1
|
Jochum K, Miccoli A, Sommersdorf C, Poetz O, Braeuning A, Tralau T, Marx-Stoelting P. Comparative case study on NAMs: towards enhancing specific target organ toxicity analysis. Arch Toxicol 2024; 98:3641-3658. [PMID: 39207506 PMCID: PMC11489238 DOI: 10.1007/s00204-024-03839-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Accepted: 08/08/2024] [Indexed: 09/04/2024]
Abstract
Traditional risk assessment methodologies in toxicology have relied upon animal testing, despite concerns regarding interspecies consistency, reproducibility, costs, and ethics. New Approach Methodologies (NAMs), including cell culture and multi-level omics analyses, hold promise by providing mechanistic information rather than assessing organ pathology. However, NAMs face limitations, like lacking a whole organism and restricted toxicokinetic interactions. This is an inherent challenge when it comes to the use of omics data from in vitro studies for the prediction of organ toxicity in vivo. One solution in this context are comparative in vitro-in vivo studies as they allow for a more detailed assessment of the transferability of the respective NAM data. Hence, hepatotoxic and nephrotoxic pesticide active substances were tested in human cell lines and the results subsequently related to the biology underlying established effects in vivo. To this end, substances were tested in HepaRG and RPTEC/tERT1 cells at non-cytotoxic concentrations and analyzed for effects on the transcriptome and parts of the proteome using quantitative real-time PCR arrays and multiplexed microsphere-based sandwich immunoassays, respectively. Transcriptomics data were analyzed using three bioinformatics tools. Where possible, in vitro endpoints were connected to in vivo observations. Targeted protein analysis revealed various affected pathways, with generally fewer effects present in RPTEC/tERT1. The strongest transcriptional impact was observed for Chlorotoluron in HepaRG cells (increased CYP1A1 and CYP1A2 expression). A comprehensive comparison of early cellular responses with data from in vivo studies revealed that transcriptomics outperformed targeted protein analysis, correctly predicting up to 50% of in vivo effects.
Collapse
Affiliation(s)
- Kristina Jochum
- Department of Pesticides Safety, German Federal Institute for Risk Assessment, Berlin, Germany
| | - Andrea Miccoli
- Department of Pesticides Safety, German Federal Institute for Risk Assessment, Berlin, Germany
- Institute for Marine Biological Resources and Biotechnology (IRBIM), National Research Council, Ancona, Italy
- Department of Food Safety, German Federal Institute for Risk Assessment, Berlin, Germany
| | | | - Oliver Poetz
- Signatope GmbH, Tübingen, Germany
- NMI Natural and Medical Sciences Institute at the University of Tübingen, Reutlingen, Germany
| | - Albert Braeuning
- Department of Food Safety, German Federal Institute for Risk Assessment, Berlin, Germany
| | - Tewes Tralau
- Department of Pesticides Safety, German Federal Institute for Risk Assessment, Berlin, Germany
| | - Philip Marx-Stoelting
- Department of Pesticides Safety, German Federal Institute for Risk Assessment, Berlin, Germany.
| |
Collapse
|
2
|
Murphy C, Jennings P, Wilmes A. Transcriptomic profile of human iPSC-derived podocyte-like cells exposed to a panel of xenobiotics. Toxicol In Vitro 2024; 97:105804. [PMID: 38447685 DOI: 10.1016/j.tiv.2024.105804] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Revised: 02/27/2024] [Accepted: 02/28/2024] [Indexed: 03/08/2024]
Abstract
Podocytes play a critical role in the formation and maintenance of the glomerular filtration barrier and injury to these cells can lead to a breakdown of the glomerular barrier causing permanent damage leading to progressive chronic kidney disease. Matured podocytes have little proliferative potential, which makes them critical cells from a health perspective, but also challenging cells to maintain in vitro. Differentiating podocyte-like cells from induced pluripotent stem cells (iPSC) provides a novel and continuous source of cells. Here, we investigated the effect of a 24-h exposure to eight compounds, including the known glomerular toxins doxorubicin and pamidronate, on transcriptomic alterations in iPSC derived podocytes. Doxorubicin (50 nM), pamidronate (50 μM), sodium arsenite (10 μM), and cyclosporine A (15 μM) had a strong impact on the transcriptome, gentamicin (450 μg/ml), lead chloride (15 μM) and valproic acid (500 μM) had a mild impact and busulfan (50 μM) exhibited no impact. Gene alterations and pathways analysis provided mechanistic insight for example, doxorubicin exposure affected the p53 pathway and dedifferentiation, pamidronate activated several pathways including HIF1alpha and sodium arsenite up-regulated oxidative stress and metal responses. The results demonstrate the applicability of iPSC derived podocytes for toxicological and mechanistic investigations.
Collapse
Affiliation(s)
- Cormac Murphy
- Division of Molecular and Computational Toxicology, Department of Chemistry and Pharmaceutical Sciences, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands; Amsterdam Institute of Molecular and Life Sciences (AIMMS), Faculty of Science, Vrije Universiteit, Amsterdam, the Netherlands
| | - Paul Jennings
- Division of Molecular and Computational Toxicology, Department of Chemistry and Pharmaceutical Sciences, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands; Amsterdam Institute of Molecular and Life Sciences (AIMMS), Faculty of Science, Vrije Universiteit, Amsterdam, the Netherlands.
| | - Anja Wilmes
- Division of Molecular and Computational Toxicology, Department of Chemistry and Pharmaceutical Sciences, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands; Amsterdam Institute of Molecular and Life Sciences (AIMMS), Faculty of Science, Vrije Universiteit, Amsterdam, the Netherlands.
| |
Collapse
|
3
|
Yu P, Zhu H, Bosholm CC, Beiner D, Duan Z, Shetty AK, Mou SS, Kramer PA, Barroso LF, Liu H, Cheng K, Ihnat M, Gorris MA, Aloi JA, Woldemichael JA, Bleyer A, Zhang Y. Precision nephrotoxicity testing using 3D in vitro models. Cell Biosci 2023; 13:231. [PMID: 38129901 PMCID: PMC10740310 DOI: 10.1186/s13578-023-01187-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Accepted: 12/15/2023] [Indexed: 12/23/2023] Open
Abstract
Nephrotoxicity is a significant concern during the development of new drugs or when assessing the safety of chemicals in consumer products. Traditional methods for testing nephrotoxicity involve animal models or 2D in vitro cell cultures, the latter of which lack the complexity and functionality of the human kidney. 3D in vitro models are created by culturing human primary kidney cells derived from urine in a 3D microenvironment that mimics the fluid shear stresses of the kidney. Thus, 3D in vitro models provide more accurate and reliable predictions of human nephrotoxicity compared to existing 2D models. In this review, we focus on precision nephrotoxicity testing using 3D in vitro models with human autologous urine-derived kidney cells as a promising approach for evaluating drug safety.
Collapse
Affiliation(s)
- Pengfei Yu
- Wake Forest Institute for Regenerative Medicine, Wake Forest University Health Sciences, Winston-Salem, NC, USA
- The Fourth Department of Liver Disease, Beijing You An Hospital, Capital Medical University, Beijing, China
| | - Hainan Zhu
- Wake Forest Institute for Regenerative Medicine, Wake Forest University Health Sciences, Winston-Salem, NC, USA
| | - Carol Christine Bosholm
- Wake Forest Institute for Regenerative Medicine, Wake Forest University Health Sciences, Winston-Salem, NC, USA
| | - Daniella Beiner
- Wake Forest Institute for Regenerative Medicine, Wake Forest University Health Sciences, Winston-Salem, NC, USA
| | - Zhongping Duan
- The Fourth Department of Liver Disease, Beijing You An Hospital, Capital Medical University, Beijing, China
| | - Avinash K Shetty
- Department of Pediatrics, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Steve S Mou
- Department of Anesthesiology and Pediatrics, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Philip Adam Kramer
- Department of Internal Medicine, Section on Gerontology and Geriatrics, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Luis F Barroso
- Internal Medicine/Infectious Diseases, Wake Forest University Health Sciences, Winston-Salem, NC, USA
| | - Hongbing Liu
- Department of Pediatrics and The Tulane Hypertension and Renal Center of Excellence, Tulane University School of Medicine, Tulane Avenue, New Orleans, LA, USA
| | - Kun Cheng
- Division of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Missouri-Kansas City, 2464 Charlotte Street, Kansas City, MO, 64108, USA
| | - Michael Ihnat
- Department of Pharmaceutical Sciences, University of Oklahoma College of Pharmacy, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Matthew A Gorris
- Division of Endocrinology and Metabolism at Wake Forest Baptist Health, Winston-Salem, NC, USA
| | - Joseph A Aloi
- Division of Endocrinology and Metabolism at Wake Forest Baptist Health, Winston-Salem, NC, USA
| | - Jobira A Woldemichael
- Division of Nephrology, Wake Forest University Health Sciences, Winston-Salem, NC, USA
| | - Anthony Bleyer
- Division of Nephrology, Wake Forest University Health Sciences, Winston-Salem, NC, USA
| | - Yuanyuan Zhang
- Wake Forest Institute for Regenerative Medicine, Wake Forest University Health Sciences, Winston-Salem, NC, USA.
| |
Collapse
|
4
|
Merrick BA, Martin NP, Brooks AM, Foley JF, Dunlap PE, Ramaiahgari S, Fannin RD, Gerrish KE. Insights into Repeated Renal Injury Using RNA-Seq with Two New RPTEC Cell Lines. Int J Mol Sci 2023; 24:14228. [PMID: 37762531 PMCID: PMC10531624 DOI: 10.3390/ijms241814228] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Revised: 09/06/2023] [Accepted: 09/09/2023] [Indexed: 09/29/2023] Open
Abstract
Renal proximal tubule epithelial cells (RPTECs) are a primary site for kidney injury. We created two RPTEC lines from CD-1 mice immortalized with hTERT (human telomerase reverse transcriptase) or SV40 LgT antigen (Simian Virus 40 Large T antigen). Our hypothesis was that low-level, repeated exposure to subcytotoxic levels of 0.25-2.5 μM cisplatin (CisPt) or 12.5-100 μM aflatoxin B1 (AFB1) would activate distinctive genes and pathways in these two differently immortalized cell lines. RNA-seq showed only LgT cells responded to AFB1 with 1139 differentially expressed genes (DEGs) at 72 h. The data suggested that AFB1 had direct nephrotoxic properties on the LgT cells. However, both the cell lines responded to 2.5 μM CisPt from 3 to 96 h expressing 2000-5000 total DEGs. For CisPt, the findings indicated a coordinated transcriptional program of injury signals and repair from the expression of immune receptors with cytokine and chemokine secretion for leukocyte recruitment; robust expression of synaptic and substrate adhesion molecules (SAMs) facilitating the expression of neural and hormonal receptors, ion channels/transporters, and trophic factors; and the expression of nephrogenesis transcription factors. Pathway analysis supported the concept of a renal repair transcriptome. In summary, these cell lines provide in vitro models for the improved understanding of repeated renal injury and repair mechanisms. High-throughput screening against toxicant libraries should provide a wider perspective of their capabilities in nephrotoxicity.
Collapse
Affiliation(s)
- B. Alex Merrick
- Mechanistic Toxicology Branch, Division of Translational Toxicology, National Institute of Environmental Health Sciences, Research Triangle Park, NC 27709, USA; (J.F.F.); (P.E.D.); (S.R.)
| | - Negin P. Martin
- Viral Vector Core, Neurobiology Laboratory, Division of Intramural Research, National Institute of Environmental Health Sciences, Research Triangle Park, NC 27709, USA;
| | - Ashley M. Brooks
- Biostatistics and Computational Biology Branch, Integrative Bioinformatics Support Group, Division of Intramural Research, National Institute of Environmental Health Sciences, Research Triangle Park, NC 27709, USA;
| | - Julie F. Foley
- Mechanistic Toxicology Branch, Division of Translational Toxicology, National Institute of Environmental Health Sciences, Research Triangle Park, NC 27709, USA; (J.F.F.); (P.E.D.); (S.R.)
| | - Paul E. Dunlap
- Mechanistic Toxicology Branch, Division of Translational Toxicology, National Institute of Environmental Health Sciences, Research Triangle Park, NC 27709, USA; (J.F.F.); (P.E.D.); (S.R.)
| | - Sreenivasa Ramaiahgari
- Mechanistic Toxicology Branch, Division of Translational Toxicology, National Institute of Environmental Health Sciences, Research Triangle Park, NC 27709, USA; (J.F.F.); (P.E.D.); (S.R.)
| | - Rick D. Fannin
- Molecular Genomics Core Laboratory, Division of Intramural Research, National Institute of Environmental Health Sciences, Research Triangle Park, NC 27709, USA; (R.D.F.)
| | - Kevin E. Gerrish
- Molecular Genomics Core Laboratory, Division of Intramural Research, National Institute of Environmental Health Sciences, Research Triangle Park, NC 27709, USA; (R.D.F.)
| |
Collapse
|
5
|
Bittenbinder MA, Capinha L, Da Costa Pereira D, Slagboom J, van de Velde B, Casewell NR, Jennings P, Kool J, Vonk FJ. Development of a high-throughput in vitro screening method for the assessment of cell-damaging activities of snake venoms. PLoS Negl Trop Dis 2023; 17:e0011564. [PMID: 37590328 PMCID: PMC10465002 DOI: 10.1371/journal.pntd.0011564] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Revised: 08/29/2023] [Accepted: 07/31/2023] [Indexed: 08/19/2023] Open
Abstract
Snakebite envenoming is a globally important public health issue that has devastating consequences on human health and well-being, with annual mortality rates between 81,000 and 138,000. Snake venoms may cause different pathological effects by altering normal physiological processes such as nervous transfer and blood coagulation. In addition, snake venoms can cause severe (local) tissue damage that may result in life-long morbidities, with current estimates pointing towards an additional 450,000 individuals that suffer from permanent disabilities such as amputations, contractions and blindness. Despite such high morbidity rates, research to date has been mainly focusing on neurotoxic and haemotoxic effects of snake venoms and considerably less on venom-induced tissue damage. The molecular mechanisms underlaying this pathology include membrane disruption and extracellular matrix degradation. This research describes methods used to study the (molecular) mechanisms underlaying venom-induced cell- and tissue damage. A selection of cellular bioassays and fluorescent microscopy were used to study cell-damaging activities of snake venoms in multi-well plates, using both crude and fractionated venoms. A panel of 10 representative medically relevant snake species was used, which cover a large part of the geographical regions most heavily affected by snakebite. The study comprises both morphological data as well as quantitative data on cell metabolism and viability, which were measured over time. Based on this data, a distinction could be made in the ways by which viper and elapid venoms exert their effects on cells. We further made an effort to characterise the bioactive compounds causing these effects, using a combination of liquid chromatography methods followed by bioassaying and protein identification using proteomics. The outcomes of this study might prove valuable for better understanding venom-induced cell- and tissue-damaging pathologies and could be used in the process of developing and improving snakebite treatments.
Collapse
Affiliation(s)
- Matyas A. Bittenbinder
- Naturalis Biodiversity Center, Leiden, The Netherlands
- AIMMS, Division of BioAnalytical Chemistry, Department of Chemistry and Pharmaceutical Sciences, Faculty of Sciences, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Centre for Analytical Sciences Amsterdam (CASA), Amsterdam, The Netherlands
| | - Liliana Capinha
- Division of Molecular and Computational Toxicology, Department of Chemistry and Pharmaceutical Sciences, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Daniel Da Costa Pereira
- Division of Molecular and Computational Toxicology, Department of Chemistry and Pharmaceutical Sciences, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Julien Slagboom
- AIMMS, Division of BioAnalytical Chemistry, Department of Chemistry and Pharmaceutical Sciences, Faculty of Sciences, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Centre for Analytical Sciences Amsterdam (CASA), Amsterdam, The Netherlands
| | - Bas van de Velde
- AIMMS, Division of BioAnalytical Chemistry, Department of Chemistry and Pharmaceutical Sciences, Faculty of Sciences, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Centre for Analytical Sciences Amsterdam (CASA), Amsterdam, The Netherlands
| | - Nicholas R. Casewell
- Centre for Snakebite Research & Interventions, Liverpool School of Tropical Medicine, Liverpool, United Kingdom
| | - Paul Jennings
- Division of Molecular and Computational Toxicology, Department of Chemistry and Pharmaceutical Sciences, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Jeroen Kool
- AIMMS, Division of BioAnalytical Chemistry, Department of Chemistry and Pharmaceutical Sciences, Faculty of Sciences, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Centre for Analytical Sciences Amsterdam (CASA), Amsterdam, The Netherlands
| | - Freek J. Vonk
- Naturalis Biodiversity Center, Leiden, The Netherlands
- AIMMS, Division of BioAnalytical Chemistry, Department of Chemistry and Pharmaceutical Sciences, Faculty of Sciences, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Centre for Analytical Sciences Amsterdam (CASA), Amsterdam, The Netherlands
| |
Collapse
|
6
|
Jennings P, Carta G, Singh P, da Costa Pereira D, Feher A, Dinnyes A, Exner TE, Wilmes A. Capturing time-dependent activation of genes and stress-response pathways using transcriptomics in iPSC-derived renal proximal tubule cells. Cell Biol Toxicol 2023; 39:1773-1793. [PMID: 36586010 PMCID: PMC10425493 DOI: 10.1007/s10565-022-09783-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Accepted: 12/06/2022] [Indexed: 01/01/2023]
Abstract
Transcriptomic analysis is a powerful method in the utilization of New Approach Methods (NAMs) for identifying mechanisms of toxicity and application to hazard characterization. With this regard, mapping toxicological events to time of exposure would be helpful to characterize early events. Here, we investigated time-dependent changes in gene expression levels in iPSC-derived renal proximal tubular-like cells (PTL) treated with five diverse compounds using TempO-Seq transcriptomics with the aims to evaluate the application of PTL for toxicity prediction and to report on temporal effects for the activation of cellular stress response pathways. PTL were treated with either 50 μM amiodarone, 10 μM sodium arsenate, 5 nM rotenone, or 300 nM tunicamycin over a temporal time course between 1 and 24 h. The TGFβ-type I receptor kinase inhibitor GW788388 (1 μM) was used as a negative control. Pathway analysis revealed the induction of key stress-response pathways, including Nrf2 oxidative stress response, unfolding protein response, and metal stress response. Early response genes per pathway were identified much earlier than 24 h and included HMOX1, ATF3, DDIT3, and several MT1 isotypes. GW788388 did not induce any genes within the stress response pathways above, but showed deregulation of genes involved in TGFβ inhibition, including downregulation of CYP24A1 and SERPINE1 and upregulation of WT1. This study highlights the application of iPSC-derived renal cells for prediction of cellular toxicity and sheds new light on the temporal and early effects of key genes that are involved in cellular stress response pathways.
Collapse
Affiliation(s)
- Paul Jennings
- Division of Molecular and Computational Toxicology, Chemistry and Pharmaceutical Sciences, AIMMS, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Giada Carta
- Division of Molecular and Computational Toxicology, Chemistry and Pharmaceutical Sciences, AIMMS, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Pranika Singh
- Edelweiss Connect GmbH, Technology Park Basel, Hochbergerstrasse 60C, 4057, Basel, Switzerland
- Division of Molecular and Systems Toxicology, Department of Pharmaceutical Sciences, University of Basel, Klingelbergstrasse 50, 4056, Basel, Switzerland
| | - Daniel da Costa Pereira
- Division of Molecular and Computational Toxicology, Chemistry and Pharmaceutical Sciences, AIMMS, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Anita Feher
- BioTalentum Ltd, Aulich Lajos Street 26, Gödöllő, 2100, Hungary
| | - Andras Dinnyes
- BioTalentum Ltd, Aulich Lajos Street 26, Gödöllő, 2100, Hungary
- HCEMM-USZ Stem Cell Research Group, Hungarian Centre of Excellence for Molecular Medicine, Szeged, 6723, Hungary
| | - Thomas E Exner
- Seven Past Nine d.o.o., Hribljane 10, 1380, Cerknica, Slovenia
| | - Anja Wilmes
- Division of Molecular and Computational Toxicology, Chemistry and Pharmaceutical Sciences, AIMMS, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands.
| |
Collapse
|
7
|
Capinha L, Jennings P, Commandeur JNM. Exposure to Cis- and Trans-regioisomers of S-(1,2-dichlorovinyl)-L-cysteine and S-(1,2-dichlorovinyl)-glutathione result in quantitatively and qualitatively different cellular effects in RPTEC/TERT1 cells. Toxicol Lett 2023:S0378-4274(23)00205-9. [PMID: 37353095 DOI: 10.1016/j.toxlet.2023.06.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Revised: 06/05/2023] [Accepted: 06/20/2023] [Indexed: 06/25/2023]
Abstract
Bioactivation of trichloroethylene (TCE) via glutathione conjugation is associated with several adverse effects in the kidney and other extrahepatic tissues. Of the three regioisomeric conjugates formed, S-(1,2-trans-dichlorovinyl)-glutathione (1,2-trans-DCVG), S-(1,2-cis-dichlorovinyl)-glutathione and S-(2,2-dichlorovinyl)-glutathione, only 1,2-trans-DCVG and its corresponding cysteine-conjugate, 1,2-trans-DCVC, have been subject to extensive mechanistic studies. In the present study, the metabolism and cellular effects of 1,2-cis-DCVG, the major regioisomer formed by rat liver fractions, and 1,2-cis-DCVC were investigated for the first time using RPTEC/TERT1-cells as in vitro renal model. In contrast to 1,2-trans-DCVG/C, the cis-regioisomers showed minimal effects on cell viability and mitochondrial respiration. Transcriptomics analysis showed that both 1,2-cis-DCVC and 1,2-trans-DCVC caused Nrf2-mediated antioxidant responses, with 3µM as lowest effective concentration. An ATF4-mediated integrated stress response and p53-mediated responses were observed starting from 30µM for 1,2-trans-DCVC and 125µM for 1,2-cis-DCVC. Comparison of the metabolism of the DCVG regioisomers by LC/MS showed comparable rates of processing to their corresponding DCVC. No detectable N-acetylation was observed in RPTEC/TERT1 cells. Instead, N-glutamylation of DCVC to form N-γ-glutamyl-S-(dichlorovinyl)-L-cysteine was identified as a novel route of metabolism. The results suggest that 1,2-cis-DCVC may be of less toxicological concern for humans than 1,2-trans-DCVC, considering its lower intrinsic toxicity and lower rate of formation by human liver fractions.
Collapse
Affiliation(s)
- Liliana Capinha
- Division of Molecular and Computational Toxicology, Amsterdam Institute for Molecules, Medicines and Systems (AIMMS), Faculty of Sciences, Vrije Universiteit Amsterdam, De Boelelaan 1108, 1081 HZ Amsterdam, the Netherlands
| | - Paul Jennings
- Division of Molecular and Computational Toxicology, Amsterdam Institute for Molecules, Medicines and Systems (AIMMS), Faculty of Sciences, Vrije Universiteit Amsterdam, De Boelelaan 1108, 1081 HZ Amsterdam, the Netherlands.
| | - Jan N M Commandeur
- Division of Molecular and Computational Toxicology, Amsterdam Institute for Molecules, Medicines and Systems (AIMMS), Faculty of Sciences, Vrije Universiteit Amsterdam, De Boelelaan 1108, 1081 HZ Amsterdam, the Netherlands.
| |
Collapse
|
8
|
Hyperoside attenuates Cd-induced kidney injury via inhibiting NLRP3 inflammasome activation and ROS/MAPK/NF-κB signaling pathway in vivo and in vitro. Food Chem Toxicol 2023; 172:113601. [PMID: 36610472 DOI: 10.1016/j.fct.2023.113601] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2022] [Revised: 12/31/2022] [Accepted: 01/03/2023] [Indexed: 01/06/2023]
Abstract
Cadmium accumulates in the kidney and causes inflammation. The NLRP3 inflammasome has been linked to the pathogenesis of inflammation. Hyperoside (HYP) possesses potent nephroprotective properties against of kidney injury. This study aimed to research the effects and related mechanism of HYP on Cd-induced kidney damage. Wide-type and NLRP3-/- mice were used to determine the role of NLRP3 inflammasome in Cd-induced renal dysfunction. Female C57BL/6 were treated with Cd (50 m,g/L) and HYP (25, 50 mg/kg) for 12 weeks. In vitro experiments, the human renal proximal-tubule epithelial cells (RPTEC/TERT1) were pretreated with HYP (50-200 μM) before exposure to Cd. NLRP3 deficiency attenuated Cd-induced NLRP3 activation, inflammation and kidney injury in mice. HYP treatment significantly alleviated Cd-induced kidney injury by decreasing indexes of kidney function, reducing pro-inflammatory cytokines release, decreasing ROS production and suppressing NLRP3 inflammasome activation. Moreover, treatment with siRNA targeting NLRP3 blocked the anti-inflammatory protective effect of HYP in Cd-treated cells. Additionally, HYP markedly inhibited Cd-induced MAPK/NF-κB pathway stimulation in vitro and in vivo. The findings indicated HYP conferred protection against Cd-induced kidney inflammation via suppression of NLRP3 inflammasome mediated by ROS/MAPK/NF-κB signaling. Our results thus support the notion of developing HYP as promising therapeutic candidate for Cd-induced kidney injury.
Collapse
|
9
|
Transcriptomic-based evaluation of trichloroethylene glutathione and cysteine conjugates demonstrate phenotype-dependent stress responses in a panel of human in vitro models. Arch Toxicol 2023; 97:523-545. [PMID: 36576512 PMCID: PMC9859926 DOI: 10.1007/s00204-022-03436-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Accepted: 12/14/2022] [Indexed: 12/29/2022]
Abstract
Environmental or occupational exposure of humans to trichloroethylene (TCE) has been associated with different extrahepatic toxic effects, including nephrotoxicity and neurotoxicity. Bioactivation of TCE via the glutathione (GSH) conjugation pathway has been proposed as underlying mechanism, although only few mechanistic studies have used cell models of human origin. In this study, six human derived cell models were evaluated as in vitro models representing potential target tissues of TCE-conjugates: RPTEC/TERT1 (kidney), HepaRG (liver), HUVEC/TERT2 (vascular endothelial), LUHMES (neuronal, dopaminergic), human induced pluripotent stem cells (hiPSC) derived peripheral neurons (UKN5) and hiPSC-derived differentiated brain cortical cultures containing all subtypes of neurons and astrocytes (BCC42). A high throughput transcriptomic screening, utilizing mRNA templated oligo-sequencing (TempO-Seq), was used to study transcriptomic effects after exposure to TCE-conjugates. Cells were exposed to a wide range of concentrations of S-(1,2-trans-dichlorovinyl)glutathione (1,2-DCVG), S-(1,2-trans-dichlorovinyl)-L-cysteine (1,2-DCVC), S-(2,2-dichlorovinyl)glutathione (2,2-DCVG), and S-(2,2-dichlorovinyl)-L-cysteine (2,2-DCVC). 1,2-DCVC caused stress responses belonging to the Nrf2 pathway and Unfolded protein response in all the tested models but to different extents. The renal model was the most sensitive model to both 1,2-DCVC and 1,2-DCVG, with an early Nrf2-response at 3 µM and hundreds of differentially expressed genes at higher concentrations. Exposure to 2,2-DCVG and 2,2-DCVC also resulted in the upregulation of Nrf2 pathway genes in RPTEC/TERT1 although at higher concentrations. Of the three neuronal models, both the LUHMES and BCC42 showed significant Nrf2-responses and at higher concentration UPR-responses, supporting recent hypotheses that 1,2-DCVC may be involved in neurotoxic effects of TCE. The cell models with the highest expression of γ-glutamyltransferase (GGT) enzymes, showed cellular responses to both 1,2-DCVG and 1,2-DCVC. Little to no effects were found in the neuronal models from 1,2-DCVG exposure due to their low GGT-expression. This study expands our knowledge on tissue specificity of TCE S-conjugates and emphasizes the value of human cell models together with transcriptomics for such mechanistic studies.
Collapse
|
10
|
Vidal Yucha SE, Quackenbush D, Chu T, Lo F, Sutherland JJ, Kuzu G, Roberts C, Luna F, Barnes SW, Walker J, Kuss P. "3D, human renal proximal tubule (RPTEC-TERT1) organoids 'tubuloids' for translatable evaluation of nephrotoxins in high-throughput". PLoS One 2022; 17:e0277937. [PMID: 36409750 PMCID: PMC9678317 DOI: 10.1371/journal.pone.0277937] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Accepted: 11/07/2022] [Indexed: 11/22/2022] Open
Abstract
The importance of human cell-based in vitro tools to drug development that are robust, accurate, and predictive cannot be understated. There has been significant effort in recent years to develop such platforms, with increased interest in 3D models that can recapitulate key aspects of biology that 2D models might not be able to deliver. We describe the development of a 3D human cell-based in vitro assay for the investigation of nephrotoxicity, using RPTEC-TERT1 cells. These RPTEC-TERT1 proximal tubule organoids 'tubuloids' demonstrate marked differences in physiologically relevant morphology compared to 2D monolayer cells, increased sensitivity to nephrotoxins observable via secreted protein, and with a higher degree of similarity to native human kidney tissue. Finally, tubuloids incubated with nephrotoxins demonstrate altered Na+/K+-ATPase signal intensity, a potential avenue for a high-throughput, translatable nephrotoxicity assay.
Collapse
Affiliation(s)
- Sarah E. Vidal Yucha
- Novartis Institutes for BioMedical Research-San Diego, La Jolla, CA, United States of America
- * E-mail:
| | - Doug Quackenbush
- Novartis Institutes for BioMedical Research-San Diego, La Jolla, CA, United States of America
| | - Tiffany Chu
- Novartis Institutes for BioMedical Research-San Diego, La Jolla, CA, United States of America
| | - Frederick Lo
- Novartis Institutes for BioMedical Research-San Diego, La Jolla, CA, United States of America
| | - Jeffrey J. Sutherland
- Novartis Institutes for BioMedical Research-Cambridge, Cambridge, MA, United States of America
| | - Guray Kuzu
- Novartis Institutes for BioMedical Research-San Diego, La Jolla, CA, United States of America
| | - Christopher Roberts
- Novartis Institutes for BioMedical Research-San Diego, La Jolla, CA, United States of America
| | - Fabio Luna
- Novartis Institutes for BioMedical Research-San Diego, La Jolla, CA, United States of America
| | - S. Whitney Barnes
- Novartis Institutes for BioMedical Research-San Diego, La Jolla, CA, United States of America
| | - John Walker
- Novartis Institutes for BioMedical Research-San Diego, La Jolla, CA, United States of America
| | - Pia Kuss
- Novartis Institutes for BioMedical Research-San Diego, La Jolla, CA, United States of America
| |
Collapse
|
11
|
Chen Y, Lu S, Zhang Y, Chen B, Zhou H, Jiang H. Examination of the emerging role of transporters in the assessment of nephrotoxicity. Expert Opin Drug Metab Toxicol 2022; 18:787-804. [PMID: 36420583 DOI: 10.1080/17425255.2022.2151892] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
INTRODUCTION The kidney is vulnerable to various injuries based on its function in the elimination of many xenobiotics, endogenous substances and metabolites. Since transporters are critical for the renal elimination of those substances, it is urgent to understand the emerging role of transporters in nephrotoxicity. AREAS COVERED This review summarizes the contribution of major renal transporters to nephrotoxicity induced by some drugs or toxins; addresses the role of transporter-mediated endogenous metabolic disturbances in nephrotoxicity; and discusses the advantages and disadvantages of in vitro models based on transporter expression and function. EXPERT OPINION Due to the crucial role of transporters in the renal disposition of xenobiotics and endogenous substances, it is necessary to further elucidate their renal transport mechanisms and pay more attention to the underlying relationship between the transport of endogenous substances and nephrotoxicity. Considering the species differences in the expression and function of transporters, and the low expression of transporters in general cell models, in vitro humanized models, such as humanized 3D organoids, shows significant promise in nephrotoxicity prediction and mechanism study.
Collapse
Affiliation(s)
- Yujia Chen
- Institute of Drug Metabolism and Pharmaceutical Analysis, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, P.R. China
| | - Shuanghui Lu
- Institute of Drug Metabolism and Pharmaceutical Analysis, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, P.R. China
| | - Yingqiong Zhang
- Institute of Drug Metabolism and Pharmaceutical Analysis, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, P.R. China.,Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, P.R. China
| | - Binxin Chen
- Institute of Drug Metabolism and Pharmaceutical Analysis, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, P.R. China
| | - Hui Zhou
- Institute of Drug Metabolism and Pharmaceutical Analysis, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, P.R. China.,Jinhua Institute of Zhejiang University, Jinhua, P.R. China
| | - Huidi Jiang
- Institute of Drug Metabolism and Pharmaceutical Analysis, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, P.R. China.,Jinhua Institute of Zhejiang University, Jinhua, P.R. China
| |
Collapse
|
12
|
Bejoy J, Qian ES, Woodard LE. Tissue Culture Models of AKI: From Tubule Cells to Human Kidney Organoids. J Am Soc Nephrol 2022; 33:487-501. [PMID: 35031569 PMCID: PMC8975068 DOI: 10.1681/asn.2021050693] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
Abstract
AKI affects approximately 13.3 million people around the world each year, causing CKD and/or mortality. The mammalian kidney cannot generate new nephrons after postnatal renal damage and regenerative therapies for AKI are not available. Human kidney tissue culture systems can complement animal models of AKI and/or address some of their limitations. Donor-derived somatic cells, such as renal tubule epithelial cells or cell lines (RPTEC/hTERT, ciPTEC, HK-2, Nki-2, and CIHP-1), have been used for decades to permit drug toxicity screening and studies into potential AKI mechanisms. However, tubule cell lines do not fully recapitulate tubular epithelial cell properties in situ when grown under classic tissue culture conditions. Improving tissue culture models of AKI would increase our understanding of the mechanisms, leading to new therapeutics. Human pluripotent stem cells (hPSCs) can be differentiated into kidney organoids and various renal cell types. Injury to human kidney organoids results in renal cell-type crosstalk and upregulation of kidney injury biomarkers that are difficult to induce in primary tubule cell cultures. However, current protocols produce kidney organoids that are not mature and contain off-target cell types. Promising bioengineering techniques, such as bioprinting and "kidney-on-a-chip" methods, as applied to kidney nephrotoxicity modeling advantages and limitations are discussed. This review explores the mechanisms and detection of AKI in tissue culture, with an emphasis on bioengineered approaches such as human kidney organoid models.
Collapse
Affiliation(s)
- Julie Bejoy
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Eddie S. Qian
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Lauren E. Woodard
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
- Department of Veterans Affairs, Tennessee Valley Healthcare System, Nashville, Tennessee
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee
| |
Collapse
|
13
|
Singh P, Chandrasekaran V, Hardy B, Wilmes A, Jennings P, Exner TE. Temporal transcriptomic alterations of cadmium exposed human iPSC-derived renal proximal tubule-like cells. Toxicol In Vitro 2021; 76:105229. [PMID: 34352368 DOI: 10.1016/j.tiv.2021.105229] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Revised: 06/25/2021] [Accepted: 07/26/2021] [Indexed: 12/12/2022]
Abstract
Cadmium is a well-studied environmental pollutant where the kidney and particularly the proximal tubule cells are especially sensitive as they are exposed to higher concentrations of cadmium than other tissues. Here we investigated the temporal transcriptomic alterations (TempO-Seq) of human induced pluripotent stem cell (iPSC)-derived renal proximal tubule-like (PTL) cells exposed to 5 μM cadmium chloride for 1, 2, 4, 8, 12, 16, 20, 24, 72 and 168 h. There was an early activation (within 4 h) of the metal and oxidative stress responses (metal-responsive transcription factor-1 (MTF1) and nuclear factor erythroid-2-related factor 2 (Nrf2) genes). The Nrf2 response returned to baseline within 24 h. The Activator Protein 1 (AP-1) regulated genes HSPA6 and FOSL-1 followed the Nrf2 time course. While the MTF1 genes also spiked at 4 h, they remained strongly elevated over the entire exposure period. The data and cell culture model utilised will be useful in further research aimed at the refinement of safe human exposure limits for cadmium, other metals and their mixtures.
Collapse
Affiliation(s)
- Pranika Singh
- Edelweiss Connect GmbH, Technology Park Basel, Hochbergerstrasse 60C, 4057 Basel, Switzerland; Division of Molecular and Systems Toxicology, Department of Pharmaceutical Sciences, University of Basel, Klingelbergstrasse 50, 4056 Basel, Switzerland
| | - Vidya Chandrasekaran
- Division of Molecular and Computational Toxicology, Chemistry and Pharmaceutical Sciences, AIMMS, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | - Barry Hardy
- Edelweiss Connect GmbH, Technology Park Basel, Hochbergerstrasse 60C, 4057 Basel, Switzerland
| | - Anja Wilmes
- Division of Molecular and Computational Toxicology, Chemistry and Pharmaceutical Sciences, AIMMS, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | - Paul Jennings
- Division of Molecular and Computational Toxicology, Chemistry and Pharmaceutical Sciences, AIMMS, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands.
| | - Thomas E Exner
- Seven Past Nine d.o.o., Hribljane 10, 1380 Cerknica, Slovenia.
| |
Collapse
|
14
|
Irvine AR, van Berlo D, Shekhani R, Masereeuw R. A systematic review of in vitro models of drug-induced kidney injury. CURRENT OPINION IN TOXICOLOGY 2021. [DOI: 10.1016/j.cotox.2021.06.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
15
|
Keele GR, Prokop JW, He H, Holl K, Littrell J, Deal AW, Kim Y, Kyle PB, Attipoe E, Johnson AC, Uhl KL, Sirpilla OL, Jahanbakhsh S, Robinson M, Levy S, Valdar W, Garrett MR, Solberg Woods LC. Sept8/SEPTIN8 involvement in cellular structure and kidney damage is identified by genetic mapping and a novel human tubule hypoxic model. Sci Rep 2021; 11:2071. [PMID: 33483609 PMCID: PMC7822875 DOI: 10.1038/s41598-021-81550-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Accepted: 01/05/2021] [Indexed: 01/29/2023] Open
Abstract
Chronic kidney disease (CKD), which can ultimately progress to kidney failure, is influenced by genetics and the environment. Genes identified in human genome wide association studies (GWAS) explain only a small proportion of the heritable variation and lack functional validation, indicating the need for additional model systems. Outbred heterogeneous stock (HS) rats have been used for genetic fine-mapping of complex traits, but have not previously been used for CKD traits. We performed GWAS for urinary protein excretion (UPE) and CKD related serum biochemistries in 245 male HS rats. Quantitative trait loci (QTL) were identified using a linear mixed effect model that tested for association with imputed genotypes. Candidate genes were identified using bioinformatics tools and targeted RNAseq followed by testing in a novel in vitro model of human tubule, hypoxia-induced damage. We identified two QTL for UPE and five for serum biochemistries. Protein modeling identified a missense variant within Septin 8 (Sept8) as a candidate for UPE. Sept8/SEPTIN8 expression increased in HS rats with elevated UPE and tubulointerstitial injury and in the in vitro hypoxia model. SEPTIN8 is detected within proximal tubule cells in human kidney samples and localizes with acetyl-alpha tubulin in the culture system. After hypoxia, SEPTIN8 staining becomes diffuse and appears to relocalize with actin. These data suggest a role of SEPTIN8 in cellular organization and structure in response to environmental stress. This study demonstrates that integration of a rat genetic model with an environmentally induced tubule damage system identifies Sept8/SEPTIN8 and informs novel aspects of the complex gene by environmental interactions contributing to CKD risk.
Collapse
Affiliation(s)
| | - Jeremy W Prokop
- HudsonAlpha Institute, Huntsville, AL, USA
- Department of Pediatrics and Human Development, Department of Pharmacology, Michigan State University, Grand Rapids, MI, USA
| | - Hong He
- Departments of Pediatrics and Physiology, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Katie Holl
- Departments of Pediatrics and Physiology, Medical College of Wisconsin, Milwaukee, WI, USA
| | - John Littrell
- Departments of Pediatrics and Physiology, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Aaron W Deal
- Department of Internal Medicine, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Yunjung Kim
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Patrick B Kyle
- Department of Pharmacology, Medicine (Nephrology), Pediatrics (Genetics), University of Mississippi Medical Center, Jackson, MS, USA
- Department of Pathology, University of Mississippi Medical Center, Jackson, MS, USA
| | - Esinam Attipoe
- Department of Pharmacology, Medicine (Nephrology), Pediatrics (Genetics), University of Mississippi Medical Center, Jackson, MS, USA
| | - Ashley C Johnson
- Department of Pharmacology, Medicine (Nephrology), Pediatrics (Genetics), University of Mississippi Medical Center, Jackson, MS, USA
| | - Katie L Uhl
- Department of Pediatrics and Human Development, Department of Pharmacology, Michigan State University, Grand Rapids, MI, USA
| | - Olivia L Sirpilla
- Department of Pediatrics and Human Development, Department of Pharmacology, Michigan State University, Grand Rapids, MI, USA
| | - Seyedehameneh Jahanbakhsh
- Department of Pediatrics and Human Development, Department of Pharmacology, Michigan State University, Grand Rapids, MI, USA
| | | | - Shawn Levy
- HudsonAlpha Institute, Huntsville, AL, USA
| | - William Valdar
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Michael R Garrett
- Department of Pharmacology, Medicine (Nephrology), Pediatrics (Genetics), University of Mississippi Medical Center, Jackson, MS, USA
| | - Leah C Solberg Woods
- Department of Internal Medicine, Wake Forest School of Medicine, Winston-Salem, NC, USA.
| |
Collapse
|
16
|
Glykofridis IE, Knol JC, Balk JA, Westland D, Pham TV, Piersma SR, Lougheed SM, Derakhshan S, Veen P, Rooimans MA, van Mil SE, Böttger F, Poddighe PJ, van de Beek I, Drost J, Zwartkruis FJ, de Menezes RX, Meijers-Heijboer HE, Houweling AC, Jimenez CR, Wolthuis RM. Loss of FLCN-FNIP1/2 induces a non-canonical interferon response in human renal tubular epithelial cells. eLife 2021; 10:61630. [PMID: 33459596 PMCID: PMC7899648 DOI: 10.7554/elife.61630] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Accepted: 01/16/2021] [Indexed: 12/14/2022] Open
Abstract
Germline mutations in the Folliculin (FLCN) tumor suppressor gene cause Birt–Hogg–Dubé (BHD) syndrome, a rare autosomal dominant disorder predisposing carriers to kidney tumors. FLCN is a conserved, essential gene linked to diverse cellular processes but the mechanism by which FLCN prevents kidney cancer remains unknown. Here, we show that disrupting FLCN in human renal tubular epithelial cells (RPTEC/TERT1) activates TFE3, upregulating expression of its E-box targets, including RRAGD and GPNMB, without modifying mTORC1 activity. Surprisingly, the absence of FLCN or its binding partners FNIP1/FNIP2 induces interferon response genes independently of interferon. Mechanistically, FLCN loss promotes STAT2 recruitment to chromatin and slows cellular proliferation. Our integrated analysis identifies STAT1/2 signaling as a novel target of FLCN in renal cells and BHD tumors. STAT1/2 activation appears to counterbalance TFE3-directed hyper-proliferation and may influence immune responses. These findings shed light on unique roles of FLCN in human renal tumorigenesis and pinpoint candidate prognostic biomarkers.
Collapse
Affiliation(s)
- Iris E Glykofridis
- Amsterdam UMC, location VUmc, Vrije Universiteit Amsterdam, Clinical Genetics, Cancer Center Amsterdam, Amsterdam, Netherlands
| | - Jaco C Knol
- Amsterdam UMC, location VUmc, Vrije Universiteit Amsterdam, Medical Oncology, Cancer Center Amsterdam, Amsterdam, Netherlands
| | - Jesper A Balk
- Amsterdam UMC, location VUmc, Vrije Universiteit Amsterdam, Clinical Genetics, Cancer Center Amsterdam, Amsterdam, Netherlands
| | - Denise Westland
- University Medical Center Utrecht, Center for Molecular Medicine, Molecular Cancer Research, Universiteitsweg, Utrecht, Netherlands
| | - Thang V Pham
- Amsterdam UMC, location VUmc, Vrije Universiteit Amsterdam, Medical Oncology, Cancer Center Amsterdam, Amsterdam, Netherlands
| | - Sander R Piersma
- Amsterdam UMC, location VUmc, Vrije Universiteit Amsterdam, Medical Oncology, Cancer Center Amsterdam, Amsterdam, Netherlands
| | - Sinéad M Lougheed
- Amsterdam UMC, location VUmc, Vrije Universiteit Amsterdam, Medical Oncology, Cancer Center Amsterdam, Amsterdam, Netherlands
| | - Sepide Derakhshan
- Princess Máxima Center for Pediatric Oncology, Oncode Institute, Heidelberglaan, Utrecht, Netherlands
| | - Puck Veen
- Amsterdam UMC, location VUmc, Vrije Universiteit Amsterdam, Clinical Genetics, Cancer Center Amsterdam, Amsterdam, Netherlands
| | - Martin A Rooimans
- Amsterdam UMC, location VUmc, Vrije Universiteit Amsterdam, Clinical Genetics, Cancer Center Amsterdam, Amsterdam, Netherlands
| | - Saskia E van Mil
- Amsterdam UMC, location VUmc, Vrije Universiteit Amsterdam, Clinical Genetics, Cancer Center Amsterdam, Amsterdam, Netherlands
| | - Franziska Böttger
- Amsterdam UMC, location VUmc, Vrije Universiteit Amsterdam, Medical Oncology, Cancer Center Amsterdam, Amsterdam, Netherlands
| | - Pino J Poddighe
- Amsterdam UMC, location VUmc, Vrije Universiteit Amsterdam, Clinical Genetics, Amsterdam, Netherlands
| | - Irma van de Beek
- Amsterdam UMC, location VUmc, Vrije Universiteit Amsterdam, Clinical Genetics, Amsterdam, Netherlands
| | - Jarno Drost
- Princess Máxima Center for Pediatric Oncology, Oncode Institute, Heidelberglaan, Utrecht, Netherlands
| | - Fried Jt Zwartkruis
- University Medical Center Utrecht, Center for Molecular Medicine, Molecular Cancer Research, Universiteitsweg, Utrecht, Netherlands
| | | | - Hanne Ej Meijers-Heijboer
- Amsterdam UMC, location VUmc, Vrije Universiteit Amsterdam, Clinical Genetics, Cancer Center Amsterdam, Amsterdam, Netherlands
| | - Arjan C Houweling
- Amsterdam UMC, location VUmc, Vrije Universiteit Amsterdam, Clinical Genetics, Amsterdam, Netherlands
| | - Connie R Jimenez
- Amsterdam UMC, location VUmc, Vrije Universiteit Amsterdam, Medical Oncology, Cancer Center Amsterdam, Amsterdam, Netherlands
| | - Rob Mf Wolthuis
- Amsterdam UMC, location VUmc, Vrije Universiteit Amsterdam, Clinical Genetics, Cancer Center Amsterdam, Amsterdam, Netherlands
| |
Collapse
|
17
|
Mihevc M, Petreski T, Maver U, Bevc S. Renal proximal tubular epithelial cells: review of isolation, characterization, and culturing techniques. Mol Biol Rep 2020; 47:9865-9882. [PMID: 33170426 DOI: 10.1007/s11033-020-05977-4] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Accepted: 11/03/2020] [Indexed: 12/23/2022]
Abstract
The kidney is a complex organ, comprised primarily of glomerular, tubular, mesangial, and endothelial cells, and podocytes. The fact that renal cells are terminally differentiated at 34 weeks of gestation is the main obstacle in regeneration and treatment of acute kidney injury or chronic kidney disease. Furthermore, the number of chronic kidney disease patients is ever increasing and with it the medical community should aim to improve existing and develop new methods of renal replacement therapy. On the other hand, as polypharmacy is on the rise, thought should be given into developing new ways of testing drug safety. A possible way to tackle these issues is with isolation and culture of renal cells. Several protocols are currently described to isolate the desired cells, of which the most isolated are the proximal tubular epithelial cells. They play a major role in water homeostasis, acid-base control, reabsorption of compounds, and secretion of xenobiotics and endogenous metabolites. When exposed to ischemic, toxic, septic, or obstructive conditions their death results in what we clinically perceive as acute kidney injury. Additionally, due to renal cells' limited regenerative potential, the profibrotic environment inevitably leads to chronic kidney disease. In this review we will focus on human proximal tubular epithelial cells. We will cover human kidney culture models, cell sources, isolation, culture, immortalization, and characterization subdivided into morphological, phenotypical, and functional characterization.
Collapse
Affiliation(s)
- Matic Mihevc
- Department of Nephrology, Clinic for Internal Medicine, University Medical Centre Maribor, Ljubljanska ulica 5, 2000, Maribor, Slovenia
| | - Tadej Petreski
- Department of Nephrology, Clinic for Internal Medicine, University Medical Centre Maribor, Ljubljanska ulica 5, 2000, Maribor, Slovenia
- Faculty of Medicine, Institute of Biomedical Sciences, University of Maribor, Taborska ulica 8, 2000, Maribor, Slovenia
| | - Uroš Maver
- Faculty of Medicine, Institute of Biomedical Sciences, University of Maribor, Taborska ulica 8, 2000, Maribor, Slovenia.
- Department of Pharmacology, Faculty of Medicine, University of Maribor, Taborska ulica 8, 2000, Maribor, Slovenia.
| | - Sebastjan Bevc
- Department of Nephrology, Clinic for Internal Medicine, University Medical Centre Maribor, Ljubljanska ulica 5, 2000, Maribor, Slovenia.
- Department of Pharmacology, Faculty of Medicine, University of Maribor, Taborska ulica 8, 2000, Maribor, Slovenia.
| |
Collapse
|
18
|
Lin N, Zhou X, Geng X, Drewell C, Hübner J, Li Z, Zhang Y, Xue M, Marx U, Li B. Repeated dose multi-drug testing using a microfluidic chip-based coculture of human liver and kidney proximal tubules equivalents. Sci Rep 2020; 10:8879. [PMID: 32483208 PMCID: PMC7264205 DOI: 10.1038/s41598-020-65817-0] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2019] [Accepted: 04/15/2020] [Indexed: 11/28/2022] Open
Abstract
A microfluidic multi-organ chip emulates the tissue culture microenvironment, enables interconnection of organ equivalents and overcomes interspecies differences, making this technology a promising and powerful tool for preclinical drug screening. In this study, we established a microfluidic chip-based model that enabled non-contact cocultivation of liver spheroids and renal proximal tubule barriers in a connecting media circuit over 16 days. Meanwhile, a 14-day repeated-dose systemic administration of cyclosporine A (CsA) alone or in combination with rifampicin was performed. Toxicity profiles of the two different doses of CsA on different target organs could be discriminated and that concomitant treatment with rifampicin from day6 onwards decreased the CsA concentration and attenuated the toxicity compared with that after treatment with CsA for 14 consecutive days. The latter is manifested with the changes in cytotoxicity, cell viability and apoptosis, gene expression of metabolic enzymes and transporters, and noninvasive toxicity biomarkers. The on chip coculture of the liver and the proximal tubulus equivalents showed its potential as an effective and translational tool for repeated dose multi-drug toxicity screening in the preclinical stage of drug development.
Collapse
Affiliation(s)
- Ni Lin
- Key Laboratory of Beijing for Safety Evaluation of Drugs, National Center for Safety Evaluation of Drugs, National Institutes for Food and Drug Control, A8 Hongda Middle Street, Beijing Economic-Technological Development Area, Beijing, 100176, P. R. China.,Department of Pharmacology, Beijing Laboratory for Biomedical Detection Technology and Instrument, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China.,Beijing Institute for Drug Control, 25 Science Park Road, Changping District, Beijing, 102206, China
| | - Xiaobing Zhou
- Key Laboratory of Beijing for Safety Evaluation of Drugs, National Center for Safety Evaluation of Drugs, National Institutes for Food and Drug Control, A8 Hongda Middle Street, Beijing Economic-Technological Development Area, Beijing, 100176, P. R. China
| | - Xingchao Geng
- Key Laboratory of Beijing for Safety Evaluation of Drugs, National Center for Safety Evaluation of Drugs, National Institutes for Food and Drug Control, A8 Hongda Middle Street, Beijing Economic-Technological Development Area, Beijing, 100176, P. R. China
| | - Christopher Drewell
- Technische Universitaet Berlin, Institute of Biotechnology, Department Medical Biotechnology, Gustav-Meyer-Allee 25, 13355, Berlin, Germany
| | - Juliane Hübner
- Technische Universitaet Berlin, Institute of Biotechnology, Department Medical Biotechnology, Gustav-Meyer-Allee 25, 13355, Berlin, Germany
| | - Zuogang Li
- Key Laboratory of Beijing for Safety Evaluation of Drugs, National Center for Safety Evaluation of Drugs, National Institutes for Food and Drug Control, A8 Hongda Middle Street, Beijing Economic-Technological Development Area, Beijing, 100176, P. R. China
| | - Yingli Zhang
- Key Laboratory of Beijing for Safety Evaluation of Drugs, National Center for Safety Evaluation of Drugs, National Institutes for Food and Drug Control, A8 Hongda Middle Street, Beijing Economic-Technological Development Area, Beijing, 100176, P. R. China
| | - Ming Xue
- Department of Pharmacology, Beijing Laboratory for Biomedical Detection Technology and Instrument, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China.
| | - Uwe Marx
- TissUse GmbH, Oudenarder Strasse 16, 13347, Berlin, Germany.
| | - Bo Li
- National Institutes for Food and Drug Control, 31 Hua Tuo road, Daxing district, Beijing, 102629, China.
| |
Collapse
|
19
|
Qiu X, Miao Y, Geng X, Zhou X, Li B. Evaluation of biomarkers for in vitro prediction of drug-induced nephrotoxicity in RPTEC/TERT1 cells. Toxicol Res (Camb) 2020; 9:91-100. [PMID: 32440340 DOI: 10.1093/toxres/tfaa005] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2019] [Revised: 02/04/2020] [Accepted: 02/18/2020] [Indexed: 12/16/2022] Open
Abstract
There have been intensive efforts to identify in vivo biomarkers that can be used to monitor drug-induced kidney damage before significant impairment occurs. Kidney injury molecule-1, neutrophil gelatinase-associated lipocalin, clusterin, β2-microglobulin and cystatin C (CysC) have been validated as clinical or preclinical biomarkers in urinary and plasma predictive of acute and chronic kidney injuries and diseases. A high-throughput in vitro assay predictive of nephrotoxicity could potentially be implemented in early drug discovery stage to reduce attrition at later stages of drug development. To assess the potential of these known in vivo biomarkers for in vitro evaluation of drug-induced nephrotoxicity, we selected four nephrotoxic agents (cisplatin, cyclosporin, aristolochic acid I and gentamicin) and detected their effects on the protein levels of nephrotoxic biomarkers in RPTEC/TERT1 cells. The protein levels of clusterin, CysC, GSTπ and TIMP-1 significantly increased in the conditioned media of RPTEC/TERT1 cells treated with cisplatin, cyclosporin, aristolochic acid I and gentamicin. The messenger RNA levels of clusterin, CysC, GSTπ and TIMP-1 also increased in RPTEC/TERT1 cells treated with cisplatin, cyclosporin, aristolochic acid I and gentamicin, indicating that drug-induced upregulation involves transcriptional activation. Taken together, the results clearly demonstrate that among the known in vivo nephrotoxic biomarkers, clusterin, CysC, GSTπ and TIMP-1 can be effectively used as in vitro biomarkers for drug-induced nephrotoxicity in RPTEC/TERT1 cells.
Collapse
Affiliation(s)
- Xuan Qiu
- Chinese Academy of Medical Sciences & Peking Union Medical College, No. 9, Dongdan Santiao, Dongcheng District, Beijing 100730, China.,National Center for Safety Evaluation of Drugs, National Institutes for Food and Drug Control, A8 Hongda Middle Street, Beijing Economic-Technological Development Area, Beijing 100176, China
| | - Yufa Miao
- National Center for Safety Evaluation of Drugs, National Institutes for Food and Drug Control, A8 Hongda Middle Street, Beijing Economic-Technological Development Area, Beijing 100176, China
| | - Xingchao Geng
- National Center for Safety Evaluation of Drugs, National Institutes for Food and Drug Control, A8 Hongda Middle Street, Beijing Economic-Technological Development Area, Beijing 100176, China
| | - Xiaobing Zhou
- National Center for Safety Evaluation of Drugs, National Institutes for Food and Drug Control, A8 Hongda Middle Street, Beijing Economic-Technological Development Area, Beijing 100176, China
| | - Bo Li
- Chinese Academy of Medical Sciences & Peking Union Medical College, No. 9, Dongdan Santiao, Dongcheng District, Beijing 100730, China.,National Center for Safety Evaluation of Drugs, National Institutes for Food and Drug Control, A8 Hongda Middle Street, Beijing Economic-Technological Development Area, Beijing 100176, China
| |
Collapse
|
20
|
Abstract
Drug attrition related to kidney toxicity remains a challenge in drug discovery and development. In vitro models established over the past 2 decades to supplement in vivo studies have improved the throughput capacity of toxicity evaluation, but usually suffer from low predictive value. To achieve a paradigm shift in the prediction of drug-induced kidney toxicity, two aspects are fundamental: increased physiological relevance of the kidney model, and use of appropriate toxicity end points. Recent studies have suggested that increasing the physiological relevance of kidney models can improve their sensitivity to drug-induced damage. Here, we discuss how advanced culture models, including modified cell lines, induced pluripotent stem cells, kidney organoid cultures, and microfluidic devices enhance in vivo similarity. To this end, culture models aim to increase the proximal tubule epithelial phenotype, reconstitute multiple tissue compartments and extracellular matrix, allow exposure to fluid shear stress, and enable interaction between multiple cell types. Applying computation-aided end points and novel biomarkers to advanced culture models will further improve sensitivity and clinical relevance of in vitro drug-induced toxicity prediction. Implemented at the right stage of drug discovery and development and coupled to high-content evaluation techniques, these models have the potential to reduce attrition and aid the selection of candidate drugs with an appropriate safety profile.
Collapse
Affiliation(s)
- Tom T G Nieskens
- CVRMSafety, Drug Safety and Metabolism, IMED Biotech Unit, AstraZeneca, Gothenburg, Sweden
| | - Anna-Karin Sjögren
- CVRMSafety, Drug Safety and Metabolism, IMED Biotech Unit, AstraZeneca, Gothenburg, Sweden.
| |
Collapse
|
21
|
Faria J, Ahmed S, Gerritsen KGF, Mihaila SM, Masereeuw R. Kidney-based in vitro models for drug-induced toxicity testing. Arch Toxicol 2019; 93:3397-3418. [PMID: 31664498 DOI: 10.1007/s00204-019-02598-0] [Citation(s) in RCA: 65] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Accepted: 10/15/2019] [Indexed: 12/18/2022]
Abstract
The kidney is frequently involved in adverse effects caused by exposure to foreign compounds, including drugs. An early prediction of those effects is crucial for allowing novel, safe drugs entering the market. Yet, in current pharmacotherapy, drug-induced nephrotoxicity accounts for up to 25% of the reported serious adverse effects, of which one-third is attributed to antimicrobials use. Adverse drug effects can be due to direct toxicity, for instance as a result of kidney-specific determinants, or indirectly by, e.g., vascular effects or crystals deposition. Currently used in vitro assays do not adequately predict in vivo observed effects, predominantly due to an inadequate preservation of the organs' microenvironment in the models applied. The kidney is highly complex, composed of a filter unit and a tubular segment, together containing over 20 different cell types. The tubular epithelium is highly polarized, and the maintenance of this polarity is critical for optimal functioning and response to environmental signals. Cell polarity is dependent on communication between cells, which includes paracrine and autocrine signals, as well as biomechanic and chemotactic processes. These processes all influence kidney cell proliferation, migration, and differentiation. For drug disposition studies, this microenvironment is essential for prediction of toxic responses. This review provides an overview of drug-induced injuries to the kidney, details on relevant and translational biomarkers, and advances in 3D cultures of human renal cells, including organoids and kidney-on-a-chip platforms.
Collapse
Affiliation(s)
- João Faria
- Division of Pharmacology, Department of Pharmaceutical Sciences, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Universiteitsweg 99, 3584 CG, Utrecht, The Netherlands
| | - Sabbir Ahmed
- Division of Pharmacology, Department of Pharmaceutical Sciences, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Universiteitsweg 99, 3584 CG, Utrecht, The Netherlands
| | - Karin G F Gerritsen
- Department of Nephrology and Hypertension, University Medical Center, Utrecht, The Netherlands
| | - Silvia M Mihaila
- Division of Pharmacology, Department of Pharmaceutical Sciences, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Universiteitsweg 99, 3584 CG, Utrecht, The Netherlands.,Department of Nephrology and Hypertension, University Medical Center, Utrecht, The Netherlands
| | - Rosalinde Masereeuw
- Division of Pharmacology, Department of Pharmaceutical Sciences, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Universiteitsweg 99, 3584 CG, Utrecht, The Netherlands.
| |
Collapse
|
22
|
Zgheib E, Gao W, Limonciel A, Aladjov H, Yang H, Tebby C, Gayraud G, Jennings P, Sachana M, Beltman JB, Bois FY. Application of three approaches for quantitative AOP development to renal toxicity. ACTA ACUST UNITED AC 2019. [DOI: 10.1016/j.comtox.2019.02.001] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|
23
|
Functional transepithelial transport measurements to detect nephrotoxicity in vitro using the RPTEC/TERT1 cell line. Arch Toxicol 2019; 93:1965-1978. [DOI: 10.1007/s00204-019-02469-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Accepted: 05/02/2019] [Indexed: 12/11/2022]
|
24
|
Frazier KS, Ryan AM, Peterson RA, Obert LA. Kidney Pathology and Investigative Nephrotoxicology Strategies Across Species. Semin Nephrol 2019; 39:190-201. [DOI: 10.1016/j.semnephrol.2018.12.007] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
|
25
|
Gabelova A, Kozics K, Kapka-Skrzypczak L, Kruszewski M, Sramkova M. Nephrotoxicity: Topical issue. Mutat Res 2018; 845:402988. [PMID: 31561894 DOI: 10.1016/j.mrgentox.2018.11.012] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2018] [Revised: 11/27/2018] [Accepted: 11/29/2018] [Indexed: 12/12/2022]
Abstract
Drug-induced kidney injury is one of the most significant adverse events and dose limiting factor in chemotherapy as well a major cause of prospective drug attrition during pharmaceutical development. Moreover, kidney injury can also occur as a consequence of exposures to environmental xenobiotics such as heavy metals, fungal toxins and nanomaterials. The lack of adequate in vitro human kidney models that mimic more realistically the in vivo conditions and the absence of suitable and robust, cost-effective and predictive cell-based in vitro assays contribute to an underestimation of the kidney toxic potential of new drugs and xenobiotics. Therefore, a rapid screening system capable to detect potential nephrotoxicity at early stages of drug discovery is an urgent need. Here we provide an overview of human cell lines currently used as a surrogate in vitro kidney models in nephrotoxicity studies, including their advantages and limitations. In addition, the capacity of the single cell gel electrophoresis (SCGE)/comet assay as a potential tool in kidney toxicants screening is discussed. Despite a limited number of studies using the comet assay to evaluate the drug-induced kidney damage potential, a considerable variability in SCGE methodology (e.g. lysis, unwinding, and electrophoresis conditions) has been observed. Before the comet assay can be included in nephrotoxicity testing, a basic guideline has to be developed. To test its feasibility, additional in vitro experiments including inter-laboratory validation studies based on this guideline have to be performed.
Collapse
Affiliation(s)
- Alena Gabelova
- Cancer Research Institute, Biomedical Research Center SAS, Dubravska cesta 9, 845 05 Bratislava, Slovakia.
| | - Katarina Kozics
- Cancer Research Institute, Biomedical Research Center SAS, Dubravska cesta 9, 845 05 Bratislava, Slovakia
| | - Lucyna Kapka-Skrzypczak
- Department of Molecular Biology and Translational Research, Institute of Rural Health, Jaczewskiego 2, 20-090 Lublin, Poland; Department of Medical Biology and Translational Research, Faculty of Medicine, University of Information Technology and Management, Sucharskiego 2, 35-225, Rzeszów, Poland
| | - Marcin Kruszewski
- Department of Molecular Biology and Translational Research, Institute of Rural Health, Jaczewskiego 2, 20-090 Lublin, Poland; Department of Medical Biology and Translational Research, Faculty of Medicine, University of Information Technology and Management, Sucharskiego 2, 35-225, Rzeszów, Poland; Centre for Radiobiology and Biological Dosimetry, Institute of Nuclear Chemistry and Technology, Dorodna 16, 03-195, Warsaw, Poland
| | - Monika Sramkova
- Cancer Research Institute, Biomedical Research Center SAS, Dubravska cesta 9, 845 05 Bratislava, Slovakia
| |
Collapse
|
26
|
Zgheib E, Limonciel A, Jiang X, Wilmes A, Wink S, van de Water B, Kopp-Schneider A, Bois FY, Jennings P. Investigation of Nrf2, AhR and ATF4 Activation in Toxicogenomic Databases. Front Genet 2018; 9:429. [PMID: 30333853 PMCID: PMC6176024 DOI: 10.3389/fgene.2018.00429] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Accepted: 09/11/2018] [Indexed: 12/14/2022] Open
Abstract
Toxicological responses to chemical insult are largely regulated by transcriptionally activated pathways that may be independent, correlated and partially or fully overlapping. Investigating the dynamics of the interactions between stress responsive transcription factors from toxicogenomic data and defining the signature of each of them is an additional step toward a system level understanding of perturbation driven mechanisms. To this end, we investigated the segregation of the genes belonging to the three following transcriptionally regulated pathways: the AhR pathway, the Nrf2 pathway and the ATF4 pathway. Toxicogenomic datasets from three projects (carcinoGENOMICS, Predict-IV and TG-GATEs) obtained in various experimental conditions (in human and rat in vitro liver and kidney models and rat in vivo, with bolus administration and with repeated doses) were combined and consolidated where overlaps between datasets existed. A bioinformatic analysis was performed to refine pathways' signatures and to create chemical activation capacity scores to classify chemicals by their potency and selectivity of activation of each pathway. With some refinement such an approach may improve chemical safety classification and allow biological read across on a pathway level.
Collapse
Affiliation(s)
- Elias Zgheib
- Laboratoire de Biomécanique et Bio-ingénierie, Sorbonne Universités - Université de Technologie de Compiègne, Compiègne, France
| | - Alice Limonciel
- Division of Molecular and Computational Toxicology, Amsterdam Institute for Molecules, Medicines and Systems, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Xiaoqi Jiang
- Division of Biostatistics, German Cancer Research Center, Heidelberg, Germany
| | - Anja Wilmes
- Division of Molecular and Computational Toxicology, Amsterdam Institute for Molecules, Medicines and Systems, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Steven Wink
- Division of Drug Discovery and Safety, Leiden Cell Observatory High Content Imaging Screening Facility, Leiden Academic Center for Drug Research, Leiden University, Leiden, Netherlands
| | - Bob van de Water
- Division of Drug Discovery and Safety, Leiden Cell Observatory High Content Imaging Screening Facility, Leiden Academic Center for Drug Research, Leiden University, Leiden, Netherlands
| | | | - Frederic Y Bois
- Models for Ecotoxicology and Toxicology Unit (DRC/VIVA/METO), Institut National de l'Environnement Industriel et des Risques, Verneuil-en-Halatte, France
| | - Paul Jennings
- Division of Molecular and Computational Toxicology, Amsterdam Institute for Molecules, Medicines and Systems, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| |
Collapse
|
27
|
Bajaj P, Chowdhury SK, Yucha R, Kelly EJ, Xiao G. Emerging Kidney Models to Investigate Metabolism, Transport, and Toxicity of Drugs and Xenobiotics. Drug Metab Dispos 2018; 46:1692-1702. [PMID: 30076203 DOI: 10.1124/dmd.118.082958] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2018] [Accepted: 08/01/2018] [Indexed: 01/11/2023] Open
Abstract
The kidney is a major clearance organ of the body and is responsible for the elimination of many xenobiotics and prescription drugs. With its multitude of uptake and efflux transporters and metabolizing enzymes, the proximal tubule cell (PTC) in the nephron plays a key role in the disposition of xenobiotics and is also a primary site for toxicity. In this minireview, we first provide an overview of the major transporters and metabolizing enzymes in the PTCs responsible for biotransformation and disposition of drugs. Next, we discuss different cell sources that have been used to model PTCs in vitro, their pros and cons, and their characterization. As current technology is inadequate to evaluate reliably drug disposition and toxicity in the kidney, we then discuss recent advancements in kidney microphysiological systems (MPS) and the need to develop robust in vitro platforms that could be routinely used by pharmaceutical companies to screen compounds. Finally, we discuss the new and exciting field of stem cell-derived kidney models as potential cell sources for future kidney MPS. Given the push from both regulatory agencies and pharmaceutical companies to use more predictive "human-like" in vitro systems in the early stages of drug development to reduce attrition, these emerging models have the potential to be a game changer and may revolutionize how renal disposition and kidney toxicity in drug discovery are evaluated in the future.
Collapse
Affiliation(s)
- Piyush Bajaj
- Drug Safety Research and Evaluation (P.B.) and Drug Metabolism and Pharmacokinetics Department (S.K.C., R.Y., G.X.), Takeda Pharmaceutical International Co., Cambridge, Massachusetts; and Department of Pharmaceutics, University of Washington, Seattle, Washington (E.J.K.)
| | - Swapan K Chowdhury
- Drug Safety Research and Evaluation (P.B.) and Drug Metabolism and Pharmacokinetics Department (S.K.C., R.Y., G.X.), Takeda Pharmaceutical International Co., Cambridge, Massachusetts; and Department of Pharmaceutics, University of Washington, Seattle, Washington (E.J.K.)
| | - Robert Yucha
- Drug Safety Research and Evaluation (P.B.) and Drug Metabolism and Pharmacokinetics Department (S.K.C., R.Y., G.X.), Takeda Pharmaceutical International Co., Cambridge, Massachusetts; and Department of Pharmaceutics, University of Washington, Seattle, Washington (E.J.K.)
| | - Edward J Kelly
- Drug Safety Research and Evaluation (P.B.) and Drug Metabolism and Pharmacokinetics Department (S.K.C., R.Y., G.X.), Takeda Pharmaceutical International Co., Cambridge, Massachusetts; and Department of Pharmaceutics, University of Washington, Seattle, Washington (E.J.K.)
| | - Guangqing Xiao
- Drug Safety Research and Evaluation (P.B.) and Drug Metabolism and Pharmacokinetics Department (S.K.C., R.Y., G.X.), Takeda Pharmaceutical International Co., Cambridge, Massachusetts; and Department of Pharmaceutics, University of Washington, Seattle, Washington (E.J.K.)
| |
Collapse
|
28
|
Limonciel A, Ates G, Carta G, Wilmes A, Watzele M, Shepard PJ, VanSteenhouse HC, Seligmann B, Yeakley JM, van de Water B, Vinken M, Jennings P. Comparison of base-line and chemical-induced transcriptomic responses in HepaRG and RPTEC/TERT1 cells using TempO-Seq. Arch Toxicol 2018; 92:2517-2531. [PMID: 30008028 PMCID: PMC6063331 DOI: 10.1007/s00204-018-2256-2] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2017] [Accepted: 07/04/2018] [Indexed: 12/21/2022]
Abstract
The utilisation of genome-wide transcriptomics has played a pivotal role in advancing the field of toxicology, allowing the mapping of transcriptional signatures to chemical exposures. These activities have uncovered several transcriptionally regulated pathways that can be utilised for assessing the perturbation impact of a chemical and also the identification of toxic mode of action. However, current transcriptomic platforms are not very amenable to high-throughput workflows due to, high cost, complexities in sample preparation and relatively complex bioinformatic analysis. Thus, transcriptomic investigations are usually limited in dose and time dimensions and are, therefore, not optimal for implementation in risk assessment workflows. In this study, we investigated a new cost-effective, transcriptomic assay, TempO-Seq, which alleviates the aforementioned limitations. This technique was evaluated in a 6-compound screen, utilising differentiated kidney (RPTEC/TERT1) and liver (HepaRG) cells and compared to non-transcriptomic label-free sensitive endpoints of chemical-induced disturbances, namely phase contrast morphology, xCELLigence and glycolysis. Non-proliferating cell monolayers were exposed to six sub-lethal concentrations of each compound for 24 h. The results show that utilising a 2839 gene panel, it is possible to discriminate basal tissue-specific signatures, generate dose-response relationships and to discriminate compound-specific and cell type-specific responses. This study also reiterates previous findings that chemical-induced transcriptomic alterations occur prior to cytotoxicity and that transcriptomics provides in depth mechanistic information of the effects of chemicals on cellular transcriptional responses. TempO-Seq is a robust transcriptomic platform that is well suited for in vitro toxicity experiments.
Collapse
Affiliation(s)
- Alice Limonciel
- Division of Molecular and Computational Toxicology, Amsterdam Institute for Molecules, Medicines and Systems, Vrije Universiteit Amsterdam, De Boelelaan 1108, 1081 HZ, Amsterdam, The Netherlands
- Division of Physiology, Department of Physiology and Medical Physics, Medical University of Innsbruck, 6020, Innsbruck, Austria
| | - Gamze Ates
- Department of In Vitro Toxicology and Dermato-Cosmetology, Vrije Universiteit Brussel, Laarbeeklaan 103, 1090, Brussels, Belgium
| | - Giada Carta
- Division of Molecular and Computational Toxicology, Amsterdam Institute for Molecules, Medicines and Systems, Vrije Universiteit Amsterdam, De Boelelaan 1108, 1081 HZ, Amsterdam, The Netherlands
- Division of Physiology, Department of Physiology and Medical Physics, Medical University of Innsbruck, 6020, Innsbruck, Austria
| | - Anja Wilmes
- Division of Molecular and Computational Toxicology, Amsterdam Institute for Molecules, Medicines and Systems, Vrije Universiteit Amsterdam, De Boelelaan 1108, 1081 HZ, Amsterdam, The Netherlands
- Division of Physiology, Department of Physiology and Medical Physics, Medical University of Innsbruck, 6020, Innsbruck, Austria
| | - Manfred Watzele
- Roche Diagnostics GmbH, Nonnenwald 2, 82377, Penzberg, Germany
| | - Peter J Shepard
- BioSpyder Technologies, Inc., 5922 Farnsworth Ct Ste 102, Carlsbad, CA, 92008, USA
| | | | - Bruce Seligmann
- BioSpyder Technologies, Inc., 5922 Farnsworth Ct Ste 102, Carlsbad, CA, 92008, USA
| | - Joanne M Yeakley
- BioSpyder Technologies, Inc., 5922 Farnsworth Ct Ste 102, Carlsbad, CA, 92008, USA
| | - Bob van de Water
- Division of Toxicology, Leiden Academic Center for Drug Research, Leiden University, PO Box 9503, 2300 RA, Leiden, The Netherlands
| | - Mathieu Vinken
- Department of In Vitro Toxicology and Dermato-Cosmetology, Vrije Universiteit Brussel, Laarbeeklaan 103, 1090, Brussels, Belgium.
| | - Paul Jennings
- Division of Molecular and Computational Toxicology, Amsterdam Institute for Molecules, Medicines and Systems, Vrije Universiteit Amsterdam, De Boelelaan 1108, 1081 HZ, Amsterdam, The Netherlands.
- Division of Physiology, Department of Physiology and Medical Physics, Medical University of Innsbruck, 6020, Innsbruck, Austria.
| |
Collapse
|
29
|
Suter-Dick L, Mauch L, Ramp D, Caj M, Vormann MK, Hutter S, Lanz HL, Vriend J, Masereeuw R, Wilmer MJ. Combining Extracellular miRNA Determination with Microfluidic 3D Cell Cultures for the Assessment of Nephrotoxicity: a Proof of Concept Study. AAPS JOURNAL 2018; 20:86. [PMID: 30039346 DOI: 10.1208/s12248-018-0245-2] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/01/2018] [Accepted: 06/25/2018] [Indexed: 12/31/2022]
Abstract
Drug-induced kidney injury is often observed in the clinics and can lead to long-term organ failure. In this work, we evaluated a novel in vitro system that aims at detecting whether compounds can cause renal proximal tubule damage in man. For this, we implemented organotypic cultures of human conditionally immortalized proximal tubule epithelial cells overexpressing the organic anion transporter 1 (ciPTEC-OAT1) in a three-channel OrganoPlate under microfluidic conditions. Cells were exposed to four known nephrotoxicants (cisplatin, tenofovir, cyclosporine A, and tobramycin). The effect on cell viability and NAG release into the medium was determined. A novel panel of four miRNAs (mir-21, mir-29a, mir-34a, and mir-192) was selected as potential biomarkers of proximal tubule damage. After nephrotoxicant treatment, miRNA levels in culture medium were earlier indicators than cell viability (WST-8 assay) and outperformed NAG for proximal tubule damage. In particular, mir-29a, mir-34a, and mir-192 were highly reproducible between experiments and across compounds, whereas mir-21 showed more variability. Moreover, similar data were obtained in two different laboratories, underlining the reproducibility and technical transferability of the results, a key requirement for the implementation of novel biomarkers. In conclusion, the selected miRNAs behaved like sensitive biomarkers of damage to tubular epithelial cells caused by several nephrotoxicity mechanisms. This biomarker panel, in combination with the 3D cultures of ciPTEC-OAT1 in the OrganoPlate, represents a novel tool for in vitro nephrotoxicity detection. These results pave the way for the application of miRNAs in longitudinal, time-course in vitro toxicity studies.
Collapse
Affiliation(s)
- Laura Suter-Dick
- School of Life Sciences, University of Applied Sciences Northwestern Switzerland, Gründenstrasse 40, 4132, Muttenz, Switzerland.
| | - L Mauch
- School of Life Sciences, University of Applied Sciences Northwestern Switzerland, Gründenstrasse 40, 4132, Muttenz, Switzerland
| | - D Ramp
- School of Life Sciences, University of Applied Sciences Northwestern Switzerland, Gründenstrasse 40, 4132, Muttenz, Switzerland
| | - M Caj
- School of Life Sciences, University of Applied Sciences Northwestern Switzerland, Gründenstrasse 40, 4132, Muttenz, Switzerland
| | - M K Vormann
- MIMETAS B.V., JH Oortweg 19, 2333 CH, Leiden, The Netherlands
| | - S Hutter
- MIMETAS B.V., JH Oortweg 19, 2333 CH, Leiden, The Netherlands
| | - H L Lanz
- MIMETAS B.V., JH Oortweg 19, 2333 CH, Leiden, The Netherlands
| | - J Vriend
- Department of Pharmacology and Toxicology, Radboudumc, P.O. box 9101, 6500 HB, Nijmegen, The Netherlands
| | - R Masereeuw
- Div. Pharmacology, Department of Pharmaceutical Sciences, Utrecht University, Universiteitsweg 99, 3584 CG, Utrecht, The Netherlands
| | - M J Wilmer
- Department of Pharmacology and Toxicology, Radboudumc, P.O. box 9101, 6500 HB, Nijmegen, The Netherlands
| |
Collapse
|
30
|
Corvi R, Madia F, Guyton KZ, Kasper P, Rudel R, Colacci A, Kleinjans J, Jennings P. Moving forward in carcinogenicity assessment: Report of an EURL ECVAM/ESTIV workshop. Toxicol In Vitro 2017; 45:278-286. [PMID: 28911985 PMCID: PMC5735222 DOI: 10.1016/j.tiv.2017.09.010] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2017] [Revised: 09/08/2017] [Accepted: 09/10/2017] [Indexed: 10/27/2022]
Abstract
There is an increased need to develop novel alternative approaches to the two-year rodent bioassay for the carcinogenicity assessment of substances where the rodent bioassay is still a basic requirement, as well as for those substances where animal use is banned or limited or where information gaps are identified within legislation. The current progress in this area was addressed in a EURL ECVAM- ESTIV workshop held in October 2016, in Juan les Pins. A number of initiatives were presented and discussed, including data-driven, technology-driven and pathway-driven approaches. Despite a seemingly diverse range of strategic developments, commonalities are emerging. For example, providing insight into carcinogenicity mechanisms is becoming an increasingly appreciated aspect of hazard assessment and is suggested to be the best strategy to drive new developments. Thus, now more than ever, there is a need to combine and focus efforts towards the integration of available information between sectors. Such cross-sectorial harmonisation will aid in building confidence in new approach methods leading to increased implementation and thus a decreased necessity for the two-year rodent bioassay.
Collapse
Affiliation(s)
- Raffaella Corvi
- European Commission, Joint Research Centre (JRC), EU Reference Laboratory for Alternatives to Animal Testing (EURL ECVAM), Ispra, (VA), Italy.
| | - Federica Madia
- European Commission, Joint Research Centre (JRC), EU Reference Laboratory for Alternatives to Animal Testing (EURL ECVAM), Ispra, (VA), Italy
| | - Kathryn Z Guyton
- Monographs Programme, International Agency for Research on Cancer, Lyon, France
| | - Peter Kasper
- Federal Institute for Drugs and Medical Devices (BfArM), Bonn, Germany
| | | | - Annamaria Colacci
- Centre for Environmental Toxicology and Risk Assessment, Environmental Protection and Health Prevention Agency, Emilia Romagna Region, Italy
| | - Jos Kleinjans
- Department of Toxicogenomics, Maastricht University, Maastricht, The Netherlands
| | - Paul Jennings
- Division of Molecular and Computational Toxicology, Amsterdam Institute for Molecules, Medicines and Systems, Vrije Universiteit Amsterdam, HZ Amsterdam, The Netherlands
| |
Collapse
|
31
|
Bolt HM. Adverse outcome pathways. Arch Toxicol 2017; 91:4023-4024. [DOI: 10.1007/s00204-017-2123-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2017] [Accepted: 11/13/2017] [Indexed: 01/07/2023]
|
32
|
Hartung T, FitzGerald RE, Jennings P, Mirams GR, Peitsch MC, Rostami-Hodjegan A, Shah I, Wilks MF, Sturla SJ. Systems Toxicology: Real World Applications and Opportunities. Chem Res Toxicol 2017; 30:870-882. [PMID: 28362102 PMCID: PMC5396025 DOI: 10.1021/acs.chemrestox.7b00003] [Citation(s) in RCA: 72] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2017] [Indexed: 01/14/2023]
Abstract
Systems Toxicology aims to change the basis of how adverse biological effects of xenobiotics are characterized from empirical end points to describing modes of action as adverse outcome pathways and perturbed networks. Toward this aim, Systems Toxicology entails the integration of in vitro and in vivo toxicity data with computational modeling. This evolving approach depends critically on data reliability and relevance, which in turn depends on the quality of experimental models and bioanalysis techniques used to generate toxicological data. Systems Toxicology involves the use of large-scale data streams ("big data"), such as those derived from omics measurements that require computational means for obtaining informative results. Thus, integrative analysis of multiple molecular measurements, particularly acquired by omics strategies, is a key approach in Systems Toxicology. In recent years, there have been significant advances centered on in vitro test systems and bioanalytical strategies, yet a frontier challenge concerns linking observed network perturbations to phenotypes, which will require understanding pathways and networks that give rise to adverse responses. This summary perspective from a 2016 Systems Toxicology meeting, an international conference held in the Alps of Switzerland, describes the limitations and opportunities of selected emerging applications in this rapidly advancing field. Systems Toxicology aims to change the basis of how adverse biological effects of xenobiotics are characterized, from empirical end points to pathways of toxicity. This requires the integration of in vitro and in vivo data with computational modeling. Test systems and bioanalytical technologies have made significant advances, but ensuring data reliability and relevance is an ongoing concern. The major challenge facing the new pathway approach is determining how to link observed network perturbations to phenotypic toxicity.
Collapse
Affiliation(s)
- Thomas Hartung
- Center
for Alternatives to Animal Testing (CAAT), Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland 21205, United States
- University
of Konstanz, CAAT-Europe, 78457 Konstanz, Germany
| | - Rex E. FitzGerald
- Swiss
Centre for Applied Human Toxicology, University
of Basel, 4055 Basel, Switzerland
| | - Paul Jennings
- Division
of Physiology, Department of Physiology and Medical Physics, Medical University of Innsbruck, 6020 Innsbruck, Austria
| | - Gary R. Mirams
- Centre
for Mathematical Medicine & Biology, School of Mathematical Sciences, University of Nottingham, Nottingham NG7 2RD, U.K.
| | - Manuel C. Peitsch
- Department
of Research and Development, Philip Morris
International, 2000 Neuchâtel, Switzerland
| | - Amin Rostami-Hodjegan
- Centre
for Applied Pharmacokinetic Research, University
of Manchester, Manchester M13 9PL, U.K.
- Simcyp
Limited (a Certara Company), Blades Enterprise
Centre, Sheffield S2 4SU, U.K.
| | - Imran Shah
- National
Center for Computational Toxicology, Research Triangle Park, North Carolina 27711, United States
| | - Martin F. Wilks
- Swiss
Centre for Applied Human Toxicology, University
of Basel, 4055 Basel, Switzerland
| | - Shana J. Sturla
- Department
of Health Sciences and Technology, ETH Zurich, 8092 Zurich, Switzerland
| |
Collapse
|
33
|
Abstract
There is no good science in bad models. Cell culture is especially prone to artifacts. A number of novel cell culture technologies have become more broadly available in the 21st century, which allow overcoming limitations of traditional culture and are more physiologically relevant. These include the use of stem-cell derived human cells, cocultures of different cell types, scaffolds and extracellular matrices, perfusion platforms (such as microfluidics), 3D culture, organ-on-chip technologies, tissue architecture, and organ functionality. The physiological relevance of such models is further enhanced by the measurement of biomarkers (e.g., key events of pathways), organ specific functionality, and more comprehensive assessment cell responses by high-content methods. These approaches are still rarely combined to create microphysiological systems. The complexity of the combination of these technologies can generate results closer to the in vivo situation but increases the number of parameters to control, bringing some new challenges. In fact, we do not argue that all cell culture needs to be that sophisticated. The efforts taken are determined by the purpose of our experiments and tests. If only a very specific molecular target to cell response is of interest, a very simple model, which reflects this, might be much more suited to allow standardization and high-throughput. However, the less defined the end point of interest and cellular response are, the better we should approximate organ- or tissue-like culture conditions to make physiological responses more probable. Besides these technologic advances, important progress in the quality assurance and reporting on cell cultures as well as the validation of cellular test systems brings the utility of cell cultures to a new level. The advancement and broader implementation of Good Cell Culture Practice (GCCP) is key here. In toxicology, this is a major prerequisite for meaningful and reliable results, ultimately supporting risk assessment and product development decisions.
Collapse
Affiliation(s)
- David Pamies
- Center for Alternatives to Animal Testing (CAAT), Johns Hopkins Bloomberg School of Public Health , Baltimore, Maryland 21205, United States
| | - Thomas Hartung
- Center for Alternatives to Animal Testing (CAAT), Johns Hopkins Bloomberg School of Public Health , Baltimore, Maryland 21205, United States.,CAAT-Europe, University of Konstanz , 78464 Konstanz, Germany
| |
Collapse
|
34
|
Pfaller W, Prieto P, Dekant W, Jennings P, Blaauboer BJ. The Predict-IV project: Towards predictive toxicology using in vitro techniques. Toxicol In Vitro 2016; 30:1-3. [PMID: 26653009 DOI: 10.1016/j.tiv.2015.10.006] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Affiliation(s)
- Walter Pfaller
- Department of Physiology and Medical Physics, Division of Physiology, Renal Physiology, Medical University of Innsbruck, A-6020 Innsbruck, Fritz-Pregl-Strasse 3/1, Austria
| | - Pilar Prieto
- The European Union Reference Laboratory for Alternatives to Animal Testing (EURL ECVAM), Institute for Health and Consumer Protection, European Commission Joint Research Centre, Ispra, Italy
| | - Wolfgang Dekant
- Department of Toxicology, University of Wuerzburg, Wuerzburg, Germany
| | - Paul Jennings
- Department of Physiology and Medical Physics, Division of Physiology, Renal Physiology, Medical University of Innsbruck, A-6020 Innsbruck, Fritz-Pregl-Strasse 3/1, Austria
| | - Bas J Blaauboer
- Institute for Risk Assessment Sciences, Division of Toxicology, Utrecht University, Utrecht, The Netherlands
| |
Collapse
|
35
|
Biokinetics in repeated-dosing in vitro drug toxicity studies. Toxicol In Vitro 2015; 30:217-24. [PMID: 26362508 DOI: 10.1016/j.tiv.2015.09.005] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2015] [Revised: 08/11/2015] [Accepted: 09/04/2015] [Indexed: 12/17/2022]
Abstract
The aim of the EU FP7 Predict-IV project was to improve the predictivity of in vitro assays for unwanted effects of drugs after repeated dosing. The project assessed the added benefit of integrating long-lived in vitro organotypic cell systems with 'omics' technologies and in silico modelling, including systems biology and pharmacokinetic assessments. RPTEC/TERT1 kidney cells, primary rat and human hepatocytes, HepaRG liver cells and 2D and 3D primary brain cultures were dosed daily or every other day for 14 days to a selection of drugs varying in their mechanism of pharmacological action. Since concentration-effect relationships not only depend on the activity of the drug or the sensitivity of the target, but also on the distribution of compounds in the in vitro system, the concentration of a selection of drugs in cells, microtitre plate plastic and medium was measured over time. Results, reviewed in this paper, indicate that lipophilic drugs bind significantly to plastic labware. A few drugs, including less lipophilic drugs, bind to cell-attachment matrices. Chemicals that reach high concentrations in cells, including cyclosporin A and amiodarone, significantly accumulate over time after repeated dosing, partly explaining their increased toxicity after repeated dosing, compared to a single dose.
Collapse
|
36
|
Value of monitoring Nrf2 activity for the detection of chemical and oxidative stress. Biochem Soc Trans 2015; 43:657-62. [PMID: 26551708 PMCID: PMC4613517 DOI: 10.1042/bst20150044] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2015] [Indexed: 02/08/2023]
Abstract
Beyond specific limits of exposure, chemical entities can provoke deleterious effects in mammalian cells via direct interaction with critical macromolecules or by stimulating the accumulation of reactive oxygen species (ROS). In particular, these chemical and oxidative stresses can underpin adverse reactions to therapeutic drugs, which pose an unnecessary burden in the clinic and pharmaceutical industry. Novel pre-clinical testing strategies are required to identify, at an earlier stage in the development pathway, chemicals and drugs that are likely to provoke toxicity in humans. Mammalian cells can adapt to chemical and oxidative stress via the action of the transcription factor nuclear factor erythroid 2-related factor 2 (Nrf2), which up-regulates the expression of numerous cell defence genes and has been shown to protect against a variety of chemical toxicities. Here, we provide a brief overview of the Nrf2 pathway and summarize novel experimental models that can be used to monitor changes in Nrf2 pathway activity and thus understand the functional consequences of such perturbations in the context of chemical and drug toxicity. We also provide an outlook on the potential value of monitoring Nrf2 activity for improving the pre-clinical identification of chemicals and drugs with toxic liability in humans.
Collapse
|
37
|
Ranninger C, Rurik M, Limonciel A, Ruzek S, Reischl R, Wilmes A, Jennings P, Hewitt P, Dekant W, Kohlbacher O, Huber CG. Nephron Toxicity Profiling via Untargeted Metabolome Analysis Employing a High Performance Liquid Chromatography-Mass Spectrometry-based Experimental and Computational Pipeline. J Biol Chem 2015; 290:19121-32. [PMID: 26055719 DOI: 10.1074/jbc.m115.644146] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2015] [Indexed: 11/06/2022] Open
Abstract
Untargeted metabolomics has the potential to improve the predictivity of in vitro toxicity models and therefore may aid the replacement of expensive and laborious animal models. Here we describe a long term repeat dose nephrotoxicity study conducted on the human renal proximal tubular epithelial cell line, RPTEC/TERT1, treated with 10 and 35 μmol·liter(-1) of chloroacetaldehyde, a metabolite of the anti-cancer drug ifosfamide. Our study outlines the establishment of an automated and easy to use untargeted metabolomics workflow for HPLC-high resolution mass spectrometry data. Automated data analysis workflows based on open source software (OpenMS, KNIME) enabled a comprehensive and reproducible analysis of the complex and voluminous metabolomics data produced by the profiling approach. Time- and concentration-dependent responses were clearly evident in the metabolomic profiles. To obtain a more comprehensive picture of the mode of action, transcriptomics and proteomics data were also integrated. For toxicity profiling of chloroacetaldehyde, 428 and 317 metabolite features were detectable in positive and negative modes, respectively, after stringent removal of chemical noise and unstable signals. Changes upon treatment were explored using principal component analysis, and statistically significant differences were identified using linear models for microarray assays. The analysis revealed toxic effects only for the treatment with 35 μmol·liter(-1) for 3 and 14 days. The most regulated metabolites were glutathione and metabolites related to the oxidative stress response of the cells. These findings are corroborated by proteomics and transcriptomics data, which show, among other things, an activation of the Nrf2 and ATF4 pathways.
Collapse
Affiliation(s)
- Christina Ranninger
- From the Department of Molecular Biology, Division of Chemistry and Bioanalytics, University of Salzburg, 5020 Salzburg, Austria
| | - Marc Rurik
- the Applied Bioinformatics Group, Center for Bioinformatics, Quantitative Biology Center and Department of Computer Science, University of Tübingen, Tübingen 72076, Germany
| | - Alice Limonciel
- the Division of Physiology, Department of Physiology and Medical Physics, Medical University of Innsbruck, Innsbruck 6020, Austria
| | - Silke Ruzek
- From the Department of Molecular Biology, Division of Chemistry and Bioanalytics, University of Salzburg, 5020 Salzburg, Austria
| | - Roland Reischl
- From the Department of Molecular Biology, Division of Chemistry and Bioanalytics, University of Salzburg, 5020 Salzburg, Austria
| | - Anja Wilmes
- the Division of Physiology, Department of Physiology and Medical Physics, Medical University of Innsbruck, Innsbruck 6020, Austria
| | - Paul Jennings
- the Division of Physiology, Department of Physiology and Medical Physics, Medical University of Innsbruck, Innsbruck 6020, Austria
| | - Philip Hewitt
- Merck KGaA, Merck Serono, Nonclinical Safety, Darmstadt 64293, Germany, and
| | - Wolfgang Dekant
- the Department of Toxicology, University of Würzburg, Würzburg 97078, Germany
| | - Oliver Kohlbacher
- the Applied Bioinformatics Group, Center for Bioinformatics, Quantitative Biology Center and Department of Computer Science, University of Tübingen, Tübingen 72076, Germany
| | - Christian G Huber
- From the Department of Molecular Biology, Division of Chemistry and Bioanalytics, University of Salzburg, 5020 Salzburg, Austria,
| |
Collapse
|
38
|
Expression of xenobiotic transporters in the human renal proximal tubule cell line RPTEC/TERT1. Toxicol In Vitro 2014; 30:95-105. [PMID: 25500123 DOI: 10.1016/j.tiv.2014.12.003] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2014] [Revised: 11/17/2014] [Accepted: 12/03/2014] [Indexed: 12/20/2022]
Abstract
The kidney is a major target for drug-induced injury, primarily due the fact that it transports a wide variety of chemical entities into and out of the tubular lumen. Here, we investigated the expression of the main xenobiotic transporters in the human renal proximal tubule cell line RPTEC/TERT1 at an mRNA and/or protein level. RPTEC/TERT1 cells expressed OCT2, OCT3, OCTN2, MATE1, MATE2, OAT1, OAT3 and OAT4. The functionality of the OCTs was demonstrated by directional transport of the fluorescent dye 4-Di-1-ASP. In addition, P-glycoprotein activity in RPTEC/TERT1 cells was verified by fluorescent dye retention in presence of various P-glycoprotein inhibitors. In comparison to proliferating cells, contact inhibited RPTEC/TERT1 cells expressed increased mRNA levels of several ABC transporter family members and were less sensitive to cyclosporine A. We conclude that differentiated RPTEC/TERT1 cells are well suited for utilisation in xenobiotic transport and pharmacokinetic studies.
Collapse
|