Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
Add more filters










Publication year range
1.
Curr Mol Med ; 16(8): 710-720, 2016.
Article in English | MEDLINE | ID: mdl-27774883

ABSTRACT

Nonalcoholic steatohepatitis (NASH) is a progressive form of nonalcoholic fatty liver disease, characterized by inflammation, hepatocyte injury and fibrogenesis. Overall mortality, and liver-related mortality, are both increased in NASH patients. Considering that nonalcoholic fatty liver disease is the most prevalent hepatic abnormality in the Western world, understanding the mechanisms leading to NASH and its progression to cirrhosis is critical for a better management of these patients. Moreover, a more detailed knowledge of this condition may be helpful to identify those subjects which are more susceptible to develop progressive liver disease. Emerging data indicate that NASH progression results from parallel events originating from the liver as well as from the adipose tissue, and the gastrointestinal tract. In this review we highlight some of the most recent findings reported on the pathogenesis of NASH and its fibrogenic progression to cirrhosis, in an effort to identify possible targets for treatment or biomarkers of disease progression.


Subject(s)
Non-alcoholic Fatty Liver Disease/etiology , Adipokines/metabolism , Animals , Biomarkers , Endoplasmic Reticulum Stress , Genetic Predisposition to Disease , Hepatocytes/metabolism , Hepatocytes/pathology , Humans , Inflammasomes/metabolism , Insulin Resistance , Liver/metabolism , Liver/pathology , Microbiota , Non-alcoholic Fatty Liver Disease/metabolism , Non-alcoholic Fatty Liver Disease/pathology , Oxidative Stress , Risk Factors
2.
Gut ; 55(1): 90-7, 2006 Jan.
Article in English | MEDLINE | ID: mdl-16041064

ABSTRACT

BACKGROUND AND AIMS: Activated myofibroblast-like cells, originating from hepatic stellate cells (HSC/MFs) or other cellular sources, play a key profibrogenic role in chronic liver diseases (CLDs) that, as suggested by studies in animal models or rat HSC/MFs, may be modulated by reactive oxygen intermediates (ROI). In this study, human HSC/MFs, exposed to different levels of superoxide anion (O(2)(.-)) and, for comparison, hydrogen peroxide (H(2)O(2)), were analysed in terms of cytotoxicity, proliferative response, and migration. METHODS: Cultured human HSC/MFs were exposed to controlled O(2)(.-) generation by hypoxanthine/xanthine oxidase systems or to a range of H(2)O(2) concentrations. Induction of cell death, proliferation, and migration were investigated using morphology, molecular biology, and biochemical techniques. RESULTS: Human HSC/MFs were shown to be extremely resistant to induction of cell death by O(2)(.-) and only high rates of O(2)(.-) generation induced either necrotic or apoptotic cell death. Non-cytotoxic low levels of O(2)(.-), able to upregulate procollagen type I expression (but not tissue inhibitor of metalloproteinase 1 and 2), stimulated migration of human HSC/MFs in a Ras/extracellular regulated kinase (ERK) dependent, antioxidant sensitive way, without affecting basal or platelet derived growth factor (PDGF) stimulated cell proliferation. Non-cytotoxic levels of H(2)O(2) did not affect Ras/ERK or proliferative response. A high rate of O(2)(.-) generation or elevated levels of H(2)O(2 )induced cytoskeletal alterations, block in motility, and inhibition of PDGF dependent DNA synthesis. CONCLUSIONS: Low non-cytotoxic levels of extracellularly generated O(2)(.-) may stimulate selected profibrogenic responses in human HSC/MFs without affecting proliferation.


Subject(s)
Hepatocytes/drug effects , Liver/cytology , Superoxides/pharmacology , Apoptosis/drug effects , Cell Death/drug effects , Cell Movement/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Collagen Type I/metabolism , Dose-Response Relationship, Drug , Extracellular Signal-Regulated MAP Kinases/metabolism , Hepatocytes/cytology , Humans , Hydrogen Peroxide/pharmacology , Liver/metabolism , Signal Transduction/physiology , Superoxides/metabolism
3.
J Biol Chem ; 275(51): 40561-7, 2000 Dec 22.
Article in English | MEDLINE | ID: mdl-11007794

ABSTRACT

Hepatic stellate cells (HSC) undergo activation toward myofibroblast-like cells during early stages of liver injury associated with fibrogenesis. Platelet-derived growth factor (PDGF), particularly its BB isoform, has been identified as the most potent mitogen for HSC. 4-Hydroxy-2,3-nonenal and related 4-hydroxy-2, 3-alkenals (HAKs) have been suggested to modulate the process of HSC activation. In this study we investigated the relationship between HAKs and PDGF receptor activation in human HSC. By employing noncytotoxic concentrations (10(-6) m) of HAKs, we observed a significant inhibition of PDGF-BB-dependent DNA synthesis. HAKs inhibited relevant pathways of PDGF-BB-dependent mitogenic signaling, including autophosphorylation of PDGF receptor (PDGF-R) beta subunits and activation of phosphatidylinositol 3-kinase and extracellular regulated kinases 1/2. Inhibition of DNA synthesis was reversible, and recovery of PDGF-mediated mitogenic signaling occurred within 24-48 h and was associated with HAKs-induced up-regulation of PDGF-R beta gene expression. 4-Hydroxy-2,3-nonenal, used as a model HAK, inhibited the intrinsic tyrosine kinase activity associated with the PDGF-R beta subunit, whereas binding of PDGF to its receptor was unaffected. This study identifies a novel regulatory mechanism of reactive aldehydes on PDGF receptor signaling and biologic actions, which may be relevant in several pathophysiological conditions, including liver fibrosis.


Subject(s)
Aldehydes/metabolism , Receptor, Platelet-Derived Growth Factor beta/metabolism , Signal Transduction , Tyrosine/metabolism , Aldehydes/pharmacology , Cell Line , DNA Replication , Humans , Phosphorylation , Receptor, Platelet-Derived Growth Factor beta/chemistry , Up-Regulation/drug effects
4.
Gastroenterology ; 119(2): 479-92, 2000 Aug.
Article in English | MEDLINE | ID: mdl-10930383

ABSTRACT

BACKGROUND & AIMS: Nitrovasodilators have been proposed for the treatment of portal hypertension alone or in combination with beta-blockers. In addition to their vasodilatory properties, nitric oxide (NO) donors may exert direct antifibrogenic properties. We evaluated the effect of nitroglycerin (NTG) and S-nitroso-N-acetyl penicillamine (SNAP) on the mitogenic and chemotactic properties of platelet-derived growth factor (PDGF)-BB and the modulation of the relative intracellular signaling pathways in fully activated human hepatic stellate cells (HSCs), a cell type that plays an active role in liver fibrogenesis and portal hypertension. METHODS & RESULTS: Both NTG and SNAP induced a dose-dependent decrease in PDGF-induced DNA synthesis and cell migration, which was associated with a decrease in PDGF-induced intracellular Ca(2+) increase and extracellular signal-regulated kinase (ERK) activity. These effects were not related to activation of the classic soluble guanylate cyclase (sGC)/guanosine 3',5'-cyclic monophosphate pathway; accordingly, Western blot analysis of HSC lysates revealed the absence of the alpha(1)beta(1) ubiquitous subunits of sGC, whereas they were detectable in quiescent HSCs, freshly isolated from normal human liver. Conversely, both NTG and SNAP induced a more than 10-20-fold increase in prostaglandin E(2) in cell supernatants within 1 minute, associated with an increase in intracellular adenosine 3',5'-cyclic monophosphate levels. Accordingly, the inhibitory effects of NO donors on PDGF action and signaling were eliminated after preincubation with ibuprofen. CONCLUSIONS: These results suggest that NO donors may exert a direct antifibrogenic action by inhibiting proliferation, motility, and contractility of HSCs in addition to a reduction of fibrillar extracellular matrix accumulation.


Subject(s)
Cell Movement/drug effects , Liver/cytology , Nitroglycerin/pharmacology , Platelet-Derived Growth Factor/pharmacology , Vasodilator Agents/pharmacology , Calcium/metabolism , Cell Division/drug effects , Cells, Cultured , Chemotaxis/drug effects , Cyclic GMP/metabolism , Dinoprostone/metabolism , Guanylate Cyclase/metabolism , Hemostatics/pharmacology , Humans , Liver/enzymology , Nitric Oxide Donors/pharmacology , Penicillamine/analogs & derivatives , Penicillamine/pharmacology , Signal Transduction/drug effects , Thrombin/pharmacology , Type C Phospholipases/metabolism
5.
Gastroenterology ; 119(2): 466-78, 2000 Aug.
Article in English | MEDLINE | ID: mdl-10930382

ABSTRACT

BACKGROUND & AIMS: Proliferation and migration of hepatic stellate cells (HSCs) and expression of chemokines are involved in the pathogenesis of liver inflammation and fibrogenesis. Peroxisome proliferator-activated receptor (PPAR)-gamma is a receptor transcription factor that controls growth and differentiation in different tissues. We explored the effects of PPAR-gamma agonists on the biological actions of cultured human HSCs. METHODS: HSCs were isolated from normal human liver tissue and used in their myofibroblast-like phenotype or immediately after isolation. Activation of PPAR-gamma was induced with 15-deoxy-Delta(12, 14)-prostaglandin J(2) or with troglitazone. RESULTS: PPAR-gamma agonists dose-dependently inhibited HSC proliferation and chemotaxis induced by platelet-derived growth factor. This effect was independent of changes in postreceptor signaling or expression of c-fos and c-myc and was associated with inhibition of cell cycle progression beyond the G(1) phase. Activation of PPAR-gamma also resulted in a complete inhibition of the expression of monocyte chemotactic protein 1 at the gene and protein levels. Comparison of quiescent and culture-activated HSCs revealed a marked decrease in PPAR-gamma expression in activated cells. CONCLUSIONS: Activation of PPAR-gamma modulates profibrogenic and proinflammatory actions in HSCs. Reduced PPAR-gamma expression may contribute to confer an activated phenotype to HSCs.


Subject(s)
Hepatitis/metabolism , Liver Cirrhosis/metabolism , Liver/cytology , Liver/immunology , Receptors, Cytoplasmic and Nuclear/metabolism , Thiazolidinediones , Transcription Factors/metabolism , Antineoplastic Agents/pharmacology , Biological Transport/drug effects , Biological Transport/immunology , Cell Division/immunology , Cell Movement/immunology , Cell Survival/drug effects , Cells, Cultured , Chemokine CCL2/genetics , Chemokine CCL2/metabolism , Chromans/pharmacology , Cytotoxins/metabolism , Gene Expression/immunology , Hepatitis/immunology , Hepatitis/pathology , Humans , Interleukin-1/pharmacology , Ligands , Liver/metabolism , Liver Cirrhosis/immunology , Liver Cirrhosis/pathology , Mitogen-Activated Protein Kinases/metabolism , NF-kappa B/metabolism , Phosphorylation , Platelet-Derived Growth Factor/metabolism , Prostaglandin D2/analogs & derivatives , Prostaglandin D2/pharmacology , Proto-Oncogenes/genetics , RNA, Messenger/analysis , Thiazoles/pharmacology , Transcription Factor AP-1/metabolism , Troglitazone , Tyrosine/metabolism , Wound Healing/immunology , p38 Mitogen-Activated Protein Kinases
6.
Hepatology ; 32(1): 17-25, 2000 Jul.
Article in English | MEDLINE | ID: mdl-10869284

ABSTRACT

We studied the role of gastrin in regulating cholangiocyte proliferation induced by bile duct ligation (BDL). In purified cholangiocytes, we evaluated (1) for the presence of cholecystokinin-B (CCK-B)/gastrin receptors, (2) the effect of gastrin on D-myo-Inositol 1,4,5-triphosphate (IP(3)) levels, and (3) the effect of gastrin on DNA synthesis and adenosine 3', 5'-monophosphate (cAMP) levels in the absence or presence of CCK-A (L-364,718) and CCK-B/gastrin (L-365,260) receptor inhibitors, 1, 2-bis(2-aminophenoxy)-ethane-N,N,N',N'-tetraacetic acid tetrakis(acetxymethyl ester) (BAPTA/AM; an intracellular Ca(2+) chelator), and 2 protein kinase C (PKC) inhibitors, 1-(5-Isoquinolinylsulfonyl)-2-methylpiperazine (H7) and staurosporin. To evaluate if gastrin effects on cholangiocyte proliferation are mediated by the isoform PKCalpha, we evaluated (1) for the presence of PKCalpha in cholangiocytes and (2) the effect of gastrin on the PKCalpha protein expression in a triton-soluble (containing cytoplasm + membrane) and a triton-insoluble (containing cytoskeleton) fraction. To evaluate the effects of gastrin in vivo, immediately following BDL, gastrin or bovine serum albumin (BSA) was infused by minipumps for 7 days to rats and we measured cholangiocyte growth and cAMP levels. We found CCK-B/gastrin receptors on cholangiocytes. Gastrin increased IP(3) levels. Gastrin inhibited DNA synthesis and cAMP synthesis in cholangiocytes. Gastrin effects on cholangiocyte functions were blocked by L-365,260, BAPTA/AM, H7, and staurosporin but not by L-364,718. Gastrin induced translocation of PKCalpha from cholangiocyte cytoskeleton to membrane. In vivo, gastrin decreased cholangiocyte growth and cAMP synthesis compared with controls. We concluded that gastrin inhibits cholangiocyte growth in BDL rats by interacting with CCK-B/gastrin receptors through a signal transduction pathway involving IP(3), Ca(2+), and PKCalpha.


Subject(s)
Bile Ducts/cytology , Calcium/physiology , Cholestasis, Extrahepatic/pathology , Gastrins/pharmacology , Inositol 1,4,5-Trisphosphate/physiology , Isoenzymes/physiology , Protein Kinase C/physiology , Receptors, Cholecystokinin/physiology , Animals , Bile Ducts/drug effects , Cell Division/drug effects , Male , Rats , Rats, Inbred F344 , Receptors, Cholecystokinin/drug effects
7.
Hepatology ; 30(4): 951-8, 1999 Oct.
Article in English | MEDLINE | ID: mdl-10498647

ABSTRACT

Upon liver injury, hepatic stellate cells (HSC) show increased proliferation, motility, and extracellular matrix (ECM) production. The extracellular signal-regulated kinases (ERK) control different functions in a cell-specific manner. In this study, we evaluated the role of ERK activation in cultured HSC stimulated with platelet-derived growth factor (PDGF) and after induction of liver injury in vivo. HSC were isolated from normal human liver tissue, cultured on plastic, and used in their myofibroblast-like phenotype. In in vivo experiments, HSC were isolated from normal rats or at different time points after a single intragastric administration of CCl(4). Nontoxic concentrations of PD98059, a specific inhibitor of ERK activation, reduced PDGF-induced activation of ERK in a dose-dependent fashion. Suppression of ERK activation was associated with complete inhibition of HSC proliferation and with a 57% reduction in chemotaxis. In the presence of the ERK inhibitor, binding of the AP-1 complex and of STAT1 to the related regulatory elements was inhibited. The inhibition of the DNA binding activity of STAT1 was mediated by a reduction in PDGF-induced tyrosine phosphorylation. Expression of c-fos in response to PDGF was also reduced, but not suppressed, by treatment with PD98059. In HSC isolated from CCl(4)-treated rats, ERK activity increased as early as 6 hours following liver damage, and declined thereafter. The results of this study indicate that ERK activation regulates proliferation and chemotaxis of HSC, and modulates nuclear signaling. Acute liver damage in vivo leads to activation of ERK in HSC.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Liver Diseases/enzymology , Liver/physiology , Platelet-Derived Growth Factor/physiology , Animals , Calcium-Calmodulin-Dependent Protein Kinases/antagonists & inhibitors , Calcium-Calmodulin-Dependent Protein Kinases/physiology , Carbon Tetrachloride , Cell Division/physiology , Cell Movement/physiology , Cell Nucleus/physiology , Chemical and Drug Induced Liver Injury , Enzyme Activation/physiology , Enzyme Induction , Enzyme Inhibitors/pharmacology , Flavonoids/pharmacology , Humans , Liver/cytology , Liver/pathology , Proto-Oncogene Proteins c-fos/metabolism , Rats , Signal Transduction/drug effects , Time Factors
8.
J Hepatol ; 31(1): 100-9, 1999 Jul.
Article in English | MEDLINE | ID: mdl-10424289

ABSTRACT

BACKGROUND/AIMS: Chronic cholestasis stimulates a fibroductular reaction which may progress to secondary biliary fibrosis and cirrhosis. Since platelet-derived growth factor has been indicated as a major fibrogenic factor in chronic liver disease, we analyzed its expression and that of its receptor beta subunit in a rat model of chronic cholestasis. METHODS: Liver tissue samples collected at 7, 10, 21, and 28 days after induction of cholestasis obtained by bile duct ligation, were analyzed by immunohistochemistry, in situ hybridization and RNase protection assay for the expression of platelet-derived growth factor (PDGF)-B chain and receptor beta subunit. Furthermore, the expression of PDGF-B chain mRNA was analyzed in highly purified cholangiocytes from normal and cholestatic rat liver. RESULTS: In cholestatic liver, platelet-derived growth factor-BB and B chain mRNA expression increased up to 4 weeks in epithelial cells of proliferating bile ducts, and periductular mesenchymal cells. The increased expression of PDGF-B chain mRNA was confirmed in highly purified cholangiocytes obtained from normal and cholestatic rat liver. The expression of the receptor beta subunit progressively increased after induction of cholestasis and was mainly localized to desmin-positive periductular hepatic stellate cells. CONCLUSIONS: These data suggest that platelet-derived growth factor-B chain can be synthesized by cholangiocytes during chronic cholestasis. The presence of its receptor on periductular hepatic stellate cells raises the possibility that, in this experimental setting, this cytokine might contribute to fibrogenesis in vivo.


Subject(s)
Bile Ducts/metabolism , Cholestasis/metabolism , Gene Expression Regulation , Liver/metabolism , Animals , Becaplermin , Bile Ducts/pathology , Cholestasis/pathology , Chronic Disease , Common Bile Duct/physiology , Female , In Situ Hybridization , Liver/pathology , Platelet-Derived Growth Factor/biosynthesis , Platelet-Derived Growth Factor/genetics , Proto-Oncogene Proteins c-sis , RNA, Messenger/genetics , Rats , Rats, Wistar , Transcription, Genetic
9.
Gastroenterology ; 117(1): 191-9, 1999 Jul.
Article in English | MEDLINE | ID: mdl-10381927

ABSTRACT

BACKGROUND & AIMS: To investigate the role of the cholinergic system in regulation of cholangiocyte functions, we evaluated the effects of vagotomy on cholangiocyte proliferation and secretion in rats that underwent bile duct ligation (BDL rats). METHODS: After bile duct ligation (BDL), the vagus nerve was resected; 7 days later, expression of M3 acetylcholine receptor was evaluated. Cholangiocyte proliferation was assessed by morphometry and measurement of DNA synthesis. Apoptosis was evaluated by light microscopy and annexin-V staining. Ductal secretion was evaluated by measurement of secretin-induced choleresis, secretin receptor (SR) gene expression, and cyclic adenosine 3',5'-monophosphate (cAMP) levels. RESULTS: Vagotomy decreased the expression of M3 acetylcholine receptors in cholangiocytes. DNA synthesis and ductal mass were markedly decreased, whereas cholangiocyte apoptosis was increased by vagotomy. Vagotomy decreased ductal secretion. Forskolin treatment prevented the decrease in cAMP levels induced by vagotomy, maintained cholangiocyte proliferation, and decreased cholangiocyte apoptosis caused by vagotomy in BDL rats. Cholangiocyte secretion was also maintained by forskolin. CONCLUSIONS: Vagotomy impairs cholangiocyte proliferation and enhances apoptosis, leading to decreased ductal mass in response to BDL. Secretin-induced choleresis of BDL rats was virtually eliminated by vagotomy in association with decreased cholangiocyte cAMP levels. Maintenance of cAMP levels by forskolin administration prevents the effects of vagotomy on cholangiocyte proliferation, apoptosis, and secretion.


Subject(s)
Bile Ducts/cytology , Bile Ducts/physiology , Cholinergic Fibers/physiology , Animals , Apoptosis/physiology , Bile Ducts/drug effects , Bile Ducts/metabolism , Body Weight/physiology , Cell Division/physiology , Colforsin/pharmacology , Cyclic AMP/antagonists & inhibitors , Cyclic AMP/metabolism , Hormones/blood , Ligation , Liver/anatomy & histology , Male , Organ Size/physiology , Rats , Rats, Inbred F344 , Receptors, Cholinergic/metabolism , Vagotomy
10.
Am J Physiol ; 276(5): G1289-301, 1999 05.
Article in English | MEDLINE | ID: mdl-10330021

ABSTRACT

Bile duct damage and/or loss is limited to a range of duct sizes in cholangiopathies. We tested the hypothesis that CCl4 damages only large ducts. CCl4 or mineral oil was given to bile duct-ligated (BDL) rats, and 1, 2, and 7 days later small and large cholangiocytes were purified and evaluated for apoptosis, proliferation, and secretion. In situ, we measured apoptosis by morphometric and TUNEL analysis and the number of small and large ducts by morphometry. Two days after CCl4 administration, we found an increased number of small ducts and reduced number of large ducts. In vitro apoptosis was observed only in large cholangiocytes, and this was accompanied by loss of proliferation and secretion in large cholangiocytes and loss of choleretic effect of secretin. Small cholangiocytes de novo express the secretin receptor gene and secretin-induced cAMP response. Consistent with damage of large ducts, we detected cytochrome P-4502E1 (which CCl4 converts to its radicals) only in large cholangiocytes. CCl4 induces selective apoptosis of large ducts associated with loss of large cholangiocyte proliferation and secretion.


Subject(s)
Bile Duct Diseases/chemically induced , Bile Ducts, Intrahepatic , Bile Ducts/surgery , Carbon Tetrachloride/toxicity , Animals , Apoptosis , Bile/metabolism , Bile Duct Diseases/pathology , Bile Duct Diseases/physiopathology , Bile Ducts, Intrahepatic/pathology , Bile Ducts, Intrahepatic/physiopathology , Cell Division , Cell Separation , Cyclic AMP/metabolism , Cytochrome P-450 CYP2E1/analysis , Epithelial Cells/pathology , Gene Expression , Ligation , Male , Rats , Rats, Inbred F344 , Receptors, G-Protein-Coupled , Receptors, Gastrointestinal Hormone/genetics , Secretin/pharmacology
11.
Hepatology ; 29(2): 307-19, 1999 Feb.
Article in English | MEDLINE | ID: mdl-9918904

ABSTRACT

The aim of this study was to develop a model of selective duct damage restricted to hormone-responsive segments corresponding to the ducts damaged in primary biliary cirrhosis (PBC). Carbon tetrachloride (CCl4) was fed by gavage to rats, and 2, 7, 14, and 28 days later, small and large cholangiocytes were isolated. Apoptosis was determined in situ by morphology and in purified cholangiocytes by assessment of nuclear fragmentation by 4, 6-diamidino-2-phenylindole (DAPI) staining. Cholangiocyte proliferation was evaluated in situ by morphometry of liver sections stained for cytokeratin-19 (CK-19) and by proliferating cellular nuclear antigen (PCNA) staining in liver sections and in purified cholangiocytes by PCNA gene expression. Ductal secretion was assessed by measurement of secretin receptor (SR) gene expression and secretin-induced cyclic adenosine 3',5'-monophosphate (cAMP) synthesis and secretin-induced choleresis. Two days after CCl4 administration, there was an increased number of small ducts, but a reduction of large ducts. Apoptosis, observed only in large ducts, was associated with decreased DNA synthesis and ductal secretion. Conversely, small cholangiocytes expressed de novo the SR gene and secretin-stimulated cAMP synthesis 2 days after CCl4 treatment. Proliferation of large cholangiocytes was delayed until 7 days, which was associated with a transient increase in ductal secretion in vivo. CCl4 effects on cholangiocytes were reversed by day 28. CCl4 treatment causes a decrease in large duct mass as a result of a higher rate of apoptosis and absence of initial proliferation in large cholangiocytes. These processes were concomitant with a decrease of ductal secretion in large cholangiocytes. Small cholangiocytes appear resistant to CCl4-induced apoptosis, and proliferate and transiently compensate for loss of proliferative and secretory activity of large cholangiocytes.


Subject(s)
Bile Ducts, Intrahepatic/pathology , Carbon Tetrachloride/administration & dosage , Liver Cirrhosis, Biliary/chemically induced , Liver Cirrhosis, Biliary/pathology , Animals , Apoptosis , Bicarbonates/metabolism , Bile/physiology , Bile Ducts, Intrahepatic/metabolism , Cell Division , Cell Nucleus/pathology , Cyclic AMP/biosynthesis , DNA/biosynthesis , Disease Models, Animal , Epithelial Cells/pathology , Fluorescent Dyes , Indoles , Liver Cirrhosis, Biliary/physiopathology , Male , Proliferating Cell Nuclear Antigen/analysis , Proliferating Cell Nuclear Antigen/genetics , Rats , Receptors, G-Protein-Coupled , Receptors, Gastrointestinal Hormone/genetics , Secretin/pharmacology
12.
Gastroenterology ; 116(1): 179-86, 1999 Jan.
Article in English | MEDLINE | ID: mdl-9869616

ABSTRACT

BACKGROUND & AIMS: We have shown that taurocholate (TC) and taurolithocholate (TLC) interact in vitro with normal cholangiocytes, increasing DNA synthesis, secretin receptor (SR) gene expression, and adenosine 3',5'-cyclic monophosphate (cAMP) synthesis. To further extend these in vitro studies, we tested the hypothesis that bile acids (BAs) directly stimulate cholangiocyte proliferation and secretion in vivo. METHODS: After feeding with TC or TLC (1% for 1-4 weeks), we assessed the following in vivo: (1) ductal proliferation by both morphometry and immunohistochemistry for proliferating cell nuclear antigen (PCNA) and measurement of [3H]thymidine incorporation; and (2) the effect of secretin on bile secretion and bicarbonate secretion in vivo. Genetic expression of H3-histone and SR and intracellular cAMP levels were measured in isolated cholangiocytes. RESULTS: After BA feeding, there was an increased number of PCNA-positive cholangiocytes and an increased number of ducts compared with control rats. [3H]Thymidine incorporation, absent in control cholangiocytes, was increased in cholangiocytes from BA-fed rats. In BA-fed rats, there was increased SR gene expression (approximately 2.5-fold) and secretin-induced cAMP levels (approximately 3.0-fold) in cholangiocytes, which was associated with de novo secretin-stimulated bile flow and bicarbonate secretion. CONCLUSIONS: These data indicate that elevated BA levels stimulate ductal secretion and cholangiocyte proliferation.


Subject(s)
Bile Acids and Salts , Bile Ducts, Intrahepatic/metabolism , Bile Ducts, Intrahepatic/pathology , Animals , Bile/metabolism , Bile Acids and Salts/metabolism , Bile Acids and Salts/pharmacology , Bile Ducts, Intrahepatic/drug effects , Cell Division/drug effects , Cholagogues and Choleretics/pharmacology , Cyclic AMP/metabolism , Gene Expression Regulation/drug effects , Liver/pathology , Male , Rats , Rats, Inbred F344 , Receptors, G-Protein-Coupled , Receptors, Gastrointestinal Hormone/biosynthesis , Receptors, Gastrointestinal Hormone/genetics , Secretin/metabolism , Taurocholic Acid/pharmacology , Taurolithocholic Acid/pharmacology , Thymidine/metabolism
13.
Am J Physiol ; 275(4): G835-46, 1998 10.
Article in English | MEDLINE | ID: mdl-9756516

ABSTRACT

We studied the expression of endothelin-1 (ET-1) receptors (ETA and ETB) and the effects of ET-1 on cholangiocyte secretion. The effects of ET-1 on cholangiocyte secretion were assessed in normal and bile duct-ligated (BDL) rats by measuring 1) basal and secretin-induced choleresis in vivo, 2) secretin receptor gene expression and cAMP levels in small and large cholangiocytes, and 3) luminal expansion in response to secretin in intrahepatic bile duct units (IBDU). ETA and ETB receptors were expressed by small and large cholangiocytes. ET-1 had no effect on basal bile flow or bicarbonate secretion in normal or BDL rats but decreased secretin-induced bicarbonate-rich choleresis in BDL rats. ET-1 decreased secretin receptor gene expression and secretin-stimulated cAMP synthesis in large cholangiocytes and secretin-induced luminal expansion in IBDU from normal or BDL rats. The inhibitory effects of ET-1 on secretin-induced cAMP synthesis and luminal duct expansion were blocked by specific inhibitors of the ETA (BQ-610) receptor. ET-1 inhibits secretin-induced ductal secretion by decreasing secretin receptor and cAMP synthesis, two important determinants of ductal secretion.


Subject(s)
Bile Ducts, Intrahepatic/physiology , Bile/metabolism , Endothelin-1/pharmacology , Receptors, Endothelin/genetics , Secretin/pharmacology , Animals , Bile Ducts/physiology , Bile Ducts, Intrahepatic/cytology , Bile Ducts, Intrahepatic/drug effects , Cells, Cultured , Cyclic AMP/metabolism , DNA Primers , Gene Expression Regulation , Male , Rats , Rats, Inbred F344 , Receptor, Endothelin A , Receptor, Endothelin B , Reverse Transcriptase Polymerase Chain Reaction , Secretin/antagonists & inhibitors , Transcription, Genetic
14.
Am J Physiol ; 274(4): G767-75, 1998 04.
Article in English | MEDLINE | ID: mdl-9575860

ABSTRACT

We previously introduced the concept that intrahepatic bile duct epithelial cells, or cholangiocytes, are functionally heterogeneous. This concept is based on the observation that secretin receptor (SR) gene expression and secretin-induced cAMP synthesis are present in cholangiocytes derived from large (> 15 microns in diameter) but not small (< 15 microns in diameter) bile ducts. In work reported here, we tested the hypothesis that cholangiocytes are heterogeneous with regard to proliferative capacity. We assessed cholangiocyte proliferation in vivo by measurement of [3H]thymidine incorporation and in vitro by both [3H]thymidine incorporation and H3 histone gene expression in small (fraction 1) and large (fraction 2) cholangiocytes isolated from rats after bile duct ligation (BDL). In the two cholangiocyte subpopulations, we also studied basal somatostatin receptor (SSTR2) gene expression as well as the effects of somatostatin on 1) SR gene expression and secretin-induced cAMP synthesis and 2) [3H]thymidine incorporation and H3 histone gene expression. In normal rat liver, cholangiocytes, unlike hepatocytes, were mitotically dormant; after BDL, incorporation of [3H]thymidine markedly increased in cholangiocytes but not hepatocytes. When subpopulations of cholangiocytes were isolated after BDL, DNA synthesis assessed by both techniques was limited to large cholangiocytes, as was SSTR2 steady-state gene expression. In vitro, somatostatin inhibited SR gene expression and secretin-induced cAMP synthesis only in large cholangiocytes. Moreover, compared with no hormone, somatostatin inhibited DNA synthesis solely in large cholangiocytes. These results support the concept of the heterogeneity of cholangiocytes along the biliary tree, extend this concept to cholangiocyte proliferative activity, and imply that the proliferative compartment of cholangiocytes after BDL is located principally in the cholangiocytes lining large (> 15 microns) bile ducts.


Subject(s)
Bile Ducts, Intrahepatic/cytology , Animals , Autoradiography , Bile Ducts, Intrahepatic/drug effects , Bile Ducts, Intrahepatic/metabolism , Cell Division/physiology , Epithelial Cells/cytology , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Histones/genetics , Immunohistochemistry , Ligation , Male , RNA, Messenger/metabolism , Rats , Rats, Inbred F344 , Receptors, Somatostatin/genetics , Secretin/pharmacology , Somatostatin/pharmacology , Thymidine/pharmacokinetics
15.
Br J Pharmacol ; 122(6): 1047-54, 1997 Nov.
Article in English | MEDLINE | ID: mdl-9401768

ABSTRACT

1. Pentoxifylline (PTF) may act as a potential antifibrogenic agent by inhibiting cell proliferation and/or collagen deposition in cell type(s) responsible for the accumulation of extracellular matrix. The aim of the present study was to investigate at which level PTF may affect synthesis and degradation of type I collagen in human hepatic stellate cells (HSCs), a key source of connective tissue in fibrotic liver. 2. Procollagen type I synthesis and release were evaluated in cells maintained in serum free/insulin free medium for 48 h and then stimulated with transforming growth factor-beta 1 (TGF-beta 1) for different time periods in the presence or absence of PTF. TGF-beta 1 caused an upregulation of procollagen I mRNA levels with a peak increase after 3-6 h of stimulation. This effect was followed by an increase in both the cell associated and the extracellular levels of the corresponding protein, with a peak effect at 9-12 h after the addition of TGF-beta 1. Co-incubation with PTF slightly but consistently reduced basal as well as stimulated procollagen I mRNA levels, with negligible effects on the cell-associated expression of the corresponding protein. Conversely, PTF dose-dependently reduced procollagen type I levels detected in supernatants from unstimulated and stimulated cells. 3. Pulse-chase experiments employing L-[3H]-proline revealed that PTF was able to induce significantly the degradation of procollagen, mainly in the extracellular compartment. We next analysed the effect of PTF on the major pathway involved in type I collagen degradation. PTF did not affect the expression of metalloproteinase 1 (MMP-1) mRNA both in basal and stimulated conditions, whereas it markedly reduced the expression of tissue inhibitor of metalloproteinase 1 (TIMP-1) mRNA. Accordingly incubation with PTF increased the levels of 'activated MMP-1' in cell supernatants in both basal and stimulated conditions. 4. These results suggest that the antifibrogenic action of PTF on human HSCs is mainly mediated by extracellular collagen degradation rather than by a reduction of collagen synthesis.


Subject(s)
Liver/drug effects , Pentoxifylline/pharmacology , Procollagen/metabolism , Transforming Growth Factors/pharmacology , Cells, Cultured , Collagenases/genetics , Extracellular Space/metabolism , Humans , Hydrolysis , Liver/cytology , Liver/metabolism , Matrix Metalloproteinase 1 , Procollagen/genetics , RNA, Messenger/genetics , Tissue Inhibitor of Metalloproteinase-1/genetics
16.
Am J Physiol ; 273(5): G1061-70, 1997 11.
Article in English | MEDLINE | ID: mdl-9374703

ABSTRACT

We assessed the effect of gastrin on ductal secretion in normal and bile duct-ligated (BDL) rats. The effect of gastrin on ductal secretion was examined in the presence of proglumide, a specific antagonist for gastrin receptor (GR). We isolated pure cholangiocytes from normal and BDL rats and assessed gastrin effects on secretin receptor (SR) gene expression and intracellular adenosine 3',5'-cyclic monophosphate (cAMP) levels. We examined the presence of GR mRNA in cholangiocytes by reverse transcription polymerase chain reaction (RT-PCR). In normal or BDL rats, gastrin produced no changes in spontaneous bile secretion. Simultaneous infusion of gastrin inhibited secretin-induced choleresis and bicarbonate output in BDL rats. In the presence of proglumide gastrin did not inhibit secretin-induced choleresis in BDL rats. Gastrin decreased in cholangiocytes from BDL rats 1) SR gene expression and 2) secretin-induced cAMP levels. With the use of RT-PCR, GR mRNA was detected in cholangiocytes. Similar to what is shown for secretin and somatostatin, we propose that the opposing effects of secretin and gastrin on cholangiocyte secretory activity regulate ductal secretion in rats.


Subject(s)
Bile Ducts, Intrahepatic/physiology , Cyclic AMP/metabolism , Gastrins/pharmacology , Proglumide/pharmacology , Receptors, Cholecystokinin/physiology , Secretin/pharmacology , Animals , Bicarbonates/pharmacology , Bile/metabolism , Bile Ducts/physiology , Bile Ducts, Intrahepatic/cytology , Bile Ducts, Intrahepatic/drug effects , Gallbladder/physiology , Male , Polymerase Chain Reaction , Rats , Rats, Inbred F344 , Receptors, Cholecystokinin/antagonists & inhibitors , Receptors, Cholecystokinin/biosynthesis , Secretin/antagonists & inhibitors
17.
Gastroenterology ; 113(5): 1734-40, 1997 Nov.
Article in English | MEDLINE | ID: mdl-9352879

ABSTRACT

BACKGROUND & AIMS: Bile acids interact with cholangiocytes, resulting in cholangiocyte proliferation and increases in ductal bile secretion in large but not small cholangiocytes. It was proposed that for bile acids to exert these effects on cholangiocytes, a specific uptake mechanism must be present in cholangiocytes. The aim of this study was to show the expression of a bile acid transporter in cholangiocytes. METHODS: Small and large cholangiocytes or intrahepatic bile duct units (IBDUs) were isolated from normal rats, and gene expression for the apical Na+-dependent bile acid transporter (ABAT) and the 14-kilodalton ileal cytosolic binding protein (IBABP) was assessed by ribonuclease-protection assays. Tissue and subcellular distribution of bile acid transporters was also studied. [14C]-Taurocholate uptake into cholangiocytes was determined. RESULTS: Both ABAT and IBABP messenger RNAs were detected in large but not small cholangiocytes. By immunohistochemistry, ABAT was present in large but not small cholangiocytes. Immunofluorescence showed ABAT to be present in the apical membrane of large IBDUs. A Na+-dependent saturable uptake of taurocholate was present in large but not small cholangiocytes. CONCLUSIONS: These proteins may mediate bile acid uptake from the duct lumen in large ducts, resulting in modification of canalicular bile secretion and modulation of ductal bile secretion and growth.


Subject(s)
Bile Ducts/chemistry , Carrier Proteins/analysis , Hydroxysteroid Dehydrogenases , Membrane Glycoproteins , Sodium/pharmacology , Animals , Bile Ducts/cytology , Carrier Proteins/genetics , Immunohistochemistry , Male , RNA, Messenger/analysis , Rats , Rats, Inbred F344 , Taurocholic Acid/metabolism
18.
Am J Physiol ; 273(2 Pt 1): G518-29, 1997 Aug.
Article in English | MEDLINE | ID: mdl-9277433

ABSTRACT

Accumulation of bile acids (BA) and cholangiocyte proliferation occur in cholestasis, but BA effects on the proliferative and secretory capacity of cholangiocytes are undefined. Cholangiocyte proliferation coupled with increased expression of H3 histone and secretin receptor (SR) genes and secretin-stimulated adenosine 3',5'-cyclic monophosphate (cAMP) levels is limited to large cholangiocytes. We isolated pooled small and large cholangiocytes and studied the effect of taurocholic (TC) and taurolithocholic (TLC) acids on proliferation, by measurement of H3 histone gene expression, and secretion, by measurement of SR gene expression, cAMP levels, and Cl-/HCO3- exchanger activity. In pooled cholangiocytes, TC and TLC increased H3 histone (12-fold) and SR (3-fold) gene expression and both spontaneous (1.4-fold) and secretin-induced (4-fold) cAMP response. TC and TLC increased H3 histone (10-fold) and SR (2-fold) gene expression and secretin-induced cAMP response and Cl-/HCO3- exchanger activity (3-fold) only in large cholangiocytes. In large cholangiocytes, BA may have a signaling function in the modulation of ductal secretion.


Subject(s)
Bile Ducts/cytology , Bile Ducts/metabolism , Cholagogues and Choleretics/pharmacology , Taurocholic Acid/pharmacology , Taurolithocholic Acid/pharmacology , Animals , Antiporters/metabolism , Bile Ducts/physiology , Cell Division/drug effects , Chloride-Bicarbonate Antiporters , Cyclic AMP/biosynthesis , Cyclic AMP/metabolism , Gene Expression , Histones/genetics , Intracellular Membranes/metabolism , Male , Phenotype , Rats , Rats, Inbred F344 , Receptors, G-Protein-Coupled , Receptors, Gastrointestinal Hormone/genetics
19.
Hepatology ; 25(2): 361-7, 1997 Feb.
Article in English | MEDLINE | ID: mdl-9021948

ABSTRACT

Experimental evidence indicates that the lipid peroxidation of biological membranes is often associated with the development of liver fibrosis. We have studied the effect of neutrophil-derived reactive oxygen species (ROS) on collagen synthesis by human hepatic stellate cells (HSC), the major source of collagen in the liver, in a coculture system. Lipid peroxidation in the cocultures was evaluated in terms of either malondialdehyde (MDA) production or the formation of MDA/4-hydroxynonenal protein adducts. The expression of cellular messenger RNAs (mRNAs) was evaluated by either Northern blotting or RNAse protection assay. Nitric oxide (NO) synthase activity in cells was measured by [3H]citrulline formation from [3H]arginine. In vitro exposure of HSC to ROS resulted in the early induction of lipid peroxidation and was associated with a marked increase (threefold) of procollagen I mRNA expression and synthesis. The addition of antioxidants, such as vitamin E or superoxide dismutase (SOD), impaired this stimulation. The inhibition of neutrophil NO formation by N(G)-monomethyl-L-arginine made the ROS-induced stimulation of procollagen I more evident. The addition of xanthine/xanthine oxidase X/XO, a superoxide anion donor, to HSC cultures strongly increased procollagen I synthesis. This stimulation was hampered by the addition of both SOD and sodium nitroprusside (an NO donor). The contribution of HSC to the production of NO in our coculture system was negligible, because inducible NO synthase (iNOS) mRNA was almost undetectable in these cells, and also because the amount of NO produced by HSC stimulated with tumor necrosis factor alpha (TNF-alpha) and lipopolysaccharide (LPS) was 500 times less than that synthesized by neutrophils. In conclusion, these results indicate that neutrophil-derived ROS may contribute to the development of hepatic fibrosis associated with alcoholic hepatitis. NO produced by neutrophils may exert a "protective" antioxidant effect by operating as a scavenger of superoxide anion.


Subject(s)
Neutrophils/metabolism , Nitric Oxide/metabolism , Procollagen/metabolism , Reactive Oxygen Species/metabolism , Humans , Nitric Oxide Synthase/metabolism , RNA, Messenger/metabolism , Superoxide Dismutase/metabolism , Vitamin E/pharmacology
20.
Br J Pharmacol ; 119(6): 1117-24, 1996 Nov.
Article in English | MEDLINE | ID: mdl-8937713

ABSTRACT

1. It has been proposed that pentoxifylline (PTF) acts an antifibrogenic agent by reducing the synthesis of extracellular matrix components, and this possibility has been confirmed in animal models of hepatic fibrosis. In this study the effects of PTF on the proliferation of extracellular matrix producing cells induced by platelet-derived growth factor (PDGF) were evaluated. The study was performed on hepatic stellate cells, currently indicated as the major source of extracellular matrix in fibrotic liver. 2. PTF caused a dose-dependent reduction of PDGF-induced mitogenesis with an IC50 of 170 microM, identical to the EC50 for the increase in intracellular cyclic AMP levels. Preincubation with PTF did not affect either PDGF-receptor autophosphorylation or phosphotidylinositol 3-kinase activity, whereas it markedly reduced PDGF-stimulated extracellular signal-regulated kinase (ERK) activity and ERK isoform phosphorylation. PTF also reduced PDGF-induced c-fos mRNA expression, which is dependent on activation of the RAS/ERK pathway. In addition, the PDGF-induced increase in cytsolic-free calcium was almost completely prevented by pretreating the cells with PTF. 3. The results of the present study indicate that PTF, in addition to its effect on collagen deposition and degradation, may exert an antifibrogenic effect by reducing the PDGF-induced proliferation of extracellular matrix producing cells. This effect appears to be mediated by a reduction of PDGF-stimulated ERK activity as well as of other intracellular signalling pathways such as the PDGF-induced elevation of cytosolic-free calcium.


Subject(s)
Adipocytes/drug effects , Calcium-Calmodulin-Dependent Protein Kinases/antagonists & inhibitors , Liver/drug effects , Mitogen-Activated Protein Kinases , Pentoxifylline/pharmacology , Platelet-Derived Growth Factor/pharmacology , Adipocytes/enzymology , Calcium/metabolism , Cell Division/drug effects , Cells, Cultured , DNA/biosynthesis , Humans , Liver/cytology , Liver/enzymology , Liver Cirrhosis/drug therapy , Mitogen-Activated Protein Kinase 1 , Mitogen-Activated Protein Kinase 3 , Phosphatidylinositol 3-Kinases , Phosphorylation , Phosphotransferases (Alcohol Group Acceptor)/biosynthesis , Receptors, Platelet-Derived Growth Factor/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...