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:

Daytime-restricted feeding enhances running endurance without prior exercise in mice

Abstract

Physical endurance and energy conservation are essential for survival in the wild. However, it remains unknown whether and how meal timing regulates physical endurance and muscle diurnal rhythms. Here, we show that day/sleep time-restricted feeding (DRF) enhances running endurance by 100% throughout the circadian cycle in both male and female mice, compared to either ad libitum feeding or night/wake time-restricted feeding. Ablation of the circadian clock in the whole body or the muscle abolished the exercise regulatory effect of DRF. Multi-omics analysis revealed that DRF robustly entrains diurnal rhythms of a mitochondrial oxidative metabolism-centric network, compared to night/wake time-restricted feeding. Remarkably, muscle-specific knockdown of the myocyte lipid droplet protein perilipin-5 completely mimics DRF in enhancing endurance, enhancing oxidative bioenergetics and outputting rhythmicity to circulating energy substrates, including acylcarnitine. Together, our work identifies a potent dietary regimen to enhance running endurance without prior exercise, as well as providing a multi-omics atlas of muscle circadian biology regulated by meal timing.

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: Day/sleep time-restricted feeding enhances running endurance in sedentary and trained female mice.
Fig. 2: DRF enhances running endurance through the myocyte clock.
Fig. 3: Day/sleep time-restricted feeding entrains diurnal rhythm of mitochondrial lipid metabolism genes.
Fig. 4: Day/sleep time-restricted feeding entrains diurnal oscillation of energy substrates in the muscle and serum.
Fig. 5: Day/sleep time-restricted feeding entrains diurnal rhythms of mitochondrial functions and dynamics in muscle tissues.
Fig. 6: Day/sleep time-restricted feeding induces diurnal rhythm of muscle Plin5 in a clock-dependent manner.
Fig. 7: Day/sleep time-restricted feeding enhances running endurance via myocyte Plin5.
Fig. 8: Myocyte Plin5 knockdown mimics day/sleep time-restricted feeding in outputting rhythmicity to muscle mitochondrial lipid metabolism and circulating energy substrates.

Similar content being viewed by others

Data availability

RNA-seq data in this paper have been deposited into the Gene Expression Omnibus under accession number GSE194106. The published human transcriptome data are available at GSE28422. The MS proteomics data have been deposited to the ProteomeXchange Consortium via the iProX59 partner repository under dataset identifier PXD039355. Graphs of these multi-omics data can be found at http://www.circametdb.org.cn/. Raw quantification data are deposited to the Science Data Bank (https://www.scidb.cn/en/). Source data are provided with this paper. Any other relevant data are available from the corresponding author upon request.

Code availability

Custom code is deposited to the Science Data Bank (https://doi.org/10.57760/sciencedb.08081).

References

  1. Gabriel, B. M. & Zierath, J. R. Circadian rhythms and exercise—re-setting the clock in metabolic disease. Nat. Rev. Endocrinol. 15, 197–206 (2019).

    Google Scholar 

  2. Baskin, K. K., Winders, B. R. & Olson, E. N. Muscle as a ‘mediator’ of systemic metabolism. Cell Metab. 21, 237–248 (2015).

    CAS  PubMed Central  Google Scholar 

  3. Fan, W. et al. PPARδ promotes running endurance by preserving glucose. Cell Metab. 25, 1186–1193 (2017).

    CAS  PubMed Central  Google Scholar 

  4. Bass, J. & Lazar, M. A. Circadian time signatures of fitness and disease. Science 354, 994–999 (2016).

    CAS  Google Scholar 

  5. Dyar, K. A. et al. Muscle insulin sensitivity and glucose metabolism are controlled by the intrinsic muscle clock. Mol. Metab. 3, 29–41 (2014).

    CAS  Google Scholar 

  6. Schroder, E. A. et al. Intrinsic muscle clock is necessary for musculoskeletal health. J. Physiol. 593, 5387–5404 (2015).

    CAS  PubMed Central  Google Scholar 

  7. Woldt, E. et al. Rev-erb-α modulates skeletal muscle oxidative capacity by regulating mitochondrial biogenesis and autophagy. Nat. Med. 19, 1039–1046 (2013).

    CAS  PubMed Central  Google Scholar 

  8. Hong, S. et al. Dissociation of muscle insulin sensitivity from exercise endurance in mice by HDAC3 depletion. Nat. Med. 23, 223–234 (2017).

    CAS  Google Scholar 

  9. Takahashi, J. S. Transcriptional architecture of the mammalian circadian clock. Nat. Rev. Genet. 18, 164–179 (2017).

    CAS  Google Scholar 

  10. Jordan, S. D. et al. CRY1/2 selectively repress PPARδ and limit exercise capacity. Cell Metab. 26, 243–255 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  11. Ezagouri, S. et al. Physiological and molecular dissection of daily variance in exercise capacity. Cell Metab. 30, 78–91 (2019).

    CAS  PubMed  Google Scholar 

  12. Adamovich, Y. et al. Clock proteins and training modify exercise capacity in a daytime-dependent manner. Proc. Natl Acad. Sci. USA 118, e2101115118 (2021).

    CAS  PubMed  PubMed Central  Google Scholar 

  13. Sato, S. et al. Time of exercise specifies the impact on muscle metabolic pathways and systemic energy homeostasis. Cell Metab. 30, 92–110 (2019).

    CAS  PubMed  Google Scholar 

  14. Sato, S. et al. Atlas of exercise metabolism reveals time-dependent signatures of metabolic homeostasis. Cell Metab. 34, 329–345 (2022).

    CAS  PubMed  Google Scholar 

  15. Reinke, H. & Asher, G. Cross-talk between metabolism and circadian clocks. Nat. Rev. Mol. Cell Biol. 20, 227–241 (2019).

    CAS  Google Scholar 

  16. Guan, D. & Lazar, M. A. Interconnections between circadian clocks and metabolism. J. Clin. Invest. 131, e148278 (2021).

    CAS  PubMed Central  Google Scholar 

  17. Allada, R. & Bass, J. Circadian mechanisms in medicine. N. Engl. J. Med. 384, 550–561 (2021).

    CAS  PubMed Central  Google Scholar 

  18. Panda, S. Circadian physiology of metabolism. Science 354, 1008–1015 (2016).

    CAS  PubMed Central  Google Scholar 

  19. Chaix, A., Manoogian, E. N. C., Melkani, G. C. & Panda, S. Time-restricted eating to prevent and manage chronic metabolic diseases. Annu. Rev. Nutr. 39, 291–315 (2019).

    CAS  PubMed Central  Google Scholar 

  20. Li, M.-D. Clock-modulated checkpoints in time-restricted eating. Trends Mol. Med. 28, 25–35 (2022).

    PubMed  Google Scholar 

  21. Manoogian, E. N., Chow, L. S., Taub, P. R., Laferrère, B. & Panda, S. Time-restricted eating for the prevention and management of metabolic diseases. Endocr. Rev. 49, 405–436 (2021).

    Google Scholar 

  22. Petersen, M. C. et al. Complex physiology and clinical implications of time-restricted eating. Physiol. Rev. 102, 1991–2034 (2022).

    CAS  PubMed  Google Scholar 

  23. Chaix, A., Deota, S., Bhardwaj, R., Lin, T. & Panda, S. Sex- and age-dependent outcomes of 9-hour time-restricted feeding of a Western high-fat high-sucrose diet in C57BL/6J mice. Cell Rep. 36, 109543 (2021).

    CAS  PubMed  PubMed Central  Google Scholar 

  24. Damiola, F. et al. Restricted feeding uncouples circadian oscillators in peripheral tissues from the central pacemaker in the suprachiasmatic nucleus. Genes Dev. 14, 2950–2961 (2000).

    CAS  PubMed  PubMed Central  Google Scholar 

  25. Stokkan, K.-A., Yamazaki, S., Tei, H., Sakaki, Y. & Menaker, M. Entrainment of the circadian clock in the liver by feeding. Science 291, 490–493 (2001).

    CAS  PubMed  Google Scholar 

  26. Xin, H. et al. A multi-tissue multi-omics analysis reveals distinct kinetics in entrainment of diurnal transcriptomes by inverted feeding. iScience 24, 102335 (2021).

    CAS  PubMed  PubMed Central  Google Scholar 

  27. Manella, G. et al. The liver-clock coordinates rhythmicity of peripheral tissues in response to feeding. Nat. Metab. 3, 829–842 (2021).

    CAS  PubMed  PubMed Central  Google Scholar 

  28. Zhang, Z., Shui, G. & Li, M. -D. Time to eat reveals the hierarchy of peripheral clocks. Trends Cell Biol. 31, 869–872 (2021).

    PubMed  Google Scholar 

  29. Al-Nawaiseh, A. M., Bataineh, M. F., Kilani, H. A., Bellar, D. M. & Judge, L. W. Time-restricted feeding and aerobic performance in elite runners: Ramadan fasting as a model. Front. Nutr. 8, 718936 (2021).

    PubMed  PubMed Central  Google Scholar 

  30. Marcaletti, S., Thomas, C. & Feige, J. N. Exercise performance tests in mice. Curr. Protoc. Mouse Biol. 1, 141–154 (2011).

  31. Schiaffino, S. & Reggiani, C. Fiber types in mammalian skeletal muscles. Physiol. Rev. 91, 1447–1531 (2011).

    CAS  Google Scholar 

  32. Tabebordbar, M. et al. Directed evolution of a family of AAV capsid variants enabling potent muscle-directed gene delivery across species. Cell 184, 4919–4938 (2021).

    CAS  PubMed Central  Google Scholar 

  33. Opperhuizen, A. L. et al. Feeding during the resting phase causes profound changes in physiology and desynchronization between liver and muscle rhythms of rats. Eur. J. Neurosci. 44, 2795–2806 (2016).

    Google Scholar 

  34. Mukherji, A. et al. Shifting eating to the circadian rest phase misaligns the peripheral clocks with the master SCN clock and leads to a metabolic syndrome. Proc. Natl Acad. Sci. USA 112, E6691–E6698 (2015).

    CAS  PubMed Central  Google Scholar 

  35. Schmitt, K. et al. Circadian control of DRP1 activity regulates mitochondrial dynamics and bioenergetics. Cell Metab. 27, 657–666 (2018).

    CAS  Google Scholar 

  36. Ohta, H., Yamazaki, S. & McMahon, D. G. Constant light desynchronizes mammalian clock neurons. Nat. Neurosci. 8, 267–269 (2005).

    CAS  Google Scholar 

  37. Mason, R. R. & Watt, M. J. Unraveling the roles of PLIN5: linking cell biology to physiology. Trends Endocrinol. Metab. 26, 144–152 (2015).

    CAS  Google Scholar 

  38. Wolins, N. E. et al. OXPAT/PAT-1 is a PPAR-induced lipid droplet protein that promotes fatty acid utilization. Diabetes 55, 3418–3428 (2006).

    CAS  Google Scholar 

  39. Simcox, J. et al. Global analysis of plasma lipids identifies liver-derived acylcarnitines as a fuel source for brown fat thermogenesis. Cell Metab. 26, 509–522 (2017).

    CAS  PubMed Central  Google Scholar 

  40. Levine, D. C. et al. NADH inhibition of SIRT1 links energy state to transcription during time-restricted feeding. Nat. Metab. 3, 1621–1632 (2021).

    CAS  PubMed Central  Google Scholar 

  41. Yasumoto, Y. et al. Short-term feeding at the wrong time is sufficient to desynchronize peripheral clocks and induce obesity with hyperphagia, physical inactivity and metabolic disorders in mice. Metabolism 65, 714–727 (2016).

    CAS  Google Scholar 

  42. Arble, D. M., Bass, J., Laposky, A. D., Vitaterna, M. H. & Turek, F. W. Circadian timing of food intake contributes to weight gain. Obesity 17, 2100–2102 (2009).

    Google Scholar 

  43. Acosta-Rodríguez, 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).

    PubMed Central  Google Scholar 

  44. Acosta-Rodríguez, V. et al. Circadian alignment of early onset caloric restriction promotes longevity in male C57BL/6J mice. Science 376, 1192–1202 (2022).

    PubMed Central  Google Scholar 

  45. Murphy, R. M., Watt, M. J. & Febbraio, M. A. Metabolic communication during exercise. Nat. Metab. 2, 805–816 (2020).

    Google Scholar 

  46. Hargreaves, M. & Spriet, L. L. Skeletal muscle energy metabolism during exercise. Nat. Metab. 2, 817–828 (2020).

    CAS  Google Scholar 

  47. Meier, F. et al. diaPASEF: parallel accumulation–serial fragmentation combined with data-independent acquisition. Nat. Methods 17, 1229–1236 (2020).

    CAS  PubMed  Google Scholar 

  48. Song, J. W. et al. Omics-driven systems interrogation of metabolic dysregulation in COVID-19 pathogenesis. Cell Metab. 32, 188–202 (2020).

    CAS  PubMed Central  Google Scholar 

  49. Chen, Y. et al. Proteomic analysis identifies prolonged disturbances in pathways related to cholesterol metabolism and myocardium function in the COVID-19 recovery stage. J. Proteome Res. 20, 3463–3474 (2021).

    CAS  Google Scholar 

  50. Lundell, L. S. et al. Time-restricted feeding alters lipid and amino acid metabolite rhythmicity without perturbing clock gene expression. Nat. Commun. 11, 4643 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  51. Narkar, V. A. et al. AMPK and PPARδ agonists are exercise mimetics. Cell 134, 405–415 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  52. Xin, H. et al. Protocol for setup and circadian analysis of inverted feeding in mice. STAR Protoc. 2, 100701 (2021).

    PubMed  PubMed Central  Google Scholar 

  53. Zhang, Z. et al. Impaired function of the suprachiasmatic nucleus rescues the loss of body temperature homeostasis caused by time-restricted feeding. Sci. Bull. 65, 1268–1280 (2020).

    Google Scholar 

  54. Storch, K. F. et al. Intrinsic circadian clock of the mammalian retina: importance for retinal processing of visual information. Cell 130, 730–741 (2007).

    CAS  PubMed  PubMed Central  Google Scholar 

  55. Meng, Z. X. et al. Baf60c drives glycolytic metabolism in the muscle and improves systemic glucose homeostasis through Deptor-mediated Akt activation. Nat. Med. 19, 640–645 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  56. Tian, H. et al. Precise metabolomics reveals a diversity of aging‐associated metabolic features. Small Methods 6, e2200130 (2022).

    PubMed  Google Scholar 

  57. Lam, S. M. et al. A multi-omics investigation of the composition and function of extracellular vesicles along the temporal trajectory of COVID-19. Nat. Metab. 3, 909–922 (2021).

    CAS  PubMed  Google Scholar 

  58. Xin, H. et al. Circadian signatures of adipose tissue in diet-induced obesity. Front. Physiol. 13, 953237 (2022).

    PubMed  PubMed Central  Google Scholar 

  59. Chen, T. et al. iProX in 2021: connecting proteomics data sharing with big data. Nucleic Acids Res. 50, D1522–D1527 (2022).

    CAS  PubMed  Google Scholar 

Download references

Acknowledgements

Per1/Per2-DKO mice were gifted from E. E. Zhang, and bred by J. R. He. Bmal1fl/fl mice were gifted from W. He. We thank D. Zhu, Z. Gan, Z. Meng and L. Xie for suggestions. This study was supported by National Natural Science Foundation of China Grants (32271208 to M.-D.L., 92057109 to M.-D.L., 81900776 to M.-D.L. and 32150005 to C.C.L.W.), Chongqing Science Fund for Distinguished Young Scholars (CSTB2022NSCQ-JQX0009 to M.-D.L.), National Key Research and Development Programme of China (2022YFA0806001 to C.C.L.W.), CAMS Innovation Fund for Medical Sciences (2022-I2M-1-004 to C.C.L.W.).

Author information

Authors and Affiliations

Authors

Contributions

Conceptualization: M.-D.L.; Methodology: H.X., R.H., M.Z., J.C., G.S., C.C.L.W. and M.-D.L.; Investigation: M.-D.L., H.X., R.H., M.Z., J.C., T.Z., S.J., X.L., H.T., S.M.L., X.B., J.Z. and L.L.; Funding acquisition: M.-D.L. and C.C.L.W.; Project administration: M.-D.L.; Resources: M.-D.L., C.C.L.W., G.S., S.T., F.D. and Z.Z.; Supervision: M.-D.L., C.C.L.W. and G.S.; Writing—original draft: M.-D.L., H.X., R.H., M.Z. and J.C.; Writing—review and editing: M.-D.L., C.C.L.W. and G.S.

Corresponding authors

Correspondence to Zhihui Zhang, Catherine C. L. Wong or Min-Dian Li.

Ethics declarations

Competing interests

The authors declare no competing interests.

Peer review

Peer review information

Nature Metabolism thanks Chih-Hao Lee, Kristin Stanford and the other, anonymous, reviewer for their contribution to the peer review of this work. Primary Handling Editor: Ashley Castellanos-Jankiewicz, in collaboration with the Nature Metabolism team.

Additional information

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

Extended data

Extended Data Fig. 1 Exercise phenotyping in time-restricted fed mice.

a, Running power in 3-week TRF female mice, as measured by time to exhaustion, distance, cumulative stimulations, and maximum speed (n = 14 NRF or 16 DRF mice). b, Running endurance in the early night (ZT14, n = 15 NRF or 16 DRF mice), as measured by time to exhaustion and distance. c, Running endurance in 3-week DRF mice (Male: n = 15 mice; Female: n = 16 mice), as measured by time to exhaustion, distance, and blood glucose. d, Running power in 3-week DRF mice (Male: n = 15 AL or 16 DRF mice; Female: n = 16 mice), as measured by time to exhaustion, distance, and maximum speed. e, Immunofluorescence (IF) of slow- (type 1)/fast- (type 2B, 2 A, 2X) myofibers and histochemical staining of bioenergetic enzyme activity in soleus muscle after 3 weeks of TRF (n = 8 mice, one image per mouse). MyHC, myosin heavy chain. SDH, succinate dehydrogenase. GPDH, alpha-glycerophosphate dehydrogenase. Arrows indicate positively stained fibers. Scale bars, 200 µm. f, Total locomotor activity in 3-week TRF female mice (n = 9 AL or 8 DRF/NRF mice). g, Respiratory exchange ratio (RER) in 1-week TRF female mice (n = 4 mice). h, The scheme for testing endurance in trained female mice (n = 12 mice, slope: 15o). i, Myofiber typing in GA muscle from ad libitum (AL)-fed exercised (Exe) mice or TRF-fed sedentary (Sed) mice, as measured by IF and bioenergetic enzyme activity (n = 11 Sed-NRF, 12 Sed-DRF (IF/GPDH), 11 Sed-DRF (SDH) or 16 Exe-AL mice). TRF, time-restricted feeding; NRF, night/wake time-restricted feeding; DRF, day/sleep time-restricted feeding (food available from ZT0 to ZT12); AL, ad libitum feeding. Data were represented as the mean ± s.e.m. Statistical tests: ANOVA, post-hoc Sidak/Tukey test, Kruskal-Wallis test, or two-sided unpaired Student’s/Wilcoxon test; *P < 0.05, **P < 0.01, ***P < 0.001, ****P < 0.0001.

Source data

Extended Data Fig. 2 Striated muscle-specific knockdown of Per2 gene by systemic administration of MyoAAV does not alter running endurance under DRF in female mice.

a, Daytime locomotor activity in Per1−/− Per2−/− (Per1/2 DKO) male mice before and after 3 weeks of DRF beginning at the age of 34 weeks (3 weeks AL feeding > 3 weeks DRF) (n = 8 mice), as measured by percentage of daytime activity and 24-h beam breaks. b, Daytime locomotor activity in Per2/ female mice before and after 3 weeks of DRF starting at the age of 7 weeks (n = 10 mice). c, RT-qPCR analysis of muscle and liver tissues in MyoAAV-transduced (MKD) female mice under DRF (N = 5 Ctrl-MKD mice, N = 6 Per2-MKD mice). d, Representative immunofluorescence image of PER2 protein in TA muscle tissue. This experiment was repeated independently twice with similar results (n = 200 cells examined over two independent experiments). GA muscle from Per1/2 DKO mice, as well as mock primary antibody-treated Per2-MKD, serves as the negative control. e, Daytime locomotor activity in MKD female mice before and after 3 weeks of DRF starting at the age of 8 weeks (n = 5 Ctrl-MKD or 6 Per2-MKD mice). f, Running endurance of MKD female mice in the early day (ZT2, n = 16 mice), as measured by time to exhaustion, distance and cumulative stimulations. Mice received MyoAAV by tain vein injection at the age of 8 weeks, tested 3 weeks later (AL), and fed DRF for 3 weeks before the next running test. g, Tissue mass in Bmal1 or Per2 MKO mice under DRF (n = 8 control or 7 MKO mice). Per2-MKD, striated muscle-specific knockdown of Per2; Ctrl-MKD, control group. Data are presented as the mean ± s.e.m. or minima, maxima, median, 25th and 75th percentiles. Statistical tests: two-sided paired Student’s t-test, ANOVA, post-hoc Sidak’s test, or two-sided Wilcoxon test, *P < 0.05, **P < 0.01, ***P < 0.001.

Source data

Extended Data Fig. 3 General features and circadian clock oscillation of the multi-omics dataset.

a, Exercise performance in 1-week TRF mice as measured by time to exhaustion (n = 16 mice). b, Workflow of the multi-omics study. c-d, Diurnal expression of the muscle clock genes in TRF mice (c, female: n = 4 mice; d, male: n = 4 mice). e, Phase distribution of oscillating muscle genes and phase-shift between dual-oscillating genes in TRF mice. f-h, Diurnal expression of dual-oscillating (f), DRF-only (g), NRF-only (g), and mitochondrial (h) rhythmic genes in skeletal muscle (n = 4 mice per time-point). Data were represented as the mean ± s.e.m. or minima, maxima, median, 25th and 75th percentiles. Statistical test: ANOVA, two-sided unpaired Student’s t-test with Bonferroni correction, MetaCycle: P-values from JTK and LS methods were adjusted by Fisher’s method.

Source data

Extended Data Fig. 4 Global profiling of muscle metabolites.

Heatmap showing cycling metabolites in GA muscle during TRF. Inset shows phase distribution of muscle cycling metabolites.

Source data

Extended Data Fig. 5 Global profiling of muscle lipids and serum metabolites.

a, Heatmap showing cycling lipid species in GA muscle. Inset shows phase distribution of muscle cycling lipids. Abbreviations see Data S4. b, Diurnal expression of cycling serum metabolites (n = 7 mice for 2 time points). c, Principal component analysis of the four groups. D00, DRF ZT0; D12, DRF ZT12; N00, NRF ZT0; N12, NRF ZT12.

Source data

Extended Data Fig. 6 Global profiling of the muscle proteome.

a, 48-h profile of cycling muscle proteins from 3-week DRF mice. Samples were collected every 2 h for 48 h containing 25 time points (n = 2 mice per time point). b, Representative cycling muscle proteins (n = 2 mice per time point). Data were represented as mean and points. c, Representative immunoblots of pSer637-DRP1 in anterior hypothalamus (aHyp) of the brain in ad libitum fed mice (n = 2 mice, repeated three times with similar results). ALP, alkaline phosphatase treatment. d, Immunoblot analysis of key regulators involved in mitochondrial dynamics by DRF in GA muscle. Data were represented as the mean ± s.e.m. MetaCycle: P-values from JTK and LS methods were adjusted by Fisher’s method.

Source data

Extended Data Fig. 7 Daily rhythms of acylcarnitine and amino acids persist in the muscle of 3-week DRF female mice.

a, Heatmap showing cycling metabolites in GA muscle from 3-week DRF female mice. b, Class of cycling metabolites in muscle. c, Heatmap showing cycling acylcarnitines, amino acids and fatty acids. d, Daily abundance levels of selected cycling muscle metabolites representing acylcarnitines and amino acids. Stearoylcarnitine, 18:0-carnitine; L-Palmitoylcarnitine, 16:0-carnitine. Samples were collected every 2 h for 48 h containing 25 time points (n = 2 mice per time point). Data were shown as the mean ± s.e.m. Statistical tests: RAIN and MetaCycle.

Source data

Extended Data Fig. 8 Diurnal expression of rhythmic transcripts in DRF muscle.

a, Diurnal expression of Pdp2 and Hmgcs2 in GA muscle from TRF female mice, as measured by RNA sequencing (n = 4 mice). b, Diurnal gene expression in GA muscle from TRF female mice (n = 4 mice). c, Diurnal gene expression in GA muscle from TRF male mice (n = 4 mice). d, Diurnal gene expression in GA muscle from female mice subjected to DRF and constant lightness (n = 4 mice). e, Diurnal gene expression in GA muscle from Per1/Per2/ mutant mice subjected to constant darkness and DRF (N = 14 mice, n = 2 mice per time point for 7 time points). CT, circadian time, or hours under constant darkness; τ, period length. f, Gene expression in vastus lateralis muscle biopsies before (T1, T3) and after (T2, T4) resistance exercise. Y (young, n = 8 males/8 females), O (old, n = 6 males/6 females); T1/T2 is from the sedentary state, T3/T4 is from the trained state (Source: GSE28422). Data are presented as mean ± s.e.m. or minima, maxima, median, 25th and 75th percentiles. Statistical tests: multiple t-tests with Bonferroni correction, MetaCycle (rhythmicity), Circacompare (phase shift), or ANOVA, two-sided Wilcoxon test; ns, not significant, AR, arrhythmia.

Source data

Extended Data Fig. 9 The role of myocyte Plin5 in the regulation of exercise capacity under DRF or ad libitum.

a, Treadmill running scheme for studying the role of myocyte Plin5 in exercise capacity. KD, TA muscle-specific knockdown of Plin5 gene; WT, control group dosed with negative control miRNA. b, Representative immunoblots of PLIN5 protein in different muscle depots from the ipsilateral and contralateral sides of the AAV injection site (one technical replicate of two biological replicates). c, Running power in female mice transduced with AAV-pre-miR-Plin5 (KD) or AAV-pre-miR-Ctrl (WT) and fed ad libitum (AL) or DRF for 3 weeks (n = 16 mice), as measured by time to exhaustion, distance, maximum speed, and cumulative stimulations. d, Cumulative stimulations and body weight in the endurance test for KD and WT mice fed AL or DRF for 3 weeks (n = 16 mice). Data were represented as the mean ± s.e.m. 2-way ANOVA or two-sided unpaired Student’s t test, *P < 0.05.

Source data

Extended Data Fig. 10 Muscle phenotyping in Plin5 KD mice.

a, Running endurance in DRF Plin5 KD mice resuming AL feeding for 1 to 3 weeks (n = 16 mice), as measured by time to exhaustion, distance, and cumulative number of stimulations. b, Running endurance and histochemical staining of bioenergetic enzyme activity in GA muscle from diet reversed (DRF > AL, return to AL for 3 weeks after 3 weeks of DRF) and DRF mice (n = 16 mice, one image per mouse). Scale bars, 100 μm. c, Representative immunoblots for PLIN5 in GA muscle and heart (HT) of MKD mice (n = 5 mice). d, Distribution of phase-shift among dual-oscillating muscle genes in KD and WT mice. KD, TA-specific Plin5 knockdown. e, Diurnal expression of circadian clock genes in the muscle (n = 2 mice for 8 time points). f, Pathway analysis of differentially expressed muscle genes from ZT21 to ZT3 between KD and WT mice (n = 6 mice). Data were represented as the mean ± s.e.m. 2-way ANOVA or two-sided unpaired Student’s t test, *P < 0.05, ****P < 0.0001.

Source data

Supplementary information

Supplementary Information

Supplementary Methods and Supplementary Table 4

Reporting Summary

Supplementary Tables 1–3

Table 1: Rhythmicity results of genes, proteins, metabolites and lipids. Table 2: PSEA results of oscillating genes in skeletal muscle. Table 3: Rhythmicity results of metabolites in the serum.

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 Fig. 6

Statistical source data.

Source Data Fig. 7

Statistical source data.

Source Data Fig. 8

Statistical source data.

Source Data Extended Data Fig. 1

Statistical source data.

Source Data Extended Data Fig. 2

Statistical source data.

Source Data Extended Data Fig. 3

Statistical source data.

Source Data Extended Data Fig. 4

Statistical source data.

Source Data Extended Data Fig. 5

Statistical source data.

Source Data Extended Data Fig. 6

Statistical source data.

Source Data Extended Data Fig. 7

Statistical source data.

Source Data Extended Data Fig. 8

Unprocessed western blots and statistical source data.

Source Data Extended Data Fig. 9

Unprocessed western blots and statistical source data.

Source Data Extended Data Fig. 10

Statistical source data.

Rights and permissions

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

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Xin, H., Huang, R., Zhou, M. et al. Daytime-restricted feeding enhances running endurance without prior exercise in mice. Nat Metab 5, 1236–1251 (2023). https://doi.org/10.1038/s42255-023-00826-7

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s42255-023-00826-7

This article is cited by

Search

Quick links

Nature Briefing: Translational Research

Sign up for the Nature Briefing: Translational Research newsletter — top stories in biotechnology, drug discovery and pharma.

Get what matters in translational research, free to your inbox weekly. Sign up for Nature Briefing: Translational Research