e0016722 (2022).

    Article  PubMed  Google Scholar 

  64. Lin, H. & Peddada, S. D. Analysis of microbial compositions: a review of normalization and differential abundance analysis. NPJ Biofilms Microbiomes 6, 60 (2020).

    Article  PubMed  PubMed Central  Google Scholar 

  65. Sudlow, C. et al. UK Biobank: an open access resource for identifying the causes of a wide range of complex diseases of middle and old age. PLoS Med. 12, e1001779 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  66. Chen, G. C. et al. Association of oily and nonoily fish consumption and fish oil supplements with incident type 2 diabetes: a large population-based prospective study. Diabetes Care 44, 672–680 (2021).

    Article  PubMed  PubMed Central  Google Scholar 

  67. Eastwood, S. V. et al. Algorithms for the capture and adjudication of prevalent and incident diabetes in UK Biobank. PLoS ONE 11, e0162388 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  68. Wright, J. D. et al. The ARIC (Atherosclerosis Risk In Communities) Study: JACC Focus Seminar 3/8. J. Am. Coll. Cardiol. 77, 2939–2959 (2021).

    Article  PubMed  PubMed Central  Google Scholar 

  69. Feofanova, E. V. et al. Whole-genome sequencing analysis of human metabolome in multi-ethnic populations. Nat. Commun. 14, 3111 (2023).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  70. Yin, X. et al. Genome-wide association studies of metabolites in Finnish men identify disease-relevant loci. Nat. Commun. 13, 1644 (2022).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  71. Mahajan, A. et al. Multi-ancestry genetic study of type 2 diabetes highlights the power of diverse populations for discovery and translation. Nat. Genet. 54, 560–572 (2022).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  72. Kurki, M. I. et al. FinnGen provides genetic insights from a well-phenotyped isolated population. Nature 613, 508–518 (2023).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  73. Zhao, Q. Y., Wang, J. S., Hemani, G., Bowden, J. & Small, D. S. Statistical inference in two-sample summary-data Mendelian randomization using robust adjusted profile score. Ann. Stat. 48, 1742–1769 (2020).

    Article  Google Scholar 

Download references

Acknowledgements

The present work is supported by R01-DK119268 (to Q.Q.) and R01-DK126698 (to Q.Q.) from the National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK), and R01-MD011389 (to R.K., R.D.B. and R.C.K.) from the National Institute on Minority Health and Health Disparities. B.Y. is supported by R01-HL141824 and R01-HL142003 from the National Heart, Lung and Blood Institute (NHLBI). J.-Y.M. is supported by BioData Catalyst Fellowship. J.L. is supported by R00-DK122128 from the NIDDK. G.H. is partially supported by R01-DK132011 from the NIDDK and U54GM104940 from the National Institute of General Medical Sciences. K.L. is supported by an AHA postdoctoral fellowship award (23POST1020455). Other funding sources for this study include UM1-HG008898 from the National Human Genome Research Institute; R01-HL060712, R01-HL140976 and R01-HL136266 from the NHLBI; and R01-DK120870 and the New York Regional Center for Diabetes Translation Research (P30-DK111022) from the NIDDK. Support for metabolomics data was graciously provided by the JLH Foundation (Houston, Texas, to B.Y.).

The HCHS/SOL is a collaborative study supported by contracts from the NHLBI to the University of North Carolina (HHSN268201300001I/N01-HC-65233), University of Miami (HHSN268201300004I/N01-HC-65234), Albert Einstein College of Medicine (HHSN268201300002I/N01-HC-65235), University of Illinois at Chicago (HHSN268201300003I/N01-HC-65236 Northwestern University) and San Diego State University (HHSN268201300005I/N01-HC-65237). The following institutes/centres/offices have contributed to the HCHS/SOL through a transfer of funds to the NHLBI: National Institute on Minority Health and Health Disparities, National Institute on Deafness and Other Communication Disorders, National Institute of Dental and Craniofacial Research, NIDDK, National Institute of Neurological Disorders and Stroke and National Institutes of Health (NIH) Institution-Office of Dietary Supplements. We thank the staff and participants of HCHS/SOL for their important contributions. A complete list of staff and investigators is available on the study website at http://www.cscc.unc.edu/hchs/.

The ARIC study has been funded in whole or in part with Federal funds from the NHLBI, NIH, Department of Health and Human Services (contract numbers HHSN268201700001I, HHSN268201700002I, HHSN268201700003I, HHSN268201700004I and HHSN268201700005I), R01-HL087641, R01-HL059367 and R01-HL086694; National Human Genome Research Institute contract number U01HG004402; and NIH contract number HHSN268200625226C. We thank the staff and participants of the ARIC study for their important contributions. Infrastructure was partly supported by grant number UL1RR025005, a component of the NIH and NIH Roadmap for Medical Research. Metabolomics measurements were sponsored by the National Human Genome Research Institute (3U01HG004402-02S1). This research is part of our Diet and Cardiometabolic Health Collection. The funding agencies have no role in the data analyses and results interpretation.

Author information

Authors and Affiliations

Authors

Contributions

K.L. and Q.Q. conceptualized the study. K.L. performed most of the data analysis and drafted the paper. Q.Q. critically revised the paper. G.-C.C., Y.Z., J.-Y.M., J.X., B.A.P., M.U., Z.W., J.L., G.H. and T.W. contributed to data analysis and bioinformatic analysis. E.S., C.M.R., C.R.I., B.Y., R.K., E.B., R.D.B., R.C.K. and Q.Q. contributed to acquisition of the data and funding. All authors interpreted the results and reviewed and edited the paper. R.D.B., R.C.K. and Q.Q. supervised the study.

Corresponding author

Correspondence to Qibin Qi.

Ethics declarations

Competing interests

The authors declare no competing interests.

Peer review

Peer review information

Nature Metabolism thanks the anonymous reviewers for their contribution to the peer review of this work. Primary Handling Editor: Ashley Castellanos-Jankiewicz, in collaboration with the Nature Metabolism team.

Additional information

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

Extended data

Extended Data Fig. 1 Genotype frequency of the LCT-rs4988235 in 1000 Genomes Project Phase 3.

(a) frequencies of genotypes by subpopulations; (b) annotations of subpopulations. Data were accessed on March 27, 2023 through http://useast.ensembl.org/Homo_sapiens/Variation/Population?r=2:135850576-135851576;v=rs4988235;vdb=variation;vf=184227258#population_freq_SAS.

Source data

Extended Data Fig. 2 Abundances of milk intake-associated gut bacterial species across milk intake levels by LCT genotype.

Abundances of milk intake-associated gut bacterial species across milk intake levels (<0.5 serving/day [n = 141 in AA/AG group, and 331 in GG group], 0.5 ~ 1.0 serving/day [n = 210 in AA/AG group and 419 in GG group], > 1 serving/day [n = 286 in AA/AG group and 380 in GG group]) by LCT genotype groups. Data are the least-squares means of centered log-ratio transferred abundances (shown as points) and the corresponding 95% confidence interval (shown as error bars) adjusted for baseline age, sex, study field center, Hispanic/Latino background, physical activity, and education, annual household income, smoking, alcohol consumption, US born status, use of probiotics or antibiotics, prevalent status of hypertension, dyslipedemia, and diabetes at the time of fecal samples collection. P-interaction (Pint) was derived from the test of the product of LCT genotype and milk intake, and P-trend was derived from linear regression with categorical milk intake modeled as an ordinal variable. All statistical tests shown in this figure were two-sided.

Source data

Extended Data Fig. 3 Glycoside hydrolases (GH) family enzymes distribution patterns of identified milk-associated Bifidobacterium species.

Data in the heatmap are abundances of GH enzymes in species, calculated as the ratios of total counts of contained enzymes with GH family to the number of reference genomes (that is, the numbers following the species in the right row annotations). Values < 1 (toward white color) indicate that less than one copy per genome of the corresponding GH family for the examined species, whereas values > 1 refer that more than one copy per reference genome was detected. Annotations of GH family number are from the carbohydrate-active enzymes (CAZy enzyme) database (http://www.cazy.org/Glycoside-Hydrolases.html). The reference genomes of assessed Bifidobacterium species were accessed through the Genome Taxonomy Database (GTDB, release 89; https://gtdb.ecogenomic.org/). Of the originally identified 7 milk-associated Bifidobacterium spp., data of B.reuteri were not available. Annotations of enzyme classes shown in the figure were summarized according to the description on the CAZypedia.org website (https://www.cazypedia.org/index.php/Main_Page).

Source data

Extended Data Fig. 4 Relationship between milk associated bacteria, metabolic traits, and incident T2D.

The top heatmap was retrieved from Fig. 3 in the main text, where bacteria species highlighted in red had positive associations with milk intake, whereas those highlighted in grey were inversely associated with milk intake. Data are partial spearman correlation coefficients adjusted for baseline age, sex, field center, Hispanic/Latino background, physical activity, education, annual household income, smoking, alcohol consumption, US nativity, use of probiotics or antibiotics, prevalent status of hypertension, dyslipidemia, and T2D at the time of fecal sample collection. The bottom forest plot shows the associations of milk-associated bacteria with incident T2D among an interim subsample of HCHS/SOL participants that included 1311 individuals who were free of T2D at the time of stool sample collection but completed the 3rd follow-up visit. Of these 1,311 individuals, 139 incident T2D cases were identified at the 3rd visit. Data are relative risks (RRs, points) and 95% confidence intervals (shown as error bars) estimated from Poisson regression models adjusted for age, sex, filed center, Hispanic/Latino background, US nativity, education, annual household income, smoking, drinking, physical activity, AHEI-2010, use of probiotics and antibiotics at the time of stool sample collection. FDR-q: false discovery rate corrected P-value.

Source data

Extended Data Fig. 5 Associations of milk-associated metabolites with incident T2D in HCHS/SOL and in ARIC.

Results of 42 metabolites in both cohorts were shown. Estimates (relative risk: RR) in HCHS/SOL were derived from survey Poisson regression models adjusted for baseline age, sex, field center, Hispanic/Latino background, education, annual household income, smoking, alcohol consumption, physical activity, prevalent status of hypertension and dyslipidemia. Estimates (harzard risk:HR) in ARIC were derived from Cox proportional hazard regression models adjusted for baseline age, sex, race/ethnicity, smoking, alcohol consumption, education, prevalent status of hypertension and dyslipidemia. Metabolite levels were inverse-normal transformed. Data are effect estimates (shown as points) and 95% confidence intervals (shown as error bars). All statistical tests shown in this figure are two-sided.

Source data

Extended Data Fig. 6 Correlation between GG specific milk-associated bacteria spp. and metabolites.

Data in the left panel were partial spearman correlation adjusted for age, sex, place of birth, study site (Uppsala or Malmö), microbial DNA extraction plate, and metabolomics delivery batch from the Swedish CArdioPulmonary bioImage Study (SCAPIS, n = 8,583) (https://gutsyatlas.serve.scilifelab.se/), while data in the right panel were partial spearman correlation adjusted for baseline age, sex, field center, Hispanic/Latino background, physical activity, education, annual household income, smoking, alcohol consumption, US nativity, use of probiotics or antibiotics, and status of hypertension, dyslipidemia, and T2D at time of fecal sample collection in the present study (HCHS/SOL, n = 804). In SCAPIS, only partial correlations with false discovery rate corrected p-value: FDR-q < 0.05 were available in the summary statistics. Cells with black bold border refer to significant results in both SCAPIS and the present study.

Source data

Extended Data Fig. 7 A summary of key findings on the altered gut microbiota species, circulating metabolites, and metabolic traits associated with milk intake in lactase non-persistent individuals (LNP: LCT GG group).

Created with BioRender.com.

Supplementary information

Supplementary Information

Supplementary Methods and Supplementary Figs. 1–12.

Reporting Summary

Supplementary Tables 1–6.

Source data

Source Data Fig. 2

Statistical source data.

Source Data Fig. 3

Statistical source data.

Source Data Fig. 4

Statistical source data.

Source Data Fig. 5

Statistical source data.

Source Data Fig. 6

Statistical source data.

Source Data Extended Data Fig. 1

Statistical source data.

Source Data Extended Data Fig. 2

Statistical source data.

Source Data Extended Data Fig. 3

Statistical source data.

Source Data Extended Data Fig. 4

Statistical source data.

Source Data Extended Data Fig. 5

Statistical source data.

Source Data Extended Data Fig. 6

Statistical source data.

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

Luo, K., Chen, GC., Zhang, Y. et al. Variant of the lactase LCT gene explains association between milk intake and incident type 2 diabetes. Nat Metab 6, 169–186 (2024). https://doi.org/10.1038/s42255-023-00961-1

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s42255-023-00961-1

This article is cited by

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing