Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters











Type of study
Publication year range
2.
Hepatology ; 2024 Jun 20.
Article in English | MEDLINE | ID: mdl-38900412

ABSTRACT

Porto-sinusoidal vascular disorder (PSVD) is a rare liver disease. The pathophysiological mechanisms underlying the development of PSVD are unknown. Isolated cases of PSVD associated with gene mutations have been reported, but no overview is available. Therefore, we performed an extensive literature search to provide a comprehensive overview of gene mutations associated with PSVD. We identified 34 genes and 1 chromosomal abnormality associated with PSVD in the literature, and we describe here 1 additional gene mutation ( TBL1XR1 mutation, leading to Pierpont syndrome). These gene mutations are associated either with extrahepatic organ involvement as part of syndromes (Adams-Oliver, telomere biology disorders, retinal vasculopathy with cerebral leukoencephalopathy and systemic manifestations, immune deficiencies, cystic fibrosis, cystinosis, Williams-Beuren, Turner, Pierpont) or with isolated PSVD ( KCNN3 , DGUOK , FOPV , GIMAP5 , FCHSD1 , TRMT5 , HRG gene mutations). Most of the cases were revealed by signs or complications of portal hypertension. When analyzing the cell types in which these genes are expressed, we found that these genes are predominantly expressed in immune cells, suggesting that these cells may play a more important role in the development of PSVD than previously thought. In addition, pathway analyses suggested that there may be 2 types of PSVD associated with gene mutations: those resulting directly from morphogenetic abnormalities and those secondary to immune changes.

3.
Biochimie ; 223: 41-53, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38608750

ABSTRACT

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


Subject(s)
Estradiol , Hepatocytes , NADPH Oxidases , Reactive Oxygen Species , Animals , NADPH Oxidases/metabolism , Reactive Oxygen Species/metabolism , Rats , Hepatocytes/metabolism , Hepatocytes/drug effects , Estradiol/pharmacology , Estradiol/metabolism , Estradiol/analogs & derivatives , Female , Cholestasis/chemically induced , Cholestasis/metabolism , Cholestasis/pathology , Rats, Wistar , Acetophenones/pharmacology , Oxidative Stress/drug effects , Acetylcysteine/pharmacology , p38 Mitogen-Activated Protein Kinases/metabolism , Multidrug Resistance-Associated Proteins/metabolism , MAP Kinase Signaling System/drug effects , Cells, Cultured , Antioxidants/pharmacology , Antioxidants/metabolism , Cholestasis, Intrahepatic , Pregnancy Complications , ATP-Binding Cassette Transporters
4.
Toxicol Appl Pharmacol ; 426: 115636, 2021 09 01.
Article in English | MEDLINE | ID: mdl-34214573

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.


Subject(s)
Chemical and Drug Induced Liver Injury/drug therapy , Genistein/therapeutic use , Protective Agents/therapeutic use , 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
5.
Life Sci ; 259: 118352, 2020 Oct 15.
Article in English | MEDLINE | ID: mdl-32860804

ABSTRACT

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


Subject(s)
ATP-Binding Cassette Transporters/metabolism , Cholestasis/drug therapy , Spironolactone/therapeutic use , ATP Binding Cassette Transporter, Subfamily B, Member 11/metabolism , Animals , Bile/metabolism , Cholestasis/blood , Cholestasis/metabolism , Cytokines/blood , Gene Expression Regulation/drug effects , Lipopolysaccharides/adverse effects , Male , Rats , Rats, Wistar , Real-Time Polymerase Chain Reaction
6.
Arch Toxicol ; 93(8): 2279-2294, 2019 08.
Article in English | MEDLINE | ID: mdl-31300867

ABSTRACT

Taurolithocholate (TLC) is a cholestatic bile salt that induces disinsertion of the canalicular transporter Abcc2 (Mrp2, multidrug resistance-associated protein 2). This internalization is mediated by different intracellular signaling proteins such as PI3K, PKCε and MARCK but the initial receptor of TLC remains unknown. A few G protein-coupled receptors interact with bile salts in hepatocytes. Among them, sphingosine-1 phosphate receptor 2 (S1PR2) represents a potential initial receptor for TLC. The aim of this study was to evaluate the role of this receptor and its downstream effectors in the impairment of Abcc2 function induced by TLC. In vitro, S1PR2 inhibition by JTE-013 or its knockdown by small interfering RNA partially prevented the decrease in Abcc2 activity induced by TLC. Moreover, adenylyl cyclase (AC)/PKA and PI3K/Akt inhibition partially prevented TLC effect on canalicular transporter function. TLC produced PKA and Akt activation, which were blocked by JTE-013 and AC inhibitors, connecting S1PR2/AC/PKA and PI3K/Akt in a same pathway. In isolated perfused rat liver, injection of TLC triggered endocytosis of Abcc2 that was accompanied by a sustained decrease in the bile flow and the biliary excretion of the Abcc2 substrate dinitrophenyl-glutathione until the end of the perfusion period. S1PR2 or AC inhibition did not prevent the initial decay, but they accelerated the recovery of these parameters and the reinsertion of Abcc2 into the canalicular membrane. In conclusion, S1PR2 and the subsequent activation of AC, PKA, PI3K and Akt is partially responsible for the cholestatic effects of TLC through sustained internalization of Abcc2.


Subject(s)
ATP-Binding Cassette Transporters/metabolism , Adenylyl Cyclases/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Sphingosine-1-Phosphate Receptors/metabolism , Taurolithocholic Acid/pharmacology , Animals , Cells, Cultured , Female , Hepatocytes/drug effects , Hepatocytes/metabolism , Liver/drug effects , Liver/metabolism , Metabolic Networks and Pathways/drug effects , Organ Culture Techniques , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Pyrazoles/pharmacology , Pyridines/pharmacology , Rats, Wistar , Sphingosine-1-Phosphate Receptors/antagonists & inhibitors , Sphingosine-1-Phosphate Receptors/genetics , Taurolithocholic Acid/metabolism
7.
Biochem Pharmacol ; 168: 48-56, 2019 10.
Article in English | MEDLINE | ID: mdl-31202734

ABSTRACT

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


Subject(s)
Cholagogues and Choleretics/therapeutic use , Cholestasis/chemically induced , Cholestasis/drug therapy , Lipopolysaccharides/pharmacology , Ursodeoxycholic Acid/therapeutic use , ATP Binding Cassette Transporter, Subfamily B, Member 11/metabolism , ATP-Binding Cassette Transporters/metabolism , Alkaline Phosphatase/blood , Animals , Bile Acids and Salts/metabolism , Cholagogues and Choleretics/administration & dosage , Cholagogues and Choleretics/pharmacology , Disease Models, Animal , Hepatocytes/drug effects , Hepatocytes/metabolism , Liver/metabolism , Male , Rats , Rats, Wistar , Treatment Outcome , Ursodeoxycholic Acid/administration & dosage , Ursodeoxycholic Acid/pharmacology
8.
Biochem Pharmacol ; 164: 311-320, 2019 06.
Article in English | MEDLINE | ID: mdl-31026445

ABSTRACT

TNFα is a cytokine whose levels are increased in inflammatory pathologies that are associated with cholestasis. Endocytic internalization of Abcc2 (multidrug resistance-associated protein 2), a canalicular transporter of organic anions that is implicated in the clearance of clinically important drugs, is a phenomenon that occurs in inflammatory liver diseases, and it has been established that cytokines act as mediators. However, the intracellular mechanism involved in this effect remains unknown. The aim of the present work was to characterize the internalization of Abcc2 induced by TNFα and to study the role of ERK1/2 and reactive oxygen species as signaling mediators of transporter internalization. Using rat hepatocyte couplets, we found that TNFα (6.25 pg/ml) induced a decrease in Abcc2 activity estimated by the accumulation of the Abcc2 substrate glutathione methylfluorescein in the canalicular vacuole that was accompanied by internalization of Abcc2 from the canalicular membrane. Inhibition of MEK1/2 (upstream of ERK1/2) partially prevented TNFα effects on Abcc2 internalization and activity impairment. Reactive oxygen species (ROS) scavengers such as vitamin C and mannitol partially prevented both TNFα-induced decrease in Abcc2 activity and ERK1/2 phosphorylation. Apocynin, a NADPH oxidase inhibitor, prevented the increase in ROS and the phosphorylation of ERK1/2 produced by TNFα. Taken together, these results indicate that TNFα activates a pathway involving NADPH oxidase, ROS and MEK1/2-ERK1/2 that is partially responsible for the internalization of Abcc2. This internalization leads to an altered transport activity of Abcc2 that could impair drug disposal, enhancing drug toxicity in patients suffering from inflammatory liver diseases.


Subject(s)
ATP-Binding Cassette Transporters/metabolism , Hepatocytes/metabolism , MAP Kinase Signaling System/physiology , Tumor Necrosis Factor-alpha/pharmacology , Animals , Dose-Response Relationship, Drug , Female , Hepatocytes/drug effects , MAP Kinase Signaling System/drug effects , Rats , Rats, Wistar
9.
Toxicol In Vitro ; 58: 51-59, 2019 Aug.
Article in English | MEDLINE | ID: mdl-30876886

ABSTRACT

Many drugs can induce liver injury, characterized by hepatocellular, cholestatic or mixed hepatocellular-cholestatic lesions. While an inflammatory stress is known to aggravate hepatocellular injury caused by some drugs much less evidence exists for cholestatic features. In this study, the influence of pro-inflammatory cytokines (IL-6, IL-1ß and TNF-α), either individually or combined, on cytotoxic and cholestatic properties of antibiotics was evaluated using differentiated HepaRG cells. Six antibiotics of various chemical structures and known to cause cholestasis and/or hepatocellular injury in clinic were investigated. Caspase-3 activity was increased with all these tested hepatotoxic drugs and except with erythromycin, was further augmented in presence of cytokines mainly when these were co-added as a mixture. TNF-α and IL-1ß aggravated cytotoxicity of TVX more than IL-6. Bile canaliculi (BC) dilatation induced by cholestatic drugs was increased by co-treatment with IL-6 and IL-1ß but not with TNF-α. Reduced accumulation of carboxy-dichlorofluorescein, a substrate of the multi-drug resistance-associated protein 2, in antibiotic-induced dilatated BC, was further extended in presence of individual or mixed cytokines. In conclusion, our data demonstrate that pro-inflammatory cytokines either individually or in mixture, can modulate cholestatic and/or cytotoxic responses to antibiotics and that the extent of these effects is dependent on the cytokine and the cholestatic antibiotic.


Subject(s)
Anti-Bacterial Agents/adverse effects , Bile Canaliculi/drug effects , Cholestasis/chemically induced , Cytokines/pharmacology , Bile Canaliculi/physiology , C-Reactive Protein/metabolism , Caspase 3/metabolism , Cell Line , Cell Survival/drug effects , Cholestasis/metabolism , Fluoresceins/metabolism , Humans
10.
Arch Toxicol ; 92(2): 729-744, 2018 Feb.
Article in English | MEDLINE | ID: mdl-29090346

ABSTRACT

Estradiol-17ß-D-glucuronide (E17G), through the activation of different signaling proteins, induces acute endocytic internalization of canalicular transporters in rat, including multidrug resistance-associated protein 2 (Abcc2) and bile salt export pump (Abcb11), generating cholestasis. Insulin-like growth factor 1 receptor (IGF-1R) is a membrane-bound tyrosine kinase receptor that can potentially interact with proteins activated by E17G. The aim of this study was to analyze the potential role of IGF-1R in the effects of E17G in isolated perfused rat liver (IPRL) and isolated rat hepatocyte couplets. In vitro, IGF-1R inhibition by tyrphostin AG1024 (TYR, 100 nM), or its knock-down with siRNA, strongly prevented E17G-induced impairment of Abcc2 and Abcb11 function and localization. The protection by TYR was not additive to that produced by wortmannin (PI3K inhibitor, 100 nM), and both protections share the same dependency on microtubule integrity, suggesting that IGF-1R shared the signaling pathway of PI3K/Akt. Further analysis of the activation of Akt and IGF-1R induced by E17G indicated a sequence of activation GPR30-IGF-1R-PI3K/Akt. In IPRL, an intraportal injection of E17G triggered endocytosis of Abcc2 and Abcb11, and this was accompanied by a sustained decrease in the bile flow and the biliary excretion of Abcc2 and Abcb11 substrates. TYR did not prevent the initial decay, but it greatly accelerated the recovery to normality of these parameters and the reinsertion of transporters into the canalicular membrane. In conclusion, the activation of IGF-1R is a key factor in the alteration of canalicular transporter function and localization induced by E17G, and its activation follows that of GPR30 and precedes that of PI3K/Akt.


Subject(s)
Cholestasis/metabolism , Estradiol/analogs & derivatives , Hepatocytes/drug effects , Receptor, IGF Type 1/metabolism , Receptors, G-Protein-Coupled/metabolism , ATP Binding Cassette Transporter, Subfamily B, Member 11/metabolism , ATP-Binding Cassette Transporters/metabolism , Animals , Cells, Cultured , Cholestasis/chemically induced , Endocytosis , Estradiol/toxicity , Female , Hepatocytes/metabolism , Liver/drug effects , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , RNA Interference , Rats , Rats, Wistar , Signal Transduction , Tyrphostins/pharmacology , Wortmannin/pharmacology
11.
Toxicol Appl Pharmacol ; 304: 90-8, 2016 08 01.
Article in English | MEDLINE | ID: mdl-27180241

ABSTRACT

Oxidative stress is a frequent cause underlying drug-induced hepatotoxicity. Benznidazole (BZL) is the only trypanocidal agent available for treatment of Chagas disease in endemic areas. Its use is associated with side effects, including increases in biomarkers of hepatotoxicity. However, BZL potential to cause oxidative stress has been poorly investigated. Here, we evaluated the effect of a pharmacologically relevant BZL concentration (200µM) at different time points on redox status and the counteracting mechanisms in the human hepatic cell line HepG2. BZL increased reactive oxygen species (ROS) after 1 and 3h of exposure, returning to normality at 24h. Additionally, BZL increased glutathione peroxidase activity at 12h and the oxidized glutathione/total glutathione (GSSG/GSSG+GSH) ratio that reached a peak at 24h. Thus, an enhanced detoxification of peroxide and GSSG formation could account for ROS normalization. GSSG/GSSG+GSH returned to control values at 48h. Expression of the multidrug resistance-associated protein 2 (MRP2) and GSSG efflux via MRP2 were induced by BZL at 24 and 48h, explaining normalization of GSSG/GSSG+GSH. BZL activated the nuclear erythroid 2-related factor 2 (Nrf2), already shown to modulate MRP2 expression in response to oxidative stress. Nrf2 participation was confirmed using Nrf2-knockout mice in which MRP2 mRNA expression was not affected by BZL. In summary, we demonstrated a ROS increase by BZL in HepG2 cells and a glutathione peroxidase- and MRP2 driven counteracting mechanism, being Nrf2 a key modulator of this response. Our results could explain hepatic alterations associated with BZL therapy.


Subject(s)
Multidrug Resistance-Associated Proteins , NF-E2-Related Factor 2 , Nitroimidazoles , Oxidative Stress , Trypanocidal Agents , Animals , Humans , Male , Mice , Glutathione Disulfide/metabolism , Glutathione Peroxidase/metabolism , Hep G2 Cells , Mice, Inbred C57BL , Multidrug Resistance-Associated Protein 2 , Multidrug Resistance-Associated Proteins/biosynthesis , NF-E2-Related Factor 2/biosynthesis , Nitroimidazoles/pharmacology , Oxidation-Reduction/drug effects , Oxidative Stress/drug effects , Reactive Oxygen Species/metabolism , RNA, Small Interfering/drug effects , Trypanocidal Agents/pharmacology
12.
Curr Med Chem ; 23(13): 1370-89, 2016.
Article in English | MEDLINE | ID: mdl-27048380

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.


Subject(s)
ATP-Binding Cassette Transporters/metabolism , Genistein/pharmacology , Phytoestrogens/pharmacology , Drug Interactions , Drug Resistance, Multiple/drug effects , Drug Resistance, Neoplasm/drug effects , Genistein/metabolism , Humans , Neoplasms/drug therapy , Neoplasms/metabolism , Neoplasms/pathology , Phytoestrogens/metabolism
13.
PLoS One ; 10(3): e0119502, 2015.
Article in English | MEDLINE | ID: mdl-25781341

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.


Subject(s)
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 , 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
SELECTION OF CITATIONS
SEARCH DETAIL