119

Resolution of Liver Fibrosis: Basic Mechanisms and Clinical Relevance

1 The University of Edinburgh/ Medical Research Council (MRC) Centre

for Inflammation Research, Queen’s Medical Research Institute, Edinburgh, United Kingdom Semin Liver Dis 2015;35:119–131.

Abstract

Keywords

► matrix degradation ► myofibroblast fate ► proresolution macrophage ► antifibrotic therapy

Address for correspondence Jonathan A. Fallowfield, BSc, BM, MRCP, PhD, The University of Edinburgh/ Medical Research Council (MRC) Centre for Inflammation Research, Queen’s Medical Research Institute, 47 Little France Crescent, Room W2.30, Edinburgh, EH16 4TJ, United Kingdom (e-mail: [email protected]).

With evidence from a large number of animal models and clinical trials, it is now beyond debate that liver fibrosis and even cirrhosis are potentially reversible if the underlying cause can be successfully eliminated. However, in a significant proportion of patients cure of the underlying disease may not result in fibrosis regression or a significant reduction of the risk for hepatocellular carcinoma development. Understanding of the mechanistic pathways and regulatory factors that characterize matrix remodeling and architectural repair during fibrosis regression may provide therapeutic approaches to induce or accelerate regression as well as novel diagnostic tools. Recent seminal observations have determined that in resolving liver fibrosis a significant proportion of hepatic stellate cell-myofibroblasts (HSC-MFs) can revert to a near quiescent phenotype. Hepatic macrophages derived from inflammatory monocytes may contribute to fibrosis resolution through an in situ phenotypic switch mediated by phagocytosis. Emerging therapeutic approaches include deletion or inactivation of HSC-MFs, modulation of macrophage activity and autologous cell infusion therapies. Novel noninvasive diagnostic tests such as serum and imaging markers responsive to extracellular matrix degradation are being developed to evaluate the clinical efficacy of antifibrotic interventions.

Clinical Relevance Resolution of liver fibrosis, and even cirrhosis, is a wellestablished phenomenon that has not only been observed in models of liver fibrosis in rodents,1,2 but also in patients with chronic liver disease of diverse etiologies after cure of the underlying disease.3 The successful treatment of hepatitis B,4 hepatitis C,5 or hepatitis D6 with antivirals has been shown to lead to reversal of fibrosis, even in some patients with biopsy-proven cirrhosis. Similarly, venesection in hemochromatosis,7 chelation of copper in Wilson’s disease,8 immunomodulatory therapy in autoimmune hepatitis,9 bariatric surgery in nonalcoholic steatohepatitis (NASH),10 bone

Issue Theme Liver Fibrosis; Guest Editors, Robert Schwabe, MD, and Ramon Bataller, MD, PhD

marrow transplantation in thalassaemia,11 and reversal of biliary obstruction12 can also promote fibrosis regression. This is also observed in patients with alcohol-related liver disease after prolonged abstinence, yet specific studies are lacking. But it is recent large-scale trials in chronic viral hepatitis that have shown the most compelling results in terms of remodeling of fibrosis,13–15 such that regression of cirrhosis is no longer considered a lofty ambition or point of contention, rather it is part of the vernacular of contemporary hepatology, a rational goal of treatment, and an advertising strap-line for the new generation of antiviral drugs and surgical therapies for NASH. Critically, regression of fibrosis also appears to correlate with improved clinical outcomes. For

Copyright © 2015 by Thieme Medical Publishers, Inc., 333 Seventh Avenue, New York, NY 10001, USA. Tel: +1(212) 584-4662.

DOI http://dx.doi.org/ 10.1055/s-0035-1550057. ISSN 0272-8087.

This document was downloaded for personal use only. Unauthorized distribution is strictly prohibited.

Prakash Ramachandran, MBChB (Hons), BSc (Hons), MRCP, PhD1 John P. Iredale, BM (Hons), FMedSci, FRCP, FRSE1 Jonathan A. Fallowfield, BSc (Hons), BM (Hons), MRCP, PhD1

Resolution of Liver Fibrosis: Basic Mechanisms and Clinical Relevance example, in patients with hepatitis C virus- (HCV-) related cirrhosis treated with antivirals, “regressors” had better 10year survival rates than “nonregressors” (100% vs. 74%)16 (see also “Liver Fibrosis in the Post-HCV Era” by Pinzani in this issue). Moreover, studies investigating histological–hemodynamic associations in liver fibrosis have demonstrated that semiquantitative (small nodule size, thick fibrotic bands) and quantitative (collagen proportionate area) biopsy characteristics correlate with clinically significant portal hypertension.17–19 These studies have significant pathophysiological and clinical implications. In particular, they focus attention on linking the underlying biology of fibrosis with clinical expression of disease and identify hepatic venous pressure gradient (HVPG; or better, a noninvasive biomarker for HVPG) as a suitable measure to stratify and monitor progression or regression of fibrosis. However, robust long-term longitudinal data tracking fibrosis regression after successful treatment of chronic viral hepatitis is limited. A recent study in patients with HCV used the FibroSure (FibroTest) serum biomarker assay and showed that virological cure was associated with a modest long-term impact on fibrosis.20 After 10 years, 49% of SVR individuals with advanced baseline fibrosis had a significant improvement, but the net reduction of cirrhosis prevalence was only 5%. Disappointingly, there was a residual 5% risk of hepatocellular carcinoma (HCC) despite viral clearance. Clinical outcome data will continue to emerge from ongoing interventional studies in individuals with hepatic fibrosis due to chronic hepatitis B or NASH. Clearly, treatment of the underlying disease may not be sufficient for a proportion of patients and it is likely that as yet undefined environmental and genetic factors will influence this. Therefore, further understanding of molecular events that drive fibrosis regression is needed and may provide additional means of reducing longterm complications of chronic liver disease. In addition to clinical endpoints, antifibrotic trials are likely to require surrogate endpoints that reflect clinically meaningful benefit within shorter observation periods—their definition and validation (probably on a disease-specific basis) remains a priority for the field (see also “Diagnosis of Liver Fibrosis: Present and Future” by Patel, Bedossa, and Castera in this issue).

The Fate of Hepatic Myofibroblasts in Liver Fibrosis Resolution Liver fibrosis becomes problematic, and clinically relevant, when dysregulated and excessive scarring occurs in response to chronic injury and leads to altered tissue function. Although it is clear that the potential for regression of established liver fibrosis exists, for this to occur there must be cessation of active scar production and also degradation of the extracellular matrix (ECM) that has already been deposited to restore normal tissue architecture. As described in detail elsewhere in this issue, the principal cell type responsible for ECM deposition in the damaged liver is the activated HSC,21,22 which transdifferentiates into a MF phenotype following liver injury. In addition to ECM synthesis, activated Seminars in Liver Disease

Vol. 35

No. 2/2015

Ramachandran et al.

HSC-MFs have a range of additional profibrogenic properties,23 including immunomodulatory functions and a possible role in amplifying hepatic injury.24 Furthermore, hepatic MFs are the principal source of tissue inhibitors of metalloproteinases (TIMPs), in particular TIMP1 and to a lesser extent TIMP2.25–28 Tissue inhibitors of metalloproteinases are secreted into the extracellular milieu and bind noncovalently to the active domain and inhibit the group of matrix-degrading enzymes known as matrix metalloproteinases (MMPs), which are potentially capable of breaking down a variety of ECM components including fibrillar collagens. Importantly, even during progressive hepatic fibrosis a range of MMPs are expressed in the liver,29–32 demonstrating the inherent potential for scar degradation. However, TIMP expression by hepatic MFs inhibits this hepatic MMP activity, preventing scar degradation and producing a microenvironment favoring ECM deposition.27,33 The development of animal models of reversible liver fibrosis has enabled more detailed study of the role and fate of the HSC-MF during fibrosis regression. In rats, bile duct ligation results in a dense biliary fibrosis, with a restoration of near normal liver architecture following biliojejunal anastamosis.2 Similarly, following chronic CCl4 injury in rats, advanced liver fibrosis was remodeled to levels close to uninjured liver after the cessation of injury.1 What became clear from these initial studies is that resolution of fibrosis was associated with a profound loss of MFs1,2 from the receding hepatic scar. In addition to the loss of the ECM producing cells with a potential proinflammatory role, a decline in the MF number also removes the source of TIMPs. This has been confirmed in in vivo models, where hepatic MMP levels remained relatively unchanged and loss of TIMP1 during fibrosis resolution altered the overall MMP-TIMP balance resulting in increased hepatic collagenase activity and net ECM degradation.1,34 Indeed, in a transgenic murine model with persistent hepatic overexpression of TIMP1, there was a failure of remodeling of fibrosis even after the cessation of injury.35 Hence, the loss of activated HSCs represents a critical step for matrix degradation to occur. This has led to several pivotal studies in rodent models of fibrosis resolution investigating the fate of MFs and underlying mechanisms that could potentially be exploited therapeutically (►Fig. 1). Although the relevance of these findings will need to be confirmed in human disease, paired liver biopsies before and after long-term lamivudine treatment in patients with chronic hepatitis B did indeed demonstrate a significant reduction in α-SMA staining compared with placebo, indicating that a change in MF number or activation status was associated with regression of fibrosis.4

Myofibroblast Apoptosis Initial studies investigating MF fate demonstrated clear evidence of programmed cell death, or apoptosis, in these cells during fibrosis resolution.1,2 Subsequently, work has focused on the pathways that regulate MF apoptosis in liver.36 It has become apparent that proinflammatory signaling, acting via NF-κB activation in HSC-MFs, results in expression of profibrotic genes and promotes resistance to apoptosis.25,37 TIMP1

This document was downloaded for personal use only. Unauthorized distribution is strictly prohibited.

120

Ramachandran et al.

Fig. 1 Summary of the fate of hepatic myofibroblasts (MFs) during liver fibrosis regression. During fibrogenesis quiescent hepatic stellate cells (HSCs) become activated, adopting a MF phenotype and proliferate. During fibrosis regression these activated MFs can undergo apoptosis, induced by immune cell interactions, proapoptotic signals or loss of cell survival signals such as tissue inhibitor of metalloproteinase (TIMP). Activated MFs can also undergo senescence, induced by p53 signaling, IL-22, CCN1/CYR61, and as yet other undefined mechanisms. Recent data demonstrate that activated MFs can also revert to a quiescent HSC like phenotype, which is primed to respond to profibrogenic signals. The signals controlling this reversion process are undefined. Natural killer (NK) cells play an important role in killing activated and senescent MFs. (Adapted with permission from Pellicoro A, Ramachandran P, Iredale JP, Fallowfield JA. Liver fibrosis and repair: immune regulation of wound healing in a solid organ. Nat Rev Immunol 2014;14(3):181–194).

itself can act in an autocrine fashion on hepatic MFs to promote survival and inhibit apoptosis.38,39 Indeed, the interaction between activated HSC-MFs and ECM itself can modulate apoptosis, with collagen I binding promoting HSC activation while loss of integrin-mediated contact leads to HSC-MFs entering the apoptotic pathway.40 Further evidence for the importance of this interaction comes from hepatic injury in the r/r collagen mouse, in which collagen I is resistant to degradation. Liver fibrosis due to chronic CCl4 in these mice failed to resolve and activated HSC-MFs persisted adjacent to the scar tissue.41 A variety of signals have also been demonstrated to stimulate apoptosis in activated HSC-MFs. These include farnesoid X receptor (FXR or bile acid receptor), CEBP/β signaling, cannabinoid receptor 2 (CB2), adiponectin, and TRAIL (also known as TNFSF10).25 Nerve growth factor (NGF) can be released from infiltrating inflammatory cells or regenerating hepatocytes and is proapoptotic for activated HSC-MFs.42–44 Furthermore, direct cellular contact between macrophages and activated HSCs has been demonstrated to promote HSC death.45 The relative contribution of these various regulatory pathways in mediating HSC apoptosis in vivo remains to be elucidated, but it is likely that a complex cellular crosstalk exists that is impossible to recapitulate in vitro.

HSC-MFs in rodent and human liver fibrosis express senescence markers.46 Inhibition of cellular senescence in HSC-MFs using cell-specific p53 knockout mice resulted in increased HSC proliferation and exacerbated liver fibrosis in response to CCl4. Additionally, following the cessation of CCl4, p53 deficiency resulted in persistence of activated HSC-MFs and prevented scar resolution, suggesting that a HSC-MF senescence program is important in the removal of activated MFs during liver fibrosis resolution. Further, in vitro characterization of senescent hepatic MFs demonstrated that senescence resulted in reduced expression of ECM components, increased expression of MMPs, and upregulation of genes involved in immune surveillance, which could potentially aid the clearance of senescent HSC-MFs by NK cells.46 These findings have led authors to investigate the signals inducing MF senescence. CCN1/CYR61, an ECM-associated signaling protein, is one such molecule that has been shown to induce MF senescence and have an antifibrotic effect in vivo.47,48 Additionally, IL-22 can induce HSC-MF senescence and accelerate liver fibrosis resolution.49 Intriguingly, a recent report has suggested that atorvastatin treatment can induce HSC-MF senescence,50 consistent with studies suggesting an antifibrotic effect in humans.51 These findings clearly merit further study.

Myofibroblast Reversion Myofibroblast Senescence Senescence is a stable form of cell-cycle arrest, which has mostly been investigated in the context of malignant disease. Interestingly, it has now been identified that a proportion of

More recently, the development of transgenic mice has enabled lineage tracing of activated MFs and shown that they can also revert to a near-quiescent phenotype following the cessation of injury.52,53 Kisseleva et al utilized transgenic Seminars in Liver Disease

Vol. 35

No. 2/2015

121

This document was downloaded for personal use only. Unauthorized distribution is strictly prohibited.

Resolution of Liver Fibrosis: Basic Mechanisms and Clinical Relevance

Resolution of Liver Fibrosis: Basic Mechanisms and Clinical Relevance mice with fluorescently labeled cells that had expressed collagen I during their lifetime. Using this system, they induced liver fibrosis with CCl4 or alcohol followed by a period of fibrosis resolution. After protracted recovery, when no significant residual liver fibrosis was present, fluorescent quiescent HSCs were detected, indicating an origin from a previously activated MF population.52 Similarly, Troeger et al used transgenic mice to fluorescently label hepatic MFs during CCl4 liver injury and also identified fluorescent quiescent HSCs following fibrosis resolution. The authors estimated that 40% of HSCs derived from reversion to a quiescent phenotype,53 similar to the proportion identified by Kisseleva.52 Both groups also determined that reverted HSCs were not identical to pre-injury quiescent HSCs, with reverted HSCs remaining in a “primed” state and demonstrating an augmented response to further fibrogenic stimuli.52,53 Although the signals causing MF reversion rather than apoptosis and the factors responsible for the maintenance of this inactivated but primed HSC population remain to be elucidated, these findings may have major implications for human liver fibrosis, where periods of repetitive tissue injury are a common feature (e.g., in alcoholrelated liver disease).

Regulation of Fibrosis Resolution by Immune Cells As discussed, the activation and subsequent fate of HSC-MFs are key determinants of liver fibrosis progression and regression. There is now compelling evidence supporting the role of different immune cell populations in promoting fibrosis resolution.

The Role of the Macrophage The role of cells of the monocyte/macrophage lineage in hepatic fibrogenesis and fibrosis resolution has been most widely studied (►Fig. 2). This is perhaps unsurprising, given that a significant proportion of hepatic macrophages are found in very close proximity to MFs and scar tissue following chronic hepatic injury (so-called scar-associated macrophages [SAMs]).54–56 Investigators have used different macrophage depletion strategies in rodent models of liver fibrosis including gadolinium chloride,57 liposomal clodronate,58,59 or transgenic CD11b-DTR mice (where administration of diphtheria toxin results in depletion of CD11b-expressing macrophages),54 and consistently shown that removal of hepatic macrophages during ongoing injury results in reduced liver fibrosis. Macrophages are potent producers of proinflammatory and profibrotic mediators and this injury promoting effect may well be mediated through interactions with HSC-MFs. Specifically, macrophages can produce the archetypal profibrogenic cytokine TGFβ, which acts on MFs to promote activation and ECM synthesis.60,61 Additionally, they can express high levels of thrombospondin-1, a potent activator of TGFβ in vivo.56 Macrophages are also a potential source of other soluble mediators including TNF-α and IL-1β (which act on activated HSCs to induce NF-κB activity, promote survival, and increase liver fibrosis59), plateletSeminars in Liver Disease

Vol. 35

No. 2/2015

Ramachandran et al.

derived growth factor (which can induce MF proliferation), IL-4 and IL-13 (which can directly stimulate collagen synthesis in fibroblasts), CCL7 and CCL8 (which can recruit MFs), and a range of other chemokines that can promote further leucocyte migration and perpetuate the inflammatory response.61 Galectin-3 is a macrophage-derived lectin that has been shown to promote MF activation in both liver and kidney fibrosis models62,63 and whose inhibition has an antifibrotic effect.64 Such profibrogenic mediators are also produced by human monocytes/macrophages derived from patients with chronic liver disease.65–67 Despite this array of profibrogenic capabilities, intriguing data also describe a role for macrophages in the resolution of liver fibrosis (►Fig. 2). Depletion of macrophages during the resolution of liver fibrosis using the aforementioned CD11bDTR mice54 or MAFIA (CSF1R-FKB12) mice68 demonstrated a clear failure of matrix degradation. Similar observations have been made during the resolution of murine pulmonary fibrosis.69 The mechanisms underpinning these exciting findings are the subject of active investigation. What is clear is that macrophages, in particular SAMs, are a key source of matrix-degrading MMPs, including MMP1355 and MMP12.70 This macrophage MMP expression is upregulated by phagocytosis of dead cells,56,71 a major function of this cell type in inflamed tissue. Furthermore, macrophages are a potential source of molecules such as MMP956 and TRAIL,45 which can promote MF apoptosis. To understand the apparently dichotomous effects of macrophages in liver fibrosis, it is important to appreciate the potential heterogeneity of macrophages. It is now apparent that the traditional M1/M2 classification is wholly inadequate to describe in vivo macrophage phenotypes.56,72 Rather, a classification on the basis of ontogeny and function is more likely to be useful. In the liver, resident Kupffer cells make up the majority of hepatic macrophages during the steady state; indeed they are the largest macrophage pool in the entire body.73 A distinct population of hepatic monocyte-derived macrophages can also be identified.56,73–75 Although there is a longstanding debate as to the origin of these distinct populations, a recent series of lineage tracing murine models have demonstrated that under steady state, resident Kupffer cells derive from yolk sac macrophages and are maintained via local proliferation with no meaningful contribution from recruited monocytes.76–78 The precise function of these distinct resident macrophages following hepatic injury remains to be defined. However, following injury there is a massive expansion of the total hepatic macrophage pool, predominantly via recruitment of circulating monocytes.73 Further studies have identified a subset of this population, expressing high levels of the marker Ly6C and recruited via the CCR2-CCL2 axis, as being a principal driver of hepatic fibrogenesis.60,79,80 These Ly-6Chi monocyte-derived cells also have a profibrotic role in lung69 and renal injury.81 Our group went on to identify a population of Ly-6Clo hepatic monocyte-derived macrophages, termed the “restorative macrophage,” which was responsible for murine liver fibrosis resolution following chronic CCl4 injury.56 These cells,

This document was downloaded for personal use only. Unauthorized distribution is strictly prohibited.

122

Ramachandran et al.

Fig. 2 Schematic showing the proposed role of macrophages in hepatic fibrogenesis and fibrosis regression. In response to hepatic injury, inflammatory monocytes are recruited to the liver and form profibrotic Ly-6Chi macrophages in the liver. Ly-6C hi macrophages can express proinflammatory cytokines (IL-1β and TNF-α), which promote hepatocellular damage and myofibroblast (MF) survival, chemokines that promote recruitment of inflammatory cells/monocytes (CCL2, CXCL10) and MFs (CCL7, CCL8), TGFβ and galectin-3 that promote activation of hepatic stellate cells (HSCs) into a MF phenotype, thrombospondin-1 (thbs1) that can activate latent TGFβ, and platelet-derived growth factor (PDGF) that promotes MF proliferation. Activated MFs proliferate and secrete extracellular matrix (resulting in scar formation) and TIMP1, which inhibits both matrix-degradation by matrix metalloproteinases (MMPs) and HSC apoptosis. Within the liver these profibrogenic macrophages then change phenotype, at least partly in response to phagocytosis of cellular debris. This results in the generation of Ly-6Clo restorative hepatic macrophages, which mediate fibrosis regression. These restorative macrophages upregulate opsonins and phagocytosis receptors, enhancing the clearance of cellular debris, and hence promoting the generation of further restorative macrophages. Ly-6C lo macrophages downregulate proinflammatory cytokine expression promoting MF apoptosis, may express proapoptotic mediators such as MMP9 and TRAIL, and upregulate matrix-degrading enzymes such as MMP12, MMP13, and MMP9, which in combination with falling TIMP1 levels, results in increased hepatic matrix degrading activity and hence scar degradation. Restorative macrophage can also express Arginase-1, anti-inflammatory receptors (CX3CR1), proresolution chemokines such as CXCL9 and growth factors such as IGF-1, which may promote hepatocyte regeneration. Vascular endothelial growth factor has a dual role, promoting monocyte recruitment but also proresolution features in hepatic macrophages. (Adapted with permission from Pellicoro A, Ramachandran P, Iredale JP, Fallowfield JA. Liver fibrosis and repair: immune regulation of wound healing in a solid organ. Nat Rev Immunol 2014;14(3):181–194.)

which accumulate maximally in livers at the time of scar degradation, are derived from a phenotypic switch of profibrogenic Ly-6Chi macrophages potentially induced by macrophage phagocytosis, resulting in downregulation of proinflammatory cytokines and chemokines and increased expression of MMPs and other antifibrotic genes such as CX3CR182,83 and arginase-1.61 The fact that the proresolution macrophage subset has the same origin as the profibrogenic subset is supported by data from transgenic CCR2 deficient mice, which although partially protected from liver fibrogenesis are also unable to degrade the accumulated scar.80 Recent findings by Baeck et al shed further light on this process.84 Using a Spiegelmer technique, they inhibited CCL2 in vivo during the resolution of murine liver fibrosis from either CCl4 or MCD diet (a murine model of NASH). This intervention caused a reduction in profibrogenic Ly-6Chi monocyte-derived macrophages and an increase in restor-

ative Ly-6Clo macrophages and resulted in accelerated fibrosis resolution.84 These findings mirror our own, where the administration of liposomes during fibrosis resolution induced phagocytic behavior and caused a very similar change in hepatic macrophage subsets with a consequent increase in matrix degradation.56 Hence, it seems to be the balance of profibrotic and restorative macrophages that governs the overall tissue phenotype and a greater understanding of the mechanisms controlling this could yield novel therapeutic targets. A recent insight into this has been provided by Yang et al.68 Vascular endothelial growth factor (VEGF) has previously been thought to be a profibrogenic molecule85 and inhibition of VEGF has been suggested as a potential therapy for fibrosis.86 However, blockade of VEGF during resolution from murine BDL or CCl4 liver fibrosis prevented fibrosis regression while overexpression of VEGF accelerated it.68 The beneficial effect of VEGF during fibrosis resolution seemed to Seminars in Liver Disease

Vol. 35

No. 2/2015

123

This document was downloaded for personal use only. Unauthorized distribution is strictly prohibited.

Resolution of Liver Fibrosis: Basic Mechanisms and Clinical Relevance

Resolution of Liver Fibrosis: Basic Mechanisms and Clinical Relevance be mediated via effects on sinusoidal endothelial permeability affecting monocyte recruitment and also direct stimulation of SAMs to produce MMP13 and the antifibrotic chemokine CXCL9.68,87 This apparent dual role of VEGF in fibrogenesis and fibrosis resolution resembles that seen with macrophages overall and highlights an important point when considering translational strategies: namely that caution needs to be exercised when identifying antifibrotic targets as these molecules may also have a key role in fibrosis resolution.88 What remains to be seen is how closely this murine macrophage heterogeneity relates to human disease. Similar to murine Kupffer cells, a population of “resident” macrophages can be identified in human cirrhotic liver as CD14l ° CD16, while human hepatic monocyte-derived macrophages could be defined as CD14hiCD16 and CD14þCD16þ subsets.65,66 Interestingly, analogous to murine data on the restorative macrophage, CD14þCD16þ macrophages can derive from CD14hiCD16 cells and have a high phagocytic capacity,66 but this same population also expresses high levels of proinflammatory mediators and can directly activate HSCs.65,66 It may well be that further, as yet undefined, heterogeneity exists within these macrophage subpopulations in human liver.

Other Immune Cells Several other immune cell populations have been studied in relation to a role in hepatic fibrosis, albeit in less depth than monocytes/macrophages. We will briefly summarize the data suggesting an antifibrotic role for these cell types.

Dendritic Cells Dendritic cells (DCs) may mediate ECM degradation during liver fibrosis resolution, potentially via MMP9 expression.89 These findings merit further investigation, but a note of caution is that the distinction between macrophages and DCs is not always clear cut, particularly in the context of inflammation.90

Neutrophils

Despite being an early responder to hepatic injury,91 no clear role for neutrophils has been identified in hepatic fibrogenesis.92 Some authors have proposed a potential role for neutrophil-derived MMPs in fibrosis resolution,93 but it is likely that neutrophils are not present in livers in sufficient numbers for this to be a meaningful effect during fibrosis resolution.

Natural Killer Cells Natural killer (NK) cells have been studied in liver fibrosis and have been demonstrated to have an antifibrotic effect (►Fig. 1).94–96 This seems to be mediated via NK cell-mediated killing of activated HSCs as a result of IFN-γ expression and/or expression of death receptors or ligands such as NKG2D,94 TRAIL97 or FasL.98 Additionally, NK cells have a key role in the clearance of senescent HSCs, with NK cell depletion causing persistent senescent HSCs in vivo and a failure of scar resolution, while activation of NK cells accelerSeminars in Liver Disease

Vol. 35

No. 2/2015

Ramachandran et al.

ated removal of senescent HSCs and augmented fibrosis resolution.46

T Cells T cells play a central role in the adaptive immune response, with a variety of subtypes having differing immunomodulatory roles. T helper cells (TH cells) become activated by interactions with antigen presenting cells (APCs) and secrete cytokines that influence other immune cells. The cytokine profile expressed by the TH cells is dependent on the nature of the signaling from the APC. In liver fibrosis, the TH2 subtype, expressing IL-4, IL-5, IL-13, and IL-21, has been shown to be strongly profibrogenic.99 Conversely, a TH1 immune response is associated with IFN-γ and IL-12 expression, which have been shown to be antifibrotic.99 Thus, the relative balance of TH1 and TH2 cells may influence the degree of fibrosis progression.100 This is likely to be an overly simplistic view as our understanding of the complexities of T cell responses increases. Regulatory T cells (Treg) for example, are a specific subset that has been associated with an antifibrotic effect in animal models101 and human liver disease.102

γδ T Cells γδ T cells are a specific subpopulation of nonconventional T cells that share features of innate immune cells and conventional T cells, the liver being one of the richest sources of this subpopulation in the body. A recent study has identified that these γδ T cells, recruited to the liver via CCR6, have an antifibrotic effect potentially via the induction of HSC apoptosis in a FasL-dependent manner.103

Is Liver Fibrosis Always Reversible? Having discussed data demonstrating that liver fibrosis is reversible and some of the underlying mechanisms, a key question is whether all liver fibrosis is reversible, and if not, what factors limit ECM degradation? Critically, cirrhosis is not simply advanced fibrosis, but also encompasses architectural disruption, nodular hepatocyte regeneration, and vascular changes including angiogenesis. Although rodent models have been invaluable in defining the pathophysiology of liver fibrosis, it is worth noting that liver fibrosis is less reversible in humans, probably as a result of the dense fibrosis that typically develops over decades (rather than weeks in rodents) and the presence of prominent angioarchitectural changes such as vascularized septae that are probably irreversible.104 Indeed, the fibrotic response to chronic CCl4 in mice is weak and resolves very rapidly after withdrawal of the toxin. Outbred rats (e.g., Sprague-Dawley) exhibit a more robust fibrotic reaction and it is possible to generate cirrhosis.34 Chronic thioacetamide treatment in mice also appears to induce cirrhosis that does not reverse.105 In cirrhotic rats, although significant matrix remodeling occurred during recovery (particularly areas of recently deposited scar), fibrotic septae failed to completely regress even after 1 year, resulting in an attenuated macronodular pattern34 similar to that described in the human cirrhosis specimens by Wanless and colleagues.106 Close scrutiny of residual (compared

This document was downloaded for personal use only. Unauthorized distribution is strictly prohibited.

124

with remodeled) ECM in this model showed that persistent scars were heavily crosslinked (potentially mediated by tissue transglutaminase or lysyl oxidase enzymes), rich in elastin, and were relatively hypocellular (lacking in MFs and inflammatory cells capable of secreting proteases).34 It is unlikely that any of these mechanisms operate in isolation. For example, tissue transglutaminase seems dispensable for ECM crosslinking in murine liver fibrosis,105 while lysyl oxidase has immunomodulatory functions in addition to effects on ECM crosslinking.107 It is likely that a combination of ECM modifications, in addition to physical factors (e.g., deeper areas of dense fibrous scars may simply be inaccessible to MMPs) confer resilience to established hepatic scar tissue. The differences between liver fibrosis in mice and men may partly explain why translation of efficacious preclinical antifibrotic candidate drugs (such as INFγ or glitazones) has not always proven to be as successful in humans.108,109 However, while entirely normal liver architecture may never be fully restored, even limited matrix remodeling may be sufficient to reduce portal pressure, risk of HCC, and improve patient outcomes. Susceptibility to fibrosis regression may also differ with etiology. Moreover, etiological variation in the pattern of fibrosis distribution (i.e., postnecrotic, biliary, centrilobular, or perisinusoidal) influences the degree of associated angiogenesis, which in turn may increase the rate of disease progression to cirrhosis and limit the potential for fibrosis regression.110 Interestingly, at the time of transplantation, cirrhotic livers were shown to have different ranges of collagen proportionate area (CPA), according to etiology (e.g., median CPA in alcohol-related cirrhosis 30% compared with 16.5% in HBV-related cirrhosis).111 However, although qualitative and quantitative differences have been identified in human biopsy samples, we need better tissue markers of regressing fibrosis and “the point of no return” to distinguish patients who may reverse fibrosis (e.g., in response to disease-specific or antifibrotic drug therapy) from those who probably will not.

Resolution of Fibrosis and Liver Regeneration In contrast to other adult tissues, the liver has a remarkable regenerative capacity—a phenomenon observed in all vertebrate organisms. However, epithelial cell proliferation is severely blunted in human chronic liver disease due to several mechanisms including hepatocyte cell cycle arrest, upregulated p21 expression, and senescence.112 As a result many patients with end-stage cirrhosis will require a liver transplant. For functional restitution to occur, resolution of liver fibrosis must also initiate a robust regenerative response. Studies have shown a critical link between ECM degradation and hepatic epithelial regeneration. In the aforementioned study by Issa et al, mice bearing a mutated collagen I gene (r/r mice), which confers resistance to collagenase degradation, demonstrated less fibrosis regression, but also a blunted hepatocyte regenerative response compared with wild-type controls.41 Furthermore, using extracted collagen I from each

Ramachandran et al.

genotype as culture substrata, wild-type collagen I promoted hepatocyte proliferation via stimulation of integrin αvβ3.41 In advanced liver damage, hepatic regeneration can also occur through proliferation of a transit-amplifying population of “facultative” resident hepatic progenitor cells (HPCs), located within a stereotypical niche in close association with MFs and bone marrow (BM) derived macrophages.113 It was shown using r/r mice and MMP13 knockout mice that failure of ECM remodeling in experimental fibrosis hindered the ability of the liver to activate HPCs.114 HPCs are bipotential (i.e., can differentiate into hepatocytes or cholangiocytes) and subsequent HPC fate is determined by differential signaling cascades and underlying disease etiology.115 In fact, hepatic macrophages may also play a key role in epithelial regeneration, by expression of molecules such as TWEAK to promote HPC proliferation,116 Wnt3a to promote HPC differentiation to hepatocytes,115 and growth factors such as IGF-156 that can act directly on hepatocytes to stimulate proliferation and survival (►Fig. 2).117,118 Finally, the recent finding in mice that stimulation of 5-HT2B serotonin receptors on activated HSCs inhibited hepatocyte regeneration via increased TGFβ production119 indicates that, in addition to fibrosis resolution, exploring the relationship between cellular regeneration and fibrogenesis in the liver could also identify new therapeutic approaches as an alternative to liver transplantation.

Regression of Fibrosis: Diagnostic and Therapeutic Applications Our increasing knowledge of the underlying mechanisms and natural history of liver fibrosis/cirrhosis resolution offers opportunities to exploit these insights to develop new diagnostic tests to track disease regression and new treatments that directly promote liver repair.

Diagnostic Approaches for Monitoring Fibrosis Resolution As described by Patel et al in this issue, liver biopsy for the assessment of fibrosis has several limitations. These make it unsuitable for monitoring regression of disease in response to etiology-specific or antifibrotic treatments where large sample sizes and prolonged study duration would be required. Although currently available noninvasive serum and imaging (e.g., FibroScan) markers of hepatic fibrosis have demonstrated acceptable performance for the diagnosis of advanced versus no/mild fibrosis, they are unlikely to be accurate enough to evaluate the subtle changes expected from the pharmacological treatment of fibrosis, especially at earlier stages of disease. Furthermore, no consensus exists on what minimum change in serum marker level defines a clinically important effect and there are very limited data on the use of elastography techniques (transient or magnetic resonance imaging [MRI]) to monitor fibrosis regression. 120 The availability of a quantitative noninvasive serum or imaging biomarker, especially a dynamic measure of fibrogenesis or fibrolysis, would circumvent many of these issues. Seminars in Liver Disease

Vol. 35

No. 2/2015

125

This document was downloaded for personal use only. Unauthorized distribution is strictly prohibited.

Resolution of Liver Fibrosis: Basic Mechanisms and Clinical Relevance

Resolution of Liver Fibrosis: Basic Mechanisms and Clinical Relevance One approach that may hold promise is the detection of serum protein markers that reflect connective tissue formation and degradation (turnover). When specific MMPs degrade ECM components characteristic neo-epitopes are exposed at the site of degradation and these can be detected in serum by enzyme-linked immunosorbent assay (ELISA). Serum levels of specific ECM markers have been shown to correlate with levels of hepatic fibrosis in CCl4-treated rats.121 Furthermore, a combination of two serum ECM markers and the Model for End Stage Liver Disease (MELD) score correlated significantly with HVPG in patients with alcohol-related cirrhosis.122 These protein fingerprints, directly associated with ECM turnover, may change more rapidly than other measures of liver fibrosis (e.g., biopsy collagen, imaging stiffness) and thus could be utilized as early diagnostic, prognostic, and treatment-monitoring markers. Novel dynamic imaging methods that employ a small molecular ligand for ECM components or a cell-associated molecule coupled to a radioimaging or MRI agent could provide a sensitive indicator of change in fibrosis over the whole liver and permit early assessment of antifibrogenic effect in therapeutic trials. Gadolinium-based MRI probes targeted to collagen I123 or fibronectin124 have shown promise. The use of molecular tracers that bind to cell surface receptors overexpressed in the fibrotic liver (such as αvβ6 on activated cholangiocytes125 or serotonin receptors on HSCMFs126) is an attractive approach, but signal-to-noise ratio could be a limiting factor.

Therapeutic Application A comprehensive review of potential antifibrotic therapies is discussed in detail in the article “Antifibrotic Therapies in the

Ramachandran et al.

Liver” by Mehal and Schuppan in this issue. A variety of promising drug therapies have been shown to directly target HSC-MFs or accumulated scar ECM (►Table 1). Combined development of drug-targeting methodology could enable an oncology-type approach to deliver potent drugs selectively to the fibrotic liver in a cell-specific manner. Alternatively, cell infusion approaches could potentially induce matrix degrading and/or pro-regenerative effects in chronic liver disease (►Table 1).127 There are some concerns regarding the use of autologous unsorted bone marrow cell (BMC) infusions for the treatment of liver fibrosis as the BMCs contain mesenchymal stem cells that can differentiate into MFs in certain settings. A better strategy might be to use macrophages, hematopoietic stem cells, or BM mononuclear cells. In particular, macrophages are biologically well-defined and can be generated in large numbers from blood monocytes. Injected autologous macrophages have been shown to be antifibrotic, anti-inflammatory, and stimulate liver regeneration in models of liver injury and fibrosis.128 Several hurdles remain with respect to clinical translation of emerging antifibrotic therapies. First, standard operating procedures for the best preclinical models for proof-ofconcept studies are long overdue. History indicates that inappropriate selection of preclinical models may lead to future disappointment in clinical trials.129 In addition, antifibrotic drug development is currently hindered by the long and usually asymptomatic natural history of most fibrotic liver diseases, varying clinical phenotypes that have not been defined (e.g., the small % of patients with nonalcoholic fatty liver disease who progress to NASH), lack of well-defined and validated endpoints (both for cirrhotic and noncirrhotic trial populations), and lack of data to support the Food and Drug

Table 1 Potential antifibrotic therapies that target fibrosis regression or liver regeneration Promote apoptosis, quiescence, or senescence of hepatic myofibroblasts • Sulfasalazine or sulfasalazine analogs130 • Cannabinoid receptor (CB1) antagonists131 • TIMP1 blockade using siRNA,132 neutralizing antibody133 or inactive MMP sump134,135 • Hepatic stellate cell-myofibroblast specific targeted therapies o Anti-EGFR single chain fragment variable antibody-TRAIL fusion protein136 o Vitamin A-coupled liposomes to deliver siRNA against a collagen-specific chaperone137 o Single chain antibody (C1–3) targeted gliotoxin138 • PPARγ agonism or derepression139 • FXR agonism140,141 • Atorvastatin50,51 Stimulate degradation of accumulated scar ECM • TIMP1 blockade using siRNA,132 neutralizing antibody133 or inactive MMP sump134,135 • MMP gene therapy142,143 • Halofuginone144 • Lysyl oxidase (LoxL2) inhibitors107 • Cell therapies o Autologous granulocyte colony stimulating factor (G-CSF) mobilized CD133þ bone marrow stem cells145 o Autologous CD14þ monocyte-derived macrophages146 Target the immune response • Blockade of CCL2/CCR284 • Enhance restorative macrophages by liposome administration56 • Modulation of CXCL9/CXCR3 axis87,147 Abbreviations: ECM, extracellular matrix; EGFR, epidermal growth factor receptor; FXR, farnesoid X receptor; MMP, Matrix metalloproteinase; PPAR, peroxisome proliferator-activated receptor. Seminars in Liver Disease

Vol. 35

No. 2/2015

This document was downloaded for personal use only. Unauthorized distribution is strictly prohibited.

126

Administration’s “reasonably likely to predict clinical benefit” standard of surrogate endpoints. Although trial populations with more advanced stage disease have higher clinical event rates (4%/y), such hard endpoints may not be suitable for some antifibrotic drug targets (e.g., metabolic therapies in NASH). Moreover, fibrosis in patients with established cirrhosis will inherently be less reversible. Until these issues are resolved, translation (and prioritization) of antifibrotic candidates will be challenging. In the future, selection and stratification of trial participants should be based on those most likely to progress/respond. The “ideal” antifibrotic treatment would be potent yet specific, easily administered, and safe/well-tolerated as most patients are likely to be asymptomatic. In particular, treatment in cirrhotic patients should not increase risk of HCC.

Flt3 HCC HCV HPC HSC-MF HVPG IGF-1 MCD MMP NASH NK cell SAM SVR TIMP TRAIL

Conclusion

TWEAK

As we have discussed above, there is now compelling evidence from patients and rodent models that significant remodeling of even relatively advanced liver fibrosis is possible with net matrix degradation, a loss of the activated MF physically or functionally, and restitution of more normal liver architecture. Very recent data suggests that macrophages, recruited in the inflammatory phase but remaining in situ, orchestrate the resolution of fibrosis and may directly influence HPC proliferation and promote their specification to hepatocytes, thereby repopulating organ parenchyma. These recent steps forward in our understanding of the resolution of liver fibrosis pave the way to the design of novel treatments based on drugs, small molecules, and arguably, cell-based therapies. At the same time, research into fibrogenesis and fibrosis resolution has for the first time generated a panoply of surrogate markers of fibrosis and fibrosis degradation, which will facilitate the future evaluation of these new treatments.

VEGF Wnt3a YFP

Abbreviations α-SMA ANIT BDL BM CCl4 CCL2 CCL7 CCL8 CCN1/CYR61 CCR2 CCR6 Cd11b Cd11c CX3CR1 CXCL9 DTR ECM FasL

α-smooth muscle actin alpha-naphthylisothiocyanate bile duct ligation bone marrow carbon tetrachloride chemokine (C-C motif) Ligand 2 (MCP-1) chemokine (C-C motif) ligand 7 (MCP-3) chemokine (C-C motif) ligand 8 (MCP-2) cysteine rich protein 61 chemokine (C-C motif) receptor 2 chemokine (C-C motif) receptor 6 integrin αm integrin αx chemokine (C-X3-C motif) receptor 1 (fractalkine receptor) chemokine (C-X-C motif) ligand 9 human diphtheria toxin receptor (DTR) extracellular matrix Fas ligand (TNF superfamily, member 6)

Ramachandran et al.

FMS-like tyrosine kinase 3 hepatocellular carcinoma hepatitis C virus hepatic progenitor cell hepatic stellate cell-myofibroblast hepatic venous pressure gradient insulin-like growth factor 1 methionine choline deficient diet matrix metalloproteinase nonalcoholic steatohepatitis natural killer cell scar-associated macrophage sustained virological response tissue inhibitor of metalloproteinase tumor necrosis factor (ligand) superfamily, member 10 (Tnfsf10) tumor necrosis factor (ligand) superfamily, member 12 vascular endothelial growth factor drosophila melanogaster wingless 3a yellow fluorescent protein

References 1 Iredale JP, Benyon RC, Pickering J, et al. Mechanisms of spontane-

2

3 4

5

6

7

8

9 10

11

12

ous resolution of rat liver fibrosis. Hepatic stellate cell apoptosis and reduced hepatic expression of metalloproteinase inhibitors. J Clin Invest 1998;102(3):538–549 Issa R, Williams E, Trim N, et al. Apoptosis of hepatic stellate cells: involvement in resolution of biliary fibrosis and regulation by soluble growth factors. Gut 2001;48(4):548–557 Ellis EL, Mann DA. Clinical evidence for the regression of liver fibrosis. J Hepatol 2012;56(5):1171–1180 Kweon YO, Goodman ZD, Dienstag JL, et al. Decreasing fibrogenesis: an immunohistochemical study of paired liver biopsies following lamivudine therapy for chronic hepatitis B. J Hepatol 2001;35(6):749–755 Poynard T, McHutchison J, Manns M, et al. Impact of pegylated interferon alfa-2b and ribavirin on liver fibrosis in patients with chronic hepatitis C. Gastroenterology 2002;122(5): 1303–1313 Farci P, Roskams T, Chessa L, et al. Long-term benefit of interferon alpha therapy of chronic hepatitis D: regression of advanced hepatic fibrosis. Gastroenterology 2004;126(7):1740–1749 Niederau C, Fischer R, Pürschel A, Stremmel W, Häussinger D, Strohmeyer G. Long-term survival in patients with hereditary hemochromatosis. Gastroenterology 1996;110(4):1107–1119 Cope-Yokoyama S, Finegold MJ, Sturniolo GC, et al. Wilson disease: histopathological correlations with treatment on follow-up liver biopsies. World J Gastroenterol 2010;16(12): 1487–1494 Czaja AJ, Carpenter HA. Decreased fibrosis during corticosteroid therapy of autoimmune hepatitis. J Hepatol 2004;40(4):646–652 Dixon JB, Bhathal PS, Hughes NR, O’Brien PE. Nonalcoholic fatty liver disease: Improvement in liver histological analysis with weight loss. Hepatology 2004;39(6):1647–1654 Muretto P, Angelucci E, Lucarelli G. Reversibility of cirrhosis in patients cured of thalassemia by bone marrow transplantation. Ann Intern Med 2002;136(9):667–672 Hammel P, Couvelard A, O’Toole D, et al. Regression of liver fibrosis after biliary drainage in patients with chronic

Seminars in Liver Disease

Vol. 35

No. 2/2015

127

This document was downloaded for personal use only. Unauthorized distribution is strictly prohibited.

Resolution of Liver Fibrosis: Basic Mechanisms and Clinical Relevance

Resolution of Liver Fibrosis: Basic Mechanisms and Clinical Relevance

13

14

15

16

17

18

19

20

21 22 23 24

25 26

27

28 29

30

31 32

33

34

pancreatitis and stenosis of the common bile duct. N Engl J Med 2001;344(6):418–423 Marcellin P, Gane E, Buti M, et al. Regression of cirrhosis during treatment with tenofovir disoproxil fumarate for chronic hepatitis B: a 5-year open-label follow-up study. Lancet 2013; 381(9865):468–475 Chang TT, Liaw YF, Wu SS, et al. Long-term entecavir therapy results in the reversal of fibrosis/cirrhosis and continued histological improvement in patients with chronic hepatitis B. Hepatology 2010;52(3):886–893 Shiffman ML, Sterling RK, Contos M, et al. Long term changes in liver histology following treatment of chronic hepatitis C virus. Ann Hepatol 2014;13(4):340–349 Mallet V, Gilgenkrantz H, Serpaggi J, et al. Brief communication: the relationship of regression of cirrhosis to outcome in chronic hepatitis C. Ann Intern Med 2008;149(6):399–403 Nagula S, Jain D, Groszmann RJ, Garcia-Tsao G. Histologicalhemodynamic correlation in cirrhosis-a histological classification of the severity of cirrhosis. J Hepatol 2006;44(1):111–117 Sethasine S, Jain D, Groszmann RJ, Garcia-Tsao G. Quantitative histological-hemodynamic correlations in cirrhosis. Hepatology 2012;55(4):1146–1153 Calvaruso V, Burroughs AK, Standish R, et al. Computer-assisted image analysis of liver collagen: relationship to Ishak scoring and hepatic venous pressure gradient. Hepatology 2009;49(4): 1236–1244 Poynard T, Moussalli J, Munteanu M, et al; FibroFrance-GHPS group. Slow regression of liver fibrosis presumed by repeated biomarkers after virological cure in patients with chronic hepatitis C. J Hepatol 2013;59(4):675–683 Puche JE, Saiman Y, Friedman SL. Hepatic stellate cells and liver fibrosis. Compr Physiol 2013;3(4):1473–1492 Reeves HL, Friedman SL. Activation of hepatic stellate cells—a key issue in liver fibrosis. Front Biosci 2002;7:d808–d826 Friedman SL. Hepatic stellate cells: protean, multifunctional, and enigmatic cells of the liver. Physiol Rev 2008;88(1):125–172 Puche JE, Lee YA, Jiao J, et al. A novel murine model to deplete hepatic stellate cells uncovers their role in amplifying liver damage in mice. Hepatology 2013;57(1):339–350 Hernandez-Gea V, Friedman SL. Pathogenesis of liver fibrosis. Annu Rev Pathol 2011;6:425–456 Iredale JP, Murphy G, Hembry RM, Friedman SL, Arthur MJ. Human hepatic lipocytes synthesize tissue inhibitor of metalloproteinases-1. Implications for regulation of matrix degradation in liver. J Clin Invest 1992;90(1):282–287 Iredale JP. Models of liver fibrosis: exploring the dynamic nature of inflammation and repair in a solid organ. J Clin Invest 2007; 117(3):539–548 Iredale JP. Tissue inhibitors of metalloproteinases in liver fibrosis. Int J Biochem Cell Biol 1997;29(1):43–54 Iredale J. Defining therapeutic targets for liver fibrosis: exploiting the biology of inflammation and repair. Pharmacol Res 2008; 58(2):129–136 Arthur MJ, Iredale JP, Mann DA. Tissue inhibitors of metalloproteinases: role in liver fibrosis and alcoholic liver disease. Alcohol Clin Exp Res 1999;23(5):940–943 Benyon RC, Arthur MJP. Extracellular matrix degradation and the role of hepatic stellate cells. Semin Liver Dis 2001;21(3):373–384 Iredale JP, Thompson A, Henderson NC. Extracellular matrix degradation in liver fibrosis: Biochemistry and regulation. Biochim Biophys Acta 2013;1832(7):876–883 Hemmann S, Graf J, Roderfeld M, Roeb E. Expression of MMPs and TIMPs in liver fibrosis - a systematic review with special emphasis on anti-fibrotic strategies. J Hepatol 2007;46(5):955–975 Issa R, Zhou X, Constandinou CM, et al. Spontaneous recovery from micronodular cirrhosis: evidence for incomplete resolution associated with matrix cross-linking. Gastroenterology 2004; 126(7):1795–1808

Seminars in Liver Disease

Vol. 35

No. 2/2015

Ramachandran et al.

35 Yoshiji H, Kuriyama S, Yoshii J, et al. Tissue inhibitor of metal-

36

37

38

39

40

41

42

43

44

45

46 47

48

49

50

51

52

loproteinases-1 attenuates spontaneous liver fibrosis resolution in the transgenic mouse. Hepatology 2002;36(4 Pt 1): 850–860 Elsharkawy AM, Oakley F, Mann DA. The role and regulation of hepatic stellate cell apoptosis in reversal of liver fibrosis. Apoptosis 2005;10(5):927–939 Watson MR, Wallace K, Gieling RG, et al. NF-kappaB is a critical regulator of the survival of rodent and human hepatic myofibroblasts. J Hepatol 2008;48(4):589–597 Murphy FR, Issa R, Zhou X, 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 2002;277(13):11069–11076 Murphy F, Waung J, Collins J, et al. N-Cadherin cleavage during activated hepatic stellate cell apoptosis is inhibited by tissue inhibitor of metalloproteinase-1. Comp Hepatol 2004;3(Suppl 1): S8 Zhou X, Murphy FR, Gehdu N, Zhang J, Iredale JP, Benyon RC. Engagement of alphavbeta3 integrin regulates proliferation and apoptosis of hepatic stellate cells. J Biol Chem 2004;279(23): 23996–24006 Issa R, Zhou X, Trim N, et al. Mutation in collagen-1 that confers resistance to the action of collagenase results in failure of recovery from CCl4-induced liver fibrosis, persistence of activated hepatic stellate cells, and diminished hepatocyte regeneration. FASEB J 2003;17(1):47–49 Oakley F, Trim N, Constandinou CM, et al. Hepatocytes express nerve growth factor during liver injury: evidence for paracrine regulation of hepatic stellate cell apoptosis. Am J Pathol 2003; 163(5):1849–1858 Trim N, Morgan S, Evans M, et al. Hepatic stellate cells express the low affinity nerve growth factor receptor p75 and undergo apoptosis in response to nerve growth factor stimulation. Am J Pathol 2000;156(4):1235–1243 Kendall TJ, Hennedige S, Aucott RL, et al. p75 Neurotrophin receptor signaling regulates hepatic myofibroblast proliferation and apoptosis in recovery from rodent liver fibrosis. Hepatology 2009;49(3):901–910 Fischer R, Cariers A, Reinehr R, Häussinger D. Caspase 9-dependent killing of hepatic stellate cells by activated Kupffer cells. Gastroenterology 2002;123(3):845–861 Krizhanovsky V, Yon M, Dickins RA, et al. Senescence of activated stellate cells limits liver fibrosis. Cell 2008;134(4):657–667 Borkham-Kamphorst E, Schaffrath C, Van de Leur E, et al. The anti-fibrotic effects of CCN1/CYR61 in primary portal myofibroblasts are mediated through induction of reactive oxygen species resulting in cellular senescence, apoptosis and attenuated TGF-β signaling. Biochim Biophys Acta 2014;1843(5): 902–914 Kim KH, Chen CC, Monzon RI, Lau LF. Matricellular protein CCN1 promotes regression of liver fibrosis through induction of cellular senescence in hepatic myofibroblasts. Mol Cell Biol 2013;33(10): 2078–2090 Kong X, Feng D, Wang H, et al. Interleukin-22 induces hepatic stellate cell senescence and restricts liver fibrosis in mice. Hepatology 2012;56(3):1150–1159 Klein S, Klösel J, Schierwagen R, et al. Atorvastatin inhibits proliferation and apoptosis, but induces senescence in hepatic myofibroblasts and thereby attenuates hepatic fibrosis in rats. Lab Invest 2012;92(10):1440–1450 Simon TG, King LY, Zheng H, Chung RT. Statin use is associated with a reduced risk of fibrosis progression in chronic hepatitis C. J Hepatol 2015;62(1):18–23 Kisseleva T, Cong M, Paik Y, et al. Myofibroblasts revert to an inactive phenotype during regression of liver fibrosis. Proc Natl Acad Sci U S A 2012;109(24):9448–9453

This document was downloaded for personal use only. Unauthorized distribution is strictly prohibited.

128

Resolution of Liver Fibrosis: Basic Mechanisms and Clinical Relevance

54

55

56

57

58

59

60

61 62

63

64

65

66

67

68

69

70

71

stellate cells during liver fibrosis resolution in mice. Gastroenterology 2012;143(4):1073–83.e22 Duffield JS, Forbes SJ, Constandinou CM, et al. Selective depletion of macrophages reveals distinct, opposing roles during liver injury and repair. J Clin Invest 2005;115(1):56–65 Fallowfield JA, Mizuno M, Kendall TJ, et al. Scar-associated macrophages are a major source of hepatic matrix metalloproteinase13 and facilitate the resolution of murine hepatic fibrosis. J Immunol 2007;178(8):5288–5295 Ramachandran P, Pellicoro A, Vernon MA, et al. Differential Ly-6C expression identifies the recruited macrophage phenotype, which orchestrates the regression of murine liver fibrosis. Proc Natl Acad Sci U S A 2012;109(46):E3186–E3195 Ide M, Kuwamura M, Kotani T, Sawamoto O, Yamate J. Effects of gadolinium chloride (GdCl(3)) on the appearance of macrophage populations and fibrogenesis in thioacetamide-induced rat hepatic lesions. J Comp Pathol 2005;133(2-3):92–102 Sunami Y, Leithäuser F, Gul S, et al. Hepatic activation of IKK/NFκB signaling induces liver fibrosis via macrophage-mediated chronic inflammation. Hepatology 2012;56(3):1117–1128 Pradere JP, Kluwe J, De Minicis S, et al. Hepatic macrophages but not dendritic cells contribute to liver fibrosis by promoting the survival of activated hepatic stellate cells in mice. Hepatology 2013;58(4):1461–1473 Karlmark KR, Weiskirchen R, Zimmermann HW, et al. Hepatic recruitment of the inflammatory Gr1þ monocyte subset upon liver injury promotes hepatic fibrosis. Hepatology 2009;50(1): 261–274 Wynn TA, Barron L. Macrophages: master regulators of inflammation and fibrosis. Semin Liver Dis 2010;30(3):245–257 Henderson NC, Mackinnon AC, Farnworth SL, et al. Galectin-3 expression and secretion links macrophages to the promotion of renal fibrosis. Am J Pathol 2008;172(2):288–298 Henderson NC, Mackinnon AC, Farnworth SL, et al. Galectin-3 regulates myofibroblast activation and hepatic fibrosis. Proc Natl Acad Sci U S A 2006;103(13):5060–5065 Traber PG, Chou H, Zomer E, et al. Regression of fibrosis and reversal of cirrhosis in rats by galectin inhibitors in thioacetamide-induced liver disease. PLoS ONE 2013;8(10):e75361 Zimmermann HW, Seidler S, Nattermann J, et al. Functional contribution of elevated circulating and hepatic non-classical CD14CD16 monocytes to inflammation and human liver fibrosis. PLoS ONE 2010;5(6):e11049 Liaskou E, Zimmermann HW, Li KK, et al. Monocyte subsets in human liver disease show distinct phenotypic and functional characteristics. Hepatology 2013;57(1):385–398 Preisser L, Miot C, Le Guillou-Guillemette H, et al. IL-34 and macrophage colony-stimulating factor are overexpressed in hepatitis C virus fibrosis and induce profibrotic macrophages that promote collagen synthesis by hepatic stellate cells. Hepatology 2014;60(6):1879–1890 Yang L, Kwon J, Popov Y, et al. Vascular endothelial growth factor promotes fibrosis resolution and repair in mice. Gastroenterology 2014;146(5):1339–50.e1 Gibbons MA, MacKinnon AC, Ramachandran P, et al. Ly6Chi monocytes direct alternatively activated profibrotic macrophage regulation of lung fibrosis. Am J Respir Crit Care Med 2011; 184(5):569–581 Pellicoro A, Aucott RL, Ramachandran P, et al. Elastin accumulation is regulated at the level of degradation by macrophage metalloelastase (MMP-12) during experimental liver fibrosis. Hepatology 2012;55(6):1965–1975 Popov Y, Sverdlov DY, Bhaskar KR, et al. Macrophage-mediated phagocytosis of apoptotic cholangiocytes contributes to reversal of experimental biliary fibrosis. Am J Physiol Gastrointest Liver Physiol 2010;298(3):G323–G334

129

72 Mosser DM, Edwards JP. Exploring the full spectrum of macro-

phage activation. Nat Rev Immunol 2008;8(12):958–969 73 Tacke F, Zimmermann HW. Macrophage heterogeneity in liver

injury and fibrosis. J Hepatol 2014;60(5):1090–1096 74 Holt MP, Cheng L, Ju C. Identification and characterization of

75

76

77

78

79 80

81

82

83

84

85

86

87

88

89

90 91 92

93

infiltrating macrophages in acetaminophen-induced liver injury. J Leukoc Biol 2008;84(6):1410–1421 Zigmond E, Samia-Grinberg S, Pasmanik-Chor M, et al. Infiltrating monocyte-derived macrophages and resident kupffer cells display different ontogeny and functions in acute liver injury. J Immunol 2014;193(1):344–353 Yona S, Kim KW, Wolf Y, et al. Fate mapping reveals origins and dynamics of monocytes and tissue macrophages under homeostasis. Immunity 2013;38(1):79–91 Schulz C, Gomez Perdiguero E, Chorro L, et al. A lineage of myeloid cells independent of Myb and hematopoietic stem cells. Science 2012;336(6077):86–90 Hashimoto D, Chow A, Noizat C, et al. Tissue-resident macrophages self-maintain locally throughout adult life with minimal contribution from circulating monocytes. Immunity 2013;38(4): 792–804 Seki E, de Minicis S, Inokuchi S, et al. CCR2 promotes hepatic fibrosis in mice. Hepatology 2009;50(1):185–197 Mitchell C, Couton D, Couty JP, et al. Dual role of CCR2 in the constitution and the resolution of liver fibrosis in mice. Am J Pathol 2009;174(5):1766–1775 Lin SL, Castaño AP, Nowlin BT, Lupher ML Jr, Duffield JS. Bone marrow Ly6Chigh monocytes are selectively recruited to injured kidney and differentiate into functionally distinct populations. J Immunol 2009;183(10):6733–6743 Aoyama T, Inokuchi S, Brenner DA, Seki E. CX3CL1-CX3CR1 interaction prevents carbon tetrachloride-induced liver inflammation and fibrosis in mice. Hepatology 2010;52(4):1390–1400 Karlmark KR, Zimmermann HW, Roderburg C, et al. The fractalkine receptor CX3CR1 protects against liver fibrosis by controlling differentiation and survival of infiltrating hepatic monocytes. Hepatology 2010;52(5):1769–1782 Baeck C, Wei X, Bartneck M, et al. Pharmacological inhibition of the chemokine C-C motif chemokine ligand 2 (monocyte chemoattractant protein 1) accelerates liver fibrosis regression by suppressing Ly-6C(þ) macrophage infiltration in mice. Hepatology 2014;59(3):1060–1072 Yoshiji H, Kuriyama S, Yoshii J, et al. Vascular endothelial growth factor and receptor interaction is a prerequisite for murine hepatic fibrogenesis. Gut 2003;52(9):1347–1354 Rosmorduc O. Antiangiogenic therapies in portal hypertension: a breakthrough in hepatology. Gastroenterol Clin Biol 2010;34(89):446–449 Sahin H, Borkham-Kamphorst E, Kuppe C, et al. Chemokine Cxcl9 attenuates liver fibrosis-associated angiogenesis in mice. Hepatology 2012;55(5):1610–1619 Iredale JP, Bataller R. Identifying molecular factors that contribute to resolution of liver fibrosis. Gastroenterology 2014;146(5): 1160–1164 Jiao J, Sastre D, Fiel MI, et al. Dendritic cell regulation of carbon tetrachloride-induced murine liver fibrosis regression. Hepatology 2012;55(1):244–255 Hume DA. Macrophages as APC and the dendritic cell myth. J Immunol 2008;181(9):5829–5835 Kubes P, Mehal WZ. Sterile inflammation in the liver. Gastroenterology 2012;143(5):1158–1172 Pellicoro A, Ramachandran P, Iredale JP, Fallowfield JA. Liver fibrosis and repair: immune regulation of wound healing in a solid organ. Nat Rev Immunol 2014;14(3):181–194 Harty MW, Muratore CS, Papa EF, et al. Neutrophil depletion blocks early collagen degradation in repairing cholestatic rat livers. Am J Pathol 2010;176(3):1271–1281

Seminars in Liver Disease

Vol. 35

No. 2/2015

This document was downloaded for personal use only. Unauthorized distribution is strictly prohibited.

53 Troeger JS, Mederacke I, Gwak GY, et al. Deactivation of hepatic

Ramachandran et al.

Resolution of Liver Fibrosis: Basic Mechanisms and Clinical Relevance

Ramachandran et al.

94 Radaeva S, Sun R, Jaruga B, Nguyen VT, Tian Z, Gao B. Natural killer

113 Lorenzini S, Bird TG, Boulter L, et al. Characterisation of a stereotypi-

cells ameliorate liver fibrosis by killing activated stellate cells in NKG2D-dependent and tumor necrosis factor-related apoptosisinducing ligand-dependent manners. Gastroenterology 2006; 130(2):435–452 Melhem A, Muhanna N, Bishara A, et al. Anti-fibrotic activity of NK cells in experimental liver injury through killing of activated HSC. J Hepatol 2006;45(1):60–71 Jeong WI, Park O, Gao B. Abrogation of the antifibrotic effects of natural killer cells/interferon-gamma contributes to alcohol acceleration of liver fibrosis. Gastroenterology 2008;134(1): 248–258 Jeong WI, Park O, Suh YG, et al. Suppression of innate immunity (natural killer cell/interferon-γ) in the advanced stages of liver fibrosis in mice. Hepatology 2011;53(4):1342–1351 Glässner A, Eisenhardt M, Krämer B, et al. NK cells from HCVinfected patients effectively induce apoptosis of activated primary human hepatic stellate cells in a TRAIL-, FasL- and NKG2Ddependent manner. Lab Invest 2012;92(7):967–977 Wynn TA. Cellular and molecular mechanisms of fibrosis. J Pathol 2008;214(2):199–210 Shi Z, Wakil AE, Rockey DC. Strain-specific differences in mouse hepatic wound healing are mediated by divergent T helper cytokine responses. Proc Natl Acad Sci U S A 1997;94(20): 10663–10668 Katz SC, Ryan K, Ahmed N, et al. Obstructive jaundice expands intrahepatic regulatory T cells, which impair liver T lymphocyte function but modulate liver cholestasis and fibrosis. J Immunol 2011;187(3):1150–1156 Claassen MA, de Knegt RJ, Tilanus HW, Janssen HL, Boonstra A. Abundant numbers of regulatory T cells localize to the liver of chronic hepatitis C infected patients and limit the extent of fibrosis. J Hepatol 2010;52(3):315–321 Hammerich L, Bangen JM, Govaere O, et al. Chemokine receptor CCR6-dependent accumulation of γδ T cells in injured liver restricts hepatic inflammation and fibrosis. Hepatology 2014; 59(2):630–642 Desmet VJ, Roskams T. Cirrhosis reversal: a duel between dogma and myth. J Hepatol 2004;40(5):860–867 Popov Y, Sverdlov DY, Sharma AK, et al. Tissue transglutaminase does not affect fibrotic matrix stability or regression of liver fibrosis in mice. Gastroenterology 2011;140(5):1642–1652 Wanless IR, Nakashima E, Sherman M. Regression of human cirrhosis. Morphologic features and the genesis of incomplete septal cirrhosis. Arch Pathol Lab Med 2000;124(11): 1599–1607 Barry-Hamilton V, Spangler R, Marshall D, et al. Allosteric inhibition of lysyl oxidase-like-2 impedes the development of a pathologic microenvironment. Nat Med 2010;16(9): 1009–1017 Pockros PJ, Jeffers L, Afdhal N, et al. Final results of a double-blind, placebo-controlled trial of the antifibrotic efficacy of interferongamma1b in chronic hepatitis C patients with advanced fibrosis or cirrhosis. Hepatology 2007;45(3):569–578 McHutchison J, Goodman Z, Patel K, et al; Farglitizar Study Investigators. Farglitazar lacks antifibrotic activity in patients with chronic hepatitis C infection. Gastroenterology 2010; 138(4):1365–1373, 1373.e1–1373.e2 Fernández M, Semela D, Bruix J, Colle I, Pinzani M, Bosch J. Angiogenesis in liver disease. J Hepatol 2009;50(3):604–620 Hall A, Germani G, Isgrò G, Burroughs AK, Dhillon AP. Fibrosis distribution in explanted cirrhotic livers. Histopathology 2012; 60(2):270–277 Marshall A, Rushbrook S, Davies SE, et al. Relation between hepatocyte G1 arrest, impaired hepatic regeneration, and fibrosis in chronic hepatitis C virus infection. Gastroenterology 2005; 128(1):33–42

cal cellular and extracellular adult liver progenitor cell niche in rodents and diseased human liver. Gut 2010;59(5):645–654 Kallis YN, Robson AJ, Fallowfield JA, et al. Remodelling of extracellular matrix is a requirement for the hepatic progenitor cell response. Gut 2011;60(4):525–533 Boulter L, Govaere O, Bird TG, et al. Macrophage-derived Wnt opposes Notch signaling to specify hepatic progenitor cell fate in chronic liver disease. Nat Med 2012;18(4):572–579 Bird TG, Lu WY, Boulter L, et al. Bone marrow injection stimulates hepatic ductular reactions in the absence of injury via macrophage-mediated TWEAK signaling. Proc Natl Acad Sci U S A 2013; 110(16):6542–6547 Kundu AK, Nagaoka M, Chowdhury EH, Hirose S, Sasagawa T, Akaike T. IGF-1 induces growth, survival and morphological change of primary hepatocytes on a galactose-bared polymer through both MAPK and beta-catenin pathways. Cell Struct Funct 2003;28(4):255–263 Desbois-Mouthon C, Wendum D, Cadoret A, et al. Hepatocyte proliferation during liver regeneration is impaired in mice with liver-specific IGF-1R knockout. FASEB J 2006;20(6):773–775 Ebrahimkhani MR, Oakley F, Murphy LB, et al. Stimulating healthy tissue regeneration by targeting the 5-HT2B receptor in chronic liver disease. Nat Med 2011;17(12):1668–1673 Kim JK, Ma DW, Lee KS, Paik YH. Assessment of hepatic fibrosis regression by transient elastography in patients with chronic hepatitis B treated with oral antiviral agents. J Korean Med Sci 2014;29(4):570–575 Leeming DJ, Byrjalsen I, Jiménez W, Christiansen C, Karsdal MA. Protein fingerprinting of the extracellular matrix remodelling in a rat model of liver fibrosis—a serological evaluation. Liver Int 2013;33(3):439–447 Leeming DJ, Karsdal MA, Byrjalsen I, et al. Novel serological neoepitope markers of extracellular matrix proteins for the detection of portal hypertension. Aliment Pharmacol Ther 2013;38(9): 1086–1096 Fuchs BC, Wang H, Yang Y, et al. Molecular MRI of collagen to diagnose and stage liver fibrosis. J Hepatol 2013;59(5):992–998 Chow AM, Tan M, Gao DS, et al. Molecular MRI of liver fibrosis by a peptide-targeted contrast agent in an experimental mouse model. Invest Radiol 2013;48(1):46–54 Popov Y, Patsenker E, Stickel F, et al. Integrin alphavbeta6 is a marker of the progression of biliary and portal liver fibrosis and a novel target for antifibrotic therapies. J Hepatol 2008;48(3): 453–464 Ruddell RG, Oakley F, Hussain Z, et al. A role for serotonin (5-HT) in hepatic stellate cell function and liver fibrosis. Am J Pathol 2006;169(3):861–876 Forbes SJ, Newsome PN. New horizons for stem cell therapy in liver disease. J Hepatol 2012;56(2):496–499 Thomas JA, Pope C, Wojtacha D, et al. Macrophage therapy for murine liver fibrosis recruits host effector cells improving fibrosis, regeneration, and function. Hepatology 2011;53(6): 2003–2015 Ratziu V, Bedossa P, Francque SM, et al. Lack of efficacy of an inhibitor of PDE4 in phase 1 and 2 trials of patients with nonalcoholic steatohepatitis. Clin Gastroenterol Hepatol 2014; 12(10):1724–30.e5 Habens F, Srinivasan N, Oakley F, Mann DA, Ganesan A, Packham G. Novel sulfasalazine analogues with enhanced NF-kB inhibitory and apoptosis promoting activity. Apoptosis 2005;10(3): 481–491 Teixeira-Clerc F, Julien B, Grenard P, et al. CB1 cannabinoid receptor antagonism: a new strategy for the treatment of liver fibrosis. Nat Med 2006;12(6):671–676 Cong M, Liu T, Wang P, et al. Antifibrotic effects of a recombinant adeno-associated virus carrying small interfering RNA

95

96

97

98

99 100

101

102

103

104 105

106

107

108

109

110 111

112

Seminars in Liver Disease

Vol. 35

No. 2/2015

114

115

116

117

118

119

120

121

122

123 124

125

126

127 128

129

130

131

132

This document was downloaded for personal use only. Unauthorized distribution is strictly prohibited.

130

133

134

135

136

137

138

139

140

targeting TIMP-1 in rat liver fibrosis. Am J Pathol 2013;182(5): 1607–1616 Parsons CJ, Bradford BU, Pan CQ, et al. Antifibrotic effects of a tissue inhibitor of metalloproteinase-1 antibody on established liver fibrosis in rats. Hepatology 2004;40(5):1106–1115 Roderfeld M, Weiskirchen R, Wagner S, et al. Inhibition of hepatic fibrogenesis by matrix metalloproteinase-9 mutants in mice. FASEB J 2006;20(3):444–454 Roeb E, Behrmann I, Grötzinger J, Breuer B, Matern S. An MMP-9 mutant without gelatinolytic activity as a novel TIMP-1-antagonist. FASEB J 2000;14(12):1671–1673 Arabpour M, Poelstra K, Helfrich W, Bremer E, Haisma HJ. Targeted elimination of activated hepatic stellate cells by an anti-epidermal growth factor-receptor single chain fragment variable antibody-tumor necrosis factor-related apoptosis-inducing ligand (scFv425-sTRAIL). J Gene Med 2014;16(9-10):281–290 Sato Y, Murase K, Kato J, et al. Resolution of liver cirrhosis using vitamin A-coupled liposomes to deliver siRNA against a collagenspecific chaperone. Nat Biotechnol 2008;26(4):431–442 Douglass A, Wallace K, Parr R, et al. Antibody-targeted myofibroblast apoptosis reduces fibrosis during sustained liver injury. J Hepatol 2008;49(1):88–98 Yang MD, Chiang YM, Higashiyama R, et al. Rosmarinic acid and baicalin epigenetically derepress peroxisomal proliferator-activated receptor γ in hepatic stellate cells for their antifibrotic effect. Hepatology 2012;55(4):1271–1281 Zhang S, Wang J, Liu Q, Harnish DC. Farnesoid X receptor agonist WAY-362450 attenuates liver inflammation and fibrosis in mu-

141

142

143

144

145

146

147

Ramachandran et al.

rine model of non-alcoholic steatohepatitis. J Hepatol 2009; 51(2):380–388 Fiorucci S, Rizzo G, Antonelli E, et al. A farnesoid x receptor-small heterodimer partner regulatory cascade modulates tissue metalloproteinase inhibitor-1 and matrix metalloprotease expression in hepatic stellate cells and promotes resolution of liver fibrosis. J Pharmacol Exp Ther 2005;314(2):584–595 Kim EJ, Cho HJ, Park D, et al. Antifibrotic effect of MMP13encoding plasmid DNA delivered using polyethylenimine shielded with hyaluronic acid. Mol Ther 2011;19(2):355–361 Siller-López F, Sandoval A, Salgado S, et al. Treatment with human metalloproteinase-8 gene delivery ameliorates experimental rat liver cirrhosis. Gastroenterology 2004;126(4):1122–1133, discussion 949 Popov Y, Patsenker E, Bauer M, Niedobitek E, Schulze-Krebs A, Schuppan D. Halofuginone induces matrix metalloproteinases in rat hepatic stellate cells via activation of p38 and NFkappaB. J Biol Chem 2006;281(22):15090–15098 Fürst G, Schulte am Esch J, Poll LW, et al. Portal vein embolization and autologous CD133þ bone marrow stem cells for liver regeneration: initial experience. Radiology 2007;243(1): 171–179 Wang J, Zhou X, Cui L, et al. The significance of CD14þ monocytes in peripheral blood stem cells for the treatment of rat liver cirrhosis. Cytotherapy 2010;12(8):1022–1034 Wasmuth HE, Lammert F, Zaldivar MM, et al. Antifibrotic effects of CXCL9 and its receptor CXCR3 in livers of mice and humans. Gastroenterology 2009;137(1):309–319, 319.e1–319.e3

Seminars in Liver Disease

Vol. 35

No. 2/2015

131

This document was downloaded for personal use only. Unauthorized distribution is strictly prohibited.

Resolution of Liver Fibrosis: Basic Mechanisms and Clinical Relevance

Copyright of Seminars in Liver Disease is the property of Thieme Medical Publishing Inc. and its content may not be copied or emailed to multiple sites or posted to a listserv without the copyright holder's express written permission. However, users may print, download, or email articles for individual use.

Resolution of liver fibrosis: basic mechanisms and clinical relevance.

With evidence from a large number of animal models and clinical trials, it is now beyond debate that liver fibrosis and even cirrhosis are potentially...
386KB Sizes 1 Downloads 7 Views