Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 4 de 4
Filter
Add more filters










Database
Language
Publication year range
1.
J Hepatol ; 63(4): 952-61, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26044126

ABSTRACT

BACKGROUND & AIMS: Polycystic liver diseases (PLDs) are genetic disorders characterized by progressive biliary cystogenesis. Current therapies show short-term and/or modest beneficial effects. Cystic cholangiocytes hyperproliferate as a consequence of diminished intracellular calcium levels ([Ca(2+)]i). Here, the therapeutic value of ursodeoxycholic acid (UDCA) was investigated. METHODS: Effect of UDCA was examined in vitro and in polycystic (PCK) rats. Hepatic cystogenesis and fibrosis, and the bile acid (BA) content were evaluated from the liver, bile, serum, and kidneys by HPLC-MS/MS. RESULTS: Chronic treatment of PCK rats with UDCA inhibits hepatic cystogenesis and fibrosis, and improves their motor behaviour. As compared to wild-type animals, PCK rats show increased BA concentration ([BA]) in liver, similar hepatic Cyp7a1 mRNA levels, and diminished [BA] in bile. Likewise, [BA] is increased in cystic fluid of PLD patients compared to their matched serum levels. In PCK rats, UDCA decreases the intrahepatic accumulation of cytotoxic BA, normalizes their diminished [BA] in bile, increases the BA secretion in bile and diminishes the increased [BA] in kidneys. In vitro, UDCA inhibits the hyperproliferation of polycystic human cholangiocytes via a PI3K/AKT/MEK/ERK1/2-dependent mechanism without affecting apoptosis. Finally, the presence of glycodeoxycholic acid promotes the proliferation of polycystic human cholangiocytes, which is inhibited by both UDCA and tauro-UDCA. CONCLUSIONS: UDCA was able to halt the liver disease of a rat model of PLD through inhibiting cystic cholangiocyte hyperproliferation and decreasing the levels of cytotoxic BA species in the liver, which suggests the use of UDCA as a potential therapeutic tool for PLD patients.


Subject(s)
Apoptosis , Cysts/drug therapy , Liver Diseases/drug therapy , Liver/pathology , Ursodeoxycholic Acid/pharmacology , Animals , Bile Acids and Salts/metabolism , Bile Ducts/metabolism , Bile Ducts/pathology , Calcium/metabolism , Cell Proliferation/drug effects , Cells, Cultured , Cholagogues and Choleretics/pharmacology , Cysts/metabolism , Cysts/pathology , Disease Models, Animal , Liver/drug effects , Liver/metabolism , Liver Diseases/metabolism , Liver Diseases/pathology , Rats , Tandem Mass Spectrometry
2.
Hepatology ; 56(2): 687-97, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22383162

ABSTRACT

UNLABELLED: Cl(-) /HCO3- anion exchanger 2 (AE2) participates in intracellular pH homeostasis and secretin-stimulated biliary bicarbonate secretion. AE2/SLC4A2 gene expression is reduced in liver and blood mononuclear cells from patients with primary biliary cirrhosis (PBC). Our previous findings of hepatic and immunological features mimicking PBC in Ae2-deficient mice strongly suggest that decreased AE2 expression might be involved in the pathogenesis of PBC. Here, we tested the potential role of microRNA 506 (miR-506) - predicted as candidate to target AE2 mRNA - for the decreased expression of AE2 in PBC. Real-time quantitative polymerase chain reaction showed that miR-506 expression is increased in PBC livers versus normal liver specimens. In situ hybridization in liver sections confirmed that miR-506 is up-regulated in the intrahepatic bile ducts of PBC livers, compared with normal and primary sclerosing cholangitis livers. Precursor-mediated overexpression of miR-506 in SV40-immortalized normal human cholangiocytes (H69 cells) led to decreased AE2 protein expression and activity, as indicated by immunoblotting and microfluorimetry, respectively. Moreover, miR-506 overexpression in three-dimensional (3D)-cultured H69 cholangiocytes blocked the secretin-stimulated expansion of cystic structures developed under the 3D conditions. Luciferase assays and site-directed mutagenesis demonstrated that miR-506 specifically may bind the 3'untranslated region (3'UTR) of AE2 messenger RNA (mRNA) and prevent protein translation. Finally, cultured PBC cholangiocytes showed decreased AE2 activity, together with miR-506 overexpression, compared to normal human cholangiocytes, and transfection of PBC cholangiocytes with anti-miR-506 was able to improve their AE2 activity. CONCLUSION: miR-506 is up-regulated in cholangiocytes from PBC patients, binds the 3'UTR region of AE2 mRNA, and prevents protein translation, leading to diminished AE2 activity and impaired biliary secretory functions. In view of the putative pathogenic role of decreased AE2 in PBC, miR-506 may constitute a potential therapeutic target for this disease.


Subject(s)
Anion Transport Proteins/genetics , Anion Transport Proteins/metabolism , Antiporters/genetics , Antiporters/metabolism , Bile Ducts, Intrahepatic/physiopathology , Liver Cirrhosis, Biliary , MicroRNAs/metabolism , Bicarbonates/metabolism , Bile Ducts, Intrahepatic/cytology , Bile Ducts, Intrahepatic/metabolism , Cell Line, Tumor , Chloride-Bicarbonate Antiporters , Chlorides/metabolism , Computer Simulation , Epithelium/physiology , Humans , Liver Cirrhosis, Biliary/genetics , Liver Cirrhosis, Biliary/metabolism , Liver Cirrhosis, Biliary/physiopathology , MicroRNAs/genetics , Primary Cell Culture , Protein Biosynthesis/physiology , RNA, Messenger/metabolism , SLC4A Proteins , Up-Regulation/genetics
3.
PLoS One ; 6(12): e28717, 2011.
Article in English | MEDLINE | ID: mdl-22194894

ABSTRACT

BACKGROUND & AIMS: Secretin induces bicarbonate-rich hydrocholeresis in healthy individuals, but not in untreated patients with primary biliary cirrhosis (PBC). Ursodeoxycholic acid (UDCA)--the first choice treatment for PBC--restores the secretin response. Compared with humans, secretin has poor effect in experimental normal-rat models with biliary drainage, although it may elicit hydrocholeresis when the bile-acid pool is maintained. In view of the benefits of UDCA in PBC, we used normal-rat models to unravel the acute contribution of UDCA (and/or taurine-conjugated TUDCA) for eliciting the biliary secretin response. METHODS: Intravascular and/or intrabiliary administration of agonists and inhibitors was performed in normal rats with biliary monitoring. Secretin/bile-acid interplay was analyzed in 3D cultured rat cholangiocytes that formed expansive cystic structures with intralumenal hydroionic secretion. RESULTS: In vivo, secretin stimulates hydrocholeresis upon UDCA/TUDCA infusion, but does not modify the intrinsic hypercholeretic effect of dehydrocholic acid (DHCA). The former effect is dependent on microtubule polymerization, and involves PKCα, PI3K and MEK pathways, as shown by colchicine (i.p.) and retrograde biliary inhibitors. In vitro, while secretin alone accelerates the spontaneous expansion of 3D-cystic structures, this effect is enhanced in the presence of TUDCA, but not UDCA or DHCA. Experiments with inhibitors and Ca(2+)-chelator confirmed that the synergistic effect of secretin plus TUDCA involves microtubules, intracellular Ca(2+), PKCα, PI3K, PKA and MEK pathways. Gene silencing also demonstrated the involvement of the bicarbonate extruder Ae2. CONCLUSIONS: UDCA is conjugated in order to promote secretin-stimulated hydrocholeresis in rats through Ae2, microtubules, intracellular Ca(2+), PKCα, PI3K, PKA, and MEK.


Subject(s)
Liver Cirrhosis, Biliary/pathology , Secretin/pharmacology , Taurine/pharmacology , Taurochenodeoxycholic Acid/pharmacology , Ursodeoxycholic Acid/pharmacology , Animals , Anion Transport Proteins/metabolism , Antiporters/metabolism , Bile/drug effects , Bile/metabolism , Cells, Cultured , Choledochal Cyst/metabolism , Choledochal Cyst/pathology , Dehydrocholic Acid/pharmacology , Gene Silencing/drug effects , Humans , Liver Cirrhosis, Biliary/enzymology , Male , Microtubules/drug effects , Microtubules/metabolism , Mitogen-Activated Protein Kinase Kinases/metabolism , Models, Biological , Phosphatidylinositol 3-Kinases/metabolism , Polymerization/drug effects , Protein Kinase C/metabolism , Rats , Rats, Wistar , SLC4A Proteins
4.
J Clin Invest ; 118(2): 695-709, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18188457

ABSTRACT

Primary biliary cirrhosis (PBC) is a cholestatic disease associated with autoimmune phenomena and alterations in both biliary bicarbonate excretion and expression of the bicarbonate carrier AE2. The bile acid ursodeoxycholic acid (UCDA) is currently used in treatment of cholestatic liver diseases and is the treatment of choice in PBC; however, a subset of PBC patients respond poorly to UDCA monotherapy. In these patients, a combination of UDCA and glucocorticoid therapy appears to be beneficial. To address the mechanism of this benefit, we analyzed the effects of UDCA and dexamethasone on AE2 gene expression in human liver cells from hepatocyte and cholangiocyte lineages. The combination of UDCA and dexamethasone, but not UDCA or dexamethasone alone, increased the expression of liver-enriched alternative mRNA isoforms AE2b1 and AE2b2 and enhanced AE2 activity. Similar effects were obtained after replacing UDCA with UDCA conjugates. In in vitro and in vivo reporter assays, we found that a UDCA/dexamethasone combination upregulated human AE2 alternate overlapping promoter sequences from which AE2b1 and AE2b2 are expressed. In chromatin immunoprecipitation assays, we demonstrated that combination UCDA/dexamethasone treatment induced p300-related interactions between HNF1 and glucocorticoid receptor on the AE2 alternate promoter. Our data provide a potential molecular explanation for the beneficial effects of the combination of UDCA and glucocorticoids in PBC patients with inadequate response to UDCA monotherapy.


Subject(s)
Anion Transport Proteins/genetics , Antiporters/genetics , Cholagogues and Choleretics/pharmacology , Dexamethasone/pharmacology , Glucocorticoids/pharmacology , Hepatocytes/drug effects , Transcription, Genetic/drug effects , Ursodeoxycholic Acid/pharmacology , Animals , Anion Transport Proteins/analysis , Anion Transport Proteins/metabolism , Antiporters/analysis , Antiporters/metabolism , Cell Line , Cholagogues and Choleretics/therapeutic use , Chromatin Immunoprecipitation , Dexamethasone/therapeutic use , Drug Therapy, Combination , Gene Expression Regulation , Glucocorticoids/therapeutic use , Hepatocyte Nuclear Factor 1-alpha/genetics , Hepatocyte Nuclear Factor 1-alpha/metabolism , Hepatocyte Nuclear Factor 1-beta/metabolism , Hepatocytes/metabolism , Humans , Liver Cirrhosis, Biliary/drug therapy , Mice , Mutation , Promoter Regions, Genetic/drug effects , Promoter Regions, Genetic/genetics , SLC4A Proteins , Up-Regulation , Ursodeoxycholic Acid/therapeutic use , p300-CBP Transcription Factors/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...