Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 96
Filtrar
1.
Gastroenterology ; 160(6): 2103-2118, 2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-33465371

RESUMO

BACKGROUND & AIMS: Liver tight junctions (TJs) establish tissue barriers that isolate bile from the blood circulation. TJP2/ZO-2-inactivating mutations cause progressive cholestatic liver disease in humans. Because the underlying mechanisms remain elusive, we characterized mice with liver-specific inactivation of Tjp2. METHODS: Tjp2 was deleted in hepatocytes, cholangiocytes, or both. Effects on the liver were assessed by biochemical analyses of plasma, liver, and bile and by electron microscopy, histology, and immunostaining. TJ barrier permeability was evaluated using fluorescein isothiocyanate-dextran (4 kDa). Cholic acid (CA) diet was used to assess susceptibility to liver injury. RESULTS: Liver-specific deletion of Tjp2 resulted in lower Cldn1 protein levels, minor changes to the TJ, dilated canaliculi, lower microvilli density, and aberrant radixin and bile salt export pump (BSEP) distribution, without an overt increase in TJ permeability. Hepatic Tjp2-defcient mice presented with mild progressive cholestasis with lower expression levels of bile acid transporter Abcb11/Bsep and detoxification enzyme Cyp2b10. A CA diet tolerated by control mice caused severe cholestasis and liver necrosis in Tjp2-deficient animals. 1,4-Bis[2-(3,5-dichloropyridyloxy)]benzene ameliorated CA-induced injury by enhancing Cyp2b10 expression, and ursodeoxycholic acid provided partial improvement. Inactivating Tjp2 separately in hepatocytes or cholangiocytes showed only mild CA-induced liver injury. CONCLUSION: Tjp2 is required for normal cortical distribution of radixin, canalicular volume regulation, and microvilli density. Its inactivation deregulated expression of Cldn1 and key bile acid transporters and detoxification enzymes. The mice provide a novel animal model for cholestatic liver disease caused by TJP2-inactivating mutations in humans.


Assuntos
Canalículos Biliares/metabolismo , Doença Hepática Induzida por Substâncias e Drogas/genética , Colestase/genética , Junções Íntimas/metabolismo , Proteína da Zônula de Oclusão-2/genética , Membro 11 da Subfamília B de Transportadores de Cassetes de Ligação de ATP/genética , Membro 11 da Subfamília B de Transportadores de Cassetes de Ligação de ATP/metabolismo , Animais , Hidrocarboneto de Aril Hidroxilases/metabolismo , Ácidos e Sais Biliares/metabolismo , Canalículos Biliares/patologia , Doença Hepática Induzida por Substâncias e Drogas/tratamento farmacológico , Colagogos e Coleréticos/uso terapêutico , Ácido Cólico , Claudina-1/metabolismo , Família 2 do Citocromo P450/metabolismo , Proteínas do Citoesqueleto/metabolismo , Células Epiteliais , Feminino , Fibrose , Predisposição Genética para Doença , Hepatócitos , Masculino , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Knockout , Mutação , Oxazóis/uso terapêutico , Permeabilidade , Fatores de Proteção , RNA Mensageiro/metabolismo , Esteroide Hidroxilases/metabolismo , Junções Íntimas/ultraestrutura , Ácido Ursodesoxicólico/uso terapêutico , Proteína da Zônula de Oclusão-2/deficiência
2.
Toxicol Appl Pharmacol ; 394: 114958, 2020 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-32198022

RESUMO

Drug-induced liver injury (DILI) can cause hepatic failure and result in drug withdrawal from the market. It has host-related and compound-dependent mechanisms. Preclinical prediction of DILI risk is very challenging and safety assessments based on animals inadequately forecast human DILI risk. In contrast, human-derived in vitro cell culture-based models could improve DILI risk prediction accuracy. Here, we developed and validated an innovative method to assess DILI risk associated with various compounds. Fifty-four marketed and withdrawn drugs classified as DILI risks of "most concern", "less concern", and "no concern" were tested using a combination of four assays addressing mitochondrial injury, intrahepatic lipid accumulation, inhibition of bile canalicular network formation, and bile acid accumulation. Using the inhibitory potencies of the drugs evaluated in these in vitro tests, an algorithm with the highest available DILI risk prediction power was built by artificial neural network (ANN) analysis. It had an overall forecasting accuracy of 73%. We excluded the intrahepatic lipid accumulation assay to avoid overfitting. The accuracy of the algorithm in terms of predicting DILI risks was 62% when it was constructed by ANN but only 49% when it was built by the point-added scoring method. The final algorithm based on three assays made no DILI risk prediction errors such as "most concern " instead of "no concern" and vice-versa. Our mechanistic approach may accurately predict DILI risks associated with numerous candidate drugs.


Assuntos
Bioensaio/métodos , Doença Hepática Induzida por Substâncias e Drogas/diagnóstico , Efeitos Colaterais e Reações Adversas Relacionados a Medicamentos/diagnóstico , Valor Preditivo dos Testes , Algoritmos , Ácidos e Sais Biliares/metabolismo , Canalículos Biliares/patologia , Linhagem Celular , Humanos , Metabolismo dos Lipídeos/efeitos dos fármacos , Dose Máxima Tolerável , Mitocôndrias/efeitos dos fármacos , Reprodutibilidade dos Testes
3.
Nat Med ; 25(12): 1885-1893, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31792455

RESUMO

Early disease diagnosis is key to the effective treatment of diseases. Histopathological analysis of human biopsies is the gold standard to diagnose tissue alterations. However, this approach has low resolution and overlooks 3D (three-dimensional) structural changes resulting from functional alterations. Here, we applied multiphoton imaging, 3D digital reconstructions and computational simulations to generate spatially resolved geometrical and functional models of human liver tissue at different stages of non-alcoholic fatty liver disease (NAFLD). We identified a set of morphometric cellular and tissue parameters correlated with disease progression, and discover profound topological defects in the 3D bile canalicular (BC) network. Personalized biliary fluid dynamic simulations predicted an increased pericentral biliary pressure and micro-cholestasis, consistent with elevated cholestatic biomarkers in patients' sera. Our spatially resolved models of human liver tissue can contribute to high-definition medicine by identifying quantitative multiparametric cellular and tissue signatures to define disease progression and provide new insights into NAFLD pathophysiology.


Assuntos
Colestase/patologia , Imageamento Tridimensional , Fígado/patologia , Hepatopatia Gordurosa não Alcoólica/patologia , Canalículos Biliares/patologia , Sistema Biliar/patologia , Colestase/diagnóstico , Simulação por Computador , Progressão da Doença , Diagnóstico Precoce , Humanos , Modelos Biológicos , Hepatopatia Gordurosa não Alcoólica/diagnóstico
4.
J Hepatol ; 71(2): 344-356, 2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-30965071

RESUMO

BACKGROUND & AIMS: Hepatocyte polarity is essential for the development of bile canaliculi and for safely transporting bile and waste products from the liver. Functional studies of autologous mutated proteins in the context of the polarized hepatocyte have been challenging because of the lack of appropriate cell models. The aims of this study were to obtain a patient-specific hepatocyte model that recapitulated hepatocyte polarity and to employ this model to study endogenous mutant proteins in liver diseases that involve hepatocyte polarity. METHODS: Urine cell-derived pluripotent stem cells, taken from a patient with a homozygous mutation in ATP7B and a patient with a heterozygous mutation, were differentiated towards hepatocyte-like cells (hiHeps). HiHeps were also derived from a patient with MEDNIK syndrome. RESULTS: Polarized hiHeps that formed in vivo-like bile canaliculi could be generated from embryonic and patient urine cell-derived pluripotent stem cells. HiHeps recapitulated polarized protein trafficking processes, exemplified by the Cu2+-induced redistribution of the copper transporter protein ATP7B to the bile canalicular domain. We demonstrated that, in contrast to the current dogma, the most frequent yet enigmatic Wilson disease-causing ATP7B-H1069Q mutation per se did not preclude trafficking of ATP7B to the trans-Golgi Network. Instead, it prevented its Cu2+-induced polarized redistribution to the bile canalicular domain, which could not be reversed by pharmacological folding chaperones. Finally, we demonstrate that hiHeps from a patient with MEDNIK syndrome, suffering from liver copper overload of unclear etiology, showed no defect in the Cu2+-induced redistribution of ATP7B to the bile canaliculi. CONCLUSIONS: Functional cell polarity can be achieved in patient pluripotent stem cell-derived hiHeps, enabling, for the first time, the study of the endogenous mutant proteins, patient-specific pathogenesis and drug responses for diseases where hepatocyte polarity is a key factor. LAY SUMMARY: This study demonstrates that cells that are isolated from urine can be reprogrammed in a dish towards hepatocytes that display architectural characteristics similar to those seen in the intact liver. The application of this methodology to cells from patients diagnosed with inherited copper metabolism-related liver diseases (that is, Wilson disease and MEDNIK syndrome) revealed unexpected and novel insights into patient mutation-specific disease mechanisms and drug responses.


Assuntos
Canalículos Biliares/patologia , Polaridade Celular/genética , Eritroceratodermia Variável/genética , Hepatócitos/metabolismo , Degeneração Hepatolenticular/genética , Células-Tronco Pluripotentes/metabolismo , Complexo 1 de Proteínas Adaptadoras/genética , Subunidades sigma do Complexo de Proteínas Adaptadoras/genética , Canalículos Biliares/metabolismo , Células Cultivadas , Cobre/metabolismo , ATPases Transportadoras de Cobre/genética , Eritroceratodermia Variável/patologia , Degeneração Hepatolenticular/patologia , Humanos , Proteínas Mutantes/metabolismo , Mutação , Transporte Proteico
5.
Methods Mol Biol ; 1981: 313-323, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31016663

RESUMO

Cholestasis remains a major challenge in drug-induced liver injury, and therefore warrants identification of chemical entities that may lead to cholestasis. Recent advances in cell culture methods enable 3D spheroid models to remain viable for much longer periods of time than conventional sandwich cultures of primary human hepatocytes while maintaining native tissue-like functionality, such as drug metabolism activity, receptor signaling functionality, and physiological relevance. These spheroid models enable us to study repeated exposure effects associated with chemicals and their metabolites that may ultimately progress to cholestasis and liver injury. HepaRG cells cultured as spheroids are viable for more than 4 weeks with cytochrome P450 enzymatic activities comparable to ranges observed in freshly isolated/cryopreserved suspensions of primary human hepatocytes. HepaRG spheroids form bile canalicular structures with potential application as a model to study biliary excretion processes and intrahepatic obstruction of bile flow, leading to hepatocellular damage and death. In this chapter, we describe methods to culture 3D spheroids of HepaRG cells with extensive bile canalicular structures/networks, image transport of bile acid (cholyl-lysyl-fluorescein) to the bile canaliculi, and measure cholestatic drug-induced cytotoxicity.


Assuntos
Colestase/metabolismo , Colestase/patologia , Hepatócitos/citologia , Fígado/citologia , Canalículos Biliares/metabolismo , Canalículos Biliares/patologia , Doença Hepática Induzida por Substâncias e Drogas/metabolismo , Doença Hepática Induzida por Substâncias e Drogas/patologia , Sistema Enzimático do Citocromo P-450/metabolismo , Hepatócitos/metabolismo , Hepatócitos/patologia , Humanos , Esferoides Celulares/metabolismo , Esferoides Celulares/patologia
6.
Methods Mol Biol ; 1981: 351-362, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31016666

RESUMO

To mimic (human) cholestasis in vitro requires multiple triggers to establish a diseased phenotype. However, this is currently not simulated by existing in vitro models. Therefore, there is a high need for multicellular systems similar to the human physiology. In such an in vitro model, cell-cell interactions and intact bile canaliculi with functional bile flow should be present and preserved during long-term culture. Precision-cut liver slices represent an ex vivo tissue culture technique that replicates most of the multicellular characteristics of a whole liver in vivo. This chapter describes the preparation and culturing of (human) precision-cut liver slices. Furthermore, a protocol to use the precision-cut liver slices technique to predict drug-induced cholestatic liver injury is described.


Assuntos
Doença Hepática Induzida por Substâncias e Drogas/metabolismo , Doença Hepática Induzida por Substâncias e Drogas/patologia , Colestase/metabolismo , Colestase/patologia , Canalículos Biliares/metabolismo , Canalículos Biliares/patologia , Humanos , Fígado/metabolismo , Fígado/patologia
7.
Artigo em Inglês | MEDLINE | ID: mdl-32010639

RESUMO

Plasmodium parasites undergo a dramatic transformation during the liver stage of their life cycle, amplifying over 10,000-fold inside infected hepatocytes within a few days. Such a rapid growth requires large-scale interactions with, and manipulations of, host cell functions. Whereas hepatocyte polarity is well-known to be critical for liver function, little is presently known about its involvement during the liver stage of Plasmodium development. Apical domains of hepatocytes are critical components of their polarity machinery and constitute the bile canalicular network, which is central to liver function. Here, we employed high resolution 3-D imaging and advanced image analysis of Plasmodium-infected liver tissues to show that the parasite associates preferentially with the apical domain of hepatocytes and induces alterations in the organization of these regions, resulting in localized changes in the bile canalicular architecture in the liver tissue. Pharmacological perturbation of the bile canalicular network by modulation of AMPK activity reduces the parasite's association with bile canaliculi and arrests the parasite development. Our findings using Plasmodium-infected liver tissues reveal a host-Plasmodium interaction at the level of liver tissue organization. We demonstrate for the first time a role for bile canaliculi, a central component of the hepatocyte polarity machinery, during the liver stage of Plasmodium development.


Assuntos
Hepatócitos/parasitologia , Interações Hospedeiro-Patógeno/fisiologia , Fígado/parasitologia , Malária/parasitologia , Plasmodium berghei/fisiologia , Animais , Ácidos e Sais Biliares/análise , Canalículos Biliares/diagnóstico por imagem , Canalículos Biliares/parasitologia , Canalículos Biliares/patologia , Modelos Animais de Doenças , Imageamento Tridimensional , Estágios do Ciclo de Vida , Fígado/diagnóstico por imagem , Fígado/patologia , Malária/diagnóstico por imagem , Malária/patologia , Camundongos , Camundongos Endogâmicos C57BL
8.
J Vis Exp ; (154)2019 12 20.
Artigo em Inglês | MEDLINE | ID: mdl-31904017

RESUMO

Hepatocytes are the central cells of the liver responsible for its metabolic function. As such, they form a uniquely polarized epithelium, in which two or more hepatocytes contribute apical membranes to form a bile canalicular network through which bile is secreted. Hepatocyte polarization is essential for correct canalicular formation and depends on interactions between the hepatocyte cytoskeleton, cell-cell contacts, and the extracellular matrix. In vitro studies of hepatocyte cytoskeleton involvement in canaliculi formation and its response to pathological situations are handicapped by the lack of cell culture, which would closely resemble the canaliculi network structure in vivo. Described here is a protocol for the isolation of mouse hepatocytes from the adult mouse liver using a modified collagenase perfusion technique. Also described is the production of culture in a 3D collagen sandwich setting, which is used for immunolabeling of cytoskeletal components to study bile canalicular formation and its response to treatments in vitro. It is shown that hepatocyte 3D collagen sandwich cultures respond to treatments with toxins (ethanol) or actin cytoskeleton altering drugs (e.g., blebbistatin) and serve as a valuable tool for in vitro studies of bile canaliculi formation and function.


Assuntos
Canalículos Biliares/patologia , Colágeno/metabolismo , Citoesqueleto/metabolismo , Hepatócitos/patologia , Citoesqueleto de Actina , Actinas/metabolismo , Animais , Bile/metabolismo , Canalículos Biliares/metabolismo , Transporte Biológico , Membrana Celular/metabolismo , Células Cultivadas , Matriz Extracelular/metabolismo , Hepatócitos/metabolismo , Camundongos , Microtúbulos/metabolismo
9.
Sci Rep ; 8(1): 16115, 2018 10 31.
Artigo em Inglês | MEDLINE | ID: mdl-30382126

RESUMO

HepaRG is an original human hepatoma cell line, acquiring highly differentiated hepatic features when exposed to dimethylsulfoxide (DMSO). To search alternatives to DMSO, which may exert some toxicity, we have analyzed the effects of forskolin (FSK), a cAMP-generating agent known to favor differentiation of various cell types. FSK used at 50 µM for 3 days was found to promote polarization of high density-plated HepaRG cells, i.e., it markedly enhanced the formation of functional biliary canaliculi structures. It also increased expressions of various hepatic markers, including those of cytochrome P-450 (CYP) 3A4, of drug transporters like NTCP, OATP2B1 and BSEP, and of metabolism enzymes like glucose 6-phosphatase. In addition, FSK-treated HepaRG cells displayed enhanced activities of CYP3A4, NTCP and OATPs when compared to untreated cells. These polarizing/differentiating effects of FSK were next shown to reflect not only the generation of cAMP, but also the activation of the xenobiotic sensing receptors PXR and FXR by FSK. Co-treatment of HepaRG cells by the cAMP analog Sp-5,6-DCl-cBIMPS and the reference PXR agonist rifampicin reproduced the polarizing effects of FSK. Therefore, FSK may be considered as a relevant alternative to DMSO for getting polarized and differentiated HepaRG cells, notably for pharmacological and toxicological studies.


Assuntos
Carcinoma Hepatocelular/patologia , Polaridade Celular , Colforsina/farmacologia , Neoplasias Hepáticas/patologia , Canalículos Biliares/efeitos dos fármacos , Canalículos Biliares/patologia , Biomarcadores Tumorais/metabolismo , Carcinoma Hepatocelular/genética , Linhagem Celular Tumoral , Polaridade Celular/efeitos dos fármacos , AMP Cíclico/análogos & derivados , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Fígado/efeitos dos fármacos , Fígado/metabolismo , Fígado/patologia , Neoplasias Hepáticas/genética , Receptor de Pregnano X/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Rifampina/farmacologia , Transdução de Sinais
10.
Toxicol Lett ; 295: 187-194, 2018 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-29913214

RESUMO

Cholestasis represents a major subtype of drug-induced liver injury and novel preclinical models for its prediction are needed. Here we used primary human hepatocytes (PHH) from different donors in 2D-sandwich (2D-sw) and/or 3D-spheroid cultures to study inter-individual differences in the response towards cholestatic hepatotoxins after short-term (48-72 hours) and long-term repeated exposures (14 days). The cholestatic liabilities of drugs were determined by comparing cell viability upon exposure to the highest non-cytotoxic drug concentration in the presence and absence of a non-cytotoxic concentrated bile acid mixture. In 2D-sw culture, cyclosporine A and amiodarone presented clear cholestatic liabilities in all four PHH donors tested, whereas differences in the susceptibility of the various PHH donors towards the cholestatic toxicity of bosentan, chlorpromazine and troglitazone were observed. In PHH from one donor, the cholestatic liabilities of chlorpromazine and troglitazone could only be detected after long-term repeated exposures when maintained in 3D-spheroid culture, but not after short-term exposures in either 2D-sw or 3D-spheroid culture, suggesting that cholestatic hepatotoxicity may require time to develop. In conclusion, inter-individual susceptibility exists towards drug-induced cholestasis, which depends on the compound as well as the exposure time.


Assuntos
Canalículos Biliares/efeitos dos fármacos , Variação Biológica da População , Doença Hepática Induzida por Substâncias e Drogas/etiologia , Colestase/induzido quimicamente , Hepatócitos/efeitos dos fármacos , Idoso , Canalículos Biliares/metabolismo , Canalículos Biliares/patologia , Células Cultivadas , Doença Hepática Induzida por Substâncias e Drogas/metabolismo , Doença Hepática Induzida por Substâncias e Drogas/patologia , Colestase/metabolismo , Colestase/patologia , Relação Dose-Resposta a Droga , Feminino , Hepatócitos/metabolismo , Hepatócitos/patologia , Humanos , Masculino , Pessoa de Meia-Idade , Cultura Primária de Células , Medição de Risco , Esferoides Celulares , Fatores de Tempo
11.
Pharmacol Res ; 125(Pt B): 105-113, 2017 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-28889972

RESUMO

AMP-activated protein kinase (AMPK), recognized as an energy sensor with three heterotrimeric subunits (α, ß and γ), not only maintains basal intracellular adenosine triphosphate levels but also regulates energy-intensive pathological responses, such as neurodegenerative and metabolic diseases, through multiple signaling pathways. Recent studies open a new direction for AMPK research and demonstrate that AMPK is a critical player in the pathogenesis of cholestatic liver injury and plays paradoxical roles in the regulation of different pathological processes, including the disruption of bile acid homeostasis and the regulation of hepatic polarity, inflammation and fibrosis. In the present review, we summarize recent findings that implicate AMPK-mediated signaling pathways in the pathogenesis of cholestatic liver injury. These findings provide novel insight regarding the potential use of AMPK as a therapeutic target for the treatment of cholestatic liver injury.


Assuntos
Proteínas Quinases Ativadas por AMP/metabolismo , Colestase/metabolismo , Animais , Ácidos e Sais Biliares/metabolismo , Canalículos Biliares/patologia , Microbioma Gastrointestinal , Humanos , Cirrose Hepática/metabolismo
12.
Ann Surg Oncol ; 24(9): 2456-2464, 2017 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-28612126

RESUMO

BACKGROUND: We studied histologic changes of bile canalicular-ductule networks in the future liver remnant (FLR) while associating liver partition and portal vein occlusion for staged hepatectomy (ALPPS), since little is known about regeneration of these networks during the relatively short interval between procedures in ALPPS. METHODS: Bile canalicular-ductule networks were examined in specimens from eight patients treated with ALPPS and six patients undergoing hepatectomy following portal vein embolization (PVE). Expression of multidrug resistance-1 (MDR1), a membrane transporter in bile canaliculi (BC), was analyzed immunohistochemistcally. Morphologic changes of BC and tight junctions (TJs) adjoining BC were also assessed electron microscopically. RESULTS: Extrapolated kinetic growth of the FLR was greater during ALPPS (17.2 ± 6.8 mL/day) than after PVE (6.3 ± 3.4 mL/day; p = 0.005), and continuity of the MDR1-positive bile canalicular networks was less evident in ALPPS than PVE (p < 0.001). Electron microscopically, no significant difference was evident in numbers of BC or BC lumen size between the two groups; however, development of microvilli in BC was poorer in the ALPPS group than in the PVE group (p < 0.001). TJ/desmosome complexes were shorter in the ALPPS group (0.69 ± 0.52 µm) than in the PVE group (1.09 ± 0.50 µm; p < 0.001), and leaky TJs were seen more frequently in the ALPPS group (64.9 vs. 23.6%; p = 0.001). CONCLUSIONS: Regeneration of bile canalicular-ductule networks in the FLR was poorer in ALPPS than PVE, which may be associated with prolonged cholestasis following final hepatectomy in ALPPS.


Assuntos
Canalículos Biliares/patologia , Canalículos Biliares/ultraestrutura , Embolização Terapêutica , Hepatectomia/métodos , Neoplasias Hepáticas/terapia , Veia Porta , Membro 1 da Subfamília B de Cassetes de Ligação de ATP/metabolismo , Adulto , Idoso , Idoso de 80 Anos ou mais , Canalículos Biliares/metabolismo , Desmossomos/ultraestrutura , Feminino , Humanos , Imuno-Histoquímica , Fígado/crescimento & desenvolvimento , Masculino , Microscopia Eletrônica de Transmissão , Microvilosidades/ultraestrutura , Pessoa de Meia-Idade , Junções Íntimas/ultraestrutura
13.
Toxicol Sci ; 157(2): 451-464, 2017 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-28369585

RESUMO

Several endothelin receptor antagonists (ERAs) have been developed for the treatment of pulmonary arterial hypertension (PAH). Some of them have been related to clinical cases of hepatocellular injury (sitaxentan [SIT]) and/or cholestasis (bosentan [BOS]). We aimed to determine if ambrisentan (AMB) and macitentan (MAC), in addition to BOS and SIT, could potentially cause liver damage in man by use of human HepaRG cells. Our results showed that like BOS, MAC-induced cytotoxicity and cholestatic disorders characterized by bile canaliculi dilatation and impairment of myosin light chain kinase signaling. Macitentan also strongly inhibited taurocholic acid and carboxy-2',7'-dichlorofluorescein efflux while it had a much lower inhibitory effect on influx activity compared to BOS and SIT. Moreover, these three drugs caused decreased intracellular accumulation and parallel increased levels of total bile acids (BAs) in serum-free culture media. In addition, all drugs except AMB variably deregulated gene expression of BA transporters. In contrast, SIT was hepatotoxic without causing cholestatic damage, likely via the formation of reactive metabolites and AMB was not hepatotoxic. Together, our results show that some ERAs can be hepatotoxic and that the recently marketed MAC, structurally similar to BOS, can also cause cholestatic alterations in HepaRG cells. The absence of currently known or suspected cases of cholestasis in patients suffering from PAH treated with MAC is rationalized by the lower therapeutic doses and Cmax, and longer receptor residence time compared to BOS.


Assuntos
Colestase/induzido quimicamente , Antagonistas dos Receptores de Endotelina/toxicidade , Hepatócitos/efeitos dos fármacos , Receptores de Endotelina/metabolismo , Ácidos e Sais Biliares/metabolismo , Canalículos Biliares/efeitos dos fármacos , Canalículos Biliares/metabolismo , Canalículos Biliares/patologia , Miosinas Cardíacas/metabolismo , Técnicas de Cultura de Células , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Colestase/metabolismo , Colestase/patologia , Relação Dose-Resposta a Droga , Hepatócitos/metabolismo , Hepatócitos/patologia , Humanos , Cadeias Leves de Miosina/metabolismo
14.
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
15.
BMJ Case Rep ; 20162016 Aug 02.
Artigo em Inglês | MEDLINE | ID: mdl-27485877

RESUMO

Rift Valley fever (RVF) is a zoonotic disease that primarily affects ruminant animals and can also cause fatal disease in humans. In the current report, we present the ultrastructural changes in the liver of a man aged 60 years who died from RVF in the Aseer Central Hospital, Abha, Saudi Arabia. The main hepatic changes by transmission electron microscopy included the presence of 95-115 nm electron-dense particles consistent with RVF virions, nuclear condensation, vacuolar degeneration, lipid droplet accumulation and mitochondrial damage and dilation. There were also viral inclusion bodies with electron-dense aggregates, dilation of intercellular spaces, damage of sinusoidal microvilli with widening of space of Disse, dilation of bile canaliculi and increasing number of phagolysosomes.


Assuntos
Fígado/ultraestrutura , Febre do Vale de Rift/patologia , Vírus da Febre do Vale do Rift , Canalículos Biliares/patologia , Canalículos Biliares/ultraestrutura , Evolução Fatal , Humanos , Corpos de Inclusão Viral , Masculino , Microvilosidades , Pessoa de Meia-Idade , Fagossomos , Febre do Vale de Rift/virologia , Arábia Saudita , Vírion
16.
Nanomedicine ; 12(1): 21-32, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26542278

RESUMO

This study investigated the effects of nanoencapsulated curcumin (NEC) and praziquantel (PZQ) treatment on the resolution of periductal fibrosis (PDF) and bile canalicular (BC) abnormalities in Opisthorchis viverrini infected hamsters. Chronic O. viverrini infection (OV) was initially treated with either PZQ (OP) and subsequently treated with NEC (OP+NEC), curcumin (OP+Cur) or unloaded carriers (OP+carrier) daily for one month. OP+NEC treatment reduced the PDF by suppression of fibrotic markers (hydroxyproline content, α-SMA, CTGF, fibronectin, collagen I and III), cytokines (TGF-ß and TNF-α) and TIMP-1, 2, 3 expression and upregulation of MMP-7, 13 genes. Higher activity of NEC in reducing fibrosis compared to curcumin was also demonstrated in in vitro studies. Moreover, OP+NEC also prevented BC abnormalities and upregulated several genes involved in bile acid metabolism. These results demonstrate that NEC and PZQ treatment reduces PDF and attenuates BC defect in experimental opisthorchiasis. From the Clinical Editor: Infection by Opisthorchis viverrini leads to liver fibrosis and affects population in SE Asia. Currently, praziquantel (PZQ) is the drug of choice but this drug has significant side effects. In this study, the authors combined curcumin (NEC) and praziquantel in a nanocarrier to test the anti-oxidative effect of curcumin in an animal model. The encouraging results may pave a way for better treatment in the future.


Assuntos
Canalículos Biliares/efeitos dos fármacos , Canalículos Biliares/patologia , Curcumina/administração & dosagem , Nanocápsulas/química , Opistorquíase/tratamento farmacológico , Praziquantel/administração & dosagem , Animais , Anti-Helmínticos/administração & dosagem , Anti-Helmínticos/química , Anti-Inflamatórios não Esteroides/administração & dosagem , Anti-Inflamatórios não Esteroides/química , Canalículos Biliares/anormalidades , Cricetinae , Curcumina/química , Difusão , Combinação de Medicamentos , Fibrose/patologia , Fibrose/prevenção & controle , Nanocápsulas/administração & dosagem , Nanocápsulas/ultraestrutura , Opistorquíase/patologia , Praziquantel/química , Resultado do Tratamento
17.
Drug Metab Dispos ; 43(6): 829-35, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25788542

RESUMO

Nonalcoholic fatty liver disease (NAFLD) is the most common chronic liver disease, representing a spectrum of liver pathologies that include simple hepatic steatosis and the more advanced nonalcoholic steatohepatitis (NASH). The current study was conducted to determine whether pediatric NASH also results in altered disposition of acetaminophen (APAP) and its two primary metabolites, APAP-sulfate and APAP-glucuronide. Pediatric patients with hepatic steatosis (n = 9) or NASH (n = 3) and healthy patients (n = 12) were recruited in a small pilot study design. All patients received a single 1000-mg dose of APAP. Blood and urine samples were collected at 1, 2, and 4 hours postdose, and APAP and APAP metabolites were determined by high-performance liquid chromatography. Moreover, human liver tissues from patients diagnosed with various stages of NAFLD were acquired from the Liver Tissue Cell Distribution System to investigate the regulation of the membrane transporters, multidrug resistance-associated protein 2 and 3 (MRP2 and MRP3, respectively). Patients with the more severe disease (i.e., NASH) had increased serum and urinary levels of APAP-glucuronide along with decreased serum levels of APAP-sulfate. Moreover, an induction of hepatic MRP3 and altered canalicular localization of the biliary efflux transporter, MRP2, describes the likely mechanism for the observed increase in plasma retention of APAP-glucuronide, whereas altered regulation of sulfur activation genes may explain decreased sulfonation activity in NASH. APAP-glucuronide and APAP-sulfate disposition is altered in NASH and is likely due to hepatic membrane transporter dysregulation as well as altered intracellular sulfur activation.


Assuntos
Acetaminofen/farmacocinética , Analgésicos não Narcóticos/farmacocinética , Fígado/metabolismo , Proteínas Associadas à Resistência a Múltiplos Medicamentos/metabolismo , Hepatopatia Gordurosa não Alcoólica/metabolismo , Regulação para Cima , Acetaminofen/análogos & derivados , Acetaminofen/sangue , Acetaminofen/urina , Adolescente , Analgésicos não Narcóticos/sangue , Analgésicos não Narcóticos/urina , Canalículos Biliares/metabolismo , Canalículos Biliares/patologia , Biotransformação , Criança , Estudos de Coortes , Fígado Gorduroso/sangue , Fígado Gorduroso/metabolismo , Fígado Gorduroso/patologia , Fígado Gorduroso/urina , Feminino , Humanos , Fígado/patologia , Masculino , Proteína 2 Associada à Farmacorresistência Múltipla , Hepatopatia Gordurosa não Alcoólica/sangue , Hepatopatia Gordurosa não Alcoólica/patologia , Hepatopatia Gordurosa não Alcoólica/urina , Projetos Piloto , Transporte Proteico
18.
Am J Pathol ; 185(4): 958-68, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25688837

RESUMO

Polarized hepatocytes contain tight junctions (TJs), which are among the most important junctions for sealing the bile canalicular lumen from the sinusoidal space. Alterations in TJs are implicated in chronic cholestatic liver diseases, such as primary biliary cirrhosis and primary sclerosing cholangitis, which have lipid peroxidation marker elevations or antioxidant vitamin decreases. However, the effect of oxidative stress on hepatocyte polarity or liver morphology is unknown. We found that carbon tetrachloride (CCl4)-induced oxidative stress resulted in disassembly of TJs. Ultrastructural analysis revealed disruption in TJs, Golgi morphology, and expansion of the bile canalicular lumen size in CCl4-treated hepatocytes. The Par complex [Par-3-atypical protein kinase C (aPKC) and Par-6 ternary complex] regulates TJs and lumen formation, and the Par-3-aPKC complex formation was inhibited by CCl4 treatment. Moreover, the antioxidant compound vitamin E prohibited a CCl4-induced disturbance in TJs and Par-3-aPKC complex formation. aPKC phosphorylates Par-3 and down-regulates its own affinity with Par-3. Importantly, aPKC kinase activity and Par-3 phosphorylation were significantly increased in CCl4-treated rat livers. These results indicate that the Par-3-aPKC complex plays a crucial role in the maintenance of hepatocyte polarity and sealing of the bile canalicular lumen. Our findings suggest that bile canalicular lumen expansion might explain the presence of cholestasis in patients with primary biliary cirrhosis and primary sclerosing cholangitis.


Assuntos
Canalículos Biliares/enzimologia , Canalículos Biliares/patologia , Tetracloreto de Carbono/toxicidade , Polaridade Celular/efeitos dos fármacos , Estresse Oxidativo/efeitos dos fármacos , Proteína Quinase C/metabolismo , Animais , Canalículos Biliares/efeitos dos fármacos , Proteínas de Transporte/metabolismo , Ativação Enzimática/efeitos dos fármacos , Complexo de Golgi/efeitos dos fármacos , Complexo de Golgi/metabolismo , Complexo de Golgi/ultraestrutura , Hepatócitos/efeitos dos fármacos , Hepatócitos/metabolismo , Hepatócitos/patologia , Hepatócitos/ultraestrutura , Humanos , Fígado/efeitos dos fármacos , Fígado/patologia , Fígado/ultraestrutura , Masculino , Modelos Biológicos , Proteínas do Tecido Nervoso , Ratos Wistar , Junções Íntimas/efeitos dos fármacos , Junções Íntimas/metabolismo , Vitamina E/farmacologia
19.
J Pediatr Gastroenterol Nutr ; 60(3): 368-74, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25383786

RESUMO

OBJECTIVES: Partial external bile diversion (PEBD) is an established therapy for low-γ-glutamyl transferase (GGT) progressive familial intrahepatic cholestasis (PFIC). This study sought to determine whether the dynamics of the cholic acid (CA) and chenodeoxycholic acid (CDCA) pools in subjects with low-GGT-PFIC with successful PEBD were equivalent to those achieved with successful liver transplantation (LTX). METHODS: The kinetics of CA and CDCA metabolism were measured by stable isotope dilution in plasma samples in 5 subjects with PEBD, all with intact canalicular bile salt export pump expression and compared with subjects with low-GGT-PFIC with successful LTX. Stomal loss of bile acids was measured in subjects with PEBD. RESULTS: The fractional turnover rate for CA in the PEBD group ranged from 0.5 to 4.2/day (LTX group, range 0.2-0.9/day, P = 0.076) and for CDCA from 0.7 to 4.5/day (LTX group 0.3-0.4/day, P = 0.009). The CA and CDCA pool sizes were equivalent between groups; however, pool composition in PEBD was somewhat more hydrophilic. The CA/CDCA ratio in PEBD ranged from 0.9 to 19.5, whereas in LTX it ranged from 0.5 to 2.6. Synthesis rates computed from isotope dilution correlated well with timed output for both CA (r2 = 0.760, P = 0.024) and CDCA (r2 = 0.690, P = 0.021). CONCLUSIONS: PEBD results in bile acid fractional turnover rates greater than LTX, pool sizes equivalent to LTX, and pool composition that is at least as hydrophilic as produced by LTX.


Assuntos
Ácidos e Sais Biliares/metabolismo , Coledocostomia/efeitos adversos , Colestase Intra-Hepática/cirurgia , Fígado/metabolismo , Adolescente , Adulto , Ácidos e Sais Biliares/sangue , Canalículos Biliares/metabolismo , Canalículos Biliares/patologia , Ductos Biliares Intra-Hepáticos/cirurgia , Criança , Pré-Escolar , Colestase Intra-Hepática/sangue , Colestase Intra-Hepática/metabolismo , Colestase Intra-Hepática/patologia , Deutério , Feminino , Humanos , Interações Hidrofóbicas e Hidrofílicas , Lactente , Jejuno/cirurgia , Cinética , Fígado/patologia , Transplante de Fígado/efeitos adversos , Masculino , Técnica de Diluição de Radioisótopos , Adulto Jovem
20.
Sci Rep ; 4: 6835, 2014 Oct 30.
Artigo em Inglês | MEDLINE | ID: mdl-25355493

RESUMO

The liver is unique in its capacity to regenerate after injury, during which hepatocytes actively divide and establish cell-cell contacts through cell adhesion complexes. Here, we demonstrate that the loss of α-catenin, a well-established adhesion component, dramatically disrupts liver regeneration. Using a partial hepatectomy model, we show that regenerated livers from α-catenin knockdown mice are grossly larger than control regenerated livers, with an increase in cell size and proliferation. This increased proliferation correlated with increased YAP activation, implicating α-catenin in the Hippo/YAP pathway. Additionally, α-catenin knockdown mice exhibited a phenotype reminiscent of clinical cholestasis, with drastically altered bile canaliculi, elevated levels of bile components and signs of jaundice and inflammation. The disrupted regenerative capacity is a result of actin cytoskeletal disorganisation, leading to a loss of apical microvilli, dilated lumens in the bile canaliculi, and leaky tight junctions. This study illuminates a novel, essential role for α-catenin in liver regeneration.


Assuntos
Colestase/genética , Regeneração Hepática/fisiologia , alfa Catenina/genética , Actinas/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Animais , Canalículos Biliares/patologia , Canalículos Biliares/ultraestrutura , Proteínas de Ciclo Celular , Proliferação de Células , Colestase/sangue , Feminino , Hepatócitos/fisiologia , Camundongos , Camundongos Knockout , Microvilosidades/ultraestrutura , Modelos Animais , Fosfoproteínas/metabolismo , Proteínas de Sinalização YAP , alfa Catenina/deficiência
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA