Invited Commentary Open Access
Copyright ©The Author(s) 1999. Published by Baishideng Publishing Group Inc. All rights reserved.
World J Gastroenterol. Oct 15, 1999; 5(5): 388-390
Published online Oct 15, 1999. doi: 10.3748/wjg.v5.i5.388
Fibrodynamics-elucidation of the mechanisms and sites of liver fibrogenesis
Catherine H Wu, Department of Medicine, Division of Gastroenterology-Hepatology, University of Connecticut Health Center, Farmington, CT, USA
Author contributions: All authors contributed equally to the work.
Correspondence to: Catherine H Wu, Department of Medicine, Division of Gastroenterology-Hepatology, University of Connecticut Health Center, 263 Farmington Avenue, Farmington, Connecticut 06030, USA
Fax: (860) 679-3159
Received: July 1, 1999
Revised: August 20, 1999
Accepted: September 24, 1999
Published online: October 15, 1999

Abstract
Key Words: liver cirrhosis/pathology, liver cirrhosis/etiology, liver cirrhosis/physiopathology, fibrodynamics, hepatic stellate cells



ORIGINAL ARTICLE

Dynamic changes of type I, III and IV collagen synthesis and distribution of collagen-producing cells in carbon tetrachloride- induced rat liver fibrosis.

MAJOR POINTS OF THE COMMENTED ARTICLE

In their article that appears in this issue, Du and colleagues used a combination of immunohistochemistry and in situ hybridization to demonstrate increased levels of collagen types I, III and IV in CCl4/choline deficient rat model of hepatic fibrosis. Over the course of 20 weeks of treatment mRNA levels for all three types of collagen were increased, but there was a preferential increase of type III collagen mRNA over the other two types. This is consistent with the results of previous investigators[1,2] where increases in protein levels of collagen types I, III and IV were found in CCl4 induced liver fibrosis. The authors clearly demonstrated that collagen type I, III and IV mRNAs were localized on sinusoidal cells using in situ hybridization. This result is also consistent with previous work of Maher and co-workers[3,4] who used the same technique to demonstrate the localization of both interstitial and basement collagen mRNAs in hepatic stellate cells in normal rat and human livers. The authors have utilized state-of-the-art technology to examine an important question in liver fibrosis-the cells responsible for overproduction of liver biomatrix components. Their results are consistent with the results of other investigators in the field. However, caution should be taken not to overinterpret results. Additional controls in which the in situ localization of collagen mRNAs in untreated normal liver as compared to that seen in fibrotic liver could have given a clearer picture of changes in the fibrotic liver. Nevertheless, the results add strong confirmation to the temporal course of fibrogenesis, and localization of the products in an important model.

COMMENTARY

Liver fibrosis is the common result of chronic hepatic injury of diverse origins such as chronic viral infections (HBV, HCV), metabolic/storage diseases (hemochromatosis), helminthic infections (schistosomiasis), chronic toxin exposure (alcohol and environmental poisons) and biliary obstruction (biliary cirrhosis). In end stage liver fibrosis or cirrhosis, the liver biomatrix may contain up to six to ten times more collagen and proteoglycans than in the normal state[5,6]. Because the connective tissue support of the liver parenchyma is particularly critical to its function, research that emphasizes the nature of liver biomatrix, the molecular regulation of the tumover of components of the biomatrix, and identification of liver cells responsible for the synthesis of biomatrix proteins are especially crucial for the ultimate design of effective therapies for liver fibrosis.

In the late 50’s, Hans Popper, the eminent hepatologist, observed a correlation between the histomorphology and biochemistry of liver collagens in chronic liver diseases[7]. In the four decades following Dr. Popper’s original observation, a great deal of research on liver biomatrix has resulted in our current knowledge of the pathogenesis of liver fibrosis. Progress has been made in three areas of liver fibrosis: characterization and quantitation of matrix component s in normal and fibrotic liver; identification of hepatic cells responsible for the increased synthesis of matrix proteins; and the role of cellular mediators of fibrogenesis.

Quantitation of matrix proteins in normal and fibrotic livers

The use of animal models of liver fibrosis such as the administration of liver toxins CCl4[8], dimethylnitrosamine[9], alcohol[10], helminthic infections[11,12] have greatly helped in the characterization of the temporal expression of various components of the biomatrix during fibrogenesis. There is increase in the amounts of collagens types I, III, IV, V and VI[1]. In early fibrosis, the amounts of types III and IV collagens increase relative to other collagens. In late fibrosis, type I collagen predominates[13]. Other components of liver biomatrix such as laminin, fibronectin and proteoglycans are also increased in fibrosis[14,15]. Although most investigations have shown changes at the protein and mRNA levels of various biomatrix components, the significance of the changes in the fibrogenic process remain hotly debated[16]. The biomatrix in the normal liver changes from being rich in basement membrane collagens to interstitial collagens during fibrogenesis.

Hepatic stellate cells is the major effector cell type in hepatic fibrosis

The search for effector cells in the liver responsible for collagen synthesis became feasible with the development of molecular probes[17] and antibodies[14,18] to components of liver biomatrix. Stellate cells are responsible for the increased synthesis in liver biomatrix proteins such as basement membrane collagens, interstitial collagens, fibronectin, laminin and proteoglycans[3,4]. Activation of hepatic stellate cells is the earliest response to liver injury. Upon activation, stellate cells lose stored lipids and retinoids[19] and rapidly undergo morphological changes to myofibroblast-like phenotype[20]. Other phenotypic changes of activated stellate cells include stimulation of α-actin gene expression[21,22] and increased synthesis of hepatic biomatrix components[3]. Activation also results in loss of an important feedback regulation of collagen synthesis by its terminal propeptides. In the normal liver, stellate cells are capable of controlling the amount of collagen needed for normal biomatrix formation by a feedback inhibition of collage n synthesis by its terminal propeptides[23,24]. Following activation, stellate cells lose their normal feedback regulation of collagen synthesis leading to increased accumulation of collagen[25]. In particular, there is an increased synthesis of types I and III collagen resulting in a biomatrix rich in interstitial collagens. There is increasing evidence that accumulation of fibers in the sinusoids is not only due to increased synthesis of collagens, but also is a result of decreased synthesis of tissue collagenases and increased synthesis of inhibitors of collagenase (TIMP-1: tissue inhibitors of metalloprotein-ase)[26]. Thus fibrogenesis is a net result of increased synthesis and decreased degradation of interstitial collagens of activated stellate cells.

Cellular mediators of hepatic fibrosis

Understanding the underlying molecular mechanisms responsible for hepatic fibrosis became feasible with the availability of molecular probes to cytokines. It is now accepted that the initial liver injury results in a host of cytokine responses from liver cells. Specifically, TGFβ[27], TNFα[28], PDGF[29] and Kupffer cell soluble factors[30] have been implicated in stellate cell activation and proliferation. TGFβ mRNA and protein levels are increased in activated stellate cells[27]. Over-expression of TGFβ gene in cultured fibroblasts[27] and in stellate cells[31] results in increased synthesis of collagens. Inhibitors of TGFβ decrease collagen synthesis in vivo[32,33] while transgenic mice over expressing TGFβ have kidney and liver fibrosis[34]. Both PDGF[29] and TNFα[35] are stellate cell mitogens. PDGF-induced stellate cell proliferation and matrix protein synthesis is mediated by factors secreted by Kupffer cells[30]. TNFα acts via transcription regulation of tissue collagenase and TIMP-1 genes in activated stellate cells[28].

Current research

The elucidation of the molecular mechanisms of cytokine regulation of liver biomatrix protein synthesis continue to be a focus of current research efforts. There is increasing evidence that cytokines may act via interactions with DNA binding proteins to affect matrix proteins synthesis. Both TGFβ and TNFá interact with known transcription factors such as C/EBP[36] and NFkappaB[37,38]. Transcription factors are DNA binding proteins which act as regulators of gene transcriptions[39,40]. There is continued interest in the search for regulatory elements within genes of matrix proteins[41]. Research on interactions of DNA binding proteins to regulatory elements on matrix protein genes are underway and may provide a link between cytokines and regulation of liver biomatrix.

Future directions

Effective therapy for chronic hepatic fibrosis can be designed only with complete understanding of the molecular mechanisms that regulate matrix protein gene expression. Future research may be centered on the application of gene therapy to control hepatic fibrosis[42]. Over-expression of tissue collagenase gene, inhibition of TGFβ gene expression are potential approach in controlling and regulating hepatic fibrosis[43].

Footnotes

Edited by Wang XL

References
1.  Schuppan D. Structure of the extracellular matrix in normal and fibrotic liver: collagens and glycoproteins. Semin Liver Dis. 1990;10:1-10.  [PubMed]  [DOI]  [Cited in This Article: ]
2.  Greenwel P, Rojkind M. Accelerated development of liver fibrosis in CCl4-treated rats by the weekly induction of acute phase response episodes: upregulation of alpha1(I) procollagen and tissue inhibitor of metalloproteinase-1 mRNAs. Biochim Biophys Acta. 1997;1361:177-184.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 13]  [Cited by in F6Publishing: 12]  [Article Influence: 0.4]  [Reference Citation Analysis (0)]
3.  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;86:1641-1648.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 307]  [Cited by in F6Publishing: 332]  [Article Influence: 9.8]  [Reference Citation Analysis (0)]
4.  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;82:8681-8685.  [PubMed]  [DOI]  [Cited in This Article: ]
5.  Rojkind M, Kershenobich D. Hepatic fibrosis. Prog Liver Dis. 1976;5:294-310.  [PubMed]  [DOI]  [Cited in This Article: ]
6.  Schuppan D. Structure of the extracellular matrix in normal and fibrotic liver: collagens and glycoproteins. Semin Liver Dis. 1990;10:1-10.  [PubMed]  [DOI]  [Cited in This Article: ]
7.  Kent G, Fels IG, Dubin A, Popper H. Collagen content based on hydroxyproline determinations in human and rat livers: its rela-tion to morphologically demonstrable reticulum and collagen fibers. Lab Invest. 1959;8:48.  [PubMed]  [DOI]  [Cited in This Article: ]
8.  Rojkind M, Giambrone MA, Ehrenpreise M. Proline oxidase ac-tivity and the availability of proline for collagen biosynthesis in Livers of CCl4 treated rats. Gastroenterology. 1977;73:1243.  [PubMed]  [DOI]  [Cited in This Article: ]
9.  Risteli J, Tuderman L, Kivirikko KI. Intracellular enzymes of collagen biosynthesis in rat liver as a function of age and in hepatic injury produced by dimethylnitrosamine. Biochem J. 1976;158:369-376.  [PubMed]  [DOI]  [Cited in This Article: ]
10.  Mezey E, Potter JJ, Maddrey WC. Hepatic collagen proline hy-droxylase activity in alcoholic liver disease. Clin Chem Acta. 1976;68:313-320.  [PubMed]  [DOI]  [Cited in This Article: ]
11.  Isaka Y, Brees DK, Ikegaya K, Kaneda Y, Imai E, Noble NA, Border WA. Gene therapy by skeletal muscle expression of decorin prevents fibrotic disease in rat kidney. Nat Med. 1996;2:418-423.  [PubMed]  [DOI]  [Cited in This Article: ]
12.  Wu CH, Giambrone MA, Howard DJ, Rojkind M, Wu GY. The nature of the collagen in hepatic fibrosis in advanced murine schistosomiasis. Hepatology. 1982;2:366-371.  [PubMed]  [DOI]  [Cited in This Article: ]
13.  Rojkind M, Giambrone MA, Biempica L. Collagen types in normal and cirrhotic liver. Gastroenterology. 1979;76:710-719.  [PubMed]  [DOI]  [Cited in This Article: ]
14.  Geerts A, Geuze HJ, Slot JW, Voss B, Schuppan D, Schellinck P, Wisse E. Immunogold localization of procollagen III, fibronectin and heparan sulfate proteoglycan on ultrathin frozen sections of the normal rat liver. Histochemistry. 1986;84:355-362.  [PubMed]  [DOI]  [Cited in This Article: ]
15.  Reid LM, Fiorino AS, Sigal SH, Brill S, Holst PA. Extracellular matrix gradients in the space of Disse: relevance to liver biology. Hepatology. 1992;15:1198-1203.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 113]  [Cited by in F6Publishing: 99]  [Article Influence: 3.1]  [Reference Citation Analysis (0)]
16.  Jarnagin WR, Rockey DC, Koteliansky VE, Wang SS, Bissell DM. Expression of variant fibronectins in wound healing: cellular source and biological activity of the EIIIA segment in rat hepatic fibrogenesis. J Cell Biol. 1994;127:2037-2048.  [PubMed]  [DOI]  [Cited in This Article: ]
17.  Adams SI Regulation of collagen gene expression. in Extracellu-lar Matrix: Chemistry, Biology, and Pathobiology with emphasis on the liver. Zcrn MA and Reid LM (eds). Marcell Dekker, Inc. New York 1993; p91-119.  [PubMed]  [DOI]  [Cited in This Article: ]
18.  Becker J, Schuppan D, Benzian H, Bals T, Hahn EG, Cantaluppi C, Reichart P. Immunohistochemical distribution of collagens types IV, V, and VI and of pro-collagens types I and III in human alveolar bone and dentine. J Histochem Cytochem. 1986;34:1417-1429.  [PubMed]  [DOI]  [Cited in This Article: ]
19.  Mak KM, Leo AM, Lieber CS. Alcoholic liver injury in baboons: transformation of lipocytes to transitional cells. Gastroenterology. 1984;87:188-200.  [PubMed]  [DOI]  [Cited in This Article: ]
20.  Mak KM, Leo AM, Lieber CS. Alcoholic liver injury in baboons: transformation of lipocytes to transitional cells. Gastroenterology. 1984;87: 188-200.  [PubMed]  [DOI]  [Cited in This Article: ]
21.  Tanaka Y, Nouchi T, Yamane M, Irie T, Miyakawa H, Sato C, Marumo F. Phenotypic modulation in lipocytes in experimental liver fibrosis. J Pathol. 1991;164:273-278.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 52]  [Cited by in F6Publishing: 53]  [Article Influence: 1.6]  [Reference Citation Analysis (0)]
22.  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;24:193-203.  [PubMed]  [DOI]  [Cited in This Article: ]
23.  Wu CH, Donovan CB, Wu GY. Evidence for pretranslational regulation of collagen synthesis by procollagen propeptides. J Biol Chem. 1986;261:10482-10484.  [PubMed]  [DOI]  [Cited in This Article: ]
24.  Wu CH, Walton CM, Wu GY. Propeptide-mediated regulation of procollagen synthesis in IMR-90 human lung fibroblast cell cultures. Evidence for transcriptional control. J Biol Chem. 1991;266:2983-2987.  [PubMed]  [DOI]  [Cited in This Article: ]
25.  Ikeda H, Wu GY, Wu CH. Lipocytes from fibrotic rat liver have an impaired feedback response to procollagen propeptides. Am J Physiol. 1993;264:G157-G162.  [PubMed]  [DOI]  [Cited in This Article: ]
26.  Iredale JP, Murphy G, Hembry RM, Friedman SL, Arthur MJ. Human hepatic lipocytes synthesize tissue inhibitor of metalloproteinases-1. Implications for regulation of matrix degradation in liver. J Clin Invest. 1992;90:282-287.  [PubMed]  [DOI]  [Cited in This Article: ]
27.  Roberts AB, Sporn MB, Assoian RK, Smith JM, Roche NS, Wakefield LM, Heine UI, Liotta LA, Falanga V, Kehrl JH. Transforming growth factor type beta: rapid induction of fibrosis and angiogenesis in vivo and stimulation of collagen formation in vitro. Proc Natl Acad Sci USA. 1986;83:4167-4171.  [PubMed]  [DOI]  [Cited in This Article: ]
28.  Iredale JP. Matrix turnover in fibrogenesis. Hepatogastroenterology. 1996;43:56-71.  [PubMed]  [DOI]  [Cited in This Article: ]
29.  Pinzani M, Gesualdo L, Sabbah GM, Abboud HE. Effects of platelet-derived growth factor and other polypeptide mitogens on DNA synthesis and growth of cultured rat liver fat-storing cells. J Clin Invest. 1989;84:1786-1793.  [PubMed]  [DOI]  [Cited in This Article: ]
30.  Friedman SL, Arthur MJ. 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;84:1780-1785.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 292]  [Cited by in F6Publishing: 301]  [Article Influence: 8.6]  [Reference Citation Analysis (0)]
31.  Milani S, Schupan 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;254-263.  [PubMed]  [DOI]  [Cited in This Article: ]
32.  Border WA, Noble NA, Yamamoto T, Harper JR, Yamaguchi Yu, Pierschbacher MD, Ruoslahti E. Natural inhibitor of transforming growth factor-beta protects against scarring in experimental kidney disease. Nature. 1992;360:361-364.  [PubMed]  [DOI]  [Cited in This Article: ]
33.  Shah M, Foreman DM, Ferguson MW. Control of scarring in adult wounds by neutralising antibody to transforming growth factor beta. Lancet. 1992;339:213-214.  [PubMed]  [DOI]  [Cited in This Article: ]
34.  Sanderson N, Factor V, Nagy P, Kopp J, Kondaiah P, Wakefield L, Roberts AB, Sporn MB, Thorgeirsson SS. Hepatic expression of mature transforming growth factor beta 1 in transgenic mice results in multiple tissue lesions. Proc Natl Acad Sci USA. 1995;92:2572-2576.  [PubMed]  [DOI]  [Cited in This Article: ]
35.  Camussi G, Albano E, Tetta C, Bussolino F. The molecular action of tumor necrosis factor-alpha. Eur J Biochem. 1991;202:3-14.  [PubMed]  [DOI]  [Cited in This Article: ]
36.  Garcia-Trevijiano ER, Iraburu MJ, Fontana L, Dominguez-Rosales JA, Auster A, Covarrubias-Pinedo A, Rojkind M. Transforming growth factor betal induces the expression of alphal (I) procollagen mRNA by a hydrogen peroxide-C/EBPbeta-dependent mechanism in rate hepatic stellate cells. Hepatology. 1999;29:960-970.  [PubMed]  [DOI]  [Cited in This Article: ]
37.  Zeldin G, Yang SQ, Yin M, Lin HZ, Rai R, Diehl AM. Alcohol and cytokine-inducible transcription factors. Alcoholism, Clinical & Experimental Research. 1996;20:1639-1645.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 50]  [Cited by in F6Publishing: 46]  [Article Influence: 1.6]  [Reference Citation Analysis (0)]
38.  Yang SQ, Lin HZ, Yin M, Albrecht JH, Diehl AM. Effects of chronic ethanol consumption on cytokine regulation of liver regeneration. Am J Physiol. 1998;275:G696-G704.  [PubMed]  [DOI]  [Cited in This Article: ]
39.  Hellerbrand C, Stefanovic B, Giordano F, Burchardt ER, Brenner DA. The role of TGFbeta1 in initiating hepatic stellate cell activation in vivo. J Hepatol. 1999;30:77-87.  [PubMed]  [DOI]  [Cited in This Article: ]
40.  Hellerbrand C, Jobin C, Licato LL, Sartor RB, Brenner DA. Cytokines induce NF-kappaB in activated but not in quiescent rat hepatic stellate cells. Am J Physiol. 1998;275:G269-G278.  [PubMed]  [DOI]  [Cited in This Article: ]
41.  Stefanovic B, Hellerbrand C, Holcik M, Briendl M, Aliebhaber S, Brenner DA. Posttranscriptional regulation of collagen alpha1(I) mRNA in hepatic stellate cells. Mol Cell Biol. 1997;17:5201-5209.  [PubMed]  [DOI]  [Cited in This Article: ]
42.  Hellerbrand C, Jobin C, Iimuro Y, Licato L, Sartor RB, Brenner DA. Inhibition of NFkappaB in activated rat hepatic stellate cells by proteasome inhibitors and an IkappaB super-repressor. Hepatology. 1998;27:1285-1295.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 138]  [Cited by in F6Publishing: 148]  [Article Influence: 5.7]  [Reference Citation Analysis (0)]
43.  Isaka Y, Brees DK, Ikegaya K, Kaneda Y, Imai E, Noblc NA, Border WA. Gene therapy by skeletal muscle expression of decorin prevents fibrotic disease in rat kidney. Nature Med. 1996;2:418-423.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 349]  [Cited by in F6Publishing: 329]  [Article Influence: 11.8]  [Reference Citation Analysis (0)]