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:

Gene-by-environment modulation of lifespan and weight gain in the murine BXD family

Abstract

How lifespan and body weight vary as a function of diet and genetic differences is not well understood. Here we quantify the impact of differences in diet on lifespan in a genetically diverse family of female mice, split into matched isogenic cohorts fed a low-fat chow diet (CD, n = 663) or a high-fat diet (HFD, n = 685). We further generate key metabolic data in a parallel cohort euthanized at four time points. HFD feeding shortens lifespan by 12%: equivalent to a decade in humans. Initial body weight and early weight gains account for longevity differences of roughly 4–6 days per gram. At 500 days, animals on a HFD typically gain four times as much weight as control, but variation in weight gain does not correlate with lifespan. Classic serum metabolites, often regarded as health biomarkers, are not necessarily strong predictors of longevity. Our data indicate that responses to a HFD are substantially modulated by gene-by-environment interactions, highlighting the importance of genetic variation in making accurate individualized dietary recommendations.

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

Access options

Rent or buy this article

Prices vary by article type

from$1.95

to$39.95

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

Fig. 1: Study overview.
Fig. 2: Diet influence on lifespan in female mice.
Fig. 3: Effect of body weight on lifespan.
Fig. 4: Diet effect on serum metabolites.
Fig. 5: Diet effect on serum metabolic hormones.

Similar content being viewed by others

Data availability

All data conform fully to findable, accessible, interoperable and reusable standards94 and Supplementary Table 1 provides research resource identifiers (www.rrids.org) for all strains. Mean, median and 75% quantile lifespan data from cases and controls are available at GN (www.genenetwork.org) under the headings Species: Mouse; Group: BXD Family; Type: Traits and Cofactors and Dataset: BXD Published Phenotypes (for example, GN traits BXD_18435, 18441, 19451, 19452, 21302 and 21450). Body weight data at 6, 12, 18 and 24 months is also available in GN (for example, traits BXD_19126, 19130, 19131, 19167, 19168, 19169, 19170 and 19171). For example, the following URL with query string parameters will retrieve mean lifespan data for HFD cases: www.genenetwork.org/show_trait?trait_id=18435&dataset=BXDPublish, where the number can be replaced with other ID numbers to obtain and download any data from this work. Organ weight data for a large subset of cases and controls that were euthanized between 6 and 24 months of age (liver, heart, kidneys and brain) are available but these data are only covered briefly here (for example, GN traits BXD_20156, 20157, 20158, 20159, 20353, 20354, 20148, 20149, 20150, 20151, 20146 and 20147). Individual data are also available for all cases, both in the Supplementary table (the precise data used in all analyses here) and in GN under the headings Species: Mouse; Group: BXD NIA Longevity Study; Type: Traits and Cofactors and Dataset: BXD-NIA-Longevity Phenotypes. For example, individual data for lifespan, irrespective of diet, is accessible at www.genenetwork.org/show_trait?trait_id=10002&dataset=BXD-HarvestedPublish. Note that lifespan datasets in GeneNetwork are part of a long-term, and still active genetic study of lifespan in the BXD family, and some datasets will therefore include additional strains as well as outliers excluded from the fixed Supplementary table. Source data are provided with this paper.

Code availability

Source code and raw data used for the fixed-effects linear model, random-effects meta-analysis model, and survival analysis in R are available at https://github.com/genenetwork/bxd_gxelongevity_2020. We have generated a Jupyter notebook as well, detailing our source code with computational and statistical output.

References

  1. Hook, M. et al. Genetic cartography of longevity in humans and mice: current landscape and horizons. Biochim. Biophys. Acta 1864, 2718–2732 (2018).

    Article  CAS  PubMed Central  Google Scholar 

  2. Kuningas, M. et al. Genes encoding longevity: from model organisms to humans. Aging Cell 7, 270–280 (2008).

    Article  CAS  PubMed  Google Scholar 

  3. de Magalhães, J. P., Wuttke, D., Wood, S. H., Plank, M. & Vora, C. Genome-environment interactions that modulate aging: powerful targets for drug discovery. Pharmacol. Rev. 64, 88–101 (2012).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  4. McDaid, A. F. et al. Bayesian association scan reveals loci associated with human lifespan and linked biomarkers. Nat. Commun. 8, 15842 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Fontana, L. & Partridge, L. Promoting health and longevity through diet: from model organisms to humans. Cell 161, 106–118 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Muller, A. P., de Oliveira Dietrich, M., Martimbianco de Assis, A., Souza, D. O. & Portela, L. V. High saturated fat and low carbohydrate diet decreases lifespan independent of body weight in mice. Longev. Heal. 2, 10 (2013).

    Article  Google Scholar 

  7. Kreienkamp, R. et al. Doubled lifespan and patient-like pathologies in progeria mice fed high-fat diet. Aging Cell 18, e12852 (2019).

    Article  PubMed  CAS  Google Scholar 

  8. Heilbronn, L. K. & Ravussin, E. Calorie restriction and aging: review of the literature and implications for studies in humans. Am. J. Clin. Nutr. 78, 361–369 (2003).

    Article  CAS  PubMed  Google Scholar 

  9. Liang, Y. et al. Calorie restriction is the most reasonable anti-ageing intervention: a meta-analysis of survival curves. Sci. Rep. 8, 5779 (2018).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  10. Roberts, M. N. et al. A ketogenic diet extends longevity and healthspan in adult mice. Cell Metab. 26, 539–546.e5 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Speakman, J. R., Mitchell, S. E. & Mazidi, M. Calories or protein? The effect of dietary restriction on lifespan in rodents is explained by calories alone. Exp. Gerontol. 86, 28–38 (2016).

    Article  CAS  PubMed  Google Scholar 

  12. Barrington, W. T. et al. Improving metabolic health through precision dietetics in mice. Genetics 208, 399–417 (2018).

    Article  CAS  PubMed  Google Scholar 

  13. Vaughan, K. L. et al. Caloric restriction study design limitations in rodent and nonhuman primate studies. J. Gerontol. Ser. A 73, 48–53 (2018).

    Article  CAS  Google Scholar 

  14. Finkel, T. & Holbrook, N. J. Oxidants, oxidative stress and the biology of ageing. Nature 408, 239–247 (2000).

    Article  CAS  PubMed  Google Scholar 

  15. Keipert, S., Voigt, A. & Klaus, S. Dietary effects on body composition, glucose metabolism, and longevity are modulated by skeletal muscle mitochondrial uncoupling in mice. Aging Cell 10, 122–136 (2011).

    Article  CAS  PubMed  Google Scholar 

  16. Skorupa, D. A., Dervisefendic, A., Zwiener, J. & Pletcher, S. D. Dietary composition specifies consumption, obesity, and lifespan in Drosophila melanogaster. Aging Cell 7, 478–490 (2008).

    Article  CAS  PubMed  Google Scholar 

  17. Liao, C.-Y., Rikke, B. A., Johnson, T. E., Diaz, V. & Nelson, J. F. Genetic variation in the murine lifespan response to dietary restriction: from life extension to life shortening. Aging Cell 9, 92–95 (2010).

    Article  CAS  PubMed  Google Scholar 

  18. Mitchell, S. J. et al. Effects of sex, strain, and energy intake on hallmarks of aging in mice. Cell Metab. 23, 1093–1112 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Rikke, B. A., Liao, C.-Y., McQueen, M. B., Nelson, J. F. & Johnson, T. E. Genetic dissection of dietary restriction in mice supports the metabolic efficiency model of life extension. Exp. Gerontol. 45, 691–701 (2010).

    Article  PubMed  PubMed Central  Google Scholar 

  20. Azzu, V. & Valencak, T. G. Energy metabolism and ageing in the mouse: a mini-review. Gerontology 63, 327–336 (2017).

    Article  PubMed  Google Scholar 

  21. Pennacchio, L. A. & Rubin, E. M. Comparative genomic tools and databases: providing insights into the human genome. J. Clin. Invest. 111, 1099–1106 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Miller, R. A. et al. An aging interventions testing program: study design and interim report. Aging Cell 6, 565–575 (2007).

    Article  CAS  PubMed  Google Scholar 

  23. Strong, R. et al. Evaluation of resveratrol, green tea extract, curcumin, oxaloacetic acid, and medium-chain triglyceride oil on life span of genetically heterogeneous mice. J. Gerontol. A. Biol. Sci. Med. Sci. 68, 6–16 (2013).

    Article  CAS  PubMed  Google Scholar 

  24. Yuan, R., Peters, L. L. & Paigen, B. Mice as a mammalian model for research on the genetics of aging. ILAR J. Natl Res. Counc. Inst. Lab. Anim. Resour. 52, 4–15 (2011).

    Article  CAS  Google Scholar 

  25. Saul, M. C., Philip, V. M., Reinholdt, L. G. & Chesler, E. J. High-diversity mouse populations for complex traits. Trends Genet. 35, 501–514 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Williams, R. W. Principles of Molecular Medicine (eds. Runge M.S., Patterson C.) 2nd edition, pp 53–60 (Humana Press, 2006).

  27. Williams, R. W. & Williams, E. G. Systems Genetics (eds. Schughart, K. & Williams, R. W.) Vol. 1488, pp 3–29 (Humana Press, 2017).

  28. Williams, R. W. Herding cats: the sociology of data integration. Front. Neurosci. https://doi.org/10.3389/neuro.01.016.2009 (2009).

  29. Peirce, J. L., Lu, L., Gu, J., Silver, L. M. & Williams, R. W. A new set of BXD recombinant inbred lines from advanced intercross populations in mice. BMC Genet. 5, 7 (2004).

    Article  PubMed  PubMed Central  Google Scholar 

  30. Wang, X. et al. Joint mouse–human phenome-wide association to test gene function and disease risk. Nat. Commun. 7, 10464 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Ashbrook, D. G. et al. A platform for experimental precision medicine: the extended BXD mouse family. Cell Syst. 12, 235–247.e9 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Andreux, P. A. et al. Systems genetics of metabolism: the use of the BXD murine reference panel for multiscalar integration of traits. Cell 150, 1287–1299 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. De Haan, G. & Van Zant, G. Genetic analysis of hemopoietic cell cycling in mice suggests its involvement in organismal life span. FASEB J. Publ. Fed. Am. Soc. Exp. Biol. 13, 707–713 (1999).

    Google Scholar 

  34. Gelman, R., Watson, A., Bronson, R. & Yunis, E. Murine chromosomal regions correlated with longevity. Genetics 118, 693–704 (1988).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Houtkooper, R. H. et al. The metabolic footprint of aging in mice. Sci. Rep. https://doi.org/10.1038/srep00134 (2011).

  36. Houtkooper, R. H. et al. Mitonuclear protein imbalance as a conserved longevity mechanism. Nature 497, 451–457 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Williams, E. G. et al. An evolutionarily conserved role for the aryl hydrocarbon receptor in the regulation of movement. PLoS Genet. 10, e1004673 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  38. Lang, D. H. et al. Quantitative trait loci (QTL) analysis of longevity in C57BL/6J by DBA/2J (BXD) recombinant inbred mice. Aging Clin. Exp. Res. 22, 8–19 (2010).

    Article  CAS  PubMed  Google Scholar 

  39. Belknap, J. K. Effect of within-strain sample size on QTL detection and mapping using recombinant inbred mouse strains. Behav. Genet. 28, 29–38 (1998).

    Article  CAS  PubMed  Google Scholar 

  40. Hall, R. A. et al. Systems genetics of liver fibrosis: identification of fibrogenic and expression quantitative trait loci in the BXD murine reference population. PLoS ONE https://doi.org/10.1371/journal.pone.0089279 (2014).

  41. Williams, E. G. et al. Systems proteomics of liver mitochondria function. Science 352, aad0189 (2016).

    Article  PubMed  CAS  Google Scholar 

  42. Wu, Y. et al. Multilayered genetic and omics dissection of mitochondrial activity in a mouse reference population. Cell 158, 1415–1430 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Greenland, S. & Robins, J. M. Identifiability, exchangeability and confounding revisited. Epidemiol. Perspect. Innov. EPI 6, 4 (2009).

    Article  Google Scholar 

  44. Pearl, J. Causal inference in statistics: an overview. Stat. Surv. 3, 96–146 (2009).

    Article  Google Scholar 

  45. Flurkey, K., Currer, J. M. & Harrison, D. E. The Mouse in Biomedical Research 2nd edn Vol. 3 (Elsevier, 2007).

  46. Sandoval-Sierra, J. V. et al. Body weight and high-fat diet are associated with epigenetic aging in female members of the BXD murine family. Aging Cell 19, e13207 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Fuster José, J., Ouchi, N., Gokce, N. & Walsh, K. Obesity-induced changes in adipose tissue microenvironment and their impact on cardiovascular disease. Circ. Res. 118, 1786–1807 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  48. Tchkonia, T. et al. Mechanisms and metabolic implications of regional differences among fat depots. Cell Metab. 17, 644–656 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Austad, S. N. & Fischer, K. E. Sex differences in lifespan. Cell Metab. 23, 1022–1033 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Cheng, C. J., Gelfond, J. A. L., Strong, R. & Nelson, J. F. Genetically heterogeneous mice exhibit a female survival advantage that is age- and site-specific: results from a large multi-site study. Aging Cell 18, e12905 (2019).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  51. Morrison, K. E., Jašarević, E., Howard, C. D. & Bale, T. L. It’s the fiber, not the fat: significant effects of dietary challenge on the gut microbiome. Microbiome 8, 15 (2020).

    Article  PubMed  PubMed Central  Google Scholar 

  52. Mair, W. & Dillin, A. Aging and survival: the genetics of life span extension by dietary restriction. Annu. Rev. Biochem. 77, 727–754 (2008).

    Article  CAS  PubMed  Google Scholar 

  53. Masoro, E. J. Caloric restriction-induced life extension of rats and mice: a critique of proposed mechanisms. Biochim. Biophys. Acta 1790, 1040–1048 (2009).

    Article  CAS  PubMed  Google Scholar 

  54. Weindruch, R., Walford, R. L., Fligiel, S. & Guthrie, D. The retardation of aging in mice by dietary restriction: longevity, cancer, immunity and lifetime energy intake. J. Nutr. 116, 641–654 (1986).

    Article  CAS  PubMed  Google Scholar 

  55. Mattson, M. P. Genes and behavior interact to determine mortality in mice when food is scarce and competition fierce. Aging Cell 9, 448–449 (2010).

    Article  CAS  PubMed  Google Scholar 

  56. Mattison, J. A. et al. Caloric restriction improves health and survival of rhesus monkeys. Nat. Commun. 8, 14063 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Johnson, A. A. & Stolzing, A. The role of lipid metabolism in aging, lifespan regulation, and age-related disease. Aging Cell 18, e13048 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Nakamura, K., Fuster, J. J. & Walsh, K. Adipokines: a link between obesity and cardiovascular disease. J. Cardiol. 63, 250–259 (2014).

    Article  PubMed  Google Scholar 

  59. Calligaris, S. D. et al. Mice long-term high-fat diet feeding recapitulates human cardiovascular alterations: an animal model to study the early phases of diabetic cardiomyopathy. PLoS ONE https://doi.org/10.1371/journal.pone.0060931 (2013).

  60. Manrique, C. et al. Obesity and insulin resistance induce early development of diastolic dysfunction in young female mice fed a western diet. Endocrinology 154, 3632–3642 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Barzilai, N., Huffman, D. M., Muzumdar, R. H. & Bartke, A. The critical role of metabolic pathways in aging. Diabetes 61, 1315–1322 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. Templeman, N. M. et al. Reduced circulating insulin enhances insulin sensitivity in old mice and extends lifespan. Cell Rep. 20, 451–463 (2017).

    Article  CAS  PubMed  Google Scholar 

  63. Barzilai, N. & Ferrucci, L. Insulin resistance and aging: a cause or a protective response? J. Gerontol. Ser. A. 67, 1329–1331 (2012).

    Article  Google Scholar 

  64. Holmes, M. V., Ala-Korpela, M. & Smith, G. D. Mendelian randomization in cardiometabolic disease: challenges in evaluating causality. Nat. Rev. Cardiol. 14, 577–590 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  65. Holmes, M. V. et al. Mendelian randomization of blood lipids for coronary heart disease. Eur. Heart J. 36, 539–550 (2015).

    Article  CAS  PubMed  Google Scholar 

  66. Liu, Z. et al. Associations of triglyceride levels with longevity and frailty: a Mendelian randomization analysis. Sci. Rep. 7, 41579 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  67. Elliott, P. et al. Genetic loci influencing C-reactive protein levels and risk of coronary heart disease. JAMA J. Am. Med. Assoc. 302, 37–48 (2009).

    Article  CAS  Google Scholar 

  68. Menotti, A. & Puddu, P. E. How the Seven Countries Study contributed to the definition and development of the Mediterranean diet concept: a 50-year journey. Nutr. Metab. Cardiovasc. Dis. 25, 245–252 (2015).

    Article  CAS  PubMed  Google Scholar 

  69. Sacks Frank, M. et al. Dietary fats and cardiovascular disease: a presidential advisory from the American Heart Association. Circulation 136, e1–e23 (2017).

    CAS  PubMed  Google Scholar 

  70. Willett, W. C. Diet and cancer. Oncologist 5, 393–404 (2000).

    Article  CAS  PubMed  Google Scholar 

  71. Goncalves, M. D., Hopkins, B. D. & Cantley, L. C. Dietary fat and sugar in promoting cancer development and progression. Ann. Rev. Cancer 21, 255–273 (2019).

    Article  Google Scholar 

  72. Cozzo, A. J., Fuller, A. M. & Makowski, L. Contribution of adipose tissue to development of cancer. Compr. Physiol. 8, 237–282 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  73. Gao, C. et al. Mendelian randomization study of adiposity-related traits and risk of breast, ovarian, prostate, lung and colorectal cancer. Int. J. Epidemiol. 45, 896–908 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  74. Gáliková, M. & Klepsatel, P. Obesity and aging in the Drosophila model. Int. J. Mol. Sci. https://doi.org/10.3390/ijms19071896 (2018).

  75. Otabe, S. et al. Overexpression of human adiponectin in transgenic mice results in suppression of fat accumulation and prevention of premature death by high-calorie diet. Am. J. Physiol. - Endocrinol. Metab. 293, E210–E218 (2007).

    Article  CAS  PubMed  Google Scholar 

  76. Yen, C. A. & Curran, S. P. Gene-diet interactions and aging in C. elegans. Exp. Gerontol. 86, 106–112 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  77. Miller, R. A., Chrisp, C. & Atchley, W. Differential longevity in mouse stocks selected for early life growth trajectory. J. Gerontol. A. Biol. Sci. Med. Sci. 55, B455–B461 (2000).

    Article  CAS  PubMed  Google Scholar 

  78. Kraus, C., Pavard, S. & Promislow, D. E. L. The size–life span trade-off decomposed: why large dogs die young. Am. Nat. 181, 492–505 (2013).

    Article  PubMed  Google Scholar 

  79. Samaras, T. T., Storms, L. H. & Elrick, H. Longevity, mortality and body weight. Ageing Res. Rev. 1, 673–691 (2002).

    Article  PubMed  Google Scholar 

  80. Nüesch, E. et al. Adult height, coronary heart disease and stroke: a multi-locus Mendelian randomization meta-analysis. Int. J. Epidemiol. 45, 1927–1937 (2016).

    Article  PubMed  Google Scholar 

  81. Vitale, G., Pellegrino, G., Vollery, M. & Hofland, L. J. Role of IGF-1 system in the modulation of longevity: controversies and new insights from a centenarians’ perspective. Front. Endocrinol. https://doi.org/10.3389/fendo.2019.00027 (2019).

  82. Choi, Y. J. et al. Adult height in relation to risk of cancer in a cohort of 22,809,722 Korean adults. Br. J. Cancer 120, 668–674 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  83. Nunney, L. Size matters: height, cell number and a person’s risk of cancer. Proc. R. Soc. B. Biol. Sci. 285, 20181743 (2018).

    Article  CAS  Google Scholar 

  84. Wade, K. H., Carslake, D., Sattar, N., Davey Smith, G. & Timpson, N. J. BMI and mortality in UK Biobank: revised estimates using Mendelian randomization. Obesity 26, 1796–1806 (2018).

    Article  CAS  PubMed  Google Scholar 

  85. Jha, P. et al. Systems analyses reveal physiological roles and genetic regulators of liver lipid species. Cell Syst. 6, 722–733.e6 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  86. Jha, P. et al. Genetic regulation of plasma lipid species and their association with metabolic phenotypes. Cell Syst. 6, 709–721.e6 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  87. Williams, E. G. et al. Multi-omic profiling of the liver across diets and age in a diverse mouse population, Cell Systems. Preprint at bioRxiv https://www.biorxiv.org/content/10.1101/2020.08.20.222968v2 (2021).

  88. Almeida-Suhett, C. P., Scott, J. M., Graham, A., Chen, Y. & Deuster, P. A. Control diet in a high-fat diet study in mice: regular chow and purified low-fat diet have similar effects on phenotypic, metabolic, and behavioral outcomes. Nutr. Neurosci. 22, 19–28 (2019).

    Article  CAS  PubMed  Google Scholar 

  89. Ziebarth, J. D. & Cui, Y. Precise network modeling of systems genetics data using the Bayesian Network Webserver. Methods Mol. Biol. 1488, 319–335 (2017).

    Article  CAS  PubMed  Google Scholar 

  90. Ziebarth, J. D., Bhattacharya, A. & Cui, Y. Bayesian Network Webserver: a comprehensive tool for biological network modeling. Bioinformatics 29, 2801–2803 (2013).

    Article  CAS  PubMed  Google Scholar 

  91. Voelkl, B. et al. Reproducibility of animal research in light of biological variation. Nat. Rev. Neurosci. 21, 384–393 (2020).

    Article  CAS  PubMed  Google Scholar 

  92. Viechtbauer, W. Conducting meta-analyses in R with the metafor package. J. Stat. Softw. 36, 1–48 (2010).

    Article  Google Scholar 

  93. Therneau, T. M. & Grambsch, P. M. Modeling Survival Data: Extending the Cox Model (Springer-Verlag, 2000).

  94. Wilkinson, M. D. et al. The FAIR Guiding Principles for scientific data management and stewardship. Sci. Data 3, 160018 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank J.F. Nelson for helpful discussion on the LXS dietary restriction datasets. We thank P. Prins, Z. Sloan and other members of the GeneNetwork team for superb informatics support. Finally, we thank Dr. Elizabeth A Fitzpatrick and team at the Regional Biocontainment Laboratory at UTHSC for generating serum hormone data. This work was supported by grants from the NIH nos. R01AG043930 (R.W.W.), NIH R01AG070913 (R.W.W.), the University of Tennessee Center for Integrative and Translational Genomics (L.L.), the Ecole Polytechnique Fédérale de Lausanne, the European Research Council (AdG-787702) (J.A.), the Swiss National Science Foundation (310030B-160318) (J.A.) and the AgingX programme of the Swiss Initiative for Systems Biology (RTD 2013/153) (J.A.). S.S. was supported by NIH grant no. P30 DA044223-04. E.G.W. was supported by NIH F32 Ruth Kirchstein Fellowship (grant no. F32GM119190). K.M. was supported by NIH grant no. R21 AG055841. R.W.R. was supported by TriMetis Life Sciences, Memphis, TN, USA. L.M. was supported by the American Heart Association and Methodist Mission Support Fund. C.K. was supported by grant no. NIH R01AG054180.

Author information

Authors and Affiliations

Authors

Contributions

E.G.W., S.R., J.A., L.L. and R.W.W. were responsible for the conceptualization. Aging colony management and informatics was done by S.R., J.F.I., C.J.C., M.S.M., A.G.C. Investigations were carried out by S.R., D.G.A., J.F.I., C.J.C., M.S.M., A.G.C., K.M., M.K.M., J.D.Z., W.Z., J.H., S.M.N., L.A.W., T.M.S., C.C.K., Y.C., L.L. and R.W.W. Formal analysis and data curation were done by S.R., D.G.A., M.B.S., P.J., E.G.W., A.S., M.H., R.W.R., S.S. and R.W.W. The original draft was written by S.R. and R.W.W. Review and editing of the paper was done by S.R., M.B.S., E.G.W., K.M., L.M., D.G.A., S.S., R.A.M., J.A. and R.W.W. Companion web resources were provided by A.G.C., S.R., S.S. and R.W.W.

Corresponding author

Correspondence to Robert W. Williams.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

Peer review information Nature Metabolism thanks the anonymous reviewers for their contribution to the peer review of this work. Primary handling editor: Christoph Schmitt.

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

Extended data

Extended Data Fig. 1 Diet effect on lifespan and body weight at 500 days of age.

Related to Fig. 1 and Fig. 3. Diet effect on lifespan and body weight at 500 days of age (A) Data points represent lifespan of animals on low-fat chow diet (CD) in BXDs with n ≥ 4 per strain. Red + denotes the strain median. (B) Data points represent lifespan on the high-fat diet (HFD) in BXDs with n ≥ 4 per strain. Blue + denotes the strain median. (C) Data points represent body weight on CD at 500 days of age in BXDs with n ≥ 4 per strain. Red + denotes the strain median. (D) Data points represent body weight on HFD at 500 days of age in BXDs with n ≥ 4 per strain. Blue + denotes the strain median.

Source data

Extended Data Fig. 2 A Bayesian Network model of the impact of diet on serum metabolites and lifespan and peak body weight at 500 days age.

Edge weights in this network are the weighted fraction of best 1000 Bayesian models that have the same edge and polarity. A value of 1.0 means every one of 1000 “top-ranked models” has this edge. The upper 6 nodes are serum metabolites, and lifespan and body weight at 500 days age are the final outcomes.

Source data

Supplementary information

Source data

Source Data Fig. 1

Statistical 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 Extended Data Fig. 1

Statistical source data.

Source Data Extended Data Fig. 2

Statistical source data.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Roy, S., Sleiman, M.B., Jha, P. et al. Gene-by-environment modulation of lifespan and weight gain in the murine BXD family. Nat Metab 3, 1217–1227 (2021). https://doi.org/10.1038/s42255-021-00449-w

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s42255-021-00449-w

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