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:

EGFR has a tumour-promoting role in liver macrophages during hepatocellular carcinoma formation

Abstract

Hepatocellular carcinoma (HCC) is a frequent cancer with limited treatment options and poor prognosis. Tumorigenesis has been linked with macrophage-mediated chronic inflammation and diverse signalling pathways, including the epidermal growth factor receptor (EGFR) pathway. The precise role of EGFR in HCC is unknown, and EGFR inhibitors have shown disappointing clinical results. Here we discover that EGFR is expressed in liver macrophages in both human HCC and in a mouse HCC model. Mice lacking EGFR in macrophages show impaired hepatocarcinogenesis, whereas mice lacking EGFR in hepatocytes unexpectedly develop more HCC owing to increased hepatocyte damage and compensatory proliferation. Mechanistically, following interleukin-1 stimulation, EGFR is required in liver macrophages to transcriptionally induce interleukin-6, which triggers hepatocyte proliferation and HCC. Importantly, the presence of EGFR-positive liver macrophages in HCC patients is associated with poor survival. This study demonstrates a tumour-promoting mechanism for EGFR in non-tumour cells, which could lead to more effective precision medicine strategies.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Purchase on Springer Link

Instant access to full article PDF

Prices may be subject to local taxes which are calculated during checkout

Figure 1: HCC formation in mice lacking EGFR in hepatocytes or all liver cells.
Figure 2: EGFR expression in Kupffer cells/liver macrophages promotes HCC development.
Figure 3: EGFR expression is induced in activated Kupffer cells/liver macrophages under pathological conditions.
Figure 4: EGFR expression in Kupffer cells of HCC patients correlates with poor prognosis.
Figure 5: EGFR-dependent IL-6 production and release.

Similar content being viewed by others

References

  1. Jemal, A. et al. Global cancer statistics. CA Cancer J. Clin. 61, 69–90 (2011).

    Article  Google Scholar 

  2. Whittaker, S., Marais, R. & Zhu, A. X. The role of signaling pathways in the development and treatment of hepatocellular carcinoma. Oncogene 29, 4989–5005 (2010).

    Article  CAS  Google Scholar 

  3. Laurent-Puig, P. & Zucman-Rossi, J. Genetics of hepatocellular tumors. Oncogene 25, 3778–3786 (2006).

    Article  CAS  Google Scholar 

  4. EASL-EORTC clinical practice guidelines: management of hepatocellular carcinoma. J. Hepatol. 56, 908–943 (2012).

  5. Finkin, S. & Pikarsky, E. NF-κB in liver cancer: the plot thickens. Curr. Top. Microbiol. Immunol. 349, 185–196 (2011).

    CAS  PubMed  Google Scholar 

  6. Luedde, T. et al. Deletion of NEMO/IKKγ in liver parenchymal cells causes steatohepatitis and hepatocellular carcinoma. Cancer Cell 11, 119–132 (2007).

    Article  CAS  Google Scholar 

  7. Sakurai, T., Maeda, S., Chang, L. & Karin, M. Loss of hepatic NF-κB activity enhances chemical hepatocarcinogenesis through sustained c-Jun N-terminal kinase 1 activation. Proc. Natl Acad. Sci. USA 103, 10544–10551 (2006).

    Article  CAS  Google Scholar 

  8. Vainer, G. W., Pikarsky, E. & Ben-Neriah, Y. Contradictory functions of NF-κB in liver physiology and cancer. Cancer Lett. 267, 182–188 (2008).

    Article  CAS  Google Scholar 

  9. Coussens, L. M. & Werb, Z. Inflammation and cancer. Nature 420, 860–867 (2002).

    Article  CAS  Google Scholar 

  10. Grivennikov, S. I., Greten, F. R. & Karin, M. Immunity, inflammation, and cancer. Cell 140, 883–899 (2010).

    Article  CAS  Google Scholar 

  11. Sakurai, T. et al. Hepatocyte necrosis induced by oxidative stress and IL-1α release mediate carcinogen-induced compensatory proliferation and liver tumorigenesis. Cancer Cell 14, 156–165 (2008).

    Article  CAS  Google Scholar 

  12. Naugler, W. E. et al. Gender disparity in liver cancer due to sex differences in MyD88-dependent IL-6 production. Science 317, 121–124 (2007).

    Article  CAS  Google Scholar 

  13. Hui, L., Zatloukal, K., Scheuch, H., Stepniak, E. & Wagner, E. F. Proliferation of human HCC cells and chemically induced mouse liver cancers requires JNK1-dependent p21 downregulation. J. Clin. Invest. 118, 3943–3953 (2008).

    Article  CAS  Google Scholar 

  14. Li, H. P. et al. miR-451 inhibits cell proliferation in human hepatocellular carcinoma through direct suppression of IKK-β. Carcinogenesis 34, 2443–2451 (2013).

    Article  CAS  Google Scholar 

  15. Wang, S. N., Lee, K. T., Tsai, C. J., Chen, Y. J. & Yeh, Y. T. Phosphorylated p38 and JNK MAPK proteins in hepatocellular carcinoma. Eur. J. Clin. Invest. 42, 1295–1301 (2012).

    Article  CAS  Google Scholar 

  16. Das, M., Garlick, D. S., Greiner, D. L. & Davis, R. J. The role of JNK in the development of hepatocellular carcinoma. Genes Dev. 25, 634–645 (2011).

    Article  CAS  Google Scholar 

  17. Heinrichsdorff, J., Luedde, T., Perdiguero, E., Nebreda, A. R. & Pasparakis, M. p38α MAPK inhibits JNK activation and collaborates with IκB kinase 2 to prevent endotoxin-induced liver failure. EMBO Reports 9, 1048–1054 (2008).

    Article  CAS  Google Scholar 

  18. Hui, L. et al. p38α suppresses normal and cancer cell proliferation by antagonizing the JNK-c-Jun pathway. Nat. Genet. 39, 741–749 (2007).

    Article  CAS  Google Scholar 

  19. Maeda, S., Kamata, H., Luo, J. L., Leffert, H. & Karin, M. IKKβ couples hepatocyte death to cytokine-driven compensatory proliferation that promotes chemical hepatocarcinogenesis. Cell 121, 977–990 (2005).

    Article  CAS  Google Scholar 

  20. Buckley, A. F., Burgart, L. J., Sahai, V. & Kakar, S. Epidermal growth factor receptor expression and gene copy number in conventional hepatocellular carcinoma. Am. J. Clin. Pathol. 129, 245–251 (2008).

    Article  Google Scholar 

  21. Feitelson, M. A., Pan, J. & Lian, Z. Early molecular and genetic determinants of primary liver malignancy. Surg. Clin. North Am. 84, 339–354 (2004).

    Article  Google Scholar 

  22. Hopfner, M. et al. Targeting the epidermal growth factor receptor by gefitinib for treatment of hepatocellular carcinoma. J. Hepatol. 41, 1008–1016 (2004).

    Article  Google Scholar 

  23. Schiffer, E. et al. Gefitinib, an EGFR inhibitor, prevents hepatocellular carcinoma development in the rat liver with cirrhosis. Hepatology 41, 307–314 (2005).

    Article  CAS  Google Scholar 

  24. Mallarkey, G. & Coombes, R. C. Targeted therapies in medical oncology: successes, failures and next steps. Ther. Adv. Med. Oncol. 5, 5–16 (2013).

    Article  Google Scholar 

  25. Natarajan, A., Wagner, B. & Sibilia, M. The EGF receptor is required for efficient liver regeneration. Proc. Natl Acad. Sci. USA 104, 17081–17086 (2007).

    Article  CAS  Google Scholar 

  26. Eferl, R. et al. Liver tumor development. c-Jun antagonizes the proapoptotic activity of p53. Cell 112, 181–192 (2003).

    Article  CAS  Google Scholar 

  27. He, G. & Karin, M. NF-κB and STAT3 - key players in liver inflammation and cancer. Cell Res. 21, 159–168 (2011).

    Article  CAS  Google Scholar 

  28. Sell, S. Cellular origin of hepatocellular carcinomas. Semin. Cell Dev. Biol. 13, 419–424 (2002).

    Article  Google Scholar 

  29. Degryse, B. et al. The high mobility group (HMG) boxes of the nuclear protein HMG1 induce chemotaxis and cytoskeleton reorganization in rat smooth muscle cells. J. Cell Biol. 152, 1197–1206 (2001).

    Article  CAS  Google Scholar 

  30. Wang, K. et al. Overexpression of aspartyl-(asparaginyl)-β-hydroxylase in hepatocellular carcinoma is associated with worse surgical outcome. Hepatology 52, 164–173 (2010).

    Article  CAS  Google Scholar 

  31. Blobel, C. P. ADAMs: key components in EGFR signalling and development. Nat. Rev. Mol. Cell Biol. 6, 32–43 (2005).

    Article  CAS  Google Scholar 

  32. McElroy, S. J. et al. Transactivation of EGFR by LPS induces COX-2 expression in enterocytes. PloS ONE 7, e38373 (2012).

    Article  CAS  Google Scholar 

  33. Clausen, B. E., Burkhardt, C., Reith, W., Renkawitz, R. & Forster, I. Conditional gene targeting in macrophages and granulocytes using LysMcre mice. Transgenic Res. 8, 265–277 (1999).

    Article  CAS  Google Scholar 

  34. Lichtenberger, B. M. et al. Autocrine VEGF signaling synergizes with EGFR in tumor cells to promote epithelial cancer development. Cell 140, 268–279 (2010).

    Article  CAS  Google Scholar 

  35. Sibilia, M. et al. The EGF receptor provides an essential survival signal for SOS-dependent skin tumor development. Cell 102, 211–220 (2000).

    Article  CAS  Google Scholar 

  36. Drobits, B. et al. Imiquimod clears tumors in mice independent of adaptive immunity by converting pDCs into tumor-killing effector cells. J. Clin. Invest. 122, 575–585 (2012).

    Article  CAS  Google Scholar 

  37. Smedsrod, B. & Pertoft, H. Preparation of pure hepatocytes and reticuloendothelial cells in high yield from a single rat liver by means of Percoll centrifugation and selective adherence. J. Leukoc. Biol. 38, 213–230 (1985).

    Article  CAS  Google Scholar 

  38. Sieghart, W. et al. Osteopontin expression predicts overall survival after liver transplantation for hepatocellular carcinoma in patients beyond the Milan criteria. J. Hepatol. 54, 89–97 (2011).

    Article  CAS  Google Scholar 

  39. Mazzaferro, V. et al. Liver transplantation for the treatment of small hepatocellular carcinomas in patients with cirrhosis. N. Engl. J. Med. 334, 693–699 (1996).

    Article  CAS  Google Scholar 

  40. Wang, K. et al. Overexpression of aspartyl-(asparaginyl)-β-hydroxylase in hepatocellular carcinoma is associated with worse surgical outcome. Hepatology 52, 164–173 (2010).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

We are grateful to L. Bakiri, D. P. Barlow, R. Eferl, M. Oft and E. F. Wagner for critical reading of the manuscript. We thank T. Baykuscheva-Gentscheva and S. Bardakji for genotyping and F. Hucke and G. Heinze for statistical support. This work was supported by the EC programme LSHC-CT-2006-037731 (Growthstop), the Austrian Science Fund (FWF) grants SFB F3518-B20 (to M.S.), F3517 (to M.T.), FWF-DK W1212 and P25925 and the Austrian Federal Government’s GEN-AU program ‘Austromouse’ (GZ 200.147/1-VI/1a/2006 and 820966). H.W. acknowledges funding by the National Natural Science Foundation of China, 30921006.

Author information

Authors and Affiliations

Authors

Contributions

A.N. designed, carried out and analysed in vivo tumour experiments with the EGFRΔhep, EGFRΔMx and EGFRΔMx mice. H.L. designed, carried out and analysed in vitro experiments and some western blot analysis and carried out in vivo tumour experiments with EGFRΔhep/Δmac and EGFRΔmac mice. K.K. carried out in vivo analyses with EGFRΔhep/Δmac and EGFRΔmac mice and in vitro analyses with Kupffer cells including western blot analysis. N.A. helped with histology, immunohistochemistry and immunofluorescence. S.K.W. helped with qRT-PCRs, M.H. helped with histology, mouse colony and animal experiments. L.L. and L.C. carried out stainings on all human samples (Chinese and European cohorts) and analysed the Chinese cohort with the supervision of H.W. W.S. analysed the European cohort together with M.T. and M.P-R. R.Z. and M.S. wrote the manuscript with input from H.L., A.N., N.A., W.S., M.T., M.P-R. and H.W. and with major contributions during the revision phase from K.K. M.S. conceived and supervised the whole project.

Corresponding author

Correspondence to Maria Sibilia.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Integrated supplementary information

Supplementary Figure 1 HCC induction and liver damage in mice and hepatocytes.

(a) Tumour development was initiated in male mice of the indicated genotypes by DEN injection at 4 weeks of age (black arrow). At 8 weeks of age tumours were promoted by a diet complemented with phenobarbital (PB) until mice were sacrificed. Top: EGFRΔhep mice (EGFRf/f; Alfp-Cre). Middle: EGFRΔMx mice (EGFRf/f; Mx-Cre) that received 3 pIpC injections with 2 days intervals at 7 weeks of age to delete EGFR in the liver (grey arrow). Bottom: EGFRΔMx mice (EGFRf/f; Mx-Cre), that received pIpC injections on day 9, 11, and 13 (grey arrow) after birth. (b) Southern Blot analysis showing EGFR deletion in EGFRΔMx (left) and EGFRΔhep (right, non-recombined EGFR allele from non-parenchymal cells) livers (f: floxed EGFR allele (6 kb), Δ: Cre-deleted EGFR allele (3.9 kb). (c) Ki67-positive (left, EGFRf/f, n = 45 (six mice); EGFRΔMx, n = 48 (7 mice)) and TUNEL-positive cells (right, EGFRf/f, n = 56 (six mice); EGFRΔMx: n = 55 (six mice)) in adjacent non-tumour tissue. n = HPF. (d) Representative H&E staining of livers 0, 36, 48, and 72 h after DEN intoxication in vivo. Black arrows indicate necrotic areas. Scale bar, 500 μm. (e,f) Alanine transaminase (ALT, 0 h and 24 h: n = 5 mice per genotype, 48 h: EGFRf/f and EGFRΔMx: n = 4, EGFRΔhepn = 5 mice) and Aspartate transaminase (AST, 0 h: EGFRf/f: n = 3, EGFRΔhep: n = 4, EGFRΔMx: n = 4, 24 h: EGFRf/f: n = 3, EGFRΔhep, n = 3, EGFRΔMx: n = 4, 48 h: EGFRf/f: n = 4, EGFRΔhep, n = 4, EGFRΔMx: n = 3 mice) measured in serum 0, 24, and 48 h after DEN intoxication. (g) Representative active caspase-3 staining (Alexa 488, green) and nuclei (DAPI, blue) of EGFRf/f and EGFRΔhep liver sections 24 and 96 h after DEN intoxication, showing increased apoptosis in EGFR-deficient livers after 96 h. Scale bar indicates 500 μm. (h) HMGB1 staining of cultured EGFRf/f and EGFRΔMx hepatocytes 12 h after DEN treatment in vitro. Nuclei (DAPI, blue), actin (Phalloidin, red), HMGB1 (Alexa 488, green). Scale bar indicates 100 μm. (i) Active caspase-3 staining of cultured hepatocytes of EGFRf/f and EGFRΔMx mice incubated with TNFα/CHX for 12 h. Nuclei (DAPI, blue), actin (Phalloidin, red), active caspase-3 (Alexa 488, green). Scale bar indicates 50 μm. (c) Data represent mean ± s.e.m. (e,f) Data represent mean ± s.d. Student’s t-test for independent samples and unequal variances was used to assess statistical significance (P < 0.05,P < 0.01,P < 0.001). Original data are provided in Supplementary Table 1.

Supplementary Figure 2 EGFR deletion in EGFRΔMx mice and cytokine production on DEN injection.

(a) ELISA showing IL-1β in the supernatant of primary hepatocyte cultures 4 h after incubation with increasing amounts of DEN in vitro. (Primary hepatocyte isolates of EGFRf/f (n = 3) and EGFRΔhep (n = 3)). (b) Release of IL-1β and IL-1α to the supernatant of cultured primary hepatocytes of EGFRf/f, EGFRΔhep, and EGFRΔMx mice after incubation with TNFα quantified by ELISA. n.d. = not detectable. Result of two pooled independent experiments is shown. For each experiment hepatocytes isolated from 2 livers per genotype were pooled and analysed as 4 technical replicates (primary hepatocytes of n = 4 mice were analysed in total for each genotype). (c) Western Blot analysis of EGFR in livers of EGFRf/f and EGFRΔMx mice. (d) Representative livers of EGFRf/f and EGFRΔMx mice 46 weeks after tumour initiation. Scale bar indicates 1 cm. (e) Tumour mass (left, EGFRf/f (n = 10) and EGFRΔMx (n = 9)), area (middle, EGFRf/f (n = 7) and EGFRΔMx (n = 9)), and number (right, EGFRf/f (n = 7) and EGFRΔMx (n = 9)). Two pooled independent experiments. Data (a,b) represent mean ± s.d. Data (e) represent mean ± s.e.m. Student’s t-test for independent samples and unequal variances was used to assess statistical significance (P < 0.05, P < 0.01, P < 0.001). Original data are provided in Supplementary Table 1.

Supplementary Figure 3 EGFR expression in tumour cells and Kupffer cells of human HCCs.

(a) Representative Immunohistochemistry showing EGFR expression in tumour cells/hepatocytes of HCC. Scoring (0, +, ++, +++) was performed according to the scale described below resulting in the generation of Table 1a. Scale bar indicates 50 μm. (be) OS (b,d) and DFS (c,e) of HCC patients of the Chinese (b,c: 129 patients (n = 70 negative for EGFR; n = 59 positive for EGFR)) and European cohort (d,e: 108 patients (n = 69 negative for EGFR; n = 39 positive for EGFR)) with or without EGFR expression in hepatocytes. (f) Representative immunohistochemistry showing EGFR and CD68 staining (0, +, ++) in liver macrophages of human HCC. Scoring (0, +, ++, +++) was performed according to the scale described below resulting in the generation of Table 1b. Scale bar indicates 50 μm. (g) Representative immunofluorescent EGFR and CD68 co-staining in fresh frozen human HCC tissue (n = 12). Nuclei (DAPI, blue), CD68 (Alexa 488, green) and EGFR (Alexa 594, red), merged (bottom right). White arrows indicate double positive cells. Scale bar indicates 50 μm. Scoring system: 0 = negative staining (0%–10% positive), 1 =weak signal (10%–20% positive), 2 = intermediate signal (20%–50% positive) and 3 = strong signal (>50% positive) as previously described30. Log-rank test was used to assess statistical significance.

Supplementary Figure 4 IL-6 production by Kupffer cells after various stimuli and inhibitor treatments.

(a,b) ELISA quantifying IL-1β-induced IL-6 secretion by isolated Kupffer cells after preincubation with increasing amounts of the EGFR inhibitors Cetuximab (a) or BIBW2992 (Afatinib) (b). (n = 2 primary Kupffer cell isolates). (c) IL-6 secretion by isolated Kupffer cells following stimulation with polyIC (20 μg ml−1), imiquimod (12 μg ml−1) and LPS (10 ng ml−1). - = unstimulated. (n = 2 primary Kupffer cell isolates). (d) ELISA quantifying IL-1β- or EGF-induced IL-17A, IL-22 and IL-23 secretion by isolated Kupffer cells (n = 2 primary Kupffer cell isolates). (e,f) Representative Western Blot showing activation of the indicated proteins after 15 min stimulation with IL-1β (e) or EGF (f) in the presence of the respective inhibitors. Note: Each lane contains proteins isolated from pooling Kupffer cells of 3 different livers. Because the amount of proteins obtained from Kupffer cells from 3 pooled livers was not sufficient to perform Western blot analysis for all indicated proteins and treatments, 2 different isolates and Western blots for each series of treatment (EGF+ inhibitors and IL-1β+ inhibitors) had to be performed. (e) Blot 1: IL-1β stimulated EGFRf/f and EGFRΔMx+ inhibitors and expression of EGFR and JNK. Blot 2: IL-1β stimulated EGFRf/f and EGFRΔMx+ inhibitors and expression of p38, IKK, NF-kB, Stat3. (f) Blot 1: EGF stimulated EGFRf/f and EGFRΔMx+ inhibitors and expression of EGFR and JNK. Blot 2: EGF stimulated EGFRf/f and EGFRΔMx+ inhibitors and expression of p38, IKK, NF-kB, Stat3. The results were confirmed in a second set of isolates and Westerns. Original data are provided in Supplementary Table 1.

Supplementary Figure 5 Model of EGFR signalling in hepatocytes and Kupffer cells during HCC formation.

EGFR signalling is hepatoprotective during DEN-induced liver damage as in the absence of EGFR, hepatocytes undergo more necrosis and apoptosis thus leading to increased IL-1β production and release. IL-1β stimulation of Kupffer cells in turn leads to release of IL-6, which is required for compensatory proliferation and repair of damaged hepatocytes. IL-1β-induced IL-6 production occurs in a bimodal way involving the activation of the IL-1R/MyD88 pathway to first induce EGFR ligands and ADAM17 expression with subsequent EGFR transactivation required for IL-6 production via JNK, p38 and IKK.

Supplementary Figure 6 Uncropped images of western blots and PCR analysis.

The uncropped films and gel photographs (Fig. 3c) showing the Western blots and PCR analysis (Fig. 3c) displayed in the main figures. Boxed areas indicate the cropped regions displayed in the respective figures.

Supplementary information

Supplementary Information

Supplementary Information (PDF 1100 kb)

Supplementary Tables 1–6

Supplementary Information (XLSX 120 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Lanaya, H., Natarajan, A., Komposch, K. et al. EGFR has a tumour-promoting role in liver macrophages during hepatocellular carcinoma formation. Nat Cell Biol 16, 972–981 (2014). https://doi.org/10.1038/ncb3031

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/ncb3031

This article is cited by

Search

Quick links

Nature Briefing: Cancer

Sign up for the Nature Briefing: Cancer newsletter — what matters in cancer research, free to your inbox weekly.

Get what matters in cancer research, free to your inbox weekly. Sign up for Nature Briefing: Cancer