Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
1.
Acta Cytol ; 62(4): 295-301, 2018.
Article in English | MEDLINE | ID: mdl-29734171

ABSTRACT

OBJECTIVE: Routine bile duct brushing cytology is an important diagnostic tool in the evaluation of bile duct stricture. The purpose of this study was to evaluate the performance of the UroVysion fluorescence in situ hybridization (FISH) assay for the detection of malignant bile duct brushing specimens. STUDY DESIGN: Thirty-five bile duct brushing specimens were included in the study. The FISH assay utilized the commercially available UroVysion probes. The indeterminate cytology results were considered as negative for statistical analysis. RESULTS: Twenty-two of 35 patients were diagnosed as having malignancy based on tissue diagnosis or clinical progression of disease by image assessment. The sensitivity of routine cytology and FISH for the detection of malignancy was 14% (3/22) and 55% (12/22), respectively (p = 0.003). The specificity of routine cytology and FISH was 100% (13/13) and 62% (8/13), respectively (p = 0.025). The false-positive rate for routine cytology and FISH was 0% (0/13) and 38% (5/13), respectively. CONCLUSIONS: Our study shows that FISH is significantly more sensitive than routine cytology for the detection of malignancy in bile duct brushing specimens. However, in our study, the specificity of FISH was poor compared to the excellent specificity of routine cytology. The compromised specificity of FISH may limit its utility in the detection of malignant bile duct brushing specimens.


Subject(s)
Bile Duct Neoplasms/genetics , Bile Ducts, Extrahepatic/pathology , Biomarkers, Tumor/genetics , Cholangiocarcinoma/genetics , Cholestasis, Extrahepatic/genetics , Chromosome Aberrations , In Situ Hybridization, Fluorescence , Pancreatic Neoplasms/genetics , Adult , Aged , Aged, 80 and over , Bile Duct Neoplasms/complications , Bile Duct Neoplasms/pathology , Child , Cholangiocarcinoma/complications , Cholangiocarcinoma/pathology , Cholangiopancreatography, Endoscopic Retrograde , Cholestasis, Extrahepatic/etiology , Cholestasis, Extrahepatic/pathology , False Positive Reactions , Female , Humans , Liquid Biopsy , Male , Middle Aged , Pancreatic Neoplasms/complications , Pancreatic Neoplasms/pathology , Predictive Value of Tests , Reproducibility of Results , Young Adult
2.
Gene Expr ; 18(3): 197-207, 2018 08 22.
Article in English | MEDLINE | ID: mdl-29580318

ABSTRACT

α7-nAChR is a nicotinic acetylcholine receptor [specifically expressed on hepatic stellate cells (HSCs), Kupffer cells, and cholangiocytes] that regulates inflammation and apoptosis in the liver. Thus, targeting α7-nAChR may be therapeutic in biliary diseases. Bile duct ligation (BDL) was performed on wild-type (WT) and α7-nAChR-/- mice. We first evaluated the expression of α7-nAChR by immunohistochemistry (IHC) in liver sections. IHC was also performed to assess intrahepatic bile duct mass (IBDM), and Sirius Red staining was performed to quantify the amount of collagen deposition. Immunofluorescence was performed to assess colocalization of α7-nAChR with bile ducts (costained with CK-19) and HSCs (costained with desmin). The mRNA expression of α7-nAChR, Ki-67/PCNA (proliferation), fibrosis genes (TGF-ß1, fibronectin-1, Col1α1, and α-SMA), and inflammatory markers (IL-6, IL-1ß, and TNF-α) was measured by real-time PCR. Biliary TGF-ß1 and hepatic CD68 (Kupffer cell marker) expression was assessed using IHC. α7-nAChR immunoreactivity was observed in both bile ducts and HSCs and increased following BDL. α7-nAChR-/- BDL mice exhibited decreased (i) bile duct mass, liver fibrosis, and inflammation, and (ii) immunoreactivity of TGF-ß1 as well as expression of fibrosis genes compared to WT BDL mice. α7-nAChR activation triggers biliary proliferation and liver fibrosis and may be a therapeutic target in managing extrahepatic biliary obstruction.


Subject(s)
Cholestasis, Extrahepatic/genetics , Liver Cirrhosis/genetics , alpha7 Nicotinic Acetylcholine Receptor/genetics , Animals , Bile Ducts/metabolism , Bile Ducts/pathology , Cell Line, Tumor , Cholestasis, Extrahepatic/complications , Cholestasis, Extrahepatic/metabolism , Cytokines/genetics , Cytokines/metabolism , Humans , Hyperplasia , Ki-67 Antigen/genetics , Ki-67 Antigen/metabolism , Liver Cirrhosis/etiology , Liver Cirrhosis/metabolism , Male , Mice , Mice, Inbred C57BL , Proliferating Cell Nuclear Antigen/genetics , Proliferating Cell Nuclear Antigen/metabolism , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/metabolism , alpha7 Nicotinic Acetylcholine Receptor/metabolism
3.
World J Gastroenterol ; 21(29): 8817-25, 2015 Aug 07.
Article in English | MEDLINE | ID: mdl-26269671

ABSTRACT

AIM: To examine renal expression of organic anion transporter 5 (Oat5) and sodium-dicarboxylate cotransporter 1 (NaDC1), and excretion of citrate in rats with acute extrahepatic cholestasis. METHODS: Obstructive jaundice was induced in rats by double ligation and division of the common bile duct (BDL group). Controls underwent sham operation that consisted of exposure, but not ligation, of the common bile duct (Sham group). Studies were performed 21 h after surgery. During this period, animals were maintained in metabolic cages in order to collect urine. The urinary volume was determined by gravimetry. The day of the experiment, blood samples were withdrawn and used to measure total and direct bilirubin as indicative parameters of hepatic function. Serum and urine samples were used for biochemical determinations. Immunoblotting for Oat5 and NaDC1 were performed in renal homogenates and brush border membranes from Sham and BDL rats. Immunohistochemistry studies were performed in kidneys from both experimental groups. Total RNA was extracted from rat renal tissue in order to perform reverse transcription polymerase chain reaction. Another set of experimental animals were used to evaluate medullar renal blood flow (mRBF) using fluorescent microspheres. RESULTS: Total and direct bilirubin levels were significantly higher in BDL animals, attesting to the adequacy of biliary obstruction. An important increase in mRBF was determined in BDL group (Sham: 0.53 ± 0.12 mL/min per 100 g body weight vs BDL: 1.58 ± 0.24 mL/min per 100 g body weight, P < 0.05). An increase in the urinary volume was observed in BDL animals. An important decrease in urinary levels of citrate was seen in BDL group. Besides, a decrease in urinary citrate excretion (Sham: 0.53 ± 0.11 g/g creatinine vs BDL: 0.07 ± 0.02 g/g creatinine, P < 0.05) and an increase in urinary excretion of H(+) (Sham: 0.082 ± 0.03 µmol/g creatinine vs BDL: 0.21 ± 0.04 µmol/g creatinine, P < 0.05) were observed in BDL animals. We found upregulations of both proteins Oat5 and NaDC1 in brush border membranes where they are functional. Immunohistochemistry technique corroborated these results for both proteins. No modifications were observed in Oat5 mRNA and in NaDC1 mRNA levels in kidney from BDL group as compared with Sham ones. CONCLUSION: Citrate excretion is decreased in BDL rats, at least in part, because of the higher NaDC1 expression. Using the outward gradient of citrate generated by NaDC1, Oat5 can reabsorb/eliminate different organic anions of pathophysiological importance.


Subject(s)
Cholestasis, Extrahepatic/metabolism , Dicarboxylic Acid Transporters/metabolism , Jaundice, Obstructive/metabolism , Kidney/metabolism , Organic Anion Transporters, Sodium-Dependent/metabolism , Symporters/metabolism , Animals , Bilirubin/blood , Biomarkers/blood , Biomarkers/urine , Cholestasis, Extrahepatic/blood , Cholestasis, Extrahepatic/genetics , Cholestasis, Extrahepatic/urine , Citric Acid/urine , Common Bile Duct/surgery , Dicarboxylic Acid Transporters/genetics , Disease Models, Animal , Jaundice, Obstructive/blood , Jaundice, Obstructive/genetics , Jaundice, Obstructive/urine , Ligation , Male , Organic Anion Transporters, Sodium-Dependent/genetics , Rats, Wistar , Renal Circulation , Renal Elimination , Symporters/genetics , Time Factors , Up-Regulation
4.
J Pathol ; 237(3): 343-54, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26108453

ABSTRACT

Keratins (K) are cytoprotective proteins and keratin mutations predispose to the development of multiple human diseases. K19 represents the most widely used marker of biliary and hepatic progenitor cells as well as a marker of ductular reaction that constitutes the basic regenerative response to chronic liver injury. In the present study, we investigated the role of K19 in biliary and hepatic progenitor cells and its importance for ductular reaction. K19 wild-type (WT) and knockout (KO) mice were fed: (a) 3,5-diethoxycarbonyl-1,4-dihydrocollidine (DDC); (b) cholic acid (CA); (c) a choline-deficient, ethionine-supplemented (CDE) diet; or (d) were subjected to common bile duct ligation (CBDL). The bile composition, liver damage, bile duct proliferation, oval cell content and biliary fibrosis were analysed. In untreated animals, loss of K19 led to redistribution of the K network in biliary epithelial cells (BECs) but to no obvious biliary phenotype. After DDC feeding, K19 KO mice exhibited (compared to WTs): (a) increased cholestasis; (b) less pronounced ductular reaction with reduced ductular proliferation and fewer oval cells; (c) impaired Notch 2 signalling in BECs; (d) lower biliary fibrosis score and biliary bicarbonate concentration. An attenuated oval cell proliferation in K19 KOs was also found after feeding with the CDE diet. K19 KOs subjected to CBDL displayed lower BEC proliferation, oval cell content and less prominent Notch 2 signal. K19 deficiency did not change the extent of CA- or CBDL-induced liver injury and fibrosis. Our results demonstrate that K19 plays an important role in the ductular reaction and might be of importance in multiple chronic liver disorders that frequently display a ductular reaction.


Subject(s)
Chemical and Drug Induced Liver Injury/metabolism , Cholangitis, Sclerosing/metabolism , Cholestasis, Extrahepatic/metabolism , Common Bile Duct/metabolism , Epithelial Cells/metabolism , Keratin-19/deficiency , Liver Cirrhosis, Biliary/metabolism , Liver/metabolism , Stem Cells/metabolism , Animals , Cell Proliferation , Chemical and Drug Induced Liver Injury/etiology , Chemical and Drug Induced Liver Injury/genetics , Chemical and Drug Induced Liver Injury/pathology , Cholangitis, Sclerosing/chemically induced , Cholangitis, Sclerosing/genetics , Cholangitis, Sclerosing/pathology , Cholestasis, Extrahepatic/etiology , Cholestasis, Extrahepatic/genetics , Cholestasis, Extrahepatic/pathology , Cholic Acid , Choline Deficiency/complications , Common Bile Duct/pathology , Common Bile Duct/surgery , Disease Models, Animal , Epithelial Cells/pathology , Ethionine , Keratin-19/genetics , Ligation , Liver/pathology , Liver Cirrhosis, Biliary/chemically induced , Liver Cirrhosis, Biliary/genetics , Liver Cirrhosis, Biliary/pathology , Liver Regeneration , Male , Mice, Knockout , Phenotype , Pyridines , Signal Transduction , Stem Cells/pathology , Time Factors
5.
J Clin Invest ; 125(5): 1886-900, 2015 May.
Article in English | MEDLINE | ID: mdl-25822023

ABSTRACT

Liver cholestatic diseases, which stem from diverse etiologies, result in liver toxicity and fibrosis and may progress to cirrhosis and liver failure. We show that CCN1 (also known as CYR61), a matricellular protein that dampens and resolves liver fibrosis, also mediates cholangiocyte proliferation and ductular reaction, which are repair responses to cholestatic injury. In cholangiocytes, CCN1 activated NF-κB through integrin αvß5/αvß3, leading to Jag1 expression, JAG1/NOTCH signaling, and cholangiocyte proliferation. CCN1 also induced Jag1 expression in hepatic stellate cells, whereupon they interacted with hepatic progenitor cells to promote their differentiation into cholangiocytes. Administration of CCN1 protein or soluble JAG1 induced cholangiocyte proliferation in mice, which was blocked by inhibitors of NF-κB or NOTCH signaling. Knock-in mice expressing a CCN1 mutant that is unable to bind αvß5/αvß3 were impaired in ductular reaction, leading to massive hepatic necrosis and mortality after bile duct ligation (BDL), whereas treatment of these mice with soluble JAG1 rescued ductular reaction and reduced hepatic necrosis and mortality. Blockade of integrin αvß5/αvß3, NF-κB, or NOTCH signaling in WT mice also resulted in defective ductular reaction after BDL. These findings demonstrate that CCN1 induces cholangiocyte proliferation and ductular reaction and identify CCN1/αvß5/NF-κB/JAG1 as a critical axis for biliary injury repair.


Subject(s)
Bile Ducts/metabolism , Cysteine-Rich Protein 61/physiology , Liver/metabolism , NF-kappa B/metabolism , Receptors, Vitronectin/physiology , Animals , Bile Ducts/physiology , Calcium-Binding Proteins/biosynthesis , Calcium-Binding Proteins/genetics , Calcium-Binding Proteins/pharmacology , Calcium-Binding Proteins/therapeutic use , Cell Division , Cells, Cultured , Cholestasis, Extrahepatic/genetics , Cholestasis, Extrahepatic/metabolism , Cholestasis, Extrahepatic/pathology , Cysteine-Rich Protein 61/genetics , Cysteine-Rich Protein 61/pharmacology , Gene Expression Regulation , Gene Knock-In Techniques , Hepatic Stellate Cells/metabolism , Hepatocytes/metabolism , Humans , Integrin alphaVbeta3 , Intercellular Signaling Peptides and Proteins/biosynthesis , Intercellular Signaling Peptides and Proteins/genetics , Intercellular Signaling Peptides and Proteins/pharmacology , Intercellular Signaling Peptides and Proteins/therapeutic use , Jagged-1 Protein , Membrane Proteins/biosynthesis , Membrane Proteins/genetics , Membrane Proteins/pharmacology , Membrane Proteins/therapeutic use , Mice , Mice, Inbred C57BL , RNA Interference , Receptors, Notch/physiology , Recombinant Fusion Proteins/metabolism , Regeneration , Serrate-Jagged Proteins
6.
Am J Physiol Gastrointest Liver Physiol ; 308(8): G691-701, 2015 Apr 15.
Article in English | MEDLINE | ID: mdl-25678505

ABSTRACT

Cholangiocyte proliferation is regulated in a coordinated fashion by many neuroendocrine factors through autocrine and paracrine mechanisms. The renin-angiotensin system (RAS) is known to play a role in the activation of hepatic stellate cells and blocking the RAS attenuates hepatic fibrosis. We investigated the role of the RAS during extrahepatic cholestasis induced by bile duct ligation (BDL). In this study, we used normal and BDL rats that were treated with control, angiotensin II (ANG II), or losartan for 2 wk. In vitro studies were performed in a primary rat cholangiocyte cell line (NRIC). The expression of renin, angiotensin-converting enzyme, angiotensinogen, and angiotensin receptor type 1 was evaluated by immunohistochemistry (IHC), real-time PCR, and FACs and found to be increased in BDL compared with normal rat. The levels of ANG II were evaluated by ELISA and found to be increased in serum and conditioned media of cholangiocytes from BDL compared with normal rats. Treatment with ANG II increased biliary mass and proliferation in both normal and BDL rats. Losartan attenuated BDL-induced biliary proliferation. In vitro, ANG II stimulated NRIC proliferation via increased intracellular cAMP levels and activation of the PKA/ERK/CREB intracellular signaling pathway. ANG II stimulated a significant increase in Sirius red staining and IHC for fibronectin that was blocked by angiotensin receptor blockade. In vitro, ANG II stimulated the gene expression of collagen 1A1, fibronectin 1, and IL-6. These results indicate that cholangiocytes express a local RAS and that ANG II plays an important role in regulating biliary proliferation and fibrosis during extraheptic cholestasis.


Subject(s)
Angiotensin II/pharmacology , Bile Ducts, Extrahepatic/drug effects , Bile Ducts, Extrahepatic/surgery , Cell Proliferation/drug effects , Cholestasis, Extrahepatic/etiology , Cholestasis, Extrahepatic/metabolism , Renin-Angiotensin System/drug effects , Angiotensin II Type 1 Receptor Blockers/pharmacology , Animals , Bile Ducts, Extrahepatic/pathology , Cell Line , Cholestasis, Extrahepatic/genetics , Cholestasis, Extrahepatic/pathology , Cholestasis, Extrahepatic/prevention & control , Disease Models, Animal , Fibrosis , Gene Expression Regulation , Hyperplasia , Ligation , Losartan/pharmacology , Male , Rats, Inbred F344 , Renin-Angiotensin System/genetics , Signal Transduction/drug effects
7.
Sci Transl Med ; 6(247): 247ra100, 2014 Jul 30.
Article in English | MEDLINE | ID: mdl-25080475

ABSTRACT

Hepatic accumulation of bile acids is central to the pathogenesis of cholestatic liver diseases. Endocrine hormone fibroblast growth factor 19 (FGF19) may reduce hepatic bile acid levels through modulation of bile acid synthesis and prevent subsequent liver damage. However, FGF19 has also been implicated in hepatocellular carcinogenesis, and consequently, the potential risk from prolonged exposure to supraphysiological levels of the hormone represents a major hurdle for developing an FGF19-based therapy. We describe a nontumorigenic FGF19 variant, M70, which regulates bile acid metabolism and, through inhibition of bile acid synthesis and reduction of excess hepatic bile acid accumulation, protects mice from liver injury induced by either extrahepatic or intrahepatic cholestasis. Administration of M70 in healthy human volunteers potently reduces serum levels of 7α-hydroxy-4-cholesten-3-one, a surrogate marker for the hepatic activity of cholesterol 7α-hydroxylase (CYP7A1), the enzyme responsible for catalyzing the first and rate-limiting step in the classical bile acid synthetic pathway. This study provides direct evidence for the regulation of bile acid metabolism by FGF19 pathway in humans. On the basis of these results, the development of nontumorigenic FGF19 variants capable of modulating CYP7A1 expression represents an effective approach for the prevention and treatment of cholestatic liver diseases as well as potentially for other disorders associated with bile acid dysregulation.


Subject(s)
Bile Acids and Salts/metabolism , Cholagogues and Choleretics/therapeutic use , Cholestasis, Extrahepatic/drug therapy , Cholestasis, Intrahepatic/drug therapy , Fibroblast Growth Factors/therapeutic use , Liver/drug effects , Adult , Animals , Australia , Biomarkers/blood , Cholagogues and Choleretics/adverse effects , Cholagogues and Choleretics/pharmacokinetics , Cholestasis, Extrahepatic/genetics , Cholestasis, Extrahepatic/metabolism , Cholestasis, Extrahepatic/pathology , Cholestasis, Intrahepatic/genetics , Cholestasis, Intrahepatic/metabolism , Cholestasis, Intrahepatic/pathology , Cholestenones/blood , Cholesterol 7-alpha-Hydroxylase/genetics , Cholesterol 7-alpha-Hydroxylase/metabolism , Disease Models, Animal , Double-Blind Method , Down-Regulation , Fibroblast Growth Factors/adverse effects , Fibroblast Growth Factors/biosynthesis , Fibroblast Growth Factors/genetics , Fibroblast Growth Factors/pharmacokinetics , Gene Expression Regulation, Enzymologic , Gene Transfer Techniques , Genetic Variation , Healthy Volunteers , Humans , Liver/metabolism , Liver/pathology , Liver Neoplasms/genetics , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Male , Mice , Middle Aged , RNA, Messenger/metabolism , Recombinant Proteins/therapeutic use , Risk Assessment , Young Adult
8.
Liver Int ; 32(9): 1342-53, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22698069

ABSTRACT

BACKGROUND AND AIMS: CCN3/NOV, a matricellular protein of the CYR61-CTGF-NOV (CCN) family, comprises six secreted proteins that associate specifically with the extracellular matrix. CCN proteins lack specific high-affinity receptors; instead, they regulate crucial biological processes, such as fibrosis, by signalling via integrins and proteoglycans. Recent studies have linked overexpression of CCN3/NOV to mitigate kidney fibrosis. This study aims to investigate CCN3/NOV overexpression in liver fibrogenesis in vivo. METHODS: The biological efficacy of adenoviral expressed CCN3/NOV directed under transcriptional control of the constitutively active Cytomegalovirus promoter (Ad-NOV) was analysed in a bile duct ligation model and in cultured primary hepatocytes. RESULTS AND CONCLUSIONS: Even though Ad-NOV gene transfer in a 3-week bile duct ligation mouse model showed the expected high levels of CCN3/NOV in both mRNA and protein, it failed to reduce liver fibrogenesis, but instead enhanced hepatocyte apoptosis. Furthermore, overexpressed CCN3/NOV in cultured primary hepatocytes resulted in decreased levels of CCN2/CTGF, the profibrotic marker protein in liver fibrosis. Both Ad-NOV and Ad-CTGF induced reactive oxygen species production, enhanced p38 and JNK activation. Therefore, we conclude that CCN3/NOV overexpression in vivo is insufficient to mitigate liver fibrogenesis because of the induction of hepatocyte injury and apoptosis.


Subject(s)
Adenoviridae/genetics , Apoptosis/physiology , Hepatocytes/pathology , Liver Cirrhosis/pathology , Nephroblastoma Overexpressed Protein/genetics , Animals , Cells, Cultured , Cholestasis, Extrahepatic/complications , Cholestasis, Extrahepatic/genetics , Cholestasis, Extrahepatic/pathology , Common Bile Duct/surgery , Disease Models, Animal , Disease Progression , Gene Expression , Gene Transfer Techniques , Genetic Vectors/genetics , Hepatocytes/metabolism , Ligation , Liver Cirrhosis/etiology , Liver Cirrhosis/genetics , Mice , Mice, Inbred C57BL , Nephroblastoma Overexpressed Protein/metabolism , RNA, Messenger/metabolism , Reactive Oxygen Species/metabolism
9.
Free Radic Biol Med ; 52(9): 1543-51, 2012 May 01.
Article in English | MEDLINE | ID: mdl-22306509

ABSTRACT

Oxidative stress and mitochondrial dysfunction are involved in the pathogenesis of chronic liver cholestasis. Mitochondrial DNA (mtDNA) is highly susceptible to oxidative stress and mtDNA damage leads to mitochondrial dysfunction. This study aimed to investigate the mtDNA alterations that occurred during liver injury in patients with extrahepatic cholestasis. Along with an increase in malondialdehyde (MDA) levels and a decrease in ATP levels, extrahepatic cholestatic patients presented a significant increase in mitochondrial 8-hydroxydeoxyguanosine (8-OHdG) levels and decreases in mtDNA copy number, mtDNA transcript levels, and mtDNA nucleoid structure. In L02 cells, glycochenodeoxycholic acid (GCDCA) induced similar damage to the mtDNA and mitochondria. In line with the mtDNA alterations, the mRNA and protein levels of mitochondrial transcription factor A (TFAM) were significantly decreased both in cholestatic patients and in GCDCA-treated L02 cells. Moreover, overexpression of TFAM could efficiently attenuate the mtDNA damage induced by GCDCA in L02 cells. However, without its C-tail, ΔC-TFAM appeared less effective against the hepatotoxicity of GCDCA than the wild-type TFAM. Overall, our study demonstrates that mtDNA damage is involved in liver damage in extrahepatic cholestatic patients. The mtDNA damage is attributable to the loss of TFAM. TFAM has mtDNA-protective effects against the hepatotoxicity of bile acid during cholestasis.


Subject(s)
Cholestasis, Extrahepatic/genetics , DNA Damage , DNA, Mitochondrial/genetics , DNA-Binding Proteins/physiology , Liver/injuries , Mitochondrial Proteins/physiology , Transcription Factors/physiology , Adenosine Triphosphate/metabolism , Adult , Base Sequence , Blotting, Western , DNA Primers , Enzyme-Linked Immunosorbent Assay , Female , Humans , Male , Middle Aged , Oxidative Stress , Real-Time Polymerase Chain Reaction
10.
Liver Int ; 31(5): 630-41, 2011 May.
Article in English | MEDLINE | ID: mdl-21457436

ABSTRACT

AIM: Neural cell adhesion molecule (N-CAM) is expressed by activated hepatic stellate cells (HSC), portal fibroblasts, cholangiocytes and hepatic progenitor cells during liver injury. Its functional role in liver disease and fibrogenesis is unknown. The aim of this study was to investigate the role of N-CAM in liver fibrogenesis. METHODS: To induce fibrosis, N-CAM knockout mice and wild-type controls were subjected to bile duct ligation (BDL) or repeated carbon tetrachloride (CCl(4) ) injections. Fibrosis was quantified by hydroxyproline, immunhistochemistry staining and image analysis. Protein levels were determined with immunoblotting. HSCs were isolated by ultracentrifugation in a Larcoll gradient and thereafter in vitro stimulated with recombinant transforming growth factor (TGF)-ß1. RESULTS: Two weeks after BDL, wild-type mice had developed pronounced liver fibrosis while N-CAM-/- mice had less such alterations. N-CAM-/- mice had less deposition of collagen and fibronectin seen in immunhistochemistry. The protein levels of fibronectin were higher in the liver from the wild type, while laminin were unaltered. CCl(4) -treated N-CAM-/- and wild-type mice showed no significant difference in the extent of liver fibrosis or the expression levels of the above-mentioned genes. HSC isolated from N-CAM-/- mice showed declined levels of smooth muscle actin and desmin after stimulation in vitro with TGF-ß1. CONCLUSIONS: Loss of N-CAM results in decreased hepatic collagen and fibronectin deposition in mice subjected to BDL, but not in animals exposed to repeated CCl(4) injections. HSC isolated from N-CAM null mice show impaired activation in vitro. This indicates a role of N-CAM in cholestatic liver disease and HSC activation.


Subject(s)
Cholestasis, Extrahepatic/complications , Common Bile Duct/surgery , Hepatic Stellate Cells/metabolism , Liver Cirrhosis, Experimental/prevention & control , Liver/metabolism , Neural Cell Adhesion Molecules/deficiency , Actins/metabolism , Animals , Blotting, Western , Carbon Tetrachloride , Cell Separation , Cells, Cultured , Cholestasis, Extrahepatic/etiology , Cholestasis, Extrahepatic/genetics , Cholestasis, Extrahepatic/metabolism , Cholestasis, Extrahepatic/pathology , Collagen/metabolism , Desmin/metabolism , Fibronectins/metabolism , Hepatic Stellate Cells/pathology , Hydroxyproline/metabolism , Immunohistochemistry , Laminin/metabolism , Ligation , Liver/pathology , Liver Cirrhosis, Experimental/etiology , Liver Cirrhosis, Experimental/genetics , Liver Cirrhosis, Experimental/metabolism , Liver Cirrhosis, Experimental/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Neural Cell Adhesion Molecules/genetics , Time Factors , Transforming Growth Factor beta1/metabolism
11.
Biochem Pharmacol ; 80(11): 1718-26, 2010 Dec 01.
Article in English | MEDLINE | ID: mdl-20816672

ABSTRACT

Liver fibrosis is a consequence of chronic liver disorders which lead to the accumulation of extracellular matrix (ECM). Particularly, there is an increased accumulation of collagen in the fibrotic liver. We have therefore used a triplex forming oligonucleotide (TFO) against the type α1(I) collagen and evaluated, whether it can attenuate liver fibrosis induced by common bile duct ligation (CBDL) in rats. There was a significant decrease in hydroxyproline levels and Masson's trichrome staining for collagen in TFO-treated CBDL groups compared to non-treated CBDL group. There was over expression of type α1(I) collagen, α-smooth muscle actin (α-SMA) and TGF-ß1 expression in the CBDL group compared to TFO-treated CBDL group. Also, the serum alanine transaminase (ALT) and aspartate transaminase (AST) concentrations were less in the TFO treated group compared to non-treated CBDL group. There was also less neutrophils accumulation in TFO treated CBDL group assayed by myeloperoxidase (MPO) assay. These results suggests that TFO can be used to downregulate type 1 collagen gene expression and can alleviate liver fibrosis induced by common bile duct ligation.


Subject(s)
Cholestasis, Extrahepatic/prevention & control , Collagen Type I/genetics , Common Bile Duct/metabolism , DNA/pharmacology , Liver Cirrhosis/prevention & control , Oligonucleotides/pharmacology , Animals , Cell Line, Transformed , Cholestasis, Extrahepatic/genetics , Cholestasis, Extrahepatic/pathology , Collagen Type I/antagonists & inhibitors , Collagen Type I/biosynthesis , Common Bile Duct/pathology , DNA/chemistry , DNA/therapeutic use , Ligation , Liver Cirrhosis/genetics , Liver Cirrhosis/pathology , Male , Nucleic Acid Conformation , Oligonucleotides/chemistry , Oligonucleotides/genetics , Rats , Rats, Sprague-Dawley
12.
Hepatology ; 52(1): 204-14, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20578263

ABSTRACT

UNLABELLED: During bile duct ligation (BDL), the growth of large cholangiocytes is regulated by the cyclic adenosine monophosphate (cAMP)/extracellular signal-regulated kinase 1/2 (ERK1/2) pathway and is closely associated with increased secretin receptor (SR) expression. Although it has been suggested that SR modulates cholangiocyte growth, direct evidence for secretin-dependent proliferation is lacking. SR wild-type (WT) (SR(+/+)) or SR knockout (SR(-/-)) mice underwent sham surgery or BDL for 3 or 7 days. We evaluated SR expression, cholangiocyte proliferation, and apoptosis in liver sections and proliferating cell nuclear antigen (PCNA) protein expression and ERK1/2 phosphorylation in purified large cholangiocytes from WT and SR(-/-) BDL mice. Normal WT mice were treated with secretin (2.5 nmoles/kg/day by way of osmotic minipumps for 1 week), and biliary mass was evaluated. Small and large cholangiocytes were used to evaluate the in vitro effect of secretin (100 nM) on proliferation, protein kinase A (PKA) activity, and ERK1/2 phosphorylation. SR expression was also stably knocked down by short hairpin RNA, and basal and secretin-stimulated cAMP levels (a functional index of biliary growth) and proliferation were determined. SR was expressed by large cholangiocytes. Knockout of SR significantly decreased large cholangiocyte growth induced by BDL, which was associated with enhanced apoptosis. PCNA expression and ERK1/2 phosphorylation were decreased in large cholangiocytes from SR(-/-) BDL compared with WT BDL mice. In vivo administration of secretin to normal WT mice increased ductal mass. In vitro, secretin increased proliferation, PKA activity, and ERK1/2 phosphorylation of large cholangiocytes that was blocked by PKA and mitogen-activated protein kinase kinase inhibitors. Stable knockdown of SR expression reduced basal cholangiocyte proliferation. SR is an important trophic regulator sustaining biliary growth. CONCLUSION: The current study provides strong support for the potential use of secretin as a therapy for ductopenic liver diseases.


Subject(s)
Bile Ducts/pathology , Cholestasis, Extrahepatic/complications , Liver Diseases/etiology , Liver/pathology , Receptors, G-Protein-Coupled/physiology , Receptors, Gastrointestinal Hormone/physiology , Animals , Apoptosis , Bile Ducts/drug effects , Cell Proliferation , Cholestasis, Extrahepatic/genetics , Cholestasis, Extrahepatic/pathology , Gene Knockout Techniques , Hyperplasia/genetics , Hyperplasia/pathology , Liver/drug effects , Liver Diseases/pathology , Mice , Mice, Knockout , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Organ Size , Phosphorylation , Proliferating Cell Nuclear Antigen/metabolism , Receptors, G-Protein-Coupled/genetics , Receptors, Gastrointestinal Hormone/genetics , Secretin/pharmacology
13.
Gastroenterology ; 137(1): 297-308, 308.e1-4, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19208365

ABSTRACT

BACKGROUND & AIMS: The hepatocyte growth factor (HGF)/mesenchymal-epithelial transition factor (c-Met) system is an essential inducer of hepatocyte growth and proliferation. Although a fundamental role for the HGF receptor c-Met has been shown in acute liver regeneration, its cell-specific role in hepatocytes during chronic liver injury and fibrosis progression has not been determined. METHODS: Hepatocyte-specific c-Met knockout mice (c-Met(Delta hepa)) using the Cre-loxP system were studied in a bile duct ligation (BDL) model. Microarray analyses were performed to define HGF/c-Met-dependent gene expression. RESULTS: Two strategies for c-Met deletion in hepatocytes to generate hepatocyte-specific c-Met knockout mice were tested. Early deletion during embryonic development was lethal, whereas post-natal Cre expression was successful, leading to the generation of viable c-Met(Delta hepa) mice. BDL in these mice resulted in extensive necrosis and lower proliferation rates of hepatocytes. Gene array analysis of c-Met(Delta hepa) mice revealed a significant reduction of anti-apoptotic genes in c-Met-deleted hepatocytes. These findings could be tested functionally because c-Met(Delta hepa) mice showed a stronger apoptotic response after BDL and Jo-2 stimulation. The phenotype was associated with increased expression of proinflammatory cytokines (tumor necrosis factor-alpha and interleukin-6) and an enhanced recruitment of neutrophils. Activation of these mechanisms triggered a stronger profibrogenic response as evidenced by increased transforming growth factor-beta(1), alpha-smooth muscle actin, collagen-1alpha messenger RNA expression, and enhanced collagen-fiber staining in c-Met(Delta hepa) mice. CONCLUSIONS: Our results show that deletion of c-Met in hepatocytes leads to more liver cell damage and fibrosis in a chronic cholestatic liver injury model because c-Met triggers survival signals important for hepatocyte recovery.


Subject(s)
Apoptosis , Cholestasis, Extrahepatic/complications , Liver Cirrhosis/prevention & control , Liver/metabolism , Proto-Oncogene Proteins c-met/metabolism , Animals , Apoptosis/genetics , Cell Proliferation , Cholestasis, Extrahepatic/genetics , Cholestasis, Extrahepatic/metabolism , Cholestasis, Extrahepatic/pathology , Chronic Disease , Common Bile Duct/surgery , Cytokines/metabolism , Disease Models, Animal , Disease Progression , Extracellular Matrix Proteins/metabolism , Gene Expression Profiling/methods , Hepatic Stellate Cells/metabolism , Hepatic Stellate Cells/pathology , Hepatitis/metabolism , Hepatitis/pathology , Hepatocyte Growth Factor/metabolism , Hepatocytes/metabolism , Hepatocytes/pathology , Inflammation Mediators/metabolism , Ligation , Liver/pathology , Liver Cirrhosis/genetics , Liver Cirrhosis/metabolism , Liver Cirrhosis/pathology , Mice , Mice, Knockout , Necrosis , Neutrophil Infiltration , Oligonucleotide Array Sequence Analysis , Proto-Oncogene Proteins c-met/deficiency , Proto-Oncogene Proteins c-met/genetics , Time Factors
14.
Am J Physiol Gastrointest Liver Physiol ; 296(3): G582-92, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19136383

ABSTRACT

Liver fibrosis is characterized by excessive deposition of extracellular matrix in the liver during chronic injury. During early stages of this disease, cells begin to synthesize and secrete profibrotic proteins that stimulate matrix production and inhibit matrix degradation. Although it is clear that these proteins are important for development of fibrosis, what remains unknown is the mechanism by which chronic liver injury stimulates their production. In the present study, the hypothesis was tested that hypoxia-inducible factor-1alpha (HIF-1alpha) is activated in the liver during chronic injury and regulates expression of profibrotic proteins. To investigate this hypothesis, mice were subjected to bile duct ligation (BDL), an animal model of liver fibrosis. HIF-1alpha protein was increased in the livers of mice subjected to BDL by 3 days after surgery. To test the hypothesis that HIF-1alpha is required for the development of fibrosis, control and HIF-1alpha-deficient mice were subjected to BDL. Levels of type I collagen and alpha-smooth muscle actin mRNA and protein were increased in control mice by 14 days after BDL. These levels were significantly reduced in HIF-1alpha-deficient mice. Next, the levels of several profibrotic mediators were measured to elucidate the mechanism by which HIF-1alpha promotes liver fibrosis. Platelet-derived growth factor (PDGF)-A, PDGF-B, and plasminogen activator inhibitor-1 mRNA levels were increased to a greater extent in control mice subjected to BDL compared with HIF-1alpha-deficient mice at 7 and 14 days after BDL. Results from these studies suggest that HIF-1alpha is a critical regulator of profibrotic mediator production during the development of liver fibrosis.


Subject(s)
Cholestasis, Extrahepatic/genetics , Cholestasis, Extrahepatic/physiopathology , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Liver Cirrhosis/genetics , Liver Cirrhosis/physiopathology , Actins/metabolism , Animals , Bile Ducts , Cell Division/physiology , Cholestasis, Extrahepatic/pathology , Collagen Type I/metabolism , Disease Models, Animal , Fibroblast Growth Factor 2/genetics , Gene Expression/physiology , Hypoxia/genetics , Hypoxia/pathology , Hypoxia/physiopathology , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Ligation , Liver Cirrhosis/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Platelet-Derived Growth Factor/genetics , Serpin E2 , Serpins/genetics
15.
Med Klin (Munich) ; 98(9): 499-509, 2003 Sep 15.
Article in German | MEDLINE | ID: mdl-14551707

ABSTRACT

BACKGROUND: Cholestasis is known as an etiologically diverse clinical entity which requires a broad differential diagnostic workup. In the majority of patients, history, clinical examination, clinical chemical analysis, and abdominal ultrasound enable the differentiation between extrahepatic and intrahepatic cholestasis. This review summarizes our current knowledge in the diagnosis of cholestatic disorders. METHODS: In regard to clinical practice, diagnostic tools and new developments in imaging and molecular genetics are discussed including an algorithm for the diagnostic workup of cholestatic patients. CONCLUSION: Ultrasound and computed tomography have represented the most important primary imaging techniques in hepatobiliary disorders over the last 2 decades. The direct visualization either by endoscopic retrograde cholangiopancreatography (ERCP) or percutaneous transhepatic cholangiography (PTC) still remains the gold standard in the evaluation of the extrahepatic bile duct. In the past decade, magnetic resonance cholangiopancreatography (MRCP) has increasingly been established as a noninvasive alternative, thereby reducing the necessity of ERCP as an invasive exploration of the biliary system. Liver biopsy is indicated for the histologic grading and staging of intrahepatic cholestatic disorders. Recently, molecular genetic studies have elucidated several mutations in genes of hepatobiliary transporters which are responsible for hereditary forms of cholestasis in man. Thus, molecular genetics may be of interest in single cases of unclassified cholestasis or familial syndromes and will contribute to the routine diagnosis of hereditary cholestatic syndromes in the future. In summary, application of these diagnostic tools will finally lead to an unequivocal diagnosis in the majority of cholestatic patients with consecutive rational therapy.


Subject(s)
Cholestasis/diagnosis , Biopsy , Cholangiography , Cholangiopancreatography, Endoscopic Retrograde , Cholestasis/diagnostic imaging , Cholestasis/genetics , Cholestasis/pathology , Cholestasis, Extrahepatic/diagnosis , Cholestasis, Extrahepatic/diagnostic imaging , Cholestasis, Extrahepatic/genetics , Cholestasis, Intrahepatic/diagnosis , Cholestasis, Intrahepatic/diagnostic imaging , Cholestasis, Intrahepatic/genetics , Diagnosis, Differential , Humans , Jaundice, Obstructive/diagnosis , Jaundice, Obstructive/diagnostic imaging , Jaundice, Obstructive/genetics , Liver/pathology , Magnetic Resonance Imaging , Mutation , Tomography, X-Ray Computed , Ultrasonography
16.
Gastrointest Endosc ; 55(7): 883-8, 2002 Jun.
Article in English | MEDLINE | ID: mdl-12024145

ABSTRACT

BACKGROUND: K-ras mutations in endobiliary brush cytology are an early event in carcinogenesis and justify a suspicion of malignancy in patients with extrahepatic biliary stenosis. However, K-ras mutations have been detected in specimens obtained by brushing of clinically benign extrahepatic biliary stenosis. The aim of this study was to determine whether these findings represent an early or false-positive diagnosis of cancer. METHODS: Cytologic specimens were obtained by brushing in 312 consecutive patients with extrahepatic biliary stenosis. Mutations in the K-ras oncogene were detected by an enriched polymerase chain reaction and allele-specific oligonucleotide hybridization assay. Eight patients with a K-ras mutation and a clinically benign extrahepatic biliary stenosis were followed. RESULTS: After a median follow-up of 65 months, 6 of the 8 patients were alive without evidence of malignancy. One patient died of congestive heart failure. The other patient died 60 months after the specimen was obtained, possibly because of chronic pancreatitis, although previously there had been suspicion of malignancy. Biopsy specimens from the papilla were negative for neoplasia and the K-ras analysis harbored the same mutation as previously found in the brush specimen. CONCLUSION: Based on long-term follow-up, the K-ras mutations in all 8 patients with a clinically benign extrahepatic biliary stenosis must be considered as confirmed false-positives. Nevertheless, a false-positive result is infrequent. Therefore, patients with a positive K-ras mutation in biliary cytologic specimens obtained by brushing still require careful, continuing follow-up.


Subject(s)
Cholestasis, Extrahepatic/genetics , Cholestasis, Extrahepatic/pathology , Genes, ras/genetics , Mutation/genetics , Cholestasis, Extrahepatic/mortality , False Positive Reactions , Follow-Up Studies , Humans , Microvilli/genetics , Microvilli/pathology , Outcome Assessment, Health Care , Predictive Value of Tests , Reproducibility of Results , Survival Rate , Time Factors
17.
Biochim Biophys Acta ; 1586(2): 129-45, 2002 Mar 16.
Article in English | MEDLINE | ID: mdl-11959455

ABSTRACT

Bile formation, the exocrine function of the liver, represents a process that is unique to the hepatocyte as a polarized epithelial cell. The generation of bile flow is an osmotic process and largely depends on solute secretion by primary active transporters in the apical membrane of the hepatocyte. In recent years an impressive progress has been made in the discovery of these proteins, most of which belong to the family of ABC transporters. The number of identified ABC transporter genes has been exponentially increasing and the mammalian subfamily now counts at least 52. This development has been of crucial importance for the elucidation of the mechanism of bile formation, and it is therefore not surprising that the development in this field has run in parallel with the discovery of the ABC genes. With the identification of these transporter genes, the background of a number of inherited diseases, which are caused by mutations in these solute pumps, has now been elucidated. We now know that at least six primary active transporters are involved in canalicular secretion of biliary components (MDR1, MDR3, BSEP, MRP2, BCRP and FIC1). Four of these transporter genes are associated with inherited diseases. In this minireview we will shortly describe our present understanding of bile formation and the associated inherited defects.


Subject(s)
ATP-Binding Cassette Transporters/genetics , Bile Canaliculi/metabolism , Animals , Bile Acids and Salts/metabolism , Cholestasis, Extrahepatic/complications , Cholestasis, Extrahepatic/genetics , Cholestasis, Extrahepatic/metabolism , Cholestasis, Intrahepatic/complications , Cholestasis, Intrahepatic/genetics , Cholestasis, Intrahepatic/metabolism , Cholesterol/blood , Female , Humans , Jaundice, Chronic Idiopathic/genetics , Jaundice, Chronic Idiopathic/metabolism , Lipid Metabolism , Mutation , Pregnancy , Pregnancy Complications/metabolism
18.
Gut ; 47(3): 357-61, 2000 Sep.
Article in English | MEDLINE | ID: mdl-10940271

ABSTRACT

BACKGROUND AND AIM: The present study was undertaken to determine if detection of Ki-ras gene point mutations in bile specimens could differentiate between benign and malignant biliary strictures. PATIENTS: Bile specimens were obtained from 117 patients exhibiting a stricture of the main bile duct, the nature of which was assessed by cholangiography, histology, and follow up. METHODS: DNA from frozen bile specimens was extracted, amplified, and tested for codon 12 point mutations of Ki-ras gene using sequence specific oligonucleotide hybridisation and mutant allele specific amplification. RESULTS: DNA amplification was successful in 110/117 bile specimens (94%). Detection of Ki-ras gene mutations in bile specimens was positive in 24.4% (22/90) of patients with malignant strictures, in 31.4% (22/70) when only primary malignant tumours were considered, and in 4% (1/25) of patients with benign strictures. Of the 49 patients with histological specimens obtained before surgery, the sensitivity of histology, Ki-ras mutation analysis, and combined methods was 59.2%, 28.6%, and 73.5% respectively. CONCLUSIONS: Our study showed that Ki-ras mutations may be detected in about one third of bile specimens from patients with primary tumours invading the main bile duct. Detection of such mutations appears to be specific and may help to differentiate between benign and malignant biliary strictures.


Subject(s)
Bile Duct Neoplasms/genetics , Cholestasis, Extrahepatic/genetics , Genes, ras/genetics , Point Mutation/genetics , Bile , Bile Duct Neoplasms/diagnosis , Cholestasis, Extrahepatic/diagnosis , DNA/analysis , Diagnosis, Differential , Humans , Oligonucleotide Array Sequence Analysis , Polymerase Chain Reaction/methods , Predictive Value of Tests , Sensitivity and Specificity
19.
Biochem Mol Biol Int ; 44(3): 443-52, 1998 Mar.
Article in English | MEDLINE | ID: mdl-9556204

ABSTRACT

The hepatic canalicular membrane has transporters that play an important role as efflux pumps in the excretion of endogenous bile constituents or xenobiotics into bile canaliculi. To elucidate functional significance of canalicular transporters in the mechanism of cholestasis, mRNA expression levels of multidrug resistance (mdr) 1b, mdr2 and canalicular multispecific organic anion transporter (cMOAT) genes were analyzed by Southern blotting of reverse-transcribed PCR products of liver mRNA obtained from cholestatic rats that had been subjected to bile duct ligation. Both mdr1b and mdr2 mRNA expression increased after ligation. Immunohistochemical study revealed that the products of both mdr1b and mdr2 were present on the canaliculi, and that their levels increased after ligation. In contrast, cMOAT mRNA expression was not increased, but rather attenuated by ligation. The expression of canalicular transporters was differentially regulated in extrahepatic biliary obstruction, indicating the different roles are played by mdr and cMOAT in the pathogenesis of cholestasis.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/genetics , ATP Binding Cassette Transporter, Subfamily B , ATP-Binding Cassette Transporters/genetics , Carrier Proteins/genetics , Cholestasis, Extrahepatic/genetics , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , ATP-Binding Cassette Transporters/metabolism , Animals , Anion Transport Proteins , Bile Ducts/surgery , Bilirubin/blood , Blotting, Northern , Blotting, Southern , Cholestasis, Extrahepatic/metabolism , Disease Models, Animal , Immunohistochemistry , Ligation , Liver/metabolism , Male , Polymerase Chain Reaction/methods , Rats , Rats, Wistar
SELECTION OF CITATIONS
SEARCH DETAIL
...