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:

Fasting drives the metabolic, molecular and geroprotective effects of a calorie-restricted diet in mice

Abstract

Calorie restriction (CR) promotes healthy ageing in diverse species. Recently, it has been shown that fasting for a portion of each day has metabolic benefits and promotes lifespan. These findings complicate the interpretation of rodent CR studies, in which animals typically eat only once per day and rapidly consume their food, which collaterally imposes fasting. Here we show that a prolonged fast is necessary for key metabolic, molecular and geroprotective effects of a CR diet. Using a series of feeding regimens, we dissect the effects of calories and fasting, and proceed to demonstrate that fasting alone recapitulates many of the physiological and molecular effects of CR. Our results shed new light on how both when and how much we eat regulate metabolic health and longevity, and demonstrate that daily prolonged fasting, and not solely reduced caloric intake, is likely responsible for the metabolic and geroprotective benefits of a CR diet.

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: Prolonged fasting is required for the CR-mediated increase in insulin sensitivity and alterations in fuel source selection in male C57BL/6J mice.
Fig. 2: Distinct physiological responses to diet in different strains and sexes of mice.
Fig. 3: Fasting alone recapitulates the metabolic effects of a CR diet.
Fig. 4: Fasting and calorie restriction result in highly similar transcriptomic signatures in liver and white adipose tissue.
Fig. 5: Prolonged fasting is required for the CR-mediated increase in insulin sensitivity and fuel selection in aged mice.
Fig. 6: Fasting is required for the geroprotective effects of a CR diet.
Fig. 7: Fasting plays a critical role in the response to a CR diet.

Similar content being viewed by others

Data availability

RNA-sequencing data have been deposited with the Gene Expression Omnibus and are available under accession number GSE168262. Source data are provided with this paper. Other data that support the plots and findings of this study are available from the corresponding author upon reasonable request.

References

  1. Green, C. L., Lamming, D. W. & Fontana, L. Molecular mechanisms of dietary restriction promoting health and longevity. Nat. Rev. Mol. Cell Biol. https://doi.org/10.1038/s41580-021-00411-4 (2021).

  2. Colman, R. J. et al. Caloric restriction reduces age-related and all-cause mortality in rhesus monkeys. Nat. Commun. 5, 3557 (2014).

    Article  PubMed  CAS  Google Scholar 

  3. Kraus, W. E. et al. 2 years of calorie restriction and cardiometabolic risk (CALERIE): exploratory outcomes of a multicentre, phase 2, randomised controlled trial. Lancet Diabetes Endocrinol. 7, 673–683 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  4. Belsky, D. W., Huffman, K. M., Pieper, C. F., Shalev, I. & Kraus, W. E. Change in the rate of biological aging in response to caloric restriction: CALERIE Biobank analysis. J. Gerontol. A Biol. Sci. Med. Sci. 73, 4–10 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  5. Das, S. K. et al. Body-composition changes in the comprehensive assessment of long-term effects of reducing intake of energy (CALERIE)-2 study: a 2-year randomized controlled trial of calorie restriction in nonobese humans. Am. J. Clin. Nutr. 105, 913–927 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Balasubramanian, P., Howell, P. R. & Anderson, R. M. Aging and caloric restriction research: a biological perspective with translational potential. EBioMedicine 21, 37–44 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  7. Yu, D. et al. Calorie-restriction-induced insulin sensitivity is mediated by adipose mTORC2 and not required for lifespan extension. Cell Rep. 29, 236–248 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Solon-Biet, S. M. et al. The ratio of macronutrients, not caloric intake, dictates cardiometabolic health, aging and longevity in ad libitum-fed mice. Cell Metab. 19, 418–430 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Fontana, L. et al. Decreased consumption of branched-chain amino acids improves metabolic health. Cell Rep. 16, 520–530 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Grandison, R. C., Piper, M. D. & Partridge, L. Amino-acid imbalance explains extension of lifespan by dietary restriction in Drosophila. Nature 462, 1061–1064 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Lu, J. et al. Sestrin is a key regulator of stem cell function and lifespan in response to dietary amino acids. Nat. Aging 1, 60–72 (2021).

    Article  Google Scholar 

  12. Solon-Biet, S. M. et al. Branched chain amino acids impact health and lifespan indirectly via amino acid balance and appetite control. Nat. Metab. 1, 532–545 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Yoshida, S. et al. Role of dietary amino acid balance in diet restriction-mediated lifespan extension, renoprotection, and muscle weakness in aged mice. Aging Cell 17, e12796 (2018).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  14. 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 

  15. Acosta-Rodriguez, V. A., de Groot, M. H. M., Rijo-Ferreira, F., Green, C. B. & Takahashi, J. S. Mice under caloric restriction self-impose a temporal restriction of food intake as revealed by an automated feeder system. Cell Metab. 26, 267–277 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Bruss, M. D., Khambatta, C. F., Ruby, M. A., Aggarwal, I. & Hellerstein, M. K. Calorie restriction increases fatty acid synthesis and whole body fat oxidation rates. Am. J. Physiol. Endocrinol. Metab. 298, E108–E116 (2010).

    Article  CAS  PubMed  Google Scholar 

  17. Longo, V. D. & Mattson, M. P. Fasting: molecular mechanisms and clinical applications. Cell Metab. 19, 181–192 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Hatori, M. et al. Time-restricted feeding without reducing caloric intake prevents metabolic diseases in mice fed a high-fat diet. Cell Metab. 15, 848–860 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Chaix, A., Zarrinpar, A., Miu, P. & Panda, S. Time-restricted feeding is a preventative and therapeutic intervention against diverse nutritional challenges. Cell Metab. 20, 991–1005 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Mitchell, S. J. et al. Daily fasting improves health and survival in male mice independent of diet composition and calories. Cell Metab. 29, 221–228 (2019).

    Article  CAS  PubMed  Google Scholar 

  21. 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 

  22. 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 

  23. Turturro, A. et al. Growth curves and survival characteristics of the animals used in the biomarkers of aging program. J. Gerontol. A Biol. Sci. Med. Sci. 54, B492–B501 (1999).

    Article  CAS  PubMed  Google Scholar 

  24. Nelson, W. & Halberg, F. Meal-timing, circadian rhythms and life span of mice. J. Nutr. 116, 2244–2253 (1986).

    Article  CAS  PubMed  Google Scholar 

  25. Fernandes, G., Yunis, E. J. & Good, R. A. Influence of diet on survival of mice. Proc. Natl Acad. Sci. USA 73, 1279–1283 (1976).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Hempenstall, S., Picchio, L., Mitchell, S. E., Speakman, J. R. & Selman, C. The impact of acute caloric restriction on the metabolic phenotype in male C57BL/6 and DBA/2 mice. Mech. Ageing Dev. 131, 111–118 (2010).

    Article  CAS  PubMed  Google Scholar 

  27. Hasek, B. E. et al. Dietary methionine restriction enhances metabolic flexibility and increases uncoupled respiration in both fed and fasted states. Am. J. Physiol. Regul. Integr. Comp. Physiol. 299, R728–R739 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Abreu-Vieira, G., Xiao, C., Gavrilova, O. & Reitman, M. L. Integration of body temperature into the analysis of energy expenditure in the mouse. Mol. Metab. 4, 461–470 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Haws, S. A., Leech, C. M. & Denu, J. M. Metabolism and the epigenome: a dynamic relationship. Trends Biochem. Sci. https://doi.org/10.1016/j.tibs.2020.04.002 (2020).

  30. Haws, S. A. et al. Methyl-metabolite depletion elicits adaptive responses to support heterochromatin stability and epigenetic persistence. Mol. Cell 78, 210–223(2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Leatham-Jensen, M. et al. Lysine 27 of replication-independent histone H3.3 is required for Polycomb target gene silencing but not for gene activation. PLoS Genet. 15, e1007932 (2019).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  32. Meyer, C., Dostou, J. M., Welle, S. L. & Gerich, J. E. Role of human liver, kidney, and skeletal muscle in postprandial glucose homeostasis. Am. J. Physiol. Endocrinol. Metab. 282, E419–E427 (2002).

    Article  CAS  PubMed  Google Scholar 

  33. Wolfe, R. R. The underappreciated role of muscle in health and disease. Am. J. Clin. Nutr. 84, 475–482 (2006).

    Article  CAS  PubMed  Google Scholar 

  34. Rhoads, T. W. et al. Molecular and functional networks linked to sarcopenia prevention by caloric restriction in rhesus monkeys. Cell Syst. 10, 156–168 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. McKiernan, S. H. et al. Caloric restriction delays aging-induced cellular phenotypes in rhesus monkey skeletal muscle. Exp. Gerontol. 46, 23–29 (2011).

    Article  CAS  PubMed  Google Scholar 

  36. Pugh, T. D. et al. A shift in energy metabolism anticipates the onset of sarcopenia in rhesus monkeys. Aging Cell 12, 672–681 (2013).

    Article  CAS  PubMed  Google Scholar 

  37. Chang, J. et al. Effect of aging and caloric restriction on the mitochondrial proteome. J. Gerontol. A Biol. Sci. Med. Sci. 62, 223–234 (2007).

    Article  PubMed  Google Scholar 

  38. Parks, B. W. et al. Genetic architecture of insulin resistance in the mouse. Cell Metab. 21, 334–347 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Xia, J., Benner, M. J. & Hancock, R. E. NetworkAnalyst–integrative approaches for protein–protein interaction network analysis and visual exploration. Nucleic Acids Res. 42, W167–W174 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Xia, J. et al. INMEX–a web-based tool for integrative meta-analysis of expression data. Nucleic Acids Res. 41, W63–W70 (2013).

    Article  PubMed  PubMed Central  Google Scholar 

  41. Xia, J., Gill, E. E. & Hancock, R. E. NetworkAnalyst for statistical, visual and network-based meta-analysis of gene expression data. Nat. Protoc. 10, 823–844 (2015).

    Article  CAS  PubMed  Google Scholar 

  42. Xia, J., Lyle, N. H., Mayer, M. L., Pena, O. M. & Hancock, R. E. INVEX–a web-based tool for integrative visualization of expression data. Bioinformatics 29, 3232–3234 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Zhou, G. et al. NetworkAnalyst 3.0: a visual analytics platform for comprehensive gene expression profiling and meta-analysis. Nucleic Acids Res. 47, W234–W241 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Kane, A. E. et al. Impact of longevity interventions on a validated mouse clinical frailty index. J. Gerontol. A Biol. Sci. Med. Sci. 71, 333–339 (2016).

    Article  CAS  PubMed  Google Scholar 

  45. Bellantuono, I. et al. A toolbox for the longitudinal assessment of health span in aging mice. Nat. Protoc. 15, 540–574 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Aon, M. A. et al. Untangling determinants of enhanced health and lifespan through a multi-omics approach in mice. Cell Metab. 32, 100–116 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Duffy, P. H. et al. Effect of chronic caloric restriction on physiological variables related to energy metabolism in the male Fischer 344 rat. Mech. Ageing Dev. 48, 117–133 (1989).

    Article  CAS  PubMed  Google Scholar 

  48. Masoro, E. J., McCarter, R. J., Katz, M. S. & McMahan, C. A. Dietary restriction alters characteristics of glucose fuel use. J. Gerontol. 47, B202–B208 (1992).

    Article  CAS  PubMed  Google Scholar 

  49. Green, C. L. et al. The effects of graded levels of calorie restriction: IX. Global metabolomic screen reveals modulation of carnitines, sphingolipids and bile acids in the liver of C57BL/6 mice. Aging Cell 16, 529–540 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Green, C. L. et al. The effects of graded levels of calorie restriction: XIV. Global metabolomics screen reveals brown adipose tissue changes in amino acids, catecholamines, and antioxidants after short-term restriction in C57BL/6 mice. J. Gerontol. A Biol. Sci. Med. Sci. 75, 218–229 (2020).

    Article  CAS  PubMed  Google Scholar 

  51. Green, C. L. et al. The effects of graded levels of calorie restriction: XIII. Global metabolomics screen reveals graded changes in circulating amino acids, vitamins, and bile acids in the plasma of C57BL/6 mice. J. Gerontol. A Biol. Sci. Med. Sci. 74, 16–26 (2019).

    CAS  PubMed  Google Scholar 

  52. Green, C. L. et al. The effects of graded levels of calorie restriction: XVI. Metabolomic changes in the cerebellum indicate activation of hypothalamocerebellar connections driven by hunger responses. J. Gerontol. A Biol. Sci. Med. Sci. 76, 601–610 (2021).

    Article  CAS  PubMed  Google Scholar 

  53. Kokkonen, G. C. & Barrows, C. H. The effect of dietary cellulose on life span and biochemical variables of male mice. Age 11, 7–9 (1988).

    Article  CAS  Google Scholar 

  54. Mair, W., Piper, M. D. & Partridge, L. Calories do not explain extension of life span by dietary restriction in Drosophila. PLoS Biol. 3, e223 (2005).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  55. Greer, E. L. & Brunet, A. Different dietary restriction regimens extend lifespan by both independent and overlapping genetic pathways in C. elegans. Aging Cell 8, 113–127 (2009).

    Article  CAS  PubMed  Google Scholar 

  56. Lyn, J. C., Naikkhwah, W., Aksenov, V. & Rollo, C. D. Influence of two methods of dietary restriction on life history features and aging of the cricket Acheta domesticus. Age 33, 509–522 (2011).

    Article  PubMed  Google Scholar 

  57. Derous, D. et al. The effects of graded levels of calorie restriction: X. Transcriptomic responses of epididymal adipose tissue. J. Gerontol. A Biol. Sci. Med. Sci. 73, 279–288 (2017).

    Article  PubMed Central  CAS  Google Scholar 

  58. Derous, D. et al. The effects of graded levels of calorie restriction: XI. Evaluation of the main hypotheses underpinning the life extension effects of CR using the hepatic transcriptome. Aging 9, 1770–1824 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Froy, O. & Miskin, R. Effect of feeding regimens on circadian rhythms: implications for aging and longevity. Aging 2, 7–27 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  61. Chaix, A., Lin, T., Le, H. D., Chang, M. W. & Panda, S. Time-restricted feeding prevents obesity and metabolic syndrome in mice lacking a circadian clock. Cell Metab. 29, 303–319 (2019).

    Article  CAS  PubMed  Google Scholar 

  62. Sutton, E. F. et al. Early time-restricted feeding improves insulin sensitivity, bood pressure, and oxidative stress even without weight loss in men with prediabetes. Cell Metab. 27, 1212–1221 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  63. Colman, R. J. et al. Caloric restriction delays disease onset and mortality in rhesus monkeys. Science 325, 201–204 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  64. Mattison, J. A. et al. Impact of caloric restriction on health and survival in rhesus monkeys from the NIA study. Nature 489, 318–321 (2012).

    Article  CAS  PubMed  Google Scholar 

  65. 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 

  66. Ramsey, J. J. et al. Dietary restriction and aging in rhesus monkeys: the University of Wisconsin study. Exp. Gerontol. 35, 1131–1149 (2000).

    Article  CAS  PubMed  Google Scholar 

  67. Cienfuegos, S. et al. Effects of 4-h and 6-h time-restricted feeding on weight and cardiometabolic health: a randomized controlled trial in adults with obesity. Cell Metab. 32, 366–378 (2020).

    Article  CAS  PubMed  Google Scholar 

  68. Wilkinson, M. J. et al. Ten-hour time-restricted eating reduces weight, blood pressure, and atherogenic lipids in patients with metabolic syndrome. Cell Metab. 31, 92–104 (2020).

    Article  CAS  PubMed  Google Scholar 

  69. Yokoyama, Y. et al. Erratum for Yokoyama et al., ‘skipping breakfast and risk of mortality from cancer, circulatory diseases and all causes: findings from the Japan Collaborative Cohort Study’. Yonago Acta Med. 62, 308 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  70. Uzhova, I. et al. The importance of breakfast in atherosclerosis disease: insights from the PESA Study. J. Am. Coll. Cardiol. 70, 1833–1842 (2017).

    Article  PubMed  Google Scholar 

  71. Cornelissen, G. When you eat matters: 60 years of Franz Halberg’s nutrition chronomics. Open Nutraceuticals J. 5, 16–44 (2012).

    Article  Google Scholar 

  72. Stote, K. S. et al. A controlled trial of reduced meal frequency without caloric restriction in healthy, normal-weight, middle-aged adults. Am. J. Clin. Nutr. 85, 981–988 (2007).

    Article  CAS  PubMed  Google Scholar 

  73. Carlson, O. et al. Impact of reduced meal frequency without caloric restriction on glucose regulation in healthy, normal-weight middle-aged men and women. Metabolism 56, 1729–1734 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  74. Arnason, T. G., Bowen, M. W. & Mansell, K. D. Effects of intermittent fasting on health markers in those with type 2 diabetes: a pilot study. World J. Diabetes 8, 154–164 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  75. Dommerholt, M. B., Dionne, D. A., Hutchinson, D. F., Kruit, J. K. & Johnson, J. D. Metabolic effects of short-term caloric restriction in mice with reduced insulin gene dosage. J. Endocrinol. 237, 59–71 (2018).

    Article  CAS  PubMed  Google Scholar 

  76. Clasquin, M. F., Melamud, E. & Rabinowitz, J. D. LC-MS data processing with MAVEN: a metabolomic analysis and visualization engine. Curr. Protoc. Bioinformatics https://doi.org/10.1002/0471250953.bi1411s37 (2012).

  77. Melamud, E., Vastag, L. & Rabinowitz, J. D. Metabolomic analysis and visualization engine for LC-MS data. Anal. Chem. https://doi.org/10.1021/ac1021166 (2010).

  78. Whitehead, J. C. et al. A clinical frailty index in aging mice: comparisons with frailty index data in humans. J. Gerontol. A Biol. Sci. Med. Sci. 69, 621–632 (2014).

    Article  PubMed  Google Scholar 

  79. Liang, H. et al. Genetic mouse models of extended lifespan. Exp. Gerontol. 38, 1353–1364 (2003).

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We thank all members of the laboratory of D.W.L., as well as J. Simcox and R. Jain, for their valuable insights and comments. We thank S. Simpson and S. Solon-Biet for advice regarding animal care. We thank T. Herfel (Envigo) for assistance with the formulation of the Diluted AL diet. We thank M. Schaid for critical reading of the manuscript. The laboratory of D.W.L. is supported in part by the National Institutes of Health (NIH)/NIA (AG050135, AG051974, AG056771, AG062328 and AG061635 to D.W.L.), NIH/National Institute of Diabetes and Digestive and Kidney Diseases (DK125859 to D.W.L and J.M.D.) and start-up funds from the UW School of Medicine and Public Health and Department of Medicine (to D.W.L.). Metabolomic and histone proteomic analysis was supported in part by a grant from the NIH (R37GM059785 to J.M.D.) and a UAB Nathan Shock Center of Excellence in the Basic Biology of Aging (P30AG050886) Core Services Pilot Award (to D.W.L.). Bomb calorimetry was supported by S10OD028739 (to C.-L.E.Y.), and gut integrity analysis was supported in part by DK124696 (to C.-L.E.Y.). H.H.P. is supported in part by a NIA F31 pre-doctoral fellowship (AG066311). C.L.G. is supported by a Glenn Foundation for Medical Research Postdoctoral Fellowship and was supported in part by a generous gift from D. Philanthropies. N.E.R. was supported in part by a training grant from the UW Institute on Aging (NIA T32 AG000213). S.A.H. was supported in part by a training grant from the UW Metabolism and Nutrition Training Program (T32 DK007665). Support for this research was provided by the UW Office of the Vice Chancellor for Research and Graduate Education with funding from the Wisconsin Alumni Research Foundation. This work was supported in part by the US Department of Veterans Affairs (I01-BX004031), and this work was supported using facilities and resources from the William S. Middleton Memorial Veterans Hospital. The content is solely the responsibility of the authors and does not necessarily represent the official views of the NIH. This work does not represent the views of the Department of Veterans Affairs or the US Government.

Author information

Authors and Affiliations

Authors

Contributions

Experiments were performed in the laboratories of D.W.L., J.M.D. and C.-L.E.Y. at UW-Madison and in the UAB Nathan Shock Center Mitometabolism Core. All authors participated in the performance of the experiments and/or analysed the data. H.H.P., S.A.H., J.Z., J.M.D. and D.W.L. prepared the manuscript.

Corresponding author

Correspondence to Dudley W. Lamming.

Ethics declarations

Competing interests

D.W.L. has received funding from, and is a scientific advisory board member of, Aeovian Pharmaceuticals, which seeks to develop novel, selective mTOR inhibitors for the treatment of various diseases. J.M.D. is a consultant for FORGE Life Sciences and co-founder of Galilei Bio-Sciences. The remaining authors declare no competing interests.

Additional information

Peer review information Nature Metabolism thanks Leonie Heilbronn and the other, anonymous, reviewer(s) 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 Additional measures of glucose homeostasis in male C57BL/6J mice.

(a) Food Consumption (b) Glucose (AL, n = 12; Diluted AL, n = 10; MF.cr, n = 11; CR, n = 12 biologically independent mice) (c) and insulin (AL, n = 12; Diluted AL, n = 11; MF.cr, n = 10; CR, n = 8 biologically independent mice) tolerance tests after 13 or 14 weeks on the indicated diets. * symbol represents a significant difference versus AL-fed mice (p = 0.0009); # symbol represents a significant difference versus Diluted AL-fed mice (p = 0.0021); * symbol represents a significant difference versus MF.cr-fed mice (p = 0.0013) based on Tukey’s test post one-way ANOVA. (D-F) Fasting blood glucose (d), fasting and glucose-stimulated insulin secretion (15 minutes) (e), and calculated HOMA-IR (F). (d-f) AL, n = 9; Diluted AL, n = 8; MF.cr, n = 9; CR, n = 9 biologically independent mice; * symbol represents a significant difference versus AL-fed mice (Diluted AL, p = 0.0256; CR, p = 0.0127); insulin levels in fasted and glucose-stimulated states were analyzed separately. All data are represented as mean ± SEM.

Source data

Extended Data Fig. 2 Fasting is required for CR-mediated reprogramming of the hepatic metabolome.

Targeted metabolomics were performed on the livers of male C57BL/6J mice fed AL, Diluted AL and CR diets (n = 6 biologically independent mice per diet). a) Heatmap of 59 targeted metabolites, represented as log2-fold change vs. AL-fed mice. b) sPLS-DA of liver metabolomics with CR mice sacrificed in the fasted state. c) sPLS-DA of liver metabolomics with CR sacrificed in the fed state. (d) Relative abundance of methionine and its metabolite S-Adenosyl-Homocysteine. * symbol represents a significant difference versus AL mice (p ≤ 0.0023); # symbol represents a significant difference versus Diluted AL mice (p ≤ 0.0024) based on Tukey’s test post one-way ANOVA. Overlaid box plots show center as median and 25th-75th percentiles; whiskers represent minima and maxima.

Extended Data Fig. 3 Additional hepatic metabolomic data.

(a-c) Hepatic metabolites that showed a statistically significant difference during the fasted or fed state (n = 6 biologically independent mice per diet). a) Relative abundance of nucleotide/nucleoside metabolites. b) Relative abundance of TCA cycle metabolites. c) Relative abundance of amino acid metabolites. (a-c) * symbol represents a significant difference versus AL mice (p ≤ 0.05); # symbol represents a significant difference versus Diluted AL mice (p ≤ 0.05) based on Tukey’s test post one-way ANOVA. Overlaid box plots show center as median and 25th-75th percentiles; whiskers represent minima and maxima.

Extended Data Fig. 4 Fasting is required for CR-mediated reprogramming of the hepatic epigenome.

Histone proteomics were performed on the livers of male C57BL/6J mice fed AL, Diluted AL and CR diets (n = 6 mice per group). a) Heatmap of histone H3 and H4 peptides represented as log2-fold change from AL. B) sPLS-DA of histone modifications. c) Statistically significant modified histones. (a-c) * symbol represents a significant difference versus AL mice (p ≤ 0.05); # symbol represents a significant difference versus Diluted AL mice (p ≤ 0.05) based on Tukey’s test post one-way ANOVA. Overlaid box plots show center as median and 25th-75th percentiles; whiskers represent minima and maxima.

Extended Data Fig. 5 Metabolomic profile of skeletal muscle from AL, Diluted AL and CR mice.

Targeted metabolomics were performed on skeletal muscle from male C57BL/6J mice fed AL, Diluted AL, and CR diets (n = 10 biologically independent mice per diet). a) Heatmap of 28 targeted metabolites, represented as log2-fold change vs. AL-fed mice. b) sPLS-DA of skeletal muscle metabolites. c) Relative abundance of amino acid metabolites. d) Relative abundance of TCA cycle metabolites. (a–d) * symbol represents a significant difference versus AL mice (p ≤ 0.05); # symbol represents a significant difference versus Diluted AL mice (p ≤ 0.05) based on Tukey’s test post one-way ANOVA. Overlaid box plots show center as median and 25th-75th percentiles; whiskers represent minima and maxima.

Extended Data Fig. 6 The effect of three calorie restriction regimens on female C57BL/6J mice.

(a) Outline of feeding regimens: AL, Diluted AL, CR and MF.cr. (b-e) Body composition measurement over 16 weeks on diet (AL, n = 12; Diluted AL, n = 10; MF.cr, n = 12; CR, n = 12 biologically independent mice); total body weight (b), lean mass (c), fat mass (d) and adiposity (e). (f-g) Glucose (n = 12 biologically independent mice per diet) (f) and insulin (AL, n = 12; Diluted AL, n = 12; MF.cr, n = 11; CR, n = 10 biologically independent mice) (g) tolerance tests after 9 or 10 weeks on the indicated diets. * symbol represents a significant difference versus AL-fed mice (Diluted AL, p < 0.0001; MF.cr, p ≤ 0.0012, CR, p < 0.0001); # symbol represents a significant difference versus Diluted AL-fed mice (MF.cr, p = 0.0019; CR, p ≤ 0.0001); @ symbol represents a significant difference versus MF.cr-fed mice (CR, p = 0.0043) based on Tukey’s test post one-way ANOVA. (h-j) Metabolic chamber analysis of mice fed the indicated diets. (h) Respiratory exchange ratio vs. time (n = 12 biologically independent mice per diet) (i) Fuel utilization was calculated for the 24-hour period following the indicated (arrow) refeeding time (n = 12 biologically independent mice per diet). * symbol represents a significant difference versus AL (Diluted AL, p = 0.0255); # symbol represents a significant difference versus Diluted AL (CR, p = 0.0274) based on Tukey’s test post one-way ANOVA performed separately for FAO and C/PO). (j) Energy expenditure as a function of lean mass was calculated for the 24-hour period following the indicated (arrow) refeeding time (n = 12 biologically independent mice per diet, data for each individual mouse is plotted; slopes and intercepts were calculated using ANCOVA). k) Food consumption (AL, n = 12; Diluted AL, n = 12; MF.cr, n = 12; CR, n = 11-12). All data are represented as mean ± SEM.

Source data

Extended Data Fig. 7 The effect of three calorie restriction regimens on male DBA/2J mice.

(a) Outline of feeding regimens: AL, Diluted AL, CR and MF.cr. (b–e) Body composition measurement over 16 weeks on diet (AL, n = 11; Diluted AL, n = 11; MF.cr, n = 12; CR, n = 12 biologically independent mice); total body weight (b), lean mass (c), fat mass (d) and adiposity (e). (f-g) Glucose (AL, n = 12; Diluted AL, n = 12; MF.cr, n = 12; CR, n = 11 biologically independent mice) (f) and insulin (AL, n = 12; Diluted AL, n = 12; MF.cr, n = 12; CR, n = 11 biologically independent mice) (g) tolerance tests after 9 or 10 weeks on the indicated diets. * symbol represents a significant difference versus AL-fed mice (Diluted AL, p = 0.0059; MF.cr, p = 0.0052; CR, p < 0.0001) based on Tukey’s test post one-way ANOVA. (H-J) Metabolic chamber analysis of mice fed the indicated diets. (h) Respiratory exchange ratio vs. time (AL, n = 12; Diluted AL, n = 11; MF.cr, n = 11; CR, n = 11 biologically independent mice) (i) Fuel utilization was calculated for the 24-hour period following the indicated (arrow) refeeding time (AL, n = 12; Diluted AL, n = 11; MF.cr, n = 11; CR, n = 11 biologically independent mice). * symbol represents a significant difference versus AL (Diluted AL, p < 0.0001; MF.cr, p < 0.0001; CR, p < 0.0001); # symbol represents a significant difference versus Diluted AL (MF.cr, p ≤ 0.0002; CR, p ≤ 0.0001) based on Tukey’s test post one-way ANOVA performed separately for FAO and C/PO). (j) Energy expenditure as a function of lean mass was calculated for the 24-hour period following the indicated (arrow) refeeding time (AL, n = 12; Diluted AL, n = 11; MF.cr, n = 11; CR, n = 11 biologically independent mice, data for each individual mouse is plotted; slopes and intercepts were calculated using ANCOVA). K) Food consumption (n = 12 biologically independent mice per diet). All data are represented as mean ± SEM.

Source data

Extended Data Fig. 8 The effect of three calorie restriction regimens on female DBA/2J mice.

(a) Outline of feeding regimens: AL, Diluted AL, CR and MF.cr. (b-e) Body composition measurement over 16 weeks on diet (AL, n = 12; Diluted AL, n = 7; MF.cr, n = 11; CR, n = 12 biologically independent mice); total body weight (b), lean mass (c), fat mass (d) and adiposity (e). (f-g) Glucose (AL, n = 12; Diluted AL, n = 11; MF.cr, n = 12; CR, n = 12 biologically independent mice) (f) and insulin (AL, n = 12; Diluted AL, n = 11; MF.cr, n = 12; CR, n = 12 biologically independent mice) (g) tolerance tests after 9 or 10 weeks on the indicated diets. * symbol represents a significant difference versus AL-fed mice (Diluted AL, p = 0.0033; MF.cr, p = 0.0003; CR, p < 0.0001) based on Tukey’s test post one-way ANOVA. (H-J) Metabolic chamber analysis of mice fed the indicated diets. (h) Respiratory exchange ratio vs. time (AL, n = 11; Diluted AL, n = 11; MF.cr, n = 11; CR, n = 10 biologically independent mice) (i) Fuel utilization was calculated for the 24-hour period following the indicated (arrow) refeeding time (AL, n = 11; Diluted AL, n = 11; MF.cr, n = 11; CR, n = 10 biologically independent mice). * symbol represents a significant difference versus AL (Diluted AL, p < 0.0001; MF.cr, p < 0.0001; CR, p < 0.0001); # symbol represents a significant difference versus Diluted AL (MF.cr, p ≤ 0.0128; CR, p < 0.0001); @ symbol represents a significant difference versus MF.cr (CR, p ≤ 0.0074) based on Tukey’s test post one-way ANOVA performed separately for FAO and C/PO). (J) Energy expenditure as a function of lean mass was calculated for the 24-hour period following the indicated (arrow) refeeding time (AL, n = 11; Diluted AL, n = 11; MF.cr, n = 11; CR, n = 10 biologically independent mice, data for each individual mouse is plotted; slopes and intercepts were calculated using ANCOVA). K) Food consumption (AL, n = 12; Diluted AL, n = 7-12; MF.cr, n = 12; CR, n = 12 biologically independent mice). All data are represented as mean ± SEM.

Source data

Extended Data Fig. 9 Additional data for C57BL/6J male mice fed CR or TR.al diets.

a) Food consumption (n = 12 biologically independent mice per diet). b-e) Body composition (body weight, lean mass, fat mass and adiposity) of C57BL/6J male mice fed the indicated diets for 4 months (n = 12 biologically independent mice per diet; statistics on supplementary table 7). F-G) Glucose (n = 12 biologically independent mice per diet) (f) and insulin (AL, n = 12; TR.al, n = 12; CR, n = 11 biologically independent mice) (g) tolerance tests were performed after 13-14 weeks, respectively on the indicated diets. (h-j) Fasting blood glucose (h), fasting and glucose-stimulated insulin secretion (15 minutes) (i), and calculated HOMA2-IR (j) (AL, n = 12; TR.al, n = 11; CR, n = 12 biologically independent mice). * symbol represents a significant difference versus AL (TR.al, p ≤ 0.0018; CR, p ≤ 0.0477); # symbol represents a significant difference versus TR.al (MF.cr, p ≤ 0.0187; CR, p = 0.0478) based on Tukey’s test post one-way ANOVA. All data are represented as mean ± SEM.

Source data

Extended Data Fig. 10 Food consumption, absorption and gut integrity.

a) Food consumption (AL, n = 27-33; Diluted AL, n = 8-33; CR, n = 30-33 biologically independent mice). b) Food absorption calculation by bomb calorimetry of 19-month-old C57BL/6J male mice fed the indicated diets for 13 months (n = 6 biologically independent mice per diet) c) Gut integrity calculation by FITC-dextran of 20-month-old C57BL/6J male mice (n = 6 biologically independent mice per diet). * symbol represents a significant difference versus AL (CR, p = 0.0160); # symbol represents a significant difference versus Diluted AL (CR, p ≤ 0.0281) based on Tukey’s test post two-way ANOVA) Data are represented as mean ± SEM.

Source data

Supplementary information

Supplementary Information

Supplementary Figs. 1–6

Reporting Summary

Supplemental Tables 1–11

Supplementary Data

Source data for Supplementary Figs. 1–6.

Source data

Source Data Fig. 1

Source data for Fig. 1.

Source Data Fig. 2

Source data for Fig. 2.

Source Data Fig. 3

Source data for Fig. 3.

Source Data Fig. 5

Source data for Fig. 5.

Source Data Fig. 6

Source data for Fig. 6.

Source Data Extended Data Fig. 1

Source data for Extended Data Fig. 1.

Source Data Extended Data Fig. 6

Source data for Extended Data Fig. 6.

Source Data Extended Data Fig. 7

Source data for Extended Data Fig. 7.

Source Data Extended Data Fig. 8

Source data for Extended Data Fig. 8.

Source Data Extended Data Fig. 9

Source data for Extended Data Fig. 9.

Source Data Extended Data Fig. 10

Source data for Extended Data Fig. 10.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Pak, H.H., Haws, S.A., Green, C.L. et al. Fasting drives the metabolic, molecular and geroprotective effects of a calorie-restricted diet in mice. Nat Metab 3, 1327–1341 (2021). https://doi.org/10.1038/s42255-021-00466-9

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s42255-021-00466-9

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