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:

A nutritional memory effect counteracts the benefits of dietary restriction in old mice

Abstract

Dietary restriction (DR) during adulthood can greatly extend lifespan and improve metabolic health in diverse species. However, whether DR in mammals is still effective when applied for the first time at old age remains elusive. Here, we report results of a late-life DR-switch experiment using 800 mice. Female mice aged 24 months were switched from an ad libitum (AL) diet to DR or vice versa. Strikingly, the switch from DR to AL acutely increases mortality, whereas the switch from AL to DR causes only a weak and gradual increase in survival, suggesting the body has a memory of earlier nutrition. RNA sequencing in liver and brown and white adipose tissue (BAT and WAT, respectively) demonstrates a largely refractory transcriptional and metabolic response in fat tissue to DR after an AL diet, particularly in WAT, and a proinflammatory signature in aged preadipocytes, which is prevented by chronic DR feeding. Our results provide evidence for a ‘nutritional memory’ as a limiting factor for DR-induced longevity and metabolic remodelling of WAT in mammals.

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: Demography of dietary restriction in mice.
Fig. 2: Post-switch weight change correlates with survival outcome under late-onset DR.
Fig. 3: Detection of an age- and tissue-specific transcriptional memory of prior AL feeding.
Fig. 4: WAT-specific impairment of mitochondrial biogenesis under late-onset DR.
Fig. 5: WAT-specific transcriptional memory predicts impaired activation of de novo lipogenesis under late-onset DR.
Fig. 6: DR remodels lipid flux in adipose tissue.
Fig. 7: Chronic, but not late-onset, DR reprograms lipid synthesis to promote mitochondrial membrane synthesis.

Similar content being viewed by others

Data availability

The data that support the findings of this study are available from the corresponding authors upon request. Raw bulk RNA-sequencing data are available under accession numbers GSE92486 and GSE124772 on the NCBI Gene Expression Omnibus database. Analysed lipidomics data are available under Supplementary Table 5.

References

  1. 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 

  2. Hine, C. et al. Endogenous hydrogen sulfide production is essential for dietary restriction benefits. Cell 160, 132–144 (2015).

    Article  CAS  PubMed  Google Scholar 

  3. Fabbiano, S. et al. Caloric restriction leads to browning of white adipose tissue through type 2 immune signaling. Cell Metab. 24, 434–446 (2016).

    Article  CAS  PubMed  Google Scholar 

  4. Kobara, M. et al. Short-term caloric restriction suppresses cardiac oxidative stress and hypertrophy caused by chronic pressure overload. J. Card. Fail. 21, 656–666 (2015).

    Article  CAS  PubMed  Google Scholar 

  5. Partridge, L., Deelen, J. & Slagboom, P. E. Facing up to the global challenges of ageing. Nature 561, 45–56 (2018).

    Article  CAS  PubMed  Google Scholar 

  6. Mair, W., Goymer, P., Pletcher, S. D. & Partridge, L. Demography of dietary restriction and death in Drosophila. Science 301, 1731–1733 (2003).

    Article  CAS  PubMed  Google Scholar 

  7. Weindruch, R. The retardation of aging by caloric restriction: studies in rodents and primates. Toxicol. Pathol. 24, 742–745 (1996).

    Article  CAS  PubMed  Google Scholar 

  8. Merry, B. J., Kirk, A. J. & Goyns, M. H. Dietary lipoic acid supplementation can mimic or block the effect of dietary restriction on life span. Mech. Ageing Dev. 129, 341–348 (2008).

    Article  CAS  PubMed  Google Scholar 

  9. Forster, M. J., Morris, P. & Sohal, R. S. Genotype and age influence the effect of caloric intake on mortality in mice. FASEB J. 17, 690–692 (2003).

    Article  PubMed  CAS  Google Scholar 

  10. Dhahbi, J. M., Kim, H.-J., Mote, P. L., Beaver, R. J. & Spindler, S. R. Temporal linkage between the phenotypic and genomic responses to caloric restriction. Proc. Natl Acad. Sci. USA 101, 5524–5529 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Vaupel, J. W. et al. Biodemographic trajectories of longevity. Science 280, 855–860 (1998).

    Article  CAS  PubMed  Google Scholar 

  12. Carey, J. R. What demographers can learn from fruit fly actuarial models and biology. Demography 34, 17–30 (1997).

    Article  CAS  PubMed  Google Scholar 

  13. Jiang, T., Liebman, S. E., Lucia, M. S., Phillips, C. L. & Levi, M. Calorie restriction modulates renal expression of sterol regulatory element binding proteins, lipid accumulation, and age-related renal disease. J. Am. Soc. Nephrol. 16, 2385–2394 (2005).

    Article  CAS  PubMed  Google Scholar 

  14. Swindell, W. R. Genes and gene expression modules associated with caloric restriction and aging in the laboratory mouse. BMC Genomics 10, 585 (2009).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  15. Kuhla, A., Blei, T., Jaster, R. & Vollmar, B. Aging Is Associated With a Shift of Fatty Metabolism Toward Lipogenesis. J. Gerontol. A Biol. Sci. Med. Sci. 66A, 1192–1200 (2011).

    Article  CAS  Google Scholar 

  16. Gillespie, Z. E., Pickering, J. & Eskiw, C. H. Better living through chemistry: Caloric restriction (CR) and CR mimetics alter genome function to promote increased health and lifespan. Front. Genet. 7, 142 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  17. Rhoads, T. W. et al. Caloric restriction engages hepatic RNA processing mechanisms in rhesus monkeys. Cell Metab. 27, 677–688.e5 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Plank, M., Wuttke, D., van Dam, S., Clarke, S. A. & de Magalhães, J. P. A meta-analysis of caloric restriction gene expression profiles to infer common signatures and regulatory mechanisms. Mol. Biosyst 8, 1339–1349 (2012).

    Article  CAS  PubMed  Google Scholar 

  19. Papsdorf, K. & Brunet, A. Linking lipid metabolism to chromatin regulation in aging. Trends Cell Biol. https://doi.org/10.1016/j.tcb.2018.09.004 (2018).

    Article  CAS  PubMed  Google Scholar 

  20. 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 

  21. Liao, C.-Y. et al. Fat maintenance is a predictor of the murine lifespan response to dietary restriction. Aging Cell 10, 629–639 (2011).

    Article  CAS  PubMed  Google Scholar 

  22. Hahn, O. et al. Dietary restriction protects from age-associated DNA methylation and induces epigenetic reprogramming of lipid metabolism. Genome Biol. 18, 56 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  23. Charles, K. N. et al. Uncoupling of metabolic health from longevity through genetic alteration of adipose tissue lipid-binding proteins. Cell Rep. 21, 393–402 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. 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).

  25. Rogers, N. H. & Smith, R. G. Brown-to-white transition in subcutaneous fat: linking aging and disease. Aging (Albany NY) 4, 728–729 (2012).

    Article  CAS  Google Scholar 

  26. Berry, D. C. et al. Cellular aging contributes to failure of cold-induced beige adipocyte formation in old mice and humans. Cell Metab. 25, 166–181 (2017).

    Article  CAS  PubMed  Google Scholar 

  27. Wang, W. et al. A PRDM16-driven metabolic signal from adipocytes regulates precursor cell fate. Cell Metab. 30, 174–189.e5 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Pisco, A. O. et al. A Single cell transcriptomic atlas characterizes aging tissues in the mouse. Preprint at https://doi.org/10.1101/661728 (2019).

  29. Konishi, M. et al. Role of Fgf10 in cell proliferation in white adipose tissue. Mol. Cell. Endocrinol. 249, 71–77 (2006).

    Article  CAS  PubMed  Google Scholar 

  30. Petrus, P. et al. Transforming growth factor-β3 regulates adipocyte number in subcutaneous white adipose tissue. Cell Rep. 25, 551–560.e5 (2018).

    Article  CAS  PubMed  Google Scholar 

  31. Townsend, K. L. & Tseng, Y.-H. Brown fat fuel utilization and thermogenesis. Trends Endocrinol. Metab. 25, 168–177 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Kazak, L. et al. A creatine-driven substrate cycle enhances energy expenditure and thermogenesis in beige fat. Cell 163, 643–655 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Ikeda, K. et al. UCP1-independent signaling involving SERCA2b-mediated calcium cycling regulates beige fat thermogenesis and systemic glucose homeostasis. Nat. Med. 23, 1454–1465 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Wang, Y., Viscarra, J., Kim, S.-J. & Sul, H. S. Transcriptional regulation of hepatic lipogenesis. Nat. Rev. Mol. Cell Biol. 16, 678–689 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. van Meer, G., Voelker, D. R. & Feigenson, G. W. Membrane lipids: where they are and how they behave. Nat. Rev. Mol. Cell Biol. 9, 112–124 (2008).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  36. Shindou, H. & Shimizu, T. Acyl-CoA:Lysophospholipid acyltransferases. J. Biol. Chem. 284, 1–5 (2008).

    Article  PubMed  CAS  Google Scholar 

  37. Potting, C. et al. TRIAP1/PRELI complexes prevent apoptosis by mediating intramitochondrial transport of phosphatidic acid. Cell Metab. 18, 287–295 (2013).

    Article  CAS  PubMed  Google Scholar 

  38. Nguyen, A., Rudge, S. A., Zhang, Q. & Wakelam, M. J. Using lipidomics analysis to determine signalling and metabolic changes in cells. Curr. Opin. Biotechnol. 43, 96–103 (2017).

    Article  CAS  PubMed  Google Scholar 

  39. Zimmermann, R. et al. Fat mobilization in adipose tissue is promoted by adipose triglyceride lipase. Science 306, 1383–1386 (2004).

    Article  CAS  PubMed  Google Scholar 

  40. Haemmerle, G. et al. Defective lipolysis and altered energy metabolism in mice lacking adipose triglyceride lipase. Science 312, 734–737 (2006).

    Article  CAS  PubMed  Google Scholar 

  41. van der Veen, J. N. et al. The critical role of phosphatidylcholine and phosphatidylethanolamine metabolism in health and disease. BBA-Biomembranes 1859, 1558–1572 (2017).

    Article  PubMed  CAS  Google Scholar 

  42. Osman, C., Haag, M., Wieland, F. T., Brügger, B. & Langer, T. A mitochondrial phosphatase required for cardiolipin biosynthesis: the PGP phosphatase Gep4. EMBO J. 29, 1976–1987 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Tamura, Y. et al. Role for two conserved intermembrane space proteins, Ups1p and Ups2p, [corrected] in intra-mitochondrial phospholipid trafficking. J. Biol. Chem. 287, 15205–15218 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Saita, S. et al. PARL partitions the lipid transfer protein STARD7 between the cytosol and mitochondria. EMBO J. 37, e97909 (2018).

  45. Horibata, Y. & Sugimoto, H. StarD7 mediates the intracellular trafficking of phosphatidylcholine to mitochondria. J. Biol. Chem. 285, 7358–7365 (2010).

    Article  CAS  PubMed  Google Scholar 

  46. Horibata, Y. et al. StarD7 Protein deficiency adversely affects the phosphatidylcholine composition, respiratory activity, and cristae structure of mitochondria. J. Biol. Chem. 291, 24880–24891 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Harrison, D. E. et al. Rapamycin fed late in life extends lifespan in genetically heterogeneous mice. Nature 460, 392–395 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Ikeno, Y. et al. Do Ames dwarf and calorie-restricted mice share common effects on age-related pathology? Pathobiol. Aging Age Relat. Dis. 3, 20833 (2013).

    Article  Google Scholar 

  49. Brandhorst, S. & Longo, V. D. Fasting and caloric restriction in cancer prevention and treatment. Recent Results Cancer Res. 207, 241–266 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Zhang, C. et al. Structural modulation of gut microbiota in life-long calorie-restricted mice. Nat. Commun. 4, 2163 (2013).

    Article  PubMed  CAS  Google Scholar 

  51. Fabbiano, S. et al. Functional gut microbiota remodeling contributes to the caloric restriction-induced metabolic improvements. Cell Metab. 28, 907–921 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Florian, M. C. et al. A canonical to non-canonical Wnt signalling switch in haematopoietic stem-cell ageing. Nature 503, 392–396 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Kalamakis, G. et al. Quiescence modulates stem cell maintenance and regenerative capacity in the aging brain. Cell 176, 1407–1419.e14 (2019).

    Article  CAS  PubMed  Google Scholar 

  54. Lee, M.-W. et al. Activated type 2 innate lymphoid cells regulate beige fat biogenesis. Cell 160, 74–87 (2015).

    Article  CAS  PubMed  Google Scholar 

  55. Huang, Z. et al. The FGF21–CCL11 axis mediates beiging of white adipose tissues by coupling sympathetic nervous system to type 2 immunity. Cell Metab. 26, 493–508.e4 (2017).

    Article  CAS  PubMed  Google Scholar 

  56. Wang, W. & Seale, P. Control of brown and beige fat development. Nat. Rev. Mol. Cell Biol. 17, 691–702 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Schaum, N. et al. The murine transcriptome reveals global aging nodes with organ-specific phase and amplitude. Preprint at https://doi.org/10.1101/662254 (2019).

  58. Fujii, N. et al. Sterol regulatory element-binding protein-1c orchestrates metabolic remodeling of white adipose tissue by caloric restriction. Aging Cell 16, 508–517 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Soare, A., Cangemi, R., Omodei, D., Holloszy, J. O. & Fontana, L. Long-term calorie restriction, but not endurance exercise, lowers core body temperature in humans. Aging (Albany NY) 3, 374–379 (2011).

    Article  Google Scholar 

  60. Lapa, C. et al. Whitening and impaired glucose utilization of brown adipose tissue in a rat model of type 2 diabetes mellitus. Sci. Rep. 7, 16795 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  61. Corrales, P. et al. Long-term caloric restriction ameliorates deleterious effects of aging on white and brown adipose tissue plasticity. Aging Cell 18, e12948 (2019).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  62. Valle, A., García-Palmer, F. J., Oliver, J. & Roca, P. Sex differences in brown adipose tissue thermogenic features during caloric restriction. Cell. Physiol. Biochem. 19, 195–204 (2007).

    Article  CAS  PubMed  Google Scholar 

  63. Cao, H. et al. Identification of a lipokine, a lipid hormone linking adipose tissue to systemic metabolism. Cell 134, 933–944 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  64. Lynes, M. D. et al. Cold-activated lipid dynamics in adipose tissue highlights a role for cardiolipin in thermogenic. metabolism. Cell Rep. 24, 781–790 (2018).

    Article  CAS  PubMed  Google Scholar 

  65. Sustarsic, E. G. et al. Cardiolipin synthesis in brown and beige fat mitochondria is essential for systemic energy homeostasis. Cell Metab. 28, 159–174.e11 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  66. Tain, L. S. et al. A proteomic atlas of insulin signalling reveals tissue-specific mechanisms of longevity assurance. Mol. Syst. Biol. 13, 939 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  67. Kajimura, S., Spiegelman, B. M. & Seale, P. Brown and beige fat: physiological roles beyond heat generation. Cell Metab. 22, 546–559 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  68. Sultan, M. et al. A simple strand-specific RNA-Seq library preparation protocol combining the Illumina TruSeq RNA and the dUTP methods. Biochem. Biophys. Res. Commun. 422, 643–646 (2012).

    Article  CAS  PubMed  Google Scholar 

  69. Kim, D. et al. TopHat2: accurate alignment of transcriptomes in the presence of insertions, deletions and gene fusions. Genome Biol. 14, R36 (2013).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  70. Love, M. I., Huber, W. & Anders, S. Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2. Genome Biol. 15, 550 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  71. Alexa, A. & Rahnenfuhrer, J. Bioconductor - topGO, R package version (2010).

  72. Stuart, T. et al. Comprehensive integration of single-cell data. Cell 177, 1888–1902.e21 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  73. Schindelin, J. et al. Fiji: an open-source platform for biological-image analysis. Nat. Meth. 9, 676–682 (2012).

    Article  CAS  Google Scholar 

  74. Peck, B. et al. Inhibition of fatty acid desaturation is detrimental to cancer cell survival in metabolically compromised environments. Cancer Metab. 4, 6 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  75. Clark, J. et al. Quantification of PtdInsP3 molecular species in cells and tissues by mass spectrometry. Nat. Meth. 8, 267–272 (2011).

    Article  CAS  Google Scholar 

  76. Bligh, E. G. & Dyer, W. J. A rapid method of total lipid extraction and purification. Can. J. Biochem. Physiol. 37, 911–917 (1959).

    Article  CAS  PubMed  Google Scholar 

  77. Fabregat, A. et al. The Reactome Pathway Knowledgebase. Nucleic Acids Res. 46, D649–D655 (2018).

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We thank R. Weindruch, J. Nelson, R. Miller, C. Selman, D. Withers and F. Kiefer for their advice on the mouse dietary restriction protocol and Dietmar Vestweber and the Max Planck Institute for Molecular Biomedicine, Münster, Germany, for kindly allowing us to conduct our studies at their facilities. We further thank I. Gravemeier, U. Hill, J. Matutat, A. Mesaros and B. Neuhaus for assistance with mouse work. We thank T. Wyss-Coray and the Tabula Muris consortium for kindly granting access to their single-cell transcriptome atlas, and for advice and supervision during the corresponding analysis. We acknowledge funding from the Max Planck Society, Bundesministerium für Bildung und Forschung Grant SyBACol 0315893A-B (to AB and LP) and the European Research Council under the European Union’s Seventh Framework Programme (FP7/2007-2013)/ERC grant agreement number 268739 to L.P. M.J.O.W., A.N. and Q.Z. thank the BBSRC (BB/P013384/1) and the MRC (MR/M004821/1) for financial support.

Author information

Authors and Affiliations

Authors

Contributions

S.G., A.B., M.J.O.W. and L.P. designed the experiments and drafted the manuscript together with O. Hahn. S.P. conceptualized and performed power analyses to determine the required number of animals for the switch experiments, and guided the mortality data analysis. O. Hendrich performed the RNA-seq. L.F.D. performed qPCR and western blot experiments. T.T. and T.L. designed and conducted the in vitro pulse–chase experiments with L.F.D. and L.G. Q.Z. and M.J.O.W. conducted the lipidomic profiling. A.N. conceptualized and performed the lipidome network analysis. O. Hahn performed most of the lifespan and bioinformatic analyses. All authors read and approved the final manuscript.

Corresponding authors

Correspondence to Michael J. O. Wakelam, Andreas Beyer, Sebastian Grönke or Linda Partridge.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

Peer review information 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 Food intake of AL and DRAL mice.

Body weights for chronic and switch AL cohorts. Solid lines indicate when chronic DR and diet switches were applied. The dashed lines indicates linear fit of weight gain for both cohorts. Slope of linear fits are indicated below. n = 45 biologically independent animals per diet. b, Averaged daily food intake per mouse. Each point represents the values from one cohort, encompassing 6 cages each. n = 3 cohorts per diet. c, Average daily food uptake for 3- and 5-weeks post-switch, split by cohorts and individual cage. n = 6 biologically independent cages per diet and cohort. Two-sided Wilcoxon rank-sum test, adjusted for multiple testing. d, Averaged daily food intake per mouse after normalizing against average body weight. Each point represents the values from one cohort, encompassing six cages. n = 3 cohorts per diet. Means ± s.e.m.

Extended Data Fig. 2 Demography of dietary restriction for each of the three breeding cohorts.

a, Pre- and post-switch weight curves for chronic and switch diet mice from the 3 breeding cohorts (± 95% confidence intervals). Solid and dashed lines indicate the time point for diet switch and tissue collection, respectively. Tissues were collected from the F3 cohort only. The number above the graph indicates total cohort size at birth. n = 15 biologically independent animals per diet and cohort. b, Cohort-specific post-switch KM survival curves for chronic and switch diet cohorts. Cox regression (dashed line) was used to avoid making assumptions about the shape of the trajectories. c, Cohort-specific distribution of P values as computed from n = 1,000 Cox regression analyses with random down-sampling of DRAL and DR cohorts to match the size of AL/ALDR cohorts. Analyses were run relative to the pre- and post-switch control. The dashed line indicates significance threshold. Whiskers represent 1st and 5th quartiles, box edges represent 2nd and 4th quartiles, and the centre line represents the 3rd quartile/median. Outliers are marked by points. b,c, Biologically independent animals per cohort at start of switch: F1: n = 58 (AL) n = 69 (DR), n = 72 (DRAL), n = 57 (ALDR); F2: n = 55 (AL) n = 69 (DR), n = 69 (DRAL), n = 56 (ALDR); F3: n = 44 (AL) n = 52 (DR), n = 53 (DRAL), n = 44 (ALDR). Means ± s.e.m.

Extended Data Fig. 3 Transcriptional reprogramming in response to early-onset DR and late-onset AL.

a, Heatmap of unsupervised clustering of expression changes for ALDR switch-resistant genes in BAT (n = 3–5 per group; colour bar represents z score range). b, Boxplot representation of scaled expression levels of differentially up- and downregulated genes under chronic DR as opposed to chronic AL controls in livers of ALDR switch mice at a young age. Whiskers represent 1st and 5th quartiles, box edges represent 2nd and 4th quartiles, and the centre line represents the 3rd quartile/median. c,d, Heatmap of expression changes for ALDR switch-resistant genes in BAT (c) and WAT (d) of young ALDR switch mice (n = 3 per group; colour bar represents z score range). Biologically independent animals used for RNA-seq: Liver: n = 3 (AL, DR, ALDR, DRAL, ALDR 5m, AL 5m); BAT: n = 3 (AL, DR, DRAL, AL 5m, ALDR 5m) n = 5 (ALDR); WAT: n = 3 (AL, DR, ALDR, AL 5m) n = 5 (ALDR, DRAL).

Extended Data Fig. 4 scRNA-seq profiling of the stromal–vascular fraction in young and old WAT.

a, t-SNE visualization of scRNA-seq data (FACS Smart-seq2) from the GAT stromal–vascular fraction, split by age. Preadipocytes as annotated by the Tabula Muris Consortium are coloured by age. b, Scatterplot representation of average expression levels of genes of young and old preadipocytes. Differentially expressed genes (DEGs) are indicated in blue. c, Representative GO enrichment of the top 300 differentially expressed genes between old and young preadipocytes. Lengths of bars represent negative ln-transformed P values, calculated using two-sided Fisher’s exact test. d, t-SNE visualisation of scRNA-seq data coloured by expression of two regulated genes, Ccl7 and Tgfb3. e, Scatterplot of expression differences between old and young preadipocytes (by scRNA-seq) versus expression differences between DR and AL (by bulk RNA-seq). The number of common DEGs in each quadrant is indicated in blue, and the number of ALDR switch-resistant genes is indicated in red. There was a significant inverse association as determined by two-sided Fisher’s exact test for common DEGs (P = 0.0026) and when analysis was limited to ALDR switch-resistant genes (P = 0.003). f, Representative GO enrichment of 91 switch-resistant genes following the inverse association in e. Lengths of bars represent negative ln-transformed P values, calculated using two-sided Fisher’s exact test. The complete list of enriched GO terms can be found in Supplementary Table 1. g, Violin plot representing expression of selected genes across all profiled cell types. Points indicate cell-wise expression levels, and the violin indicates average distribution of expression split by age. h, mRNA expression (RNA-seq) of the same genes WAT. Scd1 expression is shown in Fig. 5. Two-sided Wald test, adjusted for multiple testing. All scRNA-seq data represents cells that were derived and processes from n = 4 biologically independent mice per age group, encompassing a total of n = 1,962 biologically independent cells. Biologically independent animals used for bulk RNA-seq: n=3 (AL, DR, ALDR, AL 5m) n=5 (ALDR, DRAL). Means ± s.e.m., ***P < 0.0001.

Extended Data Fig. 5 Extended functional enrichment analysis of liver.

Representative GO enrichment of ALDR switch-resistant genes in the liver. Lengths of bars represent negative ln-transformed P values, calculated using two-sided Fisher’s exact test. Biologically independent animals used for RNA-seq: n = 3.

Extended Data Fig. 6 Thermogenic marker expression in WAT and BAT.

a, Distribution of gene-wise expression changes in BAT under chronic DR and switch diets relative to chronic AL feeding for genes associated with the GO term ‘Mitochondrion’ (n = 1299 genes). Whiskers represent 1st and 5th quartiles, box edges represent 2nd and 4th quartiles, and the centre line represents the 3rd quartile/median. Two-sided Wilcoxon rank-sum test, adjusted for multiple testing. b, mRNA expression (RNA-seq) of thermogenic marker genes in BAT. c, mRNA expression (RNA-seq) of marker for thermogenesis and mitochondrial biogenesis in BAT. d, Apoe mRNA expression (RNA-seq) in BAT. Two-sided Wald test, adjusted for multiple testing. e, Whole LICOR western blot image of Fig. 4d. f, Western blot analysis of UCP1 in WAT, with α-tubulin as loading control. Tissue extract from one BAT sample (very right lane) was included as positive control for the UCP1 antibody. g,h, mRNA expression (RNA-seq) of uncoupling-independent, thermogenic marker genes in WAT for creatine cycling (g) and Ca2+ cycling (h). Two-sided Wald test, adjusted for multiple testing. i, DRAL switch-resistant genes in WAT. Lengths of bars represent negative ln-transformed P values using two-sided Fisher’s exact test. The complete list of enriched GO terms can be found in Supplementary Table 4. Biologically independent animals used for RNA-seq: BAT: n = 3 (AL, DR, DRAL, AL 5m, ALDR 5m) n = 5 (ALDR); WAT: n = 3 (AL, DR, ALDR, AL 5m) n = 5 (ALDR, DRAL). The Western blot analysis was done once using tissues of n = 4 biologically independent animals per diet that were derived from the same cohort but were not identical to the mice used for RNA-seq. Means ± s.e.m., ***q < 0.0001.

Extended Data Fig. 7 Triglyceride composition in WAT.

a, Distribution of TG species for the switch at young (left) and old (right) age classified according to the number of carbon atoms as proxy for TG-associated chain length. Values represent normalized relative abundances (0–100%) on a logarithmic scale. b, Selected TG groups classified according to associated chain length. Values are identical to the ones in a. One-way ANOVA followed by two-sided post-hoc Tukey test. c, Distribution of TG species for the switch at young (left) and old (right) age, classified according to the number of double bonds in TG-associated chains. Values represent normalized relative abundances (0–100%) on a logarithmic scale. d, Selected TG groups classified according to the number of double bonds. Values are identical to the ones in c. One-way ANOVA followed by two-sided post-hoc Tukey test. Biologically independent animals used for Lipidomics: n = 4 per diet. Means ± s.e.m., ***P < 0.001.

Extended Data Fig. 8 Fluorescence signal analysis of pulse–chase experiment outcome.

a, Cellular uptake profiles of exogenously supplied NBD-PG by explant-cultured adipocytes. Lipids were separated by TLC and analysed by fluorescence scanning. The TLC analysis was done once with lipid extracts from n = 3 biologically independent mice per diet (indicated above), with n = 3 technical replicates each. For each biological replicate, two technical replicates were co-incubated with NBD and one with just the transfection agent. The dashed line represents the paths used to quantify fluorescent signal distribution in Fig. 6d. Dashed boxes represent the areas used to quantify individual bands. Lipid species with low polarity run on top, with TGs being represented by the top band. Fluorescent lipids run slightly lower than non-fluorescent lipids. Standard phospholipids allowed the identification of lipid spots representing TG, DG and PG levels (the asterisks indicate unidentified lipid species). Applied non-fluorescent standard lipids involve: Tetra-oleoyl CL (TO-CL); CL-rich phospholipid-extract from heart; palmitoyl-oleoyl-DG (PODG), di-oleoyl-PG (DOPG); di-oleoyl-PA (DOPA). Fluorescent NBD-labelled lipids involve: PG, PA, PC, PE, PS. b, Relative fluorescent signal in each of the major bands. n = 3 biologically independent, 24-month-old animals per diet; technical replicates were averaged prior analysis. Two-sided t test. Data for the TG band are shown in Fig. 6e. c, Non-fluorescent scans of identical TLC plate after staining with CuSO4. Due to high abundance of TGs (upper band) in adipocytes, no phospholipids can be observed. Standard phospholipids allowed the identification of lipid spots.

Extended Data Fig. 9 Lipid reaction analysis in ALDR and DRAL mice.

a,b, Analysis of lipid pathway activity in WAT of ALDR (a) or DRAL (b) mice relative to AL control. Red and blue arrows show reactions with positive and negative activity, respectively. Coloured circles indicate relative log2-transformed abundance of lipid classes involved. c, mRNA expression (RNA-seq) of key genes mapping to differentially active pathways in Fig. 7a. Two-sided Wald test, adjusted for multiple testing. Biologically independent animals used: RNA-seq: n = 3 (AL, DR, ALDR, AL 5m) n = 5 (ALDR, DRAL); Lipidomics: n = 4 per diet. Means ± s.e.m., ***q < 0.0001.

Supplementary information

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Hahn, O., Drews, L.F., Nguyen, A. et al. A nutritional memory effect counteracts the benefits of dietary restriction in old mice. Nat Metab 1, 1059–1073 (2019). https://doi.org/10.1038/s42255-019-0121-0

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s42255-019-0121-0

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