Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 148
Filtrar
1.
Drug Metab Dispos ; 47(10): 1222-1230, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-31371422

RESUMO

Human hepatoma cell lines are useful for evaluation of drug-induced hepatotoxicity, hepatic drug disposition, and drug-drug interactions. However, their applicability is compromised by aberrant expression of hepatobiliary transporters. This study was designed to evaluate whether extracellular matrix (Matrigel) overlay and dexamethasone (DEX) treatment would support cellular maturation of long-term HuH-7 hepatoma cell cultures and improve the expression, localization, and activity of canalicular ATP-binding cassette (ABC) transporters, multidrug resistance protein 1 (MDR1/P-glycoprotein/ABCB1), multidrug resistance-associated protein 2 (MRP2/ABCC2), and bile salt export pump (BSEP/ABCB11). Matrigel overlay promoted the maturation of HuH-7 cells toward cuboidal, hepatocyte-like cells displaying bile canaliculi-like structures visualized by staining for filamentous actin (F-actin), colocalization of MRP2 with F-actin, and by accumulation of the MRP2 substrate 5(6)-carboxy-2',7'-dichlorofluorescein (CDF) within the tubular canaliculi. The cellular phenotype was rather homogenous in the Matrigel-overlaid cultures, whereas the standard HuH-7 cultures contained both hepatocyte-like cells and flat epithelium-like cells. Only Matrigel-overlaid HuH-7 cells expressed MDR1 at the canaliculi and excreted the MDR1 probe substrate digoxin into biliary compartments. DEX treatment resulted in more elongated and branched canaliculi and restored canalicular expression and function of BSEP. These findings suggest that hepatocyte polarity, elongated canalicular structures, and proper localization and function of canalicular ABC transporters can be recovered, at least in part, in human hepatoma HuH-7 cells by applying the modified culture conditions. SIGNIFICANCE STATEMENT: We report the first demonstration that proper localization and function of canalicular ABC transporters can be recovered in human hepatoma HuH-7 cells by modification of cell culture conditions. Matrigel overlay and dexamethasone supplementation increased the proportion of hepatocyte-like cells, strongly augmented the canalicular structures between the cells, and restored the localization and function of key canalicular ABC transporters. These results will facilitate the development of reproducible, economical, and easily achievable liver cell models for drug development.


Assuntos
Transportadores de Cassetes de Ligação de ATP/metabolismo , Canalículos Biliares/metabolismo , Técnicas de Cultura de Células/métodos , Meios de Cultura/farmacologia , Canalículos Biliares/efeitos dos fármacos , Linhagem Celular Tumoral , Doença Hepática Induzida por Substâncias e Drogas/etiologia , Doença Hepática Induzida por Substâncias e Drogas/prevenção & controle , Colágeno/farmacologia , Dexametasona/farmacologia , Combinação de Medicamentos , Avaliação Pré-Clínica de Medicamentos/métodos , Interações Medicamentosas , Humanos , Laminina/farmacologia , Proteína 2 Associada à Farmacorresistência Múltipla , Proteoglicanas/farmacologia
2.
J Cell Sci ; 129(11): 2190-201, 2016 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-27034138

RESUMO

The Cu(+) pump ATP7B plays an irreplaceable role in the elimination of excess Cu(+) by the hepatocyte into the bile. The trafficking and site of action of ATP7B are subjects of controversy. One current proposal is that an increase in intracellular Cu(+) results in the translocation of ATP7B to the lysosomes and excretion of excess Cu(+) through lysosomal-mediated exocytosis at the bile canaliculus. Here, we show that ATP7B is transported from the trans-Golgi network (TGN) to the bile canaliculus by basolateral sorting and endocytosis, and microtubule-mediated transcytosis through the subapical compartment. Trafficking ATP7B is not incorporated into lysosomes, and addition of Cu(+) does not cause relocalization of lysosomes and the appearance of lysosome markers in the bile canaliculus. Our data reveal the pathway of the Cu(+)-mediated transport of ATP7B from the TGN to the bile canaliculus and indicates that the bile canaliculus is the primary site of ATP7B action in the elimination of excess Cu(.)


Assuntos
Adenosina Trifosfatases/metabolismo , Canalículos Biliares/metabolismo , Proteínas de Transporte de Cátions/metabolismo , Cobre/farmacologia , Transcitose/efeitos dos fármacos , Animais , Canalículos Biliares/efeitos dos fármacos , Brefeldina A/farmacologia , Compartimento Celular/efeitos dos fármacos , Membrana Celular/efeitos dos fármacos , Membrana Celular/metabolismo , ATPases Transportadoras de Cobre , Fatores de Troca do Nucleotídeo Guanina/metabolismo , Células Hep G2 , Humanos , Hidrazonas/farmacologia , Lisossomos/efeitos dos fármacos , Lisossomos/metabolismo , Macrolídeos/farmacologia , Microtúbulos/efeitos dos fármacos , Microtúbulos/metabolismo , Transporte Proteico/efeitos dos fármacos , Ratos , Vesículas Secretórias/efeitos dos fármacos , Vesículas Secretórias/metabolismo , Rede trans-Golgi/efeitos dos fármacos , Rede trans-Golgi/metabolismo
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.
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
5.
Drug Metab Dispos ; 44(11): 1780-1793, 2016 11.
Artigo em Inglês | MEDLINE | ID: mdl-27538918

RESUMO

Intrahepatic cholestasis represents 20%-40% of drug-induced injuries from which a large proportion remains unpredictable. We aimed to investigate mechanisms underlying drug-induced cholestasis and improve its early detection using human HepaRG cells and a set of 12 cholestatic drugs and six noncholestatic drugs. In this study, we analyzed bile canaliculi dynamics, Rho kinase (ROCK)/myosin light chain kinase (MLCK) pathway implication, efflux inhibition of taurocholate [a predominant bile salt export pump (BSEP) substrate], and expression of the major canalicular and basolateral bile acid transporters. We demonstrated that 12 cholestatic drugs classified on the basis of reported clinical findings caused disturbances of both bile canaliculi dynamics, characterized by either dilatation or constriction, and alteration of the ROCK/MLCK signaling pathway, whereas noncholestatic compounds, by contrast, had no effect. Cotreatment with ROCK inhibitor Y-27632 [4-(1-aminoethyl)-N-(4-pyridyl) cyclohexanecarboxamide dihydrochloride] and MLCK activator calmodulin reduced bile canaliculi constriction and dilatation, respectively, confirming the role of these pathways in drug-induced intrahepatic cholestasis. By contrast, inhibition of taurocholate efflux and/or human BSEP overexpressed in membrane vesicles was not observed with all cholestatic drugs; moreover, examples of noncholestatic compounds were reportedly found to inhibit BSEP. Transcripts levels of major bile acid transporters were determined after 24-hour treatment. BSEP, Na+-taurocholate cotransporting polypeptide, and organic anion transporting polypeptide B were downregulated with most cholestatic and some noncholestatic drugs, whereas deregulation of multidrug resistance-associated proteins was more variable, probably mainly reflecting secondary effects. Together, our results show that cholestatic drugs consistently cause an early alteration of bile canaliculi dynamics associated with modulation of ROCK/MLCK and these changes are more specific than efflux inhibition measurements alone as predictive nonclinical markers of drug-induced cholestasis.


Assuntos
Canalículos Biliares/metabolismo , Colestase Intra-Hepática/metabolismo , Fígado/metabolismo , Quinase de Cadeia Leve de Miosina/metabolismo , Quinases Associadas a rho/metabolismo , Amidas/farmacologia , Ácidos e Sais Biliares/metabolismo , Canalículos Biliares/efeitos dos fármacos , Transporte Biológico/efeitos dos fármacos , Transporte Biológico/fisiologia , Proteínas de Transporte/metabolismo , Linhagem Celular , Colestase Intra-Hepática/induzido quimicamente , Humanos , Fígado/efeitos dos fármacos , Glicoproteínas de Membrana/metabolismo , Proteínas Associadas à Resistência a Múltiplos Medicamentos/metabolismo , Piridinas/farmacologia , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia , Ácido Taurocólico/metabolismo
6.
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
7.
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
8.
Arch Toxicol ; 89(10): 1861-70, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26280096

RESUMO

Many substances are hepatotoxic due to their ability to cause intrahepatic cholestasis. Therefore, there is a high demand for in vitro systems for the identification of cholestatic properties of new compounds. Primary hepatocytes cultivated in collagen sandwich cultures are known to establish bile canaliculi which enclose secreted biliary components. Cholestatic compounds are mainly known to inhibit bile excretion dynamics, but may also alter canalicular volume, or hepatocellular morphology. So far, techniques to assess time-resolved morphological changes of bile canaliculi in sandwich cultures are not available. In this study, we developed an automated system that quantifies dynamics of bile canaliculi recorded in conventional time-lapse image sequences. We validated the hepatocyte sandwich culture system as an appropriate model to study bile canaliculi in vitro by showing structural similarity measured as bile canaliculi length per hepatocyte to that observed in vivo. Moreover, bile canalicular excretion kinetics of CMFDA (5-chloromethylfluorescein diacetate) in sandwich cultures resembled closely the kinetics observed in vivo. The developed quantification technique enabled the quantification of dynamic changes in individual bile canaliculi. With this technique, we were able to clearly distinguish between sandwich cultures supplemented with dexamethasone and insulin from control cultures. In conclusion, the automated quantification system offers the possibility to systematically study the causal relationship between disturbed bile canalicular dynamics and cholestasis.


Assuntos
Canalículos Biliares/efeitos dos fármacos , Técnicas de Cultura de Células , Colágeno/química , Hepatócitos/efeitos dos fármacos , Animais , Canalículos Biliares/metabolismo , Células Cultivadas , Doença Hepática Induzida por Substâncias e Drogas/diagnóstico , Colestase Intra-Hepática/induzido quimicamente , Dexametasona/administração & dosagem , Fluoresceínas/farmacocinética , Hepatócitos/metabolismo , Insulina/administração & dosagem , Masculino , Camundongos , Camundongos Endogâmicos C57BL
9.
Arch Toxicol ; 89(6): 979-90, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24912783

RESUMO

At present, it has not been systematically evaluated whether the functional alterations induced by cholestatic compounds in canalicular transporters involved in bile formation can be reproduced in sandwich-cultured rat hepatocytes (SCRHs). Here, we focused on two clinically relevant cholestatic agents, such as estradiol 17ß-D-glucuronide (E17G) and taurolithocholate (TLC), also testing the ability of dibutyryl cyclic AMP (DBcAMP) to prevent their effects. SCRHs were incubated with E17G (200 µM) or TLC (2.5 µM) for 30 min, with or without pre-incubation with DBcAMP (10 µM) for 15 min. Then, the increase in glutathione methyl fluorescein (GS-MF)-associated fluorescence inside the canaliculi was monitored by quantitative time-lapse imaging, and Mrp2 transport activity was calculated by measuring the slope of the time-course fluorescence curves during the initial linear phase, which was considered to be the Mrp2-mediated initial transport rate (ITR). E17G and TLC impaired canalicular bile formation, as evidenced by a decrease in both the bile canaliculus volume and the bile canaliculus width, estimated from 3D and 2D confocal images, respectively. These compounds decreased ITR and induced retrieval of Mrp2, a main pathomechanism involved in their cholestatic effects. Finally, DBcAMP prevented these effects, and its well-known choleretic effect was evident from the increase in the canalicular volume/width values; this choleretic effect is associated in part with its capability to increase Mrp2 activity, evidenced here by the increase in ITR of GS-MF. Our study supports the use of SCRHs as an in vitro model useful to quantify canalicular transport function under conditions of cholestasis and choleresis.


Assuntos
Canalículos Biliares/metabolismo , Bile/metabolismo , Transporte Biológico , Colestase/metabolismo , Hepatócitos/metabolismo , Modelos Biológicos , Animais , Canalículos Biliares/efeitos dos fármacos , Bucladesina/farmacologia , Técnicas de Cultura de Células , Células Cultivadas , Colestase/induzido quimicamente , Estradiol/análogos & derivados , Estradiol/farmacologia , Hepatócitos/efeitos dos fármacos , Proteína 2 Associada à Farmacorresistência Múltipla , Proteínas Associadas à Resistência a Múltiplos Medicamentos/metabolismo , Ratos , Ácido Taurolitocólico/farmacologia
10.
Arch Toxicol ; 88(5): 1083-95, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24599296

RESUMO

Immortalized hepatocyte cell lines show only a weak resemblance to primary hepatocytes in terms of gene expression and function, limiting their value in predicting drug-induced liver injury (DILI). Furthermore, primary hepatocytes cultured on two-dimensional tissue culture plastic surfaces rapidly dedifferentiate losing their hepatocyte functions and metabolic competence. We have developed a three-dimensional in vitro model using extracellular matrix-based hydrogel for long-term culture of the human hepatoma cell line HepG2. HepG2 cells cultured in this model stop proliferating, self-organize and differentiate to form multiple polarized spheroids. These spheroids re-acquire lost hepatocyte functions such as storage of glycogen, transport of bile salts and the formation of structures resembling bile canaliculi. HepG2 spheroids also show increased expression of albumin, urea, xenobiotic transcription factors, phase I and II drug metabolism enzymes and transporters. Consistent with this, cytochrome P450-mediated metabolism is significantly higher in HepG2 spheroids compared to monolayer cultures. This highly differentiated phenotype can be maintained in 384-well microtiter plates for at least 28 days. Toxicity assessment studies with this model showed an increased sensitivity in identifying hepatotoxic compounds with repeated dosing regimens. This simple and robust high-throughput-compatible methodology may have potential for use in toxicity screening assays and mechanistic studies and may represent an alternative to animal models for studying DILI.


Assuntos
Células Hep G2/efeitos dos fármacos , Ensaios de Triagem em Larga Escala/métodos , Testes de Toxicidade/métodos , Albuminas/metabolismo , Canalículos Biliares/efeitos dos fármacos , Canalículos Biliares/metabolismo , Diferenciação Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Sistema Enzimático do Citocromo P-450/metabolismo , Humanos , Inativação Metabólica/genética , Fígado/metabolismo , Esferoides Celulares , Ureia/metabolismo
11.
Am J Physiol Gastrointest Liver Physiol ; 305(4): G286-94, 2013 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-23764895

RESUMO

The bile salt export pump (BSEP), encoded by the abcb11 gene, is the major canalicular transporter of bile acids from the hepatocyte. BSEP malfunction in humans causes bile acid retention and progressive liver injury, ultimately leading to end-stage liver failure. The natural, hydrophilic, bile acid ursodeoxycholic acid (UDCA) is efficacious in the treatment of cholestatic conditions, such as primary biliary cirrhosis and cholestasis of pregnancy. The beneficial effects of UDCA include promoting bile flow, reducing hepatic inflammation, preventing apoptosis, and maintaining mitochondrial integrity in hepatocytes. However, the role of BSEP in mediating UDCA efficacy is not known. Here, we used abcb11 knockout mice (abcb11-/-) to test the effects of acute and chronic UDCA administration on biliary secretion, bile acid composition, liver histology, and liver gene expression. Acutely infused UDCA, or its taurine conjugate (TUDC), was taken up by the liver but retained, with negligible biliary output, in abcb11-/- mice. Feeding UDCA to abcb11-/- mice led to weight loss, retention of bile acids, elevated liver enzymes, and histological damage to the liver. Semiquantitative RT-PCR showed that genes encoding Mdr1a and Mdr1b (canalicular) as well as Mrp4 (basolateral) transporters were upregulated in abcb11-/- mice. We concluded that infusion of UDCA and TUDC failed to induce bile flow in abcb11-/- mice. UDCA fed to abcb11-/- mice caused liver damage and the appearance of biliary tetra- and penta-hydroxy bile acids. Supplementation with UDCA in the absence of Bsep caused adverse effects in abcb11-/- mice.


Assuntos
Transportadores de Cassetes de Ligação de ATP/deficiência , Canalículos Biliares/efeitos dos fármacos , Colestase/metabolismo , Dieta , Fígado/efeitos dos fármacos , Ácido Tauroquenodesoxicólico/toxicidade , Ácido Ursodesoxicólico/toxicidade , Subfamília B de Transportador de Cassetes de Ligação de ATP/genética , Subfamília B de Transportador de Cassetes de Ligação de ATP/metabolismo , Membro 11 da Subfamília B de Transportadores de Cassetes de Ligação de ATP , Transportadores de Cassetes de Ligação de ATP/genética , Animais , Canalículos Biliares/metabolismo , Canalículos Biliares/patologia , Transporte Biológico , Colestase/genética , Colestase/patologia , Modelos Animais de Doenças , Regulação da Expressão Gênica , Infusões Intravenosas , Fígado/metabolismo , Fígado/patologia , Camundongos , Camundongos Knockout , Proteínas Associadas à Resistência a Múltiplos Medicamentos/genética , Proteínas Associadas à Resistência a Múltiplos Medicamentos/metabolismo , RNA Mensageiro/metabolismo , Ácido Tauroquenodesoxicólico/administração & dosagem , Ácido Tauroquenodesoxicólico/metabolismo , Fatores de Tempo , Ácido Ursodesoxicólico/administração & dosagem , Ácido Ursodesoxicólico/metabolismo , Membro 4 da Subfamília B de Transportadores de Cassetes de Ligação de ATP
12.
Mol Pharmacol ; 79(2): 241-50, 2011 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-21056966

RESUMO

Biliary secretion of bile acids and phospholipids, both of which are essential components of biliary micelles, are mediated by the bile salt export pump (BSEP/ABCB11) and multidrug resistance 3 P-glycoprotein (MDR3/ABCB4), respectively, and their genetic dysfunction leads to the acquisition of severe cholestatic diseases. In the present study, we found two patients with itraconazole (ITZ)-induced cholestatic liver injury with markedly high serum ITZ concentrations. To characterize the effect of ITZ on bile formation in vivo, biliary bile acids and phospholipids were analyzed in ITZ-treated rats, and it was revealed that biliary phospholipids, rather than bile acids, were drastically reduced in the presence of clinically relevant concentrations of ITZ. Moreover, by using MDR3-expressing LLC-PK1 cells, we found that MDR3-mediated efflux of [¹4C]phosphatidylcholine was significantly reduced by ITZ. In contrast, BSEP-mediated transport of [³H]taurocholate was not significantly affected by ITZ, which is consistent with our in vivo observations. In conclusion, this study suggests the involvement of the inhibition of MDR3-mediated biliary phospholipids secretion in ITZ-induced cholestasis. Our approach may be useful for analyzing mechanisms of drug-induced cholestasis and evaluating the cholestatic potential of clinically used drugs and drug candidates.


Assuntos
Subfamília B de Transportador de Cassetes de Ligação de ATP/antagonistas & inibidores , Antifúngicos/toxicidade , Canalículos Biliares/efeitos dos fármacos , Colestase/induzido quimicamente , Itraconazol/toxicidade , Subfamília B de Transportador de Cassetes de Ligação de ATP/metabolismo , Animais , Antifúngicos/sangue , Canalículos Biliares/metabolismo , Western Blotting , Itraconazol/sangue , Células LLC-PK1 , Masculino , Ratos , Ratos Sprague-Dawley
13.
J Hepatol ; 55(6): 1368-76, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21703191

RESUMO

BACKGROUND & AIMS: Canalicular phosphatidylcholine and cholesterol secretion requires the coordinate action of the ATP binding cassette transporters: the bile salt export pump (Bsep) for bile salts (BS) and the phosphatidylcholine translocator multidrug resistance protein 2 (Mdr2). After their secretion, phosphatidylcholine and BS form mixed micelles acting as acceptors for canalicular cholesterol. We have shown that the canalicular liver plasma membrane (cLPM) contains lipid raft enriched in sphingomyelin and cholesterol. As BS have detergent properties and their concentration in the canaliculus is very high, we tested the hypothesis that the canalicular membrane contains BS resistant microdomains. METHODS: Isolated cLPMs were extracted at 4°C with different BS or detergents and subjected to flotation in sucrose step gradients followed by Western blotting and lipid composition analysis. RESULTS: Incubating cLPMs with increasing taurocholate concentrations revealed the presence of BS resistant microdomains. These microdomains were found with different BS in the presence and absence of lipids and contained the raft markers reggie-1/-2 and caveolin-1 and canalicular transporters Bsep, Mrp2, and Abcg5, the latter independent of the presence of lipids. BS resistant microdomains contain mainly cholesterol, phosphatidylcholine, and phosphatidylethanolamine. Extraction of cLPMs with a mixture of different BS similar to rat bile revealed a comparable microdomain composition. CONCLUSIONS: cLPM contains BS resistant microdomains potentially protecting the cLPM against the detergent action of BS. Combination of different BS has no synergistic effect on microdomain composition.


Assuntos
Ácidos e Sais Biliares/metabolismo , Canalículos Biliares/metabolismo , Transportadores de Cassetes de Ligação de ATP/metabolismo , Animais , Ácidos e Sais Biliares/farmacologia , Canalículos Biliares/efeitos dos fármacos , Caveolina 1/metabolismo , Colesterol/metabolismo , Detergentes/farmacologia , Técnicas In Vitro , Masculino , Microdomínios da Membrana/metabolismo , Proteínas de Membrana/metabolismo , Ratos , Ratos Sprague-Dawley , Esfingomielinas/metabolismo , Ácido Taurocólico/metabolismo
14.
Hepatology ; 52(4): 1410-9, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-20722001

RESUMO

UNLABELLED: Beta-catenin plays important roles in liver physiology and hepatocarcinogenesis. While studying the role of ß-catenin in diet-induced steatohepatitis, we recently found that liver-specific ß-catenin knockout (KO) mice exhibit intrahepatic cholestasis. This study was undertaken to further characterize the role of ß-catenin in biliary physiology. KO mice and wild-type (WT) littermates were fed standard chow or a diet supplemented with 0.5% cholic acid for 2 weeks. Chow-fed KO mice had higher serum and hepatic total bile acid levels and lower bile flow rate than WT mice. Expression levels of bile acid biosynthetic genes were lower and levels of major bile acid exporters were similar, which therefore could not explain the KO phenotype. Despite loss of the tight junction protein claudin-2, KO mice had preserved functional integrity of tight junctions. KO mice had bile canalicular morphologic abnormalities as evidenced by staining for F-actin and zona occludens 1. Electron microscopy revealed dilated and tortuous bile canaliculi in KO livers along with decreased canalicular and sinusoidal microvilli. KO mice on a cholic acid diet had higher hepatic and serum bile acid levels, bile ductular reaction, increased pericellular fibrosis, and dilated, misshapen bile canaliculi. Compensatory changes in expression levels of several bile acid transporters and regulatory genes were found in KO livers. CONCLUSION: Liver-specific loss of ß-catenin leads to defective bile canalicular morphology, bile secretory defect, and intrahepatic cholestasis. Thus, our results establish a critical role for ß-catenin in biliary physiology.


Assuntos
Ácidos e Sais Biliares/metabolismo , Canalículos Biliares/patologia , Colestase Intra-Hepática/metabolismo , Fígado/metabolismo , beta Catenina/deficiência , Animais , Bile/metabolismo , Ácidos e Sais Biliares/sangue , Canalículos Biliares/efeitos dos fármacos , Canalículos Biliares/metabolismo , Ácido Cólico/farmacologia , Dieta , Feminino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout
15.
Hepatology ; 52(4): 1465-76, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-20815017

RESUMO

UNLABELLED: Estradiol 17ß-D-glucuronide (E(2)17G) is an endogenous, cholestatic metabolite that induces endocytic internalization of the canalicular transporters relevant to bile secretion: bile salt export pump (Bsep) and multidrug resistance-associated protein 2 (Mrp2). We assessed whether phosphoinositide 3-kinase (PI3K) is involved in E(2)17G-induced cholestasis. E(2)17G activated PI3K according to an assessment of the phosphorylation of the final PI3K effector, protein kinase B (Akt). When the PI3K inhibitor wortmannin (WM) was preadministered to isolated rat hepatocyte couplets (IRHCs), it partially prevented the reduction induced by E(2)17G in the proportion of IRHCs secreting fluorescent Bsep and Mrp2 substrates (cholyl lysyl fluorescein and glutathione methylfluorescein, respectively). 2-Morpholin-4-yl-8-phenylchromen-4-one, another PI3K inhibitor, and an Akt inhibitor (Calbiochem 124005) showed similar protective effects. IRHC immunostaining and confocal microscopy analysis revealed that endocytic internalization of Bsep and Mrp2 induced by E(2)17G was extensively prevented by WM; this effect was fully blocked by the microtubule-disrupting agent colchicine. The protection of WM was additive to that afforded by the classical protein kinase C (cPKC) inhibitor 5,6,7,13-tetrahydro-13-methyl-5-oxo-12H-indolo[2,3-a]pyrrolo[3,4-c]carbazole-12-propanenitrile (Gö6976); this suggested differential and complementary involvement of the PI3K and cPKC signaling pathways in E(2)17G-induced cholestasis. In isolated perfused rat liver, an intraportal injection of E(2)17G triggered endocytosis of Bsep and Mrp2, and this was accompanied by a sustained decrease in the bile flow and the biliary excretion of the Bsep and Mrp2 substrates [(3)H]taurocholate and glutathione until the end of the perfusion period. Unlike Gö6976, WM did not prevent the initial decay, but it greatly accelerated the recovery to normality of these parameters and the reinsertion of Bsep and Mrp2 into the canalicular membrane in a microtubule-dependent manner. CONCLUSION: The PI3K/Akt signaling pathway is involved in the biliary secretory failure induced by E(2)17G through sustained internalization of canalicular transporters endocytosed via cPKC.


Assuntos
1-Fosfatidilinositol 4-Quinase/fisiologia , Colestase/induzido quimicamente , Proteína Quinase C/fisiologia , Proteínas Proto-Oncogênicas c-akt/fisiologia , Membro 11 da Subfamília B de Transportadores de Cassetes de Ligação de ATP , Transportadores de Cassetes de Ligação de ATP/metabolismo , Androstadienos/farmacologia , Animais , Canalículos Biliares/efeitos dos fármacos , Canalículos Biliares/fisiologia , Sistema Biliar/metabolismo , Carbazóis/farmacologia , Colchicina/farmacologia , Endocitose/efeitos dos fármacos , Estradiol/análogos & derivados , Glutationa/metabolismo , Técnicas In Vitro , Masculino , Microtúbulos/efeitos dos fármacos , Microtúbulos/fisiologia , Proteína 2 Associada à Farmacorresistência Múltipla , Proteínas Associadas à Resistência a Múltiplos Medicamentos/metabolismo , Perfusão , Proteína Quinase C/antagonistas & inibidores , Ratos , Ratos Wistar , Transdução de Sinais , Ácido Taurocólico/metabolismo , Wortmanina
16.
Clin Sci (Lond) ; 121(12): 523-44, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21854363

RESUMO

UDCA (ursodeoxycholic acid) is the therapeutic agent most widely used for the treatment of cholestatic hepatopathies. Its use has expanded to other kinds of hepatic diseases, and even to extrahepatic ones. Such versatility is the result of its multiple mechanisms of action. UDCA stabilizes plasma membranes against cytolysis by tensioactive bile acids accumulated in cholestasis. UDCA also halts apoptosis by preventing the formation of mitochondrial pores, membrane recruitment of death receptors and endoplasmic-reticulum stress. In addition, UDCA induces changes in the expression of metabolizing enzymes and transporters that reduce bile acid cytotoxicity and improve renal excretion. Its capability to positively modulate ductular bile flow helps to preserve the integrity of bile ducts. UDCA also prevents the endocytic internalization of canalicular transporters, a common feature in cholestasis. Finally, UDCA has immunomodulatory properties that limit the exacerbated immunological response occurring in autoimmune cholestatic diseases by counteracting the overexpression of MHC antigens and perhaps by limiting the production of cytokines by immunocompetent cells. Owing to this multi-functionality, it is difficult to envisage a substitute for UDCA that combines as many hepatoprotective effects with such efficacy. We predict a long-lasting use of UDCA as the therapeutic agent of choice in cholestasis.


Assuntos
Colagogos e Coleréticos/farmacologia , Colestase/tratamento farmacológico , Ácido Ursodesoxicólico/farmacologia , Animais , Apoptose/efeitos dos fármacos , Ácidos e Sais Biliares/fisiologia , Canalículos Biliares/efeitos dos fármacos , Colagogos e Coleréticos/uso terapêutico , Colestase/patologia , Colestase/fisiopatologia , Humanos , Fatores Imunológicos/farmacologia , Fatores Imunológicos/uso terapêutico , Mitocôndrias Hepáticas/efeitos dos fármacos , Mitocôndrias Hepáticas/metabolismo , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Ácido Ursodesoxicólico/uso terapêutico
17.
Cells ; 10(10)2021 10 13.
Artigo em Inglês | MEDLINE | ID: mdl-34685713

RESUMO

Fat accumulation (steatosis) in ballooned hepatocytes alters the expression of membrane transporters in Zucker fatty (fa/fa) rats. The aim of the study was to quantify the functions of these transporters and their impact on hepatocyte concentrations using a clinical hepatobiliary contrast agent (Gadobenate dimeglumine, BOPTA) for liver imaging. In isolated and perfused rat livers, we quantified BOPTA accumulation and decay profiles in fa/+ (normal) and fa/fa hepatocytes by placing a gamma counter over livers. Profiles of BOPTA accumulation and decay in hepatocytes were analysed with nonlinear regressions to characterise BOPTA influx and efflux across hepatocyte transporters. At the end of the accumulation period, BOPTA hepatocyte concentrations and influx clearances were not significantly different in fa/+ and fa/fa livers. In contrast, bile clearance was significantly lower in fatty hepatocytes while efflux clearance back to sinusoids compensated the low efflux into canaliculi. The time when BOPTA cellular efflux impacts the accumulation profile of hepatocyte concentrations was slightly delayed (2 min) by steatosis, anticipating a delayed emptying of hepatocytes. The experimental model is useful for quantifying the functions of hepatocyte transporters in liver diseases.


Assuntos
Fígado Gorduroso/metabolismo , Fígado Gorduroso/patologia , Hepatócitos/metabolismo , Hepatócitos/patologia , Proteínas de Membrana Transportadoras/metabolismo , Obesidade/metabolismo , Obesidade/patologia , Animais , Ácidos e Sais Biliares/metabolismo , Canalículos Biliares/efeitos dos fármacos , Canalículos Biliares/metabolismo , Hepatócitos/efeitos dos fármacos , Fígado/efeitos dos fármacos , Fígado/metabolismo , Meglumina/análogos & derivados , Meglumina/farmacocinética , Meglumina/farmacologia , Compostos Organometálicos/farmacocinética , Compostos Organometálicos/farmacologia , Perfusão , Ratos , Ratos Zucker , Reologia/efeitos dos fármacos
18.
J Biol Chem ; 284(31): 20773-80, 2009 Jul 31.
Artigo em Inglês | MEDLINE | ID: mdl-19465483

RESUMO

Signaling via cAMP plays an important role in apical cell surface dynamics in epithelial cells. In hepatocytes, elevated levels of cAMP as well as extracellular oncostatin M stimulate apical lumen development in a manner that depends on protein kinase A (PKA) activity. However, neither the identity of PKA isoforms involved nor the mechanisms of the cross-talk between oncostatin M and cAMP/PKA signaling pathways have been elucidated. Here we demonstrate that oncostatin M and PKA signaling converge at the level of the PKA holoenzyme downstream of oncostatin M-stimulated MAPK activation. Experiments were performed with chemically modified cAMP analogues that preferentially target regulatory subunit (R) I or RII holoenzymes, respectively, in hepatocytes. The data suggest that the dissociation of RI- but not RII-containing holoenzymes, as well as catalytic activity of PKA, is required for apical lumen development in response to elevated levels of cAMP and oncostatin M. However, oncostatin M signaling does not stimulate PKA holoenzyme dissociation in living cells. Based on pharmacological and cell biological studies, it is concluded that RI-controlled PKA activity is essential for cAMP- and oncostatin M-stimulated development of apical bile canalicular lumens.


Assuntos
Canalículos Biliares/citologia , Canalículos Biliares/enzimologia , Polaridade Celular , Subunidade RIalfa da Proteína Quinase Dependente de AMP Cíclico/metabolismo , Hepatócitos/citologia , Hepatócitos/enzimologia , Inibidores de Adenilil Ciclases , Canalículos Biliares/efeitos dos fármacos , Linhagem Celular Tumoral , Núcleo Celular/efeitos dos fármacos , Núcleo Celular/enzimologia , Polaridade Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , AMP Cíclico/farmacologia , Subunidade RIIalfa da Proteína Quinase Dependente de AMP Cíclico/metabolismo , Transferência de Energia/efeitos dos fármacos , Ativação Enzimática/efeitos dos fármacos , Ativadores de Enzimas/farmacologia , Inibidores Enzimáticos/farmacologia , Hepatócitos/efeitos dos fármacos , Holoenzimas/metabolismo , Humanos , Isoenzimas/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Oncostatina M/farmacologia , Fosforilação/efeitos dos fármacos , Proteínas Recombinantes de Fusão/metabolismo
19.
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
20.
Mol Biol Cell ; 18(7): 2745-54, 2007 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-17494870

RESUMO

Oncostatin M and cAMP signaling stimulate apical surface-directed membrane trafficking and apical lumen development in hepatocytes, both in a protein kinase A (PKA)-dependent manner. Here, we show that oncostatin M, but not cAMP, promotes the A-kinase anchoring protein (AKAP)-dependent anchoring of the PKA regulatory subunit (R)IIalpha to subapical centrosomes and that this requires extracellular signal-regulated kinase 2 activation. Stable expression of the RII-displacing peptide AKAP-IS, but not a scrambled peptide, inhibits the association of RIIalpha with centrosomal AKAPs and results in the repositioning of the centrosome from a subapical to a perinuclear location. Concomitantly, common endosomes, but not apical recycling endosomes, are repositioned from a subapical to a perinuclear location, without significant effects on constitutive or oncostatin M-stimulated basolateral-to-apical transcytosis. Importantly, however, the expression of the AKAP-IS peptide completely blocks oncostatin M-, but not cAMP-stimulated apical lumen development. Together, the data suggest that centrosomal anchoring of RIIalpha and the interrelated subapical positioning of these centrosomes is required for oncostatin M-, but not cAMP-mediated, bile canalicular lumen development in a manner that is uncoupled from oncostatin M-stimulated apical lumen-directed membrane trafficking. The results also imply that multiple PKA-mediated signaling pathways control apical lumen development and that subapical centrosome positioning is important in some of these pathways.


Assuntos
Canalículos Biliares/enzimologia , Bucladesina/farmacologia , Polaridade Celular/efeitos dos fármacos , Centrossomo/efeitos dos fármacos , Centrossomo/enzimologia , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Oncostatina M/farmacologia , Proteínas Adaptadoras de Transdução de Sinal/farmacologia , Canalículos Biliares/efeitos dos fármacos , Linhagem Celular Tumoral , Membrana Celular/efeitos dos fármacos , Membrana Celular/enzimologia , Subunidade RIIalfa da Proteína Quinase Dependente de AMP Cíclico , Proteínas Quinases Dependentes de AMP Cíclico/antagonistas & inibidores , Endocitose/efeitos dos fármacos , Endossomos/efeitos dos fármacos , Humanos , Ligação Proteica/efeitos dos fármacos , Frações Subcelulares/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA