Skip to main content

Advertisement

Log in

LIFR regulates cholesterol-driven bidirectional hepatocyte–neutrophil cross-talk to promote liver regeneration

  • Article
  • Published:

From Nature Metabolism

View current issue Submit your manuscript

Abstract

Liver regeneration is under metabolic and immune regulation. Despite increasing recognition of the involvement of neutrophils in regeneration, it is unclear how the liver signals to the bone marrow to release neutrophils after injury and how reparative neutrophils signal to hepatocytes to reenter the cell cycle. Here we report that loss of the liver tumour suppressor Lifr in mouse hepatocytes impairs, whereas overexpression of leukaemia inhibitory factor receptor (LIFR) promotes liver repair and regeneration after partial hepatectomy or toxic injury. In response to physical or chemical damage to the liver, LIFR from hepatocytes promotes the secretion of cholesterol and CXCL1 in a STAT3-dependent manner, leading to the efflux of bone marrow neutrophils to the circulation and damaged liver. Cholesterol, via its receptor ERRα, stimulates neutrophils to secrete hepatocyte growth factor to accelerate hepatocyte proliferation. Altogether, our findings reveal a LIFR–STAT3–CXCL1–CXCR2 axis and a LIFR–STAT3–cholesterol–ERRα–hepatocyte growth factor axis that form bidirectional hepatocyte–neutrophil cross-talk to repair and regenerate the liver.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Fig. 1: Loss of Lifr in hepatocytes impairs liver regeneration.
Fig. 2: Hepatocytic Lifr promotes neutrophil recruitment in liver regeneration models.
Fig. 3: Hepatocytic Lifr promotes HGF production through neutrophils.
Fig. 4: LIFR upregulates hepatocyte-derived cholesterol, which acts on neutrophils to boost HGF production.
Fig. 5: Cholesterol promotes HGF transcription through ERRα in neutrophils.
Fig. 6: LIFR accelerates partial hepatectomy-induced liver injury repair and regeneration in a neutrophil-dependent manner.
Fig. 7: Hepatocytic Lifr promotes neutrophil recruitment through Cxcl1.
Fig. 8: LIFR regulates CXCL1 and cholesterol levels through STAT3, and treatment with a STAT3 inhibitor reverses LIFR-accelerated liver regeneration.

Similar content being viewed by others

Data availability

Source data are provided with this paper. All other data supporting the findings of this study are available within the article, extended data figures or Supplementary Information.

References

  1. Michalopoulos, G. K. & Bhushan, B. Liver regeneration: biological and pathological mechanisms and implications. Nat. Rev. Gastroenterol. Hepatol. 18, 40–55 (2021).

    Article  PubMed  Google Scholar 

  2. Forbes, S. J. & Rosenthal, N. Preparing the ground for tissue regeneration: from mechanism to therapy. Nat. Med. 20, 857–869 (2014).

    Article  CAS  PubMed  Google Scholar 

  3. Thorgersen, E. B. et al. The role of complement in liver injury, regeneration, and transplantation. Hepatology 70, 725–736 (2019).

    Article  PubMed  Google Scholar 

  4. Sakamoto, Y. et al. Favorable indications for hepatectomy in patients with liver metastasis from gastric cancer. J. Surg. Oncol. 95, 534–539 (2007).

    Article  PubMed  Google Scholar 

  5. Li, N. & Hua, J. Immune cells in liver regeneration. Oncotarget 8, 3628–3639 (2017).

    Article  PubMed  Google Scholar 

  6. Patijn, G. A., Lieber, A., Schowalter, D. B., Schwall, R. & Kay, M. A. Hepatocyte growth factor induces hepatocyte proliferation in vivo and allows for efficient retroviral-mediated gene transfer in mice. Hepatology 28, 707–716 (1998).

    Article  CAS  PubMed  Google Scholar 

  7. Kiso, S. et al. Liver regeneration in heparin-binding EGF-like growth factor transgenic mice after partial hepatectomy. Gastroenterology 124, 701–707 (2003).

    Article  CAS  PubMed  Google Scholar 

  8. Michalopoulos, G. K. Liver regeneration. J. Cell. Physiol. 213, 286–300 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Burn, G. L., Foti, A., Marsman, G., Patel, D. F. & Zychlinsky, A. The neutrophil. Immunity 54, 1377–1391 (2021).

    Article  CAS  PubMed  Google Scholar 

  10. Wang, J. et al. Visualizing the function and fate of neutrophils in sterile injury and repair. Science 358, 111–116 (2017).

    Article  CAS  PubMed  Google Scholar 

  11. Sas, A. R. et al. A new neutrophil subset promotes CNS neuron survival and axon regeneration. Nat. Immunol. 21, 1496–1505 (2020).

    Article  PubMed  PubMed Central  Google Scholar 

  12. Fischer, A. et al. Neutrophils direct preexisting matrix to initiate repair in damaged tissues. Nat. Immunol. 23, 518–531 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Luo, J., Yang, H. & Song, B. L. Mechanisms and regulation of cholesterol homeostasis. Nat. Rev. Mol. Cell Biol. 21, 225–245 (2020).

    Article  CAS  PubMed  Google Scholar 

  14. Delgado-Coello, B., Briones-Orta, M. A., Macias-Silva, M. & Mas-Oliva, J. Cholesterol: recapitulation of its active role during liver regeneration. Liver Int. 31, 1271–1284 (2011).

    Article  CAS  PubMed  Google Scholar 

  15. Huang, W. et al. Nuclear receptor-dependent bile acid signaling is required for normal liver regeneration. Science 312, 233–236 (2006).

    Article  CAS  PubMed  Google Scholar 

  16. Ware, C. B. et al. Targeted disruption of the low-affinity leukemia inhibitory factor receptor gene causes placental, skeletal, neural and metabolic defects and results in perinatal death. Development 121, 1283–1299 (1995).

    Article  CAS  PubMed  Google Scholar 

  17. Yao, F. et al. A targetable LIFR–NF-κB–LCN2 axis controls liver tumorigenesis and vulnerability to ferroptosis. Nat. Commun. 12, 7333 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Mitchell, C. & Willenbring, H. A reproducible and well-tolerated method for 2/3 partial hepatectomy in mice. Nat. Protoc. 3, 1167–1170 (2008).

    Article  CAS  PubMed  Google Scholar 

  19. Jia, Y. et al. In vivo CRISPR screening identifies BAZ2 chromatin remodelers as druggable regulators of mammalian liver regeneration. Cell Stem Cell 29, 372–385 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Avasarala, S. et al. A temporal study on the histopathological, biochemical and molecular responses of CCl(4)-induced hepatotoxicity in Cyp2e1-null mice. Toxicology 228, 310–322 (2006).

    Article  CAS  PubMed  Google Scholar 

  21. Bandura, D. R. et al. Mass cytometry: technique for real time single cell multitarget immunoassay based on inductively coupled plasma time-of-flight mass spectrometry. Anal. Chem. 81, 6813–6822 (2009).

    Article  CAS  PubMed  Google Scholar 

  22. Daley, J. M., Thomay, A. A., Connolly, M. D., Reichner, J. S. & Albina, J. E. Use of Ly6G-specific monoclonal antibody to deplete neutrophils in mice. J. Leukoc. Biol. 83, 64–70 (2008).

    Article  CAS  PubMed  Google Scholar 

  23. Xiao, Y. et al. Cathepsin C promotes breast cancer lung metastasis by modulating neutrophil infiltration and neutrophil extracellular trap formation. Cancer Cell 39, 423–437 e427 (2021).

    Article  CAS  PubMed  Google Scholar 

  24. Peiseler, M. & Kubes, P. More friend than foe: the emerging role of neutrophils in tissue repair. J. Clin. Invest. 129, 2629–2639 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  25. Alves-Bezerra, M. & Cohen, D. E. Triglyceride metabolism in the liver. Compr. Physiol. 8, 1–8 (2017).

    PubMed  PubMed Central  Google Scholar 

  26. Rodrigues, H. G., Takeo Sato, F., Curi, R. & Vinolo, M. A. R. Fatty acids as modulators of neutrophil recruitment, function and survival. Eur. J. Pharmacol. 785, 50–58 (2016).

    Article  CAS  PubMed  Google Scholar 

  27. Hauert, A. B., Martinelli, S., Marone, C. & Niggli, V. Differentiated HL-60 cells are a valid model system for the analysis of human neutrophil migration and chemotaxis. Int. J. Biochem. Cell Biol. 34, 838–854 (2002).

    Article  CAS  PubMed  Google Scholar 

  28. Woo, C. H. et al. Transepithelial migration of neutrophils in response to leukotriene B4 is mediated by a reactive oxygen species-extracellular signal-regulated kinase-linked cascade. J. Immunol. 170, 6273–6279 (2003).

    Article  CAS  PubMed  Google Scholar 

  29. Babatunde, K. A. et al. Chemotaxis and swarming in differentiated HL-60 neutrophil-like cells. Sci. Rep. 11, 778 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Wei, W. et al. Ligand activation of ERRα by cholesterol mediates statin and bisphosphonate effects. Cell Metab. 23, 479–491 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Nakadai, T. et al. Two target gene activation pathways for orphan ERR nuclear receptors. Cell Res. 33, 165–183 (2023).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Sallusto, F. & Baggiolini, M. Chemokines and leukocyte traffic. Nat. Immunol. 9, 949–952 (2008).

    Article  CAS  PubMed  Google Scholar 

  33. Capucetti, A., Albano, F. & Bonecchi, R. Multiple roles for chemokines in neutrophil biology. Front. Immunol. 11, 1259 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Kumar, V. et al. Cancer-Associated fibroblasts neutralize the anti-tumor effect of CSF1 receptor blockade by inducing PMN-MDSC infiltration of tumors. Cancer Cell 32, 654–668 e655 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Heinrich, P. C., Behrmann, I., Muller-Newen, G., Schaper, F. & Graeve, L. Interleukin-6-type cytokine signalling through the gp130/Jak/STAT pathway. Biochem. J. 334, 297–314 (1998).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Takahashi-Tezuka, M. et al. Gab1 acts as an adapter molecule linking the cytokine receptor gp130 to ERK mitogen-activated protein kinase. Mol. Cell. Biol. 18, 4109–4117 (1998).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Luo, Q. et al. LIFR functions as a metastasis suppressor in hepatocellular carcinoma by negatively regulating phosphoinositide 3-kinase/AKT pathway. Carcinogenesis 36, 1201–1212 (2015).

    Article  CAS  PubMed  Google Scholar 

  38. Chen, D. et al. LIFR is a breast cancer metastasis suppressor upstream of the Hippo-YAP pathway and a prognostic marker. Nat. Med. 18, 1511–1517 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Kienzl-Wagner, K. et al. The role of lipocalin-2 in liver regeneration. Liver Int. 35, 1195–1202 (2015).

    Article  CAS  PubMed  Google Scholar 

  40. Cressman, D. E. et al. Liver failure and defective hepatocyte regeneration in interleukin-6-deficient mice. Science 274, 1379–1383 (1996).

    Article  CAS  PubMed  Google Scholar 

  41. Li, W., Liang, X., Kellendonk, C., Poli, V. & Taub, R. STAT3 contributes to the mitogenic response of hepatocytes during liver regeneration. J. Biol. Chem. 277, 28411–28417 (2002).

    Article  CAS  PubMed  Google Scholar 

  42. Zhang, H. et al. IL-6 trans-signaling promotes pancreatitis-associated lung injury and lethality. J. Clin. Invest. 123, 1019–1031 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Sander, L. E. et al. Hepatic acute-phase proteins control innate immune responses during infection by promoting myeloid-derived suppressor cell function. J. Exp. Med. 207, 1453–1464 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Jung, K. H. et al. Multifunctional effects of a small-molecule STAT3 inhibitor on NASH and hepatocellular carcinoma in mice. Clin. Cancer Res. 23, 5537–5546 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Chen, Y. Y., Ge, J. Y., Zhu, S. Y., Shao, Z. M. & Yu, K. D. Copy number amplification of ENSA promotes the progression of triple-negative breast cancer via cholesterol biosynthesis. Nat. Commun. 13, 791 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. de Oliveira, S., Rosowski, E. E. & Huttenlocher, A. Neutrophil migration in infection and wound repair: going forward in reverse. Nat. Rev. Immunol. 16, 378–391 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  47. Nejak-Bowen, K., Orr, A., Bowen, W. C. Jr & Michalopoulos, G. K. Conditional genetic elimination of hepatocyte growth factor in mice compromises liver regeneration after partial hepatectomy. PLoS ONE 8, e59836 (2013).

  48. De Giovanni, M. et al. GPR35 promotes neutrophil recruitment in response to serotonin metabolite 5-HIAA. Cell 185, 815–830 e819 (2022).

    Article  PubMed  PubMed Central  Google Scholar 

  49. Passegue, E., Wagner, E. F. & Weissman, I. L. JunB deficiency leads to a myeloproliferative disorder arising from hematopoietic stem cells. Cell 119, 431–443 (2004).

    Article  CAS  PubMed  Google Scholar 

  50. Wang, G. et al. Identification of the transgene integration site and host genome changes in MRP8-Cre/ires-EGFP transgenic mice by targeted locus amplification. Front. Immunol. 13, 875991 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Hu, Z. et al. CREBZF as a key regulator of STAT3 pathway in the control of liver regeneration in mice. Hepatology 71, 1421–1436 (2020).

    Article  CAS  PubMed  Google Scholar 

  52. Zhao, Z. et al. Hepatic PPARα function is controlled by polyubiquitination and proteasome-mediated degradation through the coordinated actions of PAQR3 and HUWE1. Hepatology 68, 289–303 (2018).

    Article  CAS  PubMed  Google Scholar 

  53. Huang, M. et al. C-C motif chemokine ligand 5 confines liver regeneration by down-regulating reparative macrophage-derived hepatocyte growth factor in a forkhead box O 3a-dependent manner. Hepatology 76, 1706–1722 (2022).

    Article  CAS  PubMed  Google Scholar 

  54. Oh, H., Siano, B. & Diamond, S. Neutrophil isolation protocol. J. Vis. Exp. 23, 745 (2008).

  55. Zhu, Y. P. et al. Preparation of whole bone marrow for mass cytometry analysis of neutrophil-lineage cells. J. Vis. Exp. https://doi.org/10.3791/59617 (2019).

  56. Deng, Y. et al. Glucocorticoid receptor regulates PD-L1 and MHC-I in pancreatic cancer cells to promote immune evasion and immunotherapy resistance. Nat. Commun. 12, 7041 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank MD Anderson’s Flow Cytometry and Cellular Imaging Core, Metabolomics Core, Functional Genomics Core, Cytogenetics and Cell Authentication Core, and Advanced Technology Genome Core for technical assistance. We are grateful to all members of the laboratory of L.M. for the discussion and to C. F. Wogan (MD Anderson’s Division of Radiation Oncology) for the critical reading of the paper. L.M. is supported by US National Institutes of Health (NIH) grants R01CA166051 and R01CA269140, an American Cancer Society grant (award DBG-22-161-01-MM) and the Nylene Eckles Distinguished Professorship of MD Anderson Cancer Center. H.Z. is supported by the NIH (R01AA028791, R01DK125396), Cancer Prevention and Research Institute of Texas (CPRIT, RP220614), the Emerging Leader Award from the Mark Foundation for Cancer Research (award 21-003-ELA) and the Nancy B. and Jake L. Hamon Distinguished Chair of University of Texas Southwestern Medical Center. The core facilities are supported by MD Anderson’s Cancer Center Support Grant P30CA016672 from the NIH. The funders had no role in the study design, data collection and analysis, decision to publish, or preparation of the manuscript.

Author information

Authors and Affiliations

Authors

Contributions

Y.D., Y.S. and L.M. conceived and designed the study. Y.D. and Z.Z. performed most experiments and data analyses. M.S., Y.Z., H.T., C.M. and J.Z. performed some experiments. C.L. and F.Y. provided technical assistance and consultation. Y.S. generated some reagents and contributed protocols. M.A.C. and H.Z. reviewed the data and provided substantial intellectual input and guidance. Y.D. and L.M. wrote the paper with input from all other authors. L.M. provided scientific direction, established collaborations and allocated funding for this study.

Corresponding author

Correspondence to Li Ma.

Ethics declarations

Competing interests

H.Z. consults for Flagship Pioneering, Alnylam Pharmaceuticals, Jumble Therapeutics and Chroma Medicines, and serves on the Scientific Advisory Board of Ubiquitix. H.Z. has research support from Chroma Medicines. H.Z. owns stock in Ionis and Madrigal Pharmaceuticals. M.A.C. reports grants, personal fees and an ownership interest in ImmunoGenesis and personal fees from AstraZeneca. The above interests are not directly related to the contents of this paper. The other authors declare no competing interests.

Peer review

Peer review information

Nature Metabolism thanks Udayan Apte, Paul Kubes and Jan Tchorz for their contribution to the peer review of this work. Primary Handling Editors: Revati Dewal and Isabella Samuelson, 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 Loss of Lifr in hepatocytes impairs liver injury repair and injury-induced upregulation of proliferative genes.

a. Immunoblotting of Lifr, cyclin D1, cyclin A2, and Gapdh in mouse livers at different time points after 2/3 partial hepatectomy (PHx). b. qPCR of mRNA of Lifr, cyclin D1, cyclin A2, cyclin B1, and cyclin E1 in the livers of Lifrfl/fl and Lifrfl/fl;Alb-Cre mice at 72 hours after PHx. n = 6, 5, 6, and 6 mice. c. Immunoblotting of Lifr, cyclin D1, and Gapdh in the livers of Lifrfl/fl and Lifrfl/fl;Alb-Cre mice at 72 hours after PHx. d, e. TUNEL staining (d) and the number of TUNEL-positive hepatocytes per high-power field (HPF; e) at 72 hours after CCl4 treatment. Scale bars, 50 μm. n = 4 mice. f. qPCR of mRNA of cyclin D1, cyclin A2, and Pcna in the livers of Lifrfl/fl and Lifrfl/fl;Alb-Cre mice at 48 and 72 hours after CCl4 treatment. n = 7, 7, 4, 4, 6, 6, 8, and 8 mice. g, h. Serum ALT (g) and AST (h) levels in Lifrfl/fl and Lifrfl/fl;Alb-Cre mice at 6 and 12 hours after CCl4 treatment. n = 8, 6, 6, 7, 6, and 8 mice. i. Immunoblotting of Cyp2e1 and Gapdh in mouse livers at 6 and 12 hours after CCl4 treatment. Representative results from one of three independent experiments are shown. Statistical significance in b and e-h was determined by a two-tailed unpaired t-test. Error bars are s.e.m.

Source data

Extended Data Fig. 2 Overexpression of LIFR promotes liver injury repair and regeneration.

a-i. C57BL/6J mice received control or LIFR-expressing adenovirus 5 days before CCl4 or vehicle treatment. Analyses were done at 48 hours after treatment. a. Experimental design. b, c. Serum ALT (b) and AST (c) levels in mice after CCl4 or vehicle treatment. n = 6 mice. d, e. H&E staining (d) and percentage of necrotic areas (e) in mouse livers after CCl4 or vehicle treatment. Scale bars, 500 μm. n = 6 mice. f, g. DAPI and TUNEL staining (f) and the number of TUNEL-positive hepatocytes per high-power field (HPF; g) in mouse livers after CCl4 or vehicle treatment. Scale bars, 100 μm. n = 6 mice. h, i. Immunofluorescence staining of Ki67 (h; overlay with DAPI staining) and percentage of Ki67-positive hepatocytes (i) in mouse livers after CCl4 or vehicle treatment. Scale bars, 50 μm. n = 3 mice. j. DAPI and TUNEL staining of mouse livers 10 days after injection of control or LIFR-expressing adenovirus. Scale bars, 100 μm. Representative results from one of three independent experiments are shown. k-m. C57BL/6J mice received control or LIFR-expressing adenovirus 10 days before CCl4 treatment. Analyses were done at 48 hours after treatment. k. Experimental design. l, m. Immunofluorescence staining of Ki67 (l; overlay with DAPI staining) and percentage of Ki67-positive hepatocytes (m) after CCl4 treatment. Scale bars, 50 μm. n = 6 mice. Statistical significance in b, c, e, g, i, and m was determined by a two-tailed unpaired t-test. Error bars are s.e.m.

Source data

Extended Data Fig. 3 LIFR deficiency or overexpression does not affect hepatocyte proliferation ex vivo or in vitro.

a-e. Primary hepatocytes isolated from Lifrfl/fl and Lifrfl/fl;Alb-Cre mice were cultured for 3 hours, followed by treatment with 100 ng/mL of Hgf and/or 20 ng/mL of Egf for 48 hours. a-c. qPCR of mRNA of Lifr (a), cyclin D1 (b), and Pcna (c) in Hgf- and/or Egf-treated hepatocytes. n = 4, 4, 5, 5, 5, 5, 4, and 4 biological replicates. d, e. Immunofluorescence staining of Ki67 (d; overlay with DAPI staining) and percentage of Ki67-positive cells (e) in Hgf- and/or Egf-treated hepatocytes. Scale bars, 100 μm. n = 4 biological replicates. f-j. Primary hepatocytes isolated from C57BL/6J mice 10 days after injection with control adenovirus or LIFR-expressing adenovirus were cultured for 3 hours, followed by treatment with 100 ng/mL of Hgf and/or 20 ng/mL of Egf for 48 hours. f-h. qPCR of mRNA of Lifr (f), cyclin D1 (g), and Pcna (h) in Hgf- and/or Egf-treated hepatocytes. n = 5, 5, 4, 4, 4, 4, 5, and 5 biological replicates. i, j. Immunofluorescence staining of Ki67 (i; overlay with DAPI staining) and percentage of Ki67-positive cells (j) in Hgf- and/or Egf-treated hepatocytes. Scale bars, 100 μm. n = 5 biological replicates. k-m. qPCR of mRNA of Lifr (k), cyclin D1 (l), and Pcna (m) in Hgf- and/or Egf-treated hepatocytes isolated from C57BL/6J mice. The cells were infected with control adenovirus or LIFR-expressing adenovirus for 24 hours before Hgf and/or Egf treatment. n = 5, 5, 4, 4, 4, 4, 5, and 5 biological replicates. Statistical significance in a-c, e-h, and j-m was determined by a two-tailed unpaired t-test. Error bars are s.e.m.

Source data

Extended Data Fig. 4 Effects of LIFR on neutrophil recruitment after liver injury.

a, b. Quantification of liver-infiltrating immune cell populations in Lifrfl/fl and Lifrfl/fl;Alb-Cre mice 72 hours after PHx (a; n = 5 and 4 mice) or CCl4 treatment (b; n = 4 mice). NK: natural killer cells. KC: Kupffer cells. MoMa: monocyte-derived macrophages. PMN: polymorphonuclear neutrophils. DC: dendritic cells. c. Immunohistochemical staining of neutrophil elastase (NE) in the livers of Lifrfl/fl mice at 72 hours after PHx. Scale bars, 200 μm (top left) and 50 μm (top right and bottom). Representative results from one of three independent experiments are shown. d. Number of CD45+ cells per gram of liver in Lifrfl/fl and Lifrfl/fl;Alb-Cre mice at 72 hours after PHx. n = 4 and 5 mice. e-g. C57BL/6J mice received control or LIFR-expressing adenovirus 10 days before CCl4 treatment. Analyses were done at 48 hours after CCl4 treatment. e. Experimental design. f, g. Flow cytometry plots and percentage of neutrophils in liver (e) and blood (f) CD45+ cells from mice at 48 hours after CCl4 treatment. n = 6 and 5 mice. Statistical significance in a, b, d, f, and g was determined by a two-tailed unpaired t-test. Error bars are s.e.m.

Source data

Extended Data Fig. 5 The neutrophils from Lifrfl/fl;Alb-Cre mice have a lower ability to promote hepatocyte proliferation.

a. Immunofluorescence staining of neutrophil elastase (NE, green) and Hgf (red) on liver sections from Lifrfl/fl and Lifrfl/fl;Alb-Cre mice at 72 hours after PHx. Scale bars, 100 μm. Representative results from one of three independent experiments are shown. b. Immunofluorescence staining of Ki67 (overlay with DAPI staining) and percentage of Ki67-positive cells in primary mouse hepatocytes cultured for 48 hours with the conditioned medium of liver-infiltrating neutrophils purified from Lifrfl/fl and Lifrfl/fl;Alb-Cre mice at 72 hours after PHx. Scale bars, 100 μm. n = 5 biological replicates. c. Immunofluorescence staining of Ki67 (overlay with DAPI staining) and percentage of Ki67-positive cells in primary mouse hepatocytes cultured for 48 hours with the conditioned medium of blood neutrophils purified from Lifrfl/fl and Lifrfl/fl;Alb-Cre mice at 72 hours after PHx. Scale bars, 100 μm. n = 5 biological replicates. d. Immunoblotting of p-Met, Met, p-Erk, Erk, and Gapdh in the livers of Lifrfl/fl and Lifrfl/fl;Alb-Cre mice at 72 hours after PHx. Representative results from one of three independent experiments are shown. Statistical significance in b and c was determined by a two-tailed unpaired t-test. Error bars are s.e.m.

Source data

Extended Data Fig. 6 Knockdown of HGF in AdHL-60 cells.

a. Giemsa staining of HL-60 and all-trans retinoic acid (ATRA)-differentiated HL-60 (AdHL-60) cells. Scale bars, 50 μm. b. Flow cytometry plots of CD11b in HL-60 and AdHL-60 cells. c. Giemsa staining of control and HGF-knockdown HL-60 cells with or without ATRA-induced differentiation. Scale bars, 50 μm. d. Flow cytometry plots of CD11b in control and HGF-knockdown HL-60 cells with or without ATRA-induced differentiation. e. qPCR of HGF in control and HGF-knockdown AdHL-60 cells. n = 3 biological replicates. f. Flow cytometry plots and quantification of HGF in control and HGF-knockdown AdHL-60 cells. n = 3 biological replicates. Statistical significance in e and f was determined by a two-tailed unpaired t-test. Error bars are s.e.m.

Source data

Extended Data Fig. 7 LIFR accelerates CCl4-induced liver injury repair and regeneration in a neutrophil-dependent manner.

a-n. C57BL/6J mice received control or LIFR-expressing adenovirus 10 days before CCl4 treatment. Six hours after CCl4 treatment, the mice were treated with control IgG or anti-Ly6G. Analyses were done at 48 hours after CCl4 treatment. a. Experimental design. b, c. Flow cytometry plots (b) and percentage (c) of neutrophils in liver CD45+ cells. n = 5 mice. d, e. Flow cytometry plots (d) and percentage (e) of neutrophils in blood CD45+ cells. n = 5 mice. f, g. Flow cytometry plots (f) and percentage (g) of neutrophils in bone marrow (BM) CD45+ cells. n = 5 mice. h, i. Serum ALT (h) and AST (i) levels in control and LIFR-expressing adenovirus-infected C57BL/6J mice injected with control IgG or anti-Ly6G after CCl4 treatment. n = 5 mice. j, k. H&E staining (j) and percentage of necrotic areas (k). Scale bars, 300 μm. n = 5 mice. l, m. TUNEL staining (l) and the number of TUNEL-positive hepatocytes per high-power field (HPF; m). Scale bars, 100 μm. n = 5 mice. n, o. Immunohistochemical staining of Ki67 (n) and percentage of Ki67-positive hepatocytes (o). Scale bars, 50 μm. n = 5 mice. Statistical significance in c, e, g, h, i, k, m, and o was determined by a two-tailed unpaired t-test. Error bars are s.e.m.

Source data

Extended Data Fig. 8 Loss of hepatic Lifr impairs neutrophil recruitment, neutrophilic Hgf production, and liver regeneration in female mice.

a. Experimental design for panels b-i. All mice used were females. b. Liver-to-body weight ratio of Lifrfl/fl and Lifrfl/fl;Alb-Cre mice at 72 hours after PHx. n = 6 mice. c. Immunofluorescence staining of Ki67 and percentage of Ki67-positive hepatocytes in the livers of Lifrfl/fl and Lifrfl/fl;Alb-Cre mice at 72 hours after PHx. LPF: low-power field; HPF: high-power field. Scale bars, 100 μm (left) and 20 μm (right). n = 6 mice. d, e. Flow cytometry plots (d) and percentage (e) of neutrophils in liver, blood, and bone marrow (BM) CD45+ cells from Lifrfl/fl and Lifrfl/fl;Alb-Cre mice at 72 hours after PHx. n = 5 mice. f. Serum cholesterol levels in Lifrfl/fl and Lifrfl/fl;Alb-Cre mice at 72 hours after PHx. n = 5 mice. g. Serum Hgf levels in Lifrfl/fl and Lifrfl/fl;Alb-Cre mice at 72 hours after PHx. n = 6 mice. h. Flow cytometry plots and quantification of Hgf in liver neutrophils from Lifrfl/fl and Lifrfl/fl;Alb-Cre mice at 72 hours after PHx. n = 5 mice. i. Flow cytometry plots and quantification of Hgf in blood neutrophils from Lifrfl/fl and Lifrfl/fl;Alb-Cre mice at 72 hours after PHx. n = 5 mice. Statistical significance in b, c, and e-i was determined by a two-tailed unpaired t-test. Error bars are s.e.m.

Source data

Extended Data Fig. 9 Cxcl1 facilitates the recruitment of neutrophils to the liver after hepatectomy.

a. Immunofluorescence staining of neutrophil elastase (NE, red), Cxcr2 (green), and Hgf (cyan) in blood neutrophils from wild-type mice at 72 hours after PHx. Scale bars, 5 μm. Representative results from one of three independent experiments are shown. b, c. Schematic of the experimental design (b): at 40 hours after PHx, CD45.2 mice were treated with isotype IgG or anti-Cxcl1. At 62 hours after PHx, CD45.1 neutrophils were adoptively transferred to the antibody-treated mice. The liver infiltration of CD45.1 neutrophils was analyzed at 4 hours after adoptive transfer (c). n = 6 mice. Statistical significance in c was determined by a two-tailed unpaired t-test. Error bars are s.e.m.

Source data

Extended Data Fig. 10 Model for LIFR-mediated regulation of neutrophil recruitment and cholesterol-driven hepatocyte-neutrophil crosstalk during liver injury repair and regeneration.

Created with BioRender.com.

Supplementary information

Supplementary Information

Supplementary Figs. 1–3 and Supplementary Tables 2–5.

Reporting Summary

Supplementary Table 1

Normalized z-scores of lipids in plasma samples collected from control and Lifr conditional knockout mice at 72 h after partial hepatectomy.

Supplementary Data 1

Source data for Supplementary Figures.

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 and uncropped blots.

Source Data Fig. 8

Statistical source data and uncropped blots.

Source Data Extended Data Fig. 1

Statistical source data and uncropped blots.

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 and uncropped blots.

Source Data Extended Data Fig. 6

Statistical source data and uncropped and unprocessed images.

Source Data Extended Data Fig. 7

Statistical source data.

Source Data Extended Data Fig. 8

Statistical source data.

Source Data Extended Data Fig. 9

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

Deng, Y., Zhao, Z., Sheldon, M. et al. LIFR regulates cholesterol-driven bidirectional hepatocyte–neutrophil cross-talk to promote liver regeneration. Nat Metab (2024). https://doi.org/10.1038/s42255-024-01110-y

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1038/s42255-024-01110-y

  • Springer Nature Limited

Navigation