110 results on '"Mottino AD"'
Search Results
2. Localization status of hepatocellular transporters in cholestasis
- Author
-
Andrés Ernesto Zucchetti, Andrea Carolina Boaglio, Mottino Ad, Fernando A. Crocenzi, Sanchez Pozzi Ej, Marcelo G. Roma, and Ismael Ricardo Barosso
- Subjects
Cholestasis ,Chemistry ,Endosome ,media_common.quotation_subject ,Endocytic cycle ,Transporter ,Compartment (chemistry) ,medicine.disease ,Cell biology ,medicine ,Bile ,Humans ,Secretion ,Signal transduction ,Carrier Proteins ,Internalization ,media_common - Abstract
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.
- Published
- 2012
3. Upregulation of Multidrug Resistance-associated Protein 3 by Acetaminophen May Help to Increase Its Own Clearance
- Author
-
Ghanem, CI, primary and Mottino, AD, additional
- Published
- 2008
- Full Text
- View/download PDF
4. Downregulation of intestinal multidrug resistance transporter 1 in obese mice: Effect on its barrier function and role of TNF-α receptor 1 signaling.
- Author
-
Barranco MM, Perdomo VG, Zecchinati F, Manarin R, Massuh G, Sigal N, Vignaduzzo S, Mottino AD, Villanueva SSM, and García F
- Subjects
- Mice, Animals, Male, Mice, Obese, Rhodamine 123, Down-Regulation, Mice, Inbred C57BL, RNA, Messenger, Drug Resistance, Multiple, Tumor Necrosis Factor-alpha metabolism, Diet, High-Fat
- Abstract
Objectives: Multidrug resistance transporter 1 (Mdr-1) is a relevant component of the intestinal transcellular barrier that decreases absorption of oral drugs, thus modulating their bioavailability. Obese patients with metabolic disorders take medications that are subjected to intestinal metabolism and the Mdr-1-dependent barrier. This study evaluated the effect of a high-fat diet (HFD; 40% fat for 16 wk) on Mdr-1 expression and transport activity in C57BL/6 (C57) male mice. Comparable studies were performed in tumor necrosis factor α (TNF-α) receptor 1 knockout mice (R1KO) to delineate a possible role of TNF-α signaling., Methods: mRNA expression was evaluated by real-time polymerase chain reaction and protein levels by western blotting and immunohistochemistry. Mdr-1 activity was assessed using the everted intestinal sac model, with rhodamine 123 as the substrate. Statistical comparisons were made using the Student t test or one-way analysis of variance followed by the post hoc Tukey test., Results: Mdr-1 protein, as well as its corresponding Mdr1a and Mdr1b mRNA, was decreased in C57-HFD mice compared with controls. Immunohistochemical studies confirmed downregulation of Mdr-1 in situ. These results correlated with a 48% decrease in the basolateral to apical transport of rhodamine 123. In contrast, R1KO-HFD modified neither intestinal Mdr-1 mRNA nor its protein expression or activity. In addition, C57-HFD showed elevated intestinal TNF-α mRNA and protein (enzyme-linked immunosorbent assay) levels, whereas R1KO-HFD was undetectable or had a lower increase, respectively., Conclusions: This study demonstrated an impairment of the Mdr-1 intestinal barrier function induced by HFD as a consequence of downregulation of both Mdr-1 gene homologues, resulting in impaired Mdr-1 protein expression. Inflammatory response mediated by TNF-α receptor 1 signaling was likely involved., (Copyright © 2023 Elsevier Inc. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
5. Protective effect of genistein pre-treatment on paraquat hepatotoxicity in rats.
- Author
-
Semeniuk M, Ceré LI, Ciriaci N, Bucci-Muñoz M, Quiroga AD, Luquita MG, Roma S, Catania VA, Mottino AD, Rigalli JP, and Ruiz ML
- Subjects
- Alanine Transaminase blood, Aldehydes metabolism, Animals, Aspartate Aminotransferases blood, Bile metabolism, Chemical and Drug Induced Liver Injury blood, Chemical and Drug Induced Liver Injury metabolism, Genistein pharmacology, Glutathione metabolism, Glutathione Transferase metabolism, Herbicides, Liver drug effects, Liver metabolism, Male, Paraquat, Protective Agents pharmacology, Rats, Wistar, Rats, Chemical and Drug Induced Liver Injury drug therapy, Genistein therapeutic use, Protective Agents therapeutic use
- Abstract
Paraquat (PQ), an herbicide widely used in agriculture, is considered a highly toxic compound. In hepatocytes, P-glycoprotein (P-gp/Abcb1) is a canalicular transporter involved in PQ extrusion from the cell. Previously, we demonstrated that genistein (GNT) induces P-gp in rat liver. In this study, the protective role of GNT pretreatment towards hepatic damage in a model of acute intoxication with PQ in rats, was investigated. Wistar rats were randomized in 4 groups: Control, GNT (5 mg/kg/day sc, 4 days), PQ (50 mg/kg/day ip, last day) and GNT+ PQ. Hepatic lipoperoxidation (LPO) was evaluated by the thiobarbituric acid reactive substances method. Hepatic levels of 4-hydroxynonenal protein adducts (4-HNEp-add) and glutathione-S-transferase alpha (GSTα) protein expression were evaluated by Western blotting. Hepatic glutathione levels and plasma levels of alanine transaminase (ALT) and aspartate transaminase (AST) were also measured. Biliary excretion of PQ was studied in vivo and in isolated perfused liver. PQ was quantified by HPLC. PQ significantly increased AST and ALT activities, malondialdehyde and 4-HNEp-add levels, whereby pretreatment with GNT ameliorated this effect. PQ biliary excretion remained unchanged after treatments in both experimental models. Hepatic GSTα expression was augmented in GNT group. GNT pretreatment increased hepatic glutathione levels in PQ + GNT group. These results agree with the lower content of 4-HNEp-adds in GNT + PQ group respect to PQ group. Unexpectedly, increased activity of P-gp did not enhance PQ biliary excretion. Thus, GNT protective mechanism is likely through the induction of GSTα which results in increased 4-HNE metabolism before formation of protein adducts., (Copyright © 2021. Published by Elsevier Inc.)
- Published
- 2021
- Full Text
- View/download PDF
6. Multidrug resistance-associated protein 2 is negatively regulated by oxidative stress in rat intestine via a posttranslational mechanism. Impact on its membrane barrier function.
- Author
-
Zecchinati F, Barranco MM, Tocchetti GN, Domínguez CJ, Arana MR, Perdomo VG, Mottino AD, García F, and Villanueva SSM
- Subjects
- Animals, Dose-Response Relationship, Drug, Intestinal Mucosa drug effects, Jejunum drug effects, Male, Microvilli drug effects, Oxidative Stress drug effects, Protein Processing, Post-Translational drug effects, Rats, Rats, Wistar, tert-Butylhydroperoxide toxicity, ATP-Binding Cassette Transporters metabolism, Intestinal Mucosa metabolism, Jejunum metabolism, Microvilli metabolism, Oxidative Stress physiology, Protein Processing, Post-Translational physiology
- Abstract
Oxidative stress (OS) is a key factor in the development of gastrointestinal disorders, in which the intestinal barrier is altered. However, the Multidrug resistance-associated protein 2 (Mrp2) status, an essential component of the intestinal transcellular barrier exhibiting pharmaco-toxicological relevance by limiting the orally ingested toxicants and drugs absorption, has not been investigated. We here evaluated the short-term effect of OS on Mrp2 by treatment of isolated rat intestinal sacs with tert-butyl hydroperoxide (TBH) for 30 min. OS induction by TBH (250 and 500 μM) was confirmed by increased lipid peroxidation end products, decreased reduced glutathione (GSH) content and altered antioxidant enzyme activities. Under this condition, assessment of Mrp2 distribution between brush border (BBM) and intracellular (IM) membrane fractions, showed that Mrp2 protein decreased in BBM and increased in IM, consistent with an internalization process. This was associated with decreased efflux activity and, consequently, impaired barrier function. Subsequent incubation with N-Acetyl-L-Cysteine (NAC, 1 mM) reestablished GSH content and reverted concomitantly the alteration in Mrp2 localization and function induced by TBH. Cotreatment with a specific inhibitor of classic calcium-dependent Protein Kinase C (cPKC) implicated this kinase in TBH-effects. In conclusion, we demonstrated a negative posttranslational regulation of rat intestinal Mrp2 after short-term exposition to OS, a process likely mediated by cPKC and dependent on intracellular GSH content. The concomitant impairment of the Mrp2 barrier function may have implications in xenobiotic absorption and toxicity in a variety of human diseases linked to OS, with notable consequences on the toxicity/safety of therapeutic agents., (Copyright © 2021 Elsevier B.V. All rights reserved.)
- Published
- 2021
- Full Text
- View/download PDF
7. Sirtuin 1 and 2 inhibitors enhance the inhibitory effect of sorafenib in hepatocellular carcinoma cells.
- Author
-
Ceballos MP, Angel A, Delprato CB, Livore VI, Ferretti AC, Lucci A, Comanzo CG, Alvarez ML, Quiroga AD, Mottino AD, and Carrillo MC
- Subjects
- ATP Binding Cassette Transporter, Subfamily G, Member 2 metabolism, Apoptosis drug effects, Apoptosis Regulatory Proteins metabolism, Carcinoma, Hepatocellular enzymology, Carcinoma, Hepatocellular pathology, Cell Cycle Proteins metabolism, Cell Movement drug effects, Cell Proliferation drug effects, Drug Resistance, Neoplasm, Hep G2 Cells, Humans, Liver Neoplasms enzymology, Liver Neoplasms pathology, Multidrug Resistance-Associated Proteins metabolism, Neoplasm Invasiveness, Neoplasm Proteins metabolism, Signal Transduction, Sirtuin 1 metabolism, Sirtuin 2 metabolism, Spheroids, Cellular, Antineoplastic Combined Chemotherapy Protocols pharmacology, Carbazoles pharmacology, Carcinoma, Hepatocellular drug therapy, Histone Deacetylase Inhibitors pharmacology, Liver Neoplasms drug therapy, Naphthalenes pharmacology, Pyrimidinones pharmacology, Sirtuin 1 antagonists & inhibitors, Sirtuin 2 antagonists & inhibitors, Sorafenib pharmacology
- Abstract
Multidrug resistance (MDR) counteracts the efficiency of sorafenib, an important first-line therapy for hepatocellular carcinoma (HCC). Sirtuins (SIRTs) 1 and 2 are associated with tumor progression and MDR. We treated 2D and 3D cultures (which mimic the features of in vivo tumors) from HCC cells with sorafenib alone or in the presence of SIRTs 1 and 2 inhibitors (cambinol or EX-527; combined treatments). Cultures subjected to combined treatments showed a greater fall in cellular viability, proliferation (PCNA, cyclin D1 and Ki-67 expression and cell cycle analysis), migration and invasion when compared with cultures treated only with sorafenib. Similarly, combined treatments produced more apoptosis (annexin V/PI, caspase-3/7 activity) than sorafenib alone. Since cell cycle dysregulation and apoptotic blockage are reported mechanisms of MDR, the modulation found in PCNA, cyclin D1, Ki-67 and caspase-3/7 proteins by cambinol and EX-527 are probably playing a role in enhancing the sensitivity of HCC cell lines to sorafenib. EX-527 reduced MRP3 and BCRP expression in sorafenib-treated HCC cells. Since ABC transporters contribute to MDR, MRP3 and BCRP could be also influencing in the response of HCC cells to sorafenib. Overall, 2D and 3D cultures behave similarly except that 3D cultures were less sensitive to treatments, reinforcing the clinical relevance of the current study. Findings presented in this manuscript support a potential application for SIRTs 1 and 2 inhibitors since we demonstrated that these compounds enhance the inhibitory effect of sorafenib upon treatment of hepatocellular carcinoma cells lines., (Copyright © 2020 Elsevier B.V. All rights reserved.)
- Published
- 2021
- Full Text
- View/download PDF
8. Acute regulation of apical ABC transporters in the gut. Potential influence on drug bioavailability.
- Author
-
Domínguez CJ, Tocchetti GN, Rigalli JP, and Mottino AD
- Subjects
- Animals, Biological Availability, Humans, ATP-Binding Cassette Transporters metabolism, Gastrointestinal Tract metabolism, Pharmaceutical Preparations metabolism
- Abstract
The extensive intestinal surface offers an advantage regarding nutrient, ion and water absorptive capacity but also brings along a high exposition to xenobiotics, including drugs of therapeutic use and food contaminants. After absorption of these compounds by the enterocytes, apical ABC transporters play a key role in secreting them back to the intestinal lumen, hence acting as a transcellular barrier. Rapid and reversible modulation of their activity is a subject of increasing interest for pharmacologists. On the one hand, a decrease in transporter activity may result in increased absorption of therapeutic agents given orally. On the other hand, an increase in transporter activity would decrease their absorption and therapeutic efficacy. Although of less relevance, apical ABC transporters also contribute to disposition of drugs systemically administered. This review article summarizes the present knowledge on the mechanisms aimed to rapidly regulate the activity of the main apical ABC transporters of the gut: multidrug resistance protein 1 (MDR1), multidrug resistance-associated protein 2 (MRP2) and breast cancer resistance protein (BCRP). Regulation of these mechanisms by drugs, drug delivery systems, drug excipients and nutritional components are particularly considered. This information could provide the basis for controlled regulation of bioavailability of therapeutic agents and at the same time would help to prevent potential drug-drug interactions., (Copyright © 2020 Elsevier Ltd. All rights reserved.)
- Published
- 2021
- Full Text
- View/download PDF
9. Intraluminal nutrients acutely strengthen rat intestinal MRP2 barrier function by a glucagon-like peptide-2-mediated mechanism.
- Author
-
Tocchetti GN, Domínguez CJ, Zecchinati F, Arana MR, Rigalli JP, Ruiz ML, Villanueva SSM, and Mottino AD
- Subjects
- Animals, Caco-2 Cells, Humans, Intestinal Mucosa, Nutrients, Rats, Rats, Wistar, ATP-Binding Cassette Transporters, Glucagon-Like Peptide 2
- Abstract
Aim: MRP2 is an intestinal ABC transporter that prevents the absorption of dietary xenobiotics. The aims of this work were: (1) to evaluate whether a short-term regulation of intestinal MRP2 barrier function takes place in vivo after luminal incorporation of nutrients and (2) to explore the underlying mechanism., Methods: MRP2 activity and localization were assessed in an in vivo rat model with preserved irrigation and innervation. Nutrients were administered into distal jejunum. After 30-minutes treatments, MRP2 activity was assessed in proximal jejunum by quantifying the transport of the model substrate 2,4-dinitrophenyl-S-glutathione. MRP2 localization was determined by quantitative confocal microscopy. Participation of extracellular mediators was evaluated using selective inhibitors and by immunoneutralization. Intracellular pathways were explored in differentiated Caco-2 cells., Results: Oleic acid, administered intraluminally at dietary levels, acutely stimulated MRP2 insertion into brush border membrane. This was associated with increased efflux activity and, consequently, enhanced barrier function. Immunoneutralization of the gut hormone glucagon-like peptide-2 (GLP-2) prevented oleic acid effect on MRP2, demonstrating the participation of this trophic factor as a main mediator. Further experiments using selective inhibitors demonstrated that extracellular adenosine synthesis and its subsequent binding to enterocytic A2B adenosine receptor (A2BAR) take place downstream GLP-2. Finally, studies in intestinal Caco-2 cells revealed the participation of A2BAR/cAMP/PKA intracellular pathway, ultimately leading to increased MRP2 localization in apical domains., Conclusion: These findings reveal an on-demand, acute regulation of MRP2-associated barrier function, constituting a novel physiological mechanism of protection against the absorption of dietary xenobiotics in response to food intake., (© 2020 Scandinavian Physiological Society. Published by John Wiley & Sons Ltd.)
- Published
- 2020
- Full Text
- View/download PDF
10. Role of interleukin 1 beta in the regulation of rat intestinal multidrug resistance-associated protein 2 under conditions of experimental endotoxemia.
- Author
-
Arana MR, Dominguez CJ, Zecchinati F, Tocchetti GN, Mottino AD, and Villanueva SSM
- Subjects
- Animals, Blotting, Western, Endotoxemia pathology, Female, Intestinal Mucosa ultrastructure, Microscopy, Confocal, Rats, Rats, Wistar, Real-Time Polymerase Chain Reaction, ATP-Binding Cassette Transporters metabolism, Endotoxemia metabolism, Interleukin-1beta metabolism, Intestinal Mucosa metabolism
- Abstract
Multidrug resistance-associated protein 2 (Mrp2), expressed at the brush border membrane (BBM) of the enterocyte, is an ABC transporter with relevant intestinal barrier function. Its toxicological relevance lies in preventing absorption and tissue accumulation of dietary contaminants, drugs, and potentially harmful endogenous metabolites. Expression and activity of intestinal Mrp2 is downregulated in LPS-induced endotoxemia. In addition, confocal microscopy studies demonstrated internalization of the transporter to endocytic vesicles. Since IL-1β plays an important role as early mediator of LPS-inflammatory responses, we evaluated whether IL-1β mediates LPS-induced impairment of Mrp2 function. Two protocols were used: I) In vivo administration of LPS (5 mg/kg b.wt., i.p., single dose) to rats in simultaneous with administration of anti-IL-1β (25 μg/kg b.wt., i.p., 4 doses), followed by studies of Mrp2 expression, localization and activity, 24 h after LPS administration; II) In vitro incubation of isolated intestinal sacs with IL-1β (10 ng/mL) for 30 min, followed by analysis of Mrp2 activity and localization. We found that in vivo immunoneutralization of IL-1β partially prevented the decrease of Mrp2 protein expression and activity as well as its internalization to intracellular domains induced by LPS. Involvement of IL-1β in the alteration of Mrp2 localization and activity was more directly demonstrated in isolated intestinal sacs, as incubation with IL-1β resulted in detection of Mrp2 in intracellular regions of the enterocyte in simultaneous with alteration of transport activity. In conclusion, IL-1β induces early internalization of intestinal Mrp2, which could partially explain loss of expression at the BBM under conditions of experimental endotoxemia. Concomitant impairment of Mrp2-dependent barrier function may have pathophysiological relevance since IL-1β mediates the effect of many local and systemic inflammatory processes., Competing Interests: Declaration of Competing Interest The authors declare that they have no conflict of interest, (Copyright © 2020 Elsevier B.V. All rights reserved.)
- Published
- 2020
- Full Text
- View/download PDF
11. Regulation of hepatic P-gp expression and activity by genistein in rats.
- Author
-
Semeniuk M, Ceré LI, Ciriaci N, Bucci-Muñoz M, Villanueva SSM, Mottino AD, Catania VA, Rigalli JP, and Ruiz ML
- Subjects
- ATP-Binding Cassette Transporters metabolism, Animals, Carcinoma, Hepatocellular metabolism, Liver metabolism, Liver Neoplasms metabolism, Male, RNA, Messenger metabolism, Rats, ATP Binding Cassette Transporter, Subfamily B, Member 1 metabolism, Anticarcinogenic Agents toxicity, Genistein toxicity
- Abstract
P-glycoprotein (P-gp) is an ABC transporter exhibiting high pharmacotoxicological relevance by extruding a wide range of cytotoxic compounds out of the cells. Previously, we demonstrated that the phytoestrogen genistein (GNT) modulates P-gp expression in hepatocellular carcinoma in vitro. Although several beneficial effects (e.g., antioxidant, antimutagenic, anticancer) have been attributed to GNT, the molecular mechanisms have not been totally elucidated. In the present work, we evaluated the effect of GNT on P-gp expression in rat liver, kidney and ileum. We found that GNT (5 mg/kg daily s.c. 3 days) increased hepatic P-gp expression and also Mdr1a (one of the genes encoding P-gp) mRNA levels. Renal and intestinal P-gp remained unchanged after GNT treatment. Hepatic P-gp activity measured with rhodamine-123 and digoxin, both well-known P-gp substrates, was also increased. In vitro experiments using hepatocyte primary cell culture demonstrated that inhibition of ER-α with ICI182/780 did not prevent Mdr1a mRNA up-regulation by GNT (10 µM). In contrast, Mdr1a induction was suppressed after pregnane X receptor (PXR) inhibition by sulforaphane and knockdown of this nuclear receptor. These findings were confirmed in vivo by using the PXR antagonist ketoconazole. In conclusion, we demonstrated the induction of hepatic P-gp expression and activity by GNT in vivo, with PXR being a likely mediator. This suggests that GNT, at concentrations observed in plasma of individuals consuming the phytoestrogen in the diet or through supplements, could affect the clearance of relevant P-gp substrates of therapeutic use as well as toxicity of environmental and food toxicants.
- Published
- 2020
- Full Text
- View/download PDF
12. Improved hepatic MRP2/ABCC2 transport activity in LPS-induced cholestasis by aquaporin-1 gene transfer.
- Author
-
Marrone J, Tocchetti GN, Danielli M, Mottino AD, and Marinelli RA
- Subjects
- Animals, Aquaporin 1 genetics, Cholestasis therapy, Gene Transfer Techniques, Hepatocytes cytology, Male, Multidrug Resistance-Associated Protein 2, Rats, Wistar, ATP-Binding Cassette Transporters metabolism, Aquaporin 1 physiology, Bile metabolism, Cholestasis metabolism, Hepatocytes metabolism
- Abstract
Multidrug resistance-associated protein 2 (MRP2/ABCC2), a hepatocyte canalicular transporter involved in bile secretion, is downregulated in cholestasis triggered by lipopolysaccharide. The human aquaporin-1 (hAQP1) adenovirus-mediated gene transfer to liver improves cholestasis by incompletely defined mechanisms. Here we found that hAQP1 did not affect MRP2/ABCC2 expression, but significantly increased its transport activity assessed in situ with endogenous and exogenous substrates, likely by a hAQP1-induced increase in canalicular membrane cholesterol amount. Our results suggest that hAQP1-induced MRP2/ABCC2 activation contributes to the cholestasis improvement., (Copyright © 2019 Elsevier B.V. and Société Française de Biochimie et Biologie Moléculaire (SFBBM). All rights reserved.)
- Published
- 2019
- Full Text
- View/download PDF
13. Reversion of down-regulation of intestinal multidrug resistance-associated protein 2 in fructose-fed rats by geraniol and vitamin C: Potential role of inflammatory response and oxidative stress.
- Author
-
Zecchinati F, Barranco MM, Arana MR, Tocchetti GN, Domínguez CJ, Perdomo VG, Ruiz ML, Mottino AD, García F, and Villanueva SSM
- Subjects
- Animals, Anti-Inflammatory Agents, Non-Steroidal pharmacology, Antioxidants pharmacology, Body Weight drug effects, Down-Regulation drug effects, Eating drug effects, Glucose metabolism, Inflammation, Insulin Resistance, Intestinal Mucosa metabolism, Male, Oxidative Stress drug effects, Rats, Wistar, Triglycerides blood, ATP-Binding Cassette Transporters metabolism, Acyclic Monoterpenes pharmacology, Ascorbic Acid pharmacology, Fructose adverse effects, Intestinal Mucosa drug effects
- Abstract
Intestinal multidrug resistance-associated protein 2 is an ABC transporter that limits the absorption of xenobiotics ingested orally, thus acting as essential component of the intestinal biochemical barrier. Metabolic Syndrome (MetS) is a pathological condition characterized by dyslipidemia, hyperinsulinemia, insulin resistance, chronic inflammation, and oxidative stress (OS). In a previous study we demonstrated that MetS-like conditions induced by fructose in drinking water (10% v/v, during 21 days), significantly reduced the expression and activity of intestinal Mrp2 in rats. We here evaluated the potential beneficial effect of geraniol or vitamin C supplementation, natural compounds with anti-inflammatory and anti-oxidant properties, in reverse fructose-induced Mrp2 alterations. After MetS-like conditions were induced (21 days), animals were cotreated with geraniol or vitamin C or vehicle for another 14 days. Decreased expression of Mrp2 protein and mRNA due to fructose administration was reversed by geraniol and by vitamin C, consistent with restoration of Mrp2 activity evaluated in everted intestinal sacs. Concomitantly, increased intestinal IL-1β and IL-6 levels induced by fructose were totally and partially counterbalanced, respectively, by geraniol administration. The intestinal redox unbalance generated by fructose was improved by geraniol and vitamin C, as evidenced by decreasing lipid peroxidation products and activity of Superoxide Dismutase and by normalizing glutathione reduced/oxidized glutathione ratio. The restoration effects exhibited by geraniol and vitamin C suggest that local inflammatory response and OS generated under MetS-like conditions represent important mediators of the intestinal Mrp2 down-regulation. Additionally, both agents could be considered of potential therapeutic value to preserve Mrp2 function under MetS conditions., (Copyright © 2019. Published by Elsevier Inc.)
- Published
- 2019
- Full Text
- View/download PDF
14. Inhibition of multidrug resistance-associated protein 2 (MRP2) activity by the contraceptive nomegestrol acetate in HepG2 and Caco-2 cells.
- Author
-
Tocchetti GN, Domínguez CJ, Zecchinati F, Arana MR, Ruiz ML, Villanueva SSM, Mottino AD, Weiss J, and Rigalli JP
- Subjects
- Caco-2 Cells, Cell Survival drug effects, Hep G2 Cells, Humans, Multidrug Resistance-Associated Protein 2, Multidrug Resistance-Associated Proteins metabolism, Contraceptive Agents pharmacology, Megestrol pharmacology, Multidrug Resistance-Associated Proteins antagonists & inhibitors, Norpregnadienes pharmacology
- Abstract
Multidrug resistance-associated protein 2 (MRP2) plays a key role in hepatic and intestinal disposition of endo- and xenobiotics. Several therapeutic agents modulate MRP2 activity resulting in pharmacological interactions. Nomegestrol acetate (NMGA) is a progestogen increasingly used in contraceptive formulations. The aim of this work was to evaluate the effect of NMGA on MRP2 activity in HepG2 and Caco-2 cells as models of human hepatocytes and enterocytes, respectively. NMGA (5, 50 and 500 nM; 48 h) decreased MRP2-mediated transport of 2,4-dinitrophenyl-S-glutathione in HepG2 cells, with no effect on MRP2 protein expression. Acute exposure (1 h) to the same concentrations of NMGA failed to affect MRP2 activity, ruling out an inhibitory action directly induced by the drug. In contrast, acute incubation with a lysate of HepG2 cells pre-treated with NMGA, containing potential metabolites, reproduced MRP2 inhibition. Preincubation of lysates with sulfatase but not with β-glucuronidase abolished the inhibitory action, strongly suggesting participation of NMGA sulfated derivatives. Western blot studies in plasma vs. intracellular membrane fractions ruled out internalization of MRP2 to be responsible for the impairment of transport activity. MRP2-mediated transport of 5(6)-carboxy-2',7'-dichlorofluorescein was not affected in Caco-2 cells incubated for 48 h with either 5, 50 or 500 nM NMGA. Conversely, acute exposure (1 h) of Caco-2 cells to NMGA-treated HepG2 lysates decreased MRP2 activity, being this effect also prevented by pre-treatment of the lysates with sulfatase. Taken together, these findings demonstrate an inhibitory effect of NMGA sulfated metabolites on hepatic and intestinal MRP2 function. Extrapolated to the in vivo situation, they suggest the possibility of pharmacological interactions with coadministered drugs., (Copyright © 2018 Elsevier B.V. All rights reserved.)
- Published
- 2018
- Full Text
- View/download PDF
15. Biphasic modulation of cAMP levels by the contraceptive nomegestrol acetate. Impact on P-glycoprotein expression and activity in hepatic cells.
- Author
-
Tocchetti GN, Domínguez CJ, Zecchinati F, Arana MR, Ruiz ML, Villanueva SSM, Weiss J, Mottino AD, and Rigalli JP
- Subjects
- ATP Binding Cassette Transporter, Subfamily B, Member 1 agonists, Cyclic AMP antagonists & inhibitors, Dose-Response Relationship, Drug, Gene Expression, Hep G2 Cells, Hepatocytes drug effects, Humans, ATP Binding Cassette Transporter, Subfamily B, Member 1 biosynthesis, Contraceptive Agents pharmacology, Cyclic AMP metabolism, Hepatocytes metabolism, Megestrol pharmacology, Norpregnadienes pharmacology
- Abstract
ABC transporters are key players in drug excretion with alterations in their expression and activity by therapeutic agents potentially leading to drug-drug interactions. The interaction potential of nomegestrol acetate (NMGA), a synthetic progestogen increasingly used as oral contraceptive, had never been explored. In this work we evaluated (1) the effect of NMGA on ABC transporters in the human hepatic cell line HepG2 and (2) the underlying molecular mechanism. NMGA (5, 50 and 500 nM) increased P-glycoprotein (P-gp) expression at both protein and mRNA levels and reduced intracellular calcein accumulation, indicating an increase also in transporter activity. This up-regulation of P-gp was corroborated in Huh7 cells and was independent of the classical progesterone receptor. Instead, using a siRNA-mediated silencing approach, we demonstrated the involvement of membrane progesterone receptor α. Moreover, we found that the activation of this receptor by NMGA led to a falling-rising profile in intracellular cAMP levels and protein kinase A activity over time, ultimately leading to transcriptional P-gp up-regulation. Finally, we identified inhibitory G protein and phosphodiesterases as mediators of this novel biphasic modulation. These results demonstrate the ability of NMGA to selectively up-regulate hepatic P-gp expression and activity and constitute the first report of ABC transporter modulation by membrane progesterone receptor α. If a similar regulation took place in vivo, decreased bioavailability and therapeutic efficacy of NMGA-coadministered P-gp substrates could be expected. This holds special importance considering long-term administration of NMGA and broad substrate specificity of P-gp., (Copyright © 2018 Elsevier Inc. All rights reserved.)
- Published
- 2018
- Full Text
- View/download PDF
16. Inhibition of sirtuins 1 and 2 impairs cell survival and migration and modulates the expression of P-glycoprotein and MRP3 in hepatocellular carcinoma cell lines.
- Author
-
Ceballos MP, Decándido G, Quiroga AD, Comanzo CG, Livore VI, Lorenzetti F, Lambertucci F, Chazarreta-Cifre L, Banchio C, Alvarez ML, Mottino AD, and Carrillo MC
- Subjects
- ATP Binding Cassette Transporter, Subfamily B antagonists & inhibitors, ATP Binding Cassette Transporter, Subfamily B genetics, Acetylation drug effects, Antineoplastic Agents pharmacology, Carbazoles pharmacology, Carcinoma, Hepatocellular enzymology, Carcinoma, Hepatocellular metabolism, Cell Line, Tumor, Cell Movement drug effects, Cell Survival drug effects, Drug Resistance, Multiple, Drug Resistance, Neoplasm, Gene Expression Regulation, Neoplastic drug effects, Humans, Liver Neoplasms enzymology, Liver Neoplasms metabolism, Multidrug Resistance-Associated Proteins antagonists & inhibitors, Multidrug Resistance-Associated Proteins genetics, Naphthalenes pharmacology, Neoplasm Proteins antagonists & inhibitors, Neoplasm Proteins genetics, Neoplasm Proteins metabolism, Protein Processing, Post-Translational drug effects, Pyrimidinones pharmacology, Sirtuin 1 metabolism, Sirtuin 2 metabolism, ATP Binding Cassette Transporter, Subfamily B metabolism, Carcinoma, Hepatocellular drug therapy, Histone Deacetylase Inhibitors pharmacology, Liver Neoplasms drug therapy, Multidrug Resistance-Associated Proteins metabolism, Sirtuin 1 antagonists & inhibitors, Sirtuin 2 antagonists & inhibitors
- Abstract
Sirtuins (SIRTs) 1 and 2 deacetylases are overexpressed in hepatocellular carcinoma (HCC) and are associated with tumoral progression and multidrug resistance (MDR). In this study we analyzed whether SIRTs 1 and 2 activities blockage was able to affect cellular survival and migration and to modulate p53 and FoxO1 acetylation in HepG2 and Huh7 cells. Moreover, we analyzed ABC transporters P-glycoprotein (P-gp) and multidrug resistance-associated protein 3 (MRP3) expression. We used cambinol and EX-527 as SIRTs inhibitors. Both drugs reduced cellular viability, number of colonies and cellular migration and augmented apoptosis. In 3D cultures, SIRTs inhibitors diminished spheroid growth and viability. 3D culture was less sensitive to drugs than 2D culture. The levels of acetylated p53 and FoxO1 increased after treatments. Drugs induced a decrease in ABC transporters mRNA and protein levels in HepG2 cells; however, only EX-527 was able to reduce MRP3 mRNA and protein levels in Huh7 cells. This is the first work demonstrating the regulation of MRP3 by SIRTs. In conclusion, both drugs decreased HCC cells survival and migration, suggesting SIRTs 1 and 2 activities blockage could be beneficial during HCC therapy. Downregulation of the expression of P-gp and MRP3 supports the potential application of SIRTs 1 and 2 inhibitions in combination with conventional chemotherapy., (Copyright © 2018 Elsevier B.V. All rights reserved.)
- Published
- 2018
- Full Text
- View/download PDF
17. Acute regulation of multidrug resistance-associated protein 2 localization and activity by cAMP and estradiol-17β-D-glucuronide in rat intestine and Caco-2 cells.
- Author
-
Tocchetti GN, Arias A, Arana MR, Rigalli JP, Domínguez CJ, Zecchinati F, Ruiz ML, Villanueva SSM, and Mottino AD
- Subjects
- Animals, Caco-2 Cells, Cell Membrane metabolism, Cyclic AMP, Estradiol pharmacology, Humans, Intestinal Mucosa metabolism, Male, Multidrug Resistance-Associated Protein 2, Rats, Rats, Wistar, ATP-Binding Cassette Transporters metabolism, Bucladesine pharmacology, Estradiol analogs & derivatives, Intestinal Mucosa drug effects, Multidrug Resistance-Associated Proteins metabolism
- Abstract
Multidrug resistance-associated protein 2 (MRP2) is an ATP-dependent transporter expressed at the brush border membrane of the enterocyte that confers protection against absorption of toxicants from foods or bile. Acute, short-term regulation of intestinal MRP2 activity involving changes in its apical membrane localization was poorly explored. We evaluated the effects of dibutyryl-cAMP (db-cAMP), a permeable analog of cAMP, and estradiol-17β-D-glucuronide (E
2 17G), an endogenous derivative of estradiol, on MRP2 localization and activity using isolated rat intestinal sacs and Caco-2 cells, a model of human intestinal epithelium. Changes in MRP2 localization were studied by Western blotting of plasma membrane (PM) vs. intracellular membrane (IM) fractions in both experimental models, and additionally, by confocal microscopy in Caco-2 cells. After 30 min of exposure, db-cAMP-stimulated sorting of MRP2 from IM to PM both in rat jejunum and Caco-2 cells at 10 and 100 µM concentrations, respectively, with increased excretion of the model substrate 2,4-dinitrophenyl-S-glutathione. In contrast, E2 17G (400 µM) induced internalization of MRP2 together with impairment of transport activity. Confocal microscopy analysis performed in Caco-2 cells confirmed Western blot results. In the particular case of E2 17G, MRP2 exhibited an unusual pattern of staining compatible with endocytic vesiculation. Use of selective inhibitors demonstrated the participation of cAMP-dependent protein kinase and classic calcium-dependent protein kinase C in db-cAMP and E2 17G effects, respectively. We conclude that localization of MRP2 in intestine may be subjected to a dynamic equilibrium between plasma membrane and intracellular domains, thus allowing for rapid regulation of MRP2 function.- Published
- 2018
- Full Text
- View/download PDF
18. Glucagon-like peptide 2 prevents down-regulation of intestinal multidrug resistance-associated protein 2 and P-glycoprotein in endotoxemic rats.
- Author
-
Arana MR, Tocchetti GN, Zecchinati F, Londero AS, Dominguez C, Perdomo V, Rigalli JP, Villanueva SSM, and Mottino AD
- Subjects
- ATP Binding Cassette Transporter, Subfamily B genetics, ATP Binding Cassette Transporter, Subfamily B metabolism, ATP-Binding Cassette Transporters genetics, Animals, Antioxidants metabolism, Disease Models, Animal, Down-Regulation, Drug Administration Schedule, Endotoxemia chemically induced, Endotoxemia metabolism, Female, Glutathione metabolism, Injections, Subcutaneous, Interleukin-1beta metabolism, Intestinal Absorption, Lipopolysaccharides, Oxidation-Reduction, Oxidative Stress drug effects, Permeability, Rats, Wistar, Time Factors, ATP-Binding Cassette Transporters metabolism, Endotoxemia prevention & control, Glucagon-Like Peptide 2 administration & dosage, Jejunum metabolism
- Abstract
Multidrug resistance-associated protein 2 (Mrp2, ABCC2) and P-glycoprotein (P-gp, ABCB1) constitute essential components of the intestinal biochemical barrier that prevent incorporation of food contaminants, drugs or toxic metabolites into the blood stream. Endotoxemia induced in rats by administration of bacterial lipopolysaccharide (LPS) results in elevated intestinal permeability and toxicity of xenobiotics in part associated with down-regulation of expression and activity of Mrp2 and P-gp. We evaluated the protective effect of glucagon-like peptide 2 (GLP-2), a peptide hormone with enterotrophic properties, on Mrp2 and P-gp alterations induced by single i.p. injection of LPS (5mg/kg b.wt.) to rats. Two different protocols of GLP-2 administration, namely prevention and reversion, were examined. The prevention protocol consisted of 7s.c. injections of GLP-2 (125μg/kg b.wt.) administered every 12h, starting 60h before LPS administration. The reversion protocol consisted of 2 doses of GLP-2, starting 3h after LPS injection. Intestinal samples were collected 24h after LPS administration and expression (protein and mRNA) and activity of Mrp2 were evaluated in proximal jejunum whereas those of P-gp were studied in ileum. GLP-2 completely neutralized down-regulation of expression of Mrp2 and P-gp and loss of their respective activities induced by LPS under prevention protocol. GLP-2 was also able to prevent internalization of both transporters from the apical membrane of the enterocyte to intracellular compartments, as detected by confocal microscopy. LPS induced an increase in IL-1β and oxidized glutathione tissue levels, which were also counterbalanced by GLP-2 administration. In contrast, the reversion protocol failed to attenuate Mrp2 and P-gp down-regulation induced by LPS. We conclude that GLP-2 can prevent down-regulation of intestinal expression and activity of Mrp2 and P-gp in endotoxemic rats and that IL-1β and oxidative stress constitute potential targets of GLP-2 protective effects., (Copyright © 2017 Elsevier B.V. All rights reserved.)
- Published
- 2017
- Full Text
- View/download PDF
19. Intestinal multidrug resistance-associated protein 2 is down-regulated in fructose-fed rats.
- Author
-
Londero AS, Arana MR, Perdomo VG, Tocchetti GN, Zecchinati F, Ghanem CI, Ruiz ML, Rigalli JP, Mottino AD, García F, and Villanueva SSM
- Subjects
- ATP-Binding Cassette Transporters genetics, Animals, Antioxidants metabolism, Body Weight drug effects, Cytokines metabolism, Disease Models, Animal, Down-Regulation drug effects, Glutathione Transferase metabolism, Intestinal Mucosa metabolism, Lipid Peroxidation drug effects, Male, Metabolic Syndrome chemically induced, Rats, Wistar, Superoxide Dismutase metabolism, ATP-Binding Cassette Transporters metabolism, Fructose adverse effects, Intestines drug effects
- Abstract
Expression and activity of jejunal multidrug resistance-associated protein 2 (Mrp2) and glutathione-S-transferase (GST) were examined in fructose fed Wistar rats, an experimental model of metabolic syndrome. Animals were fed on (a) control diet or (b) control diet plus 10% w/vol fructose in the drinking water. Mrp2 and the α class of GST proteins as well as their corresponding mRNAs were decreased, suggesting a transcriptional regulation by fructose. Confocal microscopy studies reaffirmed down-regulation of Mrp2. Everted intestinal sacs were incubated with 1-chloro-2,4-dinitrobenzene in the mucosal compartment, and the glutathione-conjugated derivative, dinitrophenyl- S-glutathione (DNP-SG; model Mrp2 substrate), was measured in the same compartment to estimate Mrp2 activity. Excretion of DNP-SG was substantially decreased by fructose treatment, consistent with simultaneous down-regulation of Mrp2 and GST. In addition, the effect of fructose on intestinal barrier function exerted by Mrp2 was evaluated in vivo using valsartan, a recognized Mrp2 substrate of therapeutic use. After intraduodenal administration as a bolus, intestinal absorption of valsartan was increased in fructose-drinking animals. Fructose administration also induced oxidative stress in intestinal tissue as demonstrated by significant increases of intestinal lipid peroxidation end products and activity of the antioxidant enzyme superoxide dismutase, by a decreased GSH/GSSG ratio. Moreover, fructose treatment conduced to increased intestinal levels of the proinflammatory cytokines IL-β1 and IL-6. Collectively, our results demonstrate that metabolic syndrome-like conditions, induced by a fructose-rich diet, result in down-regulation of intestinal Mrp2 expression and activity and consequently in an impairment of its barrier function., (Copyright © 2016 Elsevier Inc. All rights reserved.)
- Published
- 2017
- Full Text
- View/download PDF
20. Modulation of expression and activity of intestinal multidrug resistance-associated protein 2 by xenobiotics.
- Author
-
Tocchetti GN, Rigalli JP, Arana MR, Villanueva SSM, and Mottino AD
- Subjects
- Animals, Constitutive Androstane Receptor, Humans, Intestinal Mucosa metabolism, Multidrug Resistance-Associated Protein 2, Multidrug Resistance-Associated Proteins chemistry, Multidrug Resistance-Associated Proteins metabolism, Xenobiotics pharmacology
- Abstract
The multidrug resistance-associated protein 2 (MRP2/ABCC2) is a transporter that belongs to the ATP-binding cassette (ABC) superfamily. In the intestine, it is localized to the apical membrane of the enterocyte and plays a key role in limiting the absorption of xenobiotics incorporated orally. MRP2 may also play a role in systemic clearance of xenobiotics available from the serosal side of the intestine. MRP2 transports a wide range of substrates, mainly organic anions conjugated with glucuronic acid, glutathione and sulfate and its expression can be modulated by xenobiotics at transcriptional- and post-transcriptional levels. Transcriptional regulation is usually mediated by a group of nuclear receptors. The pregnane X receptor (PXR) is a major member of this group. Relevant drugs described to up-regulate intestinal MRP2 via PXR are rifampicin, spironolactone and carbamazepine, among others. The constitutive androstane receptor (CAR, NR1I3) was also reported to modulate MRP2 expression, phenobarbital being a typical activator. Dietary compounds, including micronutrients and other natural products, are also capable of regulating intestinal MRP2 expression transcriptionally. We have given them particular attention since the composition of the food ingested daily is not necessarily supervised and may result in interactions with therapeutic drugs. Post-transcriptional regulation of MRP2 activity by xenobiotics, e.g. as a consequence of inhibitory actions, is also described in this review. Unfortunately, only few studies report on drug-drug or nutrient-drug interactions as a consequence of modulation of intestinal MRP2 activity by xenobiotics. Future clinical studies are expected to identify additional interactions resulting in changes in efficacy or safety of therapeutic drugs., (Copyright © 2016 Elsevier Inc. All rights reserved.)
- Published
- 2016
- Full Text
- View/download PDF
21. Acetaminophen from liver to brain: New insights into drug pharmacological action and toxicity.
- Author
-
Ghanem CI, Pérez MJ, Manautou JE, and Mottino AD
- Subjects
- Acetylcysteine therapeutic use, Animals, Brain metabolism, Chemical and Drug Induced Liver Injury drug therapy, Chemical and Drug Induced Liver Injury etiology, Chemical and Drug Induced Liver Injury metabolism, Humans, Liver metabolism, Acetaminophen pharmacology, Acetaminophen toxicity, Brain drug effects, Liver drug effects
- Abstract
Acetaminophen (APAP) is a well-known analgesic and antipyretic drug. It is considered to be safe when administered within its therapeutic range, but in cases of acute intoxication, hepatotoxicity can occur. APAP overdose is the leading cause of acute liver failure in the northern hemisphere. Historically, studies on APAP toxicity have been focused on liver, with alterations in brain function attributed to secondary effects of acute liver failure. However, in the last decade the pharmacological mechanism of APAP as a cannabinoid system modulator has been documented and some articles have reported "in situ" toxicity by APAP in brain tissue at high doses. Paradoxically, low doses of APAP have been reported to produce the opposite, neuroprotective effects. In this paper we present a comprehensive, up-to-date overview of hepatic toxicity as well as a thorough review of both toxic and beneficial effects of APAP in brain., (Copyright © 2016 Elsevier Ltd. All rights reserved.)
- Published
- 2016
- Full Text
- View/download PDF
22. Physiological and pathophysiological factors affecting the expression and activity of the drug transporter MRP2 in intestine. Impact on its function as membrane barrier.
- Author
-
Arana MR, Tocchetti GN, Rigalli JP, Mottino AD, and Villanueva SS
- Subjects
- Animals, Humans, Multidrug Resistance-Associated Protein 2, Intestines physiology, Intestines physiopathology, Multidrug Resistance-Associated Proteins physiology
- Abstract
The gastrointestinal epithelium functions as a selective barrier to absorb nutrients, electrolytes and water, but at the same time restricts the passage into the systemic circulation of intraluminal potentially toxic compounds. This epithelium maintains its selective barrier function through the presence of very selective and complex intercellular junctions and the ability of the absorptive cells to reject those compounds. Accordingly, the enterocytes metabolize orally incorporated xenobiotics and secrete the hydrophilic metabolites back into the intestinal lumen through specific transporters localized apically. In the recent decades, there has been increasing recognition of the existence of the intestinal cellular barrier. In the present review we focus on the role of the multidrug resistance-associated protein 2 (MRP2, ABCC2) in the apical membrane of the enterocytes, as an important component of this intestinal barrier, as well as on its regulation. We provide a detailed compilation of significant contributions demonstrating that MRP2 expression and function vary under relevant physiological and pathophysiological conditions. Because MRP2 activity modulates the availability and pharmacokinetics of many therapeutic drugs administered orally, their therapeutic efficacy and safety may vary as well., (Copyright © 2016 Elsevier Ltd. All rights reserved.)
- Published
- 2016
- Full Text
- View/download PDF
23. Modulation of Expression and Activity of ABC Transporters by the Phytoestrogen Genistein. Impact on Drug Disposition.
- Author
-
Rigalli JP, Ciriaci N, Mottino AD, Catania VA, and Ruiz ML
- Subjects
- Drug Interactions, Drug Resistance, Multiple drug effects, Drug Resistance, Neoplasm drug effects, Genistein metabolism, Humans, Neoplasms drug therapy, Neoplasms metabolism, Neoplasms pathology, Phytoestrogens metabolism, ATP-Binding Cassette Transporters metabolism, Genistein pharmacology, Phytoestrogens pharmacology
- Abstract
ATP binding cassette (ABC) transporters are involved in drug absorption, distribution and elimination. They also mediate multidrug resistance in cancer cells. Isoflavones, such as genistein (GNT), belong to a class of naturally-occurring compounds found at high concentrations in commonly consumed soya based-foods and dietary supplements. GNT and its metabolites interact with ABC transporters as substrates, inhibitors and/or modulators of their expression. This review compiles information about regulation of ABC transporters by GNT with special emphasis on the three major groups of ABC transporters involved in excretion of endo- and xenobiotics as follows: Pglycoprotein (MDR1, ABCB1), a group of multidrug resistance associated proteins (MRPs, ABCC subfamily) and ABCG2 (BCRP), an ABC half-transporter. The impact of these regulations on potential GNT-drug interactions is further considered.
- Published
- 2016
- Full Text
- View/download PDF
24. Coordinated induction of GST and MRP2 by cAMP in Caco-2 cells: Role of protein kinase A signaling pathway and toxicological relevance.
- Author
-
Arana MR, Tocchetti GN, Domizi P, Arias A, Rigalli JP, Ruiz ML, Luquita MG, Banchio C, Mottino AD, and Villanueva SSM
- Subjects
- CREB-Binding Protein metabolism, Caco-2 Cells, Colforsin pharmacology, Dinitrochlorobenzene pharmacology, Dose-Response Relationship, Drug, Humans, Multidrug Resistance-Associated Protein 2, Real-Time Polymerase Chain Reaction, Signal Transduction, Transcription Factor AP-1 metabolism, Cyclic AMP metabolism, Cyclic AMP-Dependent Protein Kinases metabolism, Glutathione Transferase biosynthesis, Multidrug Resistance-Associated Proteins biosynthesis
- Abstract
The cAMP pathway is a universal signaling pathway regulating many cellular processes including metabolic routes, growth and differentiation. However, its effects on xenobiotic biotransformation and transport systems are poorly characterized. The effect of cAMP on expression and activity of GST and MRP2 was evaluated in Caco-2 cells, a model of intestinal epithelium. Cells incubated with the cAMP permeable analog dibutyryl cyclic AMP (db-cAMP: 1,10,100 μM) for 48 h exhibited a dose-response increase in GST class α and MRP2 protein expression. Incubation with forskolin, an activator of adenylyl cyclase, confirmed the association between intracellular cAMP and upregulation of MRP2. Consistent with increased expression of GSTα and MRP2, db-cAMP enhanced their activities, as well as cytoprotection against the common substrate 1-chloro-2,4-dinitrobenzene. Pretreatment with protein kinase A (PKA) inhibitors totally abolished upregulation of MRP2 and GSTα induced by db-cAMP. In silico analysis together with experiments consisting of treatment with db-cAMP of Caco-2 cells transfected with a reporter construct containing CRE and AP-1 sites evidenced participation of these sites in MRP2 upregulation. Further studies involving the transcription factors CREB and AP-1 (c-JUN, c-FOS and ATF2) demonstrated increased levels of total c-JUN and phosphorylation of c-JUN and ATF2 by db-cAMP, which were suppressed by a PKA inhibitor. Co-immunoprecipitation and ChIP assay studies demonstrated that db-cAMP increased c-JUN/ATF2 interaction, with further recruitment to the region of the MRP2 promoter containing CRE and AP-1 sites. We conclude that cAMP induces GSTα and MRP2 expression and activity in Caco-2 cells via the PKA pathway, thus regulating detoxification of specific xenobiotics., (Copyright © 2015 Elsevier Inc. All rights reserved.)
- Published
- 2015
- Full Text
- View/download PDF
25. Regulation of multidrug resistance proteins by genistein in a hepatocarcinoma cell line: impact on sorafenib cytotoxicity.
- Author
-
Rigalli JP, Ciriaci N, Arias A, Ceballos MP, Villanueva SS, Luquita MG, Mottino AD, Ghanem CI, Catania VA, and Ruiz ML
- Subjects
- Antineoplastic Agents pharmacology, Blotting, Western, Carcinoma, Hepatocellular drug therapy, Carcinoma, Hepatocellular metabolism, Cell Proliferation drug effects, Humans, Liver Neoplasms drug therapy, Liver Neoplasms metabolism, Liver Neoplasms pathology, Membrane Transport Proteins drug effects, Membrane Transport Proteins genetics, MicroRNAs genetics, Niacinamide pharmacology, Phytoestrogens pharmacology, RNA, Messenger genetics, Real-Time Polymerase Chain Reaction, Reverse Transcriptase Polymerase Chain Reaction, Sorafenib, Tumor Cells, Cultured, Apoptosis drug effects, Carcinoma, Hepatocellular pathology, Drug Resistance, Neoplasm drug effects, Gene Expression Regulation, Neoplastic drug effects, Genistein pharmacology, Membrane Transport Proteins metabolism, Niacinamide analogs & derivatives, Phenylurea Compounds pharmacology
- Abstract
Hepatocellular carcinoma (HCC) is the fifth most frequent cancer worldwide. Sorafenib is the only drug available that improves the overall survival of HCC patients. P-glycoprotein (P-gp), Multidrug resistance-associated proteins 2 and 3 (MRP2 and 3) and Breast cancer resistance protein (BCRP) are efflux pumps that play a key role in cancer chemoresistance. Their modulation by dietary compounds may affect the intracellular accumulation and therapeutic efficacy of drugs that are substrates of these transporters. Genistein (GNT) is a phytoestrogen abundant in soybean that exerts its genomic effects through Estrogen-Receptors and Pregnane-X-Receptor (PXR), which are involved in the regulation of the above-mentioned transporters. We evaluated the effect of GNT on the expression and activity of P-gp, MRP2, MRP3 and BCRP in HCC-derived HepG2 cells. GNT (at 1.0 and 10 μM) increased P-gp and MRP2 protein expression and activity, correlating well with an increased resistance to sorafenib cytotoxicity as detected by the methylthiazole tetrazolium (MTT) assay. GNT induced P-gp and MRP2 mRNA expression at 10 but not at 1.0 μM concentration suggesting a different pattern of regulation depending on the concentration. Induction of both transporters by 1.0 μM GNT was prevented by cycloheximide, suggesting translational regulation. Downregulation of expression of the miR-379 by GNT could be associated with translational regulation of MRP2. Silencing of PXR abolished P-gp induction by GNT (at 1.0 and 10 μM) and MRP2 induction by GNT (only at 10 μM), suggesting partial mediation of GNT effects by PXR. Taken together, the data suggest the possibility of nutrient-drug interactions leading to enhanced chemoresistance in HCC when GNT is ingested with soy rich diets or dietary supplements.
- Published
- 2015
- Full Text
- View/download PDF
26. Regulation of expression and activity of multidrug resistance proteins MRP2 and MDR1 by estrogenic compounds in Caco-2 cells. Role in prevention of xenobiotic-induced cytotoxicity.
- Author
-
Arias A, Rigalli JP, Villanueva SS, Ruiz ML, Luquita MG, Perdomo VG, Vore M, Catania VA, and Mottino AD
- Subjects
- ATP Binding Cassette Transporter, Subfamily B, ATP Binding Cassette Transporter, Subfamily B, Member 1 genetics, ATP Binding Cassette Transporter, Subfamily B, Member 1 metabolism, ATP Binding Cassette Transporter, Subfamily G, Member 2, ATP-Binding Cassette Transporters genetics, ATP-Binding Cassette Transporters metabolism, Caco-2 Cells, Dinitrochlorobenzene toxicity, Dose-Response Relationship, Drug, Estrogen Antagonists pharmacology, Estrogen Receptor beta genetics, Ethinyl Estradiol administration & dosage, Gene Expression Regulation drug effects, Genistein administration & dosage, Humans, Intestinal Mucosa drug effects, Intestinal Mucosa metabolism, Multidrug Resistance-Associated Protein 2, Multidrug Resistance-Associated Proteins genetics, Multidrug Resistance-Associated Proteins metabolism, Neoplasm Proteins genetics, Neoplasm Proteins metabolism, Paraquat toxicity, RNA, Messenger metabolism, Glycine max chemistry, Up-Regulation drug effects, Xenobiotics toxicity, ATP Binding Cassette Transporter, Subfamily B, Member 1 drug effects, ATP-Binding Cassette Transporters drug effects, Ethinyl Estradiol pharmacology, Genistein pharmacology, Multidrug Resistance-Associated Proteins drug effects, Neoplasm Proteins drug effects
- Abstract
ABC transporters including MRP2, MDR1 and BCRP play a major role in tissue defense. Epidemiological and experimental studies suggest a cytoprotective role of estrogens in intestine, though the mechanism remains poorly understood. We evaluated whether pharmacologic concentrations of ethynylestradiol (EE, 0.05pM to 5nM), or concentrations of genistein (GNT) associated with soy ingestion (0.1-10μM), affect the expression and activity of multidrug resistance proteins MRP2, MDR1 and BCRP using Caco-2 cells, an in vitro model of intestinal epithelium. We found that incubation with 5pM EE and 1μM GNT for 48h increased expression and activity of both MRP2 and MDR1. Estrogens did not affect expression of BCRP protein at any concentration studied. Irrespective of the estrogen tested, up-regulation of MDR1 and MRP2 protein was accompanied by increased levels of MDR1 mRNA, whereas MRP2 mRNA remained unchanged. Cytotoxicity assays demonstrated association of MRP2 and MDR1 up-regulation with increased resistance to cell death induced by 1-chloro-2,4-dinitrobenzene, an MRP2 substrate precursor, and by paraquat, an MDR1 substrate. Experiments using an estrogen receptor (ER) antagonist implicate ER participation in MRP2 and MDR1 regulation. GNT but not EE increased the expression of ERβ, the most abundant form in human intestine and in Caco-2 cells, which could lead in turn to increased sensitivity to estrogens. We conclude that specific concentrations of estrogens can confer resistance against cytotoxicity in Caco-2 cells, due in part to positive modulation of ABC transporters involved in extrusion of their toxic substrates. Although extrapolation of these results to the in vivo situation must be cautiously done, the data could explain tentatively the cytoprotective role of estrogens against chemical injury in intestine., (Copyright © 2014 Elsevier Ireland Ltd. All rights reserved.)
- Published
- 2014
- Full Text
- View/download PDF
27. Physiological concentrations of unconjugated bilirubin prevent oxidative stress-induced hepatocanalicular dysfunction and cholestasis.
- Author
-
Basiglio CL, Toledo FD, Boaglio AC, Arriaga SM, Ochoa JE, Sánchez Pozzi EJ, Mottino AD, and Roma MG
- Subjects
- ATP Binding Cassette Transporter, Subfamily B, Member 11, ATP-Binding Cassette Transporters metabolism, Actins metabolism, Animals, Bile Acids and Salts metabolism, Bilirubin metabolism, Cholestasis metabolism, Glutathione metabolism, Hepatocytes drug effects, Hepatocytes metabolism, Liver metabolism, Male, Organ Culture Techniques, Protein Kinase C-alpha metabolism, Rats, Rats, Wistar, tert-Butylhydroperoxide pharmacology, Bilirubin pharmacology, Cholestasis prevention & control, Liver drug effects, Liver physiopathology, Oxidative Stress drug effects
- Abstract
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.
- Published
- 2014
- Full Text
- View/download PDF
28. Acetaminophen inhibits intestinal p-glycoprotein transport activity.
- Author
-
Novak A, Carpini GD, Ruiz ML, Luquita MG, Rubio MC, Mottino AD, and Ghanem CI
- Subjects
- Animals, Cell Line, Tumor, Cyclosporins pharmacology, Digoxin pharmacology, Enterocytes drug effects, Enterocytes metabolism, Hep G2 Cells, Humans, Intestinal Absorption drug effects, Intestinal Mucosa drug effects, Intestinal Mucosa metabolism, Male, Permeability drug effects, Rats, Rats, Wistar, Rhodamine 123 pharmacology, Verapamil pharmacology, ATP Binding Cassette Transporter, Subfamily B, Member 1 antagonists & inhibitors, ATP Binding Cassette Transporter, Subfamily B, Member 1 metabolism, Acetaminophen pharmacology, Biological Transport, Active drug effects, Intestines drug effects
- Abstract
Repeated acetaminophen (AP) administration modulates intestinal P-glycoprotein (P-gp) expression. Whether AP can modulate P-gp activity in a short-term fashion is unknown. We investigated the acute effect of AP on rat intestinal P-gp activity in vivo and in vitro. In everted intestinal sacs, AP inhibited serosal-mucosal transport of rhodamine 123 (R123), a prototypical P-gp substrate. R123 efflux plotted against R123 concentration adjusted well to a sigmoidal curve. Vmax decreased 50% in the presence of AP, with no modification in EC50, or slope, ruling out the possibility of inhibition to be competitive. Inhibition by AP was absent at 0°C, consistent with interference of the active transport of R123 by AP. Additionally, AP showed no effect on normal localization of P-gp at the apical membrane of the enterocyte and neither affected paracellular permeability. Consistent with absence of a competitive inhibition, two further strategies strongly suggested that AP is not a P-gp substrate. First, serosal-mucosal transport of AP was not affected by the classical P-gp inhibitors verapamil or Psc 833. Second, AP accumulation was not different between P-gp knock-down and wild-type HepG2 cells. In vivo intestinal absorption of digoxin, another substrate of P-gp, was assessed in the presence or absence of AP (100 μM). Portal digoxin concentration was increased by 214%, in average, by AP, as compared with digoxin alone. In conclusion, AP inhibited P-gp activity, increasing intestinal absorption of digoxin, a prototypical substrate. These results suggest that therapeutic efficacy of P-gp substrates can be altered if coadministered with AP., (© 2013 Wiley Periodicals, Inc. and the American Pharmacists Association.)
- Published
- 2013
- Full Text
- View/download PDF
29. Hormonal regulation of hepatic drug biotransformation and transport systems.
- Author
-
Ruiz ML, Mottino AD, Catania VA, and Vore M
- Subjects
- Animals, Female, Humans, Male, Sex Characteristics, Biotransformation, Gonadal Hormones metabolism, Ion Pumps metabolism, Liver metabolism
- Abstract
The human body is constantly exposed to many xenobiotics including environmental pollutants, food additives, therapeutic drugs, etc. The liver is considered the primary site for drug metabolism and elimination pathways, consisting in uptake, phase I and II reactions, and efflux processes, usually acting in this same order. Modulation of biotransformation and disposition of drugs of clinical application has important therapeutic and toxicological implications. We here provide a compilation and analysis of relevant, more recent literature reporting hormonal regulation of hepatic drug biotransformation and transport systems. We provide additional information on the effect of hormones that tentatively explain differences between sexes. A brief discussion on discrepancies between experimental models and species, as well as a link between gender-related differences and the hormonal mechanism explaining such differences, is also presented. Finally, we include a comment on the pathophysiological, toxicological, and pharmacological relevance of these regulations.
- Published
- 2013
- Full Text
- View/download PDF
30. Estrogen receptor-α mediates human multidrug resistance associated protein 3 induction by 17α-ethynylestradiol. Role of activator protein-1.
- Author
-
Ruiz ML, Rigalli JP, Arias A, Villanueva SS, Banchio C, Vore M, Mottino AD, and Catania VA
- Subjects
- Base Sequence, Hep G2 Cells, Humans, Molecular Sequence Data, Multidrug Resistance-Associated Proteins genetics, Estrogen Receptor alpha physiology, Ethinyl Estradiol pharmacology, Multidrug Resistance-Associated Proteins biosynthesis, Transcription Factor AP-1 physiology
- Abstract
Previously, we have demonstrated that 17α-ethynylestradiol (EE) induces rat multidrug-resistance associated protein 3 (Mrp3, Abcc3) expression transcriptionally through estrogen receptor-α (ER-α) activation. We explored the effect of EE on MRP3 expression of human origin. HepG2 cells were transfected with ER-α and incubated with EE (1-10-50 μM) for 48 h. MRP3 protein and mRNA levels were measured by Western blotting and Real time PCR, respectively. EE up-regulated MRP3 protein and mRNA at 50 μM only in ER-α(+)-HepG2 cells. The in silico analysis of mrp3 promoter region demonstrated absence of estrogen response elements, but showed several Ap-1 binding sites. We further evaluated the potential involvement of the transcription factors c-JUN and c-FOS (members of Ap-1) in MRP3 up-regulation. ER-α(+) HepG2 cells were incubated with EE and c-FOS and c-JUN levels measured by Western blotting in nuclear extracts. EE up-regulated only c-JUN. Experiments of overexpression and knock-down of c-JUN by siRNA further demonstrated that this transcription factor is indeed implicated in MRP3 upregulation by EE. Co-immunoprecipitation assay demonstrated that EE induces c-JUN/ER-α interaction, and chromatin immunoprecipitation assay showed that this complex is recruited to the AP-1 binding consensus element present at the position (-1300/-1078 bp) of human mrp3 promoter. We conclude that EE induces MRP3 expression through ER-α, with recruitment of ER-α in complex with c-JUN to the human mrp3 promoter., (Copyright © 2013 Elsevier Inc. All rights reserved.)
- Published
- 2013
- Full Text
- View/download PDF
31. Induction of hepatic multidrug resistance-associated protein 3 by ethynylestradiol is independent of cholestasis and mediated by estrogen receptor.
- Author
-
Ruiz ML, Rigalli JP, Arias A, Villanueva S, Banchio C, Vore M, Mottino AD, and Catania VA
- Subjects
- Animals, Bile metabolism, Bile Acids and Salts metabolism, Cells, Cultured, Cholestasis genetics, Dactinomycin pharmacology, Estradiol analogs & derivatives, Estradiol pharmacology, Estrogen Antagonists pharmacology, Estrogen Receptor alpha antagonists & inhibitors, Estrogen Receptor alpha metabolism, Fulvestrant, Glutathione metabolism, Liver metabolism, Male, Multidrug Resistance-Associated Proteins genetics, Multidrug Resistance-Associated Proteins metabolism, Nucleic Acid Synthesis Inhibitors pharmacology, Phosphorylation, Primary Cell Culture, Proto-Oncogene Proteins c-jun metabolism, RNA, Messenger metabolism, Rats, Rats, Wistar, Real-Time Polymerase Chain Reaction, Time Factors, Up-Regulation, Cholestasis metabolism, Estrogen Receptor alpha agonists, Estrogens pharmacology, Ethinyl Estradiol pharmacology, Liver drug effects, Multidrug Resistance-Associated Proteins drug effects
- Abstract
Multidrug resistance-associated protein 3 (Mrp3; Abcc3) expression and activity are up-regulated in rat liver after in vivo repeated administration of ethynylestradiol (EE), a cholestatic synthetic estrogen, whereas multidrug resistance-associated protein 2 (Mrp2) is down-regulated. This study was undertaken to determine whether Mrp3 induction results from a direct effect of EE, independent of accumulation of any endogenous common Mrp2/Mrp3 substrates resulting from cholestasis and the potential mediation of estrogen receptor (ER). In in vivo studies, male rats were given a single, noncholestatic dose of EE (5 mg/kg s.c.), and basal bile flow and the biliary excretion rate of bile salts and glutathione were measured 5 hours later. This treatment increased Mrp3 mRNA by 4-fold, detected by real-time polymerase chain reaction, despite the absence of cholestasis. Primary culture of rat hepatocytes incubated with EE (1-10 µM) for 5 hours exhibited a 3-fold increase in Mrp3 mRNA (10 µM), consistent with in vivo findings. The increase in Mrp3 mRNA by EE was prevented by actinomycin D, indicating transcriptional regulation. When hepatocytes were incubated with an ER antagonist [7α,17β-[9-[(4,4,5,5,5-Pentafluoropentyl)sulfinyl]nonyl]estra-1,3,5(10)-triene-3,17-diol (ICI182/780), 1 µM], in addition to EE, induction of Mrp3 mRNA was abolished, implicating ER as a key mediator. EE induced an increase in ER-α phosphorylation at 30 minutes and expression of c-Jun, a well-known ER target gene, at 60 minutes, as detected by Western blotting of nuclear extracts. These increases were prevented by ICI182/780. In summary, EE increased the expression of hepatic Mrp3 transcriptionally and independently of any cholestatic manifestation and required participation of an ER, most likely ER-α, through its phosphorylation.
- Published
- 2013
- Full Text
- View/download PDF
32. Effect of glucagon-like peptide 2 on hepatic, renal, and intestinal disposition of 1-chloro-2,4-dinitrobenzene.
- Author
-
Villanueva SS, Perdomo VG, Ruiz ML, Rigalli JP, Arias A, Luquita MG, Vore M, Catania VA, and Mottino AD
- Subjects
- ATP-Binding Cassette Transporters genetics, ATP-Binding Cassette Transporters metabolism, Animals, Bile metabolism, Dinitrobenzenes metabolism, Dinitrochlorobenzene pharmacology, Down-Regulation drug effects, Female, Glutathione analogs & derivatives, Glutathione metabolism, Glutathione Transferase genetics, Glutathione Transferase metabolism, Isoenzymes genetics, Isoenzymes metabolism, Jejunum drug effects, Kidney drug effects, Liver drug effects, RNA, Messenger genetics, Rats, Rats, Wistar, Up-Regulation drug effects, gamma-Glutamyltransferase metabolism, Dinitrochlorobenzene pharmacokinetics, Glucagon-Like Peptide 2 pharmacology, Jejunum metabolism, Kidney metabolism, Liver metabolism
- Abstract
The ability of the liver, small intestine, and kidney to synthesize and subsequently eliminate dinitrophenyl-S-glutathione (DNP-SG), a substrate for multidrug resistance-associated protein 2 (Mrp2), was assessed in rats treated with glucagon-like peptide 2 (GLP-2, 12 μg/100 g b.wt. s.c. every 12 h for 5 consecutive days). An in vivo perfused jejunum model with simultaneous bile and urine collection was used. A single intravenous dose of 30 μmol/kg b.wt. 1-chloro-2,4-dinitrobenzene (CDNB) was administered, and its conjugate, DNP-SG, and dinitrophenyl cysteinyl glycine (DNP-CG), resulting from the action of γ-glutamyltransferase on DNP-SG, were determined in bile, intestinal perfusate, and urine by high-performance liquid chromatography. Tissue content of DNP-SG was also assessed in liver, intestine, and kidneys. Biliary excretion of DNP-SG+DNP-CG was decreased in GLP-2 rats with respect to controls. In contrast, their intestinal excretion was substantially increased, whereas urinary elimination was not affected. Western blot and real-time polymerase chain reaction studies revealed preserved levels of Mrp2 protein and mRNA in liver and renal cortex and a significant increase in intestine in response to GLP-2 treatment. Tissue content of DNP-SG detected 5 min after CDNB administration was decreased in liver, increased in intestine, and unchanged in kidney in GLP-2 versus control group, consistent with GLP-2-induced down-regulation of expression of glutathione transferase (GST) Mu in liver and up-regulation of GST-Alpha in intestine at both protein and mRNA levels. In conclusion, GLP-2 induced selective changes in hepatic and intestinal disposition of a common GST and Mrp2 substrate administered systemically that could be of pharmacological or toxicological relevance under therapeutic treatment conditions.
- Published
- 2012
- Full Text
- View/download PDF
33. Regulation of biotransformation systems and ABC transporters by benznidazole in HepG2 cells: involvement of pregnane X-receptor.
- Author
-
Rigalli JP, Perdomo VG, Luquita MG, Villanueva SS, Arias A, Theile D, Weiss J, Mottino AD, Ruiz ML, and Catania VA
- Subjects
- Biotransformation, Blotting, Western, Gene Expression Profiling, Hep G2 Cells, Humans, Metabolic Networks and Pathways genetics, Pregnane X Receptor, Real-Time Polymerase Chain Reaction, Up-Regulation, ATP Binding Cassette Transporter, Subfamily B, Member 1 biosynthesis, Antiprotozoal Agents metabolism, Hepatocytes drug effects, Hepatocytes metabolism, Nitroimidazoles metabolism, Receptors, Steroid metabolism
- Abstract
Background: Benznidazole (BZL) is the only antichagasic drug available in most endemic countries. Its effect on the expression and activity of drug-metabolizing and transporter proteins has not been studied yet., Methodology/principal Findings: Expression and activity of P-glycoprotein (P-gp), Multidrug resistance-associated protein 2 (MRP2), Cytochrome P450 3A4 (CYP3A4), and Glutathione S-transferase (GST) were evaluated in HepG2 cells after treatment with BZL. Expression was estimated by immunoblotting and real time PCR. P-gp and MRP2 activities were estimated using model substrates rhodamine 123 and dinitrophenyl-S-glutathione (DNP-SG), respectively. CYP3A4 and GST activities were evaluated through their abilities to convert proluciferin into luciferin and 1-chloro-2,4-dinitrobenzene into DNP-SG, respectively. BZL (200 µM) increased the expression (protein and mRNA) of P-gp, MRP2, CYP3A4, and GSTπ class. A concomitant enhancement of activity was observed for all these proteins, except for CYP3A4, which exhibited a decreased activity. To elucidate if pregnane X receptor (PXR) mediates BZL response, its expression was knocked down with a specific siRNA. In this condition, the effect of BZL on P-gp, MRP2, CYP3A4, and GSTπ protein up-regulation was completely abolished. Consistent with this, BZL was able to activate PXR, as detected by reporter gene assay. Additional studies, using transporter inhibitors and P-gp-knock down cells, demonstrated that P-gp is involved in BZL extrusion. Pre-treatment of HepG2 cells with BZL increased its own efflux, as a consequence of P-gp up-regulation., Conclusions/significance: Modifications in the activity of biotransformation and transport systems by BZL may alter the pharmacokinetics and efficiency of drugs that are substrates of these systems, including BZL itself.
- Published
- 2012
- Full Text
- View/download PDF
34. Localization status of hepatocellular transporters in cholestasis.
- Author
-
Crocenzi FA, Zucchetti AE, Boaglio AC, Barosso IR, Sanchez Pozzi EJ, Mottino AD, and Roma MG
- Subjects
- Bile physiology, Humans, Carrier Proteins physiology, Cholestasis
- Abstract
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.
- Published
- 2012
- Full Text
- View/download PDF
35. Pregnane X receptor mediates the induction of P-glycoprotein by spironolactone in HepG2 cells.
- Author
-
Rigalli JP, Ruiz ML, Perdomo VG, Villanueva SS, Mottino AD, and Catania VA
- Subjects
- ATP Binding Cassette Transporter, Subfamily B, Member 1 drug effects, ATP Binding Cassette Transporter, Subfamily B, Member 1 genetics, Blotting, Western, Dose-Response Relationship, Drug, Doxorubicin toxicity, Drug Interactions, Hep G2 Cells, Humans, Mineralocorticoid Receptor Antagonists administration & dosage, Polymerase Chain Reaction, Pregnane X Receptor, RNA, Messenger metabolism, Rhodamine 123 pharmacokinetics, Spironolactone administration & dosage, Verapamil pharmacology, ATP Binding Cassette Transporter, Subfamily B, Member 1 metabolism, Mineralocorticoid Receptor Antagonists pharmacology, Receptors, Steroid metabolism, Spironolactone pharmacology, Up-Regulation drug effects
- Abstract
We evaluated the effect of spironolactone (SL), a well-known inducer of biotransformation and elimination pathways, on the expression and activity of P-glycoprotein (P-gp/ABCB1/MDR1), a major xenobiotic transporter, in HepG2 cells, as well as the potential mediation of pregnane X nuclear receptor (PXR). Cells were exposed to SL (1, 5, 10, 20 or 50 μM) for 48 h. Expression of P-gp and its mRNA levels were estimated by Western blotting and real time PCR, respectively. P-gp activity was inversely correlated with the ability of the cells to accumulate the model substrate rhodamine 123 (Rh123, 5 μM), in the presence or absence of verapamil (50 μM), a P-gp inhibitor. At the highest dose of SL tested, P-gp and MDR1 mRNA levels were significantly increased (73 and 108%) with respect to control cells. Rh123 accumulation was concomitantly reduced and verapamil was able to abolish this effect, confirming P-gp participation. Additionally, we tested the cytotoxicity of doxorubicin, a model substrate of P-gp, under inducing conditions. HepG2 cells treated with SL exhibited higher viability, i.e. less doxorubicin toxicity, than control cells, consistent with P-gp up-regulation. When HepG2 cells were treated with SL in the presence of ketoconazole (KTZ), a non-specific nuclear receptor inhibitor, the up-regulation of P-gp was suppressed. To further identify the nuclear receptor involved, cells were transfected with a siRNA directed against human PXR, leading to a 74% decrease in PXR protein levels, which totally abolished SL induction of P-gp. We conclude that SL up-regulates P-gp expression, likely at transcriptional level, and its efflux activity in HepG2 cells. This effect is mediated by PXR. Thus, ligands of PXR such as SL may alter the disposition and toxicity of other xenobiotics, including drugs of therapeutic use, that are P-gp substrates., (Copyright © 2011 Elsevier Ireland Ltd. All rights reserved.)
- Published
- 2011
- Full Text
- View/download PDF
36. Acetaminophen-induced stimulation of MDR1 expression and activity in rat intestine and in LS 174T human intestinal cell line.
- Author
-
Ghanem CI, Arias A, Novak A, Carpini GD, Villanueva S, Blazquez AG, Marin JJ, Mottino AD, and Rubio MC
- Subjects
- ATP Binding Cassette Transporter, Subfamily B, Member 1 genetics, Acetaminophen administration & dosage, Analgesics, Non-Narcotic administration & dosage, Animals, Biological Transport, Cardiotonic Agents metabolism, Cell Line, Digoxin metabolism, Dose-Response Relationship, Drug, Humans, Male, Rats, Rats, Wistar, ATP Binding Cassette Transporter, Subfamily B, Member 1 metabolism, Acetaminophen pharmacology, Analgesics, Non-Narcotic pharmacology, Gene Expression Regulation drug effects, Intestines cytology
- Abstract
The well-known analgesic and antipyretic drug N-acetyl-p-aminophenol (acetaminophen; APAP) has been previously reported to affect MDR1 expression in rat liver. In this study, we have investigated the effect of subtoxic doses of APAP on MDR1 expression and activity in rat intestine and human intestinal cells. Administration of APAP at increasing doses of 0.2, 0.3, and 0.6g/kg b.w., i.p. over three consecutive days, induced MDR1 expression in rat duodenum (+240%) and ileum (+160%) as detected by western blotting. This was accompanied by preserved localization of the protein at the surface of the villus, as detected by confocal immunofluorescence microscopy. MDR1 activity was increased by 50% in APAP treated rats, as evaluated by serosal to mucosal secretion of rhodamine 123 in everted intestinal sacs. Treatment with APAP also decreased by 65% the portal vein concentrations of digoxin found in anesthetized rats after intraduodenal administration of this drug, which is consistent with an APAP-induced increased efficacy of intestinal barrier for digoxin net absorption. Exposure of LS 174T human colon adenocarcinoma cells to subtoxic APAP concentration (5mM) induced an increase in MDR1 mRNA expression (+46%), which was accompanied with an enhanced ability (+78%) to reduce intracellular content of rhodamine 123. Taken together these data suggest the existence of APAP-induced stimulation of MDR1 transcription in the intestinal epithelium. These findings are of clinical relevance, as co-administration of APAP with other MDR1 substrates could indirectly inhibit the net intestinal absorption of these drugs, leading to changes in their pharmacokinetics and therapeutic efficacy., (2010 Elsevier Inc. All rights reserved.)
- Published
- 2011
- Full Text
- View/download PDF
37. Tauroursodeoxycholate counteracts hepatocellular lysis induced by tensioactive bile salts by preventing plasma membrane-micelle transition.
- Author
-
Basiglio CL, Mottino AD, and Roma MG
- Subjects
- Animals, Cell Death drug effects, Cell Membrane chemistry, Detergents pharmacology, Digitonin pharmacology, Lipid Bilayers chemistry, Male, Osmosis drug effects, Phase Transition drug effects, Polyethylene Glycols pharmacology, Rats, Solubility drug effects, Cell Membrane drug effects, Hepatocytes cytology, Hepatocytes drug effects, Micelles, Taurochenodeoxycholic Acid antagonists & inhibitors, Taurochenodeoxycholic Acid pharmacology
- Abstract
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., (Copyright © 2010 Elsevier Ireland Ltd. All rights reserved.)
- Published
- 2010
- Full Text
- View/download PDF
38. Induction of intestinal multidrug resistance-associated protein 2 by glucagon-like Peptide 2 in the rat.
- Author
-
Villanueva SS, Arias A, Ruiz ML, Rigalli JP, Pellegrino JM, Vore M, Catania VA, and Mottino AD
- Subjects
- Adenylyl Cyclase Inhibitors, Animals, Blotting, Western, Chromatography, High Pressure Liquid, Cyclic AMP metabolism, Dideoxyadenosine pharmacology, Enterocytes drug effects, Enterocytes enzymology, Enterocytes metabolism, Enterocytes pathology, Female, Fluorescent Antibody Technique, Glucagon-Like Peptide 2 physiology, Glutathione Transferase biosynthesis, Intestinal Mucosa enzymology, Intestinal Mucosa metabolism, Intestinal Mucosa pathology, Jejunum enzymology, Jejunum metabolism, Jejunum pathology, Lactation metabolism, Rats, Rats, Wistar, Reverse Transcriptase Polymerase Chain Reaction, ATP-Binding Cassette Transporters biosynthesis, Glucagon-Like Peptide 2 pharmacology, Intestinal Mucosa drug effects, Jejunum drug effects
- Abstract
The effects of glucagon-like peptide 2 (GLP-2) on expression and activity of jejunal multidrug resistance-associated protein 2 (Mrp2; Abcc2) and glutathione transferase (GST) were evaluated. After GLP-2 treatment (12 μg/100 g b.wt. s.c., every 12 h, for 5 consecutive days), Mrp2 and the α class of GST proteins and their corresponding mRNAs were increased, suggesting a transcriptional regulation. Mrp2 was localized at the apical membrane of the enterocyte in control and GLP-2 groups, as detected by confocal immunofluorescence microscopy. As a functional assay, everted intestinal sacs were incubated in the presence of 1-chloro-2,4-dinitrobenzene in the mucosal compartment, and the glutathione-conjugated derivative, dinitrophenyl-S-glutathione (DNP-SG; model Mrp2 substrate), was detected in the same compartment by high-performance liquid chromatography. A significant increase in apical secretion of DNP-SG was detected in the GLP-2 group, consistent with simultaneous up-regulation of Mrp2 and GST. GLP-2 also promoted an increase in cAMP levels as detected in homogenates of intestinal mucosa. Treatment of rats with 2',3'-dideoxyadenosine (DDA), a specific inhibitor of adenylyl cyclase, abolished the increase in cAMP levels and Mrp2 protein promoted by GLP-2, suggesting cAMP as a mediator of Mrp2 modulation. Increased expression of Mrp2 and cAMP levels in response to GLP-2 occurred not only at the tip but also at the middle region of the villus, where constitutive expression of Mrp2 is normally low. In conclusion, our study suggests a role for GLP-2 in the prevention of cell toxicity of the intestinal mucosa by increasing Mrp2 chemical barrier function.
- Published
- 2010
- Full Text
- View/download PDF
39. Phosphoinositide 3-kinase/protein kinase B signaling pathway is involved in estradiol 17β-D-glucuronide-induced cholestasis: complementarity with classical protein kinase C.
- Author
-
Boaglio AC, Zucchetti AE, Sánchez Pozzi EJ, Pellegrino JM, Ochoa JE, Mottino AD, Vore M, Crocenzi FA, and Roma MG
- Subjects
- ATP Binding Cassette Transporter, Subfamily B, Member 11, ATP-Binding Cassette Transporters metabolism, Androstadienes pharmacology, Animals, Bile Canaliculi drug effects, Bile Canaliculi physiology, Biliary Tract metabolism, Carbazoles pharmacology, Colchicine pharmacology, Endocytosis drug effects, Estradiol analogs & derivatives, Glutathione metabolism, In Vitro Techniques, Male, Microtubules drug effects, Microtubules physiology, Multidrug Resistance-Associated Protein 2, Multidrug Resistance-Associated Proteins metabolism, Perfusion, Protein Kinase C antagonists & inhibitors, Rats, Rats, Wistar, Signal Transduction, Taurocholic Acid metabolism, Wortmannin, 1-Phosphatidylinositol 4-Kinase physiology, Cholestasis chemically induced, Protein Kinase C physiology, Proto-Oncogene Proteins c-akt physiology
- Abstract
Unlabelled: Estradiol 17β-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.
- Published
- 2010
- Full Text
- View/download PDF
40. Induction of intestinal multidrug resistance-associated protein 2 (Mrp2) by spironolactone in rats.
- Author
-
Ruiz ML, Villanueva SS, Luquita MG, Pellegrino JM, Rigalli JP, Arias A, Sánchez Pozzi EJ, Mottino AD, and Catania VA
- Subjects
- Animals, Biological Transport drug effects, Cell Polarity, Dinitrochlorobenzene metabolism, Dinitrochlorobenzene pharmacokinetics, Glutathione analogs & derivatives, Glutathione analysis, Intestine, Small metabolism, Male, Microvilli metabolism, Microvilli ultrastructure, Multidrug Resistance-Associated Protein 2, Multidrug Resistance-Associated Proteins metabolism, Organ Specificity, Polymerase Chain Reaction, Pregnane X Receptor, RNA, Messenger metabolism, Rats, Rats, Wistar, Receptors, Steroid antagonists & inhibitors, Gene Expression Regulation drug effects, Intestine, Small drug effects, Multidrug Resistance-Associated Proteins genetics, Spironolactone pharmacology
- Abstract
The effect of spironolactone (SL) pretreatment (200micromol/kg b.w./day, 3 consecutive days) on intestinal multidrug resistance-associated protein 2 (Mrp2) was evaluated in rats. A significant increase in protein levels in upper regions of small intestine, where Mrp2 is mainly present, was detected by western blotting. Real time PCR studies suggest a transcriptional regulation. The administration of ketoconazole, a pregnane X receptor (PXR) antagonist, was able to prevent the increase in Mrp2 mRNA levels induced by SL. The serosal to mucosal transport of dinitrophenyl S-glutathione, a model substrate of Mrp2 was evaluated in jejunal sac model. The data indicate that SL increased Mrp2 activity, well correlating with its up-regulation. We conclude that SL is able to induce intestinal Mrp2 transcriptionally, PXR being a potential mediator. We propose that SL could be of potential therapeutic application particularly in situations of down-regulation of intestinal Mrp2.
- Published
- 2009
- Full Text
- View/download PDF
41. Complement activation and disease: protective effects of hyperbilirubinaemia.
- Author
-
Basiglio CL, Arriaga SM, Pelusa F, Almará AM, Kapitulnik J, and Mottino AD
- Subjects
- Antioxidants pharmacology, Bilirubin pharmacology, Bilirubin physiology, Complement C1q metabolism, Complement Inactivating Agents pharmacology, Complement Pathway, Classical drug effects, Cytoprotection physiology, Humans, Inflammation immunology, Oxidative Stress immunology, Complement Pathway, Classical immunology, Hyperbilirubinemia immunology, Inflammation prevention & control
- Abstract
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.
- Published
- 2009
- Full Text
- View/download PDF
42. Unconjugated bilirubin inhibits C1 esterase activity.
- Author
-
Arriaga SM, Basiglio CL, Mottino AD, and Almará AM
- Subjects
- Enzyme Activation drug effects, Humans, Immunoblotting, Bilirubin chemistry, Bilirubin pharmacology, Complement C1s metabolism
- Abstract
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.
- Published
- 2009
- Full Text
- View/download PDF
43. Regulation of expression and activity of rat intestinal multidrug resistance-associated protein 2 by cholestatic estrogens.
- Author
-
Arias A, Villanueva SS, Ruiz ML, Luquita MG, Veggi LM, Pellegrino JM, Vore M, Catania VA, and Mottino AD
- Subjects
- ATP-Binding Cassette Transporters genetics, ATP-Binding Cassette Transporters metabolism, Animals, Biological Transport, Active drug effects, Biomarkers metabolism, Body Weight drug effects, Down-Regulation drug effects, Estradiol analogs & derivatives, Estradiol pharmacology, Intestinal Mucosa metabolism, Male, Multidrug Resistance-Associated Protein 2, Multidrug Resistance-Associated Proteins metabolism, Rats, Rats, Wistar, Cholestasis metabolism, Estrogens pharmacology, Gene Expression drug effects, Multidrug Resistance-Associated Proteins genetics
- Abstract
The effect of the cholestatic estrogens ethynylestradiol (EE) and estradiol 17beta-D-glucuronide (E2-17G) on expression and activity of intestinal multidrug resistant-associated protein 2 (Mrp2, Abcc2) was studied in rats. Expression and localization of Mrp2 were evaluated by Western blotting, real-time polymerase chain reaction, and confocal immunofluorescence microscopy. Mrp2 transport activity toward dinitrophenyl-S-glutathione (DNP-SG) was assessed in vitro in intestinal sacs. EE, administered subcutaneously at a 5 mg/kg b.wt. dose, for 5 consecutive days, produced a marked decrease in Mrp2 expression at post-transcriptional level, without affecting its normal localization at the apical membrane of the enterocyte. This effect was selective because expression of other ATP-binding cassette proteins such as breast cancer resistance protein and Mrp3 were not affected and that of multidrug resistance protein 1 was only minimally impaired. Consistent with down-regulation of expression of Mrp2, a significant impairment in serosal to mucosal transport of DNP-SG and in protection against absorption of this same compound were registered. Simultaneous administration of EE with spironolactone (200 micromol/kg b.wt./day for 3 days), an Mrp2 inducer, prevented these alterations, confirming down-regulation of expression of Mrp2 by EE as a major component of functional changes. Incorporation of E2-17G (30 microM) in the serosal medium of intestinal sacs decreased serosal to mucosal transport of DNP-SG, probably because of competitive inhibition, without affecting normal Mrp2 expression or localization. Our data indicate impairment of function of intestinal Mrp2 by both cholestatic estrogens, although through a different mechanism. This finding represents an aggravation of deteriorated hepatic Mrp2 function that could further increase bioavailability of specific xenobiotics after oral exposure.
- Published
- 2009
- Full Text
- View/download PDF
44. Differential effects of silymarin and its active component silibinin on plasma membrane stability and hepatocellular lysis.
- Author
-
Basiglio CL, Sánchez Pozzi EJ, Mottino AD, and Roma MG
- Subjects
- Animals, Cell Membrane metabolism, Dose-Response Relationship, Drug, Drug Evaluation, Preclinical, Hepatocytes metabolism, Male, Octoxynol toxicity, Rats, Rats, Wistar, Silybin, Taurochenodeoxycholic Acid toxicity, Cell Membrane drug effects, Hepatocytes cytology, Hepatocytes drug effects, Silymarin pharmacology
- Abstract
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.
- Published
- 2009
- Full Text
- View/download PDF
45. Effect of repeated administration with subtoxic doses of acetaminophen to rats on enterohepatic recirculation of a subsequent toxic dose.
- Author
-
Ghanem CI, Ruiz ML, Villanueva SS, Luquita M, Llesuy S, Catania VA, Bengochea LA, and Mottino AD
- Subjects
- ATP-Binding Cassette Transporters biosynthesis, Acetaminophen administration & dosage, Acetaminophen pharmacokinetics, Acetaminophen toxicity, Analgesics, Non-Narcotic administration & dosage, Animals, Blotting, Western, Charcoal administration & dosage, Dose-Response Relationship, Drug, Drug Administration Schedule, Glutathione metabolism, Injections, Intraperitoneal, Liver metabolism, Liver pathology, Male, Microscopy, Fluorescence, Multidrug Resistance-Associated Proteins biosynthesis, Oxidative Stress drug effects, Rats, Rats, Wistar, Acetaminophen analogs & derivatives, Analgesics, Non-Narcotic pharmacokinetics, Analgesics, Non-Narcotic toxicity, Liver drug effects
- Abstract
Development of resistance to toxic effects of acetaminophen (APAP) was reported in rodents and humans, though the mechanism is only partially understood. We examined in rats the effect of administration with subtoxic daily doses (0.2, 0.3, and 0.6g/kg, i.p.) of APAP on enterohepatic recirculation and liver toxicity of a subsequent i.p. toxic dose of 1g/kg, given 24h after APAP pre-treatment. APAP and its major metabolite APAP-glucuronide (APAP-Glu) were determined in bile, urine, serum and liver homogenate. APAP pre-treatment was not toxic, as determined by serum markers of liver damage and neither induced oxidative stress as demonstrated by assessment of ROS generation in liver or glutathione species in liver and bile. APAP pre-treatment induced a partial shift from biliary to urinary elimination of APAP-Glu after administration with the toxic dose, and decreased hepatic content and increased serum content of this conjugate, consistent with a marked up-regulation of its basolateral transporter Mrp3 relative to apical Mrp2. Preferential secretion of APAP-glu into blood decreased enterohepatic recirculation of APAP, thus attenuating liver exposition to the intact drug, as demonstrated 6h after administration with the toxic dose. The beneficial effect of interfering the enterohepatic recirculation was alternatively tested in animals receiving activated charcoal by gavage to adsorb APAP of biliary origin. The data indicated decreased liver APAP content and glutathione consumption. We conclude that selective up-regulation of Mrp3 expression by APAP pre-treatment may contribute to development of resistance to APAP hepatotoxicity, at least in part by decreasing its enterohepatic recirculation.
- Published
- 2009
- Full Text
- View/download PDF
46. Hepatic drug transporters and nuclear receptors: regulation by therapeutic agents.
- Author
-
Mottino AD and Catania VA
- Subjects
- ATP Binding Cassette Transporter, Subfamily B metabolism, Acetaminophen pharmacology, Acetaminophen therapeutic use, Glucocorticoids pharmacology, Glucocorticoids therapeutic use, Humans, Multidrug Resistance-Associated Protein 2, Receptors, Cytoplasmic and Nuclear metabolism, Steroids pharmacology, Steroids therapeutic use, ATP Binding Cassette Transporter, Subfamily B drug effects, Liver Diseases drug therapy, Receptors, Cytoplasmic and Nuclear drug effects
- Abstract
The canalicular membrane represents the excretory pole of hepatocytes. Bile is an important route of elimination of potentially toxic endo- and xenobiotics (including drugs and toxins), mediated by the major canalicular transporters: multidrug resistance protein 1 (MDR1, ABCB1), also known as P-glycoprotein, multidrug resistance-associated protein 2 (MRP2, ABCC2), and the breast cancer resistance protein (BCRP, ABCG2). Their activities depend on regulation of expression and proper localization at the canalicular membrane, as regulated by transcriptional and post-transcriptional events, respectively. At transcriptional level, specific nuclear receptors (NR)s modulated by ligands, co-activators and co-repressors, mediate the physiological requirements of these transporters. This complex system is also responsible for alterations occurring in specific liver pathologies. We briefly describe the major Class II NRs, pregnane X receptor (PXR) and constitutive androstane receptor (CAR), and their role in regulating expression of multidrug resistance proteins. Several therapeutic agents regulate the expression of relevant drug transporters through activation/inactivation of these NRs. We provide some representative examples of the action of therapeutic agents modulating liver drug transporters, which in addition, involve CAR or PXR as mediators.
- Published
- 2008
- Full Text
- View/download PDF
47. Ca(2+)-dependent protein kinase C isoforms are critical to estradiol 17beta-D-glucuronide-induced cholestasis in the rat.
- Author
-
Crocenzi FA, Sánchez Pozzi EJ, Ruiz ML, Zucchetti AE, Roma MG, Mottino AD, and Vore M
- Subjects
- ATP Binding Cassette Transporter, Subfamily B, Member 11, ATP-Binding Cassette Transporters metabolism, Animals, Carbazoles pharmacology, Cells, Cultured, Disease Models, Animal, Dose-Response Relationship, Drug, Enzyme Inhibitors pharmacology, Estradiol adverse effects, Estradiol pharmacology, Female, Hepatocytes drug effects, Hepatocytes metabolism, Liver drug effects, Liver metabolism, Protein Kinase C-alpha antagonists & inhibitors, Protein Kinase C-epsilon antagonists & inhibitors, Rats, Rats, Sprague-Dawley, Calcium metabolism, Cholestasis chemically induced, Cholestasis metabolism, Estradiol analogs & derivatives, Protein Kinase C-alpha metabolism, Protein Kinase C-epsilon metabolism
- Abstract
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.
- Published
- 2008
- Full Text
- View/download PDF
48. Dynamic localization of hepatocellular transporters in health and disease.
- Author
-
Roma MG, Crocenzi FA, and Mottino AD
- Subjects
- Animals, Bile metabolism, Cell Membrane metabolism, Cholagogues and Choleretics therapeutic use, Cholestasis drug therapy, Disease Models, Animal, Endocytosis, Endoplasmic Reticulum, Rough metabolism, Endosomes metabolism, Exocytosis, Humans, Liver drug effects, Membrane Transport Proteins drug effects, Protein Transport, Signal Transduction, Xenobiotics metabolism, Cholestasis metabolism, Liver metabolism, Membrane Transport Proteins metabolism
- Abstract
Vesicle-based trafficking of hepatocellular transporters involves delivery of the newly-synthesized carriers from the rough endoplasmic reticulum to either the plasma membrane domain or to an endosomal, submembrane compartment, followed by exocytic targeting to the plasma membrane. Once delivered to the plasma membrane, the transporters usually undergo recycling between the plasma membrane and the endosomal compartment, which usually serves as a reservoir of pre-existing transporters available on demand. The balance between exocytic targeting and endocytic internalization from/to this recycling compartment is therefore a chief determinant of the overall capability of the liver epithelium to secrete bile and to detoxify endo and xenobiotics. Hence, it is a highly regulated process. Impaired regulation of this balance may lead to abnormal localization of these transporters, which results in bile secretory failure due to endocytic internalization of key transporters involved in bile formation. This occurs in several experimental models of hepatocellular cholestasis, and in most human cholestatic liver diseases. This review describes the molecular bases involved in the biology of the dynamic localization of hepatocellular transporters and its regulation, with a focus on the involvement of signaling pathways in this process. Their alterations in different experimental models of cholestasis and in human cholestatic liver disease are reviewed. In addition, the causes explaining the pathological condition (e.g. disorganization of actin or actin-transporter linkers) and the mediators involved (e.g. activation of cholestatic signaling transduction pathways) are also discussed. Finally, several experimental therapeutic approaches based upon the administration of compounds known to stimulate exocytic insertion of canalicular transporters (e.g. cAMP, tauroursodeoxycholate) are described.
- Published
- 2008
- Full Text
- View/download PDF
49. Dapsone induces oxidative stress and impairs antioxidant defenses in rat liver.
- Author
-
Veggi LM, Pretto L, Ochoa EJ, Catania VA, Luquita MG, Taborda DR, Sánchez Pozzi EJ, Ikushiro S, Coleman MD, Roma MG, and Mottino AD
- Subjects
- Animals, Female, Glucuronosyltransferase metabolism, Glutathione metabolism, Glutathione Transferase metabolism, Hepatocytes drug effects, Hepatocytes metabolism, Lipid Peroxidation drug effects, Liver metabolism, Male, Rats, Rats, Wistar, Reactive Oxygen Species metabolism, Dapsone pharmacology, Liver drug effects, Oxidative Stress drug effects
- Abstract
Dapsone (DDS) is currently used in the treatment of leprosy, malaria and in infections with Pneumocystis jirovecii and Toxoplasma gondii in AIDS patients. Adverse effects of DDS involve methemoglobinemia and hemolysis and, to a lower extent, liver damage, though the mechanism is poorly characterized. We evaluated the effect of DDS administration to male and female rats (30 mg/kg body wt, twice a day, for 4 days) on liver oxidative stress through assessment of biliary output and liver content of reduced (GSH) and oxidized (GSSG) glutathione, lipid peroxidation, and expression/activities of the main antioxidant enzymes glutathione peroxidase, superoxide dismutase, catalase and glutathione S-transferase. The influence of DDS treatment on expression/activity of the main DDS phase-II-metabolizing system, UDP-glucuronosyltransferase (UGT), was additionally evaluated. The involvement of dapsone hydroxylamine (DDS-NHOH) generation in these processes was estimated by comparing the data in male and female rats since N-hydroxylation of DDS mainly occurs in males. Our studies revealed an increase in the GSSG/GSH biliary output ratio, a sensitive indicator of oxidative stress, and in lipid peroxidation, in male but not in female rats treated with DDS. The activity of all antioxidant enzymes was significantly impaired by DDS treatment also in male rats, whereas UGT activity was not affected in any sex. Taken together, the evidence indicates that DDS induces oxidative stress in rat liver and that N-hydroxylation of DDS was the likely mediator. Impairment in the activity of enzymatic antioxidant systems, also associated with DDS-NHOH formation, constituted a key aggravating factor.
- Published
- 2008
- Full Text
- View/download PDF
50. Hepatic synthesis and urinary elimination of acetaminophen glucuronide are exacerbated in bile duct-ligated rats.
- Author
-
Villanueva SS, Ruiz ML, Ghanem CI, Luquita MG, Catania VA, and Mottino AD
- Subjects
- ATP-Binding Cassette Transporters metabolism, Acetaminophen administration & dosage, Acetaminophen urine, Animals, Bile Ducts surgery, Dose-Response Relationship, Drug, Intestinal Mucosa metabolism, Kidney metabolism, Ligation, Male, Multidrug Resistance-Associated Proteins metabolism, Rats, Rats, Wistar, Acetaminophen analogs & derivatives, Acetaminophen metabolism, Liver metabolism
- Abstract
Renal and intestinal disposition of acetaminophen glucuronide (APAP-GLU), a common substrate for multidrug resistance-associated proteins 2 and 3 (Mrp2 and Mrp3), was assessed in bile duct-ligated rats (BDL) 7 days after surgery using an in vivo perfused jejunum model with simultaneous urine collection. Doses of 150 mg/kg b.w. (i.v.) or 1 g/kg b.w. (i.p.) of acetaminophen (APAP) were administered, and its glucuronide was determined in bile (only Shams), urine, and intestinal perfusate throughout a 150-min period. Intestinal excretion of APAP-GLU was unchanged or decreased (-58%) by BDL for the 150 mg and 1 g/kg b.w. doses of APAP, respectively. In contrast, renal excretion was increased by 200 and 320%, respectively. Western studies revealed decreased levels of apical Mrp2 in liver and jejunum but increased levels in renal cortex from BDL animals, whereas Mrp3 was substantially increased in liver and not affected in kidney or intestine. The global synthesis of APAP-GLU, determined as the sum of cumulative excretions, was higher in BDL rats (+51 and +110%) for these same doses of APAP as a consequence of a significant increase in functional liver mass, with no changes in specific glucuronidating activity. Expression of apical breast cancer resistance protein, which also transports nontoxic metabolites of APAP, was decreased by BDL in liver and renal cortex, suggesting a minor participation of this route. We demonstrate a more efficient hepatic synthesis and basolateral excretion of APAP-GLU followed by its urinary elimination in BDL group, the latter two processes consistent with up-regulation of liver Mrp3 and renal Mrp2.
- Published
- 2008
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.