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:

Pre-operative exercise therapy triggers anti-inflammatory trained immunity of Kupffer cells through metabolic reprogramming

Abstract

Pre-operative exercise therapy improves outcomes for many patients who undergo surgery. Despite the well-known effects on tolerance to systemic perturbation, the mechanisms by which pre-operative exercise protects the organ that is operated on from inflammatory injury are unclear. Here, we show that four-week aerobic pre-operative exercise significantly attenuates liver injury and inflammation from ischaemia and reperfusion in mice. Remarkably, these beneficial effects last for seven more days after completing pre-operative exercising. We find that exercise specifically drives Kupffer cells toward an anti-inflammatory phenotype with trained immunity via metabolic reprogramming. Mechanistically, exercise-induced HMGB1 release enhances itaconate metabolism in the tricarboxylic acid cycle that impacts Kupffer cells in an NRF2-dependent manner. Therefore, these metabolites and cellular/molecular targets can be investigated as potential exercise-mimicking pharmaceutical candidates to protect against liver injury during surgery.

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: Exercise attenuates hepatic I/R injury by altering the immune microenvironment.
Fig. 2: Exercise shifts KCs towards an anti-inflammatory transcriptomic profile.
Fig. 3: Exercise drives KCs to be an anti-inflammatory phenotype.
Fig. 4: Exercise promotes metabolic reprogramming in KCs.
Fig. 5: Exercise-induced HMGB1 activates itaconate/IRG1/NRF2 signalling in KCs.
Fig. 6: Exercise promotes an anti-inflammatory trained immunity in KCs.

Similar content being viewed by others

Data availability

We have deposited the scRNA-seq data associated with this study with the Gene Expression Omnibus under accession no. GSE173429. Source data are provided with this paper. All other data that support the findings of this study are available from the corresponding author upon request.

Code availability

The code for the single-cell RNA sequencing data is provided with the paper in the supplementary dataset.

References

  1. Veenhof, A. A. et al. Surgical stress response and postoperative immune function after laparoscopy or open surgery with fast track or standard perioperative care: a randomized trial. Ann. Surg. 255, 216–221 (2012).

    Article  CAS  PubMed  Google Scholar 

  2. Onuma, A. E., Zhang, H., Gil, L., Huang, H. & Tsung, A. Surgical stress promotes tumor progression: a focus on the impact of the immune response. J. Clin. Med. 9, 4096 (2020).

    Article  CAS  PubMed Central  Google Scholar 

  3. Krall, J. A. et al. The systemic response to surgery triggers the outgrowth of distant immune-controlled tumors in mouse models of dormancy. Sci. Transl. Med. 10, eaan3464 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  4. Koelwyn, G. J., Zhuang, X., Tammela, T., Schietinger, A. & Jones, L. W. Exercise and immunometabolic regulation in cancer. Nat. Metab. 2, 849–857 (2020).

    Article  PubMed  Google Scholar 

  5. Dobson, G. P. Addressing the global burden of trauma in major surgery. Front. Surg. 2, 43 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  6. Tashiro, H., Kuroda, S., Mikuriya, Y. & Ohdan, H. Ischemia–reperfusion injury in patients with fatty liver and the clinical impact of steatotic liver on hepatic surgery. Surg. Today 44, 1611–1625 (2014).

    Article  CAS  PubMed  Google Scholar 

  7. Weigand, K. et al. Ischemia/reperfusion injury in liver surgery and transplantation: pathophysiology. HPB Surg. 2012, 176723 (2012).

    Article  PubMed  PubMed Central  Google Scholar 

  8. van Golen, R. F., Reiniers, M. J., Olthof, P. B., van Gulik, T. M. & Heger, M. Sterile inflammation in hepatic ischemia/reperfusion injury: present concepts and potential therapeutics. J. Gastroenterol. Hepatol. 28, 394–400 (2013).

    Article  CAS  PubMed  Google Scholar 

  9. Zhai, Y., Petrowsky, H., Hong, J. C., Busuttil, R. W. & Kupiec-Weglinski, J. W. Ischaemia–reperfusion injury in liver transplantation—from bench to bedside. Nat. Rev. Gastroenterol. Hepatol. 10, 79–89 (2013).

    Article  CAS  PubMed  Google Scholar 

  10. Hilmi, I. A. et al. N-acetylcysteine does not prevent hepatorenal ischaemia–reperfusion injury in patients undergoing orthotopic liver transplantation. Nephrol. Dial. Transplant. 25, 2328–2333 (2010).

    Article  CAS  PubMed  Google Scholar 

  11. Aliakbarian, M. et al. Effects of N-acetylcysteine addition to University of Wisconsin solution on the rate of ischemia–reperfusion injury in adult orthotopic liver transplant. Exp. Clin. Transplant. 15, 432–436 (2017).

    PubMed  Google Scholar 

  12. Vermillion, S. A. et al. Preoperative exercise therapy for gastrointestinal cancer patients: a systematic review. Syst. Rev. 7, 103 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  13. Nakajima, H. et al. Clinical benefit of preoperative exercise and nutritional therapy for patients undergoing hepato-pancreato-biliary surgeries for malignancy. Ann. Surg. Oncol. 26, 264–272 (2019).

    Article  PubMed  Google Scholar 

  14. Pouwels, S. et al. Preoperative exercise therapy for elective major abdominal surgery: a systematic review. Int. J. Surg. 12, 134–140 (2014).

    Article  PubMed  Google Scholar 

  15. Koelwyn, G. J., Khouri, M., Mackey, J. R., Douglas, P. S. & Jones, L. W. Running on empty: cardiovascular reserve capacity and late effects of therapy in cancer survivorship. J. Clin. Oncol. 30, 4458–4461 (2012).

    Article  PubMed  PubMed Central  Google Scholar 

  16. Scott, J. M., Nilsen, T. S., Gupta, D. & Jones, L. W. Exercise therapy and cardiovascular toxicity in cancer. Circulation 137, 1176–1191 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  17. Scott, J. M. et al. Efficacy of exercise therapy on cardiorespiratory fitness in patients with cancer: a systematic review and meta-analysis. J. Clin. Oncol. 36, 2297–2305 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Koelwyn, G. J., Quail, D. F., Zhang, X., White, R. M. & Jones, L. W. Exercise-dependent regulation of the tumour microenvironment. Nat. Rev. Cancer 17, 620–632 (2017).

    Article  PubMed  Google Scholar 

  19. Mela, V. et al. Exercise-induced re-programming of age-related metabolic changes in microglia is accompanied by a reduction in senescent cells. Brain Behav. Immun. 87, 413–428 (2020).

    Article  CAS  PubMed  Google Scholar 

  20. O’Neill, L. A. J. & Artyomov, M. N. Itaconate: the poster child of metabolic reprogramming in macrophage function. Nat. Rev. Immunol. 19, 273–281 (2019).

    Article  PubMed  Google Scholar 

  21. Netea, M. G. et al. Defining trained immunity and its role in health and disease. Nat. Rev. Immunol. 20, 375–388 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Saeed, S. et al. Epigenetic programming of monocyte-to-macrophage differentiation and trained innate immunity. Science 345, 1251086 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  23. Bekkering, S. et al. Metabolic induction of trained immunity through the mevalonate pathway. Cell 172, 135–146.e9 (2018).

    Article  CAS  PubMed  Google Scholar 

  24. Domínguez-Andrés, J. et al. The itaconate pathway is a central regulatory node linking innate immune tolerance and trained immunity. Cell Metab. 29, 211–220.e5 (2019).

    Article  PubMed  Google Scholar 

  25. Chen, B. P. et al. Four-week prehabilitation program is sufficient to modify exercise behaviors and improve preoperative functional walking capacity in patients with colorectal cancer. Support Care Cancer 25, 33–40 (2017).

    Article  PubMed  Google Scholar 

  26. Cabilan, C. J., Hines, S. & Munday, J. The effectiveness of prehabilitation or preoperative exercise for surgical patients: a systematic review. JBI Database System Rev. Implement. Rep. 13, 146–187 (2015).

    Article  CAS  PubMed  Google Scholar 

  27. Zhai, Y., Busuttil, R. W. & Kupiec-Weglinski, J. W. Liver ischemia and reperfusion injury: new insights into mechanisms of innate-adaptive immune-mediated tissue inflammation. Am. J. Transplant. 11, 1563–1569 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Behbehani, G. K. Immunophenotyping by mass cytometry. Methods Mol. Biol. 2032, 31–51 (2019).

    Article  CAS  PubMed  Google Scholar 

  29. Zhang, Q. et al. Landscape and dynamics of single immune cells in hepatocellular carcinoma. Cell 179, 829–845.e20 (2019).

    Article  CAS  PubMed  Google Scholar 

  30. Halpern, K. B. et al. Single-cell spatial reconstruction reveals global division of labour in the mammalian liver. Nature 542, 352–356 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. MacParland, S. A. et al. Single cell RNA sequencing of human liver reveals distinct intrahepatic macrophage populations. Nat. Commun. 9, 4383 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  32. Bradshaw, G. et al. Facilitated replacement of Kupffer cells expressing a paraoxonase-1 transgene is essential for ameliorating atherosclerosis in mice. Proc. Natl Acad. Sci. USA 102, 11029–11034 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Tsung, A. et al. Hepatic ischemia/reperfusion injury involves functional TLR4 signaling in nonparenchymal cells. J. Immunol. 175, 7661–7668 (2005).

    Article  CAS  PubMed  Google Scholar 

  34. Sato, A. et al. Involvement of the TNF and FasL produced by CD11b Kupffer cells/macrophages in CCl4-induced acute hepatic injury. PLoS ONE 9, e92515 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  35. Kozicky, L. K. & Sly, L. M. Depletion and reconstitution of macrophages in mice. Methods Mol. Biol. 1960, 101–112 (2019).

    Article  CAS  PubMed  Google Scholar 

  36. Stienstra, R., Netea-Maier, R. T., Riksen, N. P., Joosten, L. A. B. & Netea, M. G. Specific and complex reprogramming of cellular metabolism in myeloid cells during innate immune responses. Cell Metab. 26, 142–156 (2017).

    Article  CAS  PubMed  Google Scholar 

  37. Mulder, W. J. M., Ochando, J., Joosten, L. A. B., Fayad, Z. A. & Netea, M. G. Therapeutic targeting of trained immunity. Nat. Rev. Drug Discov. 18, 553–566 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Huang, H. et al. Histones activate the NLRP3 inflammasome in Kupffer cells during sterile inflammatory liver injury. J. Immunol. 191, 2665–2679 (2013).

    Article  CAS  PubMed  Google Scholar 

  39. Li, X. et al. O-GlcNAc transferase suppresses inflammation and necroptosis by targeting receptor-interacting serine/threonine-protein kinase 3. Immunity 50, 576–590.e6 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Lampropoulou, V. et al. Itaconate links inhibition of succinate dehydrogenase with macrophage metabolic remodeling and regulation of inflammation. Cell Metab. 24, 158–166 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Yi, Z. et al. Immune-responsive gene 1/itaconate activates nuclear factor erythroid 2-related factor 2 in hepatocytes to protect against liver ischemia–reperfusion injury. Hepatology 72, 1394–1411 (2020).

    Article  CAS  PubMed  Google Scholar 

  42. Swain, A. et al. Comparative evaluation of itaconate and its derivatives reveals divergent inflammasome and type I interferon regulation in macrophages. Nat. Metab. 2, 594–602 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Mills, E. L. et al. Itaconate is an anti-inflammatory metabolite that activates Nrf2 via alkylation of KEAP1. Nature 556, 113–117 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Deng, M. et al. The endotoxin delivery protein HMGB1 mediates caspase-11-dependent lethality in sepsis. Immunity 49, 740–753.e7 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Goh, J. & Behringer, M. Exercise alarms the immune system: a HMGB1 perspective. Cytokine 110, 222–225 (2018).

    Article  CAS  PubMed  Google Scholar 

  46. Shiau, D.-J. et al. Hepatocellular carcinoma-derived high mobility group box 1 triggers M2 macrophage polarization via a TLR2/NOX2/autophagy axis. Sci. Rep. 10, 13582 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Braza, M. S. et al. Inhibiting inflammation with myeloid cell-specific nanobiologics promotes organ transplant acceptance. Immunity 49, 819–828.e6 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Warnatsch, A., Ioannou, M., Wang, Q. & Papayannopoulos, V. Inflammation. Neutrophil extracellular traps license macrophages for cytokine production in atherosclerosis. Science 349, 316–320 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Penkov, S., Mitroulis, I., Hajishengallis, G. & Chavakis, T. Immunometabolic crosstalk: an ancestral principle of trained immunity? Trends Immunol. 40, 1–11 (2019).

    Article  CAS  PubMed  Google Scholar 

  50. Gleeson, M. et al. The anti-inflammatory effects of exercise: mechanisms and implications for the prevention and treatment of disease. Nat. Rev. Immunol. 11, 607–615 (2011).

    Article  CAS  PubMed  Google Scholar 

  51. Fiuza-Luces, C. et al. Exercise benefits in cardiovascular disease: beyond attenuation of traditional risk factors. Nat. Rev. Cardiol. 15, 731–743 (2018).

    Article  CAS  PubMed  Google Scholar 

  52. Ai, L., Luo, W., Yuan, P., Liu, L. & Zhou, Y. Liver macrophages mediate effects of downhill running and caloric restriction on nonalcoholic fatty liver disease of high fat diet-fed mice. Life Sci. 256, 117978 (2020).

    Article  CAS  PubMed  Google Scholar 

  53. Kolahdouzi, S., Talebi-Garakani, E., Hamidian, G. & Safarzade, A. Exercise training prevents high-fat diet-induced adipose tissue remodeling by promoting capillary density and macrophage polarization. Life Sci. 220, 32–43 (2019).

    Article  CAS  PubMed  Google Scholar 

  54. Galván-Peña, S. & O’Neill, L. A. J. Metabolic reprograming in macrophage polarization. Front. Immunol. 5, 420 (2014).

    PubMed  PubMed Central  Google Scholar 

  55. Kelly, B. & O’Neill, L. A. J. Metabolic reprogramming in macrophages and dendritic cells in innate immunity. Cell Res. 25, 771–784 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  56. Christ, A. et al. Western diet triggers NLRP3-dependent innate immune reprogramming. Cell 172, 162–175.e14 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Liu, P.-S. et al. α-ketoglutarate orchestrates macrophage activation through metabolic and epigenetic reprogramming. Nat. Immunol. 18, 985–994 (2017).

    Article  CAS  PubMed  Google Scholar 

  58. Zhou, B. et al. The angiocrine Rspondin3 instructs interstitial macrophage transition via metabolic–epigenetic reprogramming and resolves inflammatory injury. Nat. Immunol. 21, 1430–1443 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Hooftman, A. et al. The immunomodulatory metabolite itaconate modifies NLRP3 and inhibits inflammasome activation. Cell Metab. 32, 468–478.e7 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Cordes, T. et al. Itaconate modulates tricarboxylic acid and redox metabolism to mitigate reperfusion injury. Mol. Metab. 32, 122–135 (2020).

    Article  CAS  PubMed  Google Scholar 

  61. Bambouskova, M. et al. Electrophilic properties of itaconate and derivatives regulate the IκBζ–ATF3 inflammatory axis. Nature 556, 501–504 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. Piguet, A.-C. et al. Regular exercise decreases liver tumors development in hepatocyte-specific PTEN-deficient mice independently of steatosis. J. Hepatol. 62, 1296–1303 (2015).

    Article  CAS  PubMed  Google Scholar 

  63. Huang, H. et al. Damage-associated molecular pattern-activated neutrophil extracellular trap exacerbates sterile inflammatory liver injury. Hepatology 62, 600–614 (2015).

    Article  CAS  PubMed  Google Scholar 

  64. Yu, Y.-R. et al. A protocol for the comprehensive flow cytometric analysis of immune cells in normal and inflamed murine non-lymphoid tissues. PLoS ONE 11, e0150606 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  65. Nguyen-Lefebvre, A. T. et al. The innate immune receptor TREM-1 promotes liver injury and fibrosis. J. Clin. Invest. 128, 4870–4883 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  66. Chen, L. et al. CD56 expression marks human group 2 innate lymphoid cell divergence from a shared NK cell and group 3 innate lymphoid cell developmental pathway. Immunity 49, 464–476.e4 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  67. Finck, R. et al. Normalization of mass cytometry data with bead standards. Cytometry A 83, 483–494 (2013).

    Article  PubMed  PubMed Central  Google Scholar 

  68. Amir, el-A. D. et al. viSNE enables visualization of high dimensional single-cell data and reveals phenotypic heterogeneity of leukemia. Nat. Biotechnol. 31, 545–552 (2013).

    Article  CAS  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank X. Liao and J. Imam for their assistance in preparing the manuscript. This work was supported by a National Institutes of Health grant no. 2T32AI106704-06 to A.O., grant nos. R01-CA214865-01 and R01-GM95566-06 to A.T., grant no. R01-AI152044 to M.D., grant nos. R01-GM120496 and R01-GM135234 to H. Wen and grant no. R01-GM137203 and Joseph A. Patrick Research Fellowship in Transplantation to H.H.

Author information

Authors and Affiliations

Authors

Contributions

H.Z., M.D., A.T. and H.H. conceived and designed the study. H.Z., M.D. and H.H. developed the methodology. J.R., Y.X. and H.H. acquired the data (provided animals, acquired and managed patients, provided the facilities). H.Z., A.O., C.S., J.H., H.H., T.C., J.W., Y.W., H. Wang, J.Z., G.K.B. and J.M.A. analysed and interpreted the data (statistical analysis, biostatistics, computational analysis). H.Z., H. Wen, M.D., A.T. and H.H. wrote and reviewed the manuscript. J.R., J.H. and A.H. provided administrative, technical, or material support (that is, reporting or organizing data, constructing databases). M.D., A.T. and H.H. supervised the study.

Corresponding authors

Correspondence to Meihong Deng, Allan Tsung or Hai Huang.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

Peer review information Nature Metabolism thanks the anonymous reviewers for their contribution to the peer review of this work. Primary Handling Editor: Isabella Samuelson.

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

Extended data

Extended Data Fig. 1 Pre-operative exercise reduces hepatic I/R injury and inflammation.

a, Liver damage were measured by serum AST level (P = 0.7606, P = 0.3610, P = 0.0555, P = 0.0549, P < 0.0001, P < 0.0001, P < 0.0001, P < 0.0001 between exercise and sedentary group from 1day to 16 weeks) and liver histological evaluation (Suzuki scores)) (P = 0.7464, P = 0.1898, P = 0.0932, P = 0.0441, P < 0.0001, P = 0.0002, P < 0.0001, P < 0.0001 between exercise and sedentary group from 1day to 16 weeks) from mice with sedentary or pre-operative exercise treatment for 1 day, or 1 to 16 weeks which subjected to hepatic I/R (n = 6 mice per group). b, Liver damage were measured by plasma ALT (P = 0.7548 in sham group, P < 0.0001 in I/R group) and AST levels (P = 0.2743 in sham group, P < 0.0001 in I/R group) from mice with sedentary or pre-operative exercise treatment for 4 weeks which subjected to hepatic I/R (Each dot represents one mouse, n = 7 mice in Sham group, n = 11 mice in I/R group). c, Liver damage were measured by liver histological evaluation (necrotic areas and Suzuki scores) (for necrotic areas, (P = 0.1228 in sham group, P < 0.0001 in I/R group), for Suzuki scores, (P = 0.8095 in sham group, P = 0.0018 in I/R group) from mice with sedentary or pre-operative exercise treatment for 4 weeks which subjected to hepatic I/R (Each dot represents one mouse, n = 6 mice per group). d, Serum IL-1Ra (P = 0.0001 in 4 weeks group, P = 0.0008 in 8 weeks group, P = 0.0005 in 12 weeks group, P = 0.0006 in 16 weeks group), IL-10 (P = 0.0008 in 4 weeks group, P = 0.0002 in 8 weeks group P = 0.0005 in 12 weeks group P = 0.0002 in 16 weeks group), IL-1β (P = 0.0004 in 4 weeks group, P = 0.0003 in 8 weeks group, P = 0.0002 in 12 weeks group, P = 0.0005 in 16 weeks group), IL-6 (P = 0.0015 in 4 weeks group, P = 0.0011 in 8 weeks group, P = 0.0007 in 12 weeks group, P = 0.0012 in 16 weeks group) and TNF-α (P = 0.0006 in 4 weeks group, P = 0.0011 in 8 weeks group, P = 0.0005 in 12 weeks group, P = 0.0002 in 16 weeks group)concentrations were measured by ELISA from mice with sedentary or pre-operative exercise treatment for 4, 8, 12 or 16 weeks which subjected to hepatic I/R (n = 6 mice per group). e, Serum IFN-α (P = 0.0854 in sham group, P = 0.04854 in I/R group), IFN-β (P = 03053 in sham group, P = 0.1154 in I/R group)and IFN-γ (P = 0.1531 in sham group, P = 0.0015 in I/R group) concentrations were measured by ELISA from mice with sedentary or pre-operative exercise treatment for 4 weeks at baseline (SHAM group) and subjected to hepatic I/R (Each dot represents one mouse, n = 6 mice per group). f, Serum CXCL1 (P < 0.0001 in sham group, P < 0.0001 in I/R group), CXCL2 (P = 0.0040 in sham group, P = 0.0001 in I/R group) and CXCL3 (P = 0.0071 in sham group, P < 0.0001 in I/R group) concentrations were measured by ELISA from mice with sedentary or pre-operative exercise treatment for 4 weeks at baseline (SHAM group) and subjected to hepatic I/R (Each dot represents one mouse, n = 6 mice per group). g, Liver damage were measured by serum AST level (P = 0.0127 between sedentary and 0 day after exercise group, P = 0.0001 between sedentary and 3 days after exercise group, P = 0.0019 between sedentary and 5 days after exercise group, P = 0.0082 between sedentary and 7 days after exercise group, p = 0.4524 between 0 day and 3 days group, P = 0.6955 between 0 day and 5 days group, P = 0.9063 between 0 day and 7 days group, P = 0.6701 between 3 days and 5 days group, P = 0.5088 between 3 days and 7 days group, P = 0.7702 between 5 days and 7 days group) from mice with sedentary or pre-operative exercise treatment for 4 weeks and resting for 0, 3, 5, or 7 days before hepatic I/R (Each dot represents one mouse, n = 11 in sedentary group, n = 7 in 0D after Exercise group, n = 15 in 3D after Exercise group, n = 9 in 5D after Exercise group, n = 7 in 7D after Exercise group). Graphs show the mean ± SD (a to f). Circles, triangles and squares represent individual mice. ANOVA with adjustment for multiple comparisons for (a and d). Unpaired two-sample Student’s t test for (b, c, e and f). NS: not significant. All the statistical test used in this figure was two-sided and two-sided P < 0.05 was considered statistically significant.

Extended Data Fig. 2 Mass cytometry and flow cytometry characterize hepatic immune profile.

a, Mass cytometry (CyTOF) of hepatic immune microenvironment profile from exercise or sedentary mice (4 weeks) with or without hepatic I/R in t-distributed stochastic neighbor embedding (t-SNE) graph. b, Percentage of Kupffer cells (KCs), monocytes, and neutrophils out of hepatic CD45 + cells. (Each dot represents one mouse, n = 2 mice per group) c, Percentage of neutrophils. d, Percentage of natural killer (NK) cells, NKT cells, and CD3 + T cells out of hepatic CD45 + cells in each group. e, Numbers of B cells (P = 0.3397 in sham group, P = 0.8025 in I/R group), CD4 + T cells (P = 0.6370 in sham group, P = 0.3695 in I/R group), CD8 + T cells(P = 0.7879 in sham group, P = 0.6170 in I/R group), and NKT cells(P = 0.6738 in sham group, P = 0.3010 in I/R group), dendritic cells (DCs) (P = 0.3910 in sham group, P = 0.7954 in I/R group), out of hepatic CD45 + cells in each group (Each dot represents one mouse, n = 6 mice per group). Circles and triangles represent individual mice. Graphs show the mean ± SD (b and e). Unpaired two-sample Student’s t test (b and e). All the statistical test used in this figure was two-sided and two-sided P < 0.05 was considered statistically significant.

Extended Data Fig. 3 Single cell RNA sequencing reveals transcriptomic profile of hepatic immune cells.

a, Experimental workflow of single cell RNA-sequencing in hepatic CD45 + cells with sedentary or pre-operative exercise treatment for 4 weeks. b, Heat map of gene expression profile in 15,332 hepatic immune cells clustered into 18 discrete cell populations. c, Transcriptomic profile of feature genes expression.

Extended Data Fig. 4 Single cell RNA sequencing reveals transcriptional signature of Kupffer cells.

a, Based on the transcriptomic profile, 15,332 cells were clustered into 18 discrete cell populations in Uniform Manifold Approximation and Projection (UMAP). b, Cluster 0 was one unique subpopulation of KCs mainly derived from exercise mice and Cluster 2 were mainly derived from sedentary mice. c, Kupffer cells percentages in cluster 0, 2, 5 and 10. d, Preference of cluster from Kupffer cell estimated by RO/E. + + + , RO/E > 2; ++, 1.5<RO/E < = 2; +/-, 0.5 = <RO/E < = 1.5; - RO/E < 0.5. e, Volcano plot shows that the expression of 6205 genes was significantly changed with pre-operative exercise.

Extended Data Fig. 5 Kupffer cells are required for pre-operative exercise mediated hepatic protection from I/R injury.

a, Experiment outline. b, Experiment outline. c, Flow cytometry characterization of Kupffer cells percentage after mice injected with saline or different concentration of GdCl3. d, Liver damage was measured by serum AST level (P = 0.0130 in saline group, P = 0.9197 in GdCl3 group) and liver histological evaluation (Suzuki scores) (P = 0.0023 in saline group, P = 0.2513 in GdCl3 group) from mice with sedentary or pre-operative exercise treatment for 4 weeks injected with GdCl3 (25 mg/kg) or saline solution intraperitoneally 48 hours before hepatic I/R (Each dot represents one mouse, n = 6 mice per group). e, Experiment outline. f, Adoptive transfer of Kupffer cells isolated from either sedentary or pre-operative exercise treatment for 4 weeks into Kupffer cells KO mice with GdCl3. Liver damage was measured by serum AST level (P = 0.0083 in sedentary recipient group, P = 0.0092 in exercise recipient group) and liver histological evaluation (Suzuki scores) (P = 0.0154 in sedentary recipient group, P = 0.0026 in exercise recipient group) among mice from these four groups subjected to hepatic I/R (Each dot represents one mouse, n = 6 mice per group). Graphs show the mean ± SD (d and f). Circles and triangles represent individual mouse. Unpaired two-sample Student’s t test for (d and f). All the statistical test used in this figure was two-sided and two-sided P < 0.05 was considered statistically significant.

Extended Data Fig. 6 Pre-operative exercise induces metabolic reprogramming in Kupffer cells.

a, Metabolite sets enrichment between Kupffer cells with sedentary or pre-operative exercise treatment for 4 weeks. b, Relative expression between Kupffer cells with sedentary or pre-operative exercise treatment for 4 weeks.

Extended Data Fig. 7 Pre-operative exercise-induced protective effect on hepatic I/R injury is dependent on itaconate/IRG1.

a, Experiment outline. b, Liver damage for Irg1+/+ and Irg1−/− mice with sedentary or pre-operative exercise treatment for 4 weeks after hepatic I/R was measured by serum AST level (P = 0.0013 between Irg1+/+ sedentary and Irg1+/+ exercise group, P = 0.0425 between Irg1+/+ sedentary and Irg1−/− sedentary group, P < 0.0001 between Irg1+/+ exercise and Irg1−/− sedentary group, P = 0.0005 between Irg1+/+ exercise and Irg1−/− exercise group, P = 0.7057 between Irg1+/+ sedentary and Irg1−/− exercise group) (Each dot represents one mouse, n = 6 mice per group). c, Serum IL-1Ra (P = 0.3521), IL-10 (P = 0.5245), IL-1β (P = 0.1919), IL-6 (P = 0.4139) and TNF-α (P = 0.9512) concentrations from Irg1−/− exercise or sedentary mice (4 weeks) after hepatic I/R were measured by ELISA (Each dot represents one mouse, n = 6 mice per group). d, Liver damage of KCs-depleted Irg1+/+ mice were adoptively transferred with Irg1−/− or Irg1+/+ KCs from sedentary or exercise mice after hepatic I/R were measured by serum AST levels ((P = 0.0020 between KC-Irg1+/+ sedentary and KC-Irg1+/+ exercise group, P = 0.0413 between KC-Irg1+/+ sedentary and KC-Irg1−/− sedentary group, P = 0.0004 between KC-Irg1+/+ exercise and KC-Irg1−/− sedentary group, P = 0.0003 between KC-Irg1+/+ exercise and KC-Irg1−/− exercise group, P = 0.6203 between KC-Irg1+/+ sedentary and KC-Irg1−/− exercise group) (Each dot represents one mouse, n = 6 mice per group). e, Liver damage for Irg1+/+ or Irg1−/− mice injected itaconate (ITA, 100 mg/kg) for 2 hours before hepatic ischaemia liver I/R and at the time of reperfusion with sedentary or pre-operative exercise treatment for 4 weeks after hepatic I/R was measured by serum AST level (P = 0.0028 between Irg1+/+ sedentary and Irg1+/+ exercise group, P = 0.0446 between Irg1+/+ sedentary and Irg1−/− sedentary group, P < 0.0001 between Irg1+/+ exercise and Irg1−/− sedentary group, P = 0.0802 between Irg1+/+ exercise and Irg1−/− exercise group, P = 0.0013 between Irg1−/− sedentary and Irg1−/− exercise group) (Each dot represents one mouse, n = 6 mice per group). f, Liver damage for Irg1+/+ or Irg1−/− mice injected 4-OI (25 mg/kg) for 24 and 2 hours before hepatic ischeamia, and at the time of reperfusion with sedentary or pre-operative exercise treatment for 4 weeks after hepatic I/R was measured by serum AST level (P = 0.0012 between Irg1+/+ sedentary and Irg1+/+ exercise group, P = 0.0070 between Irg1+/+ sedentary and Irg1−/− sedentary group, P < 0.0001 between Irg1+/+ exercise and Irg1−/− sedentary group, P = 0.0012 between Irg1+/+ exercise and Irg1−/− exercise group, P = 0.0027 between Irg1−/− sedentary and Irg1−/− exercise group) (Each dot represents one mouse, n = 6 mice per group) g, Liver damage for Nrf2+/+ and Nrf2−/− mice with sedentary or pre-operative exercise treatment for 4 weeks after hepatic I/R was measured by serum AST level (P = 0.0131 between Nrf2+/+ sedentary and Nrf2+/+ exercise group, P = 0.0486 between Nrf2+/+ sedentary and Nrf2−/− sedentary group, P = 0.0004 between Nrf2+/+ exercise and Nrf2−/− exercise group, P = 0.9587 between Nrf2−/− sedentary and Nrf2−/− exercise group) (Each dot represents one mouse, n = 3-6 mice in both Nrf2+/+ Sedentary and Exercise group, n = 3 in Nrf2−/− Sedentary group, n = 5 in Nrf2−/− Exercise group). Graphs show the mean ± SD (b to g). Circles, triangles and squares represent individual mice. ANOVA with adjustment for multiple comparisons (b, d, e, f and g). Unpaired two-sample Student’s t test (c). All the statistical test used in this figure was two-sided and two-sided P < 0.05 was considered statistically significant.

Extended Data Fig. 8 Pre-operative exercise promotes anti-inflammatory trained immunity in Kupffer cells.

a, Experiment outline. b, Trained immunity marker IL-1Ra (P = 0.0035), IL-10 (P = 0.0207), IL-1β (P = 0.0218), IL-6 (P = 0.0069) and TNF-α (P = 0.0145) were measured from the medium of Kupffer cells which isolated from normal mice with re-stimulation of LPS by ELISA (n = 3 mice per group). c, Experiment outline. d, Trained immunity marker IL-1Ra (P = 0.6551, P = 0.0229, P = 0.9025, P = 0.0032 between sedentary and exercise group at the time point of baseline, after exercise, before hypoxia, and after hypoxia) and IL-10(P = 0.5531, P = 0.0149, P = 0.4440, P = 0.0223 between sedentary and exercise group at the time point of baseline, after exercise, before hypoxia, and after hypoxia), IL-1β (P = 0.6841, P = 0.0188, P = 0.3258, P = 0.0226 between sedentary and exercise group at the time point of baseline, after exercise, before hypoxia, and after hypoxia), IL-6 (P = 0.5076, P = 0.0148, P = 0.3301, P = 0.0084 between sedentary and exercise group at the time point of baseline, after exercise, before hypoxia, and after hypoxia) and TNF-α (P = 0.7328, P = 0.0088, P = 0.8229, P = 0.0202 between sedentary and exercise group at the time point of baseline, after exercise, before hypoxia, and after hypoxia) were measured from the medium of Kupffer cells which isolated from mice received sedentary or pre-operative exercise treatment for 4 weeks before and after the second stimulation of LPS by ELISA (Each dot represents one mouse, n = 6 mice per group). e, Experiment outline. f, Liver damage was measured by serum ALT level (P < 0.0001 between sedentary and all exercise groups, p = 0.8403 between 0 days and 3 days group, P = 0.5430 between 0 day and 5 days group, P = 0.2980 between 0 day and 7 days group, P = 0.5802 between 3 days and 5 days group, P = 0.2119 between 3 days and 7 days group, P = 0.4277 between 5 days and 7 days group) and liver histological evaluation (necrotic areas) (P = 0.0482 between sedentary and 0 day after exercise group, P = 0.0061 between sedentary and 3 days after exercise group, P = 0.0424 between sedentary and 5 days after exercise group, P = 0.0260 between sedentary and 7 days after exercise group, p = 0.1908 between 0 day and 3 days group, P = 0.8088 between 0 day and 5 days group, P = 0.8722 between 0 day and 7 days group, P = 0.0599 between 3 days and 5 days group, P = 0.1512 between 3 days and 7 days group, P = 0.6227 between 5 days and 7 days group) from mice with sedentary or pre-operative exercise treatment for 4 weeks and resting for 0, 3, 5, or 7 days before LPS injection (Each dot represents one mouse, n = 11 in sedentary group, n = 7 in 0D after Exercise group, n = 8 in 3D after Exercise group, n = 9 in 5D after Exercise group, n = 7 in 7D after Exercise group). g, IL-1Ra (P < 0.0001), IL-10 (P = 0.0023), IL-1β (P = 0.0293), IL-6 (P = 0.0003) and TNF-α (P = 0.0007) concentrations were measured from the medium of Kupffer cells which isolated from mice with sedentary or pre-operative exercise treatment for 4 weeks with LPS injection (Each dot represents one mouse, n = 6 mice per group). Graphs show the mean ± SD (b, d, f and g). Circles, triangles and squares represent individual mice. Unpaired two-sample Student’s t test for (b and g). ANOVA with adjustment for multiple comparisons for (d and f). NS: not significant. All the statistical test used in this figure was two-sided and two-sided P < 0.05 was considered statistically significant.

Extended Data Fig. 9 Anti-inflammatory trained immunity in Kupffer cells relies on anti-inflammatory metabolite, itaconate.

a, Experiment outline. b, Viability of 24 hours in unstimulated Kupffer cells treated with 4-OI and Itaconate. The treatment concentrations for all experiments listed in mM. c, IL-1Ra (P < 0.0001 between Vehicle and 4-OI treatment, P = 0.0016 between Vehicle and Itaconate treatment), IL-10 (P < 0.0001 between Vehicle and 4-OI treatment, P = 0.0497 between Vehicle and Itaconate treatment), IL-1β (P = 0.0015 between Vehicle and 4-OI treatment, P = 0.0207 between Vehicle and Itaconate treatment), IL-6 (P = 0.0002 between Vehicle and 4-OI treatment, P = 0.0013 between Vehicle and Itaconate treatment) and TNF-α (P = 0.0016 between Vehicle and 4-OI treatment, P = 0.0008 between Vehicle and Itaconate treatment) concentrations from the medium of Kupffer cells which isolated from normal mice with the stimulation of vehicle or 4-OI or Itaconate after the challenge of LPS or hypoxia were measured by ELISA (Each dot represents one mouse, n = 6 mice per group). d, Experiment outline e, IL-1Ra (P = 0.0172 between Vehicle and 4-OI treatment, P = 0.0463 between Vehicle and Itaconate treatment), IL-10 (P = 0.0264 between Vehicle and 4-OI treatment, P = 0.0015 between Vehicle and Itaconate treatment), IL-1β (P = 0.0498 between Vehicle and 4-OI treatment, P = 0.0315 between Vehicle and Itaconate treatment), IL-6 (P = 0.0431 between Vehicle and 4-OI treatment, P = 0.0193 between Vehicle and Itaconate treatment) and TNF-α (P = 0.0035 between Vehicle and 4-OI treatment, P = 0.0104 between Vehicle and Itaconate treatment) concentrations from the medium of Kupffer cells which isolated from Irg1−/− l mice with the stimulation of vehicle, 4-OI or itaconate after the challenge of hypoxia were measured by ELISA (Each dot represents one mouse, n = 6 mice per group). f, IL-1Ra (P = 0.0476 between Vehicle and 4-OI treatment, P = 0.0100 between Vehicle and Itaconate treatment), IL-10 (P = 0.0114 between Vehicle and 4-OI treatment, P = 0.0487 between Vehicle and Itaconate treatment), IL-1β (P = 0.0406 between Vehicle and 4-OI treatment, P = 0.0048 between Vehicle and Itaconate treatment), IL-6 (P = 0.0278 between Vehicle and 4-OI treatment, P = 0.0094 between Vehicle and Itaconate treatment) and TNF-α (P = 0.0208 between Vehicle and 4-OI treatment, P = 0.0184 between Vehicle and Itaconate treatment) concentrations from the medium of Kupffer cells which isolated from Irg1−/− l mice with the stimulation of vehicle, 4-OI or itaconate after the challenge of LPS were measured by ELISA (Each dot represents one mouse, n = 6 mice per group). g, Experiment outline. h, IL-1Ra (P = 0.0543), IL-10 (P = 0.0623), IL-1β (P = 0.2723), IL-6 (P = 0.06754) and TNF-α (P = 0.9481) concentrations from the medium of Kupffer cells which isolated from Irg1−/− mice with sedentary or pre-operative exercise treatment after the challenge of LPS or hypoxia for 4 weeks were measured by ELISA (Each dot represents one mouse, n = 6 mice per group). Graphs show the mean ± SD (c, e, f and h). Circles represent individual mice. Unpaired two-sample Student’s t test for (c, e, f and h). All the statistical test used in this figure was two-sided and two-sided P < 0.05 was considered statistically significant.

Supplementary information

Supplementary Information

Supplementary Fig. 1

Reporting Summary

Supplementary Data 1

Custom code used for transcriptomic profiling in Fig. 2a–c and Extended Data Fig. 3b,c.

Supplementary Data 2

Custom code used for heatmap generation in Fig. 2d.

Supplementary Data 3

Custom code used for pathway analysis in Fig. 2d.

Supplementary Data 4

Custom code used for Preference of cluster analysis in Extended Data Fig. 4d.

Supplementary Data 5

Custom code used for the volcano plot analysis in Extended Data Fig. 4e.

Source data

Source Data Fig. 1

Uncropped scans of the gels: uncropped western blots images. The dashed boxes denote cropped images that are presented in the manuscript.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Zhang, H., Chen, T., Ren, J. et al. Pre-operative exercise therapy triggers anti-inflammatory trained immunity of Kupffer cells through metabolic reprogramming. Nat Metab 3, 843–858 (2021). https://doi.org/10.1038/s42255-021-00402-x

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s42255-021-00402-x

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