Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
Biochem Pharmacol ; 214: 115681, 2023 08.
Article in English | MEDLINE | ID: mdl-37429423

ABSTRACT

Although pharmacological treatment is the best option for most patients with advanced hepatocellular carcinoma (HCC), its success is very limited, partly due to reduced uptake and enhanced efflux of antitumor drugs. Here we have explored the usefulness of vectorizing drugs towards the organic anion transporting polypeptide 1B3 (OATP1B3) to enhance their efficacy against HCC cells. In silico studies (RNA-Seq data, 11 cohorts) and immunohistochemistry analyses revealed a marked interindividual variability, together with general downregulation but still expression of OATP1B3 in the plasma membrane of HCC cells. The measurement of mRNA variants in 20 HCC samples showed the almost absence of the cancer-type variant (Ct-OATP1B3) together with marked predominance of the liver-type variant (Lt-OATP1B3). In Lt-OATP1B3-expressing cells, the screening of 37 chemotherapeutical drugs and 17 tyrosine kinase receptors inhibitors (TKIs) revealed that 10 classical anticancer drugs and 12 TKIs were able to inhibit Lt-OATP1B3-mediated transport. Lt-OATP1B3-expressing cells were more sensitive than Mock parental cells (transduced with empty lentiviral vectors) to some Lt-OATP1B3 substrates (paclitaxel and the bile acid-cisplatin derivative Bamet-UD2), but not to cisplatin, which is not transported by Lt-OATP1B3. This enhanced response was abolished by competition with taurocholic acid, a known Lt-OATP1B3 substrate. Tumors subcutaneously generated in immunodeficient mice by Lt-OATP1B3-expressing HCC cells were more sensitive to Bamet-UD2 than those derived from Mock cells. In conclusion, Lt-OATP1B3 expression should be screened before deciding the use of anticancer drugs substrates of this carrier in the personalized treatment of HCC. Moreover, Lt-OATP1B3-mediated uptake must be considered when designing novel anti-HCC targeted drugs.


Subject(s)
Antineoplastic Agents , Carcinoma, Hepatocellular , Liver Neoplasms , Organic Anion Transporters , Animals , Mice , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/metabolism , Cisplatin/metabolism , Liver Neoplasms/drug therapy , Liver Neoplasms/metabolism , Organic Anion Transporters/metabolism , Organic Anion Transporters, Sodium-Independent/metabolism , Solute Carrier Organic Anion Transporter Family Member 1B3/genetics , Humans
2.
Hepatology ; 70(4): 1246-1261, 2019 10.
Article in English | MEDLINE | ID: mdl-30972782

ABSTRACT

Although the multi-tyrosine kinase inhibitor sorafenib is useful in the treatment of several cancers, cholangiocarcinoma (CCA) is refractory to this drug. Among other mechanisms of chemoresistance, impaired uptake through human organic cation transporter type 1 (hOCT1) (gene SLC22A1) has been suggested. Here we have investigated the events accounting for this phenotypic characteristic and have evaluated the interest of selective gene therapy strategies to overcome this limitation. Gene expression and DNA methylation of SLC22A1 were analyzed using intrahepatic (iCCA) and extrahepatic (eCCA) biopsies (Copenhagen and Salamanca cohorts; n = 132) and The Cancer Genome Atlas (TCGA)-CHOL (n = 36). Decreased hOCT1 mRNA correlated with hypermethylation status of the SLC22A1 promoter. Treatment of CCA cells with decitabine (demethylating agent) or butyrate (histone deacetylase inhibitor) restored hOCT1 expression and increased sorafenib uptake. MicroRNAs able to induce hOCT1 mRNA decay were analyzed in paired samples of TCGA-CHOL (n = 9) and Copenhagen (n = 57) cohorts. Consistent up-regulation in tumor tissue was found for miR-141 and miR-330. High proportion of aberrant hOCT1 mRNA splicing in CCA was also seen. Lentiviral-mediated transduction of eCCA (EGI-1 and TFK-1) and iCCA (HuCCT1) cells with hOCT1 enhanced sorafenib uptake and cytotoxic effects. In chemically induced CCA in rats, reduced rOct1 expression was accompanied by impaired sorafenib uptake. In xenograft models of eCCA cells implanted in mouse liver, poor response to sorafenib was observed. However, tumor growth was markedly reduced by cotreatment with sorafenib and adenoviral vectors encoding hOCT1 under the control of the BIRC5 promoter, a gene highly up-regulated in CCA. Conclusion: The reason for impaired hOCT1-mediated sorafenib uptake by CCA is multifactorial. Gene therapy capable of selectively inducing hOCT1 in tumor cells can be considered a potentially useful chemosensitization strategy to improve the response of CCA to sorafenib.


Subject(s)
Bile Duct Neoplasms/drug therapy , Cholangiocarcinoma/drug therapy , Down-Regulation/genetics , Octamer Transcription Factor-1/genetics , Protein Kinase Inhibitors/pharmacology , Sorafenib/pharmacology , Animals , Bile Duct Neoplasms/genetics , Bile Duct Neoplasms/pathology , Cell Line, Tumor/drug effects , Cholangiocarcinoma/genetics , Cholangiocarcinoma/pathology , DNA Methylation/genetics , Disease Models, Animal , Drug Resistance/genetics , Genetic Therapy/methods , Humans , Immunoblotting , Male , RNA, Messenger/genetics , Random Allocation , Rats , Rats, Wistar , Real-Time Polymerase Chain Reaction/methods , Statistics, Nonparametric
3.
Chem Biol Interact ; 305: 195-202, 2019 May 25.
Article in English | MEDLINE | ID: mdl-30940451

ABSTRACT

A significant risk to the food chain is the presence of noxious pollutants in the feeds of animals whose products are used in human nutrition. Consequently, analytical methods and biosensors have been developed to detect these types of contaminates in feeds. Here we have evaluated whether the expression of TolC, a promiscuous component of several ATP-dependent efflux pumps in E. coli, up-regulated in response to chemical stress, could be a useful biomarker for this aim. Changes in TolC expression in response to toxic compounds, with different abilities to induce DNA damage, were determined using two E. coli strains with (DH5α) and without (BL21(DE3)) inactivating mutation in RecA gene. Deoxycholic acid and potassium dichromate up-regulated TolC in both strains. In contrast, cisplatin-induced TolC up-regulation was abolished in the absence of a functional RecA. When the effect of several insecticides, herbicides, antibiotics and common soil pollutants on TolC expression was analyzed, a relationship between toxicity and their ability to up-regulate TolC was observed. However, this was not a general event because the insecticide α-cipermetrin induced a reduction in cell viability, which was not accompanied by TolC up-regulation. In contrast, the soil pollutant benzene was able to stimulate TolC expression at non-toxic concentrations. When this test was used to analyze aqueous extracts from different feedstuffs, up-regulation of TolC was found in the absence of cell toxicity and was even accompanied by enhanced cell viability. In conclusion, TolC expression is partly dependent on the integrity of the RecA/LexaA system. Although toxic compounds up-regulate TolC in a dose-dependent manner, this response is also activated by non-toxic agents. Thus, owing to its poor specificity regardless of its sensitivity, the use of TolC up-regulation in E. coli to detect the presence of toxic pollutants in conventional and unconventional sources of nutrients for ruminant feeding requires supplementary biomarkers.


Subject(s)
Bacterial Outer Membrane Proteins/metabolism , Biomarkers/metabolism , Escherichia coli Proteins/metabolism , Escherichia coli/metabolism , Membrane Transport Proteins/metabolism , Soil Pollutants/toxicity , Up-Regulation/drug effects , Bacterial Outer Membrane Proteins/genetics , Cisplatin/pharmacology , Deoxycholic Acid/toxicity , Escherichia coli/drug effects , Escherichia coli Proteins/genetics , Membrane Transport Proteins/genetics , Organoplatinum Compounds/toxicity , Potassium Dichromate/toxicity , RNA, Messenger/metabolism , Soil Pollutants/chemistry
4.
Arch Toxicol ; 93(3): 623-634, 2019 03.
Article in English | MEDLINE | ID: mdl-30659321

ABSTRACT

Several ATP-binding cassette (ABC) proteins reduce intracellular concentrations of antitumor drugs and hence weaken the response of cancer cells to chemotherapy. Accordingly, the inhibition of these export pumps constitutes a promising strategy to chemosensitize highly chemoresistant tumors, such as hepatocellular carcinoma (HCC). Here, we have investigated the ability of ß-caryophyllene oxide (CRYO), a naturally occurring sesquiterpene component of many essential oils, to inhibit, at non-toxic doses, ABC pumps and improve the response of HCC cells to sorafenib. First, we have obtained a clonal subline (Alexander/R) derived from human hepatoma cells with enhanced multidrug resistance (MDR) associated to up-regulation (mRNA and protein) of MRP1 and MRP2. Analysis of fluorescent substrates export (flow cytometry) revealed that CRYO did not affect the efflux of fluorescein (MRP3, MRP4 and MRP5) but inhibited that of rhodamine 123 (MDR1) and calcein (MRP1 and MRP2). This ability was higher for CRYO than for other sesquiterpenes assayed. CRYO also inhibited sorafenib efflux, increased its intracellular accumulation (HPLC-MS/MS) and enhanced its cytotoxic response (MTT). For comparison, the effect of known ABC pumps inhibitors was also determined. They induced strong (diclofenac on MRPs), modest (verapamil on MDR1) or null (fumitremorgin C on BCRP) effect on sorafenib efflux and cytotoxicity. In the mouse xenograft model, the response to sorafenib treatment of subcutaneous tumors generated by mouse hepatoma Hepa 1-6/R cells, with marked MDR phenotype, was significantly enhanced by CRYO co-administration. In conclusion, at non-toxic dose, CRYO is able to chemosensitizating liver cancer cells to sorafenib by favoring its intracellular accumulation.


Subject(s)
Antineoplastic Agents/toxicity , Drug Resistance, Neoplasm/drug effects , Polycyclic Sesquiterpenes/metabolism , Sorafenib/toxicity , ATP-Binding Cassette Transporters/metabolism , Animals , Carcinoma, Hepatocellular , Cell Line, Tumor , Drug Resistance, Multiple , Humans , Liver Neoplasms , Mice , Neoplasm Proteins
5.
Oncotarget ; 8(21): 34617-34629, 2017 May 23.
Article in English | MEDLINE | ID: mdl-28423714

ABSTRACT

Tumor response to chemotherapy is often limited by drug export through ABC proteins. To overcome this problem, here we have investigated the usefulness of inducing the expression of the multidrug uptake transporter OATP1B1 under the control of the MRP2 promoter (MRP2pr). Human hepatoma cells (Alexander) were transfected with MRP2pr fragments of different length fused to the firefly luciferase ORF in order to select the shortest fragment with the highest response to dexamethasone, which was subsequently used to generate the chimeric construct MRP2pr-OATP1B1-V5. Hepatoma cells transduced with MRP2pr-OATP1B1-V5 resulted in dexamethasone-sensitive inducible OATP1B1 expression and enhanced selective antitumor response to OATP1B1 substrates (paclitaxel, Bamet-R2 and Bamet-UD2). In human colon cancer cells LS174T/R, used as a model of endogenous chemoresistance due to MRP2 overexpression, MRP2pr-OATP1B1 induced OATP1B1 expression together with chemosensitivity to OATP1B1 substrates. In nude mice, xenografted tumors formed by LS174T/R cells transduced with MRP2pr-OATP1B1 plus treatment with dexamethasone were markedly sensitized to Bamet-UD2. In conclusion, the induced expression of anticancer drug uptake transporters, under the control of promoters of ABC proteins involved in chemoresistance, constitutes an interesting approach to overcome the poor response of cancer to chemotherapy due to reduced drug uptake and/or enhanced drug export.


Subject(s)
Carcinoma, Hepatocellular/genetics , Colonic Neoplasms/genetics , Dexamethasone/administration & dosage , Drug Resistance, Neoplasm/drug effects , Liver Neoplasms/drug therapy , Liver-Specific Organic Anion Transporter 1/metabolism , Multidrug Resistance-Associated Proteins/genetics , Animals , Carcinoma, Hepatocellular/drug therapy , Colonic Neoplasms/drug therapy , Dexamethasone/pharmacology , Humans , Liver Neoplasms/genetics , Liver Neoplasms/metabolism , Liver-Specific Organic Anion Transporter 1/genetics , Mice , Mice, Nude , Multidrug Resistance-Associated Protein 2 , Promoter Regions, Genetic , Recombinant Proteins/administration & dosage , Recombinant Proteins/genetics , Recombinant Proteins/pharmacology , Transfection , Xenograft Model Antitumor Assays
6.
Food Chem ; 205: 81-8, 2016 Aug 15.
Article in English | MEDLINE | ID: mdl-27006217

ABSTRACT

Ash derived from energy generation is used as a source of minerals in livestock feeds. The microbial biosensor recApr-Luc2 was built to detect genotoxic hazard in recycled ash. Escherichia coli SOS gene (recA, lexA, dinI and umuC) expression in response to cisplatin-induced DNA damage led to the selection of the recA promoter. The biosensor required functional RecA expression to respond to genotoxic heavy metals (Cr>Cd≈Pb), and polluted ash induced a strong recApr-Luc2 response. In human liver and intestinal cells, heavy metals induced acute toxicity (Cr>Cd>Pb) at concentrations sufficient to activate recApr-Luc2. Cytostatic effects, including genotoxicity, were cell- and metal-dependent, apart from Cr. In agreement with the recApr-Luc2 bioassay, Cr had the strongest effect in all cells. In conclusion, recApr-Luc2 could be useful for evaluating the genotoxic risk of pollutants present in ash that might be concentrated in animal products and, thus, entering the human food chain.


Subject(s)
Biosensing Techniques/methods , Coal Ash/chemistry , Livestock/growth & development , Metals, Heavy/analysis , Mutagens/analysis , SOS Response, Genetics/drug effects , Animal Feed/analysis , Animals , Caco-2 Cells , Diet , Escherichia coli/genetics , Escherichia coli/metabolism , Escherichia coli Proteins/genetics , Food Chain , Food Contamination/analysis , Gene Expression/drug effects , Hep G2 Cells , Humans , Luciferases, Firefly/genetics , Metals, Heavy/toxicity , Mutagens/toxicity , Rec A Recombinases/genetics
7.
Mol Cancer Res ; 12(1): 91-100, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24255171

ABSTRACT

UNLABELLED: Bile acid accumulation in liver with cholangiolar neoplastic lesions may occur before cholestasis is clinically detected. Whether this favors intrahepatic cholangiocarcinoma development has been investigated in this study. The E. coli RecA gene promoter was cloned upstream from Luc2 to detect in vitro direct genotoxic ability by activation of SOS genes. This assay demonstrated that bile acids were not able to induce DNA damage. The genotoxic effect of the DNA-damaging agent cisplatin was neither enhanced nor hindered by the hepatotoxic and hepatoprotective glycochenodeoxycholic and glycoursodeoxycholic acids, respectively. In contrast, thioacetamide metabolites, but not thioacetamide itself, induced DNA damage. Thus, thioacetamide was used to induce liver cancer in rats, which resulted in visible tumors after 30 weeks. The effect of bile acid accumulation on initial carcinogenesis phase (8 weeks) was investigated in bile duct ligated (BDL) animals. Serum bile acid measurement and determination of liver-specific healthy and tumor markers revealed that early thioacetamide treatment induced hypercholanemia together with upregulation of the tumor marker Neu in bile ducts, which were enhanced by BDL. Bile acid accumulation was associated with increased expression of interleukin (IL)-6 and downregulation of farnesoid X receptor (FXR). Bile duct proliferation and apoptosis activation, with inverse pattern (BDL > thioacetamide + BDL >> thioacetamide vs. thioacetamide > thioacetamide + BDL > BDL), were observed. In conclusion, intrahepatic accumulation of bile acids does not induce carcinogenesis directly but facilitates a cocarcinogenic effect due to stimulation of bile duct proliferation, enhanced inflammation, and reduction in FXR-dependent chemoprotection. IMPLICATIONS: This study reveals that bile acids foster cocarcinogenic events that impact cholangiocarcinoma.


Subject(s)
Bile Acids and Salts/metabolism , Bile Duct Neoplasms/pathology , Bile Ducts, Intrahepatic/pathology , Cholangiocarcinoma/pathology , Animals , Apoptosis/drug effects , Bile Acids and Salts/blood , Bile Ducts, Intrahepatic/surgery , Biomarkers, Tumor/biosynthesis , Cell Proliferation/drug effects , Cells, Cultured , Cholestasis/diagnosis , Cholestasis/pathology , Cholic Acids/blood , Cisplatin/pharmacology , Cocarcinogenesis , Cross-Linking Reagents/pharmacology , DNA Damage/drug effects , DNA Damage/genetics , Glycochenodeoxycholic Acid/pharmacology , Hepatocytes/cytology , Hepatocytes/pathology , Inflammation , Interleukin-6/biosynthesis , Liver/pathology , Liver Neoplasms , Male , Promoter Regions, Genetic/genetics , Rats , Rats, Wistar , Rec A Recombinases/genetics , Receptor, ErbB-2/biosynthesis , Receptors, Cytoplasmic and Nuclear/biosynthesis , SOS Response, Genetics/genetics , Thioacetamide/pharmacology
8.
Bioorg Med Chem ; 21(14): 4432-41, 2013 Jul 15.
Article in English | MEDLINE | ID: mdl-23685181

ABSTRACT

Antitumor and antiviral properties of the antimalaria drug artemisinin from Artemisia annua have been reported. Novel artemisinin derivatives (AD1-AD8) have been synthesized and evaluated using in vitro models of liver/colon cancer and viral hepatitis B and C. Cell viability assays after treating human cell lines from hepatoblastoma (HepG2), hepatocarcinoma (SK-HEP-1), and colon adenocarcinoma (LS174T) with AD1-AD8 for a short (6h) and long (72h) period revealed that AD5 combined low acute toxicity together with high antiproliferative effect (IC50=1-5µM). Since iron-mediated activation of peroxide bond is involved in artemisinin antimalarial activity, the effect of iron(II)-glycine sulfate (ferrosanol) and iron(III)-containing protoporphyrin IX (hemin) was investigated. Ferrosanol, but not hemin, enhanced antiproliferative activity of AD5 if the cells were preloaded with AD5, but not if both compouds were added together. Five derivatives (AD1>AD2>AD7>AD3>AD8) were able to inhibit the cytopathic effect of bovine viral diarrhoea virus (BVDV), a surrogate in vitro model of hepatitis C virus (HCV), used here to evaluate the anti-Flaviviridae activity. Moreover, AD1 and AD2 inhibited the release of BVDV-RNA to the culture medium. Co-treatment with hemin or ferrosanol resulted in enhanced anti-Flaviviridae activity of AD1. In HepG2 cells permanently infected with hepatitis B virus (HBV), AD1 and AD4, at non-toxic concentrations for the host cells were able to reduce the release of HBV-DNA to the medium. In conclusion, high pharmacological interest deserving further evaluation in animal models has been identified for novel artemisinin-related drugs potentially useful for the treatment of liver cancer and viral hepatitis B and C.


Subject(s)
Artemisinins/chemistry , Artemisinins/pharmacology , Colonic Neoplasms/drug therapy , Hepatitis, Viral, Human/drug therapy , Liver Neoplasms/drug therapy , Virus Replication/drug effects , Animals , Artemisinins/chemical synthesis , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Humans , Molecular Structure
SELECTION OF CITATIONS
SEARCH DETAIL
...