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.

  • Review Article
  • Published:

Liver fibrosis and repair: immune regulation of wound healing in a solid organ

Key Points

  • The liver provides a useful generic model of inflammation and repair, showing the complex interplay between the epithelial, inflammatory, myofibroblast and extracellular matrix (ECM) components of the mammalian wound-healing response. In almost all situations, fibrosis is preceded by inflammation and elements of both the innate and adaptive immune systems are crucial in regulating the fibrotic process.

  • Following liver injury, pro-inflammatory mediators that are generated by cellular damage and stimulated immune cells, as well as growth factors and cytokines (including platelet-derived growth factor, connective tissue growth factor, transforming growth factor-β and interleukin-13), activate mesenchymal precursor cells in tissues and induce their transdifferentiation into myofibroblasts. Myofibroblasts are master regulators of the fibrotic response as a result of their acquisition of scar-producing, proliferative, migratory, contractile, immunomodulatory and phagocytic properties.

  • Recent studies have used bone marrow transplantation techniques in reporter mice to show that, regardless of the aetiology or the duration of the injury, liver myofibroblasts are almost exclusively derived from the activation of resident mesenchymal cells. Perpetuation of myofibroblast fibrogenic activity is mediated through several positive feedback loops, involving the autocrine and paracrine effects of cytokines and growth factors, and cell–cell and cell–matrix interactions. Myofibroblasts themselves function as innate immune cells.

  • The balance of T helper 1 (TH1) cell-mediated and TH2 cell-mediated adaptive immune responses, the influence of unconventional T cell subsets and the equilibrium between different pro-inflammatory (that is, pro-fibrotic) and pro-resolution macrophage populations determine whether the outcome of tissue injury is homeostatic and self-limited or whether it results in pathogenic scarring.

  • Liver fibrosis in rodents and humans is a dynamic, bidirectional process that has an inherent capacity for recovery and remodelling. The loss of myofibroblasts from the hepatic scar and a crucial switch in macrophage phenotype to a pro-resolution cell type are important events in the regression of liver fibrosis that facilitate remodelling of the ECM.

  • A considerable number of tractable therapeutic targets have been identified in liver fibrosis, but clinical trials of anti-fibrotic therapies have so far been unsuccessful. Identification of the core pathways in fibrosis is likely to yield greater success in clinical translation.

Abstract

Fibrosis is a highly conserved and co-ordinated protective response to tissue injury. The interaction of multiple pathways, molecules and systems determines whether fibrosis is self-limiting and homeostatic, or whether it is uncontrolled and excessive. Immune cells have been identified as key players in this fibrotic cascade, with the capacity to exert either injury-inducing or repair-promoting effects. A multi-organ approach was recently suggested to identify the core and regulatory pathways in fibrosis, with the aim of integrating the wealth of information emerging from basic fibrosis research. In this Review, we focus on recent advances in liver fibrosis research as a paradigm for wound healing in solid organs and the role of the immune system in regulating and balancing this response.

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

Access options

Rent or buy this article

Prices vary by article type

from$1.95

to$39.95

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

Figure 1: Natural history of chronic liver disease.
Figure 2: Cascade of signals following liver injury.
Figure 3: The duality of macrophages in liver fibrosis.
Figure 4: Hepatic stellate cell-derived myofibroblast fate in liver fibrosis.

Similar content being viewed by others

References

  1. Dechêne, A. et al. Acute liver failure is associated with elevated liver stiffness and hepatic stellate cell activation. Hepatology 52, 1008–1016 (2010).

    Article  CAS  PubMed  Google Scholar 

  2. Bourbonnais, E. et al. Liver fibrosis protects mice from acute hepatocellular injury. Gastroenterology 142, 130–139. e134 (2012).

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  4. Barry-Hamilton, V. et al. Allosteric inhibition of lysyl oxidase-like-2 impedes the development of a pathologic microenvironment. Nature Med. 16, 1009–1017 (2010).

    Article  CAS  PubMed  Google Scholar 

  5. Boulter, L., Lu, W. Y. & Forbes, S. J. Differentiation of progenitors in the liver: a matter of local choice. J. Clin. Invest. 123, 1867–1873 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Jiang, J. X., Mikami, K., Venugopal, S., Li, Y. & Török, N. J. Apoptotic body engulfment by hepatic stellate cells promotes their survival by the JAK/STAT and Akt/NF-κB-dependent pathways. J. Hepatol. 51, 139–148 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Muhanna, N. et al. Activation of hepatic stellate cells after phagocytosis of lymphocytes: a novel pathway of fibrogenesis. Hepatology 48, 963–977 (2008).

    Article  CAS  PubMed  Google Scholar 

  9. Hellerbrand, C., Stefanovic, B., Giordano, F., Burchardt, E. R. & Brenner, D. A. The role of TGFβ1 in initiating hepatic stellate cell activation in vivo. J. Hepatol. 30, 77–87 (1999).

    Article  CAS  PubMed  Google Scholar 

  10. Seki, E. et al. TLR4 enhances TGFβ signaling and hepatic fibrosis. Nature Med. 13, 1324–1332 (2007).

    Article  CAS  PubMed  Google Scholar 

  11. Friedman, S. L., Roll, F. J., Boyles, J. & Bissell, D. M. Hepatic lipocytes: the principal collagen-producing cells of normal rat liver. Proc. Natl Acad. Sci. USA 82, 8681–8685 (1985).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Dranoff, J. A. & Wells, R. G. Portal fibroblasts: Underappreciated mediators of biliary fibrosis. Hepatology 51, 1438–1444 (2010).

    Article  PubMed  Google Scholar 

  13. Brenner, D. A. et al. Origin of myofibroblasts in liver fibrosis. Fibrogenesis Tissue Repair 5, S17 (2012).

    Article  PubMed  PubMed Central  Google Scholar 

  14. Moeller, A. et al. Circulating fibrocytes are an indicator of poor prognosis in idiopathic pulmonary fibrosis. Am. J. Respir. Crit. Care Med. 179, 588–594 (2009).

    Article  PubMed  Google Scholar 

  15. Bonner, J. C. Regulation of PDGF and its receptors in fibrotic diseases. Cytokine Growth Factor Rev. 15, 255–273 (2004).

    Article  CAS  PubMed  Google Scholar 

  16. Oakley, F. et al. Angiotensin II activates IκB kinase phosphorylation of RelA at Ser 536 to promote myofibroblast survival and liver fibrosis. Gastroenterology 136, 2334–2344. e2331 (2009).

    Article  CAS  PubMed  Google Scholar 

  17. Horowitz, J. C. et al. Combinatorial activation of FAK and AKT by transforming growth factor-β1 confers an anoikis-resistant phenotype to myofibroblasts. Cell. Signal. 19, 761–771 (2007).

    Article  CAS  PubMed  Google Scholar 

  18. Mann, J. et al. MeCP2 controls an epigenetic pathway that promotes myofibroblast transdifferentiation and fibrosis. Gastroenterology 138, 705–714. e1–4 (2010). This study shows that myofibroblast transdifferentiation of hepatic stellate cells is controlled by a combination of MeCP2, EZH2 and miR-132 in a relay pathway, and that genetic and pharmacological disruptions of this pathway decrease the fibrogenic characteristics of myofibroblasts and attenuate fibrogenesis.

    Article  CAS  PubMed  Google Scholar 

  19. Perugorria, M. J. et al. Histone methyltransferase ASH1 orchestrates fibrogenic gene transcription during myofibroblast transdifferentiation. Hepatology 56, 1129–1139 (2012).

    Article  CAS  PubMed  Google Scholar 

  20. Olsen, A. L. et al. Hepatic stellate cells require a stiff environment for myofibroblastic differentiation. Am. J. Physiol. Gastrointest. Liver Physiol. 301, G110–118 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Wipff, P. J., Rifkin, D. B., Meister, J. J. & Hinz, B. Myofibroblast contraction activates latent TGFβ1 from the extracellular matrix. J. Cell. Biol. 179, 1311–1323 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Henderson, N. C. et al. Targeting of αv integrin identifies a core molecular pathway that regulates fibrosis in several organs. Nature Med. 19, 1617–1624 (2013).

    Article  CAS  PubMed  Google Scholar 

  23. Popov, Y. et al. Integrin αvβ6 is a marker of the progression of biliary and portal liver fibrosis and a novel target for antifibrotic therapies. J. Hepatol. 48, 453–464 (2008).

    Article  CAS  PubMed  Google Scholar 

  24. Viñas, O. et al. Human hepatic stellate cells show features of antigen-presenting cells and stimulate lymphocyte proliferation. Hepatology 38, 919–929 (2003).

    Article  PubMed  Google Scholar 

  25. Hughes, J., Liu, Y., Van Damme, J. & Savill, J. Human glomerular mesangial cell phagocytosis of apoptotic neutrophils: mediation by a novel CD36-independent vitronectin receptor/thrombospondin recognition mechanism that is uncoupled from chemokine secretion. J. Immunol. 158, 4389–4397 (1997).

    CAS  PubMed  Google Scholar 

  26. Seki, E. et al. CCR1 and CCR5 promote hepatic fibrosis in mice. J. Clin. Invest. 119, 1858–1870 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  27. Puche, J. E. et al. A novel murine model to deplete hepatic stellate cells uncovers their role in amplifying liver damage in mice. Hepatology 57, 339–350 (2013).

    Article  CAS  PubMed  Google Scholar 

  28. 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). This study shows that macrophages have crucial but divergent roles in experimental liver fibrosis, favouring ECM accumulation during ongoing injury but enhancing matrix degradation during recovery.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Fallowfield, J. A. et al. Scar-associated macrophages are a major source of hepatic matrix metalloproteinase-13 and facilitate the resolution of murine hepatic fibrosis. J. Immunol. 178, 5288–5295 (2007).

    Article  CAS  PubMed  Google Scholar 

  30. Ide, M., Kuwamura, M., Kotani, T., Sawamoto, O. & Yamate, J. Effects of gadolinium chloride (GdCl3) on the appearance of macrophage populations and fibrogenesis in thioacetamide-induced rat hepatic lesions. J. Comp. Pathol. 133, 92–102 (2005).

    Article  CAS  PubMed  Google Scholar 

  31. Sunami, Y. et al. Hepatic activation of IKK/NFκB signaling induces liver fibrosis via macrophage-mediated chronic inflammation. Hepatology 56, 1117–1128 (2012).

    Article  CAS  PubMed  Google Scholar 

  32. Pradere, J. P. et al. Hepatic macrophages but not dendritic cells contribute to liver fibrosis by promoting the survival of activated hepatic stellate cells in mice. Hepatology 58, 1461–1473 (2013).

    Article  CAS  PubMed  Google Scholar 

  33. Holt, M. P., Cheng, L. & Ju, C. Identification and characterization of infiltrating macrophages in acetaminophen-induced liver injury. J. Leukoc. Biol. 84, 1410–1421 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Yona, S. et al. Fate mapping reveals origins and dynamics of monocytes and tissue macrophages under homeostasis. Immunity 38, 79–91 (2013).

    Article  CAS  PubMed  Google Scholar 

  35. Seki, S. et al. The liver as a crucial organ in the first line of host defense: the roles of Kupffer cells, natural killer (NK) cells and NK1.1 Ag+ T cells in T helper 1 immune responses. Immunol. Rev. 174, 35–46 (2000).

    Article  CAS  PubMed  Google Scholar 

  36. Wiegard, C. et al. Murine liver antigen presenting cells control suppressor activity of CD4+CD25+ regulatory T cells. Hepatology 42, 193–199 (2005).

    Article  CAS  PubMed  Google Scholar 

  37. Ju, C. & Pohl, L. R. Tolerogenic role of Kupffer cells in immune-mediated adverse drug reactions. Toxicology 209, 109–112 (2005).

    Article  CAS  PubMed  Google Scholar 

  38. Ramachandran, P. et al. Differential Ly-6C expression identifies the recruited macrophage phenotype, which orchestrates the regression of murine liver fibrosis. Proc. Natl Acad. Sci. USA 109, E3186–E3195 (2012). This study identifies the pro-resolution macrophage population in resolving liver fibrosis models as a CD11bhiF4/80intLY6Clow macrophage subset, which is derived from recruited LY6Chi monocytes that undergo a phenotypical switch induced by phagocytosis.

    Article  PubMed  PubMed Central  Google Scholar 

  39. Tacke, F. & Zimmermann, H. W. Macrophage heterogeneity in liver injury and fibrosis. J. Hepatol. http://dx.doi.org/10.1016/j.jhep.2013.12.025 (2014).

  40. Karlmark, K. R. et al. Hepatic recruitment of the inflammatory Gr1+ monocyte subset upon liver injury promotes hepatic fibrosis. Hepatology 50, 261–274 (2009). This study shows that inflammatory GR1+ monocytes, which are recruited into the injured liver via CCR2-dependent bone marrow egress, promote the progression of liver fibrosis.

    Article  CAS  PubMed  Google Scholar 

  41. Lin, S. L., Castaño, A. P., Nowlin, B. T., Lupher, M. L. & Duffield, J. S. Bone marrow Ly6Chigh monocytes are selectively recruited to injured kidney and differentiate into functionally distinct populations. J. Immunol. 183, 6733–6743 (2009).

    Article  CAS  PubMed  Google Scholar 

  42. Hayashidani, S. et al. Anti-monocyte chemoattractant protein-1 gene therapy attenuates left ventricular remodeling and failure after experimental myocardial infarction. Circulation 108, 2134–2140 (2003).

    Article  CAS  PubMed  Google Scholar 

  43. Gibbons, M. A. et al. Ly6Chi monocytes direct alternatively activated profibrotic macrophage regulation of lung fibrosis. Am. J. Respir. Crit. Care Med. 184, 569–581 (2011).

    Article  CAS  PubMed  Google Scholar 

  44. Lucas, T. et al. Differential roles of macrophages in diverse phases of skin repair. J. Immunol. 184, 3964–3977 (2010).

    Article  CAS  PubMed  Google Scholar 

  45. Imamura, M., Ogawa, T., Sasaguri, Y., Chayama, K. & Ueno, H. Suppression of macrophage infiltration inhibits activation of hepatic stellate cells and liver fibrogenesis in rats. Gastroenterology 128, 138–146 (2005).

    Article  CAS  PubMed  Google Scholar 

  46. Mitchell, C. et al. Dual role of CCR2 in the constitution and the resolution of liver fibrosis in mice. Am. J. Pathol. 174, 1766–1775 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Jenkins, S. J. et al. Local macrophage proliferation, rather than recruitment from the blood, is a signature of TH2 inflammation. Science 332, 1284–1288 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Robbins, C. S. et al. Local proliferation dominates lesional macrophage accumulation in atherosclerosis. Nature Med. 19, 1166–1172 (2013).

    Article  CAS  PubMed  Google Scholar 

  49. Davies, L. C. et al. Distinct bone marrow-derived and tissue-resident macrophage lineages proliferate at key stages during inflammation. Nature Commun. 4, 1886 (2013).

    Article  CAS  Google Scholar 

  50. Wynn, T. A. & Barron, L. Macrophages: master regulators of inflammation and fibrosis. Semin. Liver. Dis. 30, 245–257 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Liaskou, E. et al. Monocyte subsets in human liver disease show distinct phenotypic and functional characteristics. Hepatology 57, 385–398 (2013).

    Article  CAS  PubMed  Google Scholar 

  52. Henderson, N. C. et al. Galectin-3 regulates myofibroblast activation and hepatic fibrosis. Proc. Natl Acad. Sci. USA 103, 5060–5065 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Zimmermann, H. W. et al. Functional contribution of elevated circulating and hepatic non-classical CD14CD16 monocytes to inflammation and human liver fibrosis. PLoS ONE 5, e11049 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Shi, Z., Wakil, A. E. & Rockey, D. C. Strain-specific differences in mouse hepatic wound healing are mediated by divergent T helper cytokine responses. Proc. Natl Acad. Sci. USA 94, 10663–10668 (1997).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Wynn, T. A. et al. An IL-12-based vaccination method for preventing fibrosis induced by schistosome infection. Nature 376, 594–596 (1995).

    Article  CAS  PubMed  Google Scholar 

  56. Wynn, T. A. Fibrotic disease and the TH1/TH2 paradigm. Nature Rev. Immunol. 4, 583–594 (2004).

    Article  CAS  Google Scholar 

  57. Chiaramonte, M. G., Donaldson, D. D., Cheever, A. W. & Wynn, T. A. An IL-13 inhibitor blocks the development of hepatic fibrosis during a T-helper type 2-dominated inflammatory response. J. Clin. Invest. 104, 777–785 (1999).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Korn, T., Bettelli, E., Oukka, M. & Kuchroo, V. K. IL-17 and TH17 Cells. Annu. Rev. Immunol. 27, 485–517 (2009).

    Article  CAS  PubMed  Google Scholar 

  59. Meng, F. et al. Interleukin-17 signaling in inflammatory, Kupffer cells, and hepatic stellate cells exacerbates liver fibrosis in mice. Gastroenterology 143, 765–776. e1–3 (2012).

    Article  CAS  PubMed  Google Scholar 

  60. Zhang, Y. et al. A proinflammatory role for interleukin-22 in the immune response to hepatitis B virus. Gastroenterology 141, 1897–1906 (2011).

    Article  CAS  PubMed  Google Scholar 

  61. Katz, S. C. et al. Obstructive jaundice expands intrahepatic regulatory T cells, which impair liver T lymphocyte function but modulate liver cholestasis and fibrosis. J. Immunol. 187, 1150–1156 (2011).

    Article  CAS  PubMed  Google Scholar 

  62. Turner, J. D. et al. CD4+CD25+ regulatory cells contribute to the regulation of colonic TH2 granulomatous pathology caused by schistosome infection. PLoS Negl. Trop. Dis. 5, e1269 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  63. Langhans, B. et al. Intrahepatic IL-8 producing Foxp3+CD4+ regulatory T cells and fibrogenesis in chronic hepatitis C. J. Hepatol. 59, 229–235 (2013).

    Article  CAS  PubMed  Google Scholar 

  64. Safadi, R. et al. Immune stimulation of hepatic fibrogenesis by CD8 cells and attenuation by transgenic interleukin-10 from hepatocytes. Gastroenterology 127, 870–882 (2004).

    CAS  PubMed  Google Scholar 

  65. Novobrantseva, T. I. et al. Attenuated liver fibrosis in the absence of B cells. J. Clin. Invest. 115, 3072–3082 (2005). This study shows that B cells have a role in CCl 4 -induced hepatic fibrosis in mice in an antibody- and T cell-independent manner.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  66. Kornek, M., Popov, Y., Libermann, T. A., Afdhal, N. H. & Schuppan, D. Human T cell microparticles circulate in blood of hepatitis patients and induce fibrolytic activation of hepatic stellate cells. Hepatology 53, 230–242 (2011).

    Article  CAS  PubMed  Google Scholar 

  67. Geissmann, F. et al. Intravascular immune surveillance by CXCR6+ NKT cells patrolling liver sinusoids. PLoS Biol. 3, e113 (2005). This study shows that CD1d-reactive NKT cells patrol the liver sinusoids, rather than extravasate like effector cells in other tissues, and that the chemokine receptor CXCR6 regulates the number of hepatic NKT cells.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  68. Wehr, A. et al. Chemokine receptor CXCR6-dependent hepatic NK T Cell accumulation promotes inflammation and liver fibrosis. J. Immunol. 190, 5226–5236 (2013).

    Article  CAS  PubMed  Google Scholar 

  69. Hammerich, L. et al. Chemokine receptor CCR6-dependent accumulation of γδ T cells in injured liver restricts hepatic inflammation and fibrosis. Hepatology 59, 630–642 (2013). This study shows that γδ T cells are recruited to the liver by CCR6 in chronic injury and have an anti-fibrotic effect by promoting hepatic stellate cell apoptosis in a cell–cell contact-dependent manner involving CD95L.

    Article  CAS  PubMed  Google Scholar 

  70. Liang, Y. et al. IL-33 induces nuocytes and modulates liver injury in viral hepatitis. J. Immunol. 190, 5666–5675 (2013).

    Article  CAS  PubMed  Google Scholar 

  71. McHedlidze, T. et al. Interleukin-33-dependent innate lymphoid cells mediate hepatic fibrosis. Immunity 39, 357–371 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  72. Hams, E. et al. IL-25 and type 2 innate lymphoid cells induce pulmonary fibrosis. Proc. Natl Acad. Sci. USA 111, 367–372 (2014).

    Article  CAS  PubMed  Google Scholar 

  73. Radaeva, S. et al. Natural killer cells ameliorate liver fibrosis by killing activated stellate cells in NKG2D-dependent and tumor necrosis factor-related apoptosis-inducing ligand-dependent manners. Gastroenterology 130, 435–452 (2006). This study shows that NK cells kill activated hepatic stellate cells through retinoic acid early inducible protein 1 (RAE1)–NK group 2 member D (NKG2D)-dependent and TRAIL-dependent mechanisms, thereby ameliorating liver fibrosis.

    Article  CAS  PubMed  Google Scholar 

  74. Saito, J. M., Bostick, M. K., Campe, C. B., Xu, J. & Maher, J. J. Infiltrating neutrophils in bile duct-ligated livers do not promote hepatic fibrosis. Hepatol. Res. 25, 180–191 (2003).

    Article  PubMed  Google Scholar 

  75. Xu, J., Lee, G., Wang, H., Vierling, J. M. & Maher, J. J. Limited role for CXC chemokines in the pathogenesis of α-naphthylisothiocyanate-induced liver injury. Am. J. Physiol. Gastrointest. Liver Physiol. 287, G734–G741 (2004).

    Article  CAS  PubMed  Google Scholar 

  76. Harty, M. W. et al. Neutrophil depletion blocks early collagen degradation in repairing cholestatic rat livers. Am. J. Pathol. 176, 1271–1281 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  77. Franceschini, B., Russo, C., Dioguardi, N. & Grizzi, F. Increased liver mast cell recruitment in patients with chronic C virus-related hepatitis and histologically documented steatosis. J. Viral Hepat. 14, 549–555 (2007).

    Article  CAS  PubMed  Google Scholar 

  78. Sugihara, A., Tsujimura, T., Fujita, Y., Nakata, Y. & Terada, N. Evaluation of role of mast cells in the development of liver fibrosis using mast cell-deficient rats and mice. J. Hepatol. 30, 859–867 (1999).

    Article  CAS  PubMed  Google Scholar 

  79. Ellis, E. L. & Mann, D. A. Clinical evidence for the regression of liver fibrosis. J. Hepatol. 56, 1171–1180 (2012).

    Article  PubMed  Google Scholar 

  80. Marcellin, P. et al. Regression of cirrhosis during treatment with tenofovir disoproxil fumarate for chronic hepatitis B: a 5-year open-label follow-up study. Lancet 381, 468–475 (2013).

    Article  CAS  PubMed  Google Scholar 

  81. Iredale, J. P. et al. Mechanisms of spontaneous resolution of rat liver fibrosis. Hepatic stellate cell apoptosis and reduced hepatic expression of metalloproteinase inhibitors. J. Clin. Invest. 102, 538–549 (1998). This study shows that apoptosis of activated hepatic stellate cells is a crucial mechanism in the resolution of liver fibrosis as it removes the cell population that is responsible for both producing fibrotic neomatrix and protecting this matrix from degradation through the production of TIMPs.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  82. Issa, R. et al. Apoptosis of hepatic stellate cells: involvement in resolution of biliary fibrosis and regulation by soluble growth factors. Gut 48, 548–557 (2001).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  83. Abu Dayyeh, B. K., Yang, M., Dienstag, J. L. & Chung, R. T. The effects of angiotensin blocking agents on the progression of liver fibrosis in the HALT-C Trial cohort. Dig. Dis. Sci. 56, 564–568 (2011).

    Article  CAS  PubMed  Google Scholar 

  84. McHutchison, J. et al. Farglitazar lacks antifibrotic activity in patients with chronic hepatitis C infection. Gastroenterology 138, 1365–1373. e1–2 (2010).

    Article  PubMed  Google Scholar 

  85. Iredale, J. Defining therapeutic targets for liver fibrosis: exploiting the biology of inflammation and repair. Pharmacol. Res. 58, 129–136 (2008).

    Article  CAS  PubMed  Google Scholar 

  86. Iredale, J. P. et al. Tissue inhibitor of metalloproteinase-1 messenger RNA expression is enhanced relative to interstitial collagenase messenger RNA in experimental liver injury and fibrosis. Hepatology 24, 176–184 (1996).

    Article  CAS  PubMed  Google Scholar 

  87. Issa, R. et al. Spontaneous recovery from micronodular cirrhosis: evidence for incomplete resolution associated with matrix cross-linking. Gastroenterology 126, 1795–1808 (2004).

    Article  CAS  PubMed  Google Scholar 

  88. Parsons, C. J. et al. Antifibrotic effects of a tissue inhibitor of metalloproteinase-1 antibody on established liver fibrosis in rats. Hepatology 40, 1106–1115 (2004).

    Article  CAS  PubMed  Google Scholar 

  89. Siller-López, F. et al. Treatment with human metalloproteinase-8 gene delivery ameliorates experimental rat liver cirrhosis. Gastroenterology 126, 1122–1133; discussion 1949 (2004).

    Article  CAS  PubMed  Google Scholar 

  90. Pellicoro, A. et al. Elastin accumulation is regulated at the level of degradation by macrophage metalloelastase (MMP-12) during experimental liver fibrosis. Hepatology 55, 1965–1975 (2012).

    Article  CAS  PubMed  Google Scholar 

  91. Popov, Y. et al. Macrophage-mediated phagocytosis of apoptotic cholangiocytes contributes to reversal of experimental biliary fibrosis. Am. J. Physiol. Gastrointest. Liver Physiol. 298, G323–G334 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  92. Karlmark, K. R. et al. The fractalkine receptor CX3CR1 protects against liver fibrosis by controlling differentiation and survival of infiltrating hepatic monocytes. Hepatology 52, 1769–1782 (2010).

    Article  CAS  PubMed  Google Scholar 

  93. Aoyama, T., Inokuchi, S., Brenner, D. A. & Seki, E. CX3CL1-CX3CR1 interaction prevents carbon tetrachloride-induced liver inflammation and fibrosis in mice. Hepatology 52, 1390–1400 (2010).

    Article  CAS  PubMed  Google Scholar 

  94. Thomas, J. A. et al. Macrophage therapy for murine liver fibrosis recruits host effector cells improving fibrosis, regeneration, and function. Hepatology 53, 2003–2015 (2011).

    Article  CAS  PubMed  Google Scholar 

  95. Jiao, J. et al. Dendritic cell regulation of carbon tetrachloride-induced murine liver fibrosis regression. Hepatology 55, 244–255 (2012).

    Article  CAS  PubMed  Google Scholar 

  96. Murphy, F. R. et al. Inhibition of apoptosis of activated hepatic stellate cells by tissue inhibitor of metalloproteinase-1 is mediated via effects on matrix metalloproteinase inhibition: implications for reversibility of liver fibrosis. J. Biol. Chem. 277, 11069–11076 (2002).

    Article  CAS  PubMed  Google Scholar 

  97. Elsharkawy, A. M., Oakley, F. & Mann, D. A. The role and regulation of hepatic stellate cell apoptosis in reversal of liver fibrosis. Apoptosis 10, 927–939 (2005).

    Article  CAS  PubMed  Google Scholar 

  98. Wright, M. C. et al. Gliotoxin stimulates the apoptosis of human and rat hepatic stellate cells and enhances the resolution of liver fibrosis in rats. Gastroenterology 121, 685–698 (2001).

    Article  CAS  PubMed  Google Scholar 

  99. Krizhanovsky, V. et al. Senescence of activated stellate cells limits liver fibrosis. Cell 134, 657–667 (2008). This study shows that senescent cells accumulate in mouse liver fibrosis and are mainly derived from activated hepatic stellate cells, and that these cells are killed by NK cells, which thereby facilitates the resolution of fibrosis.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  100. Kisseleva, T. et al. Myofibroblasts revert to an inactive phenotype during regression of liver fibrosis. Proc. Natl Acad. Sci. USA 109, 9448–9453 (2012).

    Article  PubMed  PubMed Central  Google Scholar 

  101. Troeger, J. S. et al. Deactivation of hepatic stellate cells during liver fibrosis resolution in mice. Gastroenterology 143, 1073–1083. e1022 (2012). References 100 and 101 show that reversion of hepatic stellate cell-derived myofibroblasts to an inactive intermediate phenotype accounts for about 50% of myofibroblast loss during the resolution of liver fibrosis.

    Article  CAS  PubMed  Google Scholar 

  102. Sakaida, I. et al. Transplantation of bone marrow cells reduces CCl4-induced liver fibrosis in mice. Hepatology 40, 1304–1311 (2004).

    Article  PubMed  Google Scholar 

  103. Syn, W. K. et al. Accumulation of natural killer T cells in progressive nonalcoholic fatty liver disease. Hepatology 51, 1998–2007 (2010).

    Article  CAS  PubMed  Google Scholar 

  104. Longhi, M. S. et al. Inhibition of interleukin-17 promotes differentiation of CD25 cells into stable T regulatory cells in patients with autoimmune hepatitis. Gastroenterology 142, 1526–1535. e1526 (2012).

    Article  CAS  PubMed  Google Scholar 

  105. Liu, X. et al. Genome-wide meta-analyses identify three loci associated with primary biliary cirrhosis. Nature Genet. 42, 658–660 (2010).

    Article  CAS  PubMed  Google Scholar 

  106. Liaskou, E. et al. Regulation of mucosal addressin cell adhesion molecule 1 expression in human and mice by vascular adhesion protein 1 amine oxidase activity. Hepatology 53, 661–672 (2011).

    Article  CAS  PubMed  Google Scholar 

  107. Fernández, M. et al. Angiogenesis in liver disease. J. Hepatol 50, 604–620 (2009).

    Article  CAS  PubMed  Google Scholar 

  108. Castaño, A. P. et al. Serum amyloid P inhibits fibrosis through Fc gamma R-dependent monocyte-macrophage regulation in vivo. Sci. Transl. Med. 1, 5ra13 (2009).

  109. Wasmuth, H. E. et al. Antifibrotic effects of CXCL9 and its receptor CXCR3 in livers of mice and humans. Gastroenterology 137, 309–319. e1–3 (2009).

    Article  CAS  PubMed  Google Scholar 

  110. Fallowfield, J. et al. Relaxin modulates human and rat hepatic myofibroblast function and ameliorates portal hypertension in vivo. Hepatology http://dx.doi.org/10.1002/hep.26627 (2013).

  111. Patsenker, E. et al. Cannabinoid receptor type I modulates alcohol-induced liver fibrosis. Mol. Med. 17, 1285–1294 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  112. Roderfeld, M. et al. Inhibition of hepatic fibrogenesis by matrix metalloproteinase-9 mutants in mice. FASEB J. 20, 444–454 (2006).

    Article  CAS  PubMed  Google Scholar 

  113. Ramachandran, P. & Iredale, J. P. Macrophages: central regulators of hepatic fibrogenesis and fibrosis resolution. J. Hepatol. 56, 1417–1419 (2012).

    Article  PubMed  Google Scholar 

Download references

Acknowledgements

J.A.F. is supported by grants from The Health Foundation, UK, Medical Research Council, UK, and the Wellcome Trust, UK. J.P.I. is supported by grants from the Medical Research Council, the Wellcome Trust and Children's Liver Disease Foundation, UK. A.P. and P.R. are supported by grants from the Medical Research Council and the Wellcome Trust, respectively.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Jonathan A. Fallowfield.

Ethics declarations

Competing interests

J.A.F. carries out consultancy work for Novartis, UK, and Conatus Pharmaceuticals, UK. J.P.I. carries out consultancy work for UCB Celltech, UK, and GE Healthcare, UK. A.P. and P.R. declare no competing interests.

PowerPoint slides

Glossary

Myofibroblast

Fibroblast-like cell of mesenchymal origin.

Idiopathic pulmonary fibrosis

(IPF). An interstitial lung disease of unknown aetiology that is characterized by a progressive fibrotic response driven by abnormally activated alveolar epithelial cells.

Kupffer cells

Cells of the resident hepatic macrophage population. In the steady state, Kupffer cells are thought to self-renew and to originate from fetal (yolk sac) precursor cells. These cells reside in the liver sinusoids where they regulate local immune responses and remove bacteria, bacterial endotoxins and microbial debris that are derived from the gastrointestinal tract and transported to the liver via the portal vein.

Fibrocytes

Cells of haematopoietic origin (marked by CD45 expression) that can differentiate into tissue myofibroblasts.

Pericytes

Specialized mesenchymal cells that are embedded within the basement membrane of capillaries (hepatic stellate cells are considered to be the pericytes of the liver).

Space of Disse

The perisinusoidal space in the liver, between the endothelial cells and the hepatocytes.

Integrins

A family of transmembrane receptors composed of non-covalently linked heterodimers of α-subunits and β-subunits that form at least 24 combinations in mammalian tissues. Several integrins, in particular β3 integrins, recognize a tripeptide arginine–glycine–aspartic acid (RGD) sequence in specific ECM ligands.

Cd11b–DTR transgenic mice

Transgenic mice that express the human diphtheria toxin receptor (DTR) under the control of the Cd11b promoter, which facilitates the selective depletion of monocytes and macrophages by the administration of diphtheria toxin.

Natural killer T cells

(NKT cells). A subset of T cells that have characteristics of both T cells and NK cells, expressing both a T cell receptor and NK lineage markers.

Innate-like invariant NKT cells

(iNKT cells). A subset of natural killer T cells that express a T cell receptor (TCR) with an invariant Vα14-Jα18 TCRα chain paired with a restricted subset of TCR Vβ chains. iNKT cells exclusively recognize glycolipid antigens that are presented on CD1d molecules.

γδ T cells

A subset of T cells that are defined by the genetic composition of their T cell receptor; the TCR is made up of one γ-chain and one δ-chain, rather than the one α-chain and one β-chain that are found in classical αβ T cells.

Innate lymphoid cells

Innate immune cells that produce many T helper cell-associated cytokines but do not express cell surface markers that are associated with other immune cell lineages. In addition, these cells do not express a T cell receptor and do not respond in an antigen-specific manner.

Senescent

A cellular state in which a growth arrest programme has been initiated that limits the lifespan of the cell and prevents unlimited cell proliferation.

Glitazones

Drugs used for the treatment of type 2 diabetes mellitus that function as agonists of peroxisome proliferator-activated receptor-γ (PPARγ) and that increase insulin sensitivity.

M1 macrophage

A macrophage that is activated by interferon-γ or by lipopolysaccharide; also known as a classically activated macrophage. These cells express pro-inflammatory cytokines and inducible nitric oxide synthase (among other things), and they inhibit cell proliferation and cause tissue damage.

M2 macrophage

A macrophage that is activated by the T helper 2 cell cytokines interleukin-4 (IL-4) and IL-13; also known as an alternatively activated macrophage. These cells express arginase 1, the mannose receptor CD206 and the IL-4 receptor α-chain (among other things), and they promote cell proliferation and tissue repair.

Rights and permissions

Reprints and permissions

About this article

Cite this article

Pellicoro, A., Ramachandran, P., Iredale, J. et al. Liver fibrosis and repair: immune regulation of wound healing in a solid organ. Nat Rev Immunol 14, 181–194 (2014). https://doi.org/10.1038/nri3623

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nri3623

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