Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 4 de 4
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
J Hepatol ; 63(4): 952-61, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26044126

RESUMEN

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.


Asunto(s)
Apoptosis , Quistes/tratamiento farmacológico , Hepatopatías/tratamiento farmacológico , Hígado/patología , Ácido Ursodesoxicólico/farmacología , Animales , Ácidos y Sales Biliares/metabolismo , Conductos Biliares/metabolismo , Conductos Biliares/patología , Calcio/metabolismo , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Colagogos y Coleréticos/farmacología , Quistes/metabolismo , Quistes/patología , Modelos Animales de Enfermedad , Hígado/efectos de los fármacos , Hígado/metabolismo , Hepatopatías/metabolismo , Hepatopatías/patología , Ratas , Espectrometría de Masas en Tándem
2.
Hepatology ; 56(2): 687-97, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22383162

RESUMEN

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.


Asunto(s)
Proteínas de Transporte de Anión/genética , Proteínas de Transporte de Anión/metabolismo , Antiportadores/genética , Antiportadores/metabolismo , Conductos Biliares Intrahepáticos/fisiopatología , Cirrosis Hepática Biliar , MicroARNs/metabolismo , Bicarbonatos/metabolismo , Conductos Biliares Intrahepáticos/citología , Conductos Biliares Intrahepáticos/metabolismo , Línea Celular Tumoral , Antiportadores de Cloruro-Bicarbonato , Cloruros/metabolismo , Simulación por Computador , Epitelio/fisiología , Humanos , Cirrosis Hepática Biliar/genética , Cirrosis Hepática Biliar/metabolismo , Cirrosis Hepática Biliar/fisiopatología , MicroARNs/genética , Cultivo Primario de Células , Biosíntesis de Proteínas/fisiología , ARN Mensajero/metabolismo , Proteínas SLC4A , Regulación hacia Arriba/genética
3.
PLoS One ; 6(12): e28717, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-22194894

RESUMEN

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.


Asunto(s)
Cirrosis Hepática Biliar/patología , Secretina/farmacología , Taurina/farmacología , Ácido Tauroquenodesoxicólico/farmacología , Ácido Ursodesoxicólico/farmacología , Animales , Proteínas de Transporte de Anión/metabolismo , Antiportadores/metabolismo , Bilis/efectos de los fármacos , Bilis/metabolismo , Células Cultivadas , Quiste del Colédoco/metabolismo , Quiste del Colédoco/patología , Ácido Deshidrocólico/farmacología , Silenciador del Gen/efectos de los fármacos , Humanos , Cirrosis Hepática Biliar/enzimología , Masculino , Microtúbulos/efectos de los fármacos , Microtúbulos/metabolismo , Quinasas de Proteína Quinasa Activadas por Mitógenos/metabolismo , Modelos Biológicos , Fosfatidilinositol 3-Quinasas/metabolismo , Polimerizacion/efectos de los fármacos , Proteína Quinasa C/metabolismo , Ratas , Ratas Wistar , Proteínas SLC4A
4.
J Clin Invest ; 118(2): 695-709, 2008 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-18188457

RESUMEN

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.


Asunto(s)
Proteínas de Transporte de Anión/genética , Antiportadores/genética , Colagogos y Coleréticos/farmacología , Dexametasona/farmacología , Glucocorticoides/farmacología , Hepatocitos/efectos de los fármacos , Transcripción Genética/efectos de los fármacos , Ácido Ursodesoxicólico/farmacología , Animales , Proteínas de Transporte de Anión/análisis , Proteínas de Transporte de Anión/metabolismo , Antiportadores/análisis , Antiportadores/metabolismo , Línea Celular , Colagogos y Coleréticos/uso terapéutico , Inmunoprecipitación de Cromatina , Dexametasona/uso terapéutico , Quimioterapia Combinada , Regulación de la Expresión Génica , Glucocorticoides/uso terapéutico , Factor Nuclear 1-alfa del Hepatocito/genética , Factor Nuclear 1-alfa del Hepatocito/metabolismo , Factor Nuclear 1-beta del Hepatocito/metabolismo , Hepatocitos/metabolismo , Humanos , Cirrosis Hepática Biliar/tratamiento farmacológico , Ratones , Mutación , Regiones Promotoras Genéticas/efectos de los fármacos , Regiones Promotoras Genéticas/genética , Proteínas SLC4A , Regulación hacia Arriba , Ácido Ursodesoxicólico/uso terapéutico , Factores de Transcripción p300-CBP/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...