Gastro Rvu-Slf 2008

15
Mechanisms of Hepatic Fibrogenesis Scott L. Friedman Division of Liver Diseases, Mount Sinai School of Medicine, New York, New York Substantial improvements in the treatment of chronic liver disease have accelerated interest in un- covering the mechanisms underlying hepatic fibrosis and its resolution. Activation of resident hepatic stel- late cells into proliferative, contractile, and fibrogenic cells in liver injury remains a dominant theme driving the field. However, several new areas of rapid progress in the past 5–10 years also have taken root, including: (1) identification of different fibrogenic populations apart from resident stellate cells, for ex- ample, portal fibroblasts, fibrocytes, and bone-mar- row– derived cells, as well as cells derived from epi- thelial mesenchymal transition; (2) emergence of stellate cells as finely regulated determinants of he- patic inflammation and immunity; (3) elucidation of multiple pathways controlling gene expression dur- ing stellate cell activation including transcriptional, post-transcriptional, and epigenetic mechanisms; (4) recognition of disease-specific pathways of fibrogen- esis; (5) re-emergence of hepatic macrophages as de- terminants of matrix degradation in fibrosis resolu- tion and the importance of matrix cross-linking and scar maturation in determining reversibility; and (6) hints that hepatic stellate cells may contribute to hepatic stem cell behavior, cancer, and regeneration. Clinical and translational implications of these ad- vances have become clear, and have begun to impact significantly on the management and outlook of pa- tients with chronic liver disease. T he field of hepatic fibrosis is flourishing thanks to continued experimental advances complemented by exciting progress in the treatment of chronic liver dis- ease. 1 Control of chronic hepatitis B and C by antiviral therapies has established that advanced fibrosis can re- gress in association with improved clinical outcomes, 2–4 thereby intensifying enthusiasm to uncover the mecha- nistic basis for hepatic fibrogenesis and its attenuation. At the same time, simple paradigms defining the cel- lular sources of extracellular matrix (ECM), and the roles of cytokines and paracrine interactions among resident liver cells and inflammatory cells have yielded a more nuanced understanding of how the liver re- sponds to injury. These advances have had a collateral benefit towards understanding fibrosis in other organs, particularly the pancreas. 5 Thus, a review of the mech- anisms underlying hepatic fibrosis is not only timely, but is more clinically relevant than ever. This article focuses on recent advances in the field, building on established principles from earlier reviews 6,7 while weaving in their clinical relevance, but also emphasiz- ing the molecular subtleties that have emerged through continued progress in the field. General Principles Fibrosis, or scarring of the liver, is a wound-heal- ing response that engages a range of cell types and me- diators to encapsulate injury. Although even acute injury will activate mechanisms of fibrogenesis, the sustained signals associated with chronic liver disease caused by infection, drugs, metabolic disorders, or immune attack are required for significant fibrosis to accumulate. Occa- sionally, fibrosis may be rapidly progressive over weeks to months, for example, as a result of drug injury, hepatitis C virus (HCV) after liver transplantation, 8 or human immunodeficiency virus (HIV)/HCV co-infection, 9 but for the most part this is a response that evolves over Abbreviations used in this paper: ADAMTS2, a disintegrin and met- alloproteinase with thrombospondin–type repeats metalloproteinase with thrombospondin type I motif; ASH, alcoholic steatohepatitis; CTGF/CCN2, connective tissue growth factor; ECM, extracellular ma- trix; HIV, human immunodeficiency virus; MMP, matrix metalloprotein- ase; NASH, nonalcoholic steatohepatitis; PDGF, platelet-derived growth factor; TGF, transforming growth factor; TIMP, tissue inhibitors of metalloproteinase; TLR, Toll-like receptor; VEGF, vascular endothe- lial growth factor. © 2008 by the AGA Institute 0016-5085/08/$34.00 doi:10.1053/j.gastro.2008.03.003 GASTROENTEROLOGY 2008;134:1655–1669

Transcript of Gastro Rvu-Slf 2008

Page 1: Gastro Rvu-Slf 2008

M

D

SccalctpipartspmiprettshhCvst

TeetgtnA

GASTROENTEROLOGY 2008;134:1655–1669

echanisms of Hepatic Fibrogenesis

Scott L. Friedman

ivision of Liver Diseases, Mount Sinai School of Medicine, New York, New York

lrrasbpabfewit

idwsiasmCif

awCtagol

ubstantial improvements in the treatment ofhronic liver disease have accelerated interest in un-overing the mechanisms underlying hepatic fibrosisnd its resolution. Activation of resident hepatic stel-ate cells into proliferative, contractile, and fibrogenicells in liver injury remains a dominant theme drivinghe field. However, several new areas of rapidrogress in the past 5–10 years also have taken root,

ncluding: (1) identification of different fibrogenicopulations apart from resident stellate cells, for ex-mple, portal fibroblasts, fibrocytes, and bone-mar-ow– derived cells, as well as cells derived from epi-helial mesenchymal transition; (2) emergence oftellate cells as finely regulated determinants of he-atic inflammation and immunity; (3) elucidation ofultiple pathways controlling gene expression dur-

ng stellate cell activation including transcriptional,ost-transcriptional, and epigenetic mechanisms; (4)ecognition of disease-specific pathways of fibrogen-sis; (5) re-emergence of hepatic macrophages as de-erminants of matrix degradation in fibrosis resolu-ion and the importance of matrix cross-linking andcar maturation in determining reversibility; and (6)ints that hepatic stellate cells may contribute toepatic stem cell behavior, cancer, and regeneration.linical and translational implications of these ad-ances have become clear, and have begun to impactignificantly on the management and outlook of pa-ients with chronic liver disease.

he field of hepatic fibrosis is flourishing thanks tocontinued experimental advances complemented by

xciting progress in the treatment of chronic liver dis-ase.1 Control of chronic hepatitis B and C by antiviralherapies has established that advanced fibrosis can re-ress in association with improved clinical outcomes,2– 4

hereby intensifying enthusiasm to uncover the mecha-istic basis for hepatic fibrogenesis and its attenuation.

t the same time, simple paradigms defining the cel-

ular sources of extracellular matrix (ECM), and theoles of cytokines and paracrine interactions amongesident liver cells and inflammatory cells have yielded

more nuanced understanding of how the liver re-ponds to injury. These advances have had a collateralenefit towards understanding fibrosis in other organs,articularly the pancreas.5 Thus, a review of the mech-nisms underlying hepatic fibrosis is not only timely,ut is more clinically relevant than ever. This articleocuses on recent advances in the field, building onstablished principles from earlier reviews6,7 whileeaving in their clinical relevance, but also emphasiz-

ng the molecular subtleties that have emergedhrough continued progress in the field.

General PrinciplesFibrosis, or scarring of the liver, is a wound-heal-

ng response that engages a range of cell types and me-iators to encapsulate injury. Although even acute injuryill activate mechanisms of fibrogenesis, the sustained

ignals associated with chronic liver disease caused bynfection, drugs, metabolic disorders, or immune attackre required for significant fibrosis to accumulate. Occa-ionally, fibrosis may be rapidly progressive over weeks to

onths, for example, as a result of drug injury, hepatitisvirus (HCV) after liver transplantation,8 or human

mmunodeficiency virus (HIV)/HCV co-infection,9 butor the most part this is a response that evolves over

Abbreviations used in this paper: ADAMTS2, a disintegrin and met-lloproteinase with thrombospondin–type repeats metalloproteinaseith thrombospondin type I motif; ASH, alcoholic steatohepatitis;TGF/CCN2, connective tissue growth factor; ECM, extracellular ma-rix; HIV, human immunodeficiency virus; MMP, matrix metalloprotein-se; NASH, nonalcoholic steatohepatitis; PDGF, platelet-derivedrowth factor; TGF, transforming growth factor; TIMP, tissue inhibitorsf metalloproteinase; TLR, Toll-like receptor; VEGF, vascular endothe-

ial growth factor.© 2008 by the AGA Institute

0016-5085/08/$34.00

doi:10.1053/j.gastro.2008.03.003
Page 2: Gastro Rvu-Slf 2008

dtoro

atesfivnciea

ndalaTti

setngpoalmiekfia(Hmbob

amda

kmychm

opaaahtoutfeieaoidibldp

rt2a

aicaoTttraatbowfp

f

1656 SCOTT L. FRIEDMAN GASTROENTEROLOGY Vol. 134, No. 6

ecades. The protracted nature of this response, in con-rast to the more rapid progression of fibrosis in kidneyr lung, has been classically ascribed to the liver’s uniqueegenerative capacity, but the molecular underpinningsf this capacity remain mysterious.

In many ways, the liver’s response to injury is anngiogenic one, with evidence of new blood vessel forma-ion, sinusoidal remodeling, and pericyte (ie, stellate cell)xpansion.10 Thus, mediators familiar to the angiogene-is field are equally relevant in understanding hepaticbrosis, including platelet-derived growth factor (PDGF),ascular endothelial growth factor (VEGF), and their cog-ate receptors, as well as vasoactive mediators that in-lude nitric oxide and carbon monoxide. For example,ncreased VEGF concentrations may contribute to accel-rated progression of fibrosis in smokers who have hep-titis C.11

Cirrhosis, the most advanced stage of fibrosis, con-otes not only more scar than fibrosis alone, but alsoistortion of the liver parenchyma associated with septaend nodule formation, altered blood flow, and risk ofiver failure. However, cirrhosis still remains a dynamicnd evolving state, as discussed later (see Clinical andranslational Implications section), such that interven-

ions even at these advanced stages could regress scar andmprove clinical outcomes.

Continued progress in the field also has exploitedteady refinements in both cell culture and animal mod-ls of fibrosis.12 Although there are no rodent modelshat closely mimic hepatitis B virus (HBV), HCV, oronalcoholic steatohepatitis (NASH), the development ofenetic mouse models has continued to acceleraterogress by enabling reductionist approaches that focusn the role of individual gene products in fibrogenesis,nd by permitting genetic lineage tracing to define cellu-ar phenotypes and their evolution.13 Animal and culture

odels also have benefited from improving technology,n particular the use of gene array and proteomics. Forxample, gene expression patterns from stellate cells (theey resident fibrogenic cell type) isolated from rats withbrosis from either CCl4 or bile duct ligation are remark-bly similar, but differ substantially from ultrapurifiedie, using flow cytometry) culture-activated stellate cells.14

owever, stellate cells isolated using standard gradientethods more closely resembled activated cells from fi-

rotic liver, indicating that standard cell isolation meth-ds yield gene expression data that are relevant to theiology of stellate cells during fibrosis in vivo.14

Liver Injury and Inflammation:Established Mediators and New PlayersFibrosis requires some element of liver injury,

lbeit not necessarily defined by the presence of inflam-atory cells. For example, although the most prevalent

iseases in clinical practice (viral hepatitis, alcoholic ste-

tohepatitis [ASH], and NASH) are characterized by leu- h

ocyte infiltration, metabolic diseases such as hemachro-atosis are notable for their lack of inflammatory cells,

et they too lead to cirrhosis and risk of hepatocellulararcinoma. Thus, any chronic perturbation of hepaticomeostasis, whether visible by light microscopy or not,ay elicit the signals necessary to stimulate fibrogenesis.One family of such established mediators are reactive

xygen species. These unstable compounds include su-eroxide and hydroxyl radicals, hydrogen peroxide, andldehydic end products including 4-hydroxy-2,3-nonenalnd 4-hydroxy-2,3-alkenals. These mediators are gener-ted through lipid peroxidation, and can derive fromepatocytes, macrophages, stellate cells, and inflamma-ory cells.15,16 Several substrates may enhance reactivexygen species production including ethanol, polyunsat-rated fatty acids, and iron. The classic pathway of reac-ive oxygen species generation in hepatocytes resultsrom induction of cytochrome P450 2E1, especially inither ASH or NASH,17,18 leading to pericentral (zone 3)njury. More recently, however, reduced nicotinamide ad-nine dinucleotide phosphate oxidase has emerged asnother source of oxidant stress that mediates pathwaysf fibrogenic activation in hepatic stellate cells, as well as

n Kupffer cells, the resident liver macrophages.19 Re-uced nicotinamide adenine dinucleotide phosphate ox-

dase also may mediate liver injury and fibrosis generatedy angiotensin signaling because animals genetically

acking the p47 subunit of reduced nicotinamide adenineinucleotide phosphate oxidase have reduced superoxideroduction and attenuated hepatic fibrosis after injury.20

More recently, increasing attention also has been di-ected to nitrosative stress generated by hepatocyte mi-ochondrial injury and induction of nitric oxide synthase,21,22 although links from this pathway to fibrogenesisre not as well defined.

Apoptosis of parenchymal cells is no longer viewed assilent consequence of liver injury, but rather as an

mportant inflammatory stimulus23 that activates stellateells, which display a surprising capacity to phagocytosepoptotic bodies,24 leading to induction of reduced nic-tinamide adenine dinucleotide phosphate oxidase.25

his response to apoptotic hepatocytes in part reflectshe interaction of hepatocyte DNA with Toll-like recep-or 9 (TLR9) expressed on stellate cells.26 A profibrogenicesponse also can be elicited by hepatocyte apoptosisfter disruption of the anti-apoptotic mediator Bcl-xL,27

nd by Fas.28 Thus, efforts to block hepatocyte apoptosisherapeutically are being developed as a potential antifi-rotic strategy.29 On the other hand, selective stimulationf apoptosis in stellate cells rather than hepatocytesould be antifibrotic, mediated by either tumor necrosis

actor-related apoptosis-inducing ligand,30 gliotoxin,31 orroteasome inhibitors.32

Although necrosis of cells is a classic morphologiceature of liver injury, its pathogenic contribution to

epatic fibrosis has been overlooked, largely because
Page 3: Gastro Rvu-Slf 2008

toaaitliob

tlarrcsstnabmgptttsttp

oyiataidfiwirclfIceBlmh

Eud

daihcvicutlficedafit

Fcblbobficwd

May 2008 HEPATIC FIBROGENESIS 1657

here are no classic biochemical or molecular hallmarksf necrosis similar to those that have been uncovered forpoptosis.33 Yet, in human disease both necrosis andpoptosis are evident in liver sections, although specificnflammatory pathways of necrosis have not been iden-ified. Necrosis may simply represent a more severe cel-ular response than apoptosis when concentrations ofnjurious stimuli are higher,15,16 but the relative potenciesf necrosis compared with apoptosis in stimulating fi-rogenesis are unknown.

Among the most compelling pathways of injury arehose recently uncovered for innate immune signaling iniver. Specifically, the discovery of TLRs has led to majordvances in understanding how the human organismesponds to pathogens.34 The identification of TLR4, theeceptor for bacterial lipopolysaccharide, on Kupfferells, was therefore not a surprise, but its expression ontellate cells was unexpected.35 Moreover, although TLR4ignaling in macrophages may be essential for inflamma-ory responses,36 recent studies have indicated that sig-aling by stellate cells in response to lipopolysaccharidend possibly endogenous ligands of TLR4 (eg, high-mo-ility group box 1, biglycan, and heparan sulfate) may beore important than in Kupffer cells in eliciting a fibro-

enic response by down-regulating bone morphogenicrotein (BMP) and activin membrane-bound inhibitor, aransmembrane suppressor of transforming growth fac-or �1 (TGF�1), which is the major fibrogenic cytokine inhe liver.37 This finding has converged with evidence thatpecific single-nucleotide polymorphisms of TLR4 con-ribute to the rate of fibrosis progression in HCV infec-ion,38 thereby linking a genetic risk marker to diseaseathogenesis.Evidence that stellate cells play a pivotal role in

rchestrating hepatic immune responses extends be-ond the TLR pathway and is among the most surpris-ng findings of the past 5 years. Stellate cells produce

host of chemotactic peptides (especially chemokines)hat amplify infiltration by inflammatory cells. Theylso interact directly with lymphocyte subsets, includ-ng natural killer cells (see Mehal and Friedman39 foretailed review). Indeed, CD8 T cells appear to be morebrogenic towards stellate cells than CD4 cells,40

hich could account for the increased rate of fibrosisn patients with HIV and HCV, in whom the CD4/CD8atio typically is reduced.41 In contrast, natural killerells play an important role in clearing activated stel-ate cells,42,43 an activity enhanced by gamma inter-eron and retinoic acid44 and abrogated by ethanol.45

ntriguingly, stellate cells also are antigen-presentingells,46 and may contribute to the liver’s immunotol-rant properties through T-cell suppression.47 Finally,

lymphocytes, which comprise up to 50% of the totalymphocyte pool in liver, contribute to the fibrogenic

ilieu because mice lacking B cells (JH-/- animals)

ave attenuated fibrosis, apparently by more rapid a

CM degradation after CCl4 injury, implicating annknown interaction with pathways of matrix degra-ation.48

Cellular Sources of ECM in HepaticFibrosis: An Evolving ParadigmThe discovery of stellate cell activation—a trans-

ifferentiation from a quiescent vitamin A–storing cell toproliferative myofibroblast—remains among the most

nformative discoveries to date in unlocking the basis forepatic fibrogenesis. However, the simple paradigm con-eived 15 years ago6 that all fibrosis derives from acti-ated stellate cells has grown far more multifaceted, bothn terms of the pathways of activation and the overallontribution of stellate cells to the total fibrogenic pop-lation during liver injury. It is increasingly clear thathese fibrogenic cells derive not only from resident stel-ate cells, but also from portal fibroblasts,49 –51 circulatingbrocytes,52 bone marrow,53 and epithelial–mesenchymalell transition.13 Although the relative contribution ofach source varies, these differences are likely to reflectiffering contributions with disease progression andmong different etiologies (Figure 1). For example, portalbroblasts appear to be especially important in choles-atic liver diseases and ischemia,51 where paracrine inter-

igure 1. Contributions of activated stellate cells and other fibrogenicell types to hepatic fibrosis. Quiescent stellate cell activation is initiatedy a range of soluble mediators (Figure 2). The activated cell is stimu-

ated further by key cytokines (detailed further in Figure 2) into myofi-roblasts (which contain contractile filaments). Over time, however,ther sources also contribute to fibrogenic populations in liver, includingone marrow (which likely gives rise to circulating fibrocytes), portalbroblasts, and epithelial mesenchymal transition (EMT) from hepato-ytes and cholangiocytes. Relative contributions and the stages athich these cell types add to the myofibroblast population is likely toiffer among various etiologies of liver injury (see text).

ctions between cholangiocytes and fibroblasts involve

Page 4: Gastro Rvu-Slf 2008

bcrcgrcocrccwrm

aado

tic

swuesfomf

FaaefiN dma

1658 SCOTT L. FRIEDMAN GASTROENTEROLOGY Vol. 134, No. 6

oth chemokines54 and extracellular nucleotides.55 Inontrast, progressive recruitment of bone-marrow– de-ived cells may occur over time, such that these cellsan represent a substantial fraction of the total fibro-enic population in more chronic injury. Bone marrowecruitment of mesenchymal cells remains a somewhatonfusing event because of contradictory findings thatn the one hand, bone marrow may provide fibrogenicells, yet on the other hand autologous bone mar-ow56,57 and marrow-derived endothelial progenitorells58 can be antifibrotic. It remains unclear whichells within marrow contribute to fibrogenesis andhich might be antifibrotic, and/or what mediators

egulate these apparently divergent activities of bone-arrow– derived cells.Because cellular sources of fibrogenic cells may differ

mong different etiologies, the relative value of particularntifibrotic therapies also may depend on the underlyingisease. For example, the integrin �v�6 is up-regulated

igure 2. Pathways of hepatic stellate cell activation. Features of stelland those that contribute to perpetuation. Initiation is provoked by solubpoptotic bodies, lipopolysaccharide (LPS), and paracrine stimuli from nendothelium, and hepatocytes. Perpetuation follows, characterized by abrogenesis, altered matrix degradation, chemotaxis, and inflammatoryO, nitric oxide; MT, membrane type. Modified with permission from Frie

nly on biliary epithelium during experimental choles- p

atic liver fibrosis,59 implying that antagonists to thisntegrin might be more rational in biliary than in paren-hymal liver diseases.

Pathways of Stellate Cell Activation:A Moving TargetInitiating PathwaysStellate cell activation unfolds progressively in

equential stages; this paradigm provides a useful frame-ork for defining fibrogenic events after liver injury (Fig-re 2). In particular, the initiation phase, which refers toarly events that render the quiescent stellate cell respon-ive to a range of growth factors, remains an importantocus. Rapid induction of �-PDGF receptor, developmentf a contractile and fibrogenic phenotype, as well asodulation of growth factor signaling are the cardinal

eatures of this early response. Initiating stimuli include

ll activation can be distinguished between those that stimulate initiationmuli that include oxidant stress signals (reactive oxygen intermediates),ring cell types including hepatic macrophages (Kupffer cells), sinusoidalber of specific phenotypic changes including proliferation, contractility,aling. FGF, fibroblast growth factor; ET-1, endothelin-1; NK, natural killer;n.7

te cele sti

ighbonumsign

aracrine signals such as reactive oxygen species from

Page 5: Gastro Rvu-Slf 2008

at

iotdpaoatsfdo

fipcsasrvtRttgtfilstcacn(ta

atsrbccs

dc2r

iRa

aostinsts

haLAfaa

raapcbfibr

tipitpIkaaBolp

ccccprot

May 2008 HEPATIC FIBROGENESIS 1659

poptotic hepatocytes and injured cholangiocytes, as de-ailed earlier (see Liver Injury and Inflammation section).

Changes in ECM composition. Initiating eventsn stellate cell activation are occurring on a backgroundf progressive changes in the surrounding ECM withinhe subendothelial space of Disse. Over time, the suben-othelial matrix composition changes from one com-rised of type IV collagen, heparan sulfate proteoglycan,nd laminin (the classic constituents of a basal lamina) tone rich in fibril-forming collagens, particularly types Ind III. One important yet subtle change is the deposi-ion of a specific isoform of cellular fibronectin frominusoidal endothelial cells, which has an activating ef-ect on stellate cells.60 Because this response is TGF-�ependent,61 however, induction of this cytokine mustccur first, either from autocrine or paracrine sources.

These progressive changes in ECM composition asbrosis accumulates instigate several positive feedbackathways that further amplify fibrosis. First, dynamichanges in membrane receptors, in particular integrins,ense altered matrix signals that provoke stellate cellctivation and migration through focal adhesion disas-embly62– 64 while also linking to other growth factoreceptors through integrin-linked kinase.65,66 Matrix-pro-oked signals also engage membrane-bound guanosineriphosphate binding proteins, in particular Rho67 andac,68 which transduce signals to the actin cytoskeleton

hat promote migration and contraction. Second, activa-ion of cellular matrix metalloproteases leads to release ofrowth factors from matrix-bound reservoirs in the ex-racellular space that may stimulate cellular growth andbrogenesis.69,70 Third, the enhanced density of ECM

eads to increasing matrix stiffness, which is a significanttimulus to stellate cell activation, at least in parthrough integrin signaling.71 Interestingly, however, in-reased stiffness caused by edema and inflammation innimal models and human beings may precede the in-rease in matrix content.72,73 These experimental findingsicely complement the increasing use of Fibroscan74,75

Echosens, Paris, France) and magnetic resonance elas-ography,76,77 two clinical techniques that noninvasivelyssess hepatic stiffness as a reflection of ECM content.

Molecular mechanisms underlying stellate cellctivation. Because activation occurs so rapidly, atten-ion has focused on regulatory pathways that can re-pond quickly to injurious stimuli, either by activating orepressing gene transcription, by epigenetic regulation, ory posttranscriptional control. In addition, although mi-roRNAs have emerged as important layers of regulatoryontrol in many systems,78 their roles in liver injury andtellate cell activation have not yet been explored.

Among the many target genes of transcription factorsescribed in stellate cells, those most comprehensivelyharacterized include type I collagen (alpha 1 and alpha

chains), �-smooth muscle actin, TGF�1, and TGF�

eceptors, matrix metalloproteinase (MMP)-2, and tissue R

nhibitors of metalloproteinases (TIMPs) 1 and 2 (seeippe and Brenner,79 Mann et al,80 and Tsukamoto etl81 for reviews).

Foxf1, JunD, and C/EBP� are among the clearest ex-mples of activating transcription factors. Deletion ofne Foxf1 allele reduces stellate cell activation and fibro-is.82 Similarly, knockout of JunD, a member of the AP-1ranscription factor complex,83 protects mice from CCl4-nduced hepatic fibrosis, which is associated with reducedumbers of activated stellate cells and diminished expres-ion of hepatic TIMP-1.84 Finally, phosphorylation of theranscription factor C/EBP� by the RSK kinase promotestellate cell survival.85

The LIM homeodomain protein, Lhx2, on the otherand, is a protein that preserves stellate cell quiescence,nd whose loss leads to activation of stellate cells. In fact,hx2-/- mice develop spontaneous congenital fibrosis.86

similar role has been uncovered for the transcriptionactor FoxO1; viz. Stellate cell activation and fibrogenesisre amplified in cells from mice with reduced FoxO1ctivity.87

Similar to Lhx2, the peroxisome proliferator activatedeceptor � nuclear receptor down-regulates stellate cellctivation and reduces collagen gene expression.88 Thisnd related observations89 have prompted the use oferoxisome proliferator activated receptor � ligands inlinical trials not only in fibrosis associated with NASH,ut also as a candidate antifibrotic in patients with HCVbrosis. Other nuclear receptors regulating stellate cellehavior include pregnane X receptor,90,91 and retinoideceptors.92,93

Epigenetic regulation is a tightly controlled pathwayhat modulates stellate cell activation in part throughnduction of the molecules CBF1 and MeCP2.94,95 Theseroteins repress gene expression of the inhibitory protein

nhibitor kappa beta (I�B) by CpG island methylation,hereby unleashing nuclear factor � B activity, whichromotes stellate cell survival and thus increases fibrosis.nterestingly, sulfasalazine inhibits the kinase (inhibitorappa kinase, I�K) that activates I�B, and thus mayccelerate recovery from experimental fibrosis by clear-nce of activated stellate cells through apoptosis.94,96

ecause these activated cells typically express high levelsf TIMP-1, a metalloproteinase inhibitor, their clearance

eads to increased net activity of matrix degradingroteases.Finally, messenger RNA (mRNA) stabilization also

ontributes to increased gene expression during stellateell activation. Specifically, there is a 16-fold increase inollagen alpha 1(I) mRNA stabilization during stellateell activation as a result of interaction of a specificrotein, �CP, to a specific sequence in the 3’ untranslatedegion of the mRNA,97 and also involving the interactionf a 120-kilodalton protein with the 5’ stem-loop struc-ure.98 Similarly, there is enhanced interaction of the

NA binding protein, RBMS3, with the 3’ untranslated
Page 6: Gastro Rvu-Slf 2008

ri

sasgionbsatttwsi

tatrtdhbstPPccqnwpiVgb

sPacotp

tcs

srmhs

capr

sgStdlttisc

aiedTwhta

rptpaeais(Crmfeacnt

tt

1660 SCOTT L. FRIEDMAN GASTROENTEROLOGY Vol. 134, No. 6

egion of the homeobox protein Prx1, thereby increasingts mRNA stability.99

Perpetuating PathwaysThe stellate cell that is activated by initiating

timuli then is primed to respond to a host of cytokinesnd growth factors. These signals conspire to generatecar through enhanced proliferation, contractility, fibro-enesis, matrix degradation, and proinflammatory signal-ng. Although earlier models suggested that the pathwaysf activation were identical regardless of the disease, it isow clear that there are disease-specific pathways of fi-rosis (see Disease-Specific Pathways of Hepatic Fibrosisection), and, moreover, that not all cytokine pathwaysre necessarily activated in parallel. For example, al-hough PDGF stimulation may drive cellular prolifera-ion in parallel with fibrogenic stimulation in some set-ings, TLR9 activation blocks PDGF-mediated migrationhile provoking fibrogenesis,26 thereby providing a stop

ignal that allows activated cells to accumulate at sites ofnjury where they can deposit more scar.

Proliferation. Autocrine signaling by PDGF washe first cytokine loop uncovered during stellate cellctivation and remains among the most potent.100 Bothhe PDGF ligand and the beta isoform of its receptor areapidly induced in vivo and in culture.101,102 In additiono the well-characterized A and B chains of platelet-erived growth factor (PDGF), C and D isoforms alsoave been discovered more recently; in fact, PDGF-D maye the most potent and physiologically relevant PDGFubunit in stellate cell activation.100 Interestingly, al-hough both mice with transgenic expression of eitherDGF-B103 or PDGF-C have hepatic fibrosis,104 theDGF-C transgenic animals also develop hepatocellulararcinoma,104 mimicking the progression from fibrosis toancer that occurs in human beings. Downstream conse-uences of PDGF signaling in stellate cells include sig-aling by PI3 kinase, ERK, and other pathways,105–107 asell as stimulation of Na�/H� exchange, providing aotential site for therapeutic intervention by blocking

on transport.108 Other stellate cell mitogens includeEGF,109 thrombin and its receptor,110,111 epidermalrowth factor, TGF�, keratinocyte growth factor,112 andasic fibroblast growth factor.113

Chemotaxis. Stellate cells can migrate towardsites of injury driven by chemoattractants that includeDGF,114,115 monocyte chemoattractant protein-1,116

nd CXCR3 ligands.117 Functionally, PDGF-stimulatedhemotaxis provokes cell spreading at the tip, movementf the cell body towards the stimulant, and retraction ofrailing protrusions associated with transient myosinhosphorylation.118

In contrast to PDGF, adenosine blunts chemotaxis,hereby providing a counter-regulatory pathway that fixesells at sites of injury.119 Paradoxically, enhanced adeno-

ine signaling also may contribute to alcoholic fibrosis by i

timulating stellate cell fibrogenesis,120 which not onlyepresents a potential fibrogenic mechanism, but also

ay explain the protective effect of caffeine (which in-ibits adenosine generation) reported in epidemiologicurveys.121

Fibrogenesis. Collagen type I is the prototypeonstituent of the fibril-forming matrix in fibrotic liver,nd its expression is regulated both transcriptionally andosttranscriptionally as described earlier and in severaleviews.122–124

TGF�1, derived from both paracrine and autocrineources, remains the classic fibrogenic cytokine (see Ina-aki and Okazaki124 and Breitkopf et al125 for reviews).ignals downstream of TGF� converge on Smad pro-eins, which fine-tune and enhance the effects of TGF�uring stellate cell activation; Smads 2 and 3 are stimu-

atory whereas Smad 7 is inhibitory124,126,127 and is an-agonized by Id1.128 TGF�1 also stimulates collagenranscription in stellate cells through a hydrogen perox-de– and C/EBP�-dependent mechanism.129 The re-ponse of Smads in stellate cells evolves as injury be-omes chronic, further enhancing fibrogenesis.126,130

Connective tissue growth factor (CTGF/CCN2) is alsopotent fibrogenic signal towards stellate cells131–133 that

s up-regulated by hyperglycemia and hyperinsulin-mia.134 Although stimulation of CTGF production tra-itionally has been considered TGF�-dependent,135

GF�-independent regulation is increasingly likely asell.136 Moreover, TGF� stimulates CTGF primarily inepatocytes, not stellate cells,137,138 a notable exceptiono the general rule that cytokine signaling in stellate cellctivation typically is autocrine.

Neurohumoral signaling contributes to stellate cellesponses.139 In particular, cannabinoids have emerged asotent mediators of hepatic steatosis, stellate cell activa-ion, and fibrosis (reviewed in Mallat et al140), as well asrovoking the hemodynamic alterations associated withdvanced liver disease.141 Two receptors, CB1 and CB2,xert opposing effects, with CB1 a fibrogenic pathwaynd CB2 antifibrotic. Thus, antagonism of CB1 signalingn stellate cells has emerged as a promising antifibrotictrategy, as exemplified by the clinical agent RimonabantSanofi-Aventis, Paris, France).142 Conversely, agonism ofB2 receptors, which also are expressed by stellate cells,

everses fibrosis in experimental animals.143 The funda-ental challenge of developing cannabinoid therapeutics

or liver disease is to minimize central nervous systemffects because CB1 and CB2 receptors are expressedbundantly in brain. Similarly, opioids signal in stellateells and promote fibrogenesis,144,145 which is antago-ized by naltrexone. Finally, sympathetic neurotransmit-ers also contribute to activation pathways.146

Contractility. Contraction of stellate cells con-ributes to increased portal resistance during liver fibrosishat presumably is reversible before the thickened septae,

ntrahepatic shunts, and lobular distortion of cirrhosis
Page 7: Gastro Rvu-Slf 2008

dEaamsicurie

lljatbrmrarav

esbbttltcsbnir

ssaatscpadssns

abtstra

Lacnktvii

vuhpptp

Frdadstsa

May 2008 HEPATIC FIBROGENESIS 1661

evelop, leading to fixed increases in portal pressure.ven in earlier stages of fibrosis, activated stellate cellslready show features of smooth muscle–like cells, char-cterized by expression of a number of contractile fila-ents including � smooth muscle actin147 and myo-

in,148 which generate calcium-dependent and calcium-ndependent contractile forces that contribute to cellularontractility.149 –151 Culture models increasingly recapit-late many of these smooth muscle features, in part byestoring a more physiologic substratum, as well as byncluding other resident cell types in a co-culture system,specially Kupffer cells.152

As reviewed recently, stellate cells are recognized asiver-specific pericytes that contribute to angiogenesis iniver development, regeneration, and the response to in-ury.10 After partial hepatectomy, stellate cells migratelong with endothelial cells to establish vascular connec-ions with hepatocytes, thereby creating new sinusoidalranches.153 Although it is unclear to what extent theseesponses occurring during pure regeneration are

ounted during liver injury, the 2 responses—repair andegeneration— occur concurrently in chronic liver disease,nd thus similar angiogenic behavior is likely to underlieepair. Moreover, progressive fibrosis with angiogenesislso contributes to tumor vascularization in which acti-ated stellate cells play a vital role.10,154

As fibrosis advances, the collagenous bands typical ofnd-stage cirrhosis contain large numbers of activatedtellate cells.155 These cells progressively impede portallood flow by both constricting individual sinusoids andy contracting the cirrhotic liver, mediated by pathwayshat allow interaction with the ECM.156,157 At the sameime, stellate cell density and coverage of the sinusoidalumen increases.10,153 Endothelin-1 and nitric oxide arehe key opposing counter-regulators that control stellateell contractility, in addition to angiotensinogen II, eico-anoids, atrial natriuretic peptide, somatostatin, and car-on monoxide, among others (see Rockey155 and Rey-aert et al158 for reviews). Progressive development of

ntrahepatic shunts also is likely to require angiogenicesponses driven by stellate cells.159,160

The therapeutic implications of elucidating angiogenicignaling in liver fibrosis are not so clear. Although aimple paradigm would suggest that anti-angiogenicgents, for example, VEGF inhibitors, are an effectiventi-inflammatory and antifibrotic strategy,161 their long-erm impact on regeneration is uncertain. For example,ustained and complete inhibition of angiogenesis mightompromise hepatic blood flow and oxygen delivery, es-ecially because the liver has high metabolic demandsnd receives primarily venous blood. Thus, there is aelicate balance between the requirement for angiogene-is to preserve blood flow, and its negative impact on livertructure and function. Moreover, strategies to antago-ize contractile proteins also may have unintended con-

equences. For example, mice lacking � smooth muscle m

ctin protein in myofibroblasts have increased renal fi-rosis in experimental glomerulonephritis,162 suggestinghat �-actin induction may be a counter-regulatory re-ponse to enhanced fibrogenesis. Moreover, missense mu-ations of this protein have been linked to aortic aneu-ysms in human beings,163 indicating that the proteinlso may contribute to vascular integrity.

Inflammatory signaling. As reviewed earlier (seeiver Injury and Inflammation: Established Mediatorsnd New Players section), stellate cells have emerged asentral modulators of hepatic inflammation and immu-ity, and not just passive targets of inflammatory cyto-ines. In particular, a growing list of chemokines andheir cognate receptors serve the dual function of pro-oking further fibrogenesis, as well as interacting withnflammatory cells to modify the immune response dur-ng injury.164 –166

Matrix Degradation and Resolution ofFibrosisEvidence that fibrosis and even cirrhosis are re-

ersible has intensified interest in understanding the reg-lation of matrix degradation and fibrosis resolution, inopes that therapies might exploit those endogenousathways that reverse disease (Figure 3). Simplisticallyut, this response would be considered therapeutic ma-rix degradation, whereas early liver injury is marked byathologic matrix degradation that disrupts hepatic ho-

igure 3. Pathways of matrix degradation in fibrosis progression andegression. Macrophages have assumed an important role in matrixegradation, which is profibrogenic during progression of fibrosis butntifibrotic during fibrosis resolution. Although key sources of matrixegrading activity are uncertain, it seems increasingly likely that bothcar-associated macrophages and stellate cells are sources of intersti-ial collagenases. At the same time, decreased TIMP-1 fosters apopto-is of fibrogenic myofibroblasts. Figure adapted from studies of Duffieldnd Iredale177 (and accompanying editorial, pp 29–32).

eostasis. Thus, in early liver injury matrix-degrading

Page 8: Gastro Rvu-Slf 2008

pMpfit

fmss

b(M

dmtHlStiamw

staaiTpadegtarisSlcpkwwlihta

lgtjcahp

esciIcwpvorCTh

tptn

awasqa

adp

cAfaeicgm

1662 SCOTT L. FRIEDMAN GASTROENTEROLOGY Vol. 134, No. 6

roteases with activity towards type IV collagen (eg,MP-2), degrade the low-density basement membrane

resent in the subendothelial space. Its replacement withbril-forming matrix has deleterious effects on differen-iated cell function, in particular on hepatocytes.

Enzymes controlling matrix degradation comprise aamily of matrix-metalloproteinases (also known as

atrixins), which are calcium-dependent enzymes thatpecifically degrade collagens and noncollagenous ECMubstrates.12,167

Stellate cells are a key source of the basement mem-rane proteases MMP-2,168 MMP-9,169 and stromelysinMMP-3),170 as well as the interstitial collagenase

MP-13 (the rodent equivalent of MMP-1).171

A major determinant of progressive fibrosis is failure toegrade the increased fibril-forming, or interstitial, scaratrix. MMP-1 is the main protease that can degrade

ype I collagen, the principal collagen in fibrotic liver.owever, sources of this enzyme are not as clearly estab-

ished as for the type IV collagenases MMP-2 and MMP-9.tellate cells express modest levels of MMP-1 mRNA andhus it is uncertain whether this represents the primarynterstitial collagenase responsible for matrix resorptions liver fibrosis regresses. Alternative interstitial proteasesight include either matrix type 1 MMP or even MMP-2,hich also displays some interstitial collagenase activity.The cross-linking of collagen by lysyl oxidase and tis-

ue transglutaminase, and the maturation of hepatic scarhrough the action of a disintegrin and metalloprotein-se with thrombospondin–type repeats metalloprotein-se with thrombospondin type I motif (ADAMTS2) aremportant determinants of hepatic fibrosis reversibility.he long-standing clinical dogma that the slower theace of injury, the less reversible the scar, is borne out bynimal studies in which even advanced fibrosis of shorturation is reversible, which is limited primarily by thextent of collagen cross-linking caused by tissue trans-lutaminase.172 Moreover, as advanced fibrosis resolves,he micronodules typical of active cirrhosis dissolve, co-lescing into macronodules.172 This finding correlatesemarkably well with recent clinical data showing thatncreased septal thickness and smaller nodule size areignificant predictors of poorer clinical outcomes.173

imilar studies have been performed in knockout miceacking ADAMTS2, which catalyzes the N-propeptide ex-ision of procollagens I, III, and V, which allows theolymerization of collagen fibrils.174 In the ADAMTS2nockout animals, the extent of liver injury after CCl4

as similar to that of wild-type mice, yet fibrosis reversalas slightly faster, associated with much less dense col-

agen fibrils, and smaller fibril diameters.175 Recently,ncreased proteolytic activity ascribed to ADAMS-13 alsoas been reported in activated stellate cells.176 However,he native substrate(s) and biological role of this protease

re not known. l

Hepatic macrophages are re-emerging as critical regu-ators of matrix remodeling. An elegant study using aenetic model that allows for the selective, timed deple-ion of macrophages during different stages of liver in-ury and resolution has shown divergent roles for theseells.177 During progression of liver fibrosis, macrophagesugment fibrogenesis, whereas during resolution theyasten matrix degradation, primarily through increasedroduction of MMP-13.178

Inactivation of proteases by binding to TIMPs also ismerging as an important locus of control12 becauseustained production of these proteins during liver injuryould inhibit the activity of interstitial collagenases, lead-ng to reduced degradation of the accumulating matrix.n addition, TIMP-1 is anti-apoptotic towards stellateells,179 and thus its sustained expression in liver injuryill enlarge the population of activated stellate cells byreventing their clearance. In support of TIMP’s role inivo, either transgenic over expression of TIMP-1 in liver,r administration of TIMP-neutralizing antibodies, delayegression of liver fibrosis in experimental animals.180

onversely, the use of MMP-9 mutant proteins asIMP-1 scavengers reduces fibrosis accumulation by en-ancing matrix resorption.181

Stellate cells express uroplasminogen activator recep-or and its inhibitor, as well as other components of thelasmin system.182–184 Collectively, these findings suggesthat stellate cells contain most, if not all, of the moleculesecessary to either activate or inhibit metalloproteinases.Clearance of activated stellate cells by apoptosis remains

n appealing target for antifibrotic therapy because thisould use an endogenous pathway of fibrosis regression. Inddition, data from cultured stellate cells185,186 and liverlices187 suggest that activated cells also can revert to auiescent phenotype, such that this response may be anntifibrotic pathway worth exploiting.

Disease-Specific Pathways of HepaticFibrosisAlthough key pathways of stellate cell activation

re common to all forms of liver injury and fibrosis,isease-specific pathways are being unearthed as well,articularly in ASH and NASH, and in HCV disease.

NASH and ASHThe accelerating obesity epidemic is tied to an in-

reasing prevalence of NASH and subsequent cirrhosis.188

dipokines mediate fibrogenesis and many hepatic mani-estations of obesity.189,190 Specifically, leptin, a circulatingdipogenic hormone, promotes stellate cell fibrogenesis andnhances TIMP-1 expression,191–193 which is associated withncreased leptin signaling.191 Concurrently, adiponectin, aounter-regulatory hormone that antagonizes the fibro-enic activity of leptin is reduced in hepatic fibrosis,194 andice lacking adiponectin have enhanced fibrosis after toxic

iver injury.195 Equally important to fibrosis is insulin resis-

Page 9: Gastro Rvu-Slf 2008

tHaarradseet

CdealisHllanHg

w

htbwtdfia

cefuartpcmicetshc

FtTM ech

May 2008 HEPATIC FIBROGENESIS 1663

ance per se, whether associated with steatosis196 orCV.197,198 As noted earlier, cannabinoids also mediate ste-

tosis, and daily cannabis use is a risk factor for steatosisnd fibrosis in HCV.199,200 Interestingly, a recent study di-ectly links CB1 receptor signaling to alcoholic steatosis inodents fed a liquid ethanol diet.201 Moreover, in thesenimals, activated stellate cells are a key source of the en-ogenous cannabinoid, 2-AG, which drives increased CB1ignaling.201 As noted earlier, oxidant stress associated withthanol metabolism is an important stimulus to fibrogen-sis. In contrast, aldehydes, although fibrogenic, are unlikelyo account entirely for ethanol-induced fibrosis.15

HCV and HIVStellate cells express the putative HCV receptors

D80, LDL receptor, and C1q, raising the possibility ofirect HCV infection in vivo, which has not yet beenstablished. Moreover, expression of HCV nonstructuralnd core proteins induces stellate cell proliferation, re-ease of inflammatory signals,202 and CTGF,203 althoughnteraction of HCV E2 protein stimulates MMP-2 expres-ion.204 Furthermore, hepatocytes harboring replicatingCV in culture produce fibrogenic stimuli towards stel-

ate cells.205 In HCV-infected liver, chemokines promoteymphocyte recruitment.165 HCV proteins also may inter-ct directly with sinusoidal endothelium.206 Remarkably,o studies have reported potential interactions betweenBV and stellate cells, or HBV-specific pathways of fibro-

enesis.The increased rate of fibrosis in patients co-infected

igure 4. Possible links between stellate cells, fibrosis, regeneration, ahat may promote progenitor cell expansion, the outcome of which couhe possibility also exists that stellate cells may harbor the potential tooreover, fibrosis promotes hepatocarcinogenesis through unknown m

ith HCV and HIV compared with those with HCV alone t

as been well documented.41 The reduced CD4/CD8 ra-io typical of HIV infection has been invoked as a cause,ecause CD8 cells may be relatively fibrogenic comparedith CD4 cells; however, recent preliminary data addi-

ionally suggest that hepatic stellate cells may be infectedirectly by HIV,207 which also might account for whybrosis progression is slowed when HIV is suppressed byntiretroviral therapy in co-infected patients.208

Links Between Stellate and ProgenitorCells, Fibrosis, and CancerThe remarkable phenotypic plasticity of stellate

ells, combined with the recent demonstration that theyxpress the stem cell marker CD133,209 have raised theascinating prospect that they are true progenitor cells (Fig-re 4). Further studies are required to establish bona fidend robust pluripotency, but intriguing possibilities areaised. First, activated stellate cells appear to contribute tohe stem cell niche based on histologic studies,210 raising theossibility that they are actually differentiating into stemells directly. Second, could this cellular behavior provide aissing link between fibrosis and hepatocellular cancer that

s derived from stem cells? Although epithelial to mesen-hymal transition is well established, the possibility of mes-nchymal to epithelial transition, while still quite specula-ive at this point, should not be ignored. Support for thisuggestion includes the presence in stellate cells of bothedgehog211,212 and Wnt signaling,213 two pathways impli-ated in stem cell differentiation and cancer.214 Moreover,

ncer. Upon liver injury, activated stellate cells release paracrine factorseither hepatic regeneration and/or promotion of hepatocellular cancer.sdifferentiate into progenitor cells directly, which remains speculative.anisms that may include release of survival signals.

nd cald be

tran

he near-absolute requirement for fibrosis to occur before

Page 10: Gastro Rvu-Slf 2008

hHttalftfit

ptstsintmndbpMwcdcdiB

psbnogo

FWgpse1dcih

1664 SCOTT L. FRIEDMAN GASTROENTEROLOGY Vol. 134, No. 6

epatocellular carcinoma develops in patients with chronicCV remains completely unexplained. Potential explana-

ions have included the presence of secreted survival factorshat prevent apoptosis of DNA-damaged hepatocytes andctivated stellate cells (eg, Gas6215), reduced tumor surveil-ance owing to decreasing natural killer cell number andunction, and/or the accelerated shortening of telomereshat accompanies progressive fibrosis. The question of howbrosis promotes hepatocellular carcinoma is a vital onehat demands clearer answers.

Clinical and Translational ImplicationsThe tightening links between the biology of he-

atic fibrosis and clinical expression of disease attest tohe importance of continued basic and translational re-earch into mechanisms of hepatic fibrogenesis. In par-icular, newly uncovered correlations between matrixtiffness and fibrogenesis, ECM cross-linking and revers-bility, and both cirrhotic nodule size and septal thick-ess with clinical outcomes, have emerged from cell cul-ure and animal studies, yet they lead directly to new

odes of diagnosis and therapy. Moreover, cirrhosis cano longer be viewed as a single, irreversible end stage ofisease but rather as a much broader category subdividedy progressive stages of ECM accumulation, nodule size,ortal pressure, reversibility, and clinical risk (Figure 5).ore refined and rigorous characterization of cirrhosisill be essential for accurate randomization of patients in

linical trials of antifibrotic therapies and stratification ofisease risk, possibly including the risk of hepatocellulararcinoma. Indeed, a key lesson from antifibrotic trials toate is that fibrosis may continue to accrue rapidly even

n patients with cirrhosis when therapy is not effective.216

igure 5. Cirrhosis is a series of progressive stages, not a single stage.ithin the spectrum of cirrhosis, the disease is characterized by pro-

ressive increases in hepatic venous pressure gradient (HVPG), decom-ensation, and matrix cross-linking, associated with shrinking noduleize, thickening septae, and enhanced risk of decompensation. Forach 1-mm increase in HVPG the risk of decompensation increases by1%. Concepts presented here are not rigorously supported by primaryata for all features, but rather are intended to convey the progressivehanges that underlie deterioration in patients with chronic hepatic

njury and fibrosis. Stages are based on data from D’Amico et al.218 HE,epatic encephalopathy; VH, variceal hemorrhage.

ased on this lesson, it is clear that trials in such cirrhotic

atients will need to be lengthy, with the use of moreensitive and specific biomarkers that do not rely oniopsy alone.217 Further progress in understanding, diag-osing, and treating hepatic fibrosis will continue to relyn the exploration of fundamental mechanisms of fibro-enesis, which is certain to lead to a meaningful impactn the prognosis of patients with chronic liver disease.

References

1. Friedman SL, Rockey DC, Bissell DM. Hepatic fibrosis 2006:report of the Third AASLD Single Topic Conference. Hepatology2007;45:242–249.

2. Hadziyannis SJ, Tassopoulos NC, Heathcote EJ, et al. Long-termtherapy with adefovir dipivoxil for HBeAg-negative chronic hepa-titis B for up to 5 years. Gastroenterology 2006;131:1743–1751.

3. Bruno S, Stroffolini T, Colombo M, et al. Sustained virologicalresponse to interferon-alpha is associated with improved out-come in HCV-related cirrhosis: a retrospective study. Hepatology2007;45:579–587.

4. Veldt BJ, Heathcote EJ, Wedemeyer H, et al. Sustained virologicresponse and clinical outcomes in patients with chronic hepati-tis C and advanced fibrosis. Ann Intern Med 2007;147:677–684.

5. Omary MB, Lugea A, Lowe AW, et al. The pancreatic stellate cell:a star on the rise in pancreatic diseases. J Clin Invest 2007;117:50–59.

6. Friedman SL. Seminars in medicine of the Beth Israel Hospital,Boston. The cellular basis of hepatic fibrosis. Mechanisms andtreatment strategies. N Engl J Med 1993;328:1828–1835.

7. Friedman SL. Molecular regulation of hepatic fibrosis, an inte-grated cellular response to tissue injury. J Biol Chem 2000;275:2247–2250.

8. Schluger LK, Sheiner PA, Thung SN, et al. Severe recurrentcholestatic hepatitis C following orthotopic liver transplantation.Hepatology 1996;23:971–976.

9. Bonnard P, Lescure FX, Amiel C, et al. Documented rapid courseof hepatic fibrosis between two biopsies in patients coinfectedby HIV and HCV despite high CD4 cell count. J Viral Hepat2007;14:806–811.

10. Lee JS, Semela D, Iredale J, et al. Sinusoidal remodeling andangiogenesis: a new function for the liver-specific pericyte?Hepatology 2007;45:817–825.

11. Dev A, Patel K, Conrad A, et al. Relationship of smoking andfibrosis in patients with chronic hepatitis C. Clin GastroenterolHepatol 2006;4:797–801.

12. Iredale JP. Models of liver fibrosis: exploring the dynamic natureof inflammation and repair in a solid organ. J Clin Invest 2007;117:539–548.

13. Zeisberg M, Yang C, Martino M, et al. Fibroblasts derive fromhepatocytes in liver fibrosis via epithelial to mesenchymal tran-sition. J Biol Chem 2007;282:23337–23347.

14. De Minicis S, Seki E, Uchinami H, et al. Gene expressionprofiles during hepatic stellate cell activation in culture and invivo. Gastroenterology 2007;132:1937–1946.

15. Parola M, Robino G. Oxidative stress-related molecules and liverfibrosis. J Hepatol 2001;35:297–306.

16. Jaeschke H. Mechanisms of liver injury. II. Mechanisms ofneutrophil-induced liver cell injury during hepatic ischemia-reper-fusion and other acute inflammatory conditions. Am J Physiol2006;290:G1083–G1088.

17. Castillo T, Koop DR, Kamimura S, et al. Role of cytochromeP-450 2E1 in ethanol-, carbon tetrachloride- and iron-dependent

microsomal lipid peroxidation. Hepatology 1992;16:992–996.
Page 11: Gastro Rvu-Slf 2008

May 2008 HEPATIC FIBROGENESIS 1665

18. Chitturi S, Farrell GC. Etiopathogenesis of nonalcoholic steato-hepatitis. Semin Liver Dis 2001;21:27–41.

19. De Minicis S, Brenner DA. NOX in liver fibrosis. Arch BiochemBiophys 2007;462:266–272.

20. Bataller R, Schwabe RF, Choi YH, et al. NADPH oxidase signaltransduces angiotensin II in hepatic stellate cells and is criticalin hepatic fibrosis. J Clin Invest 2003;112:1383–1394.

21. Nussler AK, Di Silvio M, Billiar TR, et al. Stimulation of the nitricoxide synthase pathway in human hepatocytes by cytokines andendotoxin. J Exp Med 1992;176:261–264.

22. Venkatraman A, Shiva S, Wigley A, et al. The role of iNOS inalcohol-dependent hepatotoxicity and mitochondrial dysfunctionin mice. Hepatology 2004;40:565–573.

23. Jaeschke H. Inflammation in response to hepatocellular apopto-sis. Hepatology 2002;35:964–966.

24. Canbay A, Friedman S, Gores GJ. Apoptosis: the nexus of liverinjury and fibrosis. Hepatology 2004;39:273–278.

25. Zhan SS, Jiang JX, Wu J, et al. Phagocytosis of apoptotic bodiesby hepatic stellate cells induces NADPH oxidase and is associ-ated with liver fibrosis in vivo. Hepatology 2006;43:435–443.

26. Watanabe A, Hashmi A, Gomes DA, et al. Apoptotic hepatocyteDNA inhibits hepatic stellate cell chemotaxis via Toll-like recep-tor 9. Hepatology 2007;46:1509–1518.

27. Takehara T, Tatsumi T, Suzuki T, et al. Hepatocyte-specific disrup-tion of Bcl-xL leads to continuous hepatocyte apoptosis and liverfibrotic responses. Gastroenterology 2004;127:1189–1197.

28. Canbay A, Higuchi H, Bronk SF, et al. Fas enhances fibrogenesisin the bile duct ligated mouse: a link between apoptosis andfibrosis. Gastroenterology 2002;123:1323–1330.

29. Pockros PJ, Schiff ER, Shiffman ML, et al. Oral IDN-6556, anantiapoptotic caspase inhibitor, may lower aminotransferaseactivity in patients with chronic hepatitis C. Hepatology 2007;46:324–329.

30. Taimr P, Higuchi H, Kocova E, et al. Activated stellate cellsexpress the TRAIL receptor-2/death receptor-5 and undergoTRAIL-mediated apoptosis. Hepatology 2003;37:87–95.

31. Wright M, Issa R, Smart D, et al. Gliotoxin stimulates theapoptosis of human and rat hepatic stellate cells and enhancesthe resolution of liver fibrosis in rats. Gastroenterology 2001;121:685–698.

32. Anan A, Baskin-Bey ES, Bronk SF, et al. Proteasome inhibitioninduces hepatic stellate cell apoptosis. Hepatology 2006;43:335–344.

33. Jaeschke H, Gujral JS, Bajt ML. Apoptosis and necrosis in liverdisease. Liver Int 2004;24:85–89.

34. Wagner H, Bauer S. All is not Toll: new pathways in DNA recog-nition. J Exp Med 2006;203:265–268.

35. Paik YH, Schwabe RF, Bataller R, et al. Toll-like receptor 4mediates inflammatory signaling by bacterial lipopolysaccharidein human hepatic stellate cells. Hepatology 2003;37:1043–1055.

36. Hua J, Qiu de K, Li JQ, et al. Expression of Toll-like receptor 4 inrat liver during the course of carbon tetrachloride-induced liverinjury. J Gastroenterol Hepatol 2007;22:862–869.

37. Seki E, De Minicis S, Osterreicher CH, et al. TLR4 enhancesTGF-beta signaling and hepatic fibrosis. Nat Med 2007;13:1324–1332.

38. Huang H, Shiffman ML, Friedman S, et al. A 7 gene signatureidentifies the risk of developing cirrhosis in patients with chronichepatitis C. Hepatology 2007;46:297–306.

39. Mehal WZ, Friedman SL. The role of inflammation and immunityin the pathogenesis of liver fibrosis. In: Gershwin ME, VeirlingJM, Manns MP, eds. Liver immunology. Vol 2. Totowa, NJ:Humana Press, 2007:99–109.

40. Safadi R, Ohta M, Alvarez CE, et al. Immune stimulation of

hepatic fibrogenesis by CD8 cells and attenuation by transgenic

interleukin-10 from hepatocytes. Gastroenterology 2004;127:870–882.

41. Benhamou Y, Bochet M, Di Martino V, et al. Liver fibrosisprogression in human immunodeficiency virus and hepatitis Cvirus coinfected patients. The Multivirc Group. Hepatology1999;30:1054–1058.

42. Radaeva S, Sun R, Jaruga B, et al. Natural killer cells ame-liorate liver fibrosis by killing activated stellate cells inNKG2D-dependent and tumor necrosis factor-related apopto-sis-inducing ligand-dependent manners. Gastroenterology2006;130:435–452.

43. Melhem A, Muhanna N, Bishara A, et al. Anti-fibrotic activity ofNK cells in experimental liver injury through killing of activatedHSC. J Hepatol 2006;45:60–71.

44. Radaeva S, Wang L, Radaev S, et al. Retinoic acid signalingsensitizes hepatic stellate cells to NK cell killing via upregula-tion of NK cell activating ligand RAE1. Am J Physiol 2007;293:G809–G816.

45. Jeong WI, Park O, Gao B. Abrogation of the anti-fibrotic effects ofnatural killer cells/interferon-gamma contributes to alcohol ac-celeration of liver fibrosis. Gastroenterology 2008;134:248–258.

46. Winau F, Hegasy G, Weiskirchen R, et al. Ito cells are liver-resident antigen-presenting cells for activating T cell responses.Immunity 2007;26:117–129.

47. Chen CH, Kuo LM, Chang Y, et al. In vivo immune modulatoryactivity of hepatic stellate cells in mice. Hepatology 2006;44:1171–1181.

48. Novobrantseva TI, Majeau GR, Amatucci A, et al. Attenuatedliver fibrosis in the absence of B cells. J Clin Invest 2005;115:3072–3082.

49. Wells RG, Kruglov E, Dranoff JA. Autocrine release of TGF-betaby portal fibroblasts regulates cell growth. FEBS Lett 2004;559:107–110.

50. Jhandier MN, Kruglov EA, Lavoie EG, et al. Portal fibroblastsregulate the proliferation of bile duct epithelia via expression ofNTPDase2. J Biol Chem 2005;280:22986–22992.

51. Beaussier M, Wendum D, Schiffer E, et al. Prominent contribu-tion of portal mesenchymal cells to liver fibrosis in ischemic andobstructive cholestatic injuries. Lab Invest 2007;87:292–303.

52. Kisseleva T, Uchinami H, Feirt N, et al. Bone marrow-derivedfibrocytes participate in pathogenesis of liver fibrosis. J Hepatol2006;45:429–438.

53. Forbes SJ, Russo FP, Rey V, et al. A significant proportion ofmyofibroblasts are of bone marrow origin in human liver fibrosis.Gastroenterology 2004;126:955–963.

54. Kruglov EA, Nathanson RA, Nguyen T, et al. Secretion of MCP-1/CCL2 by bile duct epithelia induces myofibroblastic transdif-ferentiation of portal fibroblasts. Am J Physiol 2006;290:G765–G771.

55. Dranoff JA, Ogawa M, Kruglov EA, et al. Expression of P2Ynucleotide receptors and ectonucleotidases in quiescent andactivated rat hepatic stellate cells. Am J Physiol 2004;287:G417–G424.

56. Sakaida I, Terai S, Yamamoto N, et al. Transplantation of bonemarrow cells reduces CCl(4)-induced liver fibrosis in mice. Hepa-tology 2004;40:1304–1311.

57. Terai S, Ishikawa T, Omori K, et al. Improved liver function inpatients with liver cirrhosis after autologous bone marrow cellinfusion therapy. Stem Cells 2006;24:2292–2298.

58. Nakamura T, Torimura T, Sakamoto M, et al. Significance andtherapeutic potential of endothelial progenitor cell transplanta-tion in a cirrhotic liver rat model. Gastroenterology 2007;133:91–107 e1.

59. Wang B, Dolinski BM, Kikuchi N, et al. Role of alphavbeta6integrin in acute biliary fibrosis. Hepatology 2007;46:1404–

1412.
Page 12: Gastro Rvu-Slf 2008

1666 SCOTT L. FRIEDMAN GASTROENTEROLOGY Vol. 134, No. 6

60. Jarnagin WR, Rockey DC, Koteliansky VE, et al. Expression ofvariant fibronectins in wound healing: cellular source and bio-logical activity of the EIIIA segment in rat hepatic fibrogenesis.J Cell Biol 1994;127:2037–2048.

61. George J, Wang SS, Sevcsik AM, et al. Transforming growthfactor-beta initiates wound repair in rat liver through induction ofthe EIIIA-fibronectin splice isoform. Am J Pathol 2000;156:115–124.

62. Yang C, Zeisberg M, Mosterman B, et al. Liver fibrosis: insightsinto migration of hepatic stellate cells in response to extracel-lular matrix and growth factors. Gastroenterology 2003;124:147–159.

63. Zhou X, Murphy FR, Gehdu N, et al. Engagement of alphavbeta3integrin regulates proliferation and apoptosis of hepatic stellatecells. J Biol Chem 2004;279:23996–24006.

64. Melton AC, Soon RK Jr, Park JG, et al. Focal adhesion disas-sembly is an essential early event in hepatic stellate cell che-motaxis. Am J Physiol 2007;293:G1272–G1280.

65. Shafiei MS, Rockey DC. The role of integrin-linked kinase in liverwound healing. J Biol Chem 2006;281:24863–24872.

66. Zhang Y, Ikegami T, Honda A, et al. Involvement of integrin-linked kinase in carbon tetrachloride-induced hepatic fibrosis inrats. Hepatology 2006;44:612–622.

67. Yee HF Jr. Rho directs activation-associated changes in rathepatic stellate cell morphology via regulation of the actin cy-toskeleton. Hepatology 1998;28:843–850.

68. Choi SS, Sicklick JK, Ma Q, et al. Sustained activation of Rac1in hepatic stellate cells promotes liver injury and fibrosis inmice. Hepatology 2006;44:1267–1277.

69. Schuppan D, Schmid M, Somasundaram R, et al. Collagens inthe liver extracellular matrix bind hepatocyte growth factor. Gas-troenterology 1998;114:139–152.

70. Schuppan D, Ruehl M, Somasundaram R, et al. Matrix as amodulator of hepatic fibrogenesis. Semin Liver Dis 2001;21:351–372.

71. Wells RG. The role of matrix stiffness in regulating cell behavior.Hepatology 2008;47:1394–1400.

72. Georges PC, Hui JJ, Gombos Z, et al. Increased stiffness of therat liver precedes matrix deposition: implications for fibrosis.Am J Physiol 2007;293:G1147–G1154.

73. Vizzutti F, Arena U, Romanelli RG, et al. Liver stiffness measure-ment predicts severe portal hypertension in patients with HCV-related cirrhosis. Hepatology 2007;45:1290–1297.

74. Kazemi F, Kettaneh A, N’Kontchou G, et al. Liver stiffnessmeasurement selects patients with cirrhosis at risk of bearinglarge oesophageal varices. J Hepatol 2006;45:230–235.

75. Rockey DC. Noninvasive assessment of liver fibrosis and portalhypertension with transient elastography. Gastroenterology2008;134:8–14.

76. Yin M, Talwalkar JA, Glaser KJ, et al. Assessment of hepaticfibrosis with magnetic resonance elastography. Clin Gastroen-terol Hepatol 2007;5:1207–1213 e2.

77. Talwalkar JA, Yin M, Fidler JL, et al. Magnetic resonance imagingof hepatic fibrosis: emerging clinical applications. Hepatology2008;47:332–342.

78. Valencia-Sanchez MA, Liu J, Hannon GJ, et al. Control of trans-lation and mRNA degradation by miRNAs and siRNAs. GenesDev 2006;20:515–524.

79. Rippe RA, Brenner DA. From quiescence to activation: generegulation in hepatic stellate cells. Gastroenterology 2004;127:1260–1262.

80. Mann J, Oakley F, Akiboye F, et al. Regulation of myofibroblasttransdifferentiation by DNA methylation and MeCP2: implica-tions for wound healing and fibrogenesis. Cell Death Differ

2007;14:275–285.

81. Tsukamoto H, She H, Hazra S, et al. Anti-adipogenic regulationunderlies hepatic stellate cell transdifferentiation. J Gastroen-terol Hepatol 2006;21(Suppl 3):S102–S105.

82. Kalinichenko VV, Bhattacharyya D, Zhou Y, et al. Foxf1 �/- miceexhibit defective stellate cell activation and abnormal liver re-generation following CCl4 injury. Hepatology 2003;37:107–117.

83. Jochum W, Passegue E, Wagner EF. AP-1 in mouse developmentand tumorigenesis. Oncogene 2001;20:2401–2412.

84. Smart DE, Green K, Oakley F, et al. JunD is a profibrogenictranscription factor regulated by Jun N-terminal kinase-indepen-dent phosphorylation. Hepatology 2006;44:1432–1440.

85. Buck M, Poli V, Hunter T, et al. C/EBPbeta phosphorylation byRSK creates a functional XEXD caspase inhibitory box critical forcell survival. Mol Cell 2001;8:807–816.

86. Wandzioch E, Kolterud A, Jacobsson M, et al. Lhx2-/- micedevelop liver fibrosis. Proc Natl Acad Sci U S A 2004;101:16549–16554.

87. Adachi M, Osawa Y, Uchinami H, et al. The forkhead transcrip-tion factor FoxO1 regulates proliferation and transdifferentiationof hepatic stellate cells. Gastroenterology 2007;132:1434–1446.

88. Yavrom S, Chen L, Xiong S, et al. Peroxisome proliferator-activated receptor gamma suppresses proximal alpha1(I) colla-gen promoter via inhibition of p300-facilitated NF-I binding toDNA in hepatic stellate cells. J Biol Chem 2005;280:40650–40659.

89. Galli A, Crabb DW, Ceni E, et al. Antidiabetic thiazolidinedionesinhibit collagen synthesis and hepatic stellate cell activation invivo and in vitro. Gastroenterology 2002;122:1924–1940.

90. Haughton EL, Tucker SJ, Marek CJ, et al. Pregnane x receptoractivators inhibit human hepatic stellate cell transdifferentiationin vitro. Gastroenterology 2006;131:194–209.

91. Marek CJ, Tucker SJ, Konstantinou DK, et al. Pregnenolone-16alpha-carbonitrile inhibits rodent liver fibrogenesis via PXR(pregnane X receptor)-dependent and PXR-independent mecha-nisms. Biochem J 2005;387:601–608.

92. Okuno M, Sato T, Kitamoto T, et al. Increased 9,13-di-cis-retinoic acid in rat hepatic fibrosis: implication for a potentiallink between retinoid loss and TGF-beta mediated fibrogenesisin vivo. J Hepatol 1999;30:1073–1080.

93. Hellemans K, Grinko I, Rombouts K, et al. All-trans and 9-cisretinoic acid alter rat hepatic stellate cell phenotype differen-tially. Gut 1999;45:134–142.

94. Oakley F, Meso M, Iredale JP, et al. Inhibition of inhibitor ofkappaB kinases stimulates hepatic stellate cell apoptosis andaccelerated recovery from rat liver fibrosis. Gastroenterology2005;128:108–120.

95. Mann J, Oakley F, Akiboye F, et al. Regulation of myofibroblasttransdifferentiation by DNA methylation and MeCP2: implica-tions for wound healing and fibrogenesis. Cell Death Differ2006;14:275–285.

96. Habens F, Srinivasan N, Oakley F, et al. Novel sulfasalazineanalogues with enhanced NF-kB inhibitory and apoptosis pro-moting activity. Apoptosis 2005;10:481–491.

97. Stefanovic B, Hellerbrand C, Holcik M, et al. Posttranscriptionalregulation of collagen alpha1(I) mRNA in hepatic stellate cells.Mol Cell Biol 1997;17:5201–5209.

98. Stefanovic B, Hellerbrand C, Brenner DA. Regulatory role of theconserved stem-loop structure at the 5’ end of collagenalpha1(I) mRNA. Mol Cell Biol 1999;19:4334–4342.

99. Fritz D, Stefanovic B. RNA-binding protein RBMS3 is expressedin activated hepatic stellate cells and liver fibrosis and in-creases expression of transcription factor Prx1. J Mol Biol

2007;371:585–595.
Page 13: Gastro Rvu-Slf 2008

1

1

1

1

1

1

1

1

1

1

1

1

1

1

1

1

1

1

1

1

1

1

1

1

1

1

1

1

1

1

1

1

1

1

1

1

1

1

1

1

May 2008 HEPATIC FIBROGENESIS 1667

00. Borkham-Kamphorst E, van Roeyen CR, Ostendorf T, et al.Pro-fibrogenic potential of PDGF-D in liver fibrosis. J Hepatol2007;46:1064–1074.

01. Wong L, Yamasaki G, Johnson RJ, et al. Induction of beta-platelet-derived growth factor receptor in rat hepatic lipocytesduring cellular activation in vivo and in culture. J Clin Invest1994;94:1563–1569.

02. Pinzani M, Milani S, Grappone C, et al. Expression of platelet-derived growth factor in a model of acute liver injury. Hepatology1994;19:701–707.

03. Czochra P, Klopcic B, Meyer E, et al. Liver fibrosis induced byhepatic overexpression of PDGF-B in transgenic mice. J Hepatol2006;45:419–428.

04. Campbell JS, Hughes SD, Gilbertson DG, et al. Platelet-derivedgrowth factor C induces liver fibrosis, steatosis, and hepatocel-lular carcinoma. Proc Natl Acad Sci U S A 2005;102:3389–3394.

05. Pinzani M, Marra F. Cytokine receptors and signaling in hepaticstellate cells. Semin Liver Dis 2001;21:397–416.

06. Lechuga CG, Hernandez-Nazara ZH, Hernandez E, et al. PI3K isinvolved in PDGF-beta receptor upregulation post-PDGF-BB treat-ment in mouse HSC. Am J Physiol 2006;291:G1051–G1061.

07. Rovida E, Navari N, Caligiuri A, et al. ERK5 differentially regu-lates PDGF-induced proliferation and migration of hepatic stel-late cells. J Hepatol 2008;48:107–115.

08. Di Sario A, Bendia E, Taffetani S, et al. Selective Na�/H�exchange inhibition by cariporide reduces liver fibrosis in therat. Hepatology 2003;37:256–266.

09. Yoshiji H, Kuriyama S, Yoshii J, et al. Vascular endothelialgrowth factor and receptor interaction is a prerequisite for mu-rine hepatic fibrogenesis. Gut 2003;52:1347–1354.

10. Marra F, Grandaliano G, Valente AJ, et al. Thrombin stimulatesproliferation of liver fat-storing cells and expression of monocytechemotactic protein-1: potential role in liver injury. Hepatology1995;22:780–787.

11. Marra F, DeFranco R, Grappone C, et al. Expression of thethrombin receptor in human liver: up-regulation during acute andchronic injury. Hepatology 1998;27:462–471.

12. Steiling H, Muhlbauer M, Bataille F, et al. Activated hepaticstellate cells express keratinocyte growth factor in chronic liverdisease. Am J Pathol 2004;165:1233–1241.

13. Yu C, Wang F, Jin C, et al. Role of fibroblast growth factor type1 and 2 in carbon tetrachloride-induced hepatic injury and fibro-genesis. Am J Pathol 2003;163:1653–1662.

14. Ikeda K, Wakahara T, Wang YQ, et al. In vitro migratory potentialof rat quiescent hepatic stellate cells and its augmentation bycell activation. Hepatology 1999;29:1760–1767.

15. Kinnman N, Hultcrantz R, Barbu V, et al. PDGF-mediated che-moattraction of hepatic stellate cells by bile duct segments incholestatic liver injury. Lab Invest 2000;80:697–707.

16. Marra F, Romanelli RG, Giannini C, et al. Monocyte chemotacticprotein-1 as a chemoattractant for human hepatic stellate cells.Hepatology 1999;29:140–148.

17. Bonacchi A, Romagnani P, Romanelli RG, et al. Signal transduc-tion by the chemokine receptor CXCR3: activation of Ras/ERK,Src, and phosphatidylinositol 3-kinase/Akt controls cell migra-tion and proliferation in human vascular pericytes. J Biol Chem2001;276:9945–9954.

18. Melton AC, Yee HF. Hepatic stellate cell protrusions coupleplatelet-derived growth factor-BB to chemotaxis. Hepatology2007;45:1446–1453.

19. Hashmi AZ, Hakim W, Kruglov EA, et al. Adenosine inhibitscytosolic calcium signals and chemotaxis in hepatic stellatecells. Am J Physiol 2007;292:G395–G401.

20. Chan ES, Montesinos MC, Fernandez P, et al. Adenosine A(2A)receptors play a role in the pathogenesis of hepatic cirrhosis.

Br J Pharmacol 2006;148:1144–1155.

21. Ruhl CE, Everhart JE. Coffee and tea consumption are associ-ated with a lower incidence of chronic liver disease in the UnitedStates. Gastroenterology 2005;129:1928–1936.

22. Tsukada S, Parsons CJ, Rippe RA. Mechanisms of liver fibrosis.Clin Chim Acta 2006;364:33–60.

23. Stefanovic B, Stefanovic L, Schnabl B, et al. TRAM2 proteininteracts with endoplasmic reticulum Ca2� pump Serca2b andis necessary for collagen type I synthesis. Mol Cell Biol 2004;24:1758–1768.

24. Inagaki Y, Okazaki I. Emerging insights into transforming growthfactor beta Smad signal in hepatic fibrogenesis. Gut 2007;56:284–292.

25. Breitkopf K, Godoy P, Ciuclan L, et al. TGF-beta/Smad signalingin the injured liver. Z Gastroenterol 2006;44:57–66.

26. Liu C, Gaca MD, Swenson ES, et al. Smads 2 and 3 aredifferentially activated by transforming growth factor-beta (TGF-beta ) in quiescent and activated hepatic stellate cells. Consti-tutive nuclear localization of Smads in activated cells is TGF-beta-independent. J Biol Chem 2003;278:11721–11728.

27. Uemura M, Swenson ES, Gaca MD, et al. Smad2 and Smad3play different roles in rat hepatic stellate cell function andalpha-smooth muscle actin organization. Mol Biol Cell 2005;16:4214–4224.

28. Wiercinska E, Wickert L, Denecke B, et al. Id1 is a criticalmediator in TGF-beta-induced transdifferentiation of rat hepaticstellate cells. Hepatology 2006;43:1032–1041.

29. Garcia-Trevijano ER, Iraburu MJ, Fontana L, et al. Transforminggrowth factor beta1 induces the expression of alpha1(I) procol-lagen mRNA by a hydrogen peroxide-C/EBPbeta-dependentmechanism in rat hepatic stellate cells. Hepatology 1999;29:960–970.

30. Dooley S, Delvoux B, Lahme B, et al. Modulation of transforminggrowth factor beta response and signaling during transdifferen-tiation of rat hepatic stellate cells to myofibroblasts. Hepatology2000;31:1094–1106.

31. Paradis V, Dargere D, Bonvoust F, et al. Effects and regulationof connective tissue growth factor on hepatic stellate cells. LabInvest 2002;82:767–774.

32. Rachfal AW, Brigstock DR. Connective tissue growth factor(CTGF/CCN2) in hepatic fibrosis. Hepatol Res 2003;26:1–9.

33. Gao R, Brigstock DR. Connective tissue growth factor (CCN2)induces adhesion of rat activated hepatic stellate cells by bind-ing of its C-terminal domain to integrin alpha(v)beta(3) andheparan sulfate proteoglycan. J Biol Chem 2004;279:8848–8855.

34. Paradis V, Perlemuter G, Bonvoust F, et al. High glucose andhyperinsulinemia stimulate connective tissue growth factor ex-pression: a potential mechanism involved in progression tofibrosis in nonalcoholic steatohepatitis. Hepatology 2001;34:738–744.

35. Grotendorst GR. Connective tissue growth factor: a mediator ofTGF-beta action on fibroblasts. Cytokine Growth Factor Rev1997;8:171–179.

36. Brigstock DR. The CCN family: a new stimulus package. J En-docrinol 2003;178:169–175.

37. Weng HL, Ciuclan L, Liu Y, et al. Profibrogenic transforminggrowth factor-beta/activin receptor-like kinase 5 signaling viaconnective tissue growth factor expression in hepatocytes.Hepatology 2007;46:1257–1270.

38. Gressner OA, Lahme B, Demirci I, et al. Differential effects ofTGF-beta on connective tissue growth factor (CTGF/CCN2) ex-pression in hepatic stellate cells and hepatocytes. J Hepatol2007;47:699–710.

39. Roskams T, Cassiman D, De Vos R, et al. Neuroregulation ofthe neuroendocrine compartment of the liver. Anat Rec 2004;

280:910–923.
Page 14: Gastro Rvu-Slf 2008

1

1

1

1

1

1

1

1

1

1

1

1

1

1

1

1

1

1

1

1

1

1

1

1

1

1

1

1

1

1

1

1

1

1

1

1

1

1

1

1

1

1

1668 SCOTT L. FRIEDMAN GASTROENTEROLOGY Vol. 134, No. 6

40. Mallat A, Teixeira-Clerc F, Deveaux V, et al. Cannabinoid recep-tors as new targets of antifibrosing strategies during chronicliver diseases. Expert Opin Ther Targets 2007;11:403–409.

41. Batkai S, Jarai Z, Wagner JA, et al. Endocannabinoids acting atvascular CB1 receptors mediate the vasodilated state in ad-vanced liver cirrhosis. Nat Med 2001;7:827–832.

42. Teixeira-Clerc F, Julien B, Grenard P, et al. CB1 cannabinoidreceptor antagonism: a new strategy for the treatment of liverfibrosis. Nat Med 2006;12:671–676.

43. Munoz-Luque J, Ros J, Fernandez-Varo G, et al. Regression offibrosis after chronic stimulation of cannabinoid CB2 receptor incirrhotic rats. J Pharmacol Exp Ther 2008;324:475–483.

44. Ebrahimkhani MR, Kiani S, Oakley F, et al. Naltrexone, an opioidreceptor antagonist, attenuates liver fibrosis in bile duct ligatedrats. Gut 2006.

45. De Minicis S, Candelaresi C, Marzioni M, et al. Role of endog-enous opioids in modulating Hsc activity in vitro and liver fibro-sis in vivo. Gut 2008;57:352–364.

46. Oben JA, Roskams T, Yang S, et al. Hepatic fibrogenesis re-quires sympathetic neurotransmitters. Gut 2004;53:438–445.

47. Rockey DC, Boyles JK, Gabbiani G, et al. Rat hepatic lipocytesexpress smooth muscle actin upon activation in vivo and inculture. J Submicrosc Cytol Pathol 1992;24:193–203.

48. Saab S, Tam SP, Tran BN, et al. Myosin mediates contractileforce generation by hepatic stellate cells in response to endo-thelin-1. J Biomed Sci 2002;9:607–612.

49. Bataller R, Gasull X, Gines P, et al. In vitro and in vivo activationof rat hepatic stellate cells results in de novo expression ofL-type voltage-operated calcium channels. Hepatology 2001;33:956–962.

50. Yee HF Jr. Ca2� and rho signaling pathways: two paths tohepatic stellate cell contraction. Hepatology 2001;33:1007–1008.

51. Laleman W, Van Landeghem L, Severi T, et al. Both Ca2�-dependent and -independent pathways are involved in rat he-patic stellate cell contraction and intrahepatic hyperresponsive-ness to methoxamine. Am J Physiol Gastrointest Liver Physiol2007;292:G556–G564.

52. Fischer R, Cariers A, Reinehr R, Haussinger D. Caspase 9-de-pendent killing of hepatic stellate cells by activated Kupffercells. Gastroenterology 2002;123:845–861.

53. Martinez-Hernandez A, Amenta PS. The extracellular matrix inhepatic regeneration. FASEB J 1995;9:1401–1410.

54. Olaso E, Salado C, Egilegor E, et al. Proangiogenic role oftumor-activated hepatic stellate cells in experimental mela-noma metastasis. Hepatology 2003;37:674–685.

55. Rockey DC. Vascular mediators in the injured liver. Hepatology2003;37:4–12.

56. Pinzani M, Failli P, Ruocco C, et al. Fat-storing cells as liver-specific pericytes. Spatial dynamics of agonist-stimulated intra-cellular calcium transients. J Clin Invest 1992;90:642–646.

57. Melton AC, Datta A, Yee HF Jr. [Ca2�]i-independent contractileforce generation by rat hepatic stellate cells in response toendothelin-1. Am J Physiol 2006;290:G7–G13.

58. Reynaert H, Thompson MG, Thomas T, et al. Hepatic stellatecells: role in microcirculation and pathophysiology of portalhypertension. Gut 2002;50:571–581.

59. Sherman IA, Pappas SC, Fisher MM. Hepatic microvascularchanges associated with development of liver fibrosis and cir-rhosis. Am J Physiol 1990;258:H460–H465.

60. Nakata M, Nakamura K, Koda Y, et al. Hemodynamics in themicrovasculature of thioacetamide-induced cirrhotic rat livers.Hepatogastroenterology 2002;49:652–656.

61. Tugues S, Fernandez-Varo G, Munoz-Luque J, et al. Antiangio-genic treatment with sunitinib ameliorates inflammatory infil-trate, fibrosis, and portal pressure in cirrhotic rats. Hepatology

2007;46:1919–1926.

62. Takeji M, Moriyama T, Oseto S, et al. Smooth muscle alpha-actin deficiency in myofibroblasts leads to enhanced renal tis-sue fibrosis. J Biol Chem 2006;281:40193–40200.

63. Guo DC, Pannu H, Tran-Fadulu V, et al. Mutations in smoothmuscle alpha-actin (ACTA2) lead to thoracic aortic aneurysmsand dissections. Nat Genet 2007;39:1488–1493.

64. Marra F. Chemokines in liver inflammation and fibrosis. FrontBiosci 2002;7:d1899–d1914.

65. Bonacchi A, Petrai I, Defranco RM, et al. The chemokine CCL21modulates lymphocyte recruitment and fibrosis in chronic hep-atitis C. Gastroenterology 2003;125:1060–1076.

66. Friedman SL. Hepatic stellate cells—protean, multifunctional,and enigmatic cells of the liver. Physiol Rev 2008;88:125–172.

67. Benyon RC, Arthur MJ. Extracellular matrix degradation and therole of hepatic stellate cells. Semin Liver Dis 2001;21:373–384.

68. Arthur MJ, Stanley A, Iredale JP, et al. Secretion of 72 kDa typeIV collagenase/gelatinase by cultured human lipocytes. Analy-sis of gene expression, protein synthesis and proteinase activ-ity. Biochem J 1992;287:701–707.

69. Han YP, Yan C, Zhou L, et al. A matrix metalloproteinase-9activation cascade by hepatic stellate cells in trans-differentia-tion in the three-dimensional extracellular matrix. J Biol Chem2007;282:12928–12939.

70. Vyas SK, Leyland H, Gentry J, et al. Rat hepatic lipocytessynthesize and secrete transin (stromelysin) in early primaryculture. Gastroenterology 1995;109:889–898.

71. Schaefer B, Rivas-Estilla AM, Meraz-Cruz N, et al. Reciprocalmodulation of matrix metalloproteinase-13 and type I collagengenes in rat hepatic stellate cells. Am J Pathol 2003;162:1771–1780.

72. Issa R, Zhou X, Constandinou CM, et al. Spontaneous recoveryfrom micronodular cirrhosis: evidence for incomplete resolutionassociated with matrix cross-linking. Gastroenterology 2004;126:1795–1808.

73. Nagula S, Jain D, Groszmann RJ, et al. Histological-hemody-namic correlation in cirrhosis—a histological classification ofthe severity of cirrhosis. J Hepatol 2006;44:111–117.

74. Li SW, Arita M, Fertala A, et al. Transgenic mice with inactivealleles for procollagen N-proteinase (ADAMTS-2) develop fragileskin and male sterility. Biochem J 2001;355:271–278.

75. Kesteloot F, Desmouliere A, Leclercq I, et al. ADAM metallopep-tidase with thrombospondin type 1 motif 2 inactivation reducesthe extent and stability of carbon tetrachloride-induced hepaticfibrosis in mice. Hepatology 2007;46:1620–1631.

76. Niiya M, Uemura M, Zheng XW, et al. Increased ADAMTS-13proteolytic activity in rat hepatic stellate cells upon activation invitro and in vivo. J Thromb Haemost 2006;4:1063–1070.

77. Duffield JS, Forbes SJ, Constandinou CM, et al. Selective de-pletion of macrophages reveals distinct, opposing roles duringliver injury and repair. J Clin Invest 2005;115:56–65.

78. Fallowfield JA, Mizuno M, Kendall TJ, et al. Scar-associatedmacrophages are a major source of hepatic matrix metallopro-teinase-13 and facilitate the resolution of murine hepatic fibro-sis. J Immunol 2007;178:5288–5295.

79. Murphy FR, Issa R, Zhou X, et al. Inhibition of apoptosis ofactivated hepatic stellate cells by tissue inhibitor of metallopro-teinase-1 is mediated via effects on matrix metalloproteinaseinhibition: implications for reversibility of liver fibrosis. J BiolChem 2002;277:11069–11076.

80. Yoshiji H, Kuriyama S, Yoshii J, et al. Tissue inhibitor of metal-loproteinases-1 attenuates spontaneous liver fibrosis resolu-tion in the transgenic mouse. Hepatology 2002;36:850–860.

81. Roderfeld M, Weiskirchen R, Wagner S, et al. Inhibition ofhepatic fibrogenesis by matrix metalloproteinase-9 mutants in

mice. FASEB J 2006;20:444–454.
Page 15: Gastro Rvu-Slf 2008

1

1

1

1

1

1

1

1

1

1

1

1

1

1

1

1

1

1

2

2

2

2

2

2

2

2

2

2

2

2

2

2

2

2

2

2

2

LMe

(

M

May 2008 HEPATIC FIBROGENESIS 1669

82. Knittel T, Fellmer P, Ramadori G. Gene expression and regula-tion of plasminogen activator inhibitor type I in hepatic stellatecells of rat liver. Gastroenterology 1996;111:745–754.

83. Zhang LP, Takahara T, Yata Y, et al. Increased expression ofplasminogen activator and plasminogen activator inhibitor dur-ing liver fibrogenesis of rats: role of stellate cells. J Hepatol1999;31:703–711.

84. Fibbi G, Pucci M, Grappone C, et al. Functions of the fibrinolyticsystem in human Ito cells and its control by basic fibroblast andplatelet-derived growth factor. Hepatology 1999;29:868 – 878.

85. Olaso E, Ikeda K, Eng FJ, et al. DDR2 receptor promotes MMP-2-mediated proliferation and invasion by hepatic stellate cells.J Clin Invest 2001;108:1369 –1378.

86. Gaca MD, Zhou X, Issa R, et al. Basement membrane-likematrix inhibits proliferation and collagen synthesis by activatedrat hepatic stellate cells: evidence for matrix-dependent deac-tivation of stellate cells. Matrix Biol 2003;22:229 –239.

87. Guyot C, Combe C, Balabaud C, et al. Fibrogenic cell fate duringfibrotic tissue remodelling observed in rat and human culturedliver slices. J Hepatol 2007;46:142–150.

88. Neuschwander-Tetri BA, Caldwell SH. Nonalcoholic steatohepa-titis: summary of an AASLD Single Topic Conference. Hepatol-ogy 2003;37:1202–1219.

89. London RM, George J. Pathogenesis of NASH: animal models.Clin Liver Dis 2007;11:55–74, viii.

90. Ikejima K, Okumura K, Kon K, et al. Role of adipocytokines inhepatic fibrogenesis. J Gastroenterol Hepatol 2007;22(Suppl1):S87–S92.

91. Ikejima K, Takei Y, Honda H, et al. Leptin receptor-mediatedsignaling regulates hepatic fibrogenesis and remodeling of ex-tracellular matrix in the rat. Gastroenterology 2002;122:1399 –1410.

92. Leclercq IA, Farrell GC, Schriemer R, et al. Leptin is essential forthe hepatic fibrogenic response to chronic liver injury. J Hepatol2002;37:206 –213.

93. Lin S, Saxena NK, Ding X, et al. Leptin increases tissue inhibitorof metalloproteinase I (TIMP-1) gene expression by a specificityprotein 1/signal transducer and activator of transcription 3mechanism. Mol Endocrinol 2006;20:3376 –3388.

94. Marra F, Aleffi S, Bertolani C, et al. Adipokines and liver fibrosis.Eur Rev Med Pharmacol Sci 2005;9:279 –284.

95. Kamada Y, Tamura S, Kiso S, et al. Enhanced carbon tetrachlo-ride-induced liver fibrosis in mice lacking adiponectin. Gastro-enterology 2003;125:1796 –1807.

96. Lonardo A, Adinolfi LE, Loria P, et al. Steatosis and hepatitis Cvirus: mechanisms and significance for hepatic and extrahe-patic disease. Gastroenterology 2004;126:586 –597.

97. Fartoux L, Poujol-Robert A, Guechot J, et al. Insulin resistance isa cause of steatosis and fibrosis progression in chronic hepa-titis C. Gut 2005;54:1003–1008.

98. Moucari R, Asselah T, Cazals-Hatem D, et al. Insulin resistancein chronic hepatitis C: association with genotypes I and 4,serum HCV RNA level, and liver fibrosis. Gastroenterology2008;134:416 – 423.

99. Hezode C, Roudot-Thoraval F, Nguyen S, et al. Daily cannabissmoking as a risk factor for progression of fibrosis in chronichepatitis C. Hepatology 2005;42:63–71.

00. Hezode C, Zafrani ES, Roudot-Thoraval F, et al. Daily cannabisuse: a novel risk factor of steatosis severity in patients withchronic hepatitis C. Gastroenterology 2008;134:432– 439.

01. Jeong WI, Osei-Hyiaman D, Park O, et al. Paracrine activation ofhepatic CB(1) receptors by stellate cell-derived endocannabi-noids mediates alcoholic fatty liver. Cell Metab 2008;7:227–

235. s

02. Bataller R, Paik YH, Lindquist JN, et al. Hepatitis C virus coreand nonstructural proteins induce fibrogenic effects in hepaticstellate cells. Gastroenterology 2004;126:529 –540.

03. Shin JY, Hur W, Wang JS, et al. HCV core protein promotes liverfibrogenesis via up-regulation of CTGF with TGF-beta1. Exp MolMed 2005;37:138 –145.

04. Mazzocca A, Sciammetta SC, Carloni V, et al. Binding of hepa-titis C virus envelope protein E2 to CD81 up-regulates matrixmetalloproteinase-2 in human hepatic stellate cells. J BiolChem 2005;280:11329 –11339.

05. Schulze-Krebs A, Preimel D, Popov Y, et al. Hepatitis C virus-replicating hepatocytes induce fibrogenic activation of hepaticstellate cells. Gastroenterology 2005;129:246 –258.

06. Pohlmann S, Zhang J, Baribaud F, et al. Hepatitis C virusglycoproteins interact with DC-SIGN and DC-SIGNR. J Virol2003;77:4070 – 4080.

07. Tuyama A, Hong F, Mosoian A, et al. HIV entry and replication instellate cells promotes cellular activation and fibrogenesis: im-plications for hepatic fibrosis in HIV/HCV co-infection. Hepatol-ogy 2007;46:80A.

08. Brau N, Salvatore M, Rios-Bedoya CF, et al, for the PuertoRico-New York Hepatitis Study G. Slower fibrosis progression inHIV/HCV-coinfected patients with successful HIV suppressionusing antiretroviral therapy. J Hepatol 2006;44:47–55.

09. Kordes C, Sawitza I, Muller-Marbach A, et al. CD133� hepaticstellate cells are progenitor cells. Biochem Biophys Res Com-mun 2007;352:410 – 417.

10. Roskams T. Different types of liver progenitor cells and theirniches. J Hepatol 2006;45:1– 4.

11. Sicklick JK, Li YX, Choi SS, et al. Role for hedgehog signaling inhepatic stellate cell activation and viability. Lab Invest 2005;85:1368 –1380.

12. Yang L, Wang Y, Mao H, et al. Sonic hedgehog is an autocrineviability factor for myofibroblastic hepatic stellate cells. J Hepa-tol 2008;48:98 –106.

13. Myung SJ, Yoon JH, Gwak GY, et al. Wnt signaling enhances theactivation and survival of human hepatic stellate cells. FEBSLett 2007;581:2954 –2958.

14. Taipale J, Beachy PA. The Hedgehog and Wnt signalling path-ways in cancer. Nature 2001;411:349 –354.

15. Lafdil F, Chobert MN, Couchie D, et al. Induction of Gas6 proteinin CCl4-induced rat liver injury and anti-apoptotic effect on he-patic stellate cells. Hepatology 2006;44:228 –239.

16. Goodman ZD, Becker RL Jr, Pockros PJ, et al. Progression offibrosis in advanced chronic hepatitis C: evaluation by morpho-metric image analysis. Hepatology 2007;45:886–894.

17. Pockros PJ, Jeffers L, Afdhal N, et al. Final results of a double-blind, placebo-controlled trial of the anti-fibrotic efficacy of inter-feron-gamma1b in chronic hepatitis C patients with advancedfibrosis or cirrhosis. Hepatology 2007;45:569–578.

18. D’Amico G, Garcia-Tsao G, Pagliaro L. Natural history and prog-nostic indicators of survival in cirrhosis: a systematic review of118 studies. J Hepatol 2006;44:217–231.

Received December 18, 2007. Accepted January 16, 2008.Address requests for reprints to: Scott L. Friedman, MD, Division of

iver Diseases, Box 1123, Mount Sinai School of Medicine, 1425adison Avenue, Room 11-70C, New York, New York 10029-6574.-mail: [email protected]; fax: (212) 849-2574.Supported by grants from the National Institutes of Health

DK37330, DK56621), and the Feld Trust.The author gratefully acknowledges Drs Guadalupe Garcia-Tsao andassimo Pinzani, who contributed significantly to the concepts pre-

ented in Figure 5.