Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
2.
Immunol Rev ; 291(1): 57-74, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31402505

RESUMEN

T-lymphocyte activation relies on the cognate recognition by the TCR of the MHC-associated peptide ligand (pMHC) presented at the surface of an antigen-presenting cell (APC). This leads to the dynamic formation of a cognate contact between the T lymphocyte and the APC: the immune synapse (IS). Engagement of the TCR by the pMHC in the synaptic zone induces a cascade of signaling events leading to phosphorylation and dephosphorylation of proteins and lipids, which ultimately shapes the response of T lymphocytes. Although the engagement of the T-cell receptor (TCR) takes place at the plasma membrane, the TCR/CD3 complexes and the signaling molecules involved in transduction of the TCR signal are also present in intracellular membrane pools. These pools, which are both endocytic and exocytic, have tentatively been characterized by several groups including ours. We will herein summarize what is known on the intracellular pools of TCR signaling components. We will discuss their origin and the mechanisms involved in their mobility at the IS. Finally, we will propose several hypotheses concerning the functional role(s) that these intracellular pools might play in T-cell activation. We will also discuss the tools that could be used to test these hypotheses.


Asunto(s)
Activación de Linfocitos , Receptores de Antígenos de Linfocitos T/metabolismo , Transducción de Señal , Linfocitos T/inmunología , Linfocitos T/metabolismo , Animales , Endocitosis , Endosomas/metabolismo , Humanos , Espacio Intracelular/metabolismo , Ligandos , Metabolismo de los Lípidos , Fosforilación , Transporte de Proteínas
3.
PLoS One ; 14(2): e0212215, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30789925

RESUMEN

In obstructive cholestasis, there is an integral adaptive response aimed to diminish the bile flow and minimize the injury of bile ducts caused by increased intraluminal pressure and harmful levels of bile salts and bilirrubin. Canalicular bicarbonate secretion, driven by the anion exchanger 2 (AE2), is an influential determinant of the canalicular bile salt-independent bile flow. In this work, we ascertained whether AE2 expression and/or activity is reduced in hepatocytes from rats with common bile duct ligation (BDL), as part of the adaptive response to cholestasis. After 4 days of BDL, we found that neither AE2 mRNA expression (measured by quantitative real-time PCR) nor total levels of AE2 protein (assessed by western blot) were modified in freshly isolated hepatocytes. However, BDL led to a decrease in the expression of AE2 protein in plasma membrane fraction as compared with SHAM control. Additionally, AE2 activity (JOH-, mmol/L/min), measured in primary cultured hepatocytes from BDL and SHAM rats, was decreased in the BDL group versus the control group (1.9 ± 0.3 vs. 3.1 ± 0.2, p<0.005). cAMP-stimulated AE2 activity, however, was not different between SHAM and BDL groups (3.7 ± 0.3 vs. 3.5 ± 0.3), suggesting that cAMP stimulated insertion into the canalicular membrane of AE2-containing intracellular vesicles, that had remained abnormally internalized after BDL. In conclusion, our results point to the existence of a novel adaptive mechanism in cholestasis aimed to reduce biliary pressure, in which AE2 internalization in hepatocytes might result in decreased canalicular HCO3- output and decreased bile flow.


Asunto(s)
Bicarbonatos/metabolismo , Antiportadores de Cloruro-Bicarbonato/biosíntesis , Cloruros/metabolismo , Colestasis/metabolismo , Regulación hacia Abajo , Hepatocitos/metabolismo , Animales , Colestasis/patología , Modelos Animales de Enfermedad , Hepatocitos/patología , Transporte Iónico , Masculino , Ratas , Ratas Wistar
4.
J Biol Chem ; 293(44): 17081-17094, 2018 11 02.
Artículo en Inglés | MEDLINE | ID: mdl-30228190

RESUMEN

Mitochondrial Ca2+ uptake through the mitochondrial Ca2+ uniporter (MCU) is a tightly controlled process that sustains cell functions mainly by fine-tuning oxidative metabolism to cellular needs. The kinetics of Ca2+ fluxes across the mitochondrial membranes have been studied both in vitro and in vivo for many years, and the discovery of the molecular components of the MCU has further clarified that this Ca2+ uptake mechanism is based on a complex system subject to elaborate layers of controls. Alterations in the speed or capacity of the in-and-out pathways can have detrimental consequences for both the organelle and the cell, impairing cellular metabolism and ultimately causing cell death. Here, we report that pretreatment of deenergized mitochondria with low-micromolar Ca2+ concentrations for a few minutes markedly increases the speed of mitochondrial Ca2+ uptake upon re-addition of an oxidizable substrate. We found that this phenomenon is sensitive to alterations in the level of the MCU modulator proteins mitochondrial calcium uptake 1 (MICU1) and 2 (MICU2), and is accompanied by changes in the association of MICU1-MICU2 complexes with MCU. This increased Ca2+ uptake capacity, occurring under conditions mimicking those during ischemia/reperfusion in vivo, could lead to a massive amount of Ca2+ entering the mitochondrial matrix even at relatively low levels of cytosolic Ca2+ We conclude that the phenomenon uncovered here represents a potential threat of mitochondrial Ca2+ overload to the cell.


Asunto(s)
Calcio/metabolismo , Citosol/metabolismo , Mitocondrias/metabolismo , Animales , Transporte Biológico , Canales de Calcio/genética , Canales de Calcio/metabolismo , Proteínas de Unión al Calcio/genética , Proteínas de Unión al Calcio/metabolismo , Proteínas de Transporte de Catión/genética , Proteínas de Transporte de Catión/metabolismo , Células HEK293 , Células HeLa , Humanos , Hígado/metabolismo , Ratones , Ratones Endogámicos C57BL , Proteínas de Transporte de Membrana Mitocondrial/genética , Proteínas de Transporte de Membrana Mitocondrial/metabolismo
5.
J Exp Med ; 215(4): 1245-1265, 2018 04 02.
Artículo en Inglés | MEDLINE | ID: mdl-29440364

RESUMEN

The adapter molecule linker for activation of T cells (LAT) orchestrates the formation of signalosomes upon T cell receptor (TCR) stimulation. LAT is present in different intracellular pools and is dynamically recruited to the immune synapse upon stimulation. However, the intracellular traffic of LAT and its function in T lymphocyte activation are ill defined. We show herein that LAT, once internalized, transits through the Golgi-trans-Golgi network (TGN), where it is repolarized to the immune synapse. This retrograde transport of LAT depends on the small GTPase Rab6 and the target soluble N-ethylmaleimide-sensitive factor attachment protein receptor (t-SNARE) Syntaxin-16, two regulators of the endosome-to-Golgi/TGN retrograde transport. We also show in vitro in Syntaxin-16- or Rab6-silenced human cells and in vivo in CD4+ T lymphocytes of the Rab6 knockout mouse that this retrograde traffic controls TCR stimulation. These results establish that the retrograde traffic of LAT from the plasma membrane to the Golgi-TGN controls the polarized delivery of LAT at the immune synapse and T lymphocyte activation.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Sinapsis Inmunológicas/metabolismo , Activación de Linfocitos/inmunología , Proteínas de la Membrana/metabolismo , Fosfoproteínas/metabolismo , Linfocitos T/inmunología , Proteínas de Unión al GTP rab/metabolismo , Animales , Membrana Celular/metabolismo , Endosomas/metabolismo , Humanos , Interleucina-2/metabolismo , Células Jurkat , Ratones , Modelos Biológicos , Fosforilación , Transporte de Proteínas , Proteínas R-SNARE/metabolismo , Receptores de Antígenos de Linfocitos T/metabolismo , Transducción de Señal , Sintaxina 16/metabolismo , Red trans-Golgi
6.
EMBO J ; 36(20): 3012-3028, 2017 10 16.
Artículo en Inglés | MEDLINE | ID: mdl-28923825

RESUMEN

Exosomes, nano-sized secreted extracellular vesicles (EVs), are actively studied for their diagnostic and therapeutic potential. In particular, exosomes secreted by dendritic cells (DCs) have been shown to carry MHC-peptide complexes allowing efficient activation of T lymphocytes, thus displaying potential as promoters of adaptive immune responses. DCs also secrete other types of EVs of different size, subcellular origin and protein composition, whose immune capacities have not been yet compared to those of exosomes. Here, we show that large EVs (lEVs) released by human DCs are as efficient as small EVs (sEVs), including exosomes, to induce CD4+ T-cell activation in vitro When released by immature DCs, however, lEVs and sEVs differ in their capacity to orient T helper (Th) cell responses, the former favouring secretion of Th2 cytokines, whereas the latter promote Th1 cytokine secretion (IFN-γ). Upon DC maturation, however, these functional differences are abolished, and all EVs become able to induce IFN-γ. Our results highlight the need to comprehensively compare the functionalities of EV subtypes in all patho/physiological systems where exosomes are claimed to perform critical roles.


Asunto(s)
Linfocitos T CD4-Positivos/efectos de los fármacos , Linfocitos T CD4-Positivos/inmunología , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Vesículas Extracelulares/metabolismo , Activación de Linfocitos , Humanos
7.
Arch Toxicol ; 91(6): 2391-2403, 2017 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-27913845

RESUMEN

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.


Asunto(s)
Canalículos Biliares/efectos de los fármacos , Colestasis/inducido químicamente , Hígado/efectos de los fármacos , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Estrés Oxidativo/efectos de los fármacos , terc-Butilhidroperóxido/toxicidad , Animales , Canalículos Biliares/metabolismo , Canalículos Biliares/fisiopatología , Colestasis/metabolismo , Hígado/metabolismo , Hígado/fisiopatología , Masculino , Proteína Quinasa C/metabolismo , Ratas Wistar
8.
Arch Toxicol ; 90(4): 891-903, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25813982

RESUMEN

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.


Asunto(s)
Transportadoras de Casetes de Unión a ATP/metabolismo , Receptores ErbB/metabolismo , Estradiol/análogos & derivados , Receptor alfa de Estrógeno/metabolismo , Hepatocitos/metabolismo , Animales , Canalículos Biliares/efectos de los fármacos , Canalículos Biliares/metabolismo , Canalículos Biliares/fisiopatología , Células Cultivadas , Colestasis/inducido químicamente , Colestasis/metabolismo , Receptores ErbB/genética , Estradiol/metabolismo , Estradiol/farmacología , Antagonistas del Receptor de Estrógeno/farmacología , Femenino , Fulvestrant , Técnicas de Silenciamiento del Gen , Hepatocitos/efectos de los fármacos , Quinazolinas/farmacología , Ratas , Ratas Wistar , Tirfostinos/farmacología , Familia-src Quinasas/metabolismo
9.
Arch Toxicol ; 89(6): 979-90, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24912783

RESUMEN

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.


Asunto(s)
Canalículos Biliares/metabolismo , Bilis/metabolismo , Transporte Biológico , Colestasis/metabolismo , Hepatocitos/metabolismo , Modelos Biológicos , Animales , Canalículos Biliares/efectos de los fármacos , Bucladesina/farmacología , Técnicas de Cultivo de Célula , Células Cultivadas , Colestasis/inducido químicamente , Estradiol/análogos & derivados , Estradiol/farmacología , Hepatocitos/efectos de los fármacos , Proteína 2 Asociada a Resistencia a Múltiples Medicamentos , Proteínas Asociadas a Resistencia a Múltiples Medicamentos/metabolismo , Ratas , Ácido Taurolitocólico/farmacología
10.
Hepatology ; 59(3): 1016-29, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24115158

RESUMEN

UNLABELLED: Estradiol-17ß-D-glucuronide (E17G) activates different signaling pathways (e.g., Ca(2+) -dependent protein kinase C, phosphoinositide 3-kinase/protein kinase B, mitogen-activated protein kinases [MAPKs] p38 and extracellular signal-related kinase 1/2, and estrogen receptor alpha) that lead to acute cholestasis in rat liver with retrieval of the canalicular transporters, bile salt export pump (Abcb11) and multidrug resistance-associated protein 2 (Abcc2). E17G shares with nonconjugated estradiol the capacity to activate these pathways. G-protein-coupled receptor 30 (GPR30) is a receptor implicated in nongenomic effects of estradiol, and the aim of this study was to analyze the potential role of this receptor and its downstream effectors in E17G-induced cholestasis. In vitro, GPR30 inhibition by G15 or its knockdown with small interfering RNA strongly prevented E17G-induced impairment of canalicular transporter function and localization. E17G increased cyclic adenosine monophosphate (cAMP) levels, and this increase was blocked by G15, linking GPR30 to adenylyl cyclase (AC). Moreover, AC inhibition totally prevented E17G insult. E17G also increased protein kinase A (PKA) activity, which was blocked by G15 and AC inhibitors, connecting the links of the pathway, GPR30-AC-PKA. PKA inhibition prevented E17G-induced cholestasis, whereas exchange protein activated directly by cyclic nucleotide/MAPK kinase, another cAMP downstream effector, was not implicated in cAMP cholestatic action. In the perfused rat liver model, inhibition of the GPR30-AC-PKA pathway totally prevented E17G-induced alteration in Abcb11 and Abcc2 function and localization. CONCLUSION: Activation of GPR30-AC-PKA is a key factor in the alteration of canalicular transporter function and localization induced by E17G. Interaction of E17G with GPR30 may be the first event in the cascade of signaling activation.


Asunto(s)
Adenilil Ciclasas/metabolismo , Colestasis/metabolismo , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Estradiol/análogos & derivados , Receptores Acoplados a Proteínas G/metabolismo , Miembro 11 de la Subfamilia B de Transportador de Casetes de Unión al ATP , Transportadoras de Casetes de Unión a ATP/metabolismo , Animales , Canalículos Biliares/metabolismo , Células Cultivadas , Colestasis/inducido químicamente , Estradiol/toxicidad , Técnicas de Silenciamiento del Gen , Hepatocitos/citología , Hepatocitos/efectos de los fármacos , Hepatocitos/enzimología , Ratas , Receptores Acoplados a Proteínas G/genética , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología
11.
Dig Dis Sci ; 58(6): 1602-14, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23371010

RESUMEN

BACKGROUND: Estradiol-17ß-D-glucuronide (E17G) induces cholestasis in vivo, endocytic internalization of the canalicular transporters multidrug resistance-associated protein 2 (Abcc2) and bile salt export pump (Abcb11) being a key pathomechanism. Cyclic AMP (cAMP) prevents cholestasis by targeting these transporters back to the canalicular membrane. In hepatocyte couplets, glucagon and salbutamol, both of which increase cAMP, prevented E17G action by stimulating the trafficking of these transporters by different mechanisms, namely: glucagon activates a protein kinase A-dependent pathway, whereas salbutamol activates an exchange-protein activated by cAMP (Epac)-mediated, microtubule-dependent pathway. METHODS: The present study evaluated whether glucagon and salbutamol prevent E17G-induced cholestasis in a more physiological model, i.e., the perfused rat liver (PRL). Additionally, the preventive effect of in vivo alanine administration, which induces pancreatic glucagon secretion, was evaluated. RESULTS: In PRLs, glucagon and salbutamol prevented E17G-induced decrease in both bile flow and the secretory activity of Abcc2 and Abcb11. Salbutamol prevention fully depended on microtubule integrity. On the other hand, glucagon prevention was microtubule-independent only at early time periods after E17G administration, but it was ultimately affected by the microtubule disrupter colchicine. Cholestasis was associated with endocytic internalization of Abcb11 and Abcc2, the intracellular carriers being partially colocalized with the endosomal marker Rab11a. This effect was completely prevented by salbutamol, whereas some transporter-containing vesicles remained colocalized with Rab11a after glucagon treatment. In vivo, alanine administration increased hepatic cAMP and accelerated the recovery of bile flow and Abcb11/Abcc2 transport function after E17G administration. The initial recovery afforded by alanine was microtubule-independent, but microtubule integrity was required to sustain this protective effect. CONCLUSION: We conclude that modulation of cAMP levels either by direct administration of cAMP modulators or by physiological manipulations leadings to hormone-mediated increase of cAMP levels (alanine administration), prevents estrogen-induced cholestasis in models with preserved liver architecture, through mechanisms similar to those arisen from in vitro studies.


Asunto(s)
Agonistas de Receptores Adrenérgicos beta 2/uso terapéutico , Albuterol/uso terapéutico , Colestasis/prevención & control , AMP Cíclico/agonistas , Estradiol , Glucagón/uso terapéutico , Hormonas/uso terapéutico , Miembro 11 de la Subfamilia B de Transportador de Casetes de Unión al ATP , Transportadoras de Casetes de Unión a ATP/metabolismo , Alanina/uso terapéutico , Animales , Biomarcadores/metabolismo , Colestasis/etiología , Colestasis/metabolismo , AMP Cíclico/metabolismo , Femenino , Hígado/metabolismo , Hígado/fisiopatología , Ratas , Ratas Wistar , Resultado del Tratamiento , Proteínas de Unión al GTP rab/metabolismo
12.
PLoS One ; 7(11): e50711, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23209816

RESUMEN

UNLABELLED: Estradiol 17ß-D-glucuronide (E17G) induces acute cholestasis in rat with endocytic internalization of the canalicular transporters bile salt export pump (Abcb11) and multidrug resistance-associated protein 2 (Abcc2). Classical protein kinase C (cPKC) and PI3K pathways play complementary roles in E17G cholestasis. Since non-conjugated estradiol is capable of activating these pathways via estrogen receptor alpha (ERα), we assessed the participation of this receptor in the cholestatic manifestations of estradiol glucuronidated-metabolite E17G in perfused rat liver (PRL) and in isolated rat hepatocyte couplets (IRHC). In both models, E17G activated ERα. In PRL, E17G maximally decreased bile flow, and the excretions of dinitrophenyl-glutathione, and taurocholate (Abcc2 and Abcb11 substrates, respectively) by 60% approximately; preadministration of ICI 182,780 (ICI, ERα inhibitor) almost totally prevented these decreases. In IRHC, E17G decreased the canalicular vacuolar accumulation of cholyl-glycylamido-fluorescein (Abcb11 substrate) with an IC50 of 91±1 µM. ICI increased the IC50 to 184±1 µM, and similarly prevented the decrease in the canalicular vacuolar accumulation of the Abcc2 substrate, glutathione-methylfluorescein. ICI also completely prevented E17G-induced delocalization of Abcb11 and Abcc2 from the canalicular membrane, both in PRL and IRHC. The role of ERα in canalicular transporter internalization induced by E17G was confirmed in ERα-knocked-down hepatocytes cultured in collagen sandwich. In IRHC, the protection of ICI was additive to that produced by PI3K inhibitor wortmannin but not with that produced by cPKC inhibitor Gö6976, suggesting that ERα shared the signaling pathway of cPKC but not that of PI3K. Further analysis of ERα and cPKC activations induced by E17G, demonstrated that ICI did not affect cPKC activation whereas Gö6976 prevented that of ERα, indicating that cPKC activation precedes that of ERα. CONCLUSION: ERα is involved in the biliary secretory failure induced by E17G and its activation follows that of cPKC.


Asunto(s)
Colestasis/inducido químicamente , Colestasis/metabolismo , Estradiol/análogos & derivados , Receptor alfa de Estrógeno/metabolismo , Proteína Quinasa C/metabolismo , Animales , Carbazoles/farmacología , Células Cultivadas , Estradiol/farmacología , Receptor alfa de Estrógeno/antagonistas & inhibidores , Femenino , Fulvestrant , Hepatocitos/efectos de los fármacos , Hepatocitos/metabolismo , Hígado/efectos de los fármacos , Hígado/metabolismo , Proteína Quinasa C/antagonistas & inhibidores , Ratas , Ratas Wistar
13.
PLoS One ; 7(11): e49255, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23166621

RESUMEN

OBJECTIVE: The endogenous, cholestatic metabolite estradiol 17ß-D-glucuronide (E(2)17G) induces endocytic internalization of the canalicular transporters relevant to bile formation, Bsep and Mrp2. We evaluated here whether MAPKs are involved in this effect. DESIGN: ERK1/2, JNK1/2, and p38 MAPK activation was assessed by the increase in their phosphorylation status. Hepatocanalicular function was evaluated in isolated rat hepatocyte couplets (IRHCs) by quantifying the apical secretion of fluorescent Bsep and Mrp2 substrates, and in isolated, perfused rat livers (IPRLs), using taurocholate and 2,4-dinitrophenyl-S-glutathione, respectively. Protein kinase participation in E(2)17G-induced secretory failure was assessed by co-administering selective inhibitors. Internalization of Bsep/Mrp2 was assessed by confocal microscopy and image analysis. RESULTS: E(2)17G activated all kinds of MAPKs. The PI3K inhibitor wortmannin prevented ERK1/2 activation, whereas the cPKC inhibitor Gö6976 prevented p38 activation, suggesting that ERK1/2 and p38 are downstream of PI3K and cPKC, respectively. The p38 inhibitor SB203580 and the ERK1/2 inhibitor PD98059, but not the JNK1/2 inhibitor SP600125, partially prevented E(2)17G-induced changes in transporter activity and localization in IRHCs. p38 and ERK1/2 co-inhibition resulted in additive protection, suggesting complementary involvement of these MAPKs. In IPRLs, E(2)17G induced endocytosis of canalicular transporters and a rapid and sustained decrease in bile flow and biliary excretion of Bsep/Mrp2 substrates. p38 inhibition prevented this initial decay, and the internalization of Bsep/Mrp2. Contrarily, ERK1/2 inhibition accelerated the recovery of biliary secretion and the canalicular reinsertion of Bsep/Mrp2. CONCLUSIONS: cPKC/p38 MAPK and PI3K/ERK1/2 signalling pathways participate complementarily in E(2)17G-induced cholestasis, through internalization and sustained intracellular retention of canalicular transporters, respectively.


Asunto(s)
Colestasis/inducido químicamente , Estradiol/análogos & derivados , Sistema de Señalización de MAP Quinasas/fisiología , Fosfatidilinositol 3-Quinasas/metabolismo , Proteína Quinasa C/metabolismo , Transducción de Señal/fisiología , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Miembro 11 de la Subfamilia B de Transportador de Casetes de Unión al ATP , Transportadoras de Casetes de Unión a ATP/metabolismo , Animales , Western Blotting , Estradiol/toxicidad , Femenino , Hepatocitos/metabolismo , Procesamiento de Imagen Asistido por Computador , Microscopía Confocal , Fosforilación , Ratas , Ratas Wistar , Estadísticas no Paramétricas , Ácido Taurocólico
14.
Front Biosci (Landmark Ed) ; 17(4): 1201-18, 2012 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-22201798

RESUMEN

Vectorial transport of osmotically active solutes from blood into bile is essential for bile flow generation. Therefore, the localization status of hepatocellular transporters involved in this function is critical. These transporters are localized either in the plasma membrane or in an endosomal, submembranous compartment, from where they undergo recycling to the plasma membrane. The balance between exocytic targeting/endocytic internalization from/to this recycling compartment is therefore a chief determinant of the liver capability to secrete bile. Furthermore, its impairment may lead to sustained endocytic internalization, eventually resulting in transporter degradation. Exacerbated internalization of hepatocellular transporters occurs in several experimental models of cholestasis, and also in most human cholestatic liver diseases. This review outlines the possible mechanisms explaining this alteration (e.g., alteration of the organization of actin or actin-transporter linking proteins), and the mediators involved (e.g., activation of "cholestatic" signaling pathways). Finally, several experimental therapeutic approaches based upon the administration of compounds that stimulate exocytic targeting of canalicular transporters (e.g., cAMP, tauroursodeoxycholate) are described with regard to their capability to prevent cholestatic alterations resulting from transporter internalization.


Asunto(s)
Proteínas Portadoras/fisiología , Colestasis , Bilis/fisiología , Humanos
15.
Mol Biol Cell ; 22(20): 3902-15, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21865596

RESUMEN

In estradiol 17ß-d-glucuronide (E17G)-induced cholestasis, the canalicular hepatocellular transporters bile salt export pump (Abcb11) and multidrug-resistance associated protein 2 (Abcc2) undergo endocytic internalization. cAMP stimulates the trafficking of transporter-containing vesicles to the apical membrane and is able to prevent internalization of these transporters in estrogen-induced cholestasis. Hepatocyte levels of cAMP are regulated by hormones such as glucagon and adrenaline (via the ß2 receptor). We analyzed the effects of glucagon and salbutamol (a ß2 adrenergic agonist) on function and localization of Abcb11 and Abcc2 in isolated rat hepatocyte couplets exposed to E17G and compared the mechanistic bases of their effects. Glucagon and salbutamol partially prevented the impairment in Abcb11 and Abcc2 transport capacity. E17G also induced endocytic internalization of Abcb11 and Abcc2, which partially colocalized with the endosomal marker Rab11a. This effect was completely prevented by salbutamol, whereas some transporter-containing vesicles remained internalized and mainly colocalizing with Rab11a in the perinuclear region after incubation with glucagon. Glucagon prevention was dependent on cAMP-dependent protein kinase (PKA) and independent of exchange proteins activated directly by cAMP (Epac) and microtubules. In contrast, salbutamol prevention was PKA independent and Epac/MEK and microtubule dependent. Anticholestatic effects of glucagon and salbutamol were additive in nature. Our results show that increases in cAMP could activate different anticholestatic signaling pathways, depending on the hormonal mediator involved.


Asunto(s)
Transportadoras de Casetes de Unión a ATP/metabolismo , Albuterol/farmacología , Canalículos Biliares/efectos de los fármacos , AMP Cíclico/metabolismo , Estradiol/análogos & derivados , Glucagón/farmacología , Hepatocitos/efectos de los fármacos , Transducción de Señal , Miembro 11 de la Subfamilia B de Transportador de Casetes de Unión al ATP , Agonistas de Receptores Adrenérgicos beta 2/farmacología , Animales , Canalículos Biliares/metabolismo , Colestasis/tratamiento farmacológico , Colestasis/metabolismo , Colestasis/fisiopatología , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Endocitosis/efectos de los fármacos , Epinefrina/farmacología , Estradiol/efectos adversos , Estradiol/farmacología , Femenino , Hepatocitos/metabolismo , Transporte de Proteínas/efectos de los fármacos , Ratas , Ratas Wistar , Transducción de Señal/efectos de los fármacos , Vesículas Transportadoras/efectos de los fármacos , Vesículas Transportadoras/metabolismo , Proteínas de Unión al GTP rab/metabolismo
16.
Hepatology ; 52(4): 1465-76, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20815017

RESUMEN

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.


Asunto(s)
1-Fosfatidilinositol 4-Quinasa/fisiología , Colestasis/inducido químicamente , Proteína Quinasa C/fisiología , Proteínas Proto-Oncogénicas c-akt/fisiología , Miembro 11 de la Subfamilia B de Transportador de Casetes de Unión al ATP , Transportadoras de Casetes de Unión a ATP/metabolismo , Androstadienos/farmacología , Animales , Canalículos Biliares/efectos de los fármacos , Canalículos Biliares/fisiología , Sistema Biliar/metabolismo , Carbazoles/farmacología , Colchicina/farmacología , Endocitosis/efectos de los fármacos , Estradiol/análogos & derivados , Glutatión/metabolismo , Técnicas In Vitro , Masculino , Microtúbulos/efectos de los fármacos , Microtúbulos/fisiología , Proteína 2 Asociada a Resistencia a Múltiples Medicamentos , Proteínas Asociadas a Resistencia a Múltiples Medicamentos/metabolismo , Perfusión , Proteína Quinasa C/antagonistas & inhibidores , Ratas , Ratas Wistar , Transducción de Señal , Ácido Taurocólico/metabolismo , Wortmanina
17.
Hepatology ; 48(6): 1885-95, 2008 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-18972403

RESUMEN

UNLABELLED: The endogenous estradiol metabolite estradiol 17beta-D-glucuronide (E(2)17G) induces an acute cholestasis in rat liver coincident with retrieval of the canalicular transporters bile salt export pump (Bsep, Abcc11) and multidrug resistance-associated protein 2 (Mrp2, Abcc2) and their associated loss of function. We assessed the participation of Ca(2+)-dependent protein kinase C isoforms (cPKC) in the cholestatic manifestations of E(2)17G in perfused rat liver (PRL) and in isolated rat hepatocyte couplets (IRHCs). In PRL, E(2)17G (2 mumol/liver; intraportal, single injection) maximally decreased bile flow, total glutathione, and [(3)H] taurocholate excretion by 61%, 62%, and 79%, respectively; incorporation of the specific cPKC inhibitor Gö6976 (500 nM) in the perfusate almost totally prevented these decreases. In dose-response studies using IRHC, E(2)17G (3.75-800 muM) decreased the canalicular vacuolar accumulation of the Bsep substrate cholyl-lysylfluorescein with an IC50 of 54.9 +/- 7.9 muM. Gö6976 (1 muM) increased the IC50 to 178.4 +/- 23.1 muM, and similarly prevented the decrease in the canalicular vacuolar accumulation of the Mrp2 substrate, glutathione methylfluorescein. Prevention of these changes by Gö6976 coincided with complete protection against E(2)17G-induced retrieval of Bsep and Mrp2 from the canalicular membrane, as detected both in the PRL and IRHC. E(2)17G also increased paracellular permeability in IRHC, which was only partially prevented by Gö6976. The cPKC isoform PKCalpha, but not the Ca(2+)-independent PKC isoform, PKCepsilon, translocated to the plasma membrane after E(2)17G administration in primary cultured rat hepatocytes; Gö6976 completely prevented this translocation, thus indicating specific activation of cPKC. This is consistent with increased autophosphorylation of cPKC by E(2)17G, as detected via western blotting. CONCLUSION: Our findings support a central role for cPKC isoforms in E(2)17G-induced cholestasis, by inducing both transporter retrieval from the canalicular membrane and opening of the paracellular route.


Asunto(s)
Calcio/metabolismo , Colestasis/inducido químicamente , Colestasis/metabolismo , Estradiol/análogos & derivados , Proteína Quinasa C-alfa/metabolismo , Proteína Quinasa C-epsilon/metabolismo , Miembro 11 de la Subfamilia B de Transportador de Casetes de Unión al ATP , Transportadoras de Casetes de Unión a ATP/metabolismo , Animales , Carbazoles/farmacología , Células Cultivadas , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Inhibidores Enzimáticos/farmacología , Estradiol/efectos adversos , Estradiol/farmacología , Femenino , Hepatocitos/efectos de los fármacos , Hepatocitos/metabolismo , Hígado/efectos de los fármacos , Hígado/metabolismo , Proteína Quinasa C-alfa/antagonistas & inhibidores , Proteína Quinasa C-epsilon/antagonistas & inhibidores , Ratas , Ratas Sprague-Dawley
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA