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:

c-Rel orchestrates energy-dependent epithelial and macrophage reprogramming in fibrosis

An Author Correction to this article was published on 10 December 2020

This article has been updated

Abstract

Fibrosis is a common pathological feature of chronic disease. Deletion of the NF-κB subunit c-Rel limits fibrosis in multiple organs, although the mechanistic nature of this protection is unresolved. Using cell-specific gene-targeting manipulations in mice undergoing liver damage, we elucidate a critical role for c-Rel in controlling metabolic changes required for inflammatory and fibrogenic activities of hepatocytes and macrophages and identify Pfkfb3 as the key downstream metabolic mediator of this response. Independent deletions of Rel in hepatocytes or macrophages suppressed liver fibrosis induced by carbon tetrachloride, while combined deletion had an additive anti-fibrogenic effect. In transforming growth factor-β1-induced hepatocytes, c-Rel regulates expression of a pro-fibrogenic secretome comprising inflammatory molecules and connective tissue growth factor, the latter promoting collagen secretion from HMs. Macrophages lacking c-Rel fail to polarize to M1 or M2 states, explaining reduced fibrosis in RelΔLysM mice. Pharmacological inhibition of c-Rel attenuated multi-organ fibrosis in both murine and human fibrosis. In conclusion, activation of c-Rel/Pfkfb3 in damaged tissue instigates a paracrine signalling network among epithelial, myeloid and mesenchymal cells to stimulate fibrogenesis. Targeting the c-Rel–Pfkfb3 axis has potential for therapeutic applications in fibrotic disease.

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: c-Rel is a feature of chronic liver, kidney and lung disease in humans and epithelial c-Rel signalling regulates hepatic fibrogenesis and regeneration in mice.
Fig. 2: c-Rel signalling regulates epithelial inflammatory responses via regulation of Pfkfb3.
Fig. 3: c-Rel signalling in macrophages is pro-fibrogenic and regulates macrophage plasticity.
Fig. 4: c-Rel regulates pro-fibrogenic epithelial–macrophage cross-talk accelerates fibroblast activation.
Fig. 5: Epithelial- or macrophage-specific deletion of c-Rel limits renal and pulmonary fibrosis.
Fig. 6: Epithelial and macrophage c-Rel signalling synergistically promote hepatic fibrosis but antagonistically regulate hepatic regeneration in mice.
Fig. 7: Pharmacological inhibition of c-Rel limits fibrogenesis in murine models of liver, kidney and lung injury.
Fig. 8: Pharmacological inhibition of c-Rel limits fibrogenesis in human precision-cut liver slices.

Similar content being viewed by others

Data availability

Mass spectrometric raw data are available through the PRIDE repository (https://www.ebi.ac.uk/pride/archive/) and have been assigned the identifier PXD017320. Source data are provided with this paper.

Change history

  • 10 December 2020

    A Correction to this paper has been published: https://doi.org/10.1038/s42255-020-00326-y.

References

  1. Wynn, T. A. & Ramalingam, T. R. Mechanisms of fibrosis: therapeutic translation for fibrotic disease. Nat. Med. 18, 1028–1040 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Rockey, D. C., Bell, P. D. & Hill, J. A. Fibrosis—a common pathway to organ injury and failure. N. Engl. J. Med. 373, 95–96 (2015).

    Article  Google Scholar 

  3. Bataller, R. & Brenner, D. A. Liver fibrosis. J. Clin. Invest. 115, 209–218 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Friedman, S. L. Liver fibrosis—from bench to bedside. J. Hepatol. 38, 38–53 (2003).

    Article  Google Scholar 

  5. Cox, T. R. & Erler, J. T. Molecular pathways: connecting fibrosis and solid tumor metastasis. Clin. Cancer Res. 20, 3637–3643 (2014).

    Article  CAS  PubMed  Google Scholar 

  6. Cernaro, V. et al. Fibrosis, regeneration and cancer: what is the link? Nephrol. Dial. Transplant. 27, 21–27 (2012).

    Article  PubMed  Google Scholar 

  7. Rybinski, B., Franco-Barraza, J. & Cukierman, E. The wound healing, chronic fibrosis and cancer progression triad. Physiol. Genomics 46, 223–244 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Klingler, W., Jurkat-Rott, K., Lehmann-Horn, F. & Schleip, R. The role of fibrosis in Duchenne muscular dystrophy. Acta Myol. 31, 184–195 (2012) http://www.ncbi.nlm.nih.gov/pubmed/23620650 http://www.ncbi.nlm.nih.gov/pubmed/23620650.

    CAS  PubMed  PubMed Central  Google Scholar 

  9. Torres, V. E. & Leof, E. B. Fibrosis, regeneration, and aging: playing chess with evolution. J. Am. Soc. Nephrol. 22, 1393–1396 (2011).

    Article  CAS  PubMed  Google Scholar 

  10. Hecker, L. et al. Reversal of persistent fibrosis in aging by targeting Nox4–Nrf2 redox imbalance. Sci. Transl. Med. 6, 231ra47 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  11. Mehal, W. Z., Iredale, J. & Friedman, S. L. Scraping fibrosis: expressway to the core of fibrosis. Nat. Med. 17, 552–553 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Wynn, T. Cellular and molecular mechanisms of fibrosis. J. Pathol. 214, 199–210 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Koyama, Y. & Brenner, D. A. Liver inflammation and fibrosis. J. Clin. Invest. 127, 55–64 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  14. Hayden, M. S. & Ghosh, S. NF-κB, the first quarter-century: remarkable progress and outstanding questions. Genes Dev. 26, 203–234 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Oeckinghaus, A. & Ghosh, S. The NF-κB family of transcription factors and its regulation. Cold Spring Harb. Perspect. Biol. 1, a000034 (2009).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  16. Lawrence, T. The nuclear factor NF-κB pathway in inflammation. Cold Spring Harb. Perspect. Biol. 1, a001651 (2009).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  17. Tak, P. P. & Firestein, G. S. NF-κB: a key role in inflammatory diseases. J. Clin. Invest. 107, 7–11 (2001).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Zhang, Q., Lenardo, M. J. & Baltimore, D. 30 years of NF-κB: a blossoming of relevance to human pathobiology. Cell 168, 37–57 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Luedde, T. & Schwabe, R. F. NF-κB in the liver—linking injury, fibrosis and hepatocellular carcinoma. Nat. Rev. Gastroenterol. Hepatol. 8, 108–118 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Perkins, N. D. & Gilmore, T. D. Good cop, bad cop: the different faces of NF-κB. Cell Death Differ. 13, 759–772 (2006).

    Article  CAS  PubMed  Google Scholar 

  21. Piva, R., Belardo, G. & Santoro, M. G. NF-κB: a stress-regulated switch for cell survival. Antioxid. Redox Signal. 8, 478–486 (2006).

    Article  CAS  PubMed  Google Scholar 

  22. Wong, D. et al. Extensive characterization of NF-κB binding uncovers non-canonical motifs and advances the interpretation of genetic functional traits. Genome Biol. 12, R70 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Geisler, F., Algül, H., Paxian, S. & Schmid, R. M. Genetic inactivation of RelA/p65 sensitizes adult mouse hepatocytes to TNF-induced apoptosis in vivo and in vitro. Gastroenterology 132, 2489–2503 (2007).

    Article  CAS  PubMed  Google Scholar 

  24. Rosenfeld, M. E., Prichard, L., Shiojiri, N. & Fausto, N. Prevention of hepatic apoptosis and embryonic lethality in RelA/TNFR-1 double knockout mice. Am. J. Pathol. 156, 997–1007 (2000).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Beg, A. A., Sha, W. C., Bronson, R. T., Ghosh, S. & Baltimore, D. Embryonic lethality and liver degeneration in mice lacking the RelA component of NF-κB. Nature 376, 167–170 (1995).

    Article  CAS  PubMed  Google Scholar 

  26. Lenardo, M. J. & Baltimore, D. NF-κB: a pleiotropic mediator of inducible and tissue-specific gene control. Cell 58, 227–229 (1989).

    Article  CAS  PubMed  Google Scholar 

  27. Fullard, N., Wilson, C. L. & Oakley, F. Roles of c-Rel signalling in inflammation and disease. Int. J. Biochem. Cell Biol. 44, 851–860 (2012).

    Article  CAS  PubMed  Google Scholar 

  28. Neo, W. H., Lim, J. F., Grumont, R., Gerondakis, S. & Su, I. c-Rel regulates Ezh2 expression in activated lymphocytes and malignant lymphoid cells. J. Biol. Chem. 289, 31693–31707 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Zeybel, M. et al. A proof-of-concept for epigenetic therapy of tissue fibrosis: inhibition of liver fibrosis progression by 3-deazaneplanocin A. Mol. Ther. 25, 218–231 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Fullard, N. et al. The c-Rel subunit of NF-κB regulates epidermal homeostasis and promotes skin fibrosis in mice. Am. J. Pathol. 182, 2109–2120 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Gaspar-Pereira, S. et al. The NF-κB Subunit c-Rel stimulates cardiac hypertrophy and fibrosis. Am. J. Pathol. 180, 929–939 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Luli, S. et al. A new fluorescence-based optical imaging method to non-invasively monitor hepatic myofibroblasts in vivo. J. Hepatol. 65, 75–83 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Hunter, J. E., Leslie, J. & Perkins, N. D. C-Rel and its many roles in cancer: an old story with new twists. Br. J. Cancer. 114, 1–6 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Schwabe, R. F., Tabas, I. & Pajvani, U. B. Mechanisms of fibrosis development in nonalcoholic steatohepatitis. Gastroenterology 158, 1913–1928 (2020).

    Article  CAS  PubMed  Google Scholar 

  35. Swamy, M., Jamora, C., Havran, W. & Hayday, A. Epithelial decision makers: in search of the ‘epimmunome’. Nat. Immunol. 11, 656–665 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Duffield, J. S. et al. Selective depletion of macrophages reveals distinct, opposing roles during liver injury and repair. J. Clin. Invest. 115, 56–65 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Seki, E. et al. CCR2 promotes hepatic fibrosis in mice. Hepatology 50, 185–197 (2009).

    Article  CAS  PubMed  Google Scholar 

  38. Wynn, T. A. & Vannella, K. M. Macrophages in tissue repair, regeneration and fibrosis. Immunity 44, 450–462 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Garcia-Lazaro, J. F. et al. Hepatic over-expression of TGF-β1 promotes LPS-induced inflammatory cytokine secretion by liver cells and endotoxemic shock. Immunol. Lett. 101, 217–222 (2005).

    Article  CAS  PubMed  Google Scholar 

  40. Yang, L. et al. Transforming growth factor-β signaling in hepatocytes promotes hepatic fibrosis and carcinogenesis in mice with hepatocyte-specific deletion of TAK1. Gastroenterology 144, 1042–1054 (2013).

    Article  CAS  PubMed  Google Scholar 

  41. Bird, T. G. et al. TGF-β inhibition restores a regenerative response in acute liver injury by suppressing paracrine senescence. Sci. Transl. Med. 10, eaan1230 (2018).

  42. Niu L., et al. Involvement of TGF-β1/Smad3 signaling in carbon tetrachloride-induced acute liver injury in mice. PLoS ONE 11, e0156090 (2016).

  43. Travis, M. A. & Sheppard, D. TGF-β activation and function in immunity. Annu Rev. Immunol. 32, 51–82 (2014).

    Article  CAS  PubMed  Google Scholar 

  44. Grgurevic, L. et al. Systemic inhibition of BMP1-3 decreases progression of CCl4-induced liver fibrosis in rats. Growth Factors 35, 201–215 (2017).

    Article  CAS  PubMed  Google Scholar 

  45. Lipson, K. E., Wong, C., Teng, Y. & Spong, S. CTGF is a central mediator of tissue remodeling and fibrosis and its inhibition can reverse the process of fibrosis. Fibrogenesis Tissue Repair 5, S24 (2012).

    Article  PubMed  PubMed Central  Google Scholar 

  46. Fox, C. et al. Inhibition of lysosomal protease cathepsin D reduces renal fibrosis in murine chronic kidney disease. Sci Rep. 6, 20101 (2016).

  47. Moles, A., Tarrats, N., Fernández-Checa, J. C. & Marí, M. Cathepsins B and D drive hepatic stellate cell proliferation and promote their fibrogenic potential. Hepatology 49, 1297–1307 (2009).

    Article  CAS  PubMed  Google Scholar 

  48. Ghosh, A. K. & Vaughan, D. E. PAI-1 in tissue fibrosis. J. Cell. Physiol. 227, 493–507 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Kodama, T. et al. Increases in p53 expression induce CTGF synthesis by mouse and human hepatocytes and result in liver fibrosis in mice. J. Clin. Invest. 121, 3343–3356 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Mathieu, J. & Ruohola-Baker, H. Metabolic remodeling during the loss and acquisition of pluripotency. Development 144, 541–551 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Sciacovelli, M. & Frezza, C. Metabolic reprogramming and epithelial-to-mesenchymal transition in cancer. FEBS J. 284, 3132–3144 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Nieto, M. A., Huang, R. Y.-J., Jackson, R. A. & Thiery, J. P. EMT: 2016. Cell 166, 21–45 (2016).

    Article  CAS  PubMed  Google Scholar 

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

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  54. Taura, K. et al. Hepatocytes do not undergo epithelial–mesenchymal transition in liver fibrosis in mice. Hepatology 51, 1027–1036 (2010).

    Article  PubMed  Google Scholar 

  55. Humphreys, B. D. et al. Fate tracing reveals the pericyte and not epithelial origin of myofibroblasts in kidney fibrosis. Am. J. Pathol. 176, 85–97 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Grande, M. T. et al. Snail1-induced partial epithelial-to-mesenchymal transition drives renal fibrosis in mice and can be targeted to reverse established disease. Nat. Med. 21, 989–997 (2015).

    Article  CAS  PubMed  Google Scholar 

  57. Rowe, R. G. et al. Hepatocyte-derived Snail1 propagates liver fibrosis progression. Mol. Cell. Biol. 31, 2392–2403 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Hee Kim, N. et al. Snail reprograms glucose metabolism by repressing phosphofructokinase PFKP allowing cancer cell survival under metabolic stress. Nat. Commun. 8, 14374 (2017).

  59. Mills, E. L. & O’Neill, L. A. Reprogramming mitochondrial metabolism in macrophages as an anti-inflammatory signal. Eur. J. Immunol. 46, 13–21 (2016).

    Article  CAS  PubMed  Google Scholar 

  60. Gieling, R. G. et al. The c-Rel subunit of NF-κB regulates murine liver inflammation, wound healing and hepatocyte proliferation. Hepatology 51, 922–931 (2010).

    Article  CAS  PubMed  Google Scholar 

  61. Shono, Y. et al. A small-molecule c-Rel inhibitor reduces alloactivation of T cells without compromising antitumor activity. Cancer Discov. 4, 578–591 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. Paish, H. L. et al. A bioreactor technology for modelling fibrosis in human and rodent precision-cut liver slices. Hepatology 70, 1377–1391 (2019).

  63. Nielsen, M. J. et al. Plasma Pro-C3 (N-terminal type III collagen propeptide) predicts fibrosis progression in patients with chronic hepatitis C. Liver Int. 35, 429–437 (2015).

    Article  CAS  PubMed  Google Scholar 

  64. Krawczyk, C. M. et al. Toll-like receptor-induced changes in glycolytic metabolism regulate dendritic cell activation. Blood 115, 4742–4749 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  65. Lees, J. G., Gardner, D. K. & Harvey, A. J. Mitochondrial and glycolytic remodeling during nascent neural differentiation of human pluripotent stem cells. Development 145, dev168997 (2018).

  66. Peng, M. et al. Aerobic glycolysis promotes T helper 1 cell differentiation through an epigenetic mechanism. Science 354, 481–484 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  67. Wei, Q. et al. Glycolysis inhibitors suppress renal interstitial fibrosis via divergent effects on fibroblasts and tubular cells. Am. J. Physiol. Renal Physiol. 316, F1162–F1172 (2018).

  68. Ding, H. et al. Inhibiting aerobic glycolysis suppresses renal interstitial fibroblast activation and renal fibrosis. Am. J. Physiol. Renal Physiol. 313, F561–F575 (2017).

    Article  CAS  PubMed  Google Scholar 

  69. Xie, N. et al. Glycolytic reprogramming in myofibroblast differentiation and lung fibrosis. Am. J. Respir. Crit. Care Med. 92, 1462–1474 (2015).

    Article  CAS  Google Scholar 

  70. 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  CAS  Google Scholar 

  71. Chang, N. et al. Single-cell transcriptomes reveal characteristic features of mouse hepatocytes with liver cholestatic injury. Cells. 8, 1069 (2019).

  72. Huang, G. & Brigstock, D. R. Regulation of hepatic stellate cells by connective tissue growth factor. Front. Biosci. 17, 2495–2507 (2012).

  73. Paradis, V. et al. Effects and regulation of connective tissue growth factor on hepatic stellate cells. Lab Invest. 82, 767–774 (2002).

    Article  CAS  PubMed  Google Scholar 

  74. Gressner, O. A., Lahme, B., Demirci, I., Gressner, A. M. & Weiskirchen, R. Differential effects of TGF-β on connective tissue growth factor (CTGF/CCN2) expression in hepatic stellate cells and hepatocytes. J. Hepatol. 47, 699–710 (2007).

    Article  CAS  PubMed  Google Scholar 

  75. Friedman, S. L. Hepatic stellate cells: protean, multifunctional and enigmatic cells of the liver. Physiol. Rev. 88, 125–172 (2008).

    Article  CAS  PubMed  Google Scholar 

  76. Gressner, O. A. et al. Intracrine signalling of activin A in hepatocytes upregulates connective tissue growth factor (CTGF/CCN2) expression. Liver Int. 28, 1207–1216 (2008).

    Article  CAS  PubMed  Google Scholar 

  77. Fearn, A. et al. The NF-κB1 is a key regulator of acute but not chronic renal injury. Cell Death Dis. 8, e2883 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  78. Wang, F. et al. NF-κB inhibition alleviates carbon tetrachloride-induced liver fibrosis via suppression of activated hepatic stellate cells. Exp. Ther. Med. 8, 95–99 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  79. Chan, L. K. et al. Epithelial NEMO/IKK-γ limits fibrosis and promotes regeneration during pancreatitis. Gut 66, 1995–2007 (2017).

    Article  CAS  PubMed  Google Scholar 

  80. Karin, M., Yamamoto, Y. & Wang, Q. M. The IKK NF-κB system: a treasure trove for drug development. Nat. Rev. Drug Discov. 3, 17–26 (2004).

    Article  CAS  PubMed  Google Scholar 

  81. Bennett, J. et al. NF-κB in the crosshairs: rethinking an old riddle. Int. J. Biochem. Cell Biol. 95, 108–112 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  82. Oakley, F. et al. Inhibition of inhibitor of κB kinases stimulates hepatic stellate cell apoptosis and accelerated recovery from rat liver fibrosis. Gastroenterology 128, 108–120 (2005).

  83. Oakley, F. et al. Angiotensin II activates I κB kinase phosphorylation of RelA at Ser 536 to promote myofibroblast survival and liver fibrosis. Gastroenterology 136, 2334–2344 (2009).

    Article  CAS  PubMed  Google Scholar 

  84. Chen, L.-W. et al. The two faces of IKK and NF-κB inhibition: prevention of systemic inflammation but increased local injury following intestinal ischemia–reperfusion. Nat. Med. 9, 575–581 (2003).

    Article  CAS  PubMed  Google Scholar 

  85. Li, Z.-W. et al. The IKKβ subunit of IκB Kinase (IKK) is essential for nuclear factor κB activation and prevention of apoptosis. J. Exp. Med. 189, 1839–1845 (1999).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  86. Li, Q., Van Antwerp, D., Mercurio, F., Lee, K. F. & Verma, I. M. Severe liver degeneration in mice lacking the IκB kinase 2 gene. Science 284, 321–325 (1999).

  87. Li, Q. & Verma, I. M. NF-κB regulation in the immune system. Nat. Rev. Immunol. 2, 725–734 (2002).

    Article  CAS  PubMed  Google Scholar 

  88. Perkins, N. D. Integrating cell-signalling pathways with NF-κB and IKK function. Nat. Rev. Mol. Cell Biol. 8, 49–62 (2007).

    Article  CAS  PubMed  Google Scholar 

  89. Shono, Y. et al. Characterization of a c-Rel inhibitor that mediates anticancer properties in hematologic malignancies by blocking NF-κB-controlled oxidative stress responses. Cancer Res. 76, 377–389 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  90. Grinberg-Bleyer, Y. et al. NF-κB c-Rel Is crucial for the regulatory T cell immune checkpoint in cancer. Cell 170, 1096–1108 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  91. De Bock, K. et al. Role of PFKFB3-driven glycolysis in vessel sprouting. Cell 154, 651–663 (2013).

    Article  PubMed  CAS  Google Scholar 

  92. Heise, N. et al. Germinal center B cell maintenance and differentiation are controlled by distinct NF-κB transcription factor subunits. J. Exp. Med. 211, 2103–2118 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  93. Mederacke, I. et al. Fate tracing reveals hepatic stellate cells as dominant contributors to liver fibrosis independent of its aetiology. Nat. Commun. 4, 2823 (2013).

    Article  PubMed  CAS  Google Scholar 

  94. Higgins, G. A. & Anderson, R. E. Experimental pathology of liver: restoration of liver in white rat following partial surgical removal. Arch. Pathol. 12, 186–202 (1931).

  95. Oakley, F. et al. Nuclear factor-κB1 (p50) limits the inflammatory and fibrogenic responses to chronic injury. Am. J. Pathol. 166, 695–708 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

This work was funded by a UK Medical Research Council PhD studentship to J.L. and program grants MR/K0019494/1 (to D.A.M., J.M. and F.O.) and MR/R023026/1 (to D.A.M., J.M., L.A.B. and F.O.). An Arthritis Research UK grant (20812) supports F.O., D.A.M., J.L. and J.C.W. A CRUK program grant (C18342/A23390) supports J.L. and D.A.M. The cross-council Lifelong Health and Wellbeing initiative, funded by the MRC (L016354) funds D.A.M. and F.O. C.N. is supported by CRUK Beatson Institute Core funding (A171196). T.G.B. is funded by the Wellcome Trust (WT107492Z) and a CRUK/AECC/AIRC Accelerator Award (A26813). M.Y.W.Z. has a personal Ph.D award from Newton-Mosharafa fund. P.C., L.S. and L.-A.T. are financed by Methusalem funding, FWO, ERC Proof of Concept (ERC-713758) and an Advanced ERC Research Grant (EU-ERC743074). The IVIS spectrum was purchased under a Wellcome Trust Equipment Grant (087961) awarded to D.A.M. and others. We thank the Newcastle University bioimaging unit and the Newcastle University Flow cytometry core facility for technical assistance.

Author information

Authors and Affiliations

Authors

Contributions

J.L. performed the majority of the laboratory-based work and analyses. S.L., A.K., C.B., G.C., L.A.B., J.C.W., A.L.C., R.A.B., L.M.G., R.C., S.M., M.T., B.S.B., X.X., M.Y.W.Z., W.J.R., H.L.P., C.N., T.B., C.B., M.G.M., L.S., L.-A.T., M.K., L.S., A.D.B. and C.B.N. performed a portion of the laboratory experiments and their related analyses. S.M.R., D.M.M., G.S., J.F., S.A.W., J.L.Z., U.K., R.F.S. and I.M. contributed materials and/or analysis tools. L.A.B., A.F., N.S., P.C., J.M. and D.A.M. provided advice and/or contributed to the experimental design and writing. J.L., D.A.M. and F.O. conceived the studies, designed the experiments and wrote the manuscript. All authors read and commented on the final manuscript.

Corresponding authors

Correspondence to Jack Leslie or Fiona Oakley.

Ethics declarations

Competing interests

F.O., D.A.M., J.M. and L.A.B. are directors of Fibrofind. J.L., H.P., F.O., D.A.M., J.M. and L.A.B. are shareholders in Fibrofind. C.B.N. is shareholder in GSK. M.K. is a stock owner of Nordic Bioscience.

Additional information

Peer review information Primary Handling Editor: Christoph Schmitt.

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

Extended data

Extended Data Fig. 1 c-Rel correlates with disease progression in chronic kidney disease and is elevated in chronic lung disease. Investigation of the cell specific actions of c-Rel in preclinical models of liver injury.

a, Graph showing average percentage area of c-Rel stained tissue in (n=5) normal lung and (n=7) chronic obstructive pulmonary disorder (COPD) (P value = 0.0003). b, Graph showing average percentage area of c-Rel stained tissue in (n=5) normal human kidney and patients with stable (n=4, p value =0.0185) or progressive kidney disease (n=8, p value = 0.003). Normal vs progressive p value <0.0001) (c) Graphs showing the Mean Fluorescence Intensity (MFI) of GFP in hepatocytes (p value = 0.0026), Cholangiocytes (EPCAM+, p value = 0.0026) and non-parenchymal (EPCAM-) cells from the livers of Relfl/fl and RelΔAlb mice. Data are mean ± s.e.m of 3 mice/group. d, Heatmap showing gene expression of Cxcl1, Cxcl2, Ccl2, Cxcl9, Cxcl10, Il1b, Il6 and Tnfα in olive oil vehicle control and acute CCl4 injured Relfl/fl and RelΔAlb mice. Asterisk denotes significance between CCl4 injured Relfl/fl and RelΔAlb mice; there is no significant difference between olive oil treated groups. (e) Histological assessment of αSMA, PCNA and CD68 stained liver sections in acute CCl4 injured Relfl/fl and RelΔLrat mice. Data in graphs are mean ± s.e.m of n=5. a, c, e, P values were calculated using unpaired two-sided T test. b, P value was calculated using a one-way ANOVA with Tukey post-hoc t-test (* P <0.05, ** P <0.01, *** P <0.001 and **** P <0.0001).

Extended Data Fig. 2 c-Rel regulates epithelial dedifferentiation and fibrogenic gene expression.

a, Representative images of c-Rel staining in CCl4 injured Relflfl and RelΔHep mice. Representative of n=7 mice/group. Scale bar represents 100 μm. b, Graph shows mRNA levels of inflammatory cytokines Tnfa, Il-1b (p=0.00038) and Il6 (p value = 0.022)and the inflammatory chemokines Cxcl1 (p value = 0.034), Cxcl2 (p=0.054) and Ccl2 (p=0.029), in CCl4 injured livers of Relfl/fl and RelΔHep mice. Data are mean ± s.e.m of 7 mice/group. c, Heatmap showing relative mRNA expression of Il-6, Cxcl1, Cxcl2, Ccl2, Ccl4 and Ccl5 in WT and Rel−/− proximal tubular epithelial cells (PTEC) stimulated with or without TGFβ1. d, Graphs show relative levels of Ctgf (p value = 0.034), Ctsd (p= 0.039), Serpine1 (p=0.04) and Bmp1 (p=0.0054) protein expressed as Intensity in control and TGFβ1 treated WT and Rel-/- hepatocytes. Data are from 4 independent cell isolations/group. e, Quantification of soluble collagen (pg/ml) released from precision cut liver slices (PCLS) generated from WT and Rel-/- liver, stimulated ± TGFβ1 ± CTGF where (control p value = 0.012 and TGFβ1 p value = 0.045). Data are from PCLS generated 3 different donors/genotype. b, d, P values were calculated using unpaired two-sided t-test. (d) P values were calculated using the R package LIMMA (* P <0.05, ** P <0.01 and ***P<0.001).

Extended Data Fig. 3 c-Rel regulates metabolic enzymes to induce epithelial dedifferentiation and fibrogenic gene expression.

a, Seahorse analysis of basal (p<0.0001) and maximal (p<0.0001) glycolysis (extracellular acidification rate, ECAR) and basal (p<0.0007) and maximal (p<0.0004) mitochondrial respiration (oxygen consumption rate, OCR) in WT and Rel-/- hepatocytes stimulated ± TGFβ1. b, Graph shows mRNA expression of Pfkfb1 in WT and Rel-/- hepatocytes stimulated ± TGFβ1. c, Graph shows mRNA expression of Pfkfb3 in WT and Rel-/- hepatocytes stimulated ± IL-1β. (p value=0.025) (d) Schematic representation of RelA, NF-κB1 and NF-κB binding sites in the murine Pfkfb1 and Pfkfb3 promoters. e, ChIP analysis of c-Rel at the proximal and distal regions of the Pfkfb1 promoter and the proximal (p<0.0001) and distal (p=0.0185) regions of the Pfkfb3 promoter in WT hepatocytes treated ± TGFβ1. f, Representative images show PFKFB3 immunohistochemical staining in liver sections from acute CCl4 injured Pfkfb3flfl and Pfkfb3Δhep mice. Images are representative of n=5 mice/group. Scale bar is 100μm. g, Graphs show media lactate in control and TGFβ1 (p=0.0064) stimulated and glucose levels in control (p=0.0227) and TGFβ1 (p=0.00284) stimulated in hepatocytes isolated from Pfkfb3flfl and Pfkfb3Δhep mice and stimulated ± TGFβ1. h, Heatmap showing secreated Il-6, Cxcl1, Cxcl2, Cxcl10, Ccl2, Ccl3, Ccl4 and Ccl5, measured by MSD in the media of hepatocytes isolated from Pfkfb3flfl and Pfkfb3Δhep mice and stimulated ± TGFβ1. (i) Quantification of connective tissue growth factor (CTGF) in pg/ml in the culture media of hepatocytes isolated from Pfkfb3flfl and Pfkfb3Δhep mice and stimulated ± TGFβ1 (p=0.0027) (j) Heatmap showing secreated Il-6, Cxcl1, Cxcl2, Cxcl10, Ccl2, Ccl3, Ccl4 and Ccl5, measured by MSD in the media of WT hepatocytes stimulated ± TGFβ1 ± the Pfkfb3 inhibitor 3PO. k, Quantification of connective tissue growth factor (CTGF) in pg/ml in the culture media of WT hepatocytes stimulated ± TGFβ1 ± the Pfkfb3 inhibitor 3PO (p=0.0013). Data in graphs are mean ± s.e.m. in n=3 (g, i, k), n=4 (a, b, c) or n=5 (e) independent cell isolations/condition. All p values were calculated using a two-way ANOVA with Tukey post-hoc t-test (* P <0.05, ** P <0.01 and *** P <0.001).

Extended Data Fig. 4 c-Rel dependent Snail regulation in chronic liver and kidney disease.

a, Representative images show expression of Snail in normal and chronic CCl4 injured liver. b, Representative images show Snail in normal and UUO injured kidney. c, Representative images of Snail transcript in epithelial cells detected by RNAScope in fibrotic chronic CCl4 injured fibrotic mouse liver. d, Representative images of Snail transcript in epithelial cells detected by RNAScope in fibrotic UUO mouse kidney. All representative images are representative of n=5 mice/group. a, b, Scale bars equal 100 microns (c, d) Scale bars equal 50 microns. e, Graph showing mRNA levels of Snail in TGFβ1 treated hepatocytes (p=0.0001) and proximal tubular epithelial cells (PTEC) (p=0.02) isolated from WT and Rel-/- mice. Data in graphs are mean ± s.e.m. in n=3 independent cell isolations. f, Graph showing ChIP analysis of c-Rel binding to distal (p=0.0074) and proximal (p=0.0063) regions of the Snail promoter in WT hepatocytes stimulated ± TGFβ1. Data in graphs are mean ± s.e.m. in n=5 independent cell isolations. P values were calculated using unpaired two-sided t-test (e) and a ratio paired t-test (f) (* P <0.05, ** P <0.01 and *** P <0.001).

Extended Data Fig. 5 c-Rel regulates macrophages polarisation to drive tissue fibrosis.

a, Histological assessment and representative images of αSMA stained liver sections in acute CCl4 injured in (n=13) Relflfl, (n=7) RelΔAlb (p=0.0116), and (n=10) RelΔLysM (p=0.0051) mice. Data are mean ± s.e.m. Scale bar equals 100 microns. P values were calculated using a one-way ANOVA with Tukey post- hoc t-test. b, Representative low power immuno-fluorescence images show c-Rel (red), CD68 (green) and nuclear (blue) staining in diagnosed idiopathic pulmonary fibrosis lung sections. Yellow arrows denote co-localisaton of c-Rel and CD68. Scale bar equals 50 microns. Images are representative of n=8 IPF stained sections. (c) Heat map showing relative mRNA expression of fibrogenic markers; Tgfb1, Tgfb2, Tgfb3, TIMP1, Fn1, Pdgfa, Pdgfb, Mmp3, Mmp9 and Mmp13 in M1 and M2 polarised WT and Rel-/- BMDMs. d, Graphs show, seahorse analysis of basal (p<0.0001) and maximal (p=0.0004) glycolysis (extracellular acidification rate, ECAR) and basal (p=0.0002) and maximal (p=0.0024) mitochondrial respiration (oxygen consumption rate, OCR) in M1 and M2 polarised WT and Rel-/- BMDMs. Data are mean ± s.e.m. from 3 independent cell isolations/group. e, ChIP analysis of c-Rel binding to distal and proximal (p=0.0004) regions of the Pfkfb3 promoter and to distal and proximal (p=0.003) regions of the Pfkfb1 promoters in WT BMDM in response to M1 and M2 polarisation. Data are mean ± s.e.m. from 3 independent cell isolations/group. d-e, P values were calculated using two-way ANOVA with Tukey post-hoc t-test. Denoted significance refers to comparisons between WT and Rel-/- macrophages polarised to either an M1 or M2 phenotype. f, Volcano plots show differentially expressed proteins detected by proteomic analysis of the secretome of M0, M1 and M2 polarised WT and Rel-/- BMDMs. g, Graphs show relative levels of Galectin 1 (p=0.031), Galectin 3 (p=0.013), Vimentin (p=0.0128) and Matrix Metalloproteinase 12 (MMP12) (p<0.001) expressed as Intensity x108 in M0, M1 and M2 polarised WT and Rel-/- BMDMs. Data are mean ± s.e.m. from 4 independent cell isolations/group generated 4 different donors/genotype. P values were calculated using the R package LIMMA (* P <0.05, ** P <0.01 and *** P <0.001).

Extended Data Fig. 6 Validation of epithelial specific deletion of c-Rel in kidney and lung fibrosis models.

a, Flow cytometry gating strategy to identify immune cells (CD45+), epithelial cells (EPCAM+) and endothelial cells (CD31+) isolated from the kidney or lungs of Relflfl, RelΔTEC or RelΔAEC mice respectively. b-c, Ex vivo images of GFP fluorescence signal in the left and right kidneys, heart, liver, lungs and spleen of RelΔTEC mice (b) or RelΔAEC mice (c) imaged using an In Vivo Imaging System (IVIS). Graph and flow cytometry histograms show the Mean Fluorescence Intensity (MFI) of GFP in EpCAM+, CD31+, CD45+ and EpCAM-CD31-CD45- cells from the kidney of RelΔTEC and Relfl/fl control mice (b) or from the lung of RelΔAEC and Relfl/fl control mice (c), n=5 mice/group. d, Heatmap showing mRNA levels of Tgfb1, Col1a1, Col1a2, Acta2 and Timp1 in control versus UUO kidney of Relflfl, RelΔTEC and RelΔLysM mice (left) or control versus bleomycin lung of Relflfl, RelΔAEC and RelΔLysM mice (right). Heatmap data are from 2 mice/group in control kidney or lung and 5 mice/group in the injured kidney or lung. e, Quantification of hydroxyproline levels in nM per left lobe of lung tissue from bleomycin injured in Relflfl, RelΔAEC (p=0.0105), and RelΔLysM (p=0.0006) mice. f, Histological assessment and representative images of TUNEL stained lung sections in day 3 bleomycin injured in Relflfl, RelΔAEC, and RelΔLysM mice. Scale bar is 50 microns. Data are mean ± s.e.m. in n=5 mice/group. g, Quantification of neutrophil numbers in UUO injured kidneys of Relflfl, RelΔTEC (p=0.0014) and RelΔLysM (p<0.0001) mice and macrophage numbers in UUO injured kidneys of Relflfl, RelΔTEC (p=0.00045) and RelΔLysM (p=0.0004) mice. h, Quantification of neutrophil numbers in Bleomycin injured lungs of Relflfl, RelΔAEC and RelΔLysM mice and macrophage numbers in Bleomycin injured lungs of Relflfl, RelΔAEC (p=0.0018) and RelΔLysM (p=0.0327) mice. e, g, h, Data are mean ± s.e.m. in a minimum of 7 mice/group for the kidney and 10 mice/group for the lung. P values were calculated using one-way ANOVA with Tukey post-hoc t-test (* P <0.05, **P <0.01 and ***P <0.001).

Extended Data Fig. 7 Validation of single myeloid or dual hepatocyte- and myeloid- specific deletion of c-Rel in mice and analysis of fibrogenic gene expression in these mice during chronic liver injury.

a, Flow cytometry plot and graphs show the Mean Fluorescence Intensity (MFI) of GFP expression in hepatocytes, macrophages (CD45+F4/80+CD11b+) (p=0.011), T-cells (CD45+CD3+) and non-parenchymal cells (CD45-) from the liver of Relflfl versus RelΔLysM mice, n=3 mice/group. b, Flow cytometry plot and graphs show the Mean Fluorescence Intensity (MFI) of GFP expression in hepatocytes (p=0.007), macrophages (CD45+F4/80+CD11b+) (p=0.007), T-cells (CD45+CD3+) and non-parenchymal cells (CD45-) from the liver of Relflfl versus RelΔHep/ΔLysM mice, n=3 mice/group. (c) Heatmap showing mRNA levels of Tgfb1, Col1a1, Col1a2, Acta2 and Timp1 in the CCl4 injured liver of Relflfl, RelΔHep, RelΔLysM and RelΔHep/ΔLysM mice. P values were calculated using two-sided t-test (* P <0.05).

Extended Data Fig. 8 c-Rel signaling in hepatocyte and macrophages differentially regulate liver regeneration via regulation of cell cycle genes and mitogenic factors.

a, Histological assessment and representative images of PCNA positive hepatocytes in 48h partial hepatectomy injured Relflfl, RelΔAlb (p=0.0003) and RelΔLysM (p=0.0096) mice. Scale bar is 100 microns. b, Graph shows average liver/body weight ratio 48h post partial hepatectomy in Relflfl, RelΔAlb (p=0.0318) and RelΔLysM (p=0.0178) mice. c, Heat map showing relative hepatic mRNA expression of cell cycle genes; Ccna1, Ccnd1, Ccne1, Cdk2, Cdk4 and mitogenic proteins; Hgf and Egf at 48h post partial hepatectomy in Relflfl, RelΔAlb and RelΔLysM mice. Data are mean ± s.e.m. in n=5 mice/group. P values were calculated using one-way ANOVA with Tukey post-hoc t-test (* P <0.05 and ** P <0.01).

Extended Data Fig. 9 IT-603 attenuates fibrogenesis in murine models of fibrosis.

a, U937 cells stably expressing 3xNF-κB-Luc reporter were transiently transfected with RelA or c-Rel expression plasmids. Graph shows RelA and c-Rel induced NF-κB luciferase reporter activity ± IT-603 therapy. Data are mean ± s.e.m. P value = 0.00039. P value was calculated using an unpaired two-sided t-test (***P<0.001) of 3 independent experiments. b, c, Histological quantification of αSMA stained kidney (p=0.0004) or lungs (p=0.009) following their respective injury. Data are mean ± s.e.m. in 7 and 10 mice/group for kidney and lung respectively. P values were calculated using an unpaired two-sided t-test. d, Histological quantification of αSMA stained chronic MCD diet injured livers at 2 weeks (pre-treatment) and 5 weeks ± therapeutic administration of IT-603 (p=0.0044). e, Graphs showing relative hepatic expression of the fibrogenic genes; Tgfb1 (p=0.039), Acta2 (p=0.028), Col1a1 (p=0.036), Col1a2, and Timp1(p=0.015) in 2-week (pre-treatment) and 5-week methionine choline deficient diet (MCD) fed mice ± therapeutic administration of IT-603. Data are mean ± s.e.m. in n=4 control mice 8 vehicle and n=7 IT-603 treated mice/group. f, Histological quantification of αSMA stained chronic CCl4 injured livers at 3 weeks (pre-treatment) or 8 weeks ± therapeutic administration of IT-603 (p=0.0105). g, Graphs showing relative hepatic expression of the fibrogenic genes; Tgfb1 (p=0.029), Acta2 (p=0.032), Col1a1 (p=0.04), Col1a2 (p=0.016), and Timp1 (p=0.09) in chronic CCl4 injured livers at 3 week (pre-treatment), 8 week and ± therapeutic administration of IT-603 (from weeks 3-8). Data are mean ± s.e.m. in n=4 control mice 7 experimental mice/group. c, e, P values were calculated using one-way ANOVA with Tukey post-hoc t-test. d, f, P values were calculated using two-way ANOVA with Tukey post-hoc t-test (*P<0.05, ***P<0.001).

Extended Data Fig. 10 IT-603 attenuates fibrosis in ex vivo human tissues slice models of liver and kidney fibrosis.

a, Diagrams show the experimental timelines of TGFβ1 induced fibrosis in ex vivo normal human liver and kidney precision cut slices (PCS). b, Representative images of CD68 stained liver and kidney tissue slices. c, Representative images and histological quantification of Picrosirius red stained fibrotic kidney slices ± IT-603 therapy (p<0.0001). Representative images and histological quantification of αSMA stained fibrotic kidney slices ± IT-603 therapy (p<0.0001). Red line denotes the value for the T=0 slice. e, Quantification of soluble collagen released from fibrotic kidney slices ± IT-603 therapy (p=0.0015). f, Quantification of the neo-epitope pro C3 released from fibrotic kidney slices ± IT-603 therapy (p=0.0493). g, Graph showing average LDH release in the media expressed as a percentage (%) of positive control (LDH levels in media from a PCS where maximal death was induced by multiple freeze/thaws – normalized to media volume). (IT-603 p=0.023 and IT-603+TGFβ1 p=0.02). Images representative of n=3 independent slice experiments. Data are mean ± s.e.m. and representative of slices generated from 3 independent donors performed in duplicate. Scale bars equal 100 microns. P values were calculated using two-way ANOVA with Tukey post- hoc t-test (*P<0.05, **P<0.01, ***P<0.001 and ****P<0.0001).

Supplementary information

Supplementary Information

Supplementary Fig. 1 and Supplementary Tables 1–4

Reporting Summary

Source data

Source Data Fig. 1

Individual data points.

Source Data Fig. 2

Individual data points.

Source Data Fig. 3

Individual data points.

Source Data Fig. 4

Individual data points.

Source Data Fig. 5

Individual data points.

Source Data Fig. 6

Individual data points.

Source Data Fig. 7

Individual data points.

Source Data Fig. 8

Individual data points.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Leslie, J., Macia, M.G., Luli, S. et al. c-Rel orchestrates energy-dependent epithelial and macrophage reprogramming in fibrosis. Nat Metab 2, 1350–1367 (2020). https://doi.org/10.1038/s42255-020-00306-2

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s42255-020-00306-2

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