pathogenesis of liver fibrosis - semantic scholar · pathogenesis of liver fibrosis r. alcolado, m....

10
Clinical Science (1997) 92, 103-1 12 (Printed in Great Britain) I03 Editorial Review Pathogenesis of liver fibrosis R. ALCOLADO, M. J. P. ARTHUR and J. P. IREDALE University Medicine, University of Southampton, U.K. 1. Liver fibrosis is a common sequel to diverse liver injuries. It is characterized by an accumulation of interstitial collagens and other matrix components. The hepatic stellate cell is pivotal to the pathogenic process. Fibrotic liver injury results in activation of the hepatic stellate cell which undergoes a pheno- typic change to a proliferative myofibroblast-like cell which synthesizes excess interstitial collagens and other matrix components. 2. The process of initiation of activation and its perpetuation result from complex, often interrelated series of signalling mechanisms which converge on this effector cell. Such mechanisms include altera- tions in matrix resulting in changed cell-matrix interactions and stimulation by cytokines released from damaged hepatocytes, infiltrating inflamma- tory cells, Kupffer cells and matrix. Foremost among the profibrotic cytokines is transforming growth factor PI. 3. Once the hepatic stellate cell is activated the preceding matrix changes and recurrent injurious stimuli will perpetuate the activated state. 4. Despite the accumulation of excess collagens, the liver retains a capacity for matrix degradation. This capacity may be overwhelmed and any secreted matrix remodelling enzymes may be inhibited by the concurrently secreted tissue inhibitors of metallo- proteinase-1 and az-macroglobulin. 5. Our understanding of the molecular pathogen- esis of liver fibrosis is increasing. It is anticipated that this knowledge will provide novel therapeutic avenues to treat this disease process. INTRODUCTION Liver fibrosis and cirrhosis represent the final common pathological pathway for a variety of liver diseases, irrespective of the underlying aetiology. In the fibrotic liver there is not only an increase in the total content of collagens and other matrix proteins compared with normal, but there are also qualitative changes in the nature of the matrix components and their distribution within the liver. Recent research has highlighted several key areas of the pathological process which has enhanced our understanding not only of liver fibrosis, but of the fibrotic process in other solid organs. The key cellular and molecular events involved in the pathogenesis of liver fibrosis include activation of hepatic stellate cells (stellate cells, lipocytes, fat-storing cells or Ito cells) to a myofibroblast-like phenotype, their production of excess matrix proteins and their ability to regulate matrix degrada- tion. The mechanisms controlling stellate cell activa- tion and the factors regulating the synthesis and degradation of matrix proteins have thus become the principal focus of research activity in liver fibrosis. This work encompasses analysis of inter- actions between stellate cells and both parenchymal cells (hepatocytes) and non-parenchymal cells (Kupffer cells and endothelial cells) as well as examining the cell-matrix interactions that deter- mine stellate cell and hepatocyte phenotype and behaviour, and the role of soluble cell messengers (cytokines, growth factors and other soluble messen- gers) released by inflammatory cells and resident liver cells. The purpose of this article is to review the current state of knowledge of this complex pathological process and to describe the principal events that occur as the previously ordered liver microanatomy progresses to the distorted architecture that charac- terizes fibrosis. MICROANATOMY, STRUCTURE AND CELLS OF THE HEPATIC SlNUSOlD The functional unit of the liver is the acinus which consists of a central portal tract containing an efferent biliary radical and terminal branches of the portal and arterial blood supplies. The blood supplies feed into the hepatic sinusoid, which are bounded by plates of hepatocytes, and finally drain Key words: cell-matrix interaction, hepatic stellate cells, liver fibrosis, matrix, matrix regulation, tissue inhibitor of metalloproteinase-I, transforminggrowth factor 8. Abbreviations: IGF, insulin-likegrowth factor; M6P, mannose-6-phosphate: MMP, matrix metalloproteinse; PDGF, platelet-derived growth factor; TGF-/I, transforming growth factor-8; TIMP-I, tissue inhibitor of metalloproteinase-I;uPA, urokinase plaminogen activator. Correspondence: Professor M. J. P. Arthur, University Medicine, Level D, South Block. Southampton General Hospital, Trrmona Road, Southampton. Hampshire SO1 6YD, U.K.

Upload: others

Post on 17-Jul-2020

4 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: Pathogenesis of Liver Fibrosis - Semantic Scholar · Pathogenesis of liver fibrosis R. ALCOLADO, M. J. P. ARTHUR and J. P. IREDALE University Medicine, University of Southampton,

Clinical Science (1997) 92, 103-1 12 (Printed in Great Britain) I03

Editorial Review

Pathogenesis of liver fibrosis

R. ALCOLADO, M. J. P. ARTHUR and J. P. IREDALE University Medicine, University of Southampton, U.K.

1. Liver fibrosis is a common sequel to diverse liver injuries. It is characterized by an accumulation of interstitial collagens and other matrix components. The hepatic stellate cell is pivotal to the pathogenic process. Fibrotic liver injury results in activation of the hepatic stellate cell which undergoes a pheno- typic change to a proliferative myofibroblast-like cell which synthesizes excess interstitial collagens and other matrix components. 2. The process of initiation of activation and its perpetuation result from complex, often interrelated series of signalling mechanisms which converge on this effector cell. Such mechanisms include altera- tions in matrix resulting in changed cell-matrix interactions and stimulation by cytokines released from damaged hepatocytes, infiltrating inflamma- tory cells, Kupffer cells and matrix. Foremost among the profibrotic cytokines is transforming growth factor PI. 3. Once the hepatic stellate cell is activated the preceding matrix changes and recurrent injurious stimuli will perpetuate the activated state. 4. Despite the accumulation of excess collagens, the liver retains a capacity for matrix degradation. This capacity may be overwhelmed and any secreted matrix remodelling enzymes may be inhibited by the concurrently secreted tissue inhibitors of metallo- proteinase-1 and az-macroglobulin. 5. Our understanding of the molecular pathogen- esis of liver fibrosis is increasing. It is anticipated that this knowledge will provide novel therapeutic avenues to treat this disease process.

INTRODUCTION

Liver fibrosis and cirrhosis represent the final common pathological pathway for a variety of liver diseases, irrespective of the underlying aetiology. In the fibrotic liver there is not only an increase in the total content of collagens and other matrix proteins compared with normal, but there are also qualitative

changes in the nature of the matrix components and their distribution within the liver. Recent research has highlighted several key areas of the pathological process which has enhanced our understanding not only of liver fibrosis, but of the fibrotic process in other solid organs.

The key cellular and molecular events involved in the pathogenesis of liver fibrosis include activation of hepatic stellate cells (stellate cells, lipocytes, fat-storing cells or Ito cells) to a myofibroblast-like phenotype, their production of excess matrix proteins and their ability to regulate matrix degrada- tion. The mechanisms controlling stellate cell activa- tion and the factors regulating the synthesis and degradation of matrix proteins have thus become the principal focus of research activity in liver fibrosis. This work encompasses analysis of inter- actions between stellate cells and both parenchymal cells (hepatocytes) and non-parenchymal cells (Kupffer cells and endothelial cells) as well as examining the cell-matrix interactions that deter- mine stellate cell and hepatocyte phenotype and behaviour, and the role of soluble cell messengers (cytokines, growth factors and other soluble messen- gers) released by inflammatory cells and resident liver cells.

The purpose of this article is to review the current state of knowledge of this complex pathological process and to describe the principal events that occur as the previously ordered liver microanatomy progresses to the distorted architecture that charac- terizes fibrosis.

MICROANATOMY, STRUCTURE AND CELLS OF THE HEPATIC SlNUSOlD

The functional unit of the liver is the acinus which consists of a central portal tract containing an efferent biliary radical and terminal branches of the portal and arterial blood supplies. The blood supplies feed into the hepatic sinusoid, which are bounded by plates of hepatocytes, and finally drain

Key words: cell-matrix interaction, hepatic stellate cells, liver fibrosis, matrix, matrix regulation, tissue inhibitor of metalloproteinase-I, transforming growth factor 8. Abbreviations: IGF, insulin-like growth factor; M6P, mannose-6-phosphate: MMP, matrix metalloproteinse; PDGF, platelet-derived growth factor; TGF-/I, transforming growth factor-8; TIMP-I, tissue inhibitor of metalloproteinase-I; uPA, urokinase plaminogen activator. Correspondence: Professor M. J. P. Arthur, University Medicine, Level D, South Block. Southampton General Hospital, Trrmona Road, Southampton. Hampshire SO1 6YD, U.K.

Page 2: Pathogenesis of Liver Fibrosis - Semantic Scholar · Pathogenesis of liver fibrosis R. ALCOLADO, M. J. P. ARTHUR and J. P. IREDALE University Medicine, University of Southampton,

104 R. Alcolado et al.

into the efferent central vein. Hepatocytes make up over 80% of the volume of the human liver and are the site of almost all liver synthetic and degradative functions. Normal hepatocytes are cuboidal in shape and are arranged in palisades along the sinusoid, in close contact with sinusoidal blood.

The vascular sinusoid is lined by a highly special- ized fenestrated endothelium and contains within its lumen Kupffer cells and pit cells. Kupffer cells are the resident tissue macrophages found predomi- nantly in the periportal areas of the sinusoid and are responsible for removing bacterial antigens and endotoxin from the portal blood. Endothelial cells in the liver are highly specialized, possessing many fenestrae which allow free passage of macromole- cules from sinusoidal blood into the space of Disse, thus permitting contact with hepatocytes. Pit cells bear a strong resemblance to the natural killer cells of the spleen which play a major role in clearing opsonized particles and tumour cells from the blood.

Between the sinusoidal endothelium and the brush-bordered sinusoidal surface of the hepatocyte lies the space of Disse. This contains a specialized matrix with the same principal matrix components as a basement membrane (type IV collagen, laminin, proteoglycans and the stellate cells) (Fig. 1).

Stellate cells, the major focus of this review, are located in the space of Disse and are believed to be the liver equivalent of pericytes, with many cytoplasmic processes encircling the sinusoid. They contain many lipid-rich droplets which store over 75% of the body’s store of retinoids, largely as retinyl esters. During liver injury stellate cells undergo activation to a myofibroblast-like pheno- type, proliferate and express the matrix proteins which characterize fibrosis. The pathogenic role of stellate cell activation and its relationship to liver matrix and soluble cell messengers is considered in detail below.

m 0 0 0 - mpATocm

Normal baxmcnlmcmbmc-like QUIESCENT HSC - matrix rich in urllagen N

1- PROFIBROTIC INJURY

and synlhcrk collagens I and 111

Fig. I. Major changes that occur in the sinusoid with profibrotic injury. The hepatic stellate cells (HSCs) become activated, and proliferate and synthesize collagens I and 111 which replace the normal basement membrane-like matrix. In consequence, hepatocyte and endothelial cell phenotype is altered and the sinusoid becomes capillarized.

LIVER MATRIX IN NORMAL LIVER

In normal liver, matrix proteins are distributed throughout the organ, but two principal types of matrix may be identified. Fibrillar or interstitial matrix proteins (collagens I, I11 and V) are located in the liver capsule, large and medium-sized blood vessels and the portal tracts. This contrasts with the basement membrane-like matrix located in the functionally important sinusoidal space of Disse. The latter matrix is not electron dense but is comprised of type IV collagen, laminin and proteo- glycans as determined by immunohistochemical studies [l-51.

Of the many collagen subtypes described to date only five have been detected in liver; they are types I, 111, IV, V and VI. The fibril-forming collagens make up the majority of collagen in normal liver [6] and are found predominantly in the perivascular spaces, around portal tracts, and subcapsular areas. Type I and I11 collagens form hybrid fibrils [7] condensing with type V fibres which forms the fibril core.

Type IV collagen is located in the space of Disse [8], together with type VI collagen [9], as well as in vascular and bile duct basement membranes. Type IV collagen together with laminin and proteoglycans forms a flexible lattice structure [lo, 111 while type VI collagen monomers interact with each other to form chains of molecules within the space of Disse

Periductal and perivascular areas are rich in fibro- nectin and vitreonectin. The basement membrane- like matrix in the space of Disse contains fibronectin [2, 8, 131 and laminin in the same distribution as type IV collagen. Undulin is found in small amounts in association with the fibril-forming collagens [14].

Heparan sulphate, the commonest proteoglycan in normal liver [13], is found predominantly at the portal end of the sinusoid, with heparin proteo- glycan at the venous end [15]. Dermatan and chondroitin sulphate are found in very small amounts in normal liver [15, 161.

t121.

MATRIX CHANGES IN DISEASED LIVER

The early response to liver injury is characterized by the activation of stellate cells. This event is common to a wide range of liver insults. A major consequence of stellate cell activation to a myofibro- blast-like phenotype is that the synthetic activity of these cells alters and they secrete excess collagen I and I11 in addition to other matrix molecules. These matrix proteins are laid down in the space of Disse, initially causing an alteration in the appearance of the sinusoid. With further progression of fibrosis there are alterations in the phenotype of the endothelial cells which lose their fenestrae, so-called capillarization of the sinusoid, and perturbation of hepatocyte function (Fig. 1).

Further progression of the fibrotic process is

Page 3: Pathogenesis of Liver Fibrosis - Semantic Scholar · Pathogenesis of liver fibrosis R. ALCOLADO, M. J. P. ARTHUR and J. P. IREDALE University Medicine, University of Southampton,

Pathogenesis of liver fibrosis I05

associated with a gross distortion of the sinusoidal architecture as the vascular structures at either end become linked by dense connective tissue. Connec- tive tissue septae can also be visualized extending into the parenchyma. The final result of this process is the end-stage cirrhotic liver in which the whole anatomy is macroscopically distorted by thick bands of collagen, within which nodules of hepatocytes, often with regenerative foci, are surrounded.

Cirrhotic liver contains approximately six times the amount of matrix as normal liver [6]; there is an increase in the amounts of types I, I11 [8] and IV [l] collagen in the Space of Disse, but for type I collagen this increase is disproportionate so that the ratio of type I [17, 181 to types I11 and IV increases. There is an increase in laminin [6] within the space of Disse and alterations in both the total quantity and the type of fibronectin produced [6]. The spectrum of glycoproteins expressed changes with a relative decrease in heparin and heparan sulphate proteoglycans and an increase in dermatan and chondroitin sulphate proteoglycans. The latter contain the core proteins decorin and biglycan which have a potentially important role in cytokine binding within the extracellular matrix and are discussed in more detail below. Hyaluronan, which is only present in small quantities in normal liver, is increased more than 8-fold in diseased liver and is secreted by hepatic stellate cells [19].

CELLULAR SOURCES OF MATRIX

Early studies of matrix secretion by isolated cells indicated that hepatocytes were a significant source of matrix components. More recently, access to rigorously characterized cell populations has cast doubt on this finding, suggesting that it may have resulted from overgrowth of contaminating stellate cells, and has given a clearer picture of the role of individual cell types in matrix synthesis. These findings have been confirmed by studies using in situ hybridization or analysis of cell fractions extracted from normal and injured liver to confirm the origin of matrix components.

Hepatocytes contain the mRNAs for collagens I [20] and IV although not all workers [21] have been able to reproduce these results. Cultured hepato- cytes produce plasma fibronectin [22] and the proteoglycan heparan sulphate [4].

Current studies indicate that stellate cells are the major source of matrix proteins in the diseased liver [21-231. They produce a broad range of matrix components - mRNA for collagen type I, I11 and IV [24-261 has been isolated along with mRNAs for laminin [5, 251, fibronectin [22, 241, heparan sulphate [4] and dermatan and chondroitin sulphate [4, 271 proteoglycan and the collagen synthetic enzyme, prolyl hydroxylase.

mRNA transcripts for type IV collagen [21, 281 and fibronectin [29] have been identified in endo-

thelial cells, while Kupffer cells and Pit cells do not appear to elaborate any matrix components on the basis of present data. Doubt has been cast on earlier data suggesting that hepatocytes are a major source of matrix after it was demonstrated that hepatocyte cultures are often contaminated with stellate cells ~301.

BIOLOGICAL EFFECT OF MATRIX CHANGES IN FIBROSIS AND THEIR RELATIONSHIP TO STELLATE CELL ACTIVATION

In addition to the described architectural changes, the shift in matrix constitution to one rich in collagen I and I11 is associated with specific changes in the phenotype of parenchymal and non-paren- chymal cells.

The evidence for a biological activity of matrix lies in a series of elegant experiments which have demonstrated that hepatocytes retain a differen- tiated phenotype in which they continue to express albumin and cytochrome P-450, when cultured on a model basement membrane. This biological effect of the whole-model matrix is greater than each of its constituent parts, when these are used as individual culture substrata. In contrast, culture of hepatocytes on type I collagen or tissue culture plastic results in a flattening and spreading of the cells, in association with which, tissue-specific gene expression is lost [31, 321. Interestingly, more simple matrices which result in cultured hepatocytes retaining their three- dimensional shape, including a gelatin sandwich, have been demonstrated to maintain gene-specific function, suggesting that the maintenance of normal shape is important for the persistence of a differen- tiated phenotype [33].

The implications of these studies are that hepato- cyte function may be disrupted comparatively early in the progression of liver fibrosis and before gross changes occur, by virtue of altered cell-matrix inter- actions. In addition, the findings are consistent with the hypothesis that hepatocellular dysfunction may remain a feature of liver injury even after withdrawal of the injurious agent in the absence of adequate matrix remodelling.

Evidence also exists for important cell-matrix interactions involving endothelial cells [16] and, pertinent to this article, stellate cells. Of importance for both the initiation and progression of fibrosis, stellate cells respond to a change in substratum by undergoing a phenotypic change [34]. Stellate cells in normal liver lie in the space of Disse. They are rich in retinoids and fat droplets and can be readily recognized. During liver injury these cells become activated to a myofibroblast-like phenotype [35]. They shed their retinoids after hydrolysing the esters and start to express a-smooth muscle actin [36-391. This phenotypic change is central to the pathogen- esis of fibrosis because it is in this activated pheno- type that stellate cells express and secrete excess

Page 4: Pathogenesis of Liver Fibrosis - Semantic Scholar · Pathogenesis of liver fibrosis R. ALCOLADO, M. J. P. ARTHUR and J. P. IREDALE University Medicine, University of Southampton,

I06 R. Alcolado et al.

matrix proteins during fibrosis and downregulate their subsequent degradation.

The activation of stellate cells is a general response to injury and can be demonstrated to be temporally related to an insult (after cc14 intoxica- tion, stellate cells can be demonstrated to pass through stages of activation, so-called transitional cells). However, activation is also related to the site of an injury; thus after cch intoxication, stellate cell activation will occur in association with hepatocyte degeneration pericentrally, while bile duct ligation is associated with periportal stellate cell activation. In the presence of recurrent or progressive injury, stellate cell activation becomes more generalized

By culture on a model basement-like matrix, stellate cells can be maintained in a normal quies- cent, retinoid-rich phenotype. When type I collagen or uncoated tissue culture plastic is substituted the stellate cells rapidly undergo a phenotypic trans- formation to activated cells in a manner that is very similar to the process in vivo [25, 34, 431. These findings have led to the hypothesis that alterations in the matrix substratum, specifically the degrada- tion of the normal basement membrane-like matrix and its replacement with a matrix rich in interstitial collagens, may be important in both the activation and perpetuation of activation of stellate cells. Furthermore, the demonstration that activation can be mimicked in tissue culture, by plating primary cultures of stellate cells onto plastic, has greatly facilitated the study of stellate cell expression and synthesis of matrix proteins and matrix remodelling enzymes.

Another important matrix change which occurs early in the course of experimental liver injury and is important for stellate cell activation is the produc- tion, by endothelial cells, of fibronectin spliced to contain the EIII-A variable region [29]. This isoform is virtually absent from normal liver. The peak in EIII-A fibronectin production occurs within 12 h of experimental bile duct ligation and remains elevated for 7 days. Stellate cells cultured with endothelial cells isolated from bile duct-ligated livers become activated more rapidly than equivalent controls. This action of the endothelial cell can be blocked by antibody directed against the EIII-A region of fibro- nectin [29], suggesting that the endothelial cell response to injury may directly activate stellate cells via changes in matrix environment.

The mechanisms of cell-matrix interactions involving stellate cells, hepatocytes and endothelial cells are likely to be mediated via integrin and non-integrin receptors. Integrins are transmembrane heterodimeric proteins which mediate cell adhesion and cell signalling and may be important in the secretion of ordered matrix by synthetic cells [44, 451. Normal basement membrane constituents contain well-characterized integrin recognition sequences. The repeated Arg-Gly-Asp motif which occurs in the triple helical region of the type I [46]

[21, 37, 38, 40-421.

and type IV [47] collagen molecule is recognized by the a$l integrin receptor which occurs on hepato- cytes [47]. Fibronectin [48] bears a number of integrin recognition sequences for the M5Pl [49] (Arg-Gly-Asp) and C(4Pl [50] (Leu-Asp-Val, or Arg-Glu-Asp-Val) receptors. It also contains a number of recognition sequences for other matrix components as well as for the transmembrane proteoglycan syndecan. Laminin [49] has eight cell adhesion or recognition sequences, many of which recognize other matrix components, especially fibrillar collagens, and are thought to be responsible for the arrangement of collagen into fibrils. Laminin also contains epidermal growth factor-like peptide repeats which, when hydrolysed, are capable of stimulating cellular epidermal growth factor recep- tors [51]. Proteoglycans interact with other matrix components via non-integrin receptors but their major interaction with cells may be mediated via their ability to bind cytokines. Heparan sulphate proteoglycan, found in normal liver, is capable of binding /3-fibroblast growth factor [52], which prevents its rapid degradation by membrane-bound plasmin and enhances binding to specific P-fibro- blast growth factor receptors. Decorin and biglycan, the protein core of chondroitin and dermatan sulphates, are capable of binding TGF-PI reversibly [531*

OTHER FACTORS INFLUENCING STELLATE CELL ACTlVATl ON

In addition to alterations in the matrix substratum, there is evidence that stellate cell activa- tion is influenced by a wide variety of cytokines and cell messengers. Kupffer cells are probably central to the response of the liver to many injuries. Indeed, experimental liver injury can be ameliorated by prior depletion of Kupffer cells. Foremost among the bioactive substances released by Kupffer cells is the cytokine TGF-PI (Fig. 2). Extensive studies of this cytokine elsewhere in the body indicate that its expression is fundamental to the scarring response.

Transforming growth factor P, so-called because of its ability to transform a non-proliferating fibro- blast cell line in soft agar culture [54], inhibits the proliferation of rat stellate cells [55] but promotes the platelet-derived growth factor (PDGF)-mediated proliferation of human stellate cells [56]. TGF-PI is one of a family of related peptides, TGF-Pl-TGF- Ps, of which only TGF-PI, P2 and P 3 have been detected in liver [57].

In disease models including CC14 intoxication, bile duct ligation, iron and alcohol intoxication, strepto- coccal cell wall administration and murine schistoso- miasis [58-621, an increase in the level of mRNA for TGF-PI has been documented. Fibrotic liver from patients with a variety of chronic liver diseases has been shown to contain increased levels of TGF-Dl mRNA [63]. In chronic liver injury TGF-PI

Page 5: Pathogenesis of Liver Fibrosis - Semantic Scholar · Pathogenesis of liver fibrosis R. ALCOLADO, M. J. P. ARTHUR and J. P. IREDALE University Medicine, University of Southampton,

Pathogenesis of liver fibrosis I07

PDGF - ACTIVATION - TGFP,

ACTIVATED

LIKE HSC MYOFIBROBLAST-

PROLIFERATION COLLAGEN-I SYNTHESIS

Fig. 2. Major cytokines and mechanisms believed to be important in the process of hepatic stellate cell (HSC) activation to the prolifera- tive myofibroblast-like phenotype observed in fibrogenic injury

is produced by a number of non-parenchymal cells [64]. TGF-PI may be derived from activated stellate cells [65], Kupffer cells [55] and endothelial cells [60]. In addition, TGF-PI may be produced by migrating mononuclear cells [58].

TGF-Pl has profound effects on matrix produc- tion in a number of well-characterized cell lines [66, 671 including fibroblasts, osteoblasts and, to a lesser extent, myofibroblastic stellate cells [24, 681. TGF-PI not only causes an increase in the production of type I collagen but also in types I11 and IV [24]. In addition, fibronectin [69, 701 and proteoglycan [55, 711 synthesis is enhanced. In primary stellate cell culture, addition of TGF-PI 24 h after plating results in an accelerated transformation to the activated phenotype [69].

TGF-PI also alters the balance of matrix turnover against matrix degradation and in favour of matrix accumulation, by inhibiting production of interstitial collagenase [72, 731, stromelysin and plasminogen activators, and promoting the production of the potent collagenase inhibitor, tissue inhibitor of metalloproteinase-1 (TIMP-1) [72] and plasminogen activator inhibitor-1 [74]. Fibroblasts will also respond to TGF-PI by upregulating gelatinase A [73]. However, this has not been demonstrated in stellate cells [75]. A further significant effect of TGF-PI on stellate cells is the stimulation of a positive autocrine loop [24, 651. TGF-PI increases the facility for stellate cell-matrix interactions by increased transcription of mRNA for the a and P subunits of integrin receptors and increased assembly of these subunits to form the active recep- tors 1761.

Relative levels of mRNA cannot be directly corre- lated with active TGF-PI because the protein is released in a latent form. The mechanism of activa- tion of TGF-PI in liver is probably via the insulin-

like growth factor II/mannose-6-phosphate (IGF- II/M6P) receptor. Moreover, it has been demon- strated that stellate cells extracted from CC14- treated rats but not normal liver bear the IGF-III M6P receptor on the cell surface [77].

The balance of activated versus inactivated cytokine must be of central importance as TGF-Pl is detectable in normal liver [77] and almost all normal liver cells exhibit receptors for active TGF-PI, while mRNA levels for receptor types 1, 2 and 3 are all increased in activated stellate cells [78]. Latent TGF-PI may also exist in a pool within the liver matrix. The type 3 TGF-Pl receptor has long been known to be a non-signalling receptor: it has recently been identified as membrane proteoglycan P-glycan [79, 801. In addition, the matrix component decorin binds and inactivates TGF-PI. The binding of TGF+ to these matrix constituents may both prevent activation and protect the latent form from breakdown. The characteristics of this binding in vivo are not understood, particularly the conditions required for the release of latent TGF-Pl from these receptors. Nonetheless, decorin has been demon- strated to have an anti-fibrotic effect in renal fibrosis in vivo [Sl]. Moreover, the position of TGF-Pl as a central cytokine to the fibrotic/scarring process has now been demonstrated in a series of mechanistic studies in which fibrosis was reduced by the addition of decorin and neutralizing antibodies to cell culture and in vivo models [81-841. Perhaps predictably, a transgenic line of TGF-PI overexpressor animals developed evidence of hepatic and renal fibrosis

Another very important cytokine is PDGF [86] (Fig. 2). This consists of A and B peptide chains. Three forms exist, the homodimeric AA and BB forms or the heterodimer. PDGF is a potent stellate cell mitogen [87] which is produced by endothelial cells [SS] and by activated stellate cells [89] in culture. Quiescent stellate cells express the a-PDGF receptor subunit, but do not express the b-subunit until activated. This is observed in both inflamma- tory liver injury and after bile duct ligation [90]. The b-receptor subunit only interacts with the B chain of PDGF, and the PDGF dimers containing at least one B-chain are significantly more potent to activated stellate cells than the PDGF-AA dimers. Expression of the PDGF b-receptor subunit is enhanced in response to TGF-PI. Furthermore, there is evidence that Kupffer cells may direct stellate cell PDGF receptor expression by means of an as yet uncharacterized soluble factor [91].

PDGF, like TGF-PI, is able to induce transcrip- tion of its own mRNA and PDGF BB has been shown to induce transcription of both A and B chains in culture-activated stellate cells [89].

While Kupffer cell-derived factors ' may. be important to the PDGF- and TGF-P-mediated effects on stellate cells, recent work has suggested that substances released directly from damaged hepatocytes are mitogenic for cultured stellate cells

~ 5 1 .

Page 6: Pathogenesis of Liver Fibrosis - Semantic Scholar · Pathogenesis of liver fibrosis R. ALCOLADO, M. J. P. ARTHUR and J. P. IREDALE University Medicine, University of Southampton,

I08 R Alcolado et al.

[92]. Moreover, IGF and IGF-binding protein released from injured hepatocytes may act in a paracrine manner to activate stellate cells locally. Studies of stellate cell IGF-I and I1 receptors indicate that there is a diminution of quantity (IGF-Ir) and binding affinity (IGF-IIr) with trans- formation to activated phenotype. This suggests that any IGF-mediated effect is only likely to be important early on in the process of stellate cell activation [93].

Tumour necrosis factor-a is a known mitogen [68, 691 for stellate cells and also has an effect on trans- cription of mRNA for chondroitin sulphate [24], interstitial collagenase [94, 9-51 and TIMP-1 [95].

Cytokines and cell messengers released during injury and inflammation may also moderate stellate cell activation, proliferation and synthetic function. The addition of interferon y to a culture medium of myofibroblastic stellate cells results in reversion to a more quiescent phenotype in which the cell regains its lipid droplets. It also affects collagen production [96] with falls in procollagen I and type IV collagen and significant reduction in the mRNA level for total fibronectin [96].

Addition of retinoids to stellate cells in culture can maintain a quiescent phenotype, and when added to activated stellate cells causes a reversion of phenotypic changes together with a decrease in the rate of cell proliferation [97, 981. Retinoids also modulate collagen production by culture-activated lipocytes - total collagen production is reduced in a dose-dependent fashion [98]. Further important effects of retinoids on culture-activated stellate cells include the inhibition of both TGF-PI mRNA production [98] and PDGF-mediated proliferation [99]. However, a paradox exists: athough retinoids display this potent cytoprotective role in models of fibrosis, the ingestion of large amounts of retinoids is known to induce liver fibrosis [loo].

It is apparent that there are many factors which play a role in the activation of stellate cells to the proliferating fibrogenic phenotype seen in chronic liver disease. There is a complex interaction between each individual cell type, the local matrix milieu and the production and activation of many cytokines, all of which converge on the stellate cell. This is summarized in Fig. 2. Key to the development and progression of fibrosis is the activation and persist- ence of activation of the stellate cell. The presence and persistence of particular matrix components is probably fundamental to the perpetuation of stellate cell activation and the progression of fibrosis. The role of matrix remodelling is therefore considered below.

MATRIX REGULATION

The control of matrix turnover is central to any understanding of the pathogenesis of liver fibrosis, since in profibrotic liver injury either the regulatory

mechanisms are overwhelmed by collagen produc- tion or the normal response to increasing collagen secretion is altered such that the excessive matrix is not degraded. Accumulating evidence indicates that progressive liver fibrosis is characterized by changes in the pattern of matrix degradation in addition to synthesis.

The turnover and remodelling of matrix by mesenchymal cells is mediated by a family of zinc- dependent neutral proteases, the matrix metallo- proteinases (MMPs) [loll. In addition, during inflammatory liver injury, matrix degradation may be mediated by matrix-degrading cathepsins.

In the analysis of matrix degradation during liver fibrosis two major groups of MMPs are believed to be important. The first, comprising the gelatinases and stromelysins, have activity against type IV collagen and other components of the normal basement membrane-like matrix [loll. These enzymes may therefore significantly alter stellate cell activation through degradation of the normal matrix. There is evidence that members of these groups are expressed by activated Kupffer cells (gelatinase B) [102], stellate cells (gelatinase A and stromelysin) [103-1051 and endothelial cells (stromelysin) [106]. Moreover, expression of both stromelysin and gelat- inase A has been described in acute liver injury in vivo in addition to studies of non-parenchymal cells in culture [106, 1071.

The second group comprises the collagenases [loll. These are derived from two sources: intersti- tial collagenase, derived from fibroblastic cells including activated myofibroblastic stellate cells and neutrophil collagenase, released from neutrophil phagosomes in areas of inflammation [loll. Because none of the other MMPs can initiate breakdown of interstitial collagens it is axiomatic that interstitial or neutrophil collagenase must be expressed for matrix remodelling to occur in established fibrosis [loll. Studies of cultured stellate cells indicate that these cells express interstitial collagenase in response to specific cytokines and early in models of activation [94, 95, 1081. One group suggested recently that gelatinase A has degradative activity against type I collagen [109]. Moreover, studies of mRNA expres- sion in fibrotic human liver and in animal models of liver fibrosis indicate that transcripts for interstitial collagenase are present and indeed may be upregu- lated in certain pathologies [95, 108, 1101. These studies have also indicated that fibrosis is accom- panied by a significant upregulation in mRNA transcripts of gelatinase A [110].

There is a relatively large literature suggesting that MMP activity in liver decreases as fibrosis progresses [lll-1131. This may in part result from the expression of the powerful MMP inhibitors the TIMPs and az-macroglobulin by stellate cells and hepatocytes [95, 108, 110, 114-1181. Detailed studies of culture-activated stellate cells have demonstrated that these cells express TIMP-1 and 2, and that these inhibitors exert a profound inhibition of

Page 7: Pathogenesis of Liver Fibrosis - Semantic Scholar · Pathogenesis of liver fibrosis R. ALCOLADO, M. J. P. ARTHUR and J. P. IREDALE University Medicine, University of Southampton,

Pathogenesis of liver fibrosis I09

co-expressed gelatinase activity [114]. Expression of mRNA for TIMPs in diseased human liver and animal models of fibrosis indicates that transcripts for both TIMP-1 and 2 are significantly upregulated [lo81 and, in the case of TIMP-1, are probably stellate cell derived [108].

TIMPs have been immunolocalized in diseased liver and co-localize with cells expressing interstitial collagenase [110]. In addition there are several studies which indicate that serum TIMP levels are elevated in fibrotic liver disease [119], and that TIMP-1 may be expressed by hepatocytes as a feature of the acute phase response [115]. crz-Macro- globulin has also been demonstrated to be expressed and secreted by stellate cells and hepatocytes [120].

Stellate cells may also influence both the degrada- tion of matrix and the activation of pro-MMPs by manipulating the plasmin/plasminogen cascade. Stellate cells express urokinase plasminogen activator (uPA) during activation and through expression of this protein can generate active plasmin from plasminogen [121]. The plasmin in turn cleaves the propiece from pro-stromelysin, thereby also initiating the activation of interstitial collagenase. By simultaneously expressing uPa receptors, stellate cells are able to activate plasmin in a focused pericellular manner [121]. As activation of stellate cells becomes more advanced, expression of uPA decreases and is replaced by the plasmi- nogen activator inhibitor with a resulting decrease in uPA activity [121].

Taken together, these data provide evidence that the potential for matrix degradation exists, even in comparatively advanced fibrosis. However, the process of matrix degradation is either overwhelmed by synthesis, and/or that matrix degradation is inhibited as a result of TIMP and plasminogen activator inhibitor expression.

CONCLUSION

There has been significant progress in our under- standing of the cellular and molecular events which characterize the response of the liver to injury and the consequent development of liver fibrosis. Central to the process is the stellate cell which undergoes a phenotypic change to a proliferative myofibroblast-like cell which then synthesizes excess interstitial collagens and other matrix components. The process of initiation of activation and its perpe- tuation result from a complex, often interrelated, series of signalling mechanisms which converge on this important effector cell. These mechanisms include alterations in matrix resulting in changed cell-matrix interactions and stimulation by cytokines released from damaged hepatocytes, infiltrating inflammatory cells, Kupffer cells and matrix. Once the stellate cell is activated the preceding matrix changes and any recurrent injurious stimuli will perpetuate the activated state. The activated stellate

cell is highly synthetic and secretes interstitial colla- gens which accumulate initially in the space of Disse, but ultimately result in gross liver fibrosis. Despite these effects, the liver retains a capacity/ potential for matrix degradation/remodelling. However, this capacity may be overwhelmed and any secreted matrix remodelling enzymes may be inhibited by the concurrently secreted TIMPs and crz-macroglobulin.

Future work in each of the areas highlighted in this review will ultimately provide a detailed dissec- tion of the molecular mechanism of stellate cell activation and the process of matrix secretion and assimilation. Specific targets are likely to include mechanisms to reverse stellate cell activation or inhibit matrix synthetic activity and intervention to promote matrix degradation. This may be accom- plished directly, i.e. by enhancing expression of collagenase and other MMPs, or indirectly by reducing the level of MMP inhibitory factors such as TIMPs. Such an approach, based on control of matrix degradation, is attractive because the majority of patients with chronic liver disease will present clinically when fibrosis is already established and may be relatively advanced. Tools to intervene directly in the process of experimental fibrosis (e.g. transgenic and gene knockout mice) are now becoming available, and it is anticipated that these data will highlight suitable avenues for therapeutic intervention.

REFERENCES I. Hahn EG, Wick G, Pencev D, Timpl R Distribution of basement membrane

proteins in normal and fibrotic human liver: collagen type N laminin and fibronectin. Gut 1980; 21: 63-71.

immunohistochemical studies in normal rat liver. Lab Invest 1984; 51: 57-69.

renal, splenic, intestinal, and hepatic basement membranes identified after the intravenous injection of heterologous antilaminin IgG. Lab Invest 1985; 5 2 169.

normal rat liver: lipocytes as a major source of proteoglycan. Gastroenterology

2. Martinez-Hernandez k The hepatic extracellular matrix. I. Electron

3. Abrahamson DR, Caulfield JP. Distribution of laminin within rat and mouse

4. Arenson DM, Friedman SL, Bissell DM. Formation of extracellular matrix in

1988; 95: 441-7. 5. Maher Jj, Friedman SL, Roll Fj, 8issell DM. lmmunolocalization of laminin in

normal rat liver and biosynthesis of laminin by hepatic lipocytes in primary culture. Gastroenterology 1988; 9 4 1053-62.

6. Schuppan D. Structure of extracellular matrix in normal and fibrotic liver. collagens and glycoproteins. Semin Liver Dis 1990; 10: 1-10.

7. Geem 111 occur together in hybrid fibrils in the space of Disse of normal rat liver. Hepatology 1990; I2 233-4 I.

8. Burt AD, Griffiths MR, Schuppan D, Voss B, MacSween RN. Ultrastructural localization of extracellular matrix proteins in liver biopsies using ultracryomicrotomy and immuno-gold labelling. Histopathology 1990; I 6 53-8.

Schuppan D, he roms S, De-Zanger R, Wisse E. Collagen type I and

9. Yurchenco PD. Assembly of basement membranes. Ann N Y Acad Sci 1990;

10. Siebold B, Deutzman R, Kuhn K. The arrangement of intra and intermolecular disulfide bonds in the carboxy terminal non collagenous aggregation and cross linking domain of basement membrane type IV collagen. Eur J Biochem 1988;

I I. Yurchenco PD, Furthmayr H. Self-assembly of basement membrane collagen.

12. Griffiths MR Ferrier R, Schuppan D, Burt AD. Collagen type VI: a component

5 8 0 195-214.

I76 6 I 7-24.

Biochemistry 1984; 23 1839-50.

of the perisinusoidal extracellular matrix in human liver. In: Wisse E, Knook DL.

Page 8: Pathogenesis of Liver Fibrosis - Semantic Scholar · Pathogenesis of liver fibrosis R. ALCOLADO, M. J. P. ARTHUR and J. P. IREDALE University Medicine, University of Southampton,

I10 R Alcolado et al.

McCuskey RS, eds. Cells of the hepatic sinusoid. Leiden, The Netherlands: Kupffer Cell Foundation, 1991: vol. 3, 155-60.

13. Geerts A, Geuze HJ, Slot J-W, et al. lmmunogold localization of procollagen 111, fibronectin and heparan sulfate proteoglycan on ultrathin frozen sections of the normal rat liver. Histochemistry 1986; 8 4 355-62.

14. Schuppan D, Cantaluppi MC, Becker J, et al. Undulin, an extracellular matrix glycoprotein associated with collagen fibrils. J Biol Chem 1990; 265: 8823-32.

IS. Reid LM, Fiorino AS, Sigal SH, Brill S, Holst PA Extracellular matrix gradients in the space of Disse: relevance to liver biology [Editorial]. Hepatology 1992; 1 5 1198-203.

16. McGuire RF. Bissell DM, Boyles J, Roll FJ. Role of extracellular matrix in regulating fenestrations of endothelial cells isolated from normal rat liver. Hepatology 1992; 15: 989-97.

liver. Gastroenterology 1979; 7 6 7 10- 19.

cirrhotic human liver. J Clin Invest 1977; 5 9 241-8.

fat storing cells (perisinusoidal lipocytes) in culture. Biochem Biophys Res Commun 1988; 151: 222-9.

20. Clement B, Laurent M, Guguen-Guillouzo C, Lebeau G, Guillouzo A Types I and IV procollagen gene expression in cultured rat hepatocytes. Coll Relat Res

17. Rojkind M, Giambrone M-A, Biempica L. Collagen types in normal and cirrhotic

18. Seyer JM, Huhenon ET, Kang AH. Collagen polymorphism in normal and

19. Gressner AM, Haarmann R Hyaluronic acid synthesis and secretion by rat liver

1988; 8: 349-59. 21. Maher JJ, McGuire RF. Extracellular matrix gene expression increases

preferentially in rat lipocytes and sinusoidal endothelial cells during hepatic fibrosis in vivo. J Clin Invest 1990; 8 6 I641 -8.

22. Ramadori G, Rieder H, Knittel T, Dienes HP, Meyer zum Buschenfelde K-H. Fat storing cells (FSC) of rat liver synthesize and secrete fibronectin. J Hepatol 1987; 4 190-7.

23. Friedman SL, Roll FJ, Boyles J, Bissell DM. Hepatic lipocytes: the principal collagen-producing cells of normal rat liver. Proc Natl Acad Sci USA 1985; 8 2 868 1-5.

24. Weiner FR, Giambrone MA, Czaja MJ, et al. Ito-cell gene expression and collagen regulation. Hepatology 1990; I I: I I 1-17.

25. Friedman SL, Rockey DC, McGuire RF, Maher JJ, Boyles JK, Yamasaki G. Isolated hepatic lipocytes and Kupffer cells from normal human liver: morphological and functional characteristics in primary culture. Hepatology

26. Kawase T, Shiratori Y, Sugimoto T. Collagen production by rat liver fat storing

27. Schafer S, Zerbe 0, Gressner AM. The synthesis of proteoglycans in fat-storing

28. ltving MG, Roll FJ, Huang S, Bissell DM. Characterisation and culture of

1992; IS: 234-43.

cells in I degree culture. Exp Cell Biol 1986; 5 4 183-92.

cells of rat liver. Hepatology (Baltimore) 1987; 7: 680-7.

sinusoidal endothelium from normal rat liver, lipoprotein uptake and collagen phenotype. Gastroenterology 1984; 87: 1233-47.

29. Jarnagin WR, Rockey DC, Koteliansky VE, Wang SS, Bissell DM. Expression of variant fibronectins in wound healing: cellular sources and biological activity of the ElllA segment in rat hepatic fibrogenesis. J Cell Biol 1994; 127: 2037-48.

cultures of normal rat hepatocytes derives from lipocytes within the monolayer. J Clin Invest 1988; 8 2 450-9.

3 I. Bissell DM, Arenson DM, Maher JJ, Roll FJ. Support of cultured hepatocytes by a laminin-rich gel. J Clin Invest 1987; 79: 801 - 12.

32. Bissell DM, Caron JM, Babiss LE, Friedman JM. Transcriptional regulation of the albumin gene in cultured rat hepatocytes. Role of basement-membrane matrix. Mol Biol Med 1990; 7: 187-97.

30. Maher JJ, Bissell DM, Friedman SL, Roll FJ. Collagen measured in primary

33. lredale JP, Arthur MJP. Hepatocyte-matrix interactions. Gut 1994; 35: 729-32. 34. Friedman SL, Roll FJ, Boyles J, Arenson DM, Biaell DM. Maintenance of

differentiated phenotype of cultured rat hepatic lipocytes by basement membrane matrix. J Biol Chem 1989; 264 10756-62.

strategies. N Engl J Med 1993: 328 1828-35.

lipocytes in acinar zone 3 and correlation to degree of collagen formation in Disse space. J Hepatol 1986; 3: 333-40.

37. Mak KM, Lieber CS. Lipocytes and transitional cells in alcoholic liver disease: a morphometric study. Hepatology (Baltimore) 1988; 8 1027-33.

38. McGee 10, Patrick Rs. The role of perisinusoidal cells in hepatic fibrogenesis. An electron microscopic study of acute carbon tetrachloride liver injury. Lab Invest 1972; 2 6 429-40.

39. Rockey DC, Boyles JK, Gabbiani G, Friedman SL. Rat hepatic lipocytes express smooth muscle actin upon activation in vivo and in culture. J Submicrosc Cytol Pathol 1992; 2 4 193-203.

35. Friedman SL. The cellular basis of hepatic fibrosis: mechanisms and treatment

36. Horn T, lunge J, Christoffenen P. Early alcoholic liver injury. Activation of

40. Mak KM, Leo AM, Lieber CS. Alcoholic liver injury in baboons: transformation

41. Leo MA, Mak KM, Savolainen ER, Lieber CS. Isolation and culture of of lipocytes to transitional cells. Gastroenterology 1984; 87: 188-200.

myofibroblasts from rat liver (42193). Proc Soc Exp Biol Med 1985; 180

42. Shiratori Y, lchida T. Geerts A, Wisse E. Modulation of collagen synthesis by 382-9 I.

fat-storing cells, isolated from CC14- or vitamin A-treated rats. Dig Dis Sci 1987; 3 2 1281-9.

43. Geerts A, Vrijsen R, Rauterberg J, Burt A, Schellinck P, Wise E. In vitro differentiation of fat-storing cells parallels marked increase of collagen synthesis and secretion. J Hepatol 1989; 9 59-68.

44. Ruoslahti E. Integrins. J Clin Invest 1991; 87: 1-5. 45. Wu C, Keivens VM, 07ooIe TE, McDonald JA, Ginsberg MH. lntegrin

activation and cytoskeletal interaction are essential for the assembly of a fibronectin matrix. Cell 1995; 83: 715-24.

collagen type I recognizes the Arg-Gly-kp sequence. J Cell Biol 1987; 104

47. Stamatoglou SC, Bawumia S, Johansson S, Forsberg E, Hughes RC. Affinity of integrin alpha-I beta-I from liver sinusoidal membranes for type IV collagen. FEBS Lett 1991; 288: 241-3.

48. Ruoslahti E, Pierschbacher MD. New perspectives in cell adhesion: RGD and integrins. Science (Washington DC) 1987; 238: 491-7.

49. Argraves WS, Suzuki S, Arai H, Thompson K, Pienchbacher MD, Ruoslahti E. Amino acid sequence of the human fibronectin receptor. J Cell Biol 1987; 105

46. Dedhar S, Ruoslahti E, Pierschbacher MD. A cell surface receptor complex for

585-93.

I 183-90. 50. Wayner EA, Garcia-Pardo A, Humphreys MJ, McDonald ]A, Carter WG.

Identification of the T-lymphocyte adhesion receptor for an alternative cell attachment domain (CS-I) in plasma fibronectin. J Cell Biol 1989; 109: 1321-30.

5 I. Panayotou G, End P, Aumailley M, et al. Domains of laminin with growth factor

52. Klagsbrunn M, Baird A. A dual receptor system is required for basic fibroblast

53. Andres JL, Stanley K, Cheifea S, Massague J. Membrane-anchored and soluble

activity. Cell 1989; 48: 989-96.

growth factor activity. Cell I99 I ; 67: 229-3 I.

forms of betaglycan, a polymorphic proteoglycan that binds transforming growth factor-beta. J Cell Biol 1989; 109 3 137-45.

transforming growth factor potentiated by epidermal growth factor. Proc Natl Acad Sci USA 1981; 78: 5339-41.

proteoglycan synthesis and proliferation by Kupffer cell-derived transforming growth factors type beta I and type alpha. Biochem Biophys Res Commun

56. Win KM, Charlotte F. Mallat A et al. Mitogenic effect of transforming growth factor-beta1 on human It0 cells in culture: evidence for mediation by endogenous platelet-derived growth factor. Hepatology 1993; 18: 137-4s.

Transforming growth factor beta (TGF-beta) isoforms in rat liver regeneration: messenger RNA expression and activation of latent TGF-beta. Cell Regulation

54. Roberts AB, Anzano MA, Lamb LC, Smith JM, Sporn MB. New class of

55. Meyer DH, Bachem MG, Gressner AM. Modulation of hepatic lipocyte

1990 171: 1122-9.

57. Jakowlew SB, Mead JE, Damelpour D, Wu J , Roberts AB, Fausto N.

1991; 2 535-48. 58. Nakatsukasa H, Nagy P, E m RP, Hsia CC, Marxlen E, Thorgeinson SS.

Cellular distribution of transforming growth factor-beta I and procollagen types I, 111, and IV transcripts in carbon tetrachloride-induced rat liver fibrosis. J Clin Invest 1990; 85: 1833-43.

59. Manthey CL, Allan JB, Ellingsworth LR, Wahl SM. In situ expression of transforming growth factor-beta in streptococcal cell wall-induced granulomatous inflammation and hepatic fibrosis. Growth Factors 1990; 4 17-26.

60. Czaja MJ, Weiner FR, Flanders KC, et al. In vitro and in vivo association of transforming growth factor beta-I with hepatic fibrosis. J Cell Biol 1989; 108: 2477-82.

induced by alcohol and iron. J Clin Invest 1995; 9 6 620-30.

transforming growth factor-beta in rat liver. Evidence for autocrine regulation of hepatocyte proliferation. J Clin Invest 1995; 9 6 447-55.

63. Castilla A, Prieto J, Fausto N. Transforming growth factors beta-I and alpha in chronic liver disease: Effects of interferon therapy. N Engl J Med I99 I ; 324

64. Milani S, Schuppan D, Herbst H, Surrenti C. Expression of transforming growth factor beta, in normal and fibrotic human liver. In: Gressner A, Ramadori G, eds. Molecular and cell biology of liver fibrogenesis. Dordrecht, The Netherlands: Kluwer Academic Publishers, 1992 254-62.

6 I. Tsukamoto H, Horne W, Kamimura S, et al. Experimental liver cirrhosis

62. Bissell DM. Wang SS, Jarnagin WR, Roll FJ. Cell-specific expression of

933-940.

Page 9: Pathogenesis of Liver Fibrosis - Semantic Scholar · Pathogenesis of liver fibrosis R. ALCOLADO, M. J. P. ARTHUR and J. P. IREDALE University Medicine, University of Southampton,

Pathogenesis of liver fibrosis I l l

65. Bachem MG, Meyer D, Melchior R, Sell K-M, Gressner AM. Activation of rat liver perisinusoidal lipocytes by transforming growth factors derived from myofibroblast-like cells. J Clin Invest 1992; 8 9 19-27.

66. Masague J. The transforming growth factor beta family. Annu Rev Cell Biol

67. Roberts AB, Spom MB. The transforming growth factors-beta. In: Sporn MB, Roberts AB. ed. Handbook of experimental pharmacology. Peptide growth factors and their receptors. Heidelberg, Germany: Springer-Verlag, 1990: vol. 95/L, 41 9-72.

68. Matsuoka M, Pham NT, Tsukamoto H. Differential effects of interleukin I alpha, tumour necrosis factor alpha, and transforming growth factor beta-I on cell proliferation and collagen formation by cultured fat storing cells. Liver 1989; 9

1990; 6 597-641.

71-8. 69. Bachem MH, Sell KM, Melchior R, Kropf J. Eller T. Gressner AM. TNF-alpha

and TGF beta-I stimulate fibronectin synthesis and the transdifferentiation of fat storing cells in rat liver to myofibroblasts. Virchows Archiv B Cell Pathol 1993;

70. Casini A, Pinzani M, Milani S, et al. Regulation of extracellular matrix synthesis 6 3 123-30.

by transforming growth factor beta I in human fat-storing cells. Gastroenterology 1993; 105: 245-53.

71. Bachem MG, Riess U, Melchior R, Sell KM, Gressner AM. Transforming growth factors (TGF alpha and TGF beta I) stimulate chondroitin sulfate and hyluronate synthesis in cultured rat liver fat storing cells. FEBS Lett 1989; 257: 134-7.

modulates the expression of collagenase and metalloproteinase inhibitor. EMBO 72. Edwards DR, Murphy G, Reynolds JJ, et al. Transforming growth factor beta

J 1987: 6 1899-904. 73. Overall CM, Wrana JL, Sudek J. Independent regulation of collagenase, 72 kD

progelatinase, and metalloendoproteinase inhibitor expression in human fibroblasts by transforming growth factor-beta. J Biol Chem 1989; 264 1860-9.

74. Kojima S, W i n DB. Mechanism of retinoid-induced activation of latent transforming growth factor beta in bovine endothelial cells. J Cell Physiol 1993; 155: 323-32.

obtained by outgrowth are representative of the fibrogenic cells in the liver. Hepatology 1995; 22: 788-97.

76. Roberts CJ. Birkenmeir TM, McQuillan JJ* et al. Transforming growth factor- beta stimulates the expression of fibronectin and of both subunits of the human fibronectin receptor by cultured human fibroblasts. J Biol Chem 1988; 263:

77. De Bleser PJ, Jannes P, van Bud-Offers SC, et al. Insulin-like growth factor- Il/mannose 6-phosphate receptor is expressed on CCI4-exposed rat fat-storing cells and facilitates activation of latent transforming growth factor-beta in cocultures with sinusoidal endothelial cells. Hepatology 1995; 21: 1429-37.

78. Friedman SL, Yamasaki G. Wong L. Modulation of transforming growth factor beta receptors of rat lipocytes during the hepatic wound healing response. J Biol Chem 1994: 269 I055 1-8.

79. Cheifetz S, Andres JL, Massague J. The transforming growth factor-beta receptor type 111 is a membrane proteoglycan. Domain structure of the receptor. J Biol Chem 1988; 263: 16984-91.

80. Lopez-Casillas F. Cheifetz 5, Doody J. Andres JL, Lane WS, Massague J. Structure and expression of the membrane proteoglycan betaglycan, a component of the TGF-beta receptor system. Cell 1991; 67: 785-95.

growth factor-beta protects against scarring in experimental kidney disease. Nature (London) 1992; 360 361-4.

82. Shah M, Foreman DM, Ferguson MWJ. Control of scarring in adult wounds by neutralising antibody to transforming growth factor beta. Lancet 1992; 339

83. Border WA, Okuda 5, Languino LR, Sporn MB. Ruoslahti E. Suppression of experimental glomerulonephritis by antiserum against transforming growth factor beta I. Nature (London) 1990; 346 371-4.

84. lsaka Y, Brees DK. lkegaya K, et al. Gene therapy by skeletal muscle expression of decorin prevents fibrotic disease in rat kidney. Nature Med 1996; 2: 418-23.

85. Sanderson N, Factor V, Nagy P, et al. Hepatic expression of mature transforming growth factor beta I in transgenic mice results in multiple tissue lesions. Proc Natl Acad Sci USA 1995; 9 2 2572-6.

factor. Cell 1986; 4 6 155-69.

growth factor and other polypeptide mitogens on DNA synthesis and growth of cultured rat liver fat storing cells. J Clin Invest 1989; 84: 1786-93.

secrete PDGF like factors stimulating the proliferation of cultured liver myofibroblastic cells. In: Gressner AM, Ramadori G, eds. Molecular and Cell

75. Blazejewski S, Preaux AM, Mallat A, et al. Human myofibroblast like cells

4586-92.

81. Border WA, Noble NA, Yamamoto T, et al. Natural inhibitor of transforming

21 3- 14.

86. Ross R, Raines EW, Bower-Pope DF. The biology of platelet derived growth

87. Pinzani M, Gesukado L, Sabbah GM, Abboud HE. Effects of platelet derived

88. Rieder H, Ramadori G, Meyer zum Buschenfelde K-H. Sinusoidal cells in vitro

biology of liver fibrogenesis. Dordrecht, The Netherlands: Kluwer Academic Publishers, I992 172-6.

89. Marra F. Choudhury GG, Pinzani M, Abboud HE. Regulation of platelet-derived growth factor secretion and gene expression in human liver fat-storing cells. Gastroenterology 1994; 107: I I 10- 17.

derived growth factor receptor in rat hepatic lipocytes during cellular activation in vivo and in culture. J Clin Invest 1994; 9 4 1563-9.

91. Friedman SL. Arthur MJP. Activation of cultured rat hepatic lipocytes by Kupffer cell conditioned medium. Direct enhancement of matrix synthesis and stimulation of cell proliferation via induction of platelet derived growth factor receptors. J Clin Invest 1989; 84: 1780-5.

92. Gressner AM, Hoffmann C, Lahme B, Scheckel L. Hepatocyte injury stimulates the mitogenic activation of cultured fat storing cells. In: Wise E, Wake K, Knook DL, eds. Cells of the hepatic sinusoid. Leiden, The Netherlands: The Kupffer Cell Foundation, 1995 vol. 5, 380-2.

like growth factor (IGF)-I- and IGF-ll-binding sites in rat liver fat storing cells (FSC) during in vitro transformation to myofibroblasts (MFB). In: Wise E, Wake K, Knook DL, eds. Cells of the hepatic sinusoid. Leiden, The Netherlands: The Kupffer Cell Foundation, 1995 vol. 5, 386-90.

capacity for synthesizing interstitial collagenase in vitro. Cell Mol Biol 1990; 3 6

90. Wong L, Yamasaki G, Johnson RJ, Friedman SL. Induction of beta-platelet-

93. Brenzel A, Weiner OH, Gressner AM. Stage-dependent expression of insulin-

94. Emonard H, Guillouzo A, Lapiere CM, Grimaud JA Human liver fibroblast

46 I -7. 95. lredale JP, Goddard S, Murphy G, Benyon RC, Arthur MJP. Tissue inhibitor of

metalloproteinase- I and interstitial collagenase expression in autoimmune chronic active hepatitis and activated human hepatic lipocytes. Clin Sci 1995: 89: 75-8 I.

96. Rockey DC, Maher JJ, Jarnagin WR, Gabbiani G, Friedman SL. Inhibition of rat hepatic lipocyte activation in culture by interferon-gamma. Hepatology 1992;

97. Geerts A, Vrijsen R, Schelliack P, Wisse E. Retinol affects the phenotype and 16 776-84.

protein synthesis of fat storing cell derived myofibroblasts in vitro. In: Wisse E, Knook DL, Decker K, eds. Cells of the hepatic sinusoid. Leiden, The Netherlands: The Kupffer Cell Foundation, I989 vol. 2, 20-4.

98. Davis BH, Kramer RT, Davidson NO. Retinoic acid modulates rat Ito cell proliferation. collagen, and transforming growth factor beta production. J Clin Invest 1990; 8 6 2062-70.

factor beta differentially inhibit platelet-derived-growth-factor-induced ito-cell activation. Biochem J 1991; 278 43-7.

hypovitaminosis A and the relationships with liver fibrosis. Semin Liver Dis 1993;

99. Davis BH, Rapp UR, Davidson NO. Retinoic acid and transforming growth

100. Hendriks HFJ, Bosma A, Brouwer A. Fat-storing cells: Hyper- and

13: 72-80. 101. Murphy G, Docherty AJP, Hembry RM, Reynolds JJ. Metalloproteinases and

tissue damage. Br J Rheumatol 1991; 30 (Suppl. I): 25-3 I. 102. Winwood PJ, Schuppan D, lredale JP, Kawser CA, Docherty AJP. Arthur MJP.

Kupffer cell-derived 95 kDa type IV collagenase/gelatinase B characterisation and expression in cultured cells. Hepatology 1995: 22: 304- 15.

103. Vyas SK, Leyland H, Gentry J, Arthur MJP. Transin (stromelysin) is expressed in early rat lipocyte primary culture. Gastroenterology 1995; 109 889-98.

104. Arthur MJP, Friedman SL, Roll FJ, Bissell DM. Lipocytes from normal rat liver release a neutral metalloproteinase that degrades basement membrane (type IVj collagen. J Clin Invest 1989; 8 4 1076-85.

Secretion of 72 kDa type IV collagenase/gelatinase by cultured human lipocytes: analysis of gene expression, protein synthesis and proteinase activity. Biochem

105. Arthur MJP, Stanley A, lredale JP, Rafferty JA, Hembry RM, Friedman SL.

J 1992; 287: 701 -7. 106. Herbst H, Heinrichs 0, Schuppan D, Milani S, Stein H. Temporal and spatial

patterns of transin/stromelysin RNA expression following toxic injury in rat liver. Virchows Archiv B Cell Pathol 1991; 60: 295-300.

C. porvum model of macrophage-induced liver injury is associated with expression of 72 kDa type IV collagenase/gelatinase. In: Knook DL, Wisse E, eds. Cells of the hepatic sinusoid. Leiden, The Netherlands: Kupffer Cell Foundation, 1993: vol. 4, 105-8.

108. lredale JP, Ferris WF, Goddard S, et al. TIMP-I expression is enhanced relative to interstitial collagenase messenger RNA in experimental liver injury and fibrosis. Hepatology 1996; 2 4 176-84.

109. Aimes RT. Quigley JP. Matrix metalloproteinase-2 is an interstitial collagenase - Inhibitor-free enzyme catalyzes the cleavage of collagen fibrils and soluble native

107. lredale JP, Winwood PJ, Kawser CA, Arthur MJP. Lipocyte proliferation in the

Page 10: Pathogenesis of Liver Fibrosis - Semantic Scholar · Pathogenesis of liver fibrosis R. ALCOLADO, M. J. P. ARTHUR and J. P. IREDALE University Medicine, University of Southampton,

I12 R Alcolado et al.

type I collagen generating the specific 3/4-and I/4-length fragments. J Biol Chem 1995; 2 7 0 5872-6.

I 10. Benyon RC, lredale JP, Goddard S, Winwood PJ, Arthur MJP. Increased expression of tissue inhibitor of metalloproteinases-l and -2 relative to interstitial collagenase in fibrotic human liver. Gastroenterology I996 I I0 821 -3 I.

I I I. Okazaki I, Maruyama K. Collagenase activity in experimental hepatic fibrosis.

I 12. Perez-Tamayo R, Montfort I, Gonzalez E. Collagenolytic activity in experimental

I 13. Maruyama K, Feinman L, Fainsilber Z, Nakano M, Okazaki I, Lieber CS.

Nature (London) 1974; 252: 49-SO.

cirrhosis of the liver. Exp Mol Pathol 1987; 47: 300-8.

Mammalian collagenase increases in early alcoholic liver disease and decreases with cirrhosis. Life Sci 1982; 3 0 1379-84.

I 14. lredale JP, Murphy G, Hembry RM, Friedman SL, Arthur MJP. Human hepatic lipocytes synthesize tissue inhibitor of metalloproteinases- I (TIMP- I): implications for regulation of matrix degradation in liver. J Clin Invest 1992; 9 0 282-7.

I 15. Kordula T, Guttgemann I, Rosejohn S, et al. Synthesis of tissue inhibitor of metalloproteinase-I (TIMP-I) in human hepatoma cells (HepG2) -

up-regulation by interleukin-6 and transforming growth factor-betal. FEBS Lett 1992; 313: 143-7.

Regulation of tissue inhibitor of metalloproteinases- I gene expression by cytokines and dexamethasone in rat hepatocyte primary cultures. Hepatology 1993; 18: 1437-42.

(TIMP-2) in rat liver cells is increased by lipopolysaccharide and prostaglandin E(2). FEBS Lett 1995; 357: 33-6.

I 18. Truden JL, Boros DL. Detection of alpha-2-macroglobulin. alpha-I-protease inhibitor, and neutral protease-antiprotease complexes within liver granulomas of schistosoma mansoni-infected mice. Am J Pathol 1988 130 281-8.

119. Murawaki Y, Yamarnoto H, Kawasaki H, Shima H. Serum tissue inhibitor of metalloproteinases in patients with chronic liver disease and with hepatocellular carcinoma. Clin Chim Acta 1993; 218: 47-58.

120. Andus T, Gross V. Tran-Thi T-A, Schreiber G, Nagashima M, Heinrich PC. The biosynthesis of acute-phase proteins in primary cultures of rat hepatocytes. Eur J Biochem 1983; 133: 561-71.

system in hepatic stellate cells. Hepatology I996 2 4 I 172-8.

I 16. Roeb E, Graeve L, Hoffmann R, Decker K, Edwards DR Heinrich PC.

I 17. Roeb E, Rosejohn S, Erren A, et al. Tissue inhibitor of metalloproteinases-2

I2 I. Leyland H, Gentry J. Arthur MJP, Benyon RC. The plasminogen activating