Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










Publication year range
1.
Radiat Environ Biophys ; 47(3): 389-97, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18493784

ABSTRACT

In a previous publication, we were able to show that irradiation of Kupffer cells, the liver resident macrophages, leads to an increased TNF-alpha concentration in the culture medium. The pathomechanisms underlying this phenomenon, however, remained to be elucidated. Here, we show that following irradiation of Kupffer cells, the apoptosis rate increased drastically within 48 h. At the same time, the total TNF-alpha concentration in cell lysates of Kupffer cells attached to the culture plate decreased. However, normalization of the TNF-alpha concentration with respect to cell number revealed that TNF-alpha concentration per attached cell remained constant during the observation period. Western blot analysis showed that heat shock protein 27 (Hsp27) is strongly downregulated and bax is upregulated in irradiated Kupffer cells as compared to sham-irradiated cells. Overexpression of Hsp27 in Kupffer cells was shown to prevent the effect of irradiation on bax expression, apoptosis and, at the same time, on increase of TNF-alpha concentration in the Kupffer cell medium. We conclude that irradiation of Kupffer cells leads to apoptosis because of downregulation of Hsp27 and consecutive upregulation of bax expression. Furthermore, we suggest that apoptosis of Kupffer cells leads to an increase of TNF-alpha concentration in the culture medium which may be due to cell death rather than active release or synthesis.


Subject(s)
Apoptosis/radiation effects , Heat-Shock Proteins/physiology , Kupffer Cells/radiation effects , Neoplasm Proteins/physiology , Tumor Necrosis Factor-alpha/metabolism , Animals , Caspases/metabolism , Cell Survival/radiation effects , Cells, Cultured , HSP27 Heat-Shock Proteins , Heat-Shock Proteins/analysis , Male , Neoplasm Proteins/analysis , Rats , Rats, Wistar , bcl-2-Associated X Protein/analysis
2.
Z Gastroenterol ; 45(1): 77-86, 2007 Jan.
Article in English | MEDLINE | ID: mdl-17236124

ABSTRACT

Liver fibrosis is defined as an excessive deposition of extracellular matrix. It is the main complication of chronic liver damage and its endpoint, the liver cirrhosis, is responsible for impressive morbidity and mortality. The accumulation of extracellular matrix proteins in liver fibrosis and cirrhosis is due to different cell types which acquire a myofibroblastic phenotype--the hepatic stellate cells, located in the space of Disse, portal fibroblasts as well as myofibroblasts of the portal and pericentral areas. Further studies also suggest an impressive role of bone marrow-derived myofibroblasts. Differences have been reported between the two cell populations with respect to myofibroblastic differentiation, activation and "deactivation", proliferation and apoptosis. However, in most cases additional confirmation may be required; thus the biological and biochemical characterization of these cells, their interactions with inflammatory cells and the cytokine environment leading to their activation or cell death are essential to understand the mechanisms underlying the progressive development of excessive scarring in the liver as well as the ability of the liver for tissue repair and regeneration. All this information is required to estimate the value of already suggested possible treatments to specifically and efficiently target the cells responsible for the development of liver fibrosis/cirrhosis and as well as for liver regeneration.


Subject(s)
Cytokines/immunology , Hepatitis/immunology , Hepatitis/pathology , Immunologic Factors/immunology , Liver Cirrhosis/immunology , Liver Cirrhosis/pathology , Liver Regeneration/immunology , Animals , Cell Communication/immunology , Hepatocytes/immunology , Humans , Models, Immunological , T-Lymphocytes/immunology
4.
Anat Embryol (Berl) ; 208(5): 359-66, 2004 Aug.
Article in English | MEDLINE | ID: mdl-15232737

ABSTRACT

Prox1 is a transcription factor with two highly conserved domains, a homeobox and a prospero domain. It has been shown that Prox1 knock-out mice die during early embryonic stages and display a rudimentary liver. We have studied the expression of Prox1 at RNA and protein levels in chick, rat, mouse and human liver and in transformed and non-transformed hepatic cell lines. Prox1 is expressed in early embryonic hepatoblasts and is still expressed in adult hepatocytes. Prox1 protein is located in the nuclei of hepatoblasts, which grow into the neighboring embryonic mesenchyme. The expression pattern in chick, mouse, rat and human embryos is highly conserved. Besides albumin and alpha-fetal protein, Prox1 belongs to the earliest markers of the developing liver. In adult liver, Prox1 is expressed in hepatocytes but is absent from bile duct epithelial and non-parenchymal cells (Kupffer cells, hepatic stellate cells, sinusoidal endothelial cells and myofibroblasts). Isolated primary hepatocytes and hepatoma cell lines (HepG2, Hep3B) are Prox1 positive, whereas the immortalized murine liver cell-line MMH, which constitutively expresses the receptor c-met, is Prox1 negative. Transfection of MMH with Prox1 cDNA increases the expression level significantly as compared to control transfectants. In HepG2 and Hep3B, the Prox1 levels are even up to 100 times higher. Our studies show that Prox1 is a highly conserved transcription factor, expressed in hepatocytes from the earliest stages of development into adulthood and over-expressed in hepatoma cell lines. Its absence from bile duct epithelial cells suggests a function for the specification of hepatoblasts into hepatocytes. The genes controlled by Prox1 need to be studied in the future.


Subject(s)
Bile Ducts/embryology , Bile Ducts/metabolism , Hepatocytes/metabolism , Homeodomain Proteins/metabolism , Liver/embryology , Liver/metabolism , Animals , Bile Ducts/cytology , Cell Line , Chick Embryo , DNA, Complementary/genetics , Epithelial Cells/cytology , Epithelial Cells/metabolism , Gene Expression Regulation, Developmental/genetics , Hepatocytes/cytology , Homeodomain Proteins/genetics , Humans , Liver/cytology , Mice , Mice, Inbred C57BL , Proto-Oncogene Proteins c-met/metabolism , RNA, Messenger/metabolism , Rats , Rats, Inbred F344 , Rats, Wistar , Stem Cells/cytology , Stem Cells/metabolism , Transfection , Tumor Suppressor Proteins , Up-Regulation/genetics
5.
Liver ; 22(4): 283-94, 2002 Aug.
Article in English | MEDLINE | ID: mdl-12296961

ABSTRACT

The wall of the liver sinusoid is made of highly specialized cells, the hepatic stellate cells (HSC) which together with the sinusoidal endothelial cells represent a loose barrier to the corpusculate part of the blood flowing through the liver. Quiescent stellate cells (quiescent HSC) store Vitamin A; "activated" stellate cells become involved in the reaction to acute or chronic noxae damaging the liver parenchyma. Activated HSC show increased protein synthesis capacity, increased DNA-synthesis and acquire a myofibroblast-like phenotype. Under similar conditions liver myofibroblasts (MF) of the portal field and of the pericentral area may also become "activated" by increasing protein synthesis, DNA synthesis and cell division. They express the fibulin-2 gene and produce large amounts of IL-6. In contrast to "activated" HSC they do not undergo spontaneous apoptosis in vitro and do not express the CD95-ligand gene. So far no definite prove has been found for a "transdifferentiation" of HSC to myofibroblasts. On the contrary an increasing amount of data support the conviction that HSC and MF represent two similar but not identical cell populations the latter being comparable to those of other organs.


Subject(s)
Endothelium, Vascular/cytology , Kupffer Cells/cytology , Liver/cytology , Mesoderm/cytology , Animals , Endothelium, Vascular/immunology , Endothelium, Vascular/metabolism , Fibroblasts/cytology , Fibroblasts/immunology , Fibroblasts/metabolism , Humans , Kupffer Cells/immunology , Kupffer Cells/metabolism , Liver/immunology , Liver/metabolism , Macrophage Activation , Mesoderm/immunology , Mesoderm/metabolism , Rats
6.
Eur J Cell Biol ; 80(8): 554-61, 2001 Aug.
Article in English | MEDLINE | ID: mdl-11561906

ABSTRACT

Activated hepatic stellate cells (HSC) are thought to play a pivotal role in development of liver fibrosis which takes place in chronic liver diseases. Previous studies have shown that "activated" rat HSC undergo spontaneous apoptosis probably through the CD95/CD95L pathway. TGF-beta as well as TNF-alpha reduced spontaneous apoptosis and CD95L expression. The aim of this study was to investigate the possible mechanisms responsible for the spontaneous apoptosis and for the anti-apoptotic effect of TGF-beta and TNF-alpha on activated HSC. While bcl-2, bax, NFkappaB and p53 gene expression were spontaneously upregulated, bcl-xL and p21WAF1 gene expression decreased and IkappaB remained unchanged during the activation process in vitro. TGF-beta as well as TNF-alpha induced activation of NFKB and upregulated bcl-xL. The latter was inhibited by overexpression of IkappaB. By suppressing spontaneous apoptosis TGF-beta as well as TNF-alpha inhibited p53 gene expression while that of the p21WAF1 gene was increased. We conclude that TGF-beta as well as TNF-alpha may act as surviving factors for activated rat HSC not only through reduction of CD95L gene expression but also by upregulating the anti-apoptotic factors NFKB, bcl-xL and p21WAF1 and by downregulating the proapoptotic factor p53. The interaction with these factors may lead to the generation of new antifibrotic drugs.


Subject(s)
Apoptosis , Cyclins/genetics , Genes, bcl-2 , Genes, p53 , NF-kappa B/genetics , Proto-Oncogene Proteins c-bcl-2 , Transforming Growth Factor beta/pharmacology , Tumor Necrosis Factor-alpha/pharmacology , Animals , Cells, Cultured , Cyclin-Dependent Kinase Inhibitor p21 , Down-Regulation , Gene Expression Regulation , Liver/metabolism , Proto-Oncogene Proteins/genetics , Rats , Rats, Wistar , Transfection , Up-Regulation , bcl-2-Associated X Protein
7.
Can J Gastroenterol ; 15(3): 187-93, 2001 Mar.
Article in English | MEDLINE | ID: mdl-11264572

ABSTRACT

Liver fibrosis represents the uniform response of liver to toxic, infectious or metabolic agents. The process leading to liver fibrosis resembles the process of wound healing, including the three phases following tissue injury: inflammation, synthesis of collagenous and noncollagenous extracellular matrix components, and tissue remodelling (scar formation). While a single liver tissue injury can be followed by an almost complete restitution ad integrum, the persistence of the original damaging noxa results in tissue damage. During the establishment of liver fibrosis, the basement membrane components collagen type IV, entactin and laminin increase and form a basement membrane-like structure within the space of Disse. The number of endothelial fenestrae of the sinusoids decreases. These changes of the sinusoids are called 'capillarization' because the altered structure of the sinusoids resembles that of capillaries. At the cellular level, origin of liver fibrogenesis is initiated by the damage of hepatocytes, resulting in the recruitment of inflammatory cells and platelets, and activation of Kupffer cells, with subsequent release of cytokines and growth factors. The hepatic stellate cells seem to be the primary target cells for these inflammatory stimuli, because during fibrogenesis, they undergo an activation process to a myofibroblast-like cell, which represents the major matrix-producing cell. Based on this pathophysiological mechanism, therapeutic methods are developed to inhibit matrix synthesis or stimulate matrix degradation. A number of substances are currently being tested that either neutralize fibrogenic stimuli and prevent the activation of hepatic stellate cells, or directly modulate the matrix metabolism. However, until now, the elimination of the hepatotoxins has been the sole therapeutic concept available for the treatment of liver fibrogenesis in humans.


Subject(s)
Extracellular Matrix/metabolism , Hepatitis/metabolism , Liver Cirrhosis/metabolism , Acute Disease , Chronic Disease , Hepatitis/complications , Humans , Liver/injuries , Liver Cirrhosis/etiology , Wound Healing
8.
Immunol Lett ; 74(2): 153-64, 2000 Oct 03.
Article in English | MEDLINE | ID: mdl-10996391

ABSTRACT

UNLABELLED: Platelet-endothelial cell adhesion molecule (PECAM-1), a member of the Ig superfamily is strongly expressed at endothelial cell-cell junctions, on most leukocytes and on monocytes. PECAM-1 has been implicated as a key mediator of the transendothelial migration of leukocytes and monocytes. To further define the physiological role of PECAM-1, we studied the modulation of PECAM-1-expression in a model of liver inflammation in both mononuclear cells (MCs) and sinusoidal endothelial cells (SECs). In normal rat liver sections, PECAM-1 immunohistology indicated a sinusoidal pattern similar to the ICAM-1 staining. Both, SECs, small and large MCs isolated from control rats express PECAM-1 as demonstrated by immunocytochemistry, flow cytometry, and Northern blot analysis. Immunohistochemical studies on liver sections from CCl(4)-treated animals indicated, that in the areas of necrosis 24-48 h after a single administration of the toxin, there was an accumulation of LFA-1-, ED1- (marker for rat monocytes) and ICAM-1-positive, but ED2-(marker for tissue macrophages)-negative inflammatory cells. Most of these cells were PECAM-1-negative. In situ hybridization indicated that there is no accumulation of PECAM-1 specific transcripts after CCl(4) treatment within the pericentral region. Immunocytology, flow cytometry, and Northern blot analysis of MCs and SECs isolated at different times after the administration of CCl(4) revealed a decrease of PECAM-1 gene expression in MCs and in SECs, whereas ICAM-1 expression increased. As TNFalpha has been shown to be upregulated early after CCl(4) administration, the influence of TNFalpha on PECAM-gene-expression was analyzed. TNFalpha treatment of cultured rat SECs and of small and large MCs from normal liver decreased PECAM-1 specific transcript level in parallel to the increase of ICAM-1 transcript level. CONCLUSIONS: Early production of TNFalpha after liver injury could induce an increased ICAM-1-expression and a decreased PECAM-1 expression, which might be essential for the transmigration of inflammatory cells into the parenchyma.


Subject(s)
Chemical and Drug Induced Liver Injury/metabolism , Endothelium/metabolism , Leukocytes, Mononuclear/metabolism , Platelet Endothelial Cell Adhesion Molecule-1/physiology , Tumor Necrosis Factor-alpha/pharmacology , Animals , Carbon Tetrachloride , Cell Culture Techniques , Endothelium/cytology , Endothelium/drug effects , Gene Expression , Immunohistochemistry , In Situ Hybridization , Intercellular Adhesion Molecule-1/genetics , Intercellular Adhesion Molecule-1/metabolism , Kupffer Cells/drug effects , Kupffer Cells/metabolism , Leukocytes, Mononuclear/drug effects , Lymphocyte Function-Associated Antigen-1/metabolism , Male , Platelet Endothelial Cell Adhesion Molecule-1/genetics , RNA, Messenger/analysis , Rats , Rats, Wistar
9.
Histochem Cell Biol ; 113(6): 443-53, 2000 Jun.
Article in English | MEDLINE | ID: mdl-10933221

ABSTRACT

Matrix metalloproteinases (MMPs) and their specific inhibitors (TIMPs) are thought to play an essential role in liver injury associated with tissue remodeling. However, their distinct expression profile in different liver repair models still remains to be established. Hepatic expression of collagenase (MMP-13), gelatinases A and B (MMP-2, -9), stromelysin-1 and -2 (MMP-3, -10), membrane-type MMP-1 (MMP-14), and TIMP-1 and -2 was studied following single and repeated CCl4-mediated injury and after partial hepatectomy. Expression was analyzed by reverse transcription-PCR (RT-PCR), northern blot analysis, zymography, and immunohistochemistry. Following a single toxic liver injury, MMPs and TIMPs were induced in a distinct time frame in that expression of most MMPs was induced during the early phase of liver injury, was maximal during the inflammatory reaction, and was diminished in the recovery phase. In contrast, TIMP and MMP-2 steady state mRNA levels remained constant in the early phase, were strongly induced during tissue inflammation, and remained increased until the recovery phase. Interestingly, hepatic TNF-alpha expression paralleled the MMP induction profile, while the increase of TGF-beta1 expression mapped to the increase of TIMPs. Chronic liver injury was accompanied by an increase in the steady state mRNA levels of MMP-2 and TIMPs, while other MMPs remained more or less unchanged or were diminished. Partial hepatectomy was followed by a dramatic increase of MMP-14 and to a lesser extent also of TIMP-1 expression; other MMPs and TIMPs were not significantly induced. Liver injury is accompanied by profound changes in hepatic MMP/TIMP expression, the latter being critically dependent on the type of injury. Single toxic injury resulting in complete restoration was characterized by a sequential induction of MMPs and TIMPs suggesting initial matrix breakdown and matrix restoration thereafter. Chronic liver injury leading to fibrosis displays overall diminished matrix degradation mainly through TIMP induction, while liver regeneration induced by partial hepatectomy caused an induction of MMP-14 and TIMP-1 only, which might be unrelated to matrix turnover but connected to pericellular fibrinolysis or fibrolysis required for hepatocellular replication.


Subject(s)
Collagenases/genetics , Liver Regeneration/physiology , Liver/enzymology , Matrix Metalloproteinase 2/genetics , Tissue Inhibitor of Metalloproteinase-1/genetics , Acute Disease , Animals , Blotting, Northern , Collagenases/analysis , Gene Expression Regulation, Enzymologic/physiology , Hepatectomy , Hepatitis, Animal/metabolism , Immunohistochemistry , Liver/surgery , Liver Cirrhosis/metabolism , Matrix Metalloproteinase 1/analysis , Matrix Metalloproteinase 1/genetics , Matrix Metalloproteinase 10 , Matrix Metalloproteinase 13 , Matrix Metalloproteinase 2/analysis , Matrix Metalloproteinase 3/analysis , Matrix Metalloproteinase 3/genetics , Matrix Metalloproteinase 9/analysis , Matrix Metalloproteinase 9/genetics , Metalloendopeptidases/analysis , Metalloendopeptidases/genetics , RNA, Messenger/analysis , Rats , Rats, Wistar , Tissue Inhibitor of Metalloproteinase-1/analysis , Tissue Inhibitor of Metalloproteinase-2/analysis , Tissue Inhibitor of Metalloproteinase-2/genetics
10.
Am J Pathol ; 155(6): 1841-8, 1999 Dec.
Article in English | MEDLINE | ID: mdl-10595913

ABSTRACT

During liver tissue repair, hepatic stellate cells (HSCs), a pericyte-like nonparenchymal liver cell population, transform from a quiescent status (resting HSCs) into myofibroblast like cells (activated HSCs); the latter is the principal matrix-synthesizing cell of the liver. Although several factors have been shown to be involved in this important process, the molecular mechanisms regulating HSC activation are still under investigation. To identify key regulatory proteins involved in the HSC activation process, we used different mRNA display technologies, with cDNAs prepared from HSCs at different stages of in vitro activation. With the latter technique, the transcription factor Ets-1 was detected through its down-regulation during activation. As confirmed by Northern blot and reverse transcriptase-polymerase chain reaction (RT-PCR) analysis, mRNAs coding for Ets-1 were present in the highest amounts in freshly isolated HSCs and in HSCs 2 days after plating (classified as resting HSCs/early activated HSCs) and were diminished in HSCs 7 days after plating (activated cells). Ets-1 protein was present in HSC-lysates, as assessed by Western blot, and bound to an oligonucleotide containing the Ets-1 consensus cis-acting motif, as demonstrated by electrophoretic mobility shift assay. Ets-1 binding activity peaked in nuclear extracts prepared from resting/early activated cells and was diminished in extracts derived from fully activated cells. In contrast, binding activity of the transcription factors TFIID, AP-1, and SP-1 was highest in activated HSCs and only barely detectable in resting/early activated HSCs. By Northern blot and RT-PCR analysis, Ets-1-specific transcripts were present in parenchymal and other nonparenchymal liver cells too, illustrating that hepatic Ets-1 expression is not specific or restricted to HSCs. However, the unique pattern of Ets-1 binding activity present in resting versus activated HSCs and its known implications for cellular differentiation and tissue remodeling suggest that Ets-1 could be of crucial importance for HSC activation and hepatic tissue repair.


Subject(s)
Gene Expression/physiology , Liver/cytology , Liver/metabolism , Proto-Oncogene Proteins/physiology , Transcription Factors/physiology , Animals , Blotting, Northern , Blotting, Western , Cell Differentiation , Cell Division , Cell Line , DNA, Complementary/analysis , Down-Regulation , Nuclear Proteins/metabolism , Protein Binding , Proto-Oncogene Protein c-ets-1 , Proto-Oncogene Proteins/biosynthesis , Proto-Oncogene Proteins c-ets , RNA, Messenger/analysis , Rats , Rats, Wistar , Reverse Transcriptase Polymerase Chain Reaction , Sequence Analysis, DNA , Transcription Factors/biosynthesis
11.
Histochem Cell Biol ; 112(5): 387-401, 1999 Nov.
Article in English | MEDLINE | ID: mdl-10603079

ABSTRACT

Previous in vitro studies indicated that hepatic stellate cells (HSC) and rat liver myofibroblasts (rMF) have to be regarded as different cell populations of the myofibroblastic lineage with fibrogenic potential. Employing the discrimination features defined by these studies the localization of HSC and rMF was analyzed in diseased livers. Normal and acutely as well as chronically carbon tetrachloride-injured livers were analyzed by immunohistochemistry and by in situ hybridization. In normal livers HSC [desmin/glial fibrillary acid protein (GFAP)-positive cells] were distributed in the hepatic parenchyma, while rMF (desmin/smooth muscle alpha actin-positive, GFAP-negative cells colocalized with fibulin-2) were located in the portal field, the walls of central veins, and only occasionally in the parenchyma. Acute liver injury was characterized almost exclusively by an increase in the number of HSC, while the amount of rMF was nearly unchanged. In early stages of fibrosis, HSC and rMF were detected within the developing scars. In advanced stages of fibrosis, HSC were mainly present at the scar-parenchymal interface, while rMF accounted for the majority of the cells located within the scar. At every stage of fibrogenesis, rMF, in contrast to HSC, were only occasionally detected in the hepatic parenchyma. HSC and rMF are present in normal and diseased livers in distinct compartments and respond differentially to tissue injury. Acute liver injury is followed by an almost exclusive increase in the number of HSC, while in chronically injured livers not only HSC but also rMF are involved in scar formation.


Subject(s)
Chemical and Drug Induced Liver Injury/pathology , Liver Regeneration/physiology , Liver/cytology , Liver/pathology , Actins/metabolism , Acute Disease , Animals , Carbon Tetrachloride Poisoning/pathology , Chronic Disease , Desmin/metabolism , Fibroblasts , Fluorescent Antibody Technique , Glial Fibrillary Acidic Protein/metabolism , Immunohistochemistry , In Situ Hybridization , Interleukin-6/metabolism , Liver/ultrastructure , Rats , Rats, Wistar
12.
J Hepatol ; 31(4): 692-702, 1999 Oct.
Article in English | MEDLINE | ID: mdl-10551394

ABSTRACT

BACKGROUND/AIMS: It is suggested that during fibrogenesis as well as during carcinogenesis of the liver, the hepatic microvascular phenotype is transformed from sinusoids - which lack a basement membrane--into continuous capillaries which rest on a basement membrane. As transforming growth factor (TGF)-beta1 seems to be the most effective mediator in the stimulation of matrix protein synthesis, we were interested in the modulation of basement membrane proteins collagen type IV, laminin, and entactin expression by TGF-beta1 in liver sinusoidal endothelial cells (SECs), especially since a stimulation of the synthesis of collagen type IV but not of entactin and laminin by TGF-beta1 has been demonstrated in a fibrosarcoma cell line. METHODS: The synthesis of the basement membrane (BM) proteins entactin, laminin, and collagen type IV and of the extracellular matrix (ECM) proteins tenascin and fibronectin with or without TGF-beta1--stimulation was analyzed by immunostaining, immunoprecipitation of endogenously labeled proteins and Northern blot analysis of total RNA extracted from freshly isolated or cultured SECs from rat or guinea pig livers. Furthermore, SECs were isolated from acutely and chronically CCl4-damaged rat livers and were analyzed for matrix protein expression. RESULTS: SECs were adherent 24 h after isolation and formed confluent monolayers on day 4 of primary culture. Specific immunoprecipitates and specific transcripts for the BM proteins entactin, laminin, and collagen type IV and for ECM proteins tenascin and fibronectin were detectable in freshly isolated or cultured SECs. The synthesis of all tested BM proteins and ECM proteins was stimulated at least 3-fold by TGF-beta1. In SECs isolated after CCl4-induced acute and chronic liver damage, increased levels of matrix protein transcripts were detectable. CONCLUSIONS: The stimulation of the synthesis of all BM-proteins by TGF-beta1 in vitro and the accumulation of ECM transcripts in SECs isolated from CCl4-treated livers, suggests that SECs are involved in the formation of a basement membrane during the "capillarization" of the sinusoids during liver disease.


Subject(s)
Collagen/metabolism , Endothelium, Vascular/metabolism , Laminin/metabolism , Liver Circulation/physiology , Membrane Glycoproteins/metabolism , Transforming Growth Factor beta/pharmacology , Animals , Basement Membrane/metabolism , Blood Vessels/cytology , Blood Vessels/drug effects , Blood Vessels/metabolism , Blotting, Northern , Carbon Tetrachloride/pharmacology , Cells, Cultured , Collagen/genetics , Endothelium, Vascular/cytology , Extracellular Matrix Proteins/genetics , Extracellular Matrix Proteins/metabolism , Gene Expression , Guinea Pigs , Immunologic Techniques , Laminin/genetics , Male , Membrane Glycoproteins/genetics , RNA, Messenger/metabolism , Rats , Rats, Wistar
13.
Gastroenterology ; 117(5): 1205-21, 1999 Nov.
Article in English | MEDLINE | ID: mdl-10535885

ABSTRACT

BACKGROUND & AIMS: Hepatic stellate cells (HSCs) are considered the principal matrix-producing cells of the damaged liver. However, other cell types of the fibroblast lineage that have not yet been characterized are also involved in liver tissue repair and fibrogenesis. METHODS: We established cultures of cells of the fibroblast lineage, termed rat liver myofibroblasts, and analyzed their phenotypical and functional properties in comparison with HSCs. RESULTS: HSCs and rat liver myofibroblasts were discernible by morphological criteria and growth behavior. Prolonged subcultivation of rat liver myofibroblasts was achieved, but HSCs were maintained in culture at maximum until second passage. HSCs were characterized by expression of glial fibrillary acidic protein, desmin, and vascular cell adhesion molecule 1, which were almost completely absent in rat liver myofibroblasts. For synthetic properties, HSCs and rat liver myofibroblasts displayed mostly overlapping properties with 4 striking differences. The complement-activating protease P100 and the protease inhibitor alpha(2)-macroglobulin were preferentially expressed by HSCs, whereas interleukin 6-coding messenger RNAs and the extracellular matrix protein fibulin 2 were almost exclusively detectable in rat liver myofibroblasts. CONCLUSIONS: The data show that morphologically and functionally different fibroblastic populations, HSCs and rat liver myofibroblasts, can be derived from liver tissue. HSCs may not represent the single matrix-producing cell type of the fibroblast lineage in the liver.


Subject(s)
Fibroblasts/cytology , Fibroblasts/physiology , Liver Cirrhosis, Experimental/etiology , Liver/cytology , Muscle, Smooth/cytology , Animals , Biomarkers , Calcium-Binding Proteins/metabolism , Cell Differentiation/physiology , Cell Line , Cytokines/metabolism , Cytoskeletal Proteins/metabolism , Extracellular Matrix Proteins/metabolism , Fibroblasts/metabolism , Liver/metabolism , Muscle, Smooth/metabolism , Rats , Rats, Wistar , Receptors, Cell Surface/metabolism
14.
Hepatology ; 30(1): 196-202, 1999 Jul.
Article in English | MEDLINE | ID: mdl-10385656

ABSTRACT

Transforming growth factor beta (TGF-beta) as well as tumor necrosis factor alpha (TNF-alpha) gene expression are up-regulated in chronically inflamed liver. These cytokines were investigated for their influence on apoptosis and proliferation of activated hepatic stellate cells (HSCs). Spontaneous apoptosis in activated HSC was significantly down-regulated by 53% +/- 8% (P <.01) under the influence of TGF-beta and by 28% +/- 2% (P <.05) under the influence of TNF-alpha. TGF-beta and TNF-alpha significantly reduced expression of CD95L in activated HSCs, whereas CD95 expression remained unchanged. Furthermore, HSC apoptosis induced by CD95-agonistic antibodies was reduced from 96% +/- 2% to 51 +/- 7% (P <.01) by TGF-beta, and from 96% +/- 2% to 58 +/- 2% (P <.01) by TNF-alpha, suggesting that intracellular antiapoptotic mechanisms may also be activated by both cytokines. During activation, HSC cultures showed a reduced portion of cells in the G0/G1 phase and a strong increment of G2-phase cells. This increment was significantly inhibited (G1 arrest) by administration of TGF-beta and/or TNF-alpha to activated cells. In liver sections of chronically damaged rat liver (CCl4 model), using desmin and CD95L as markers for activated HSC, most of these cells did not show apoptotic signs (TUNEL-negative). Taken together, these findings indicate that TGF-beta and/or TNF-alpha both inhibit proliferation and also apoptosis in activated HSC in vitro. Both processes seem to be linked to each other, and their inhibition could represent the mechanism responsible for prolonged survival of activated HSC in chronic liver damage in vivo.


Subject(s)
Apoptosis/physiology , Liver/cytology , Liver/physiology , Transforming Growth Factor beta/pharmacology , Tumor Necrosis Factor-alpha/pharmacology , Animals , Apoptosis/drug effects , Carbon Tetrachloride Poisoning/pathology , Carbon Tetrachloride Poisoning/physiopathology , Cell Cycle/drug effects , Cell Division/drug effects , Cells, Cultured , Fas Ligand Protein , Gene Expression Regulation , Kinetics , Liver/drug effects , Membrane Glycoproteins/genetics , Rats , Rats, Wistar , Time Factors , fas Receptor/genetics
15.
J Hepatol ; 30(1): 48-60, 1999 Jan.
Article in English | MEDLINE | ID: mdl-9927150

ABSTRACT

BACKGROUND/AIMS: Although matrix metalloproteinases (MMPs) and their specific inhibitors (TIMPs) play an essential role in liver injury associated with tissue remodeling, the cellular origin of MMPs/TMPs within the liver remains to be clarified. METHODS: Different liver cell populations were analysed with respect to their expression by reverse transcription-polymerase chain reaction, Northern blot analysis and zymography. RESULTS: MMP and TIMP coding transcripts were detectable in all liver cell types by reverse transcription-polymerase chain reaction; however, the cellular expression levels were markedly different as assessed by Northern blot analysis. Gelatinase-B was predominantly expressed in Kupffer cells, gelatinase-A in hepatic stellate cells and rat liver myofibroblasts and stromelysins-1, -2 as well as collagenase in hepatic stellate cells. Membrane type-1 MMP (MMP-14) was found in significant amounts in all liver cells. TIMP-1 coding m-RNAs were present mainly in hepatic stellate cells and rat liver myofibroblasts, TIMP-2 additionally in Kupffer cells, while TIMP-3 expression was detectable only in hepatocytes. During in vitro activation of hepatic stellate cells, MMP expression was mostly downregulated, while TIMP expression was enhanced, thereby providing an explanation for matrix accumulation co-localised with these cells during chronic liver injury. In general, TNF-alpha stimulated both MMP and TIMP expression of hepatic stellate cells, while TGF-beta1 induced TIMP expression only. CONCLUSIONS: Collectively these data demonstrate that all resident liver cells are involved in matrix degradation to some extent and that hepatic stellate cells play an important role in matrix breakdown in addition to matrix synthesis. The cytokine-specific regulation of MMP/TIMP expression in hepatic stellate cells suggests that the initial matrix breakdown following liver injury might be enhanced by TNF-alpha, while diminished matrix degradation during chronic tissue injury might be due to the action of TGF-beta1 through TIMP induction.


Subject(s)
Liver/metabolism , Metalloendopeptidases/metabolism , Tissue Inhibitor of Metalloproteinases/metabolism , Transforming Growth Factor beta/pharmacology , Tumor Necrosis Factor-alpha/pharmacology , Animals , Liver/cytology , Liver/drug effects , Liver/enzymology , Rats , Rats, Wistar
16.
Am J Physiol ; 276(2): E262-8, 1999 02.
Article in English | MEDLINE | ID: mdl-9950785

ABSTRACT

The mechanisms that regulate cell turnover in the intestinal epithelium are incompletely understood. Here we tested the hypothesis that proinsulin, present in serum and pancreatic juice in picomolar concentrations, stimulates growth of the rat small intestinal crypt-like cell line IEC-6 under serum-free conditions. Proinsulin binding was assessed by competitive ligand binding studies. Proinsulin and insulin-like growth factor I (IGF-I) stimulated cell proliferation up to threefold above controls, with half-maximal action already in the picomolar range and with additive effects. In early confluent cell monolayers, proinsulin bound with higher affinity (IC50 1.3 +/- 0.05 nM) and capacity (87,200 +/- 2,500 receptors/cell) than IGF-I (4.0 +/- 0.6; 23,700 +/- 2,200, P < 0. 05). C-peptide competed with 10-fold lower affinity for binding of 125I-proinsulin but not for 125I-IGF-I or 125I-insulin, suggesting a specific binding epitope of the proinsulin molecule within or close to the C-peptide region. In contrast, insulin showed approximately 100-fold lower binding affinity and growth-promoting potency than proinsulin or IGF-I. We conclude that proinsulin stimulates growth of small intestinal crypt cells through specific binding and may play a physiological role in the regulation of intestinal epithelial cell proliferation.


Subject(s)
Intestine, Small/cytology , Intestine, Small/drug effects , Proinsulin/pharmacology , Receptors, Cell Surface/physiology , Animals , Binding, Competitive/physiology , C-Peptide/metabolism , Cell Division/drug effects , Cell Division/physiology , Cell Line , Cell Membrane/metabolism , Humans , Insulin/metabolism , Insulin/pharmacology , Insulin-Like Growth Factor I/metabolism , Insulin-Like Growth Factor I/pharmacology , Insulin-Like Growth Factor II/metabolism , Proinsulin/metabolism , Rats
18.
Hepatology ; 27(5): 1275-84, 1998 May.
Article in English | MEDLINE | ID: mdl-9581681

ABSTRACT

The insulin-like growth factors I and II (IGF-I, -II) are circulating peptides known to participate in the regulation of metabolism, growth, and cellular differentiation. In the present study, "early cultured" (days 2-3 of culture) and "culture-activated" (days 6-7 of culture) rat hepatic stellate cells (HSCs) were analyzed for expression of individual components of the IGF axis. Northern blot analysis of IGF-I messenger RNA (mRNA) revealed transcripts of 7.5, 4, 2, and 1.0 to 1.5 kb in culture-activated HSCs, while early cultured HSCs did not express IGF-I mRNA. In culture-activated HSCs, an IGF-I secretion of 8.3+/-2.5 ng/10(6) cells per 24 hours was determined radioimmunologically. In media from early cultured HSCs, IGF-I was not detectable. The IGF-I receptor (IGF-I-R) mRNA expression was three-fold higher in early cultured HSCs than in culture-activated HSCs. By immunohistochemistry, a decrease of IGF-I-R expression of HSCs in vivo following CCl4-induced liver damage was noted as well. IGF binding proteins (IGFBPs) were detected in conditioned media from HSCs by 125I-IGF-I ligand blotting at apparent molecular masses of 24 and 41 to 45 kd that were immunologically identified as IGFBP-4 and -3, respectively. Synthesis of these IGFBPs increased with time of culture. At neutral pH, no IGFBP proteolysis was observed in conditioned media of early cultured and culture-activated HSCs, whereas at acidic pH, protease activities against IGFBP-3 and -4 were detectable. IGFBP protease activities were completely abolished by inhibitors of aspartyl and cysteine proteases. Addition of 100 nmol/L IGF-I stimulated cell proliferation of early cultured HSCs 5.6+/-1.1- and 4.6+/-0.2-fold as measured by [3H]thymidine and 5-bromo-2'-deoxyuridine incorporation, respectively. In culture-activated HSCs, proliferation was increased 1.2+/-0.1-fold in the presence of 100 nmol/L IGF-I in both proliferation assays. It can be concluded that due to a higher expression of the IGF-I-R and lower levels of IGFBPs, early cultured HSCs are more susceptible to the mitogenic actions of IGFs than the culture-activated HSCs. The present data suggest a role for the IGF axis components in the initiation rather than the perpetuation of HSC proliferation during hepatic fibrogenesis.


Subject(s)
Insulin-Like Growth Factor Binding Proteins/physiology , Insulin-Like Growth Factor II/physiology , Insulin-Like Growth Factor I/physiology , Liver/metabolism , Receptor, IGF Type 1/physiology , Receptor, IGF Type 2/physiology , Animals , Carbon Tetrachloride Poisoning/metabolism , Cell Division , Cells, Cultured , Gene Expression , Hydrogen-Ion Concentration , Immunohistochemistry , Liver/cytology , RNA, Messenger/genetics , Rats , Time Factors
20.
Am J Pathol ; 151(5): 1265-72, 1997 Nov.
Article in English | MEDLINE | ID: mdl-9358752

ABSTRACT

During liver tissue repair, hepatic stellate cells (HSC), a pericyte-like mesenchymal liver cell population, transform from a "quiescent" status ("resting" HSC) into myofibroblast-like cells ("activated" HSC) with the latter representing the principle matrix synthesizing cell of the liver. Presently, the mechanisms that terminate HSC cell proliferation when tissue repair is concluded are poorly understood. Controlled cell death known as apoptosis could be a mechanism underlying this phenomenon. Therefore, apoptosis and its regulation were studied in HSC using an in vitro and in vivo approach. Spontaneous apoptosis became detectable in parallel with HSC activation because resting cells (2 days after isolation) displayed no sign of apoptosis, whereas apoptosis was present in 8% (+/- 5%) of "transitional" cells (day 4) and in 18% (+/- 8%) of fully activated cells (day 7). Both CD95 (APO-1/Fas) and CD95L (APO-1-/Fas-ligand) became increasingly expressed during the course of activation. Apoptosis could be fully blocked by CD95-blocking antibodies in normal cells and HSC already entering the apoptotic cycle. Using CD95-activating antibodies, transition of more than 95% cells into apoptosis was evident at each activation step. The apoptosis-regulating proteins Bcl-2 and p53 could not be detected in resting cells but were found in increasing amounts at days 4 and 7 of cultivation. Whereas p53 expression was induced by the CD95-activating antibody, no change was inducible in Bcl-2 expression. The Bcl-2-related protein bax could be found at days 2 and 4 in similar expression, was considerably up-regulated at day 7, but was not regulated by CD95-agonistic antibodies. In vivo, acute tissue damage was first accompanied by activation and proliferation of HSC displaying no sign of apoptosis. In the recovery phase, apoptotic HSC were detectable in parallel to a reduction in the total number of HSC present in the liver tissue. The data demonstrate that apoptosis becomes detectable in parallel with HSC activation, which suggests that apoptosis might represent an important mechanism terminating proliferation of activated HSC.


Subject(s)
Apoptosis/physiology , Liver Regeneration/physiology , Liver/physiology , fas Receptor/physiology , Animals , Antibodies/physiology , Cell Count , Cell Division/physiology , Liver/cytology , Liver/immunology , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Rats , Rats, Wistar , Solubility , bcl-2-Associated X Protein , fas Receptor/analysis
SELECTION OF CITATIONS
SEARCH DETAIL
...