Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 72
Filter
1.
Gut ; 54(1): 142-51, 2005 Jan.
Article in English | MEDLINE | ID: mdl-15591520

ABSTRACT

BACKGROUND: Hepatic stellate cells (HSCs) are a major fibrogenic cell type that contributes to collagen accumulation during chronic liver disease. With increasing interest in developing antifibrotic therapies, there is a need for cell lines that preserve the in vivo phenotype of human HSCs to elucidate pathways of human hepatic fibrosis. We established and characterised two human HSC cell lines termed LX-1 and LX-2, and compared their features with those of primary human stellate cells. METHODS AND RESULTS: LX-1 and LX-2 were generated by either SV40 T antigen immortalisation (LX-1) or spontaneous immortalisation in low serum conditions (LX-2). Both lines express alpha smooth muscle actin, vimentin, and glial fibrillary acid protein, as visualised by immunocytochemistry. Similar to primary HSCs, both lines express key receptors regulating hepatic fibrosis, including platelet derived growth factor receptor beta (betaPDGF-R), obese receptor long form (Ob-RL), and discoidin domain receptor 2 (DDR2), and also proteins involved in matrix remodelling; matrix metalloproteinase (MMP)-2, tissue inhibitor of matrix metalloproteinase (TIMP)-2, and MT1-MMP, as determined by western analyses. LX-2 have reduced expression of TIMP-1. LX-2, but not LX-1, proliferate in response to PDGF. Both lines express mRNAs for alpha1(I) procollagen and HSP47. Transforming growth factor beta1 stimulation increased their alpha1(I) procollagen mRNA expression, as determined by quantitative reverse transcription-polymerase chain reaction. LX-2, but not LX-1, cells are highly transfectable. Both lines had a retinoid phenotype typical of stellate cells. Microarray analyses showed strong similarity in gene expression between primary HSCs and either LX-1 (98.4%) or LX-2 (98.7%), with expression of multiple neuronal genes. CONCLUSIONS: LX-1 and LX-2 human HSC lines provide valuable new tools in the study of liver disease. Both lines retain key features of HSCs. Two unique advantages of LX-2 are their viability in serum free media and high transfectability.


Subject(s)
Adipocytes/cytology , Cell Line/metabolism , Liver Cirrhosis/pathology , Liver/cytology , Collagen Type I/metabolism , Culture Media , Culture Media, Serum-Free , Gene Expression , Humans , Intermediate Filament Proteins/metabolism , Matrix Metalloproteinases/metabolism , Transfection , Vitamin A/metabolism
2.
Gut ; 49(5): 713-9, 2001 Nov.
Article in English | MEDLINE | ID: mdl-11600477

ABSTRACT

BACKGROUND: Activation of hepatic stellate cells (HSCs) to a myofibroblastic phenotype is a key event in liver fibrosis. Identification of transcription factors with activities that are modulated during HSC activation will improve our understanding of the molecular events controlling HSC activation. AIMS: To determine if changes in E-box DNA binding activity occur during in vitro and in vivo activation of rat and human HSCs and to investigate mechanisms underlying any observed changes. METHODS: Nuclear extracts were prepared from rat HSCs isolated and cultured from normal and carbon tetrachloride injured rat livers and from HSCs isolated from human liver. EMSA analysis of E-box DNA binding activity was performed on nuclear extracts to determine changes during HSC activation. Western and northern blot analysis of MyoD and Id1 basic helix-loop-helix (bHLH) proteins was performed to confirm expression in HSC. RESULTS: HSC activation was associated with inducible expression of two low mobility E-box binding complexes that were immunoreactive with an anti-MyoD antibody. MyoD mRNA expression was found at similar levels in freshly isolated and activated HSCs; in contrast, MyoD protein expression was elevated in activated HSCs. Activation of rat HSCs was accompanied by reduced expression of the inhibitory bHLH protein Id1. CONCLUSIONS: In vitro and in vivo activation of rat and human HSCs is accompanied by induction of MyoD binding to E-box DNA sequences which appears to be mechanistically associated with elevated MyoD protein expression and reduced expression of the inhibitory Id1 protein. Clarification of the role of MyoD and Id1 proteins in HSC activation and liver fibrogenesis is now required.


Subject(s)
DNA-Binding Proteins/physiology , E-Box Elements/physiology , Liver/cytology , Repressor Proteins , Adult , Animals , Blotting, Northern/methods , Blotting, Western/methods , Carbon Tetrachloride , Cell Differentiation , Cells, Cultured , Helix-Loop-Helix Motifs/physiology , Humans , Inhibitor of Differentiation Protein 1 , Liver/drug effects , Male , MyoD Protein/physiology , RNA, Messenger/analysis , Rats , Rats, Sprague-Dawley , Transcription Factors/physiology
3.
Semin Liver Dis ; 21(3): 373-84, 2001 Aug.
Article in English | MEDLINE | ID: mdl-11586466

ABSTRACT

Following liver injury, hepatic stellate cells (HSCs) become activated and express a combination of matrix metalloproteinases (MMPs) and their specific tissue inhibitors (TIMPs). In the early phases of liver injury (and primary cell culture), HSCs transiently express MMP-3, MMP-13, and uroplasminogen activator (uPA) and exhibit a matrix-degrading phenotype. In the later stages of liver injury and HSC activation, the pattern changes and the cells express a combination of MMPs that have the ability to degrade normal liver matrix, while inhibiting degradation of the fibrillar collagens that accumulate in liver fibrosis. This pattern is characterized by the combination of pro-MMP-2 and membrane type 1 (MT1)-MMP expression, which drive pericellular generation of active MMP-2 and local degradation of normal liver matrix. In addition there is a marked increase in expression of TIMP-1 leading to a more global inhibition of degradation of fibrillar liver collagens by interstitial collagenases (MMP-1/MMP-13). These pathways play a significant role in the progression of liver fibrosis. Following cessation of liver injury, the pattern reverses and TIMP-1 in particular is rapidly downregulated. This phase is characterized by increasing activity of collagenases, degradation of liver matrix, and regression of liver fibrosis.


Subject(s)
Extracellular Matrix/metabolism , Liver Cirrhosis/physiopathology , Liver/cytology , Liver/pathology , Matrix Metalloproteinases/biosynthesis , Cell Culture Techniques , Collagenases/biosynthesis , Collagenases/metabolism , Down-Regulation , Humans , Matrix Metalloproteinases/metabolism , Signal Transduction , Tissue Inhibitor of Metalloproteinases/metabolism
4.
Gut ; 49(4): 577-83, 2001 Oct.
Article in English | MEDLINE | ID: mdl-11559657

ABSTRACT

BACKGROUND: Following liver injury, hepatic stellate cells (HSC) transform into myofibroblast-like cells (activation) and are the major source of type I collagen and the potent collagenase inhibitors tissue inhibitors of metalloproteinases 1 and 2 (TIMP-1 and TIMP-2) in the fibrotic liver. The reproductive hormone relaxin has been reported to reduce collagen and TIMP-1 expression by dermal and lung fibroblasts and thus has potential antifibrotic activity in liver fibrosis. AIMS: To determine the effects of relaxin on activated HSC. METHODS: Following isolation, HSC were activated by culture on plastic and exposed to relaxin (1-100 ng/ml). Collagen deposition was determined by Sirius red dye binding and radiolabelled proline incorporation. Matrix metalloproteinase (MMP) and TIMP expression were assessed by zymography and northern analysis. Transforming growth factor beta1 (TGF-beta1) mRNA and protein levels were quantified by northern analysis and ELISA, respectively. RESULTS: Exposure of activated HSC to relaxin resulted in a concentration dependent decrease in both collagen synthesis and deposition. There was a parallel decrease in TIMP-1 and TIMP-2 secretion into the HSC conditioned media but no change in gelatinase expression was observed. Northern analysis demonstrated that primary HSC, continuously exposed to relaxin, had decreased TIMP-1 mRNA expression but unaltered type I collagen, collagenase (MMP-13), alpha smooth muscle actin, and TGF-beta1 mRNA expression. CONCLUSION: These data demonstrate that relaxin modulates effective collagen deposition by HSC, at least in part, due to changes in the pattern of matrix degradation.


Subject(s)
Collagen/metabolism , Hepatocytes/drug effects , Liver Cirrhosis/metabolism , Relaxin/pharmacology , Animals , Blotting, Northern , Cells, Cultured , Dose-Response Relationship, Drug , Enzyme-Linked Immunosorbent Assay , Hepatocytes/metabolism , Male , Matrix Metalloproteinases/metabolism , RNA, Messenger/analysis , Rats , Rats, Sprague-Dawley , Tissue Inhibitor of Metalloproteinases/metabolism , Transforming Growth Factor beta/metabolism
5.
Gastroenterology ; 121(3): 685-98, 2001 Sep.
Article in English | MEDLINE | ID: mdl-11522753

ABSTRACT

BACKGROUND & AIMS: Hepatic stellate cells (HSCs) play a pivotal role in liver fibrosis and stimulating their apoptosis could be an effective treatment for liver fibrosis. METHODS: Activated HSCs, hepatocytes, and rats with liver fibrosis were treated with gliotoxin. RESULTS: Addition of gliotoxin to activated (alpha-smooth muscle actin positive) rat and human HSCs resulted in morphologic alterations typical of apoptosis. Within 2-3 hours of incubation, caspase 3 activity was markedly induced and caspase inhibitor 1 (Z-VAD-FMK)-sensitive oligonucleosome-length DNA fragments were detectable by gel electrophoresis of low molecular weight DNA. Apoptosis was widespread as judged by fluorescence-activated cell sorter analysis and terminal deoxynucleotidyl transferase-mediated deoxyuridine triphosphate nick-end labeling staining in both rat and human HSCs at concentrations that had no effect on the viability of rat hepatocytes. Gliotoxin treatment significantly reduced the number of activated stellate cells and mean thickness of bridging fibrotic septae in livers from rats treated with carbon tetrachloride. CONCLUSIONS: These data demonstrate proof-of-concept that by up-regulating HSC apoptosis, the extent of fibrosis can be decreased in inflammatory liver injury.


Subject(s)
Apoptosis/drug effects , Gliotoxin/pharmacology , Immunosuppressive Agents/pharmacology , Liver Cirrhosis/drug therapy , Liver/pathology , Animals , Anti-Allergic Agents/pharmacology , Calcium/metabolism , Carbon Tetrachloride , Chlorpromazine/pharmacology , Collagen/analysis , Cycloheximide/pharmacology , Disease Models, Animal , Dopamine Antagonists/pharmacology , Gliotoxin/chemistry , Humans , Immunosuppressive Agents/chemistry , In Situ Nick-End Labeling , In Vitro Techniques , Liver/chemistry , Liver Cirrhosis/chemically induced , Liver Cirrhosis/pathology , Male , Methapyrilene/pharmacology , Mitochondria/metabolism , NF-kappa B/metabolism , Protein Synthesis Inhibitors/pharmacology , Rats , Rats, Sprague-Dawley , Tumor Necrosis Factor-alpha/pharmacology
6.
J Biol Chem ; 276(26): 24414-21, 2001 Jun 29.
Article in English | MEDLINE | ID: mdl-11337499

ABSTRACT

Activation of hepatic stellate cells (HSCs) to a myofibroblast-like phenotype is the pivotal event in hepatic wound healing and fibrosis. Rat HSCs activated in vitro express JunD, Fra2, and FosB as the predominant AP-1 DNA-binding proteins, and all three associate with an AP-1 sequence that is essential for activity of the tissue inhibitor of metalloproteinases-1 (TIMP-1) promoter. In this study, we used expression vectors for wild-type, dominant-negative, and forced homodimeric (Jun/eb1 chimeric factors) forms of JunD and other Fos and Jun proteins to determine the requirement for JunD in the transcriptional regulation of the TIMP-1 and interleukin-6 (IL-6) genes. JunD activity was required for TIMP-1 gene promoter activity, whereas overexpression of Fra2 or FosB caused a repression of promoter activity. The ability of homodimeric JunD/eb1 to elevate TIMP-1 promoter activity supports a role for JunD homodimers as the major AP-1-dependent transactivators of the TIMP-1 gene. IL-6 promoter activity was induced upon activation of HSCs and also required JunD activity; however, expression of JunD/eb1 homodimers resulted in transcriptional repression. Mutagenesis of the IL-6 promoter showed that an AP-1 DNA-binding site previously reported to be an activator of transcription in fibroblasts functions as a suppressor of promoter activity in HSCs. We conclude that JunD activates IL-6 gene transcription as a heterodimer and operates at an alternative DNA-binding site in the promoter. The relevance of these findings to events occurring in the injured liver was addressed by showing that AP-1 DNA-binding complexes are induced during HSC activation and contain JunD as the predominant Jun family protein. JunD is therefore an important transcriptional regulator of genes responsive to Jun homo- and heterodimers in activated HSCs.


Subject(s)
Hepatocytes/metabolism , Interleukin-6/genetics , Proto-Oncogene Proteins c-jun/physiology , Tissue Inhibitor of Metalloproteinase-1/genetics , Animals , Carbon Tetrachloride , Cells, Cultured , Dimerization , Interleukin-6/biosynthesis , Liver Cirrhosis/chemically induced , Liver Cirrhosis/metabolism , Male , Mutation , Promoter Regions, Genetic , Proto-Oncogene Proteins c-fos/metabolism , Proto-Oncogene Proteins c-jun/genetics , RNA, Messenger/biosynthesis , Rats , Rats, Sprague-Dawley , Trans-Activators/physiology , Transcription Factor AP-1/metabolism , Transcription, Genetic
7.
Gut ; 48(4): 548-57, 2001 Apr.
Article in English | MEDLINE | ID: mdl-11247901

ABSTRACT

BACKGROUND: Activated hepatic stellate cells (HSC) are central to the pathogenesis of liver fibrosis, both as a source of fibrillar collagens that characterise fibrosis and matrix degrading metalloproteinases and their tissue inhibitors, the TIMPs. AIMS: To test the hypothesis that HSC apoptosis is critical to recovery from biliary fibrosis and that soluble growth factors may regulate HSC survival and apoptosis. METHODS: Rats (n=15) were subjected to bile duct ligation for 21 days, after which biliodigestive anastomosis was undertaken (n=13). Livers were harvested at fixed time points of recovery for periods of up to 42 days. Numbers of activated HSCs were quantified after alpha smooth muscle actin staining and HSC apoptosis was detected by terminal UDP-nick end labelling (TUNEL) staining and quantified at each time point. HSC apoptosis was quantified in vitro in the presence or absence of insulin-like growth factor (IGF)-1, IGF-2, platelet derived growth factor (PDGF), and transforming growth factor beta1 (TGF-beta1). RESULTS: Following biliodigestive anastomosis after 21 days of bile duct ligation, rat liver demonstrated a progressive resolution of biliary fibrosis over 42 days, associated with a fivefold decrease in activated HSC determined by alpha smooth muscle actin staining. TUNEL staining indicated that loss of activated HSC resulted from an increase in the rate of apoptosis during the first two days post biliodigestive anastomosis. Serum deprivation and culture in the presence of 50 microM cycloheximide was associated with an increase in HSC apoptosis which was significantly inhibited by addition of 10 ng/ml and 100 ng/ml IGF-1, respectively (0.05>p, n=5). In contrast, 1 and 10 ng/ml of TGF-beta1 caused a significant increase in HSC apoptosis compared with serum free controls (p<0.05, n=4). PDGF and IGF-2 were neutral with respect to their effect on HSC apoptosis. CONCLUSION: HSC apoptosis plays a critical role in the spontaneous recovery from biliary fibrosis. Both survival and apoptosis of HSC are regulated by growth factors expressed during fibrotic liver injury.


Subject(s)
Apoptosis/physiology , Growth Substances/physiology , Hepatocytes/physiology , Liver Cirrhosis, Biliary/pathology , Animals , Apoptosis/drug effects , Cell Count , Cycloheximide/pharmacology , Hepatocytes/drug effects , In Situ Nick-End Labeling , Insulin-Like Growth Factor I/physiology , Insulin-Like Growth Factor II/physiology , Male , Platelet-Derived Growth Factor/physiology , Rats , Rats, Sprague-Dawley , Transforming Growth Factor beta/physiology
8.
Am J Physiol Gastrointest Liver Physiol ; 279(2): G245-9, 2000 Aug.
Article in English | MEDLINE | ID: mdl-10915630

ABSTRACT

Liver fibrosis is characterized by activation of hepatic stellate cells, which are then involved in synthesis of matrix proteins and in regulating matrix degradation. In the acute phases of liver injury and as liver fibrosis progresses, there is increased expression of matrix metalloproteinases (MMPs) and their tissue inhibitors (TIMPs). Among the changes described, striking features include increased expression of gelatinase A (MMP-2) and membrane type 1-MMP (MT(1)-MMP; MMP-14) as well as TIMP-1 and TIMP-2. These molecules and other family members are involved in regulating degradation of both normal and fibrotic liver matrix. This article outlines recent progress in this field and discusses the mechanisms by which MMPs and TIMPs may contribute to the progression and regression of liver fibrosis. Recently described properties of MMPs and TIMPs of relevance to the pathogenesis of liver fibrosis are outlined. The proposal that regression of liver fibrosis is mediated by decreased expression of TIMPs and involves degradation of fibrillar collagens by a combination of MT(1)-MMP and gelatinase A, in addition to interstitial collagenase, is explored.


Subject(s)
Liver Cirrhosis/metabolism , Liver/enzymology , Metalloendopeptidases/antagonists & inhibitors , Metalloendopeptidases/metabolism , Humans , Liver/pathology , Liver Cirrhosis/pathology , Tissue Inhibitor of Metalloproteinase-1/metabolism , Tissue Inhibitor of Metalloproteinase-2/metabolism
9.
J Hepatol ; 32(5): 754-61, 2000 May.
Article in English | MEDLINE | ID: mdl-10845662

ABSTRACT

BACKGROUND/AIMS: Connective tissue growth factor is a recently described mitogenic protein implicated in a variety of fibrotic disorders. Connective tissue growth factor may be a downstream mediator of the pro-fibrotic and mitogenic actions of transforming growth factor-beta, promoting extracellular matrix deposition and fibrogenesis. As transforming growth factor-beta is considered important to the pathogenesis of hepatic fibrosis, we examined the possible contribution of connective tissue growth factor to this process. METHODS: Connective tissue growth factor expression was examined in normal and fibrotic human and rat livers using RT-PCR and ribonuclease protection assays, and in primary cultures of rat hepatic stellate cells by Northern and Western blotting. RESULTS: Ribonuclease protection assays demonstrated connective tissue growth factor mRNA was increased 3-5-fold in human fibrotic liver compared with normal. RT-PCR showed this mRNA was increased in carbon-tetrachloride-treated rat liver. Northern analysis showed connective tissue growth factor mRNA was increasingly expressed during progressive activation of cultured rat hepatic stellate cells. Western analysis confirmed that freshly isolated hepatic stellate cells secreted relatively little connective tissue growth factor compared with hepatic stellate cells activated in culture. Hepatic stellate cells stimulated with transforming growth factor-beta showed increased expression of connective tissue growth factor mRNA and protein. CONCLUSIONS: Connective tissue growth factor mRNA is consistently upregulated in human liver cirrhosis of various aetiologies, supporting a role for this growth factor in hepatic fibrogenesis. Our studies suggest that hepatic stellate cells may be an important source of hepatic connective tissue growth factor in vivo, particularly following stimulation with transforming growth factor-beta.


Subject(s)
Connective Tissue Cells/metabolism , Growth Substances/biosynthesis , Immediate-Early Proteins/biosynthesis , Intercellular Signaling Peptides and Proteins , Liver Cirrhosis, Experimental/metabolism , Liver Cirrhosis/metabolism , Animals , Cells, Cultured , Connective Tissue Cells/pathology , Connective Tissue Growth Factor , Humans , Liver/metabolism , Liver/pathology , RNA, Messenger/analysis , Rats
10.
J Biol Chem ; 275(9): 6657-63, 2000 Mar 03.
Article in English | MEDLINE | ID: mdl-10692475

ABSTRACT

Elevated expression of the tissue inhibitor of metalloproteinases-1 (TIMP-1) protein and mRNA has been reported in human diseases including cancers and tissue fibrosis. Regulation of TIMP-1 gene expression is mainly mediated at the level of gene transcription and involves the activation of several well known transcription factors including those belonging to the AP-1, STAT, and Pea3/Ets families. In the current study, we have used DNase-1 footprinting to identify a new regulatory element (5'-TGTGGTTTCCG-3') present in the human TIMP-1 gene promoter. Mutagenesis and transfection studies in culture-activated rat hepatic stellate cells and the human Jurkat T cell line demonstrated that the new element named upstream TIMP-1 element-1 (UTE-1) is essential for transcriptional activity of the human TIMP-1 promoter. Electrophoretic mobility shift assay studies revealed that UTE-1 can form protein-DNA complexes of distinct mobilities with nuclear extracts from a variety of mammalian cell types and showed that induction of a high mobility UTE-1 complex is associated with culture activation of freshly isolated rat hepatic stellate cells. A combination of UV-cross-linking and Southwestern blotting techniques demonstrated that UTE-1 directly interacts with a 30-kDa nuclear protein that appears to be present in all cell types tested. We conclude that UTE-1 is a novel regulatory element that in combination with its cellular binding proteins may be an important component of the mechanisms controlling TIMP-1 expression in normal and pathological states.


Subject(s)
Gene Expression Regulation, Enzymologic/genetics , Nuclear Proteins/metabolism , Promoter Regions, Genetic , Regulatory Sequences, Nucleic Acid/genetics , Tissue Inhibitor of Metalloproteinase-1/genetics , Animals , Cells, Cultured , DNA Footprinting , DNA-Binding Proteins/analysis , Genes, Reporter , Humans , Liver/metabolism , Rats , Rats, Sprague-Dawley , Transcription Factors/metabolism , Ultraviolet Rays
11.
Hepatology ; 30(4): 977-86, 1999 Oct.
Article in English | MEDLINE | ID: mdl-10498650

ABSTRACT

Activated hepatic stellate cells (HSCs) are a potential source of gelatinase A, which accumulates in fibrotic livers. Progelatinase A activation requires its binding to a complex of membrane-type matrix metalloproteinase (MT-MMP) and tissue inhibitor of metalloproteinases (TIMP)-2. These studies examine gelatinase A, MT1-MMP, and TIMP-2 synthesis by HSCs during activation in vitro and the potential role of gelatinase A in promoting HSC proliferation. Gelatinase A, MT1-MMP, and TIMP-2 messenger RNA (mRNA) were all upregulated in HSCs activated on plastic over 5 to 14 days. Gelatinase A expression was maximal at 7 days of culture, coinciding with the peak of HSC proliferation and the onset of procollagen I and alpha-smooth muscle actin (alpha-SMA) mRNA expression. Active forms of gelatinase A of 62 kd and 66 kd were secreted by activated HSCs and reached a maximum of 12.1% of total enzyme in 14-day culture supernatants. Treatment of HSCs with concanavalin A (con A) induced activation of MT1-MMP and enhanced secretion of activated gelatinase A, which reached a maximum of 44.4% of the total enzyme secreted into culture supernatants using 30 microgram/mL con A. [(14)C]-gelatin degradation assays confirmed the presence of gelatinolytic activity in activated HSC supernatants, which reached a maximum level at 7 days of culture. Antisense oligonucleotide inhibition of endogenous progelatinase A production, or the MMP inhibitor 1,10-phenanthroline inhibited (3)H-thymidine incorporation into HSC DNA by greater than 50%. We conclude that HSCs produce progelatinase A during activation in vitro and activate this enzyme coincident with MT1-MMP and TIMP-2 synthesis. Gelatinase A activity is required for maximal proliferation of HSCs in vitro suggesting this metalloproteinase is an autocrine proliferation factor for HSCs.


Subject(s)
Enzyme Precursors/metabolism , Gelatinases/metabolism , Liver/enzymology , Metalloendopeptidases/metabolism , Animals , Biocompatible Materials , Cell Division/physiology , Cells, Cultured , Collagen , Drug Combinations , Enzyme Activation , Enzyme Precursors/biosynthesis , Enzyme Precursors/genetics , Enzyme Precursors/physiology , Gelatinases/biosynthesis , Gelatinases/genetics , Gelatinases/physiology , Isoenzymes/metabolism , Laminin , Liver/cytology , Male , Matrix Metalloproteinases, Membrane-Associated , Metalloendopeptidases/biosynthesis , Metalloendopeptidases/genetics , Metalloendopeptidases/physiology , Proteoglycans , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Tissue Inhibitor of Metalloproteinase-2/metabolism
12.
Eur J Gastroenterol Hepatol ; 11(9): 949-55, 1999 Sep.
Article in English | MEDLINE | ID: mdl-10503830

ABSTRACT

Acute liver failure arises from an imbalance between liver cell death and regeneration. The severity of the insult determines whether liver cells die by apoptosis or necrosis, and this in turn affects the magnitude of the inflammatory reaction. Liver cell death occurs through complex cellular interactions and is mediated by immunological, inflammatory and chemical components. Regeneration of the liver cell mass also depends on cellular interactions, often involving those same mediators implicated in the injury itself. A greater understanding of these processes will lead ultimately to targeted rational therapy in acute liver failure.


Subject(s)
Liver Failure, Acute/physiopathology , Liver/cytology , Cell Death/immunology , Humans , Liver/immunology , Liver/pathology , Liver Failure, Acute/immunology , T-Lymphocytes, Cytotoxic/immunology , fas Receptor
13.
J R Coll Physicians Lond ; 33(4): 359-64, 1999.
Article in English | MEDLINE | ID: mdl-10472025

ABSTRACT

Three schemes are presented for discussion whereby physicians undergoing postdoctoral training can combine a period of research training with their clinical training and so enable those who wish to follow a career in academic medicine to do so, or alternatively to revert to a clinical career. The training arrangements for those wishing to take up clinical academic medicine have hitherto been uncertain and hence unattractive to some. As well as encouraging more high-calibre trainees into academic medicine, the training programmes described are intended to bring greater clarity to those responsible for academic and clinical training and to those who fund research.


Subject(s)
Education, Medical, Graduate , Internal Medicine/education , Academic Medical Centers , Humans , Medical Staff, Hospital , Research , United Kingdom
14.
Hepatology ; 30(3): 761-9, 1999 Sep.
Article in English | MEDLINE | ID: mdl-10462383

ABSTRACT

Rat hepatic stellate cells (HSC) cultured in serum-containing medium underwent a rapid (3-hour) classical induction of p50:p65 and p65:p65 nuclear factor-kappaB (NF-kappaB) dimers. Subsequent culturing was associated with prolonged expression of active p50:p65 and persistent induction of a high-mobility NF-kappaB DNA binding complex consisting of potentially novel Rel-like protein(s). Formation of the latter complex was competed for by specific double-stranded oligonucleotides, was up-regulated by treatment of HSCs with tumor necrosis factor alpha (TNF-alpha), and was maintained at basal levels of expression by a soluble HSC-derived factor. An NF-kappaB-responsive CAT reporter gene was highly active in early cultured HSCs but was also trans-activated at a lower but significant level in longer-term cultured cells and could be completely suppressed by expression of dominant negative IkappaB-alpha. Physiological significance of the lower persistent NF-kappaB activities was also demonstrated by the ability of long-term cultured HSCs to support the activity of the NF-kappaB-dependent human intercellular adhesion molecule-1 (ICAM-1) promoter. Freshly isolated HSCs expressed high levels of IkappaB-alpha and IkappaB-beta. Culture activation was accompanied by a long-term reduction in levels of IkappaB-alpha with no detectable expression in the nuclear fraction of cells, under these conditions p50:p65 was detected in the nucleus. IkappaB-beta expression was transiently reduced and, upon replenishment, was associated with appearance of a lower-mobility IkappaB-beta antibody-reactive species. Bcl3 expression was absent in freshly isolated HSC but was induced during culturing and became a persistent feature of the activated HSC. Inhibition of NF-kappaB DNA binding activity by gliotoxin was associated with increased numbers of apoptotic cells. We suggest that activation of NF-kappaB in cultured HSC is required for expression of specific genes associated with the activated phenotype such as ICAM-1 and may be antiapoptotic for rat HSCs.


Subject(s)
Adipocytes/metabolism , DNA-Binding Proteins/biosynthesis , I-kappa B Proteins , Liver/metabolism , NF-kappa B/metabolism , Proto-Oncogene Proteins/biosynthesis , Transcription Factors/biosynthesis , Animals , Apoptosis/drug effects , Cells, Cultured , DNA/metabolism , Gliotoxin/pharmacology , Intercellular Adhesion Molecule-1/genetics , Liver/cytology , Male , NF-KappaB Inhibitor alpha , Rats , Rats, Sprague-Dawley , Transcription Factor RelB , Transcription, Genetic
15.
J Hepatol ; 30(5): 850-8, 1999 May.
Article in English | MEDLINE | ID: mdl-10365812

ABSTRACT

BACKGROUND/AIMS: Mast cell numbers are markedly increased in advanced liver fibrosis. Stem cell factor may recruit mast cells to the liver following injury as it induces mast cell proliferation, survival and differentiation from resident tissue precursors. This study examines stem cell factor production in human fibrotic liver and by hepatic stellate cells during culture in vitro. METHODS: Stem cell factor production was examined in human fibrotic livers by ELISA and in human and rat hepatic stellate cell cultures using reverse transcription-polymerase chain reaction (RT-PCR), Northern blotting, Western blotting and immunocytochemistry. Co-culture studies examined adhesion between hepatic stellate cells and purified mast cells. RESULTS: RT-PCR showed stem cell factor mRNA was more consistently expressed in fibrotic human livers relative to normal, and ELISA confirmed this by showing stem cell factor protein was significantly increased 2-3-fold in homogenates of human cirrhotic liver (primary biliary cirrhosis, primary sclerosing cholangitis) relative to normal. RT-PCR detected stem cell factor mRNA in human and rat hepatic stellate cells activated by culture on plastic. This was confirmed by Western blotting, which showed that freshly isolated hepatic stellate cells expressed relatively little 30 kD stem cell factor compared to late primary culture activated hepatic stellate cells (14 day) and passaged hepatic stellate cells. As assessed by fluorescence immunocytochemistry, stem cell factor protein was homogeneously expressed by populations of culture-activated rat hepatic stellate cells. During co-culture, purified human skin mast cells adhered to hepatic stellate cell monolayers on plastic, and this adherence was inhibited >50% by addition of antibodies against stem cell factor. CONCLUSIONS: Hepatic stellate cells activated in vitro produce stem cell factor. These cells may play an important role in recruiting mast cells to liver during injury and fibrosis.


Subject(s)
Liver Cirrhosis/physiopathology , Liver/cytology , Liver/physiology , Mast Cells/physiology , Stem Cell Factor/genetics , Transcription, Genetic , Animals , Cells, Cultured , Cholangitis, Sclerosing/genetics , Cholangitis, Sclerosing/metabolism , Cholangitis, Sclerosing/pathology , Humans , Liver/pathology , Liver Cirrhosis/pathology , Liver Cirrhosis, Biliary/genetics , Liver Cirrhosis, Biliary/metabolism , Liver Cirrhosis, Biliary/pathology , Male , Protein Biosynthesis , RNA, Messenger/genetics , Rats , Rats, Sprague-Dawley , Reverse Transcriptase Polymerase Chain Reaction
16.
Alcohol Clin Exp Res ; 23(5): 940-3, 1999 May.
Article in English | MEDLINE | ID: mdl-10371419

ABSTRACT

In liver fibrosis, activated hepatic stellate cells (HSC) play a major role in the deposition of excess extracellular matrix, including fibrillar collagens type I and type III. In addition to matrix protein synthesis, HSC regulate matrix degradation in the liver. This is mediated via a combination of synthesis of matrix (pro)metalloproteinases, which activate these zymogens via specific mechanisms and by inhibiting the active matrix-degrading enzymes via expression of tissue inhibitors of metalloproteinases (TIMPs). There are currently four members of the TIMP family described and of these, both TIMP-1 and TIMP-2 are synthesised by HSC. These observations have led to the suggestion that inhibition of matrix degradation mediated by a change in HSC-expression of TIMPs relative to metalloproteinases, such as interstitial collagenase, may contribute to progression of liver fibrosis. This hypothesis is supported by studies of human liver disease in which TIMP-1 expression is upregulated 5-fold in cirrhotic compared with normal liver. TIMP-1 and TIMP-2 expression is also upregulated in animal models of progressive fibrosis, whereas expression of collagenase is unchanged. In a model which is characterized by natural resolution of liver fibrosis, degradation of the deposited fibrillar liver matrix is accompanied by rapid down-regulation of TIMP-1 expression. In alcoholic liver disease, the role of TIMPs has not been studied exhaustively, but the evidence currently available supports a role for inhibition of matrix degradation by TIMPs in this progressive fibrotic liver disease.


Subject(s)
Liver Cirrhosis/physiopathology , Metalloendopeptidases/antagonists & inhibitors , Humans
17.
Hepatology ; 29(3): 839-48, 1999 Mar.
Article in English | MEDLINE | ID: mdl-10051488

ABSTRACT

In the injured liver hepatic stellate cells (HSCs) undergo a dramatic phenotypic transformation known as "activation" in which they become myofibroblast-like and express high levels of the tissue inhibitor of metalloproteinase 1 (TIMP-1). HSC activation is accompanied by transactivation of the TIMP-1 promoter. Truncation mutagenesis studies delineated a minimal active promoter consisting of nucleotides -102 to +60 relative to the major start site for transcription. Removal of an AP-1 site located at nucleotides -93 to -87 caused almost a complete loss of promoter activity. Analysis of AP-1 DNA binding activities during culture activation of HSCs initially indicated transient expression of proteins capable of forming a low mobility AP-1 DNA binding complex (LMAP-1). LMAP-1 was maximally induced at 24 hours of culture and then fell to undetectable levels at 120 hours. Western blot studies showed that both c-Fos and c-Jun underwent similar transient inductions. These temporal changes in c-Fos and c-Jun activities were unexpected because TIMP-1 mRNA expression is not detected in HSCs until culture day 3 to 5 and is thereafter sustained at a high level. Previous work in other cell lineages has established a key role for Pea3 binding proteins (Ets-1) in AP-1 mediated transactivation of the TIMP-1 promoter. We show that HSCs express relatively low levels Ets-1 and Ets-2 and show that mutagenesis of the Pea3 DNA binding site in the TIMP-1 promoter has less than a twofold effect on its activity in activated HSCs. Further analysis of AP-1 DNA binding activities in 7- to 14-day culture activated HSCs led to the discovery of high mobility AP-1 complexes (HMAP-1). HMAP-1 DNA binding activities were sequence specific with respect to AP-1 and absent from freshly isolated HSCs. Supershift EMSA and Western blot studies identified JunD, Fra2, and FosB as potential components of the HMAP-1. Mutations of the AP-1 site of the TIMP-1 promoter that prevented formation of HMAP-1 caused a 70% loss of activity in transfected activated HSCs. Taken together the data indicate that sustained upregulation of TIMP-1 gene expression may be at least partially controlled by a novel AP-1 dependent regulation of TIMP-1 promoter activity.


Subject(s)
DNA-Binding Proteins/physiology , Liver/physiology , Promoter Regions, Genetic/physiology , Tissue Inhibitor of Metalloproteinase-1/genetics , Transcription Factor AP-1/physiology , Animals , Base Sequence , Binding Sites/physiology , Cells, Cultured , DNA/metabolism , DNA-Binding Proteins/metabolism , Liver/cytology , Liver/metabolism , Molecular Sequence Data , Mutation/physiology , Protein Processing, Post-Translational/physiology , Proto-Oncogene Proteins c-fos/metabolism , Proto-Oncogene Proteins c-jun/metabolism , Rats , Rats, Sprague-Dawley , Transcription Factors/metabolism , Transcriptional Activation/physiology
18.
Hepatology ; 28(6): 1518-24, 1998 Dec.
Article in English | MEDLINE | ID: mdl-9828215

ABSTRACT

Activation of local tissue macrophages (Kupffer cells) and of quiescent hepatic stellate cells (HSCs) to a myofibroblast phenotype are two key events in liver inflammation and fibrosis. It is known that products of activated macrophages may activate stellate cells. We have hypothesized that the products of activated HSCs may also modulate the activity of Kupffer cells. The cytokine interleukin-10 (IL-10), produced by lymphocytes and macrophages, has profound inhibitory actions on macrophages. Normal rat and mouse HSCs that differentiate in vivo and in vitro to activated myofibroblasts were isolated by enzyme perfusion and density centrifugation with or without centrifugal elutriation, confirmed by vitamin A autofluorescence and positive immunostaining for the myofibroblast markers desmin and smooth muscle actin (SMA). Conditioned media and lysates from these cells were found to down-regulate lipopolysaccharide (LPS)-induced tumor necrosis factor- (TNF-) secretion by the mouse macrophage line RAW 267.4. In highly purified preparations of rat HSCs, messenger RNA (mRNA) for IL-10 was detected by reverse-transcription polymerase chain reaction (RT-PCR), from the time of isolation to up to 120 days of culture on plastic. Long-term cultures of unstimulated mouse HSCs secreted IL-10 protein as detected by immunoblotting and specific enzyme-linked immunosorbent assay (ELISA). IL-10 protein was undetectable by immunohistochemistry in mouse HSCs for the first 3 days in culture. After this, the percentage of IL-10-positive cells increased to 45% at day 7 and 100% by day 14, and expression of IL-10 continued in long-term cultures of up to 120 days. The expression of IL-10 by the stromal cells that govern the fibrotic process in the liver may have important implications for the regulation of inflammation and fibrosis in the liver.


Subject(s)
Interleukin-10/physiology , Liver/metabolism , Macrophages/physiology , Animals , Base Sequence , Immunohistochemistry , Interleukin-10/genetics , Interleukin-10/metabolism , Kupffer Cells/metabolism , Liver/cytology , Macrophages/metabolism , Mice , Mice, Inbred Strains , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Tumor Necrosis Factor-alpha/metabolism
19.
J Gastroenterol Hepatol ; 13 Suppl: S33-8, 1998 Sep.
Article in English | MEDLINE | ID: mdl-9792032

ABSTRACT

Hepatic stellate cells (HSC) play a central role in the pathogenesis of liver fibrosis. Following liver injury, these cells proliferate and are activated to a profibrogenic myofibroblastic phenotype. In addition to increased matrix protein synthesis, there is evidence to indicate that these cells are able to regulate matrix degradation. In the early phases of their cellular activation, HSC release matrix metalloproteinases with the ability to degrade the normal liver matrix. When HSC are fully activated, there is a net down-regulation of matrix degradation mediated by increased synthesis and extracellular release of tissue inhibitors of metalloproteinase (TIMP)-1 and -2. These studies in cell culture have been complemented by in vivo studies of hepatic TIMP-1 and TIMP-2 gene expression. In advanced human liver disease of various aetiologies, there is increased TIMP-1-mRNA and protein and increased TIMP-2-mRNA in fibrotic liver compared with control liver. Temporal studies of progressive rat liver fibrosis caused by bile duct ligation or by carbon tetrachloride, indicate an important role for increased TIMP-1 and TIMP-2 expression in pathogenesis. Moreover, in a rat model of reversible liver fibrosis, matrix remodelling and resolution of liver fibrosis is closely associated, temporally, with a marked decrease in TIMP-1 and TIMP-2 expression. These combined cell culture and in vivo findings have led us to investigate the mechanisms of regulation of TIMP-1 gene expression in hepatic stellate cells. Our recent data indicate that transcriptional regulation of TIMP-1 gene expression in HSC is mediated via a mechanism which differs considerably from that previously identified in skin fibroblasts. We conclude that increased TIMP-1 and TIMP-2 expression by HSC plays an important role in the pathogenesis of liver fibrosis. This may represent an important therapeutic target in the design of anti-fibrotic strategies for chronic liver disease.


Subject(s)
Liver Cirrhosis, Experimental/etiology , Liver/cytology , Metalloendopeptidases/antagonists & inhibitors , Protease Inhibitors/metabolism , Tissue Inhibitor of Metalloproteinase-1/metabolism , Animals , Cells, Cultured , Humans , Liver/metabolism , Metalloendopeptidases/metabolism , Rats , Tissue Inhibitor of Metalloproteinase-2/metabolism
20.
J Clin Invest ; 102(3): 538-49, 1998 Aug 01.
Article in English | MEDLINE | ID: mdl-9691091

ABSTRACT

Liver fibrosis results from the excessive secretion of matrix proteins by hepatic stellate cells (HSC), which proliferate during fibrotic liver injury. We have studied a model of spontaneous recovery from liver fibrosis to determine the biological mechanisms mediating resolution. Livers were harvested from rats at 0, 3, 7, and 28 d of spontaneous recovery from liver fibrosis induced by 4 wk of twice weekly intraperitoneal injections with CCl4. Hydroxyproline analysis and histology of liver sections indicated that the advanced septal fibrosis observed at time 0 (peak fibrosis) was remodeled over 28 d of recovery to levels close to control (untreated liver). alpha-Smooth muscle actin staining of liver sections demonstrated a 12-fold reduction in the number of activated HSC over the same time period with evidence of HSC apoptosis. Ribonuclease protection analysis of liver RNA extracted at each recovery time point demonstrated a rapid decrease in expression of the collagenase inhibitors TIMP-1 and TIMP-2, whereas collagenase mRNA expression remained at levels comparable to peak fibrosis. Collagenase activity in liver homogenates increased through recovery. We suggest that apoptosis of activated HSC may vitally contribute to resolution of fibrosis by acting as a mechanism for removing the cell population responsible for both producing fibrotic neomatrix and protecting this matrix from degradation via their production of TIMPs.


Subject(s)
Apoptosis , Liver Cirrhosis, Experimental/pathology , Tissue Inhibitor of Metalloproteinase-1/biosynthesis , Tissue Inhibitor of Metalloproteinase-2/biosynthesis , Animals , Carbon Tetrachloride/toxicity , Chemical and Drug Induced Liver Injury/complications , Collagenases/biosynthesis , Collagenases/genetics , Gene Expression Regulation , Hydroxyproline/analysis , Liver Cirrhosis, Experimental/enzymology , Male , Matrix Metalloproteinase 1 , Matrix Metalloproteinase 13 , Matrix Metalloproteinase Inhibitors , Procollagen/biosynthesis , Procollagen/genetics , RNA, Messenger/biosynthesis , Rats , Rats, Sprague-Dawley , Remission, Spontaneous , Tissue Inhibitor of Metalloproteinase-1/genetics , Tissue Inhibitor of Metalloproteinase-2/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...