TY - JOUR AU - Kaci, Hana AU - Bakos, Éva AU - Needs, Paul W. AU - Kroon, Paul A. AU - Valentová, Kateřina AU - Poór, Miklós AU - Laczka, Csilla TI - The 2-aminoethyl diphenylborinate-based fluorescent method identifies quercetin and luteolin metabolites as substrates of Organic anion transporting polypeptides, OATP1B1 and OATP2B1 JF - EUROPEAN JOURNAL OF PHARMACEUTICAL SCIENCES J2 - EUR J PHARM SCI PY - 2024 SN - 0928-0987 DO - 10.1016/j.ejps.2024.106740 UR - https://m2.mtmt.hu/api/publication/34719089 ID - 34719089 AB - Organic anion transporting polypeptides (OATPs), OATP1B1 and OATP2B1 are membrane proteins mediating the cellular uptake of chemically diverse organic compounds. OATP1B1 is exclusively expressed in hepatocytes and plays a key role in hepatic detoxification. The ubiquitously expressed OATP2B1 promotes the intestinal absorption of orally administered drugs. Flavonoids are widely found in foods and beverages, and many of them can inhibit OATP function, resulting in food-drug interactions. In our previous work, we have shown that not only luteolin (LUT) and quercetin (Q), but also some of their metabolites can inhibit OATP1B1 and OATP2B1 activity. However, data about the potential direct transport of these flavonoids by OATPs have been incomplete. Hence, in the current study, we developed a simple, fluorescence-based method for the measurement of intracellular flavonoid levels. The method applies a cell-permeable small molecule (2-aminoethyl diphenylborinate, 2-APB), that, upon forming a complex with flavonoids, results in their fluorescence enhancement. This way the direct uptake of LUT and Q, and also their metabolites could be investigated both by confocal microscopy and in a fluorescence plate reader in living cells. With this approach we identified quercetin-3'-O-sulfate, luteolin-3'-O-glucuronide, luteolin-7-O-glucuronide and luteolin-3'-O-sulfate as substrates of both OATP1B1 and OATP2B1. Our results highlight that OATP1B1 and OATP2B1 can be key participants in the transmembrane movement of cell-permeable LUT and Q conjugates with otherwise low cell permeability. In addition, the novel method developed in this study can be a good completion to existing fluorescence-based assays to investigate OATP function. LA - English DB - MTMT ER - TY - JOUR AU - Kaci, Hana AU - Dombi, Ágnes AU - Gömbös, Patrik AU - Szabó, András AU - Bakos, Éva AU - Laczka, Csilla AU - Poór, Miklós TI - Interaction of mycotoxins zearalenone, α-zearalenol, and β-zearalenol with cytochrome P450 (CYP1A2, 2C9, 2C19, 2D6, and 3A4) enzymes and organic anion transporting polypeptides (OATP1A2, OATP1B1, OATP1B3, and OATP2B1) JF - TOXICOLOGY IN VITRO J2 - TOXICOL IN VITRO VL - 96 PY - 2024 PG - 8 SN - 0887-2333 DO - 10.1016/j.tiv.2024.105789 UR - https://m2.mtmt.hu/api/publication/34575448 ID - 34575448 N1 - Export Date: 3 May 2024 CODEN: TIVIE Correspondence Address: Poór, M.; Department of Laboratory Medicine, Ifjúság útja 13, Hungary; email: poor.miklos@pte.hu LA - English DB - MTMT ER - TY - JOUR AU - Ungvári, Orsolya AU - Bakos, Éva AU - Kovacsics, Daniella AU - Laczka, Csilla TI - The fluorescence‐based competitive counterflow assay developed for organic anion transporting polypeptides 1A2 , 1B1 , 1B3 and 2B1 identifies pentamidine as a selective OATP1A2 substrate JF - FASEB JOURNAL J2 - FASEB J VL - 37 PY - 2023 IS - 11 SN - 0892-6638 DO - 10.1096/fj.202300530RR UR - https://m2.mtmt.hu/api/publication/34173508 ID - 34173508 AB - Organic anion transporting polypeptides OATP1A2, OATP1B1, OATP1B3 and OATP2B1 are Na + ‐ and ATP‐independent exchangers of large, organic compounds, encompassing structurally diverse xenobiotics, including various drugs. These OATPs influence intestinal absorption (OATP2B1), hepatic clearance (OATP1B1/3) and blood to brain penetration (OATP1A2, OATP2B1) of their drug substrates. Consequently, OATP‐mediated drug or food interactions may lead to altered pharmacokinetics and toxicity. During drug development, investigation of hepatic OATP1B1 and OATP1B3 is recommended by international regulatory agencies. Most frequently, OATP‐drug interactions are investigated in an indirect assay, i.e., by examining uptake inhibition of a radioactive or fluorescent probe. However, indirect assays do not distinguish between transported substrates and non‐transported OATP inhibitors. To fill this hiatus, a novel assay, termed competitive counterflow (CCF) has been developed and has since been applied for several OATPs to differentiate between substrates and non‐transported inhibitors. However, previous OATP CCF assays, with the exception of that for OATP1B1, used radioactive probes. In the current study, we demonstrate that sulforhodamine 101 or pyranine can be used as fluorescent probes in a CCF assay to identify transported substrates of OATP1A2, or OATPs 1B1, 1B3 and 2B1, respectively. With the help of the newly developed fluorescence‐based CCF method, we identify the FDA‐approved anti‐protozoal drug, pentamidine as a unique substrate of OATP1A2. Furthermore, we confirm the selective, OATP1A2‐mediated uptake of pentamidine in a cytotoxicity assay. Based on our results, OATP1A2 may be an important determinant of pentamidine transport through the blood–brain barrier. LA - English DB - MTMT ER - TY - JOUR AU - Bakos, Éva AU - Temesszentandrási-Ambrus, Csilla AU - Laczka, Csilla AU - Gáborik, Z. AU - Sarkadi, Balázs AU - Telbisz, Ágnes Mária TI - Interactions of the Anti-SARS-CoV-2 Agents Molnupiravir and Nirmatrelvir/Paxlovid with Human Drug Transporters JF - INTERNATIONAL JOURNAL OF MOLECULAR SCIENCES J2 - INT J MOL SCI VL - 24 PY - 2023 IS - 14 SN - 1661-6596 DO - 10.3390/ijms241411237 UR - https://m2.mtmt.hu/api/publication/34095016 ID - 34095016 N1 - Research Centre for Natural Sciences—RCNS, Magyar Tudósok krt 2, Budapest, 1117, Hungary Charles River Laboratories, Irinyi József u. 4-20, Budapest, 1117, Hungary Export Date: 14 August 2023 Correspondence Address: Telbisz, Á.; Research Centre for Natural Sciences—RCNS, Magyar Tudósok krt 2, Hungary; email: telbisz.agnes@ttk.hu Funding details: Hungarian Scientific Research Fund, OTKA, FK 128751, K 138518 Funding details: Magyar Tudományos Akadémia, MTA, PC2022-7/2022 Funding details: Nemzeti Kutatási Fejlesztési és Innovációs Hivatal, NKFIH Funding text 1: This work was supported by the National Research Development and Innovation Office (NKFIH, OTKA) Grant number: FK 128751 (C.Ö.-L.) and K 138518 (C.Ö.-L.) and by a grant from the Hungarian Academy of Sciences (B.S., PC2022-7/2022). AB - Orally administered small molecules may have important therapeutic potential in treating COVID-19 disease. The recently developed antiviral agents, Molnupiravir and Nirmatrelvir, have been reported to be efficient treatments, with only moderate side effects, especially when applied in the early phases of this disease. However, drug–drug and drug–transporter interactions have already been noted by the drug development companies and in the application notes. In the present work, we have studied some of the key human transporters interacting with these agents. The nucleoside analog Molnupiravir (EIDD-2801) and its main metabolite (EIDD-1931) were found to inhibit CNT1,2 in addition to the ENT1,2 nucleoside transporters; however, it did not significantly influence the relevant OATP transporters or the ABCC4 nucleoside efflux transporter. The active component of Paxlovid (PF-07321332, Nirmatrelvir) inhibited the function of several OATPs and of ABCB1 but did not affect ABCG2. However, significant inhibition was observed only at high concentrations of Nirmatrelvir and probably did not occur in vivo. Paxlovid, as used in the clinic, is a combination of Nirmatrelvir (viral protease inhibitor) and Ritonavir (a “booster” inhibitor of Nirmatrelvir metabolism). Ritonavir is known to inhibit several drug transporters; therefore, we have examined these compounds together, in relevant concentrations and ratios. No additional inhibitory effect of Nirmatrelvir was observed compared to the strong transporter inhibition caused by Ritonavir. Our current in vitro results should help to estimate the potential drug–drug interactions of these newly developed agents during COVID-19 treatment. © 2023 by the authors. LA - English DB - MTMT ER - TY - JOUR AU - Hlogyik, Tamás AU - Laczkó-Rigó, Réka AU - Bakos, Éva AU - Poór, Miklós AU - Kele, Zoltán AU - Laczka, Csilla AU - Mernyák, Erzsébet TI - Synthesis and in vitro photodynamic activity of aza-BODIPY-based photosensitizers JF - ORGANIC & BIOMOLECULAR CHEMISTRY J2 - ORG BIOMOL CHEM VL - 21 PY - 2023 IS - 29 SP - 6018 EP - 6027 PG - 10 SN - 1477-0520 DO - 10.1039/d3ob00699a UR - https://m2.mtmt.hu/api/publication/34067941 ID - 34067941 N1 - * Megosztott szerzőség AB - Aza-BODIPY dyes have recently come to attention owing to their excellent chemical and photophysical properties. In particular, their absorption and emission maxima can efficiently be shifted to the red or even to the NIR spectral region. On this basis, aza-BODIPY derivatives are widely investigated as fluorescent probes or phototherapeutic agents. Here we report the synthesis of a set of novel aza-BODIPY derivatives as potential photosensitizers for use in photodynamic therapy. Triazolyl derivatives were obtained via Cu(I)-catalyzed azide-alkyne cycloaddition as the key step. In vitro photodynamic activities of the newly synthesized compounds were evaluated on the A431 human epidermoid carcinoma cell line. Structural differences influenced the light-induced toxicity of the test compounds markedly. Compared to the initial tetraphenyl aza-BODIPY derivative, the compound bearing two hydrophilic triethylene glycol side chains showed substantial, more than 250-fold, photodynamic activity with no dark toxicity. Our newly synthesized aza-BODIPY derivative, acting in the nanomolar range, might serve as a promising candidate for the design of more active and selective photosensitizers. LA - English DB - MTMT ER - TY - JOUR AU - Németh, Krisztina AU - László, Zsófia AU - Biró, Adrienn AU - Szatmári, Ágnes AU - Cserép, Balázs Gergely AU - Várady, György AU - Bakos, Éva AU - Laczka, Csilla AU - Kele, Péter TI - Organic anion transporting polypeptide 3A1 (OATP3A1)-gated bioorthogonal labeling of intracellular proteins JF - MOLECULES J2 - MOLECULES VL - 28 PY - 2023 IS - 6 PG - 10 SN - 1420-3049 DO - 10.3390/molecules28062521 UR - https://m2.mtmt.hu/api/publication/33688209 ID - 33688209 N1 - Chemical Biology Research Group, Institute of Organic Chemistry, RCNS, Magyar Tudósok Krt. 2, Budapest, H-1117, Hungary Molecular Cell Biology Research Group, Institute of Enzymology, RCNS, Magyar Tudósok Krt. 2, Budapest, H-1117, Hungary Membrane Protein Research Group, Institute of Enzymology, RCNS, Magyar Tudósok Krt. 2, Budapest, H-1117, Hungary Export Date: 19 April 2023 CODEN: MOLEF Correspondence Address: Németh, K.; Chemical Biology Research Group, Magyar Tudósok Krt. 2, Hungary; email: nemeth.krisztina@ttk.hu Correspondence Address: Kele, P.; Chemical Biology Research Group, Magyar Tudósok Krt. 2, Hungary; email: kele.peter@ttk.hu Chemicals/CAS: protein, 67254-75-5; Fluorescent Dyes; Organic Anion Transporters; Proteins Funding details: Nemzeti Kutatási Fejlesztési és Innovációs Hivatal, NKFIH, NKFIH-K-138518, NKFIH-K-143581, VEKOP-2.3.3-15-2016-00011 Funding text 1: Present work was supported by the National Research, Development and Innovation Office of Hungary (NKFIH-K-143581, NKFIH-K-138518 and VEKOP-2.3.3-15-2016-00011). AB - Organic anion transporting polypeptides (OATPs) were found to readily deliver membrane impermeable, tetrazine bearing fluorescent probes into cells. This feature was explored in OATP3A1 conditioned bio-orthogonal labeling schemes of various intracellular proteins in live cells. Confocal microscopy and super-resolution microscopy (STED) studies have shown that highly specific and efficient staining of the selected intracellular proteins can be achieved with the otherwise non-permeable probes when OATP3A1 is present in the cell membrane of cells. Such a transport protein linked bio-orthogonal labeling scheme is believed to be useful in OATP3A1 activity-controlled protein expression studies in the future. © 2023 by the authors. LA - English DB - MTMT ER - TY - JOUR AU - Laczka, Csilla AU - Ungvári, Orsolya AU - Bakos, Éva TI - Fluorescence-based methods for studying activity and drug-drug interactions of hepatic solute carrier and ATP binding cassette proteins involved in ADME-Tox JF - BIOCHEMICAL PHARMACOLOGY J2 - BIOCHEMIC PHARMACOL VL - 209 PY - 2023 SN - 0006-2952 DO - 10.1016/j.bcp.2023.115448 UR - https://m2.mtmt.hu/api/publication/33633514 ID - 33633514 N1 - Institute of Enzymology, RCNS, Eötvös Loránd Research Network, H-1117 Budapest, Magyar tudósok krt. 2., Hungary Doctoral School of Biology, Institute of Biology, ELTE Eötvös Loránd University, Budapest, Hungary Export Date: 12 January 2024 CODEN: BCPCA Correspondence Address: Özvegy-Laczka, C.; Institute of Enzymology, H-1117 Budapest, Magyar tudósok krt. 2., Hungary; email: laczka.csilla@ttk.hu Chemicals/CAS: ABC transporter subfamily B, 149200-37-3, 208997-77-7; bilirubin, 18422-02-1, 635-65-4; heme, 14875-96-8; indocyanine green, 3599-32-4; methylene blue, 61-73-4; rhodamine 123, 62669-70-9; adenosine triphosphate, 15237-44-2, 56-65-5, 987-65-5; carrier protein, 80700-39-6; Adenosine Triphosphate; ATP-Binding Cassette Transporters; Membrane Transport Proteins; Pharmaceutical Preparations LA - English DB - MTMT ER - TY - JOUR AU - Kaci, Hana AU - Bodnárová, Slávka AU - Fliszár-Nyúl, Eszter AU - Lemli, Beáta AU - Pelantová, Helena AU - Valentová, Kateřina AU - Bakos, Éva AU - Laczka, Csilla AU - Poór, Miklós TI - Interaction of luteolin, naringenin, and their sulfate and glucuronide conjugates with human serum albumin, cytochrome P450 (CYP2C9, CYP2C19, and CYP3A4) enzymes and organic anion transporting polypeptide (OATP1B1 and OATP2B1) transporters JF - BIOMEDICINE & PHARMACOTHERAPY J2 - BIOMED PHARMACOTHER VL - 157 PY - 2023 PG - 12 SN - 0753-3322 DO - 10.1016/j.biopha.2022.114078 UR - https://m2.mtmt.hu/api/publication/33294693 ID - 33294693 N1 - Drug Resistance Research Group, Institute of Enzymology, Research Centre for Natural Sciences, Eötvös Loránd Research Network, Magyar tudósok krt. 2, Budapest, H-1117, Hungary Doctoral School of Biology, Institute of Biology, Eötvös Loránd University, Pázmány P. stny. 1/C, Budapest, H-1117, Hungary Department of Pharmacology, Faculty of Pharmacy, University of Pécs, Rókus u. 2, Pécs, H-7624, Hungary Food Biotechnology Research Group, János Szentágothai Research Centre, University of Pécs, Ifjúság útja 20, Pécs, H-7624, Hungary Green Chemistry Research Group, János Szentágothai Research Center, University of Pécs, Ifjúság útja 20, Pécs, H-7624, Hungary Institute of Microbiology of the Czech Academy of Sciences, Vídeňská 1083, Prague, CZ-142, Czech Republic Cited By :1 Export Date: 4 September 2023 CODEN: BIPHE Correspondence Address: Poór, M.; Department of Pharmacology, Rókus u. 2, Hungary; email: poor.miklos@pte.hu AB - Luteolin and naringenin are flavonoids found in various foods/beverages and present in certain dietary supplements. After a high intake of these flavonoids, their sulfate and glucuronide conjugates reach micromolar concentrations in the bloodstream. Some pharmacokinetic interactions of luteolin and naringenin have been investigated in previous studies; however, only limited data are available in regard to their metabolites. In this study, we aimed to investigate the interactions of the sulfate and glucuronic acid conjugates of luteolin and naringenin with human serum albumin, cytochrome P450 (CYP2C9, 2C19, and 3A4) enzymes, and organic anion transporting polypeptide (OATP1B1 and OATP2B1) transporters. Our main findings are as follows: (1) Sulfate conjugates formed more stable complexes with albumin than the parent flavonoids. (2) Luteolin and naringenin conjugates showed no or only weak inhibitory action on the CYP enzymes examined. (3) Certain conjugates of luteolin and naringenin are potent inhibitors of OATP1B1 and/or OATP2B1 enzymes. (4) Conjugated metabolites of luteolin and naringenin may play an important role in the pharmacokinetic interactions of these flavonoids. LA - English DB - MTMT ER - TY - JOUR AU - Bakos, Éva AU - Nemet, Orsolya AU - Kucsma, Nóra AU - Tökési, Natália AU - Stieger, Bruno AU - Rushing, Elisabeth AU - Tőkés, Anna-Mária AU - Kele, Péter AU - Tusnády, Gábor AU - Laczka, Csilla TI - Cloning and characterization of a novel functional organic anion transporting polypeptide 3A1 isoform highly expressed in the human brain and testis JF - FRONTIERS IN PHARMACOLOGY J2 - FRONT PHARMACOL VL - 13 PY - 2022 PG - 12 SN - 1663-9812 DO - 10.3389/fphar.2022.958023 UR - https://m2.mtmt.hu/api/publication/33128050 ID - 33128050 AB - Organic anion transporting polypeptide 3A1 (OATP3A1, encoded by the SLCO3A1 gene) is a prostaglandin, oligopeptide, and steroid/thyroid hormone transporter with wide tissue distribution, expressed, e.g., in the human brain and testis. Although the physiological importance of OATP3A1 has not yet been clarified, based on its expression pattern, substrate recognition, and evolutionary conservation, OATP3A1 is a potential pharmacological target. Previously, two isoforms of OATP3A1, termed as V1 and V2, have been characterized. Here, we describe the cloning and functional characterization of a third isoform, OATP3A1_V3. The mRNA of isoform V3 is formed by alternative splicing and results in an OATP3A1 protein with an altered C-terminus compared to isoforms V1 and V2. Based on quantitative PCR, we demonstrate the widespread expression of SLCO3A1_V3 mRNA in human organs, with the highest expression in the brain and testis. By generation of an isoform V3-specific antibody and immunostaining, we show that the encoded protein is expressed in the human choroid plexus, neurons, and both germ and Sertoli cells of the testis. Moreover, we demonstrate that in contrast to isoform V1, OATP3A1_V3 localizes to the apical membrane of polarized MDCKII cells. Using HEK-293 cells engineered to overexpress OATP3A1_V3, we verify the protein's functionality and identify dehydroepiandrosterone sulfate as a novel OATP3A1 substrate. Based on their distinct expression patterns but overlapping functions, OATP3A1 isoforms may contribute to transcellular (neuro)steroid transport in the central nervous system. LA - English DB - MTMT ER - TY - JOUR AU - Temesszentandrási-Ambrus, Csilla AU - Bakos, Éva AU - Sarkadi, Balázs AU - Laczka, Csilla AU - Telbisz, Ágnes Mária TI - Interactions of anti-COVID-19 drug candidates with hepatic transporters may cause liver toxicity and affect pharmacokinetics JF - SCIENTIFIC REPORTS J2 - SCI REP VL - 11 PY - 2021 IS - 1 PG - 10 SN - 2045-2322 DO - 10.1038/s41598-021-97160-3 UR - https://m2.mtmt.hu/api/publication/32216442 ID - 32216442 N1 - These authors contributed equally: Csilla Ambrus and Éva Bakos AB - Transporters in the human liver play a major role in the clearance of endo- and xenobiotics. Apical (canalicular) transporters extrude compounds to the bile, while basolateral hepatocyte transporters promote the uptake of, or expel, various compounds from/into the venous blood stream. In the present work we have examined the in vitro interactions of some key repurposed drugs advocated to treat COVID-19 (lopinavir, ritonavir, ivermectin, remdesivir and favipiravir), with the key drug transporters of hepatocytes. These transporters included ABCB11/BSEP, ABCC2/MRP2, and SLC47A1/MATE1 in the canalicular membrane, as well as ABCC3/MRP3, ABCC4/MRP4, SLC22A1/OCT1, SLCO1B1/OATP1B1, SLCO1B3/OATP1B3, and SLC10A1/NTCP, residing in the basolateral membrane. Lopinavir and ritonavir in low micromolar concentrations inhibited BSEP and MATE1 exporters, as well as OATP1B1/1B3 uptake transporters. Ritonavir had a similar inhibitory pattern, also inhibiting OCT1. Remdesivir strongly inhibited MRP4, OATP1B1/1B3, MATE1 and OCT1. Favipiravir had no significant effect on any of these transporters. Since both general drug metabolism and drug-induced liver toxicity are strongly dependent on the functioning of these transporters, the various interactions reported here may have important clinical relevance in the drug treatment of this viral disease and the existing co-morbidities. © 2021, The Author(s). LA - English DB - MTMT ER -