Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Hepatology ; 69(2): 666-683, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30102412

RESUMO

Bile duct ligation (BDL) is an experimental procedure that mimics obstructive cholestatic disease. One of the early consequences of BDL in rodents is the appearance of so-called bile infarcts that correspond to Charcot-Gombault necrosis in human cholestasis. The mechanisms causing bile infarcts and their pathophysiological relevance are unclear. Therefore, intravital two photon-based imaging of BDL mice was performed with fluorescent bile salts (BS) and non-BS organic anion analogues. Key findings were followed up by matrix-assisted laser desorption ionization imaging, clinical chemistry, immunostaining, and gene expression analyses. In the acute phase, 1-3 days after BDL, BS concentrations in bile increased and single-cell bile microinfarcts occurred in dispersed hepatocytes throughout the liver caused by the rupture of the apical hepatocyte membrane. This rupture occurred after loss of mitochondrial membrane potential, followed by entry of bile, cell death, and a "domino effect" of further death events of neighboring hepatocytes. Bile infarcts provided a trans-epithelial shunt between bile canaliculi and sinusoids by which bile constituents leaked into blood. In the chronic phase, ≥21 days after BDL, uptake of BS tracers at the sinusoidal hepatocyte membrane was reduced. This contributes to elevated concentrations of BS in blood and decreased concentrations in the biliary tract. Conclusion: Bile microinfarcts occur in the acute phase after BDL in a limited number of dispersed hepatocytes followed by larger infarcts involving neighboring hepatocytes, and they allow leakage of bile from the BS-overloaded biliary tract into blood, thereby protecting the liver from BS toxicity; in the chronic phase after BDL, reduced sinusoidal BS uptake is a dominant protective factor, and the kidney contributes to the elimination of BS until cholemic nephropathy sets in.


Assuntos
Canalículos Biliares/fisiopatologia , Colestase/fisiopatologia , Modelos Animais de Doenças , Hepatócitos/metabolismo , Subfamília B de Transportador de Cassetes de Ligação de ATP/genética , Animais , Ácidos e Sais Biliares/sangue , Colestase/diagnóstico por imagem , Colestase/metabolismo , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Imagem Óptica , Membro 4 da Subfamília B de Transportadores de Cassetes de Ligação de ATP
2.
J Hepatol ; 66(6): 1231-1240, 2017 06.
Artigo em Inglês | MEDLINE | ID: mdl-28189756

RESUMO

BACKGROUND & AIMS: A wide range of liver diseases manifest as biliary obstruction, or cholestasis. However, the sequence of molecular events triggered as part of the early hepatocellular homeostatic response in obstructive cholestasis is poorly elucidated. Pericanalicular actin is known to accumulate during obstructive cholestasis. Therefore, we hypothesized that the pericanalicular actin cortex undergoes significant remodeling as a regulatory response to obstructive cholestasis. METHODS: In vivo investigations were performed in a bile duct-ligated mouse model. Actomyosin contractility was assessed using sandwich-cultured rat hepatocytes transfected with various fluorescently labeled proteins and pharmacological inhibitors of actomyosin contractility. RESULTS: Actomyosin contractility induces transient deformations along the canalicular membrane, a process we have termed inward blebbing. We show that these membrane intrusions are initiated by local ruptures in the pericanalicular actin cortex; and they typically retract following repair by actin polymerization and actomyosin contraction. However, above a certain osmotic pressure threshold, these inward blebs pinch away from the canalicular membrane into the hepatocyte cytoplasm as large vesicles (2-8µm). Importantly, we show that these vesicles aid in the regurgitation of bile from the bile canaliculi. CONCLUSION: Actomyosin contractility induces the formation of bile-regurgitative vesicles, thus serving as an early homeostatic mechanism against increased biliary pressure during cholestasis. LAY SUMMARY: Bile canaliculi expand and contract in response to the amount of secreted bile, and resistance from the surrounding actin bundles. Further expansion due to bile duct blockade leads to the formation of inward blebs, which carry away excess bile to prevent bile build up in the canaliculi.


Assuntos
Actomiosina/fisiologia , Ductos Biliares/fisiopatologia , Colestase/fisiopatologia , Animais , Canalículos Biliares/patologia , Canalículos Biliares/fisiopatologia , Refluxo Biliar/fisiopatologia , Fenômenos Biomecânicos , Colestase/patologia , Modelos Animais de Doenças , Masculino , Camundongos , Camundongos Transgênicos , Pressão , Ratos , Ratos Wistar
3.
Arch Toxicol ; 91(6): 2391-2403, 2017 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-27913845

RESUMO

In previous studies, we showed that the pro-oxidant model agent tert-butyl hydroperoxide (tBuOOH) induces alterations in hepatocanalicular secretory function by activating Ca2+-dependent protein kinase C isoforms (cPKC), via F-actin disorganization followed by endocytic internalization of canalicular transporters relevant to bile formation (Mrp2, Bsep). Since mitogen-activated protein kinases (MAPKs) may be downstream effectors of cPKC, we investigated here the involvement of the MAPKs of the ERK1/2, JNK1/2, and p38MAPK types in these deleterious effects. tBuOOH (100 µM, 15 min) increased the proportion of the active, phosphorylated forms of ERK1/2, JNK1/2, and p38MAPK, and panspecific PKC inhibition with bisindolylmaleimide-1 (100 nM) or selective cPKC inhibition with Gö6976 (1 µM) prevented the latter two events. In isolated rat hepatocyte couplets, tBuOOH (100 µM, 15 min) decreased the canalicular vacuolar accumulation of the fluorescent Bsep and Mrp2 substrates, cholylglycylamido fluorescein, and glutathione-methylfluorescein, respectively, and selective inhibitors of ERK1/2 (PD098059), JNK1/2 (SP600125), and p38MAPK (SB203580) partially prevented these alterations. In in situ perfused rat livers, these three MAPK inhibitors prevented tBuOOH (75 µM)-induced impairment of bile flow and the decrease in the biliary output of the Bsep and Mrp2 substrates, taurocholate, and dinitrophenyl-S-glutathione, respectively. The changes in Bsep/Mrp2 and F-actin localization induced by tBuOOH, as assessed by (immuno)fluorescence staining followed by analysis of confocal images, were prevented total or partially by the MAPK inhibitors. We concluded that MAPKs of the ERK1/2, JNK1/2, and p38MAPK types are all involved in cholestasis induced by oxidative stress, by promoting F-actin rearrangement and further endocytic internalization of canalicular transporters critical for bile formation.


Assuntos
Canalículos Biliares/efeitos dos fármacos , Colestase/induzido quimicamente , Fígado/efeitos dos fármacos , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Estresse Oxidativo/efeitos dos fármacos , terc-Butil Hidroperóxido/toxicidade , Animais , Canalículos Biliares/metabolismo , Canalículos Biliares/fisiopatologia , Colestase/metabolismo , Fígado/metabolismo , Fígado/fisiopatologia , Masculino , Proteína Quinase C/metabolismo , Ratos Wistar
4.
Arch Toxicol ; 90(4): 891-903, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25813982

RESUMO

Estradiol-17ß-D-glucuronide (E17G) induces acute endocytic internalization of canalicular transporters, including multidrug resistance-associated protein 2 (Abcc2) in rat, generating cholestasis. Several proteins organized in at least two different signaling pathways are involved in E17G cholestasis: one pathway involves estrogen receptor alpha (ERα), Ca(2+)-dependent protein kinase C and p38-mitogen activated protein kinase, and the other pathway involves GPR30, PKA, phosphoinositide 3-kinase/AKT and extracellular signal-related kinase 1/2. EGF receptor (EGFR) can potentially participate in both pathways since it interacts with GPR30 and ERα. Hence, the aim of this study was to analyze the potential role of this receptor and its downstream effectors, members of the Src family kinases in E17G-induced cholestasis. In vitro, EGFR inhibition by Tyrphostin (Tyr), Cl-387785 or its knockdown with siRNA strongly prevented E17G-induced impairment of Abcc2 function and localization. Activation of EGFR was necessary but not sufficient to impair the canalicular transporter function, whereas the simultaneous activation of EGFR and GPR30 could impair Abcc2 transport. The protection of Tyr was not additive to that produced by the ERα inhibitor ICI neither with that produced by Src kinase inhibitors, suggesting that EGFR shared the signaling pathway of ERα and Src. Further analysis of ERα, EGFR and Src activations induced by E17G, demonstrated that ERα activation precedes that of EGFR and EGFR activation precedes that of Src. In conclusion, activation of EGFR is a key factor in the alteration of canalicular transporter function and localization induced by E17G and it occurs before that of Src and after that of ERα.


Assuntos
Transportadores de Cassetes de Ligação de ATP/metabolismo , Receptores ErbB/metabolismo , Estradiol/análogos & derivados , Receptor alfa de Estrogênio/metabolismo , Hepatócitos/metabolismo , Animais , Canalículos Biliares/efeitos dos fármacos , Canalículos Biliares/metabolismo , Canalículos Biliares/fisiopatologia , Células Cultivadas , Colestase/induzido quimicamente , Colestase/metabolismo , Receptores ErbB/genética , Estradiol/metabolismo , Estradiol/farmacologia , Antagonistas do Receptor de Estrogênio/farmacologia , Feminino , Fulvestranto , Técnicas de Silenciamento de Genes , Hepatócitos/efeitos dos fármacos , Quinazolinas/farmacologia , Ratos , Ratos Wistar , Tirfostinas/farmacologia , Quinases da Família src/metabolismo
5.
Zebrafish ; 10(2): 199-210, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23697887

RESUMO

Fatty liver disease in humans can progress from steatosis to hepatocellular injury, fibrosis, cirrhosis, and liver failure. We developed a series of straightforward assays to determine whether zebrafish larvae with either tunicamycin- or ethanol-induced steatosis develop hepatic dysfunction. We found altered expression of genes involved in acute phase response and hepatic function, and impaired hepatocyte secretion and disruption of canaliculi in both models, but glycogen deficiency in hepatocytes and dilation of hepatic vasculature occurred only in ethanol-treated larvae. Hepatic stellate cells (HSCs) become activated during liver injury and HSC numbers increased in both models. Whether the excess lipids in hepatocytes are a direct cause of hepatocyte dysfunction in fatty liver disease has not been defined. We prevented ethanol-induced steatosis by blocking activation of the sterol response element binding proteins (Srebps) using gonzo(mbtps1) mutants and scap morphants and found that hepatocyte dysfunction persisted even in the absence of lipid accumulation. This suggests that lipotoxicity is not the primary cause of hepatic injury in these models of fatty liver disease. This study provides a panel of parameters to assess liver disease that can be easily applied to zebrafish mutants, transgenics, and for drug screening in which liver function is an important consideration.


Assuntos
Canalículos Biliares/fisiopatologia , Modelos Animais de Doenças , Fígado Gorduroso/fisiopatologia , Células Estreladas do Fígado/patologia , Hepatócitos/patologia , Peixe-Zebra , Animais , Bile/metabolismo , Etanol/farmacologia , Fígado Gorduroso/induzido quimicamente , Humanos , Larva/crescimento & desenvolvimento , Metabolismo dos Lipídeos , Hepatopatias Alcoólicas , Tunicamicina/farmacologia , Peixe-Zebra/crescimento & desenvolvimento
6.
Acta Bioeng Biomech ; 15(4): 9-17, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24479556

RESUMO

The aim of the present paper is to develop an analytical model for description of the pathological bile flow in the major duodenal papilla duct with a calculus. The problem is separated into two parts. The first part deals with determination of bile behaviour and constitutive relation parameters of the pathological bile. The viscosity vs. shear rate, the viscosity vs. time, and shear stress vs. shear rate dependences are obtained for different types of bile taken from patients of different age and sex. As a result, the approximation of curves described by the Casson equation was obtained. It was shown that the pathological bile is a thixotropic non-Newtonian fluid. The second part is directly related to modelling of the bile flow in the duct with a calculus. As a result of solving the problem, the bile velocity profile, flow rate vs. time, and bile pressure vs. calculus radius were obtained. The dependences obtained may play an important role in the assessment of an indication to operation.


Assuntos
Canalículos Biliares/patologia , Canalículos Biliares/fisiopatologia , Ductos Biliares/patologia , Ductos Biliares/fisiopatologia , Bile/metabolismo , Modelos Biológicos , Reologia , Fenômenos Biomecânicos , Vesícula Biliar/patologia , Vesícula Biliar/fisiopatologia , Humanos , Resistência ao Cisalhamento , Esfíncter da Ampola Hepatopancreática/patologia , Estresse Mecânico
7.
Acta Gastroenterol Belg ; 75(4): 405-10, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23402083

RESUMO

Progressive familial intrahepatic cholestasis (PFIC) and benign recurrent intrahepatic cholestasis (BRIC) are two rare autosomal recessive disorders, characterized by cholestasis. They are related to mutations in hepatocellular transport system genes involved in bile formation. The differentiation between PFIC and BRIC is based on phenotypic presentation: PFIC is a progressive disease, with evolution to end-stage liver disease. BRIC is characterized by intermittent recurrent cholestatic episodes, with irresistible pruritus, mostly without evident liver damage. Between symptomatic periods, patients are completely asymptomatic. In this article, a short overview of the aetiology, the clinical and diagnostic characteristics and the therapy of both PFIC and BRIC are given.


Assuntos
Subfamília B de Transportador de Cassetes de Ligação de ATP/metabolismo , Transportadores de Cassetes de Ligação de ATP/metabolismo , Adenosina Trifosfatases/metabolismo , Bile/metabolismo , Colestase Intra-Hepática , Membro 11 da Subfamília B de Transportadores de Cassetes de Ligação de ATP , Canalículos Biliares/metabolismo , Canalículos Biliares/fisiopatologia , Colestase Intra-Hepática/diagnóstico , Colestase Intra-Hepática/genética , Colestase Intra-Hepática/fisiopatologia , Colestase Intra-Hepática/terapia , Doença Crônica , Diagnóstico Diferencial , Gerenciamento Clínico , Progressão da Doença , Predisposição Genética para Doença , Humanos , Conduta do Tratamento Medicamentoso , Recidiva , Resultado do Tratamento
8.
Cell Death Dis ; 2: e183, 2011 Jul 21.
Artigo em Inglês | MEDLINE | ID: mdl-21776021

RESUMO

Morphological changes of hepatocyte death have so far only been described on cells in culture or in tissue sections. Using a high-resolution and high-magnification multiphoton microscopic system, we recorded in living mice serial changes of acetaminophen (APAP)-induced hepatocyte necrosis in relevance to metabolism of a fluorogenic bile solute. Initial changes of hepatocyte injury included basal membrane disruption and loss of mitochondrial membrane potential. An overwhelming event of rupture at adjacent apical membrane resulting in flooding of bile into these hepatocytes might ensue. Belbs formed on basal membrane and then dislodged into the sinusoid circulation. Transmission electron microscopy disclosed a necrotic hepatocyte depicting well the changes after apical membrane rupture and bile flooding. Administration of the antidote N-acetylcysteine dramatically reduced the occurrence of apical membrane rupture. The present results demonstrated a hidden but critical step of apical membrane rupture leading to irreversible APAP-induced hepatocyte injury.


Assuntos
Acetaminofen/toxicidade , Analgésicos não Narcóticos/toxicidade , Canalículos Biliares/efeitos dos fármacos , Membrana Celular/efeitos dos fármacos , Fígado/efeitos dos fármacos , Necrose/induzido quimicamente , Acetilcisteína/farmacologia , Acetilcisteína/toxicidade , Animais , Antídotos/farmacologia , Canalículos Biliares/fisiopatologia , Membrana Celular/ultraestrutura , Fígado/citologia , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos C57BL , Microscopia Eletrônica de Transmissão , Microscopia de Fluorescência por Excitação Multifotônica
9.
J Hepatol ; 52(2): 258-71, 2010 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-20034695

RESUMO

Bile formation at the canalicular membrane is a delicate process. This is illustrated by inherited liver diseases due to mutations in ATP8B1, ABCB11, ABCB4, ABCC2 and ABCG5/8, all encoding hepatocanalicular transporters. Effective treatment of these canalicular transport defects is a clinical and scientific challenge that is still ongoing. Current evidence indicates that ursodeoxycholic acid (UDCA) can be effective in selected patients with PFIC3 (ABCB4 deficiency), while rifampicin reduces pruritus in patients with PFIC1 (ATP8B1 deficiency) and PFIC2 (ABCB11 deficiency), and might abort cholestatic episodes in BRIC (mild ATP8B1 or ABCB11 deficiency). Cholestyramine is essential in the treatment of sitosterolemia (ABCG5/8 deficiency). Most patients with PFIC1 and PFIC2 will benefit from partial biliary drainage. Nevertheless liver transplantation is needed in a substantial proportion of these patients, as it is in PFIC3 patients. New developments in the treatment of canalicular transport defects by using nuclear receptors as a target, enhancing the expression of the mutated transporter protein by employing chaperones, or by mutation specific therapy show substantial promise. This review will focus on the therapy that is currently available as well as on those developments that are likely to influence clinical practice in the near future.


Assuntos
Canalículos Biliares/fisiopatologia , Hepatopatias/genética , Hepatopatias/terapia , Subfamília B de Transportador de Cassetes de Ligação de ATP/deficiência , Subfamília B de Transportador de Cassetes de Ligação de ATP/genética , Membro 11 da Subfamília B de Transportadores de Cassetes de Ligação de ATP , Membro 5 da Subfamília G de Transportadores de Cassetes de Ligação de ATP , Membro 8 da Subfamília G de Transportadores de Cassetes de Ligação de ATP , Transportadores de Cassetes de Ligação de ATP/genética , Adenosina Trifosfatases/deficiência , Adenosina Trifosfatases/genética , Bile/fisiologia , Procedimentos Cirúrgicos do Sistema Biliar , Transporte Biológico Ativo , Resina de Colestiramina/uso terapêutico , Terapia Genética , Humanos , Lipoproteínas/deficiência , Lipoproteínas/genética , Hepatopatias/fisiopatologia , Transplante de Fígado , Modelos Biológicos , Proteína 2 Associada à Farmacorresistência Múltipla , Proteínas Associadas à Resistência a Múltiplos Medicamentos/deficiência , Proteínas Associadas à Resistência a Múltiplos Medicamentos/genética , Mutação , Rifampina/uso terapêutico , Ácido Ursodesoxicólico/uso terapêutico
10.
Gastroenterology ; 136(3): 1060-9, 2009 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-19027009

RESUMO

BACKGROUND & AIMS: Progressive familial intrahepatic cholestasis 1 (PFIC1) results from mutations in ATP8B1, a putative aminophospholipid flippase. Conflicting hypotheses have been proposed for the pathogenesis of PFIC1. The aim of this study was to determine whether ATP8B1 deficiency produces cholestasis by altering the activity of the farnesoid X receptor (FXR) or by impairing the structure of the canalicular membrane. METHODS: ATP8B1/Atp8b1 was knocked down in human and rat hepatocytes and Caco2 cells using adenoviral and oligonucleotide small interfering RNAs. RESULTS: ATP8B1 messenger RNA and protein expression was greatly reduced in human and rat cells. In contrast, FXR expression and several FXR-dependent membrane transporters (bile salt export pump [BSEP], multidrug resistance-associated protein [MRP] 2) were unchanged at messenger RNA or protein levels in ATP8B1-deficient cells, whereas Mrp3 and Mrp4 were up-regulated in rat hepatocytes. FXR activity remained intact in these cells, as evidenced by 6alpha-ethyl chenodeoxycholic acid-mediated induction of small heterodimer partner, BSEP, and multidrug-resistant protein (MDR) 3/Mdr2. Fluorescent substrate excretion assays indicate that Bsep function was significantly reduced in Atp8b1-deficient rat hepatocytes, although Bsep remained localized to the canalicular membrane. Exposure to the hydrophobic bile acid CDCA resulted in focal areas of canalicular membrane disruption by electron microscopy and luminal accumulation of NBD-phosphatidylserine, consistent with the function of Atp8b1 as an aminophospholipid flippase. CONCLUSIONS: ATP8B1 deficiency predisposes to cholestasis by favoring bile acid-induced injury in the canalicular membrane but does not directly affect FXR expression, which may occur in PFIC1 as a secondary phenomenon associated with cholestasis.


Assuntos
Adenosina Trifosfatases/genética , Canalículos Biliares/metabolismo , Canalículos Biliares/fisiopatologia , Proteínas de Ligação a DNA/genética , Hepatócitos/fisiologia , Receptores Citoplasmáticos e Nucleares/genética , Fatores de Transcrição/genética , 4-Cloro-7-nitrobenzofurazano/análogos & derivados , 4-Cloro-7-nitrobenzofurazano/farmacocinética , Membro 11 da Subfamília B de Transportadores de Cassetes de Ligação de ATP , Transportadores de Cassetes de Ligação de ATP/metabolismo , Adenosina Trifosfatases/deficiência , Animais , Canalículos Biliares/efeitos dos fármacos , Células CACO-2 , Ácido Quenodesoxicólico/farmacologia , Proteínas de Ligação a DNA/metabolismo , Fármacos Gastrointestinais/farmacologia , Expressão Gênica/fisiologia , Hepatócitos/citologia , Humanos , Proteína 2 Associada à Farmacorresistência Múltipla , Fosfatidilserinas/farmacocinética , Proteínas de Transferência de Fosfolipídeos , RNA Interferente Pequeno , Ratos , Receptores Citoplasmáticos e Nucleares/metabolismo , Fatores de Transcrição/metabolismo , Transfecção
11.
Wien Med Wochenschr ; 158(19-20): 542-8, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-18998069

RESUMO

Alterations in bile secretion at the hepatocellular and cholangiocellular levels may cause cholestasis. Formation of 'toxic bile' may be the consequence of abnormal bile composition and can result in hepatocellular and/or bile duct injury. The canalicular phospholipid flippase (Mdr2/MDR3) normally mediates biliary excretion of phospholipids, which normally form mixed micelles with bile acids and cholesterol to protect the bile duct epithelium from the detergent properties of bile acids. Mdr2 knockout mice are not capable of excreting phospholipids into bile and spontaneously develop bile duct injury with macroscopic and microscopic features closely resembling human sclerosing cholangitis. MDR3 mutations have been linked to a broad spectrum of hepatobiliary disorders in humans ranging from progressive familial intrahepatic cholestasis in neonates to intrahepatic cholestasis of pregnancy, drug-induced cholestasis, intrahepatic cholelithiasis, sclerosing cholangitis and biliary cirrhosis in adults. Other examples for bile injury due to the formation of toxic bile include the cholangiopathy seen in cystic fibrosis, after lithocholate feeding (in mice) and vanishing bile duct syndromes induced by drugs and xenobiotics. Therapeutic strategies for cholangiopathies may target bile composition/toxicity and the affected bile duct epithelium itself, and ideally should also have anti-cholestatic, anti-fibrotic and anti-neoplastic properties. Ursodeoxycholic acid (UDCA) shows some of these properties, but is of limited efficacy in the treatment of human cholangiopathies. By contrast to UDCA, its side chain-shortened homologue norUDCA undergoes cholehepatic shunting leading to a bicarbonate-rich hypercholeresis. Moreover, norUDCA has anti-inflammatory, anti-fibrotic and anti-proliferative effects, and stimulates bile acid detoxification. Upcoming clinical trials will have to demonstrate whether norUDCA or other side chain-modified bile acids are also clinically effective in humans. Finally, drugs for the treatment of cholangiopathies may target bile toxicity via nuclear receptors (FXR, PPARalpha) regulating biliary phospholipid and bile acid excretion.


Assuntos
Ácidos e Sais Biliares/toxicidade , Neoplasias dos Ductos Biliares/fisiopatologia , Ductos Biliares Intra-Hepáticos/fisiopatologia , Bile/fisiologia , Carcinoma Hepatocelular/fisiopatologia , Colangiocarcinoma/fisiopatologia , Colangite Esclerosante/fisiopatologia , Colestase Intra-Hepática/fisiopatologia , Neoplasias Hepáticas/fisiopatologia , Subfamília B de Transportador de Cassetes de Ligação de ATP/genética , Animais , Canalículos Biliares/fisiopatologia , Neoplasias dos Ductos Biliares/genética , Carcinoma Hepatocelular/genética , Transformação Celular Neoplásica/genética , Colangiocarcinoma/genética , Colangite Esclerosante/genética , Colestase Intra-Hepática/genética , Ácidos Cólicos , Modelos Animais de Doenças , Humanos , Neoplasias Hepáticas/genética , Camundongos , Camundongos Knockout , Mutação/genética , Noresteroides , Ácido Ursodesoxicólico/fisiologia , Membro 4 da Subfamília B de Transportadores de Cassetes de Ligação de ATP
12.
Transplantation ; 77(3): 373-9, 2004 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-14966410

RESUMO

BACKGROUND: Although it is generally considered that livers with moderate steatosis can be safely used in the setting of living-donor liver transplantation, the effect of the regenerative process of such a graft on postoperative liver function is incompletely understood. We assessed the morphologic and functional alterations during the regeneration of fatty liver, with special reference to the biliary system. METHODS: Wistar rats with normal or fatty livers induced by a choline-deficient diet were subjected to 70% partial hepatectomy (PH). The regenerated liver weight and serum parameters were compared. Furthermore, to assess the spatial alterations of bile canalicular networks, the distribution of AGp110, a fibronectin receptor that localizes on the apical (bile canalicular) membrane of the hepatocytes, was analyzed immunohistochemically. RESULTS: The serum albumin levels of the fatty-liver rats decreased significantly after 24 hours, and this continued until day 7. The increase in the total bile acid levels of the fatty-liver group was higher and more prolonged compared with that of the normal-liver group. At 24 hours after PH, discontinuity of the AGp110-positive canalicular network was evident in both groups. At 7 days after PH, the typical AGp110-positive canalicular network was almost restored in the normal-liver group. In contrast, the fatty-liver group showed sustained discontinuity of canalicular networks at the same time point. CONCLUSIONS: The livers with moderate steatosis are associated with prolonged cholestasis after 70% PH, and this was caused, in part, by sustained spatial disturbance of bile canalicular networks during the regenerative process.


Assuntos
Canalículos Biliares/fisiopatologia , Fígado Gorduroso/fisiopatologia , Regeneração Hepática , Fenômenos Fisiológicos da Nutrição Animal , Animais , Ácidos e Sais Biliares/metabolismo , Sistema Biliar/fisiopatologia , Hepatectomia/métodos , Imuno-Histoquímica , Fígado/patologia , Fígado/fisiopatologia , Masculino , Tamanho do Órgão , Ratos , Ratos Wistar , Receptores de Fibronectina/metabolismo , Albumina Sérica/análise , Fatores de Tempo
14.
Artigo em Inglês | MEDLINE | ID: mdl-11294287

RESUMO

It is well known that the liver plays a major role in the clearance of systemic toxemia and is postulated as a regulational organ in the host-defense system. The well-controlled interaction between hepatic parenchymal cells and sinusoidal lining cells including macrophages and Kupffer cells can systematically regulate even critical infections. However, when patients are under the overload condition caused by severe infection, rejection of a transplanted liver and other hapatic dysfunction often are experienced following surgery. Among various signs and symptoms of hepatic dysfunction, progressive cholestasis is recognized as a polarized representation of the irreversible changes in hepatic constitutional cellular functions, especially in hepatic parenchymal cells. Bile canaliculi, the smallest components of the biliary tree, lie between the apical surfaces of adjacent hepatocytes. Septic cholestasis might be a result of disturbance of the total bile canalicular system, i.e., bile secretion, canalicular contraction, and so on. Recently, the molecular biology of the hepatocellular transport system has become better understood, and the pathophysiological condition of cholestasis can be explained as a representation of the intracellular molecular transcriptional system. Cellular changes in surgical cholestasis and molecular findings concerning the bile canaliculus are introduced in this article.


Assuntos
Canalículos Biliares/fisiopatologia , Colestase Intra-Hepática/fisiopatologia , Fígado/fisiopatologia , Animais , Colestase Intra-Hepática/patologia , Hepatócitos/fisiologia , Humanos , Mediadores da Inflamação/fisiologia , Células de Kupffer/patologia , Células de Kupffer/fisiologia , Fígado/patologia
15.
Dig Dis Sci ; 44(8): 1662-8, 1999 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-10492150

RESUMO

Cholestasis, which affects phospholipid trafficking, therefore would be expected to alter canalicular membrane phospholipid composition and fluidity, as well as fatty acid composition of membrane phospholipid. These alterations may affect transcytotic vesicle fusion and would be expected to cause variety of cholestatic phenomena. The aim of this study was to determine the effect of cholestasis on transcytotic vesicle fusion. Sprague-Dawley rats with extrahepatic and intrahepatic cholestasis were prepared by bile duct ligation (6 hr or three days) and phalloidin infusion (0.4 mg/kg body weight), respectively. Liposomes of phosphatidylserine/phosphatidylcholine were labeled with octadecyl rhodamine B chloride. Fusion of liposomes to canalicular membrane vesicle preparations from cholestatic and control rats was induced by the addition of calcium. The degree of fusion was evaluated by measuring the increase in rhodamine fluorescence. Membrane phospholipid content also was analyzed. Rates of liposomal fusion to membranes from cholestatic rats were decreased compared to controls. The saturated/unsaturated and saturated/polyunsaturated fatty acid ratios were increased in membrane phosphatidylcholine and decreased in membrane sphingomyelin from cholestatic rats. Cholesterol/phospholipid ratios were increased. Thus, in the presence of cholestasis, a redistribution of phospholipid species within canalicular membranes is associated with decreased transcytotic vesicle fusion. Cholestasis likely decreases membrane fluidity. The regulation of phospholipid species within hepatocellular membranes may play an important role in intrahepatic lipid transport.


Assuntos
Canalículos Biliares/fisiopatologia , Colestase/fisiopatologia , Fusão de Membrana , Fosfolipídeos/metabolismo , Animais , Canalículos Biliares/metabolismo , Colestase/metabolismo , Colesterol/metabolismo , Ácidos Graxos/metabolismo , Lipossomos/fisiologia , Masculino , Membranas/metabolismo , Membranas/fisiopatologia , Fosfatidilcolinas/metabolismo , Ratos , Ratos Sprague-Dawley , Esfingomielinas/metabolismo , Distribuição Tecidual
16.
Yale J Biol Med ; 70(4): 365-78, 1997.
Artigo em Inglês | MEDLINE | ID: mdl-9626757

RESUMO

The discovery of unidirectional, ATP-dependent canalicular transport systems (also termed "export pumps") for bile salts, amphiphilic anionic conjugates, lipophilic cations, and phospholipids has opened new opportunities for understanding biliary physiology and the pathophysiology of cholestasis. In addition, ATP-independent canalicular transport systems for glutathione and bicarbonate contribute to (bile acid-independent) bile formation. Canalicular excretion of bile salts and several non-bile acid organic anions is impaired in various experimental models of cholestasis. Recent cloning of several canalicular transport systems now facilitates studies on their molecular regulation in cholestasis. Although the picture is far from complete, experimental evidence now exists that decreased or even absent expression of canalicular transport proteins may explain impaired transport function resulting in hyperbilirubinemia and cholestasis. With the increasing availability of molecular probes for these transport systems in humans, new information on the molecular regulation of canalicular transport proteins in human cholestatic liver diseases is beginning to emerge and should bring new insights into their pathophysiology and treatment. This article gives an overview on molecular alterations of canalicular transport systems in experimental models of cholestasis and discusses the potential implications of these changes for the pathophysiology of cholestasis.


Assuntos
Canalículos Biliares/fisiopatologia , Colestase/fisiopatologia , Membro 1 da Subfamília B de Cassetes de Ligação de ATP/metabolismo , Trifosfato de Adenosina/metabolismo , Animais , Proteínas de Transporte de Ânions , Bicarbonatos/metabolismo , Ácidos e Sais Biliares/metabolismo , Transporte Biológico Ativo , Proteínas de Transporte/metabolismo , Colestase/etiologia , Colestase/genética , Colesterol/metabolismo , Modelos Animais de Doenças , Glutationa/metabolismo , Humanos , Fosfolipídeos/metabolismo
17.
J Pediatr Surg ; 28(6): 851-6, 1993 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-8331519

RESUMO

The role of pathological changes of the intrahepatic bile canaliculi in determining postportoenterostomy bile drainage in biliary atresia (BA) patients remains unestablished. It is known that canalicular contraction contributes an active force for bile flow in normal ductal systems. This motility is controlled by the bile canalicular membrane-associated filaments (BCMF), particularly actin and myosin. Wedge resected specimens of liver from 13 patients with BA were studied using histopathological markers of BCMF in order to clarify the morphological and functional changes of bile canaliculi. Histopathological data were also compared with clinical outcomes after portoenterostomy. Patients who showed sufficient bile flow after the operation showed an almost normal shape and level of BCMF accumulation around bile canaliculi, whereas there was markedly increased BCMF volume in patients who did not exhibit bile flow after surgery. The clinical staining patterns of BCMF in BA patients appears closely related to clinical outcome. These findings suggest that BCMF plays an important role in determining the rate of postoperative bile flow in BA patients.


Assuntos
Actinas/análise , Canalículos Biliares/patologia , Atresia Biliar/patologia , Miosinas/análise , Canalículos Biliares/química , Canalículos Biliares/fisiopatologia , Atresia Biliar/metabolismo , Atresia Biliar/fisiopatologia , Atresia Biliar/cirurgia , Movimento Celular , Humanos , Imuno-Histoquímica , Lactente , Microscopia Eletrônica , Portoenterostomia Hepática , Prognóstico
19.
Proc Natl Acad Sci U S A ; 87(9): 3557-61, 1990 May.
Artigo em Inglês | MEDLINE | ID: mdl-2333302

RESUMO

TR- mutant Wistar rats secrete markedly fewer organic anions other than bile acids from the liver into the bile than do control rats. Fluorescence-image analysis of isolated normal and TR- hepatocyte "doublets", which retain a bile canaliculus between them, revealed that normal hepatocytes readily transport a fluorescent bile acid (fluorescein isothiocyanate glycocholate) and a nonbile acid organic anion (carboxydichlorofluorescein diacetate) into the canaliculus. Hepatocyte doublets from TR- rats also transported fluorescein isothiocyanate glycocholate normally, but transport of carboxydichlorofluorescein diacetate into the canaliculus was negligible. Vesicles derived from the canicular domain of the plasma membrane of hepatocytes (CMV) from control and TR- rats were used to characterize the transport process for 35S-labeled bromosulphthalein and 35S-labeled bromosulphthalein glutathione, which represent nonbile acid organic anions. CMV from normal rat hepatocytes had an ATP- and temperature-dependent, saturable transport process for these 35S-labeled compounds that was absent in CMV from TR- rats. CMV from TR- rats retained normal ATP-dependent transport of daunomycin, and immunologic blots with a monoclonal antibody against the multidrug resistance gene product, P-glycoprotein, revealed no difference between normal and TR-CMV. These studies reveal that the bile canaliculus in normal rats contains an ATP-dependent organic anion transport system that is functionally absent in TR- mutant rats. The defect in TR- mutant rats is phenotypically similar to that seen in mutant Corriedale sheep and in the Dubin-Johnson syndrome in man.


Assuntos
Trifosfato de Adenosina/metabolismo , Canalículos Biliares/fisiopatologia , Ductos Biliares Intra-Hepáticos/fisiopatologia , Bile/metabolismo , Hiperbilirrubinemia Hereditária/fisiopatologia , Fígado/metabolismo , Sulfobromoftaleína/metabolismo , Animais , Ânions , Transporte Biológico , Membrana Celular/metabolismo , Células Cultivadas , Daunorrubicina/metabolismo , Glutationa/metabolismo , Hiperbilirrubinemia Hereditária/metabolismo , Cinética , Masculino , Ratos , Ratos Endogâmicos , Ratos Mutantes , Valores de Referência
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...