Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Toxicol In Vitro ; 56: 52-61, 2019 Apr.
Article in English | MEDLINE | ID: mdl-30639138

ABSTRACT

Chronic kidney disease (CKD) is accompanied by accumulating levels of uremic solutes in the circulation. Changes in the size and composition of the bile acid pool have also been observed. We investigated via which mechanisms uremic solutes may interfere with hepatocyte function and thus contribute to altered bile acid handling. We studied interference on the level of bile acid synthesis by cytochrome P450 7A1 (CYP7A1), explored effects on hepatic bile acid transporters, and investigated effects on mitochondrial function. In HEK293 cells overexpressing bile salt transporters, we observed that p-cresyl sulfate inhibited Na+-taurocholate cotransporting polypeptide (NTCP)-mediated uptake of taurocholic acid (TCA), whereas organic anion-transporting polypeptide 1B1 (OATP1B1)-mediated TCA uptake was increased. Assays in transporter-overexpressing membrane vesicles revealed that kynurenic acid inhibited TCA transport via the bile salt efflux pump (BSEP), whereas p-cresyl glucuronide and hippuric acid increased TCA efflux via multidrug resistance-associated protein 3 (MRP3). Moreover, indoxyl sulfate decreased mRNA expression of NTCP, OATP1B3 and CYP7A1 in primary human hepatocytes. Transport studies confirmed a decreased TCA uptake in indoxyl sulfate-exposed hepatocytes. Decreased hepatocyte viability was found for all seven uremic solutes tested, whereas five out of seven also decreased intracellular ATP levels and mitochondrial membrane potential. In conclusion, uremic solutes affect hepatic bile acid transport and mitochondrial function. This can contribute to the altered bile acid homeostasis observed in CKD patients.


Subject(s)
Hepatocytes/metabolism , Mitochondria, Liver/metabolism , Taurocholic Acid/metabolism , Uremia/metabolism , Adenosine Triphosphate/metabolism , HEK293 Cells , Humans , Lactic Acid/metabolism , Membrane Transport Proteins/metabolism , Renal Insufficiency, Chronic/metabolism
2.
Toxicol Sci ; 162(2): 655-666, 2018 04 01.
Article in English | MEDLINE | ID: mdl-29329425

ABSTRACT

Primary human hepatocytes (PHHs) are commonly used for in vitro studies of drug-induced liver injury. However, when cultured as 2D monolayers, PHH lose crucial hepatic functions within hours. This dedifferentiation can be ameliorated when PHHs are cultured in sandwich configuration (2Dsw), particularly when cultures are regularly re-overlaid with extracellular matrix, or as 3D spheroids. In this study, the 6 participating laboratories evaluated the robustness of these 2 model systems made from cryopreserved PHH from the same donors considering both inter-donor and inter-laboratory variability and compared their suitability for use in repeated-dose toxicity studies using 5 different hepatotoxins with different toxicity mechanisms. We found that expression levels of proteins involved in drug absorption, distribution, metabolism, and excretion, as well as catalytic activities of 5 different CYPs, were significantly higher in 3D spheroid cultures, potentially affecting the exposure of the cells to drugs and their metabolites. Furthermore, global proteomic analyses revealed that PHH in 3D spheroid configuration were temporally stable whereas proteomes from the same donors in 2Dsw cultures showed substantial alterations in protein expression patterns over the 14 days in culture. Overall, spheroid cultures were more sensitive to the hepatotoxic compounds investigated, particularly upon long-term exposures, across testing sites with little inter-laboratory or inter-donor variability. The data presented here suggest that repeated-dosing regimens improve the predictivity of in vitro toxicity assays, and that PHH spheroids provide a sensitive and robust system for long-term mechanistic studies of drug-induced hepatotoxicity, whereas the 2Dsw system has a more dedifferentiated phenotype and lower sensitivity to detect hepatotoxicity.


Subject(s)
Chemical and Drug Induced Liver Injury/pathology , Hepatocytes/drug effects , Pharmaceutical Preparations/administration & dosage , Spheroids, Cellular/drug effects , Toxicity Tests/methods , Cell Survival/drug effects , Cells, Cultured , Coculture Techniques , Cryopreservation , Cytochrome P-450 Enzyme System/metabolism , Dose-Response Relationship, Drug , Hepatocytes/metabolism , Hepatocytes/pathology , Humans , Male , Middle Aged , Models, Biological , Predictive Value of Tests , Primary Cell Culture , Spheroids, Cellular/metabolism , Spheroids, Cellular/pathology , Time Factors , Toxicity Tests/standards
3.
J Appl Toxicol ; 38(2): 160-171, 2018 Feb.
Article in English | MEDLINE | ID: mdl-28960351

ABSTRACT

Exposure of humans to metal oxide nanoparticles (NPs) occurs mainly via air, and inhaled metal oxide NPs may generate inflammation. The aim of this study was to investigate the proinflammatory potential of six metal oxide NPs (CeO2 , Mn2 O3 , CuO, ZnO, Co3 O4 and WO3 ; 27-108 µg ml-1 ) using human primary 3-dimensional airway epithelium (MucilAir™) and dendritic cell (DC) models. Metal oxide NPs were mainly aggregated/agglomerated in the cell media, as determined by dynamic light scattering, scanning electron microscopy and differential centrifugal sedimentation. WO3 and ZnO were highly soluble, both with and without respiratory mucus. Proinflammatory signalling by the epithelium was evaluated after a 24 hour exposure by increased interleukin-6 and -8 and monocyte chemoattractant protein 1 cytokine release, which occurred only for CuO. Moreover, maturation of immature human DCs, which play a key role in the lung immune system, were evaluated by expression of surface markers HLA-DR, CD80, CD83 and CD86 after a 48 hour exposure. Only Mn2 O3 consistently upregulated DC maturation markers. Furthermore, by addition of medium from metal oxide NP-exposed 3-dimensional airway cultures to metal oxide NP-exposed DC cultures, the interplay between lung epithelium and DCs was studied. Such an interplay was again only observed for Mn2 O3 and in one of five DC donors. Our results show that, even when using dosages that represent very high in vivo exposure levels, up to 27 hours of constant human airway exposure, metal oxide NPs cause minimal proinflammatory effects and that epithelial cells not necessarily interfere with DC maturation upon metal oxide NP exposure. The present approach exemplifies a relevant translation towards human safety assessment.


Subject(s)
Dendritic Cells/drug effects , Metal Nanoparticles/toxicity , Metals, Heavy/toxicity , Respiratory Mucosa/drug effects , Administration, Inhalation , Cell Survival/drug effects , Cytokines/metabolism , Dendritic Cells/immunology , Dose-Response Relationship, Drug , Humans , Metal Nanoparticles/chemistry , Metals, Heavy/chemistry , Models, Biological , Oxides/toxicity , Respiratory Mucosa/cytology , Respiratory Mucosa/immunology
4.
Sci Rep ; 6: 25187, 2016 05 04.
Article in English | MEDLINE | ID: mdl-27143246

ABSTRACT

Liver biology and function, drug-induced liver injury (DILI) and liver diseases are difficult to study using current in vitro models such as primary human hepatocyte (PHH) monolayer cultures, as their rapid de-differentiation restricts their usefulness substantially. Thus, we have developed and extensively characterized an easily scalable 3D PHH spheroid system in chemically-defined, serum-free conditions. Using whole proteome analyses, we found that PHH spheroids cultured this way were similar to the liver in vivo and even retained their inter-individual variability. Furthermore, PHH spheroids remained phenotypically stable and retained morphology, viability, and hepatocyte-specific functions for culture periods of at least 5 weeks. We show that under chronic exposure, the sensitivity of the hepatocytes drastically increased and toxicity of a set of hepatotoxins was detected at clinically relevant concentrations. An interesting example was the chronic toxicity of fialuridine for which hepatotoxicity was mimicked after repeated-dosing in the PHH spheroid model, not possible to detect using previous in vitro systems. Additionally, we provide proof-of-principle that PHH spheroids can reflect liver pathologies such as cholestasis, steatosis and viral hepatitis. Combined, our results demonstrate that the PHH spheroid system presented here constitutes a versatile and promising in vitro system to study liver function, liver diseases, drug targets and long-term DILI.


Subject(s)
Chemical and Drug Induced Liver Injury/pathology , Chemical and Drug Induced Liver Injury/physiopathology , Hepatocytes/drug effects , Hepatocytes/physiology , Spheroids, Cellular/drug effects , Spheroids, Cellular/physiology , Arabinofuranosyluracil/analogs & derivatives , Arabinofuranosyluracil/toxicity , Cells, Cultured , Humans , Models, Biological , Proof of Concept Study , Proteome/analysis
5.
Toxicol In Vitro ; 29(7): 1868-77, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26216510

ABSTRACT

The uremic solutes p-cresyl sulfate (pCS) and p-cresyl glucuronide (pCG) accumulate in patients with chronic kidney disease (CKD), and might contribute to disease progression. Moreover, retention of these solutes may directly be related to renal tubular function. Here, we investigated the role of the efflux transporters Multidrug Resistance Protein 4 (MRP4) and Breast Cancer Resistance Protein (BCRP) in pCS and pCG excretion, and studied the impact of both solutes on the phenotype of human conditionally immortalized renal proximal tubule epithelial cells (ciPTEC). Our results show that p-cresol metabolites accumulate during CKD, with a shift from sulfation to glucuronidation upon progression. Moreover, pCS inhibited the activity of MRP4 by 40% and BCRP by 25%, whereas pCG only reduced MRP4 activity by 75%. Moreover, BCRP-mediated transport of both solutes was demonstrated. Exposure of ciPTEC to pCG caused epithelial-to-mesenchymal transition, indicated by increased expression of vimentin and Bcl-2, and diminished E-cadherin. This was associated with altered expression of key tubular transporters. In conclusion, BCRP is likely involved in the renal excretion of both solutes, and pCG promotes phenotypical changes in ciPTEC, supporting the notion that uremic toxins may be involved in CKD progression by negatively affecting renal tubule cell phenotype and functionality.


Subject(s)
ATP-Binding Cassette Transporters/metabolism , Cresols/metabolism , Glucuronides/metabolism , Kidney Tubules, Proximal/metabolism , Multidrug Resistance-Associated Proteins/metabolism , Neoplasm Proteins/metabolism , Sulfuric Acid Esters/metabolism , ATP Binding Cassette Transporter, Subfamily G, Member 2 , ATP-Binding Cassette Transporters/genetics , Adult , Aged , Baculoviridae/genetics , Cell Line , Female , HEK293 Cells , Humans , Male , Middle Aged , Multidrug Resistance-Associated Proteins/genetics , Neoplasm Proteins/genetics , Renal Elimination , Renal Insufficiency, Chronic/metabolism , Transduction, Genetic
6.
Toxicol Sci ; 136(2): 382-91, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24014645

ABSTRACT

Endocrine-disrupting chemicals (EDCs) are considered to cause testicular toxicity primarily via interference with steroid hormone function. Alternatively, EDCs could possibly exert their effects by interaction with ATP-binding cassette (ABC) transporters that are expressed in the blood-testis barrier. In this study, we investigated the effects of bisphenol A (BPA), tetrabromobisphenol A (TBBPA), bis(2-ethylhexyl) phthalate, mono(2-ethylhexyl) phthalate, perfluorooctanoic acid (PFOA), and perfluorooctanesulfonic acid (PFOS) on breast cancer resistance protein (BCRP), multidrug resistance proteins 1 and 4 (MRP1 and MRP4), and P-glycoprotein (P-gp) using membrane vesicles overexpressing these transporters. BPA solely inhibited BCRP activity, whereas TBBPA, PFOA, and PFOS inhibited all transporters tested. No effect was observed for the phthalates. Using transporter-overexpressing Madin-Darby canine kidney cells, we show that BPA and PFOA, but not TBBPA, are transported by BCRP, whereas none of the compounds were transported by P-gp. To investigate the toxicological implications of these findings, testosterone secretion and expression of steroidogenic genes were determined in murine Leydig (MA-10) cells upon exposure to the selected EDCs. Only BPA and TBBPA concentration dependently increased testosterone secretion by MA-10 cells to 6- and 46-fold of control levels, respectively. Inhibition of the Mrp's by MK-571 completely blocked testosterone secretion elicited by TBBPA, which could not be explained by coinciding changes in expression of steroidogenic genes. Therefore, we hypothesize that transporter-mediated efflux of testosterone precursors out of MA-10 cells is inhibited by TBBPA resulting in higher availability for testosterone production. Our data show the toxicological and clinical relevance of ABC transporters in EDC risk assessment related to testicular toxicity.


Subject(s)
ATP-Binding Cassette Transporters/metabolism , Blood-Testis Barrier/drug effects , Endocrine Disruptors/toxicity , Leydig Cells/drug effects , Testosterone/metabolism , Animals , Cells, Cultured , Dogs , HEK293 Cells , Humans , Leydig Cells/metabolism , Male , Mice
7.
Biochim Biophys Acta ; 1832(10): 1715-22, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23665398

ABSTRACT

Hyperuricemia is related to a variety of pathologies, including chronic kidney disease (CKD). However, the pathophysiological mechanisms underlying disease development are not yet fully elucidated. Here, we studied the effect of hyperuricemia on tryptophan metabolism and the potential role herein of two important uric acid efflux transporters, multidrug resistance protein 4 (MRP4) and breast cancer resistance protein (BCRP). Hyperuricemia was induced in mice by treatment with the uricase inhibitor oxonic acid, confirmed by the presence of urate crystals in the urine of treated animals. A transport assay, using membrane vesicles of cells overexpressing the transporters, revealed that uric acid inhibited substrate-specific transport by BCRP at clinically relevant concentrations (calculated IC50 value: 365±13µM), as was previously reported for MRP4. Moreover, we identified kynurenic acid as a novel substrate for MRP4 and BCRP. This finding was corroborated by increased plasma levels of kynurenic acid observed in Mrp4(-/-) (107±19nM; P=0.145) and Bcrp(-/-) mice (133±10nM; P=0.0007) compared to wild type animals (71±11nM). Hyperuricemia was associated with >1.5 fold increase in plasma kynurenine levels in all strains. Moreover, hyperuricemia led to elevated plasma kynurenic acid levels (128±13nM, P=0.005) in wild type mice but did not further increase kynurenic acid levels in knockout mice. Based on our results, we postulate that elevated uric acid levels hamper MRP4 and BCRP functioning, thereby promoting the retention of other potentially toxic substrates, including kynurenic acid, which could contribute to the development of CKD.


Subject(s)
ATP-Binding Cassette Transporters/antagonists & inhibitors , Hyperuricemia/metabolism , Multidrug Resistance-Associated Proteins/antagonists & inhibitors , Neoplasm Proteins/antagonists & inhibitors , Tryptophan/metabolism , ATP Binding Cassette Transporter, Subfamily G, Member 2 , Acute-Phase Proteins/metabolism , Biological Transport , HEK293 Cells , Humans , Hyperuricemia/chemically induced , Kynurenic Acid/metabolism , Lipocalin-2 , Lipocalins/metabolism , Oxonic Acid/administration & dosage , Proto-Oncogene Proteins/metabolism , Uric Acid/metabolism
8.
Curr Pharm Des ; 19(15): 2698-707, 2013.
Article in English | MEDLINE | ID: mdl-23092328

ABSTRACT

Heme is the functional group of diverse hemoproteins and crucial for many cellular processes. However, heme is increasingly recognized as a culprit for a wide variety of pathologies, including sepsis, malaria, and kidney failure. Excess of free heme can be detrimental to tissues by mediating oxidative and inflammatory injury. Protective mechanisms against free heme are therefore pivotal for cellular survival. We postulated that overexpression of Heme Oxygenase-1 (HO-1) and Breast Cancer Resistance Protein (BCRP) would protect against heme-induced cytotoxicity. HO-1 is a heme-degrading enzyme generating carbon monoxide, iron, and biliverdin/bilirubin, while BCRP is a heme efflux transporter. Human embryonic kidney cells were transduced using a baculovirus system as a novel strategy to efficiently overexpress HO-1 and BCRP. Exposing cells to heme resulted in a dose-dependent increase in reactive oxygen species formation, DNA damage and cell death. Heme-induced cell death was significantly attenuated when cells overexpressed HO-1, BCRP, or both. The protective effects of HO-1 overexpression were most pronounced, while co-treatment with the HO-activity inhibitor tin mesoporphyrin reversed these protective effects. Also cells treated with the anti-oxidants N-acetylcysteine or HO-effector molecule bilirubin showed protection against heme insults, which may explain the increased protection by HO-1 compared to BCRP. In conclusion, both HO-1 and BCRP protect against heme-induced toxicity and may thus form novel therapeutic targets for heme-mediated pathologies.


Subject(s)
ATP-Binding Cassette Transporters/physiology , Heme Oxygenase-1/metabolism , Heme/toxicity , Neoplasm Proteins/physiology , ATP Binding Cassette Transporter, Subfamily G, Member 2 , Benzimidazoles/metabolism , Blotting, Western , DNA Damage , HEK293 Cells , Humans , Oxidants/toxicity , Reactive Oxygen Species/metabolism
9.
Cell Tissue Res ; 349(2): 551-63, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22581381

ABSTRACT

Breast cancer resistance protein (BCRP) is known for its protective function against the toxic effects of exogenous compounds. In addition to this, a role in the transport of endogenous compounds has been described. Since BCRP in the plasma membrane was shown to be regulated by sex steroids, we investigated the presence and possible role of BCRP in steroid hormone-producing organs. Therefore, the presence and localization of Bcrp was investigated in endocrine organs of wild-type mice. Furthermore, the interaction of various steroid hormones with human BCRP activity was studied. Quantitative PCR revealed Bcrp mRNA in the pituitary and adrenal glands, pancreas, ovary, testis and adipose tissue. Immunohistochemistry revealed the presence of Bcrp in the cortex of the adrenal gland and in plasma membranes of adipocytes. In the pituitary gland, pancreas, ovary and testis, Bcrp was mainly located in the capillaries. The interaction between BCRP and 12 steroid hormones was studied using membrane vesicles of HEK293-BCRP cells. Estradiol, testosterone, progesterone and androstenedione inhibited BCRP-mediated uptake of (3)H-estrone sulphate (E(1)S) most potently, with calculated inhibitory constant (Ki) values of 5.0 ± 0.2, 36 ± 14, 14.7 ± 1.3 and 217 ± 13 µM, respectively. BCRP function was attenuated non-competitively, which implies an allosteric inhibition of BCRP-mediated E(1)S transport by these steroids. In conclusion, localization of Bcrp in endocrine organs together with the efficient allosteric inhibition of the efflux pump by steroid hormones are suggestive for a role for BCRP in steroid hormone regulation.


Subject(s)
ATP-Binding Cassette Transporters/analysis , ATP-Binding Cassette Transporters/metabolism , Breast Neoplasms/metabolism , Hormones/metabolism , Neoplasm Proteins/analysis , Neoplasm Proteins/metabolism , Steroids/metabolism , ATP Binding Cassette Transporter, Subfamily G, Member 2 , ATP-Binding Cassette Transporters/genetics , Animals , Biological Transport, Active , Breast Neoplasms/genetics , Female , HEK293 Cells , Humans , Mice , Neoplasm Proteins/genetics , RNA, Messenger/analysis , RNA, Messenger/genetics , Transduction, Genetic
10.
PLoS One ; 6(4): e18438, 2011 Apr 04.
Article in English | MEDLINE | ID: mdl-21483698

ABSTRACT

During chronic kidney disease (CKD), there is a progressive accumulation of toxic solutes due to inadequate renal clearance. Here, the interaction between uremic toxins and two important efflux pumps, viz. multidrug resistance protein 4 (MRP4) and breast cancer resistance protein (BCRP) was investigated. Membrane vesicles isolated from MRP4- or BCRP-overexpressing human embryonic kidney cells were used to study the impact of uremic toxins on substrate specific uptake. Furthermore, the concentrations of various uremic toxins were determined in plasma of CKD patients using high performance liquid chromatography and liquid chromatography/tandem mass spectrometry. Our results show that hippuric acid, indoxyl sulfate and kynurenic acid inhibit MRP4-mediated [(3)H]-methotrexate ([(3)H]-MTX) uptake (calculated Ki values: 2.5 mM, 1 mM, 25 µM, respectively) and BCRP-mediated [(3)H]-estrone sulfate ([(3)H]-E1S) uptake (Ki values: 4 mM, 500 µM and 50 µM, respectively), whereas indole-3-acetic acid and phenylacetic acid reduce [(3)H]-MTX uptake by MRP4 only (Ki value: 2 mM and IC(50) value: 7 mM, respectively). In contrast, p-cresol, p-toluenesulfonic acid, putrescine, oxalate and quinolinic acid did not alter transport mediated by MRP4 or BCRP. In addition, our results show that hippuric acid, indole-3-acetic acid, indoxyl sulfate, kynurenic acid and phenylacetic acid accumulate in plasma of end-stage CKD patients with mean concentrations of 160 µM, 4 µM, 129 µM, 1 µM and 18 µM, respectively. Moreover, calculated Ki values are below the maximal plasma concentrations of the tested toxins. In conclusion, this study shows that several uremic toxins inhibit active transport by MRP4 and BCRP at clinically relevant concentrations.


Subject(s)
ATP-Binding Cassette Transporters/metabolism , Multidrug Resistance-Associated Proteins/metabolism , Neoplasm Proteins/metabolism , Toxins, Biological/toxicity , Uremia/metabolism , ATP Binding Cassette Transporter, Subfamily G, Member 2 , Biological Transport/drug effects , Cell Membrane/drug effects , Cell Membrane/metabolism , Dose-Response Relationship, Drug , Estrone/analogs & derivatives , Estrone/metabolism , Gene Expression Regulation/drug effects , HEK293 Cells , Humans , Kidney Failure, Chronic/blood , Methotrexate/metabolism , Substrate Specificity , Toxins, Biological/blood
SELECTION OF CITATIONS
SEARCH DETAIL
...