Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Mais filtros












Base de dados
Intervalo de ano de publicação
1.
Biochimie ; 223: 41-53, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-38608750

RESUMO

The endogenous metabolite of estradiol, estradiol 17ß-D-glucuronide (E17G), is considered the main responsible of the intrahepatic cholestasis of pregnancy. E17G alters the activity of canalicular transporters through a signaling pathway-dependent cellular internalization, phenomenon that was attributed to oxidative stress in different cholestatic conditions. However, there are no reports involving oxidative stress in E17G-induced cholestasis, representing this the aim of our work. Using polarized hepatocyte cultures, we showed that antioxidant compounds prevented E17G-induced Mrp2 activity alteration, being this alteration equally prevented by the NADPH oxidase (NOX) inhibitor apocynin. The model antioxidant N-acetyl-cysteine prevented, in isolated and perfused rat livers, E17G-induced impairment of bile flow and Mrp2 activity, thus confirming the participation of reactive oxygen species (ROS) in this cholestasis. In primary cultured hepatocytes, pretreatment with specific inhibitors of ERK1/2 and p38MAPK impeded E17G-induced ROS production; contrarily, NOX inhibition did not affect ERK1/2 and p38MAPK phosphorylation. Both, knockdown of p47phox by siRNA and preincubation with apocynin in sandwich-cultured rat hepatocytes significantly prevented E17G-induced internalization of Mrp2, suggesting a crucial role for NOX in this phenomenon. Concluding, E17G-induced cholestasis is partially mediated by NOX-generated ROS through internalization of canalicular transporters like Mrp2, being ERK1/2 and p38MAPK necessary for NOX activation.


Assuntos
Estradiol , Hepatócitos , NADPH Oxidases , Espécies Reativas de Oxigênio , Animais , NADPH Oxidases/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Ratos , Hepatócitos/metabolismo , Hepatócitos/efeitos dos fármacos , Estradiol/farmacologia , Estradiol/metabolismo , Estradiol/análogos & derivados , Feminino , Colestase/induzido quimicamente , Colestase/metabolismo , Colestase/patologia , Ratos Wistar , Acetofenonas/farmacologia , Estresse Oxidativo/efeitos dos fármacos , Acetilcisteína/farmacologia , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Proteínas Associadas à Resistência a Múltiplos Medicamentos/metabolismo , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Células Cultivadas , Antioxidantes/farmacologia , Antioxidantes/metabolismo , Colestase Intra-Hepática , Complicações na Gravidez , Transportadores de Cassetes de Ligação de ATP
2.
Antioxid Redox Signal ; 35(10): 808-831, 2021 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-34293961

RESUMO

Significance: Most hepatopathies are primarily or secondarily cholestatic in nature. Oxidative stress (OS) is a frequent trait among them, and impairs the machinery to generate bile by triggering endocytic internalization of hepatocellular transporters, thus causing cholestasis. This is critical, since it leads to accelerated transporter degradation, which could explain the common post-transcriptional downregulation of transporter expression in human cholestatic diseases. Recent Advances: The mechanisms involved in OS-induced hepatocellular transporter internalization are being revealed. Filamentous actin (F-actin) cytoskeleton disorganization and/or detachment of crosslinking actin proteins that afford transporter stability have been characterized as causal factors. Activation of redox-sensitive signaling pathways leading to changes in phosphorylation status of these structures is involved, including Ca2+-mediated activation of "classical" and "novel" protein kinase C (PKC) isoforms or redox-signaling cascades downstream of NADPH oxidase. Critical Issues: Despite the well-known occurrence of hepatocellular transporter internalization in human hepatopathies, the cholestatic implications of this phenomenon have been overlooked. Accordingly, no specific treatment has been established in the clinical practice for its prevention/reversion. Future Directions: We need to improve our knowledge on the pro-oxidant triggering factors and the multiple signaling pathways that mediate this oxidative injury in each cholestatic hepatopathy, so as to envisage tailor-made therapeutic strategies for each case. Meanwhile, administration of antioxidants or heme oxygenase-1 induction to elevate the hepatocellular levels of the endogenous scavenger bilirubin are promising alternatives that need to be re-evaluated and implemented. They may complement current treatments in cholestasis aimed to enhance transcriptional carrier expression, by providing membrane stability to the newly synthesized carriers. Antioxid. Redox Signal. 35, 808-831.


Assuntos
Bile/metabolismo , Colestase/metabolismo , Hepatócitos/metabolismo , Fatores de Transcrição/metabolismo , Animais , Humanos , Estresse Oxidativo , Transdução de Sinais
3.
Life Sci ; 259: 118352, 2020 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-32860804

RESUMO

AIMS: Lipopolysaccharide (LPS) induces inflammatory cholestasis by impairing expression, localization, and function of carriers involved in bile formation, e.g. bile salt export pump (Bsep) and multidrug resistance-associated protein 2 (Mrp2). A specific therapy against this disease is still lacking. Therefore, we evaluated the anticholestatic effects of spironolactone (SL), a PXR ligand that regulates bile salt homeostasis, up-regulates Mrp2, and bears anti-inflammatory properties. MAIN METHODS: Male Wistar rats were divided into four groups: Control, SL (83.3 mg/kg/day of SL, i.p., for 3 days), LPS (2.5 mg/kg/day, i.p., at 8 am of the last 2 days, and 1.5 mg/kg/day at 8 pm of the last day), and SL + LPS. Biliary and plasma parameters and the expression, function, and localization of Mrp2 and Bsep were evaluated. KEY FINDINGS: SL partially prevented LPS-induced drop of basal bile flow by normalizing the bile salt-independent fraction of bile flow (BSIBF), via improvement of glutathione output. This was due to a recovery in Mrp2 transport function, the major canalicular glutathione transporter, estimated by monitoring the output of its exogenously administered substrate dibromosulfophthalein. SL counteracted the LPS-induced downregulation of Mrp2, but not that of Bsep, at both mRNA and protein levels. LPS induced endocytic internalization of both transporters, visualized by immunofluorescence followed by confocal microscopy, and SL partially prevented this relocalization. SL did not prevent the increase in IL-1ß, IL-6, and TNF-α plasma levels. SIGNIFICANCE: SL prevents the impairment in Mrp2 expression and localization, and the resulting recovery of Mrp2 function normalizes the BSIBF by improving glutathione excretion.


Assuntos
Transportadores de Cassetes de Ligação de ATP/metabolismo , Colestase/tratamento farmacológico , Espironolactona/uso terapêutico , Membro 11 da Subfamília B de Transportadores de Cassetes de Ligação de ATP/metabolismo , Animais , Bile/metabolismo , Colestase/sangue , Colestase/metabolismo , Citocinas/sangue , Regulação da Expressão Gênica/efeitos dos fármacos , Lipopolissacarídeos/efeitos adversos , Masculino , Ratos , Ratos Wistar , Reação em Cadeia da Polimerase em Tempo Real
4.
Biochem Pharmacol ; 168: 48-56, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-31202734

RESUMO

Lipopolysaccharide (LPS) from Gram (-) bacteria induces inflammatory cholestasis by impairing the expression/localization of transporters involved in bile formation (e.g., Bsep, Mrp2). Therapeutic options for this disease are lacking. Ursodeoxycholic acid (UDCA) is the first choice therapy in cholestasis, but its anticholestatic efficacy in this hepatopathy remains to be evaluated. To asses it, male Wistar rats received UDCA for 5 days (25 mg/Kg/day, i.p.) with or without LPS, administered at 8 a.m. of the last 2 days (4 mg/Kg/day, i.p.), plus half of this dose at 8 p.m. of the last day. Then, plasma alkaline phosphatase (ALP), bile flow, basal and taurocholate-stimulated bile acid output, total glutathione output, and total/plasma membrane liver protein expression of Bsep and Mrp2 by confocal microscopy were assessed. mRNA levels of both transporters were assessed by Real-Time PCR. Plasma pro-inflammatory cytokines (IL-6 and TNF-α) were measured by ELISA. Our results showed that UDCA attenuated LPS-induced ALP plasma release and the impairment in the excretion of the Bsep substrate, taurocholate. This was associated with an improved Bsep expression at both mRNA and protein levels, and by an improved localization of Bsep in plasma membrane. UDCA failed to reduce the increase in plasma pro-inflammatory cytokines induced by LPS and Mrp2 expression/function. In conclusion, UDCA protects the hepatocyte against the damaging effect of bile acids accumulated by the LPS-induced secretory failure. This involved an enhanced synthesis of Bsep and an improved membrane stability of the newly synthesized transporters.


Assuntos
Colagogos e Coleréticos/uso terapêutico , Colestase/induzido quimicamente , Colestase/tratamento farmacológico , Lipopolissacarídeos/farmacologia , Ácido Ursodesoxicólico/uso terapêutico , Membro 11 da Subfamília B de Transportadores de Cassetes de Ligação de ATP/metabolismo , Transportadores de Cassetes de Ligação de ATP/metabolismo , Fosfatase Alcalina/sangue , Animais , Ácidos e Sais Biliares/metabolismo , Colagogos e Coleréticos/administração & dosagem , Colagogos e Coleréticos/farmacologia , Modelos Animais de Doenças , Hepatócitos/efeitos dos fármacos , Hepatócitos/metabolismo , Fígado/metabolismo , Masculino , Ratos , Ratos Wistar , Resultado do Tratamento , Ácido Ursodesoxicólico/administração & dosagem , Ácido Ursodesoxicólico/farmacologia
5.
Clin Sci (Lond) ; 133(1): 117-134, 2019 01 15.
Artigo em Inglês | MEDLINE | ID: mdl-30538149

RESUMO

We previously demonstrated in in vitro and ex vivo models that physiological concentrations of unconjugated bilirubin (BR) prevent oxidative stress (OS)-induced hepatocanalicular dysfunction and cholestasis. Here, we aimed to ascertain, in the whole rat, whether a similar cholestatic OS injury can be counteracted by heme oxygenase-1 (HO-1) induction that consequently elevates endogenous BR levels. This was achieved through the administration of hemin, an inducer of HO-1, the rate-limiting step in BR generation. We found that BR peaked between 6 and 8 h after hemin administration. During this time period, HO-1 induction fully prevented the pro-oxidant tert-butylhydroperoxide (tBuOOH)-induced drop in bile flow, and in the biliary excretion of bile salts and glutathione, the two main driving forces of bile flow; this was associated with preservation of the membrane localization of their respective canalicular transporters, bile salt export pump (Bsep) and multidrug resistance-associated protein 2 (Mrp2), which are otherwise endocytosed by OS. HO-1 induction counteracted the oxidation of intracellular proteins and membrane lipids induced by tBuOOH, and fully prevented the increase in the oxidized-to-total glutathione (GSHt) ratio, a sensitive parameter of hepatocellular OS. Compensatory elevations of the activity of the antioxidant enzymes catalase (CAT) and superoxide dismutase (SOD) were also prevented. We conclude that in vivo HO-1 induction protects the liver from acute oxidative injury, thus preventing consequent cholestasis. This reveals an important role for the induction of HO-1 and the consequently elevated levels of BR in preserving biliary secretory function under OS conditions, thus representing a novel therapeutic tool to limit the cholestatic injury that bears an oxidative background.


Assuntos
Antioxidantes/farmacologia , Colestase/prevenção & controle , Heme Oxigenase (Desciclizante)/biossíntese , Hemina/farmacologia , Fígado/efeitos dos fármacos , Estresse Oxidativo , Animais , Bile/metabolismo , Bilirrubina/metabolismo , Catalase/metabolismo , Colestase/induzido quimicamente , Colestase/enzimologia , Colestase/patologia , Modelos Animais de Doenças , Indução Enzimática , Glutationa/metabolismo , Fígado/enzimologia , Fígado/patologia , Masculino , Ratos Wistar , Superóxido Dismutase/metabolismo , terc-Butil Hidroperóxido
6.
Ann Clin Biochem ; 54(5): 577-583, 2017 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-27638930

RESUMO

Background Antiphospholipid syndrome is an autoimmune disease characterized by thrombosis, fetal losses and thrombocytopenia associated to antiphospholipid antibodies. They are directed to phospholipids, such as cardiolipins (anticardiolipin) and lupus anticoagulant or to complexes formed by phospholipids and protein cofactors, such as ß2 glycoprotein 1 (a-ß2GP1) and annexin V (a-annexin V). These auto-antibodies may be considered as a family of antibodies involved in thrombotic events and antiphospholipid activity. On the other hand, some proangiogenic factors are involved in the normal development of placental vasculature, such as the vascular endothelial growth factor. Overexpression of vascular endothelial growth factor receptor in its soluble form (sVEGFR-1) has been associated to a higher antiangiogenic activity. Our aim was to analyse the association between anticardiolipin, lupus anticoagulant, a-ß2GP1, a-annexin V and sVEGFR-1 with recurrent miscarriage before week 10 of gestation in females with antiphospholipid syndrome. Methods We studied 24 females (primary or secondary antiphospholipid syndrome), who were divided into two groups: females with recurrent miscarriage before week 10 of gestation (M; n = 12) and females with no history of fetal loss (NM; n = 12). Anticardiolipin, a-ß2GP1, a-annexin V and sVEGF-R1 concentrations were assessed by ELISA, while lupus anticoagulant was assessed by screening and confirmatory tests. Results A significant association was observed between the number of positive biomarkers and the belonging group ( P < 0.05). Besides, a positive result for lupus anticoagulant and a-ß2GP1 was found to be significantly associated to the M group ( P < 0.05). Conclusions Lupus anticoagulant and a-ß2GP1 may be implicated in pregnancies complicated by recurrent miscarriage in females with antiphospholipid syndrome.


Assuntos
Aborto Habitual/sangue , Aborto Habitual/fisiopatologia , Anticorpos Antifosfolipídeos/sangue , Síndrome Antifosfolipídica/complicações , Neovascularização Fisiológica , Aborto Habitual/imunologia , Anexina A5/imunologia , Anticorpos Anticardiolipina/sangue , Feminino , Humanos , Inibidor de Coagulação do Lúpus/sangue , Gravidez , Estudos Retrospectivos , Receptor 1 de Fatores de Crescimento do Endotélio Vascular/sangue , beta 2-Glicoproteína I/imunologia
7.
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
8.
Arch Toxicol ; 88(9): 1695-709, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-24614978

RESUMO

Oxidative stress (OS) is a common event in most hepatopathies, leading to mitochondrial permeability transition pore (MPTP) formation and further exacerbation of both OS from mitochondrial origin and cell death. Intracellular Ca²âº increase plays a permissive role in these events, but the underlying mechanisms are poorly known. We examined in primary cultured rat hepatocytes whether the Ca²âº/calmodulin (CaM)-dependent protein kinase II (CaMKII) signaling pathway is involved in this process, by using tert-butyl hydroperoxide (tBOOH) as a pro-oxidant, model compound. tBOOH (500 µM, 15 min) induced MPTP formation, as assessed by measuring mitochondrial membrane depolarization as a surrogate marker, and increased lipid peroxidation in a cyclosporin A (CsA)-sensitive manner, revealing the involvement of MPTPs in tBOOH-induced radical oxygen species (ROS) formation. Intracellular Ca²âº sequestration with BAPTA/AM, CaM blockage with W7 or trifluoperazine, and CaMKII inhibition with KN-62 all fully prevented tBOOH-induced MPTP opening and reduced tBOOH-induced lipid peroxidation to a similar extent to CsA, suggesting that Ca²âº/CaM/CaMKII signaling pathway fully mediates MPTP-mediated mitochondrial ROS generation. tBOOH-induced apoptosis, as shown by flow cytometry of annexin V/propidium iodide, mitochondrial release of cytochrome c, activation of caspase-3 and increase in the Bax-to-Bcl-xL ratio, and the Ca²âº/CaM/CaMKII signaling antagonists fully prevented these effects. Intramitochondrial CaM and CaMKII were partially involved in tBOOH-induced MPTP formation, since W7 and KN-62 both attenuated the tBOOH-induced, MPTP-mediated swelling of isolated mitochondria. We concluded that Ca²âº/CaM/CaMKII signaling pathway is a key mediator of OS-induced MPTP formation and the subsequent exacerbation of OS from mitochondrial origin and apoptotic cell death.


Assuntos
Sinalização do Cálcio , Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina/metabolismo , Calmodulina/metabolismo , Hepatócitos/metabolismo , Mitocôndrias Hepáticas/metabolismo , Proteínas de Transporte da Membrana Mitocondrial/metabolismo , Estresse Oxidativo , Animais , Apoptose/efeitos dos fármacos , Sinalização do Cálcio/efeitos dos fármacos , Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina/antagonistas & inibidores , Calmodulina/antagonistas & inibidores , Células Cultivadas , Hepatócitos/citologia , Hepatócitos/efeitos dos fármacos , Peroxidação de Lipídeos/efeitos dos fármacos , Masculino , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Mitocôndrias Hepáticas/efeitos dos fármacos , Mitocôndrias Hepáticas/enzimologia , Proteínas de Transporte da Membrana Mitocondrial/agonistas , Proteínas de Transporte da Membrana Mitocondrial/antagonistas & inibidores , Poro de Transição de Permeabilidade Mitocondrial , Dilatação Mitocondrial/efeitos dos fármacos , Oxidantes/antagonistas & inibidores , Oxidantes/toxicidade , Estresse Oxidativo/efeitos dos fármacos , Inibidores de Proteínas Quinases/farmacologia , Ratos Wistar , Espécies Reativas de Oxigênio/metabolismo , terc-Butil Hidroperóxido/antagonistas & inibidores , terc-Butil Hidroperóxido/toxicidade
9.
Arch Toxicol ; 88(2): 501-14, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24306262

RESUMO

Bilirubin is an endogenous antioxidant with cytoprotective properties, and several studies highlight its potential in the treatment of pro-oxidant diseases. We demonstrated that oxidative stress (OS), a key feature in most hepatopathies, induces cholestasis by actin cytoskeleton disarrangement and further endocytic internalization of key canalicular transporters, such as the bile salt export pump (Bsep) and the multidrug resistance-associated protein 2 (Mrp2) . Here, we evaluated the capability of physiological concentrations of unconjugated bilirubin (UB) to limit OS and the impairment in biliary secretory function induced by the model pro-oxidant agent, tert-butylhydroperoxide (tBuOOH). UB fully prevented the formation of reactive oxygen species and membrane lipid peroxidation induced by tBuOOH in isolated rat hepatocytes. In the isolated rat hepatocyte couplet model, UB (17.1 µM) prevented the endocytic internalization of Bsep and Mrp2 and the impairment in their secretory function induced by tBuOOH. UB also prevented actin disarrangement, as evaluated by both plasma membrane bleb formation and actin fluorescent staining. Finally, UB prevented tBuOOH-induced cPKC activation. Experiments in isolated perfused rat livers showed that UB prevents the increase in oxidized glutathione biliary excretion and the drop in bile flow and the biliary excretion of specific Bsep and Mrp2 substrates. We conclude that physiological concentrations of UB are sufficient to prevent the biliary secretory failure induced by OS, by counteracting actin disarrangement and the consequent internalization of canalicular transporters relevant to normal bile formation. This reveals an important role for UB in preserving biliary secretory function under OS conditions.


Assuntos
Bilirrubina/farmacologia , Colestase/prevenção & controle , Fígado/efeitos dos fármacos , Fígado/fisiopatologia , Estresse Oxidativo/efeitos dos fármacos , Membro 11 da Subfamília B de Transportadores de Cassetes de Ligação de ATP , Transportadores de Cassetes de Ligação de ATP/metabolismo , Actinas/metabolismo , Animais , Ácidos e Sais Biliares/metabolismo , Bilirrubina/metabolismo , Colestase/metabolismo , Glutationa/metabolismo , Hepatócitos/efeitos dos fármacos , Hepatócitos/metabolismo , Fígado/metabolismo , Masculino , Técnicas de Cultura de Órgãos , Proteína Quinase C-alfa/metabolismo , Ratos , Ratos Wistar , terc-Butil Hidroperóxido/farmacologia
10.
Hepatology ; 59(3): 1016-29, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24115158

RESUMO

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.


Assuntos
Adenilil Ciclases/metabolismo , Colestase/metabolismo , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Estradiol/análogos & derivados , Receptores Acoplados a Proteínas G/metabolismo , Membro 11 da Subfamília B de Transportadores de Cassetes de Ligação de ATP , Transportadores de Cassetes de Ligação de ATP/metabolismo , Animais , Canalículos Biliares/metabolismo , Células Cultivadas , Colestase/induzido quimicamente , Estradiol/toxicidade , Técnicas de Silenciamento de Genes , Hepatócitos/citologia , Hepatócitos/efeitos dos fármacos , Hepatócitos/enzimologia , Ratos , Receptores Acoplados a Proteínas G/genética , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia
11.
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
12.
Chem Biol Interact ; 188(3): 386-92, 2010 Dec 05.
Artigo em Inglês | MEDLINE | ID: mdl-20797393

RESUMO

Ursodeoxycholic acid is widely used as a therapeutic agent for the treatment of cholestatic liver diseases. In these hepatopathies, the bile secretory failure produces accumulation of endogenous, tensioactive bile salts, leading to plasma membrane damage and, eventually, hepatocellular lysis. In the present study, we analyzed the capacity of the ursodeoxycholic acid endogenous metabolite, tauroursodeoxycholate (TUDC), to stabilize the hepatocellular plasma membrane against its transition to the micellar phase induced by the tensioactive bile salt taurochenodeoxycholate (TCDC), the main endogenous bile salt accumulated in cholestasis. The disruption of the plasma membrane was evaluated (i) in isolated hepatocytes, through the release of the enzyme lactate dehydrogenase to the incubation medium and (ii) in isolated plasma membranes, through the self-quenching assay of the membranotropic probe octadecylrhodamine B; this assay allows for detergent-induced transition from membrane bilayer to micelle to be monitored. Our results showed that isolated hepatocytes treated with TUDC are more resistant to TCDC-induced cell lysis. When this effect was evaluated in isolated plasma membranes, the TCDC concentration necessary to reach half of the transition from bilayer to micelle was increased by 22% (p<0.05). This difference remained even when TUDC was removed from the incubation medium before adding TCDC, thus indicating that TUDC exerted its effect directly on the plasma membrane. When the same experiments were carried out using the non-ionic detergent TX-100 or the cholesterol-complexing detergent digitonin, no protective effect was observed. In conclusion, TUDC prevents selectively the bilayer to micelle transition of the hepatocellular plasma membrane induced by hydrophobic bile salts that typically build up and accumulate in cholestatic processes. Our results suggest that formation of a complex between negatively charged TUDC and cholesterol in the membrane favours repulsion of negatively charged detergent bile salts, thus providing a basis for the understanding of the TUDC protective effects.


Assuntos
Membrana Celular/efeitos dos fármacos , Hepatócitos/citologia , Hepatócitos/efeitos dos fármacos , Micelas , Ácido Tauroquenodesoxicólico/antagonistas & inibidores , Ácido Tauroquenodesoxicólico/farmacologia , Animais , Morte Celular/efeitos dos fármacos , Membrana Celular/química , Detergentes/farmacologia , Digitonina/farmacologia , Bicamadas Lipídicas/química , Masculino , Osmose/efeitos dos fármacos , Transição de Fase/efeitos dos fármacos , Polietilenoglicóis/farmacologia , Ratos , Solubilidade/efeitos dos fármacos
13.
Clin Sci (Lond) ; 118(2): 99-113, 2009 Oct 12.
Artigo em Inglês | MEDLINE | ID: mdl-19807696

RESUMO

Complement, an important effector mechanism of the immune system, is an enzymatic cascade of approx. 30 serum proteins leading to the amplification of a specific humoral response. It can be activated through the classical or alternative pathways, or through the mannose-binding lectin pathway. The activation of the classical pathway is initiated by the binding of the C1 component to antigen-bound antibodies, known as immunocomplexes. C1 is a complex of one molecule of C1q, two molecules of C1r and two molecules of C1s. C1q contains three copies of a Y-shaped fundamental unit with globular heads included in its structure, which play a major role in the interaction with the Fc portion of immunoglobulins. Deficient or exacerbated activation of the complement system leads to diseases of variable severity, and pharmacological inhibition of the complement system is considered as a therapeutic strategy to ameliorate the inflammatory effects of exacerbated complement activation. Bilirubin is a product of haem degradation by the concerted action of haem oxygenase, which converts haem into biliverdin, and biliverdin reductase, which reduces biliverdin to UCB (unconjugated bilirubin). UCB exerts both cytoprotective and cytotoxic effects in a variety of tissues and cells, acting either as an antioxidant at low concentrations or as an oxidant at high concentrations. In the present review, we describe in detail the anti-complement properties of bilirubin, occurring at levels above the UCB concentrations found in normal human serum, as a beneficial effect of potential clinical relevance. We provide evidence that UCB interferes with the interaction between C1q and immunoglobulins, thus inhibiting the initial step in the activation of complement through the classical pathway. A molecular model is proposed for the interaction between UCB and C1q.


Assuntos
Via Clássica do Complemento/imunologia , Hiperbilirrubinemia/imunologia , Inflamação/prevenção & controle , Antioxidantes/farmacologia , Bilirrubina/farmacologia , Bilirrubina/fisiologia , Complemento C1q/metabolismo , Inativadores do Complemento/farmacologia , Via Clássica do Complemento/efeitos dos fármacos , Citoproteção/fisiologia , Humanos , Inflamação/imunologia , Estresse Oxidativo/imunologia
14.
Chem Biol Interact ; 179(2-3): 297-303, 2009 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-19135039

RESUMO

Silymarin (SIL) is a natural extract with hepatoprotective properties composed mainly of flavonolignans, with silibinin (SB) being its principal constituent. SB is thought to be the main responsible for SIL hepatoprotective properties, although this has not been corroborated systematically. We analysed comparatively the effects of SIL and SB on hepatocellular plasma membrane stability. SIL (500 microM concentration in SB) protected significantly the plasma membrane disruption induced by Triton X-100 (TX-100) and taurochenodeoxycholate (TCDC), both in isolated plasma membrane (assessed by recording the plasma membrane transition from bilayer to micelle using the R18 self-quenching assay) and in isolated rat hepatocytes (assessed by the release into the incubation medium of the cytosolic enzymes lactate dehydrogenase and alanine aminotransferase). Contrarily, SB (500 microM) exacerbated plasma membrane disruption induced by TX-100 in both systems at detergent concentrations relevant to induce hepatocellular lysis, although it displayed some stabilizing properties at higher concentrations. SB showed a lower stabilizing effect against TCDC-induced plasma membrane disruption when assayed in both models. In addition, SB exposure made the plasma membrane more labile to disruption induced by osmotic lysis. These results show that SIL and SB have differential effects on membrane stability; whereas SIL shows consistently stabilizing effects, SB exacerbates hepatocellular lysis or exerts only minimal stabilizing effects. This differential behaviour should be taken into account when considering the pros and cons of using purified SB vs. the whole SIL extract in medicinal formulations for liver diseases.


Assuntos
Membrana Celular/efeitos dos fármacos , Hepatócitos/citologia , Hepatócitos/efeitos dos fármacos , Silimarina/farmacologia , Animais , Membrana Celular/metabolismo , Relação Dose-Resposta a Droga , Avaliação Pré-Clínica de Medicamentos , Hepatócitos/metabolismo , Masculino , Octoxinol/toxicidade , Ratos , Ratos Wistar , Silibina , Ácido Tauroquenodesoxicólico/toxicidade
15.
Clin Biochem ; 42(9): 919-21, 2009 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19150444

RESUMO

OBJECTIVE: To evaluate if unconjugated bilirubin (UB) inhibits C1 esterase activity. DESIGN AND METHODS: Esterase activity was evaluated by C1-mediated hydrolysis of N-acetyl-L-tyrosine ethyl ester, and binding of UB to C1r and C1s was assessed by dot-blot analysis. RESULTS: UB inhibited C1 enzymatic activity. C1r, C1s and human serum albumin bound [(14)C]-UB to a similar extent. CONCLUSIONS: UB inhibits C1 esterase activity, apparently due to a direct pigment-protein interaction. This could explain the inhibitory action of UB on complement activation.


Assuntos
Bilirrubina/química , Bilirrubina/farmacologia , Complemento C1s/metabolismo , Ativação Enzimática/efeitos dos fármacos , Humanos , Immunoblotting
16.
Biochim Biophys Acta ; 1770(7): 1003-10, 2007 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-17442495

RESUMO

Hyperbilirubinemia and complement-mediated immune attack on hepatocyte membrane are common features of certain hepatic diseases. To assess whether unconjugated bilirubin (UB) counteracts complement-mediated hepatocytolysis, we first generated a rabbit polyclonal antibody (Ab) against rat hepatocyte plasma membrane (RHPM). An assay performed with isolated rat hepatocytes in the presence of the polyclonal Ab and rat serum as complement donor demonstrated that UB inhibits cell lysis, as lactate dehydrogenase release into the medium was inhibited by the pigment in a dose-dependent manner. Immunofluorescence microscopy studies showed that UB significantly attenuates the binding of C3 to the hepatocyte-Ab complex. Further enzyme immunoassay studies showed that UB interferes the binding of C1q to purified anti-RHPM IgG, also in a dose-dependent manner. A dot-blot assay showed that [14C]-UB binds to C1q and human serum albumin (HSA) to a similar extent. A differential spectrum analysis of UB in the presence of C1q further confirmed that the pigment interacts with this protein. In conclusion, we demonstrated an inhibitory action of UB on complement-mediated Ab-induced hepatocytolysis, this action being evidenced at pathophysiological pigment concentrations (171 microM and higher). A direct binding of the pigment to C1q is likely involved.


Assuntos
Bilirrubina/farmacologia , Membrana Celular/efeitos dos fármacos , Complemento C1q/metabolismo , Proteínas Inativadoras do Complemento/farmacologia , Hepatócitos/efeitos dos fármacos , Animais , Anticorpos/imunologia , Bilirrubina/metabolismo , Membrana Celular/imunologia , Células Cultivadas , Complemento C1q/imunologia , Relação Dose-Resposta Imunológica , Técnicas Imunoenzimáticas , L-Lactato Desidrogenase/metabolismo , Masculino , Microscopia de Fluorescência , Ratos , Ratos Wistar
17.
Biochem Pharmacol ; 69(7): 1113-20, 2005 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-15763547

RESUMO

Estradiol-17beta-d-glucuronide (E(2)17G) and taurolithocholate (TLC) induce acute cholestasis-associated with retrieval of the bile salt export pump (Bsep), which parallels with alteration in transport activity. cAMP stimulates the apically directed vesicular trafficking of transporters, partially preventing these alterations. The hepatoprotector, silymarin, which inhibits cAMP-phosphodiesterase, prevents the cholestasis induced in vivo by both estrogens and TLC. We aimed to assess the ability of silibinin (Sil), the silymarin active component, to prevent the retrieval of Bsep induced by TLC and E(2)17G, and the associated alteration in its transport function. The possible involvement of cAMP as a second messenger and the intracellular signalling pathways implicated were also evaluated. Functional studies were performed analysing the proportion of isolated rat hepatocyte couplets (IRHC) accumulating the fluorescent bile salt analogue, cholyl-lysylfluorescein (CLF), into their sealed canalicular vacuoles. Cellular localisation of Bsep was assessed by immunofluorescent staining. Intracellular levels of cAMP were measured by ELISA. Sil (2.5microM) elevated by 40+/-3% intracellular cAMP, and mimicked the ability of dibutyryl-cAMP (10microM) to prevent internalisation of Bsep and the TLC (2.5microM)- and E(2)17G (50microM)-induced impairment in the capacity of IRHC to accumulate CLF apically. Preventive effects of Sil and dibutyryl-cAMP were not abolished by the specific protein kinase A inhibitors, KT5720 and H89. Contrarily, the intracellular Ca(2+) chelator, BAPTA/AM, significantly blocked the protective effect of both compounds. We conclude that Sil prevented TLC- and E(2)17G-induced bile salt secretory failure, at least in part, by avoiding redistribution of Bsep, by a mechanism probably involving cAMP-induced cytosolic Ca(2+) elevations.


Assuntos
Transportadores de Cassetes de Ligação de ATP/fisiologia , Colestase/fisiopatologia , AMP Cíclico/fisiologia , Estradiol/análogos & derivados , Hepatócitos/fisiologia , Silimarina/farmacologia , Membro 11 da Subfamília B de Transportadores de Cassetes de Ligação de ATP , Transportadores de Cassetes de Ligação de ATP/efeitos dos fármacos , Animais , Bucladesina/farmacologia , Técnicas de Cultura de Células , Colestase/prevenção & controle , Estradiol/toxicidade , Hepatócitos/efeitos dos fármacos , Masculino , Silybum marianum , Ratos , Ratos Wistar , Silibina , Ácido Taurolitocólico/toxicidade
18.
Toxicol Sci ; 83(1): 114-25, 2005 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-15496497

RESUMO

Hydrophobic bile salts induce either necrosis or apoptosis depending on the severity of the injury caused by them. Since bile salt-induced apoptosis is influenced by Ca2+- and protein kinase-signaling pathways, and both necrosis and apoptosis share common initiating mechanisms, we analyzed whether these signaling cascades also influence bile salt-induced necrosis in isolated rat hepatocytes. Taurochenodeoxycholate (TCDC, 0.25-1.50 mM, 2 h) reduced, in a dose-dependent manner, the percentage of viable hepatocytes, and increased the release of the cytosolic enzyme, lactate dehydrogenase (LDH) and alanine aminotransferase (ALAT), and that of the plasma membrane enzyme, alkaline phosphatase (AP). The PKC inhibitors, H7 (100 microM) and chelerythrine (2.5 microM), both prevented significantly TCDC-induced necrosis. On the contrary, the PKA activator, dibutyryl-cAMP, exacerbated TCDC-induced cell damage in a dose-dependent manner; this effect was more likely due to cAMP-mediated PKA activation, as the PKA inhibitor, KT5720 (1 microM), counteracted this effect. Instead, the intracellular Ca2+ chelator, BAPTA/AM (20 microM), was without effect. TCDC (1 mM) increased lipid peroxidation from 0.7 +/- 0.2 to 7.5 +/- 0.9 nmol of malondialdehyde per mg of protein, p < 0.001; the addition of the free radical scavenger, diphenyl-p-phenylendiamine, completely blocked this increase and prevented significantly TCDC-induced necrosis. PKC inhibition induced only a slight attenuation of TCDC-induced lipid peroxidation. Possible mechanisms accounting for the modulatory effect of signal transduction pathways on TCDC-induced necrosis, including signaling influence on TCDC transport events and TCDC-induced oxidative stress, are discussed.


Assuntos
Apoptose/efeitos dos fármacos , Ácidos e Sais Biliares/farmacologia , Hepatócitos/efeitos dos fármacos , Necrose , Transdução de Sinais/efeitos dos fármacos , Animais , Cálcio/metabolismo , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Relação Dose-Resposta a Droga , Ativação Enzimática , Inibidores Enzimáticos/farmacologia , Hepatócitos/enzimologia , Hepatócitos/patologia , Peróxidos Lipídicos/metabolismo , Masculino , Necrose/enzimologia , Proteína Quinase C/metabolismo , Ratos , Ratos Wistar , Ácido Tauroquenodesoxicólico/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...