1
|
Masenga SK, Desta S, Hatcher M, Kirabo A, Lee DL. How PPAR-alpha mediated inflammation may affect the pathophysiology of chronic kidney disease. Curr Res Physiol 2024; 8:100133. [PMID: 39665027 PMCID: PMC11629568 DOI: 10.1016/j.crphys.2024.100133] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Revised: 10/03/2024] [Accepted: 11/12/2024] [Indexed: 12/13/2024] Open
Abstract
Chronic kidney disease (CKD) is a major risk factor for death in adults. Inflammation plays a role in the pathogenesis of CKD, but the mechanisms are poorly understood. Peroxisome proliferator-activated receptor alpha (PPAR-α) is a nuclear receptor and one of the three members (PPARα, PPARβ/δ, and PPARγ) of the PPARs that plays an important role in ameliorating pathological processes that accelerate acute and chronic kidney disease. Although other PPARs members are well studied, the role of PPAR-α is not well described and its role in inflammation-mediated chronic disease is not clear. Herein, we review the role of PPAR-α in chronic kidney disease with implications for the immune system.
Collapse
Affiliation(s)
- Sepiso K. Masenga
- HAND Research Group, School of Medicine and Health Sciences, Mulungushi University, Livingstone Campus, Zambia
- Vanderbilt Institute for Global Health, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Selam Desta
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
- Department of Physiology and Biophysics, College of Medicine, Howard University, Washington, DC, USA
| | - Mark Hatcher
- Department of Physiology and Biophysics, College of Medicine, Howard University, Washington, DC, USA
| | - Annet Kirabo
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
- Vanderbilt Center for Immunobiology, Vanderbilt University Medical Center, Nashville, TN, USA
- Vanderbilt Institute for Infection, Immunology, and Inflammation, Vanderbilt University Medical Center, Nashville, TN, USA
- Vanderbilt Institute for Global Health, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Dexter L. Lee
- Department of Physiology and Biophysics, College of Medicine, Howard University, Washington, DC, USA
| |
Collapse
|
2
|
Meijer T, da Costa Pereira D, Klatt OC, Buitenhuis J, Jennings P, Wilmes A. Characterization of Organic Anion and Cation Transport in Three Human Renal Proximal Tubular Epithelial Models. Cells 2024; 13:1008. [PMID: 38920639 PMCID: PMC11202273 DOI: 10.3390/cells13121008] [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: 05/24/2024] [Accepted: 06/05/2024] [Indexed: 06/27/2024] Open
Abstract
The polarised expression of specific transporters in proximal tubular epithelial cells is important for the renal clearance of many endogenous and exogenous compounds. Thus, ideally, the in vitro tools utilised for predictions would have a similar expression of apical and basolateral xenobiotic transporters as in vivo. Here, we assessed the functionality of organic cation and anion transporters in proximal tubular-like cells (PTL) differentiated from human induced pluripotent stem cells (iPSC), primary human proximal tubular epithelial cells (PTEC), and telomerase-immortalised human renal proximal tubular epithelial cells (RPTEC/TERT1). Organic cation and anion transport were studied using the fluorescent substrates 4-(4-(dimethylamino)styryl)-N-methylpyridinium iodide (ASP) and 6-carboxyfluorescein (6-CF), respectively. The level and rate of intracellular ASP accumulation in PTL following basolateral application were slightly lower but within a 3-fold range compared to primary PTEC and RPTEC/TERT1 cells. The basolateral uptake of ASP and its subsequent apical efflux could be inhibited by basolateral exposure to quinidine in all models. Of the three models, only PTL showed a modest preferential basolateral-to-apical 6-CF transfer. These results show that organic cation transport could be demonstrated in all three models, but more research is needed to improve and optimise organic anion transporter expression and functionality.
Collapse
Affiliation(s)
- Tamara Meijer
- Department of Chemistry and Pharmaceutical Sciences, Vrije Universiteit Amsterdam, De Boelelaan 1108, 1081 HZ Amsterdam, The Netherlands; (T.M.); (D.d.C.P.); (O.C.K.); (P.J.)
- Amsterdam Institute of Molecular and Life Sciences (AIMMS), Vrije Universiteit Amsterdam, De Boelelaan 1108, 1081 HZ Amsterdam, The Netherlands
| | - Daniel da Costa Pereira
- Department of Chemistry and Pharmaceutical Sciences, Vrije Universiteit Amsterdam, De Boelelaan 1108, 1081 HZ Amsterdam, The Netherlands; (T.M.); (D.d.C.P.); (O.C.K.); (P.J.)
- Amsterdam Institute of Molecular and Life Sciences (AIMMS), Vrije Universiteit Amsterdam, De Boelelaan 1108, 1081 HZ Amsterdam, The Netherlands
| | - Olivia C. Klatt
- Department of Chemistry and Pharmaceutical Sciences, Vrije Universiteit Amsterdam, De Boelelaan 1108, 1081 HZ Amsterdam, The Netherlands; (T.M.); (D.d.C.P.); (O.C.K.); (P.J.)
- Amsterdam Institute of Molecular and Life Sciences (AIMMS), Vrije Universiteit Amsterdam, De Boelelaan 1108, 1081 HZ Amsterdam, The Netherlands
| | - Joanne Buitenhuis
- Department of Chemistry and Pharmaceutical Sciences, Vrije Universiteit Amsterdam, De Boelelaan 1108, 1081 HZ Amsterdam, The Netherlands; (T.M.); (D.d.C.P.); (O.C.K.); (P.J.)
- Amsterdam Institute of Molecular and Life Sciences (AIMMS), Vrije Universiteit Amsterdam, De Boelelaan 1108, 1081 HZ Amsterdam, The Netherlands
| | - Paul Jennings
- Department of Chemistry and Pharmaceutical Sciences, Vrije Universiteit Amsterdam, De Boelelaan 1108, 1081 HZ Amsterdam, The Netherlands; (T.M.); (D.d.C.P.); (O.C.K.); (P.J.)
- Amsterdam Institute of Molecular and Life Sciences (AIMMS), Vrije Universiteit Amsterdam, De Boelelaan 1108, 1081 HZ Amsterdam, The Netherlands
| | - Anja Wilmes
- Department of Chemistry and Pharmaceutical Sciences, Vrije Universiteit Amsterdam, De Boelelaan 1108, 1081 HZ Amsterdam, The Netherlands; (T.M.); (D.d.C.P.); (O.C.K.); (P.J.)
- Amsterdam Institute of Molecular and Life Sciences (AIMMS), Vrije Universiteit Amsterdam, De Boelelaan 1108, 1081 HZ Amsterdam, The Netherlands
| |
Collapse
|
3
|
Pye K, Tasinato E, Shuttleworth S, Devlin C, Brown C. Comparison of the Impact of VRP-034 and Polymyxin B upon Markers of Kidney Injury in Human Proximal Tubule Monolayers In Vitro. Antibiotics (Basel) 2024; 13:530. [PMID: 38927196 PMCID: PMC11201133 DOI: 10.3390/antibiotics13060530] [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: 05/16/2024] [Revised: 06/01/2024] [Accepted: 06/01/2024] [Indexed: 06/28/2024] Open
Abstract
In this study, we assessed the impact of commercially available polymyxin B against VRP-034 (novel formulation of polymyxin B) using a validated in vitro human renal model, aProximateTM. Freshly isolated primary proximal tubule cells (PTCs) were cultured in Transwell plates and treated with various concentrations of the formulations for up to 48 h. The functional expression of megalin-cubilin receptors in PTC monolayers was validated using FITC-conjugated albumin uptake assays. Polymyxin B and VRP-034 were evaluated at six concentrations (0.3, 1, 3, 10, 30, and 60 µM), and nephrotoxicity was assessed through measurements of transepithelial electrical resistance (TEER), intracellular adenosine triphosphate (ATP) levels, lactate dehydrogenase (LDH) release, and novel injury biomarkers [kidney injury molecule-1 (KIM-1), neutrophil gelatinase-associated lipocalin (NGAL), and clusterin]. Additionally, histological analysis using annexin V apoptosis staining was performed. Our results indicated a significant decrease in TEER with polymyxin B at concentrations ≥10 μM compared to VRP-034. Toxic effects were observed from ATP and LDH release only at concentrations ≥30 μM for both formulations. Furthermore, injury biomarker release was higher with polymyxin B compared to VRP-034, particularly at concentrations ≥10 µM. Histologically, polymyxin B-treated PTCs showed increased apoptosis compared to VRP-034-treated cells. Overall, VRP-034 demonstrated improved tolerance in the aProximateTM model compared to polymyxin B, suggesting its potential as a safer alternative for renal protection.
Collapse
Affiliation(s)
- Keith Pye
- Newcells Biotech Ltd., The Biosphere, Newcastle-upon-Tyne NE4 5BX, UK
| | | | | | | | | |
Collapse
|
4
|
Farhang Doost N, Srivastava SK. A Comprehensive Review of Organ-on-a-Chip Technology and Its Applications. BIOSENSORS 2024; 14:225. [PMID: 38785699 PMCID: PMC11118005 DOI: 10.3390/bios14050225] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Revised: 04/09/2024] [Accepted: 04/23/2024] [Indexed: 05/25/2024]
Abstract
Organ-on-a-chip (OOC) is an emerging technology that simulates an artificial organ within a microfluidic cell culture chip. Current cell biology research focuses on in vitro cell cultures due to various limitations of in vivo testing. Unfortunately, in-vitro cell culturing fails to provide an accurate microenvironment, and in vivo cell culturing is expensive and has historically been a source of ethical controversy. OOC aims to overcome these shortcomings and provide the best of both in vivo and in vitro cell culture research. The critical component of the OOC design is utilizing microfluidics to ensure a stable concentration gradient, dynamic mechanical stress modeling, and accurate reconstruction of a cellular microenvironment. OOC also has the advantage of complete observation and control of the system, which is impossible to recreate in in-vivo research. Multiple throughputs, channels, membranes, and chambers are constructed in a polydimethylsiloxane (PDMS) array to simulate various organs on a chip. Various experiments can be performed utilizing OOC technology, including drug delivery research and toxicology. Current technological expansions involve multiple organ microenvironments on a single chip, allowing for studying inter-tissue interactions. Other developments in the OOC technology include finding a more suitable material as a replacement for PDMS and minimizing artefactual error and non-translatable differences.
Collapse
Affiliation(s)
| | - Soumya K. Srivastava
- Department of Chemical and Biomedical Engineering, West Virginia University, Morgantown, WV 26506, USA;
| |
Collapse
|
5
|
Tawade P, Mastrangeli M. Integrated Electrochemical and Optical Biosensing in Organs-on-Chip. Chembiochem 2024; 25:e202300560. [PMID: 37966365 DOI: 10.1002/cbic.202300560] [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: 08/09/2023] [Revised: 11/13/2023] [Accepted: 11/13/2023] [Indexed: 11/16/2023]
Abstract
Demand for biocompatible, non-invasive, and continuous real-time monitoring of organs-on-chip has driven the development of a variety of novel sensors. However, highest accuracy and sensitivity can arguably be achieved by integrated biosensing, which enables in situ monitoring of the in vitro microenvironment and dynamic responses of tissues and miniature organs recapitulated in organs-on-chip. This paper reviews integrated electrical, electrochemical, and optical sensing methods within organ-on-chip devices and platforms. By affording precise detection of analytes and biochemical reactions, these methods expand and advance the monitoring capabilities and reproducibility of organ-on-chip technology. The integration of these sensing techniques allows a deeper understanding of organ functions, and paves the way for important applications such as drug testing, disease modeling, and personalized medicine. By consolidating recent advancements and highlighting challenges in the field, this review aims to foster further research and innovation in the integration of biosensing in organs-on-chip.
Collapse
Affiliation(s)
- Pratik Tawade
- Electronic Components, Technology and Materials, Department of Microelectronics, Delft University of Technology, Mekelweg 4, 2628CD, Delft, Netherlands
| | - Massimo Mastrangeli
- Electronic Components, Technology and Materials, Department of Microelectronics, Delft University of Technology, Mekelweg 4, 2628CD, Delft, Netherlands
| |
Collapse
|
6
|
Mathialagan S, Chung G, Pye K, Rodrigues AD, Varma MVS, Brown C. Significance of Organic Anion Transporter 2 and Organic Cation Transporter 2 in Creatinine Clearance: Mechanistic Evaluation Using Freshly Prepared Human Primary Renal Proximal Tubule Cells. J Pharmacol Exp Ther 2024; 388:201-208. [PMID: 37977812 DOI: 10.1124/jpet.123.001890] [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: 08/18/2023] [Revised: 10/27/2023] [Accepted: 10/30/2023] [Indexed: 11/19/2023] Open
Abstract
Creatinine, a clinical marker for kidney function, is predominantly cleared by glomerular filtration, with active tubular secretion contributing to about 30% of its renal clearance. Recent studies suggested the potential involvement of organic anion transporter (OAT)2, in addition to the previously known organic cation transporter (OCT)2-mediated basolateral uptake, in creatinine active secretion. Here we characterized the transport mechanisms of creatinine using transfected human embryonic kidney (HEK)293 cells and freshly prepared human primary renal proximal tubule epithelial cells (hPTCs). Creatinine showed transport by OAT2 in transfected HEK293 cells. In addition, both creatinine and metformin showed transport by OCT2 and multidrug and toxin extrusion pump (MATE)1 and MATE2K, while penciclovir was selective for OAT2. Time-dependent cell accumulation was observed for creatinine and metformin in hPTCs. Their accumulation was increased by pyrimethamine but inhibited by decynium-22, likely due to differential inhibition of OCT2 versus MATEs. Additionally, indomethacin (an OAT2 inhibitor) reduced penciclovir uptake (∼75%) in hPTCs illustrating functional OAT2 activity. However, no modulation of creatinine and metformin cell accumulation was apparent with indomethacin. Creatinine transport characteristics in the presence of inhibitors approached those of metformin, an OCT2/MATE substrate, but were distinct from those of penciclovir, an OAT2-selective substrate. Moreover, indomethacin showed no significant effect on the basolateral-to-apical transport and net secretion of creatinine across hPTC monolayers. Collectively, the functional studies suggest OCT2 as the primary basolateral uptake mechanism and that OAT2 has a minimal role, in creatinine renal secretion. Our results highlight the utility of hPTCs to enable the functional assessment of renal transport mechanisms. SIGNIFICANCE STATEMENT: Our results obtained with primary hPTCs indicate that OCT2/MATE (vs. OAT2) play a major role in the active renal secretion of creatinine. Quantitative pharmacokinetic models should therefore focus on OCT2/MATE when describing serum creatinine and creatinine clearance modulation by inhibitor drugs and genotype- or disease-related activity changes. The present study highlights the utility of freshly isolated hPTCs to support solute carrier phenotyping to enable the functional assessment of renal transport mechanisms.
Collapse
Affiliation(s)
- Sumathy Mathialagan
- Medicine Design, Pfizer Research and Development, Pfizer Inc., Groton, Connecticut (S.M., A.D.R., M.V.S.V.); and Newcells Biotech Limited, The Biosphere, Newcastle Upton Tyne, United Kingdom (G.C., K.P., C.B.)
| | - Git Chung
- Medicine Design, Pfizer Research and Development, Pfizer Inc., Groton, Connecticut (S.M., A.D.R., M.V.S.V.); and Newcells Biotech Limited, The Biosphere, Newcastle Upton Tyne, United Kingdom (G.C., K.P., C.B.)
| | - Keith Pye
- Medicine Design, Pfizer Research and Development, Pfizer Inc., Groton, Connecticut (S.M., A.D.R., M.V.S.V.); and Newcells Biotech Limited, The Biosphere, Newcastle Upton Tyne, United Kingdom (G.C., K.P., C.B.)
| | - A David Rodrigues
- Medicine Design, Pfizer Research and Development, Pfizer Inc., Groton, Connecticut (S.M., A.D.R., M.V.S.V.); and Newcells Biotech Limited, The Biosphere, Newcastle Upton Tyne, United Kingdom (G.C., K.P., C.B.)
| | - Manthena V S Varma
- Medicine Design, Pfizer Research and Development, Pfizer Inc., Groton, Connecticut (S.M., A.D.R., M.V.S.V.); and Newcells Biotech Limited, The Biosphere, Newcastle Upton Tyne, United Kingdom (G.C., K.P., C.B.)
| | - Colin Brown
- Medicine Design, Pfizer Research and Development, Pfizer Inc., Groton, Connecticut (S.M., A.D.R., M.V.S.V.); and Newcells Biotech Limited, The Biosphere, Newcastle Upton Tyne, United Kingdom (G.C., K.P., C.B.)
| |
Collapse
|
7
|
Pisapia F, O’Brien D, Tasinato E, Garner KL, Brown CDA. Development of a Highly Differentiated Human Primary Proximal Tubule MPS Model (aProximate MPS Flow). Bioengineering (Basel) 2023; 11:7. [PMID: 38275575 PMCID: PMC10813028 DOI: 10.3390/bioengineering11010007] [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: 11/20/2023] [Revised: 12/14/2023] [Accepted: 12/19/2023] [Indexed: 01/27/2024] Open
Abstract
The kidney proximal tubule (PT) mediates renal drug elimination in vivo and is a major site of drug-induced toxicity. To reliably assess drug efficacy, it is crucial to construct a model in which PT functions are replicated. Current animal studies have proven poorly predictive of human outcome. To address this, we developed a physiologically relevant micro-physiological system (MPS) model of the human PT, the aProximate MPS Flow platform (Patent No: G001336.GB). In this model, primary human PT cells (hPTCs) are subjected to fluidic media flow and a shear stress of 0.01-0.2 Pa. We observe that these cells replicate the polarity of hPTCs and exhibit a higher expression of all the key transporters of SLC22A6 (OAT1), SLC22A8 (OAT3), SLC22A2 (OCT2), SLC47A1 (MATE1), SLC22A12 (URAT1), SLC2A9 (GLUT9), ABCB1 (MDR1), ABCC2 (MRP2), LRP2 (megalin), CUBN (cubilin), compared with cells grown under static conditions. Immunofluorescence microscopy confirmed an increase in OAT1, OAT3, and cilia protein expression. Increased sensitivity to nephrotoxic protein cisplatin was observed; creatinine and FITC-albumin uptake was significantly increased under fluidic shear stress conditions. Taken together, these data suggest that growing human PT cells under media flow significantly improves the phenotype and function of hPTC monolayers and has benefits to the utility and near-physiology of the model.
Collapse
Affiliation(s)
- Francesca Pisapia
- Newcells Biotech Ltd., The Biosphere, Draymans Way, Newcastle Helix, Newcastle upon Tyne NE4 5BX, UK; (D.O.); (E.T.); (C.D.A.B.)
| | - Donovan O’Brien
- Newcells Biotech Ltd., The Biosphere, Draymans Way, Newcastle Helix, Newcastle upon Tyne NE4 5BX, UK; (D.O.); (E.T.); (C.D.A.B.)
| | - Elena Tasinato
- Newcells Biotech Ltd., The Biosphere, Draymans Way, Newcastle Helix, Newcastle upon Tyne NE4 5BX, UK; (D.O.); (E.T.); (C.D.A.B.)
- Institute of Genetic Medicine, Newcastle University, International Centre for Life, Central Parkway, Newcastle upon Tyne NE1 3BZ, UK
| | - Kathryn L. Garner
- Newcells Biotech Ltd., The Biosphere, Draymans Way, Newcastle Helix, Newcastle upon Tyne NE4 5BX, UK; (D.O.); (E.T.); (C.D.A.B.)
| | - Colin D. A. Brown
- Newcells Biotech Ltd., The Biosphere, Draymans Way, Newcastle Helix, Newcastle upon Tyne NE4 5BX, UK; (D.O.); (E.T.); (C.D.A.B.)
| |
Collapse
|
8
|
Veiga-Matos J, Morales AI, Prieto M, Remião F, Silva R. Study Models of Drug-Drug Interactions Involving P-Glycoprotein: The Potential Benefit of P-Glycoprotein Modulation at the Kidney and Intestinal Levels. Molecules 2023; 28:7532. [PMID: 38005253 PMCID: PMC10673607 DOI: 10.3390/molecules28227532] [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: 10/11/2023] [Revised: 11/03/2023] [Accepted: 11/03/2023] [Indexed: 11/26/2023] Open
Abstract
P-glycoprotein (P-gp) is a crucial membrane transporter situated on the cell's apical surface, being responsible for eliminating xenobiotics and endobiotics. P-gp modulators are compounds that can directly or indirectly affect this protein, leading to changes in its expression and function. These modulators can act as inhibitors, inducers, or activators, potentially causing drug-drug interactions (DDIs). This comprehensive review explores diverse models and techniques used to assess drug-induced P-gp modulation. We cover several approaches, including in silico, in vitro, ex vivo, and in vivo methods, with their respective strengths and limitations. Additionally, we explore the therapeutic implications of DDIs involving P-gp, with a special focus on the renal and intestinal elimination of P-gp substrates. This involves enhancing the removal of toxic substances from proximal tubular epithelial cells into the urine or increasing the transport of compounds from enterocytes into the intestinal lumen, thereby facilitating their excretion in the feces. A better understanding of these interactions, and of the distinct techniques applied for their study, will be of utmost importance for optimizing drug therapy, consequently minimizing drug-induced adverse and toxic effects.
Collapse
Affiliation(s)
- Jéssica Veiga-Matos
- UCIBIO-Applied Molecular Biosciences Unit, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal;
- Associate Laboratory i4HB—Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
- Toxicology Unit (Universidad de Salamanca), Group of Translational Research on Renal and Cardiovascular Diseases (TRECARD), Institute of Biomedical Research of Salamanca (IBSAL), 37007 Salamanca, Spain; (A.I.M.); (M.P.)
| | - Ana I. Morales
- Toxicology Unit (Universidad de Salamanca), Group of Translational Research on Renal and Cardiovascular Diseases (TRECARD), Institute of Biomedical Research of Salamanca (IBSAL), 37007 Salamanca, Spain; (A.I.M.); (M.P.)
| | - Marta Prieto
- Toxicology Unit (Universidad de Salamanca), Group of Translational Research on Renal and Cardiovascular Diseases (TRECARD), Institute of Biomedical Research of Salamanca (IBSAL), 37007 Salamanca, Spain; (A.I.M.); (M.P.)
| | - Fernando Remião
- UCIBIO-Applied Molecular Biosciences Unit, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal;
- Associate Laboratory i4HB—Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
| | - Renata Silva
- UCIBIO-Applied Molecular Biosciences Unit, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal;
- Associate Laboratory i4HB—Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
| |
Collapse
|
9
|
Caetano-Pinto P, Stahl SH. Renal Organic Anion Transporters 1 and 3 In Vitro: Gone but Not Forgotten. Int J Mol Sci 2023; 24:15419. [PMID: 37895098 PMCID: PMC10607849 DOI: 10.3390/ijms242015419] [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: 07/26/2023] [Revised: 10/12/2023] [Accepted: 10/17/2023] [Indexed: 10/29/2023] Open
Abstract
Organic anion transporters 1 and 3 (OAT1 and OAT3) play a crucial role in kidney function by regulating the secretion of multiple renally cleared small molecules and toxic metabolic by-products. Assessing the activity of these transporters is essential for drug development purposes as they can significantly impact drug disposition and safety. OAT1 and OAT3 are amongst the most abundant drug transporters expressed in human renal proximal tubules. However, their expression is lost when cells are isolated and cultured in vitro, which is a persistent issue across all human and animal renal proximal tubule cell models, including primary cells and cell lines. Although it is well known that the overall expression of drug transporters is affected in vitro, the underlying reasons for the loss of OAT1 and OAT3 are still not fully understood. Nonetheless, research into the regulatory mechanisms of these transporters has provided insights into the molecular pathways underlying their expression and activity. In this review, we explore the regulatory mechanisms that govern the expression and activity of OAT1 and OAT3 and investigate the physiological changes that proximal tubule cells undergo and that potentially result in the loss of these transporters. A better understanding of the regulation of these transporters could aid in the development of strategies, such as introducing microfluidic conditions or epigenetic modification inhibitors, to improve their expression and activity in vitro and to create more physiologically relevant models. Consequently, this will enable more accurate assessment for drug development and safety applications.
Collapse
Affiliation(s)
- Pedro Caetano-Pinto
- Department of Urology, University Medicine Greifswald, Ferdinand-Sauerbruch-Straße, 17475 Greifswald, Germany
| | - Simone H. Stahl
- CVRM Safety, Clinical Pharmacology and Safety Sciences, R&D, AstraZeneca, 310 Darwin Building, Cambridge Science Park, Milton Road, Cambridge CB4 0WG, UK;
| |
Collapse
|
10
|
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
|
11
|
The next frontier in ADME science: Predicting transporter-based drug disposition, tissue concentrations and drug-drug interactions in humans. Pharmacol Ther 2022; 238:108271. [DOI: 10.1016/j.pharmthera.2022.108271] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2022] [Revised: 08/05/2022] [Accepted: 08/17/2022] [Indexed: 12/25/2022]
|
12
|
Apical Medium Flow Influences the Morphology and Physiology of Human Proximal Tubular Cells in a Microphysiological System. BIOENGINEERING (BASEL, SWITZERLAND) 2022; 9:bioengineering9100516. [PMID: 36290484 PMCID: PMC9598399 DOI: 10.3390/bioengineering9100516] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Accepted: 09/16/2022] [Indexed: 12/28/2022]
Abstract
There is a lack of physiologically relevant in vitro human kidney models for disease modelling and detecting drug-induced effects given the limited choice of cells and difficulty implementing quasi-physiological culture conditions. We investigated the influence of fluid shear stress on primary human renal proximal tubule epithelial cells (RPTECs) cultured in the micro-physiological Vitrofluid device. This system houses cells seeded on semipermeable membranes and can be connected to a regulable pump that enables controlled, unidirectional flow. After 7 days in culture, RPTECs maintained physiological characteristics such as barrier integrity, protein uptake ability, and expression of specific transporters (e.g., aquaporin-1). Exposure to constant apical side flow did not cause cytotoxicity, cell detachment, or intracellular reactive oxygen species accumulation. However, unidirectional flow profoundly affected cell morphology and led to primary cilia lengthening and alignment in the flow direction. The dynamic conditions also reduced cell proliferation, altered plasma membrane leakiness, increased cytokine secretion, and repressed histone deacetylase 6 and kidney injury molecule 1 expression. Cells under flow also remained susceptible to colistin-induced toxicity. Collectively, the results suggest that dynamic culture conditions in the Vitrofluid system promote a more differentiated phenotype in primary human RPTECs and represent an improved in vitro kidney model.
Collapse
|
13
|
Differentiated kidney tubular cell-derived extracellular vesicles enhance maturation of tubuloids. J Nanobiotechnology 2022; 20:326. [PMID: 35841001 PMCID: PMC9284832 DOI: 10.1186/s12951-022-01506-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Accepted: 06/09/2022] [Indexed: 12/04/2022] Open
Abstract
The prevalence of end-stage kidney disease (ESKD) is rapidly increasing with the need for regenerative therapies. Adult stem cell derived kidney tubuloids have the potential to functionally mimic the adult kidney tubule, but still lack the expression of important transport proteins needed for waste removal. Here, we investigated the potential of extracellular vesicles (EVs) obtained from matured kidney tubular epithelial cells to modulate in vitro tubuloids functional maturation. We focused on organic anion transporter 1 (OAT1), one of the most important proteins involved in endogenous waste excretion. First, we show that EVs from engineered proximal tubule cells increased the expression of several transcription factors and epithelial transporters, resulting in improved OAT1 transport capacity. Next, a more in-depth proteomic data analysis showed that EVs can trigger various biological pathways, including mesenchymal-to-epithelial transition, which is crucial in the tubular epithelial maturation. Moreover, we demonstrated that the combination of EVs and tubuloid-derived cells can be used as part of a bioartificial kidney to generate a tight polarized epithelial monolayer with formation of dense cilia structures. In conclusion, EVs from kidney tubular epithelial cells can phenotypically improve in vitro tubuloid maturation, thereby enhancing their potential as functional units in regenerative or renal replacement therapies.
Collapse
|
14
|
Gledhill A, Bowen R, Bartels M, Bond A, Chung G, Brown CDA, Pye K, Vora T. The chlorophenoxy herbicide MCPA: A mechanistic basis for the observed differences in toxicological profile in humans and rats versus dogs. Xenobiotica 2022; 52:498-510. [PMID: 35822285 DOI: 10.1080/00498254.2022.2100842] [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: 10/17/2022]
Abstract
1. Metabolism data for MCPA in rat, dog and human shows a single oral dose is quantitatively and rapidly absorbed with evidence of non-linear kinetics at >100 mg/kg bw. The extent of metabolism is low and consistent between rat and human, with substantially higher metabolic conversion in dog. Parent accounts for 50-67% dose in rat, ∼40% in human and 2-27% in dog. No dog specific metabolite is apparent.2. In rat and human, MCPA and metabolites are rapidly eliminated in urine (65-70% within 24 hours) but in dog, excretion is via urine and faeces (20-30% within 24 hours), with renal excretion saturating between 5 and 100 mg.kg bw.3. The species difference in excretion is reflected in pharmacokinetics. Terminal half-life is similar in rat and human (15-17 hour) but higher in dog (47 hour). Modelling shows species differences in single dose kinetics profoundly affect systemic exposure following repeat dosing.4. The difference in renal excretion and systemic exposure of MCPA between dogs and rats has been attributed to species differences in active transporters (OAT1/OAT3). A new in vitro flux study in renal proximal tubules supports this hypothesis with net secretion in rat and human of a similar magnitude but significantly less in dog.
Collapse
Affiliation(s)
| | | | | | | | - Git Chung
- Newcells Biotech, Newcastle upon Tyne, UK
| | | | - Keith Pye
- Newcells Biotech, Newcastle upon Tyne, UK
| | - Tarang Vora
- Simulations Plus, Lancaster, California, USA
| |
Collapse
|
15
|
Uda J, Ashizawa N, Iwanaga T. An evaluation method for uric acid uptake inhibition using primary human proximal tubule epithelial cells treated with insulin. NUCLEOSIDES, NUCLEOTIDES & NUCLEIC ACIDS 2022; 41:724-735. [PMID: 35770496 DOI: 10.1080/15257770.2022.2070204] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 04/07/2022] [Accepted: 04/20/2022] [Indexed: 06/15/2023]
Abstract
The effects of uricosuric agents have been evaluated in vitro with indices of uric acid uptake into human urate transporter 1 (URAT1)-overexpressed oocytes or cells. In the present study, we evaluated a method using primary human renal proximal tubule epithelial cells (RPTECs). Pretreatment of RPTECs with insulin significantly increased the uptake of uric acid into these cells. The uric acid uptake was inhibited in a concentration-dependent manner by the URAT1 inhibitors benzbromarone and dotinurad. Therefore, effects of uricosuric agents can be evaluated by the novel method, which is closer to the physiological system compared with previous methods.
Collapse
Affiliation(s)
- Junichiro Uda
- Medicinal Chemistry Research Department, Research Institute, FUJIYAKUHIN CO., Ltd, Saitama, Japan
| | - Naoki Ashizawa
- Biological Research Department, Research Institute, FUJIYAKUHIN CO., Ltd, Saitama, Japan
| | - Takashi Iwanaga
- Biological Research Department, Research Institute, FUJIYAKUHIN CO., Ltd, Saitama, Japan
| |
Collapse
|
16
|
Hewitt NJ, Troutman J, Przibilla J, Schepky A, Ouédraogo G, Mahony C, Kenna G, Varçin M, Dent MP. Use of in vitro metabolism and biokinetics assays to refine predicted in vivo and in vitro internal exposure to the cosmetic ingredient, phenoxyethanol, for use in risk assessment. Regul Toxicol Pharmacol 2022; 131:105132. [PMID: 35217105 DOI: 10.1016/j.yrtph.2022.105132] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Revised: 12/20/2021] [Accepted: 01/31/2022] [Indexed: 01/04/2023]
Abstract
A novel approach was developed to help characterize the biokinetics of the cosmetic ingredient, phenoxyethanol, to help assess the safety of the parent and its major stable metabolite. In the first step of this non-animal tiered approach, primary human hepatocytes were used to confirm or refute in silico predicted metabolites, and elucidate the intrinsic clearance of phenoxyethanol. A key result was the identification of the major metabolite, phenoxyacetic acid (PAA), the exposure to which in the kidney was subsequently predicted to far exceed that of phenoxyethanol in blood or other tissues. Therefore, a novel aspect of this approach was to measure in the subsequent step the formation of PAA in the cells dosed with phenoxyethanol that were used to provide points of departure (PoDs) and express the intracellular exposure as the Cmax and AUC24. This enabled the calculation of the intracellular concentrations of parent and metabolite at the PoD in the cells used to derive this value. These concentrations can be compared with in vivo tissue levels to conclude on the safety margin. The lessons from this case study will help to inform the design of other non-animal safety assessments.
Collapse
Affiliation(s)
- Nicola J Hewitt
- Cosmetics Europe, Avenue Herrmann-Debroux 40, 1160, Auderghem, Belgium
| | | | - Julia Przibilla
- Pharmacelsus GmbH, Science Park 2, D-66123, Saarbrücken, Germany
| | | | - Gladys Ouédraogo
- L'Oréal, Research & Innovation, 9 rue Pierre Dreyfus, 92110, Clichy, France
| | | | - Gerry Kenna
- Drug Safety Consultant, 2 Farmfield Drive, Macclesfield, Cheshire, SK10 2TJ, UK
| | - Mustafa Varçin
- Cosmetics Europe, Avenue Herrmann-Debroux 40, 1160, Auderghem, Belgium
| | - Mathew P Dent
- Unilever Safety and Environmental Assurance Centre, Colworth Science Park, Sharnbrook, Bedfordshire, MK44 1LQ, UK
| |
Collapse
|
17
|
Interdependent Transcription of a Natural Sense/Antisense Transcripts Pair (SLC34A1/PFN3). Noncoding RNA 2022; 8:ncrna8010019. [PMID: 35202092 PMCID: PMC8877773 DOI: 10.3390/ncrna8010019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 01/17/2022] [Accepted: 02/08/2022] [Indexed: 11/17/2022] Open
Abstract
Natural antisense transcripts (NATs) constitute a significant group of regulatory, long noncoding RNAs. They are prominently expressed in testis but are also detectable in other organs. NATs are transcribed at low levels and co-expressed with related protein coding sense transcripts. Nowadays NATs are generally considered as regulatory, long noncoding RNAs without closer focus on the inevitable interference between sense and antisense expression. This work describes a cellular system where sense and antisense transcription of a specific locus (SLC34A1/PFN3) is induced using epigenetic modifiers and CRISPR-Cas9. The renal cell lines HEK293 and HKC-8 do not express SLC34A1/PFN3 under normal culture conditions. Five-day exposure to dexamethasone significantly stimulates sense transcript (SLC34A1) levels and antisense (PFN3) minimally; the effect is only seen in HEK293 cells. Enhanced expression is paralleled by reduced sense promoter methylation and an increase in activating histone marks. Expression is further modulated by cassettes that stimulate the expression of sense or antisense transcript but disrupt protein coding potential. Constitutive expression of a 5′-truncated SLC34A1 transcript increases sense expression independent of dexamethasone induction but also stimulates antisense expression. Concordant expression is confirmed with the antisense knock-in that also enhances sense expression. The antisense effect acts on transcription in cis since transient transfection with sense or antisense constructs fails to stimulate the expression of the opposite transcript. These results suggest that bi-directional transcription of the SLC34A1/PFN3 locus has a stimulatory influence on the expression of the opposite transcript involving epigenetic changes of the promoters. In perspective of extensive, previous research into bi-directionally transcribed SLC34A loci, the findings underpin a hypothesis where NATs display different biological roles in soma and germ cells. Accordingly, we propose that in somatic cells, NATs act like lncRNAs–with the benefit of close proximity to a potential target gene. In germ cells, however, recent evidence suggests different biological roles for NATs that require RNA complementarity and double-stranded RNA formation.
Collapse
|
18
|
Youhanna S, Kemas AM, Preiss L, Zhou Y, Shen JX, Cakal SD, Paqualini FS, Goparaju SK, Shafagh RZ, Lind JU, Sellgren CM, Lauschke VM. Organotypic and Microphysiological Human Tissue Models for Drug Discovery and Development-Current State-of-the-Art and Future Perspectives. Pharmacol Rev 2022; 74:141-206. [PMID: 35017176 DOI: 10.1124/pharmrev.120.000238] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Accepted: 10/12/2021] [Indexed: 12/11/2022] Open
Abstract
The number of successful drug development projects has been stagnant for decades despite major breakthroughs in chemistry, molecular biology, and genetics. Unreliable target identification and poor translatability of preclinical models have been identified as major causes of failure. To improve predictions of clinical efficacy and safety, interest has shifted to three-dimensional culture methods in which human cells can retain many physiologically and functionally relevant phenotypes for extended periods of time. Here, we review the state of the art of available organotypic culture techniques and critically review emerging models of human tissues with key importance for pharmacokinetics, pharmacodynamics, and toxicity. In addition, developments in bioprinting and microfluidic multiorgan cultures to emulate systemic drug disposition are summarized. We close by highlighting important trends regarding the fabrication of organotypic culture platforms and the choice of platform material to limit drug absorption and polymer leaching while supporting the phenotypic maintenance of cultured cells and allowing for scalable device fabrication. We conclude that organotypic and microphysiological human tissue models constitute promising systems to promote drug discovery and development by facilitating drug target identification and improving the preclinical evaluation of drug toxicity and pharmacokinetics. There is, however, a critical need for further validation, benchmarking, and consolidation efforts ideally conducted in intersectoral multicenter settings to accelerate acceptance of these novel models as reliable tools for translational pharmacology and toxicology. SIGNIFICANCE STATEMENT: Organotypic and microphysiological culture of human cells has emerged as a promising tool for preclinical drug discovery and development that might be able to narrow the translation gap. This review discusses recent technological and methodological advancements and the use of these systems for hit discovery and the evaluation of toxicity, clearance, and absorption of lead compounds.
Collapse
Affiliation(s)
- Sonia Youhanna
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden (S.Y., A.M.K., L.P., Y.Z., J.X.S., S.K.G., R.Z.S., C.M.S., V.M.L.); Department of Drug Metabolism and Pharmacokinetics (DMPK), Merck KGaA, Darmstadt, Germany (L.P.); Department of Health Technology, Technical University of Denmark, Lyngby, Denmark (S.D.C., J.U.L.); Synthetic Physiology Laboratory, Department of Civil Engineering and Architecture, University of Pavia, Pavia, Italy (F.S.P.); Division of Micro- and Nanosystems, KTH Royal Institute of Technology, Stockholm, Sweden (Z.S.); and Dr Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany (V.M.L.)
| | - Aurino M Kemas
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden (S.Y., A.M.K., L.P., Y.Z., J.X.S., S.K.G., R.Z.S., C.M.S., V.M.L.); Department of Drug Metabolism and Pharmacokinetics (DMPK), Merck KGaA, Darmstadt, Germany (L.P.); Department of Health Technology, Technical University of Denmark, Lyngby, Denmark (S.D.C., J.U.L.); Synthetic Physiology Laboratory, Department of Civil Engineering and Architecture, University of Pavia, Pavia, Italy (F.S.P.); Division of Micro- and Nanosystems, KTH Royal Institute of Technology, Stockholm, Sweden (Z.S.); and Dr Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany (V.M.L.)
| | - Lena Preiss
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden (S.Y., A.M.K., L.P., Y.Z., J.X.S., S.K.G., R.Z.S., C.M.S., V.M.L.); Department of Drug Metabolism and Pharmacokinetics (DMPK), Merck KGaA, Darmstadt, Germany (L.P.); Department of Health Technology, Technical University of Denmark, Lyngby, Denmark (S.D.C., J.U.L.); Synthetic Physiology Laboratory, Department of Civil Engineering and Architecture, University of Pavia, Pavia, Italy (F.S.P.); Division of Micro- and Nanosystems, KTH Royal Institute of Technology, Stockholm, Sweden (Z.S.); and Dr Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany (V.M.L.)
| | - Yitian Zhou
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden (S.Y., A.M.K., L.P., Y.Z., J.X.S., S.K.G., R.Z.S., C.M.S., V.M.L.); Department of Drug Metabolism and Pharmacokinetics (DMPK), Merck KGaA, Darmstadt, Germany (L.P.); Department of Health Technology, Technical University of Denmark, Lyngby, Denmark (S.D.C., J.U.L.); Synthetic Physiology Laboratory, Department of Civil Engineering and Architecture, University of Pavia, Pavia, Italy (F.S.P.); Division of Micro- and Nanosystems, KTH Royal Institute of Technology, Stockholm, Sweden (Z.S.); and Dr Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany (V.M.L.)
| | - Joanne X Shen
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden (S.Y., A.M.K., L.P., Y.Z., J.X.S., S.K.G., R.Z.S., C.M.S., V.M.L.); Department of Drug Metabolism and Pharmacokinetics (DMPK), Merck KGaA, Darmstadt, Germany (L.P.); Department of Health Technology, Technical University of Denmark, Lyngby, Denmark (S.D.C., J.U.L.); Synthetic Physiology Laboratory, Department of Civil Engineering and Architecture, University of Pavia, Pavia, Italy (F.S.P.); Division of Micro- and Nanosystems, KTH Royal Institute of Technology, Stockholm, Sweden (Z.S.); and Dr Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany (V.M.L.)
| | - Selgin D Cakal
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden (S.Y., A.M.K., L.P., Y.Z., J.X.S., S.K.G., R.Z.S., C.M.S., V.M.L.); Department of Drug Metabolism and Pharmacokinetics (DMPK), Merck KGaA, Darmstadt, Germany (L.P.); Department of Health Technology, Technical University of Denmark, Lyngby, Denmark (S.D.C., J.U.L.); Synthetic Physiology Laboratory, Department of Civil Engineering and Architecture, University of Pavia, Pavia, Italy (F.S.P.); Division of Micro- and Nanosystems, KTH Royal Institute of Technology, Stockholm, Sweden (Z.S.); and Dr Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany (V.M.L.)
| | - Francesco S Paqualini
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden (S.Y., A.M.K., L.P., Y.Z., J.X.S., S.K.G., R.Z.S., C.M.S., V.M.L.); Department of Drug Metabolism and Pharmacokinetics (DMPK), Merck KGaA, Darmstadt, Germany (L.P.); Department of Health Technology, Technical University of Denmark, Lyngby, Denmark (S.D.C., J.U.L.); Synthetic Physiology Laboratory, Department of Civil Engineering and Architecture, University of Pavia, Pavia, Italy (F.S.P.); Division of Micro- and Nanosystems, KTH Royal Institute of Technology, Stockholm, Sweden (Z.S.); and Dr Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany (V.M.L.)
| | - Sravan K Goparaju
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden (S.Y., A.M.K., L.P., Y.Z., J.X.S., S.K.G., R.Z.S., C.M.S., V.M.L.); Department of Drug Metabolism and Pharmacokinetics (DMPK), Merck KGaA, Darmstadt, Germany (L.P.); Department of Health Technology, Technical University of Denmark, Lyngby, Denmark (S.D.C., J.U.L.); Synthetic Physiology Laboratory, Department of Civil Engineering and Architecture, University of Pavia, Pavia, Italy (F.S.P.); Division of Micro- and Nanosystems, KTH Royal Institute of Technology, Stockholm, Sweden (Z.S.); and Dr Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany (V.M.L.)
| | - Reza Zandi Shafagh
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden (S.Y., A.M.K., L.P., Y.Z., J.X.S., S.K.G., R.Z.S., C.M.S., V.M.L.); Department of Drug Metabolism and Pharmacokinetics (DMPK), Merck KGaA, Darmstadt, Germany (L.P.); Department of Health Technology, Technical University of Denmark, Lyngby, Denmark (S.D.C., J.U.L.); Synthetic Physiology Laboratory, Department of Civil Engineering and Architecture, University of Pavia, Pavia, Italy (F.S.P.); Division of Micro- and Nanosystems, KTH Royal Institute of Technology, Stockholm, Sweden (Z.S.); and Dr Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany (V.M.L.)
| | - Johan Ulrik Lind
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden (S.Y., A.M.K., L.P., Y.Z., J.X.S., S.K.G., R.Z.S., C.M.S., V.M.L.); Department of Drug Metabolism and Pharmacokinetics (DMPK), Merck KGaA, Darmstadt, Germany (L.P.); Department of Health Technology, Technical University of Denmark, Lyngby, Denmark (S.D.C., J.U.L.); Synthetic Physiology Laboratory, Department of Civil Engineering and Architecture, University of Pavia, Pavia, Italy (F.S.P.); Division of Micro- and Nanosystems, KTH Royal Institute of Technology, Stockholm, Sweden (Z.S.); and Dr Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany (V.M.L.)
| | - Carl M Sellgren
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden (S.Y., A.M.K., L.P., Y.Z., J.X.S., S.K.G., R.Z.S., C.M.S., V.M.L.); Department of Drug Metabolism and Pharmacokinetics (DMPK), Merck KGaA, Darmstadt, Germany (L.P.); Department of Health Technology, Technical University of Denmark, Lyngby, Denmark (S.D.C., J.U.L.); Synthetic Physiology Laboratory, Department of Civil Engineering and Architecture, University of Pavia, Pavia, Italy (F.S.P.); Division of Micro- and Nanosystems, KTH Royal Institute of Technology, Stockholm, Sweden (Z.S.); and Dr Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany (V.M.L.)
| | - Volker M Lauschke
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden (S.Y., A.M.K., L.P., Y.Z., J.X.S., S.K.G., R.Z.S., C.M.S., V.M.L.); Department of Drug Metabolism and Pharmacokinetics (DMPK), Merck KGaA, Darmstadt, Germany (L.P.); Department of Health Technology, Technical University of Denmark, Lyngby, Denmark (S.D.C., J.U.L.); Synthetic Physiology Laboratory, Department of Civil Engineering and Architecture, University of Pavia, Pavia, Italy (F.S.P.); Division of Micro- and Nanosystems, KTH Royal Institute of Technology, Stockholm, Sweden (Z.S.); and Dr Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany (V.M.L.)
| |
Collapse
|
19
|
Yu P, Duan Z, Liu S, Pachon I, Ma J, Hemstreet GP, Zhang Y. Drug-Induced Nephrotoxicity Assessment in 3D Cellular Models. MICROMACHINES 2021; 13:mi13010003. [PMID: 35056167 PMCID: PMC8780064 DOI: 10.3390/mi13010003] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/31/2021] [Revised: 12/11/2021] [Accepted: 12/17/2021] [Indexed: 12/19/2022]
Abstract
The kidneys are often involved in adverse effects and toxicity caused by exposure to foreign compounds, chemicals, and drugs. Early predictions of these influences are essential to facilitate new, safe drugs to enter the market. However, in current drug treatments, drug-induced nephrotoxicity accounts for 1/4 of reported serious adverse reactions, and 1/3 of them are attributable to antibiotics. Drug-induced nephrotoxicity is driven by multiple mechanisms, including altered glomerular hemodynamics, renal tubular cytotoxicity, inflammation, crystal nephropathy, and thrombotic microangiopathy. Although the functional proteins expressed by renal tubules that mediate drug sensitivity are well known, current in vitro 2D cell models do not faithfully replicate the morphology and intact renal tubule function, and therefore, they do not replicate in vivo nephrotoxicity. The kidney is delicate and complex, consisting of a filter unit and a tubular part, which together contain more than 20 different cell types. The tubular epithelium is highly polarized, and maintaining cellular polarity is essential for the optimal function and response to environmental signals. Cell polarity depends on the communication between cells, including paracrine and autocrine signals, as well as biomechanical and chemotaxis processes. These processes affect kidney cell proliferation, migration, and differentiation. For drug disposal research, the microenvironment is essential for predicting toxic reactions. This article reviews the mechanism of drug-induced kidney injury, the types of nephrotoxicity models (in vivo and in vitro models), and the research progress related to drug-induced nephrotoxicity in three-dimensional (3D) cellular culture models.
Collapse
Affiliation(s)
- Pengfei Yu
- Difficult & Complicated Liver Diseases and Artificial Liver Center, Fourth Department of Liver Disease, Beijing Youan Hospital, Capital Medical University, Beijing 100069, China; (P.Y.); (Z.D.); (S.L.)
- Beijing Municipal Key Laboratory of Liver Failure and Artificial Liver Treatment Research, Beijing Youan Hospital, Capital Medical University, Beijing 100069, China
| | - Zhongping Duan
- Difficult & Complicated Liver Diseases and Artificial Liver Center, Fourth Department of Liver Disease, Beijing Youan Hospital, Capital Medical University, Beijing 100069, China; (P.Y.); (Z.D.); (S.L.)
- Beijing Municipal Key Laboratory of Liver Failure and Artificial Liver Treatment Research, Beijing Youan Hospital, Capital Medical University, Beijing 100069, China
| | - Shuang Liu
- Difficult & Complicated Liver Diseases and Artificial Liver Center, Fourth Department of Liver Disease, Beijing Youan Hospital, Capital Medical University, Beijing 100069, China; (P.Y.); (Z.D.); (S.L.)
- Beijing Municipal Key Laboratory of Liver Failure and Artificial Liver Treatment Research, Beijing Youan Hospital, Capital Medical University, Beijing 100069, China
| | - Ivan Pachon
- Wake Forest Institute for Regenerative Medicine, Wake Forest University Health Sciences, Winston-Salem, NC 27157, USA;
| | - Jianxing Ma
- Department of Biochemistry, Wake Forest University Health Sciences, Winston-Salem, NC 27157, USA;
| | | | - Yuanyuan Zhang
- Wake Forest Institute for Regenerative Medicine, Wake Forest University Health Sciences, Winston-Salem, NC 27157, USA;
- Correspondence: ; Tel.: +1-336-713-1189
| |
Collapse
|
20
|
Imaoka T, Huang W, Shum S, Hailey DW, Chang SY, Chapron A, Yeung CK, Himmelfarb J, Isoherranen N, Kelly EJ. Bridging the gap between in silico and in vivo by modeling opioid disposition in a kidney proximal tubule microphysiological system. Sci Rep 2021; 11:21356. [PMID: 34725352 PMCID: PMC8560754 DOI: 10.1038/s41598-021-00338-y] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Accepted: 09/27/2021] [Indexed: 11/22/2022] Open
Abstract
Opioid overdose, dependence, and addiction are a major public health crisis. Patients with chronic kidney disease (CKD) are at high risk of opioid overdose, therefore novel methods that provide accurate prediction of renal clearance (CLr) and systemic disposition of opioids in CKD patients can facilitate the optimization of therapeutic regimens. The present study aimed to predict renal clearance and systemic disposition of morphine and its active metabolite morphine-6-glucuronide (M6G) in CKD patients using a vascularized human proximal tubule microphysiological system (VPT-MPS) coupled with a parent-metabolite full body physiologically-based pharmacokinetic (PBPK) model. The VPT-MPS, populated with a human umbilical vein endothelial cell (HUVEC) channel and an adjacent human primary proximal tubular epithelial cells (PTEC) channel, successfully demonstrated secretory transport of morphine and M6G from the HUVEC channel into the PTEC channel. The in vitro data generated by VPT-MPS were incorporated into a mechanistic kidney model and parent-metabolite full body PBPK model to predict CLr and systemic disposition of morphine and M6G, resulting in successful prediction of CLr and the plasma concentration–time profiles in both healthy subjects and CKD patients. A microphysiological system together with mathematical modeling successfully predicted renal clearance and systemic disposition of opioids in CKD patients and healthy subjects.
Collapse
Affiliation(s)
- Tomoki Imaoka
- Department of Pharmaceutics, School of Pharmacy, University of Washington, HSB Room H272, 1959 NE Pacific Street, Seattle, WA, 98195, USA
| | - Weize Huang
- Department of Pharmaceutics, School of Pharmacy, University of Washington, HSB Room H272, 1959 NE Pacific Street, Seattle, WA, 98195, USA
| | - Sara Shum
- Department of Pharmaceutics, School of Pharmacy, University of Washington, HSB Room H272, 1959 NE Pacific Street, Seattle, WA, 98195, USA
| | - Dale W Hailey
- Lynn and Mike Garvey Imaging Core, Institute for Stem Cell and Regenerative Medicine, Seattle, WA, 98109, USA.,Department of Laboratory Medicine and Pathology, School of Medicine, University of Washington, Seattle, WA, 98195, USA
| | - Shih-Yu Chang
- Department of Pharmacy, School of Pharmacy, University of Washington, Seattle, WA, 98195, USA
| | - Alenka Chapron
- Department of Pharmaceutics, School of Pharmacy, University of Washington, HSB Room H272, 1959 NE Pacific Street, Seattle, WA, 98195, USA
| | - Catherine K Yeung
- Department of Pharmacy, School of Pharmacy, University of Washington, Seattle, WA, 98195, USA.,Division of Nephrology, Department of Medicine, Kidney Research Institute, University of Washington, 1959 NE Pacific Street, HSB Room H272, Seattle, WA, 98195, USA
| | - Jonathan Himmelfarb
- Division of Nephrology, Department of Medicine, Kidney Research Institute, University of Washington, 1959 NE Pacific Street, HSB Room H272, Seattle, WA, 98195, USA
| | - Nina Isoherranen
- Department of Pharmaceutics, School of Pharmacy, University of Washington, HSB Room H272, 1959 NE Pacific Street, Seattle, WA, 98195, USA
| | - Edward J Kelly
- Department of Pharmaceutics, School of Pharmacy, University of Washington, HSB Room H272, 1959 NE Pacific Street, Seattle, WA, 98195, USA. .,Division of Nephrology, Department of Medicine, Kidney Research Institute, University of Washington, 1959 NE Pacific Street, HSB Room H272, Seattle, WA, 98195, USA.
| |
Collapse
|
21
|
Hermann R, Krajcsi P, Fluck M, Seithel-Keuth A, Bytyqi A, Galazka A, Munafo A. Review of Transporter Substrate, Inhibitor, and Inducer Characteristics of Cladribine. Clin Pharmacokinet 2021; 60:1509-1535. [PMID: 34435310 PMCID: PMC8613159 DOI: 10.1007/s40262-021-01065-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/28/2021] [Indexed: 12/23/2022]
Abstract
Cladribine is a nucleoside analog that is phosphorylated in its target cells (B- and T-lymphocytes) to its active adenosine triphosphate form (2-chlorodeoxyadenosine triphosphate). Cladribine tablets 10 mg (Mavenclad®) administered for up to 10 days per year in 2 consecutive years (3.5-mg/kg cumulative dose over 2 years) are used to treat patients with relapsing multiple sclerosis. The ATP-binding cassette, solute carrier, and nucleoside transporter substrate, inhibitor, and inducer characteristics of cladribine are reviewed in this article. Available evidence suggests that the distribution of cladribine across biological membranes is facilitated by a number of uptake and efflux transporters. Among the key ATP-binding cassette efflux transporters, only breast cancer resistance protein has been shown to be an efficient transporter of cladribine, while P-glycoprotein does not transport cladribine well. Intestinal absorption, distribution throughout the body, and intracellular uptake of cladribine appear to be exclusively mediated by equilibrative and concentrative nucleoside transporters, specifically by ENT1, ENT2, ENT4, CNT2 (low affinity), and CNT3. Renal excretion of cladribine appears to be most likely driven by breast cancer resistance protein, ENT1, and P-glycoprotein. The latter may play a role despite its poor cladribine transport efficiency in view of the renal abundance of P-glycoprotein. There is no evidence that solute carrier uptake transporters such as organic anion transporting polypeptides, organic anion transporters, and organic cation transporters are involved in the transport of cladribine. Available in vitro studies examining the inhibitor characteristics of cladribine for a total of 13 major ATP-binding cassette, solute carrier, and CNT transporters indicate that in vivo inhibition of any of these transporters by cladribine is unlikely.
Collapse
Affiliation(s)
- Robert Hermann
- Clinical Research Appliance (cr.appliance), Heinrich-Vingerhut-Weg 3, 63571, Gelnhausen, Germany.
| | | | | | | | | | | | - Alain Munafo
- Institute of Pharmacometrics, an Affiliate of Merck KGaA, Lausanne, Switzerland
| |
Collapse
|
22
|
Kumar D, Baligar P, Srivastav R, Narad P, Raj S, Tandon C, Tandon S. Stem Cell Based Preclinical Drug Development and Toxicity Prediction. Curr Pharm Des 2021; 27:2237-2251. [PMID: 33076801 DOI: 10.2174/1381612826666201019104712] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Accepted: 08/22/2020] [Indexed: 01/09/2023]
Abstract
Stem cell based toxicity prediction plays a very important role in the development of the drug. Unexpected adverse effects of the drugs during clinical trials are a major reason for the termination or withdrawal of drugs. Methods for predicting toxicity employ in vitro as well as in vivo models; however, the major drawback seen in the data derived from these animal models is the lack of extrapolation, owing to interspecies variations. Due to these limitations, researchers have been striving to develop more robust drug screening platforms based on stem cells. The application of stem cells based toxicity testing has opened up robust methods to study the impact of new chemical entities on not only specific cell types, but also organs. Pluripotent stem cells, as well as cells derived from them, can be evaluated for modulation of cell function in response to drugs. Moreover, the combination of state-of-the -art techniques such as tissue engineering and microfluidics to fabricate organ- on-a-chip, has led to assays which are amenable to high throughput screening to understand the adverse and toxic effects of chemicals and drugs. This review summarizes the important aspects of the establishment of the embryonic stem cell test (EST), use of stem cells, pluripotent, induced pluripotent stem cells and organoids for toxicity prediction and drug development.
Collapse
Affiliation(s)
- Dhruv Kumar
- Amity Institute of Molecular Medicine & Stem Cell Research, Amity University, Noida, Uttar Pradesh 201313, India
| | - Prakash Baligar
- Amity Institute of Molecular Medicine & Stem Cell Research, Amity University, Noida, Uttar Pradesh 201313, India
| | - Rajpal Srivastav
- Amity Institute of Biotechnology, Amity University, Noida, Uttar Pradesh 201313, India
| | - Priyanka Narad
- Amity Institute of Biotechnology, Amity University, Noida, Uttar Pradesh 201313, India
| | - Sibi Raj
- Amity Institute of Molecular Medicine & Stem Cell Research, Amity University, Noida, Uttar Pradesh 201313, India
| | - Chanderdeep Tandon
- Amity Institute of Biotechnology, Amity University, Noida, Uttar Pradesh 201313, India
| | - Simran Tandon
- Amity Institute of Molecular Medicine & Stem Cell Research, Amity University, Noida, Uttar Pradesh 201313, India
| |
Collapse
|
23
|
Bondue T, Arcolino FO, Veys KRP, Adebayo OC, Levtchenko E, van den Heuvel LP, Elmonem MA. Urine-Derived Epithelial Cells as Models for Genetic Kidney Diseases. Cells 2021; 10:cells10061413. [PMID: 34204173 PMCID: PMC8230018 DOI: 10.3390/cells10061413] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Revised: 05/28/2021] [Accepted: 06/02/2021] [Indexed: 12/11/2022] Open
Abstract
Epithelial cells exfoliated in human urine can include cells anywhere from the urinary tract and kidneys; however, podocytes and proximal tubular epithelial cells (PTECs) are by far the most relevant cell types for the study of genetic kidney diseases. When maintained in vitro, they have been proven extremely valuable for discovering disease mechanisms and for the development of new therapies. Furthermore, cultured patient cells can individually represent their human sources and their specific variants for personalized medicine studies, which are recently gaining much interest. In this review, we summarize the methodology for establishing human podocyte and PTEC cell lines from urine and highlight their importance as kidney disease cell models. We explore the well-established and recent techniques of cell isolation, quantification, immortalization and characterization, and we describe their current and future applications.
Collapse
Affiliation(s)
- Tjessa Bondue
- Department of Development and Regeneration, KU Leuven, 3000 Leuven, Belgium; (T.B.); (F.O.A.); (K.R.P.V.); (O.C.A.); (E.L.); (L.P.v.d.H.)
| | - Fanny O. Arcolino
- Department of Development and Regeneration, KU Leuven, 3000 Leuven, Belgium; (T.B.); (F.O.A.); (K.R.P.V.); (O.C.A.); (E.L.); (L.P.v.d.H.)
| | - Koenraad R. P. Veys
- Department of Development and Regeneration, KU Leuven, 3000 Leuven, Belgium; (T.B.); (F.O.A.); (K.R.P.V.); (O.C.A.); (E.L.); (L.P.v.d.H.)
- Department of Pediatrics, Division of Pediatric Nephrology, University Hospitals Leuven, 3000 Leuven, Belgium
| | - Oyindamola C. Adebayo
- Department of Development and Regeneration, KU Leuven, 3000 Leuven, Belgium; (T.B.); (F.O.A.); (K.R.P.V.); (O.C.A.); (E.L.); (L.P.v.d.H.)
- Centre for Molecular and Vascular Biology, Department of Cardiovascular Sciences, KU Leuven, 3000 Leuven, Belgium
| | - Elena Levtchenko
- Department of Development and Regeneration, KU Leuven, 3000 Leuven, Belgium; (T.B.); (F.O.A.); (K.R.P.V.); (O.C.A.); (E.L.); (L.P.v.d.H.)
- Department of Pediatrics, Division of Pediatric Nephrology, University Hospitals Leuven, 3000 Leuven, Belgium
| | - Lambertus P. van den Heuvel
- Department of Development and Regeneration, KU Leuven, 3000 Leuven, Belgium; (T.B.); (F.O.A.); (K.R.P.V.); (O.C.A.); (E.L.); (L.P.v.d.H.)
- Department of Pediatric Nephrology, Radboud University Medical Center, 6500 Nijmegen, The Netherlands
| | - Mohamed A. Elmonem
- Department of Clinical and Chemical Pathology, Faculty of Medicine, Cairo University, Cairo 11628, Egypt
- Correspondence:
| |
Collapse
|
24
|
Vriend J, Pye KR, Brown C. In vitro models for accurate prediction of renal tubular xenobiotic transport in vivo. CURRENT OPINION IN TOXICOLOGY 2021. [DOI: 10.1016/j.cotox.2020.12.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
|
25
|
Clerin V, Saito H, Filipski KJ, Nguyen AH, Garren J, Kisucka J, Reyes M, Jüppner H. Selective pharmacological inhibition of the sodium-dependent phosphate cotransporter NPT2a promotes phosphate excretion. J Clin Invest 2020; 130:6510-6522. [PMID: 32853180 PMCID: PMC7685737 DOI: 10.1172/jci135665] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Accepted: 08/20/2020] [Indexed: 12/16/2022] Open
Abstract
The sodium-phosphate cotransporter NPT2a plays a key role in the reabsorption of filtered phosphate in proximal renal tubules, thereby critically contributing to phosphate homeostasis. Inadequate urinary phosphate excretion can lead to severe hyperphosphatemia as in tumoral calcinosis and chronic kidney disease (CKD). Pharmacological inhibition of NPT2a may therefore represent an attractive approach for treating hyperphosphatemic conditions. The NPT2a-selective small-molecule inhibitor PF-06869206 was previously shown to reduce phosphate uptake in human proximal tubular cells in vitro. Here, we investigated the acute and chronic effects of the inhibitor in rodents and report that administration of PF-06869206 was well tolerated and elicited a dose-dependent increase in fractional phosphate excretion. This phosphaturic effect lowered plasma phosphate levels in WT mice and in rats with CKD due to subtotal nephrectomy. PF-06869206 had no effect on Npt2a-null mice, but promoted phosphate excretion and reduced phosphate levels in normophophatemic mice lacking Npt2c and in hyperphosphatemic mice lacking Fgf23 or Galnt3. In CKD rats, once-daily administration of PF-06869206 for 8 weeks induced an unabated acute phosphaturic and hypophosphatemic effect, but had no statistically significant effect on FGF23 or PTH levels. Selective pharmacological inhibition of NPT2a thus holds promise as a therapeutic option for genetic and acquired hyperphosphatemic disorders.
Collapse
Affiliation(s)
- Valerie Clerin
- Pfizer Inc., Worldwide Research, Development and Medical, Cambridge, Massachusetts, USA
| | | | - Kevin J. Filipski
- Pfizer Inc., Worldwide Research, Development and Medical, Cambridge, Massachusetts, USA
| | - An Hai Nguyen
- Pfizer Inc., Worldwide Research, Development and Medical, Cambridge, Massachusetts, USA
| | - Jeonifer Garren
- Pfizer Inc., Worldwide Research, Development and Medical, Cambridge, Massachusetts, USA
| | - Janka Kisucka
- Pfizer Inc., Worldwide Research, Development and Medical, Cambridge, Massachusetts, USA
| | | | - Harald Jüppner
- Endocrine Unit and
- Pediatric Nephrology Unit, Massachusetts General Hospital (MGH) and Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
26
|
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
|
27
|
Nieskens TTG, Persson M, Kelly EJ, Sjögren AK. A Multicompartment Human Kidney Proximal Tubule-on-a-Chip Replicates Cell Polarization-Dependent Cisplatin Toxicity. Drug Metab Dispos 2020; 48:1303-1311. [PMID: 33020068 DOI: 10.1124/dmd.120.000098] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Accepted: 09/23/2020] [Indexed: 02/06/2023] Open
Abstract
Drug-induced kidney injury is a major clinical problem and causes drug attrition in the pharmaceutical industry. To better predict drug-induced kidney injury, kidney in vitro cultures with enhanced physiologic relevance are developed. To mimic the proximal tubule, the main site of adverse drug reactions in the kidney, human-derived renal proximal tubule epithelial cells (HRPTECs) were injected in one of the channels of dual-channel Nortis chips and perfused for 7 days. Tubes of HRPTECs demonstrated expression of tight junction protein 1 (zona occludens-1), lotus lectin, and primary cilia with localization at the apical membrane, indicating an intact proximal tubule brush border. Gene expression of cisplatin efflux transporters multidrug and toxin extrusion transporter (MATE) 1 (SLC47A1) and MATE2-k (SLC47A2) and megalin endocytosis receptor increased 19.9 ± 5.0-, 23.2 ± 8.4-, and 106 ± 33-fold, respectively, in chip cultures compared with 2-dimensional cultures. Moreover, organic cation transporter 2 (OCT2) (SLC22A2) was localized exclusively on the basolateral membrane. When infused from the basolateral compartment, cisplatin (25 µM, 72 hours) induced toxicity, which was evident as reduced cell number and reduced barrier integrity compared with vehicle-treated chip cultures. Coexposure with the OCT2 inhibitor cimetidine (1 mM) abolished cisplatin toxicity. In contrast, infusion of cisplatin from the apical compartment did not induce toxicity, which was in line with polarized localization of cisplatin uptake transport proteins, including OCT2. In conclusion, we developed a dual channel human kidney proximal tubule-on-a-chip with a polarized epithelium, restricting cisplatin sensitivity to the basolateral membrane and suggesting improved physiologic relevance over single-compartment models. Its implementation in drug discovery holds promise to improve future in vitro drug-induced kidney injury studies. SIGNIFICANCE STATEMENT: Human-derived kidney proximal tubule cells retained characteristics of epithelial polarization in vitro when cultured in the kidney-on-a-chip, and the dual-channel construction allowed for drug exposure using the physiologically relevant compartment. Therefore, cell polarization-dependent cisplatin toxicity could be replicated for the first time in a kidney proximal tubule-on-a-chip. The use of this physiologically relevant model in drug discovery has potential to aid identification of safe novel drugs and contribute to reducing attrition rates due to drug-induced kidney injury.
Collapse
Affiliation(s)
- Tom T G Nieskens
- CVRM Safety, Clinical Pharmacology and Safety Sciences, R&D, AstraZeneca, Gothenburg, Sweden (T.T.G.N., M.P., A.-K.S.) and Department of Pharmaceutics and Kidney Research Institute, University of Washington, Seattle, Washington (E.J.K.)
| | - Mikael Persson
- CVRM Safety, Clinical Pharmacology and Safety Sciences, R&D, AstraZeneca, Gothenburg, Sweden (T.T.G.N., M.P., A.-K.S.) and Department of Pharmaceutics and Kidney Research Institute, University of Washington, Seattle, Washington (E.J.K.)
| | - Edward J Kelly
- CVRM Safety, Clinical Pharmacology and Safety Sciences, R&D, AstraZeneca, Gothenburg, Sweden (T.T.G.N., M.P., A.-K.S.) and Department of Pharmaceutics and Kidney Research Institute, University of Washington, Seattle, Washington (E.J.K.)
| | - Anna-Karin Sjögren
- CVRM Safety, Clinical Pharmacology and Safety Sciences, R&D, AstraZeneca, Gothenburg, Sweden (T.T.G.N., M.P., A.-K.S.) and Department of Pharmaceutics and Kidney Research Institute, University of Washington, Seattle, Washington (E.J.K.)
| |
Collapse
|
28
|
Bajaj P, Chung G, Pye K, Yukawa T, Imanishi A, Takai Y, Brown C, Wagoner MP. Freshly isolated primary human proximal tubule cells as an in vitro model for the detection of renal tubular toxicity. Toxicology 2020; 442:152535. [PMID: 32622972 DOI: 10.1016/j.tox.2020.152535] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Revised: 06/18/2020] [Accepted: 07/01/2020] [Indexed: 02/08/2023]
Abstract
Drug induced kidney injury (DIKI) is a common reason for compound attrition in drug development pipelines with proximal tubule epithelial cells (PTECs) most commonly associated with DIKI. Here, we investigated freshly isolated human (hPTECs) as an in vitro model for assessing renal tubular toxicity. The freshly isolated hPTECs were first characterized to confirm gene expression of important renal transporters involved in drug handling which was further corroborated by confirming the functional activity of organic cation transporter 2 and organic anion transporter 1 by using transporter specific inhibitors. Additionally, functionality of megalin/cubilin endocytic receptors was also confirmed. A training set of 36 compounds was used to test the ability of the model to classify them using six different endpoints which included three biomarkers (Kidney Injury Molecule-1, Neutrophil gelatinase-associated lipocalin, and Clusterin) and three non-specific injury endpoints (ATP depletion, LDH leakage, and barrier permeability via transepithelial electrical resistance) in a dose-dependent manner across two independent kidney donors. In general, biomarkers showed higher predictivity than non-specific endpoints, with Clusterin showing the highest predictivity (Sensitivity/Specificity - 65.0/93.8 %). By using the thresholds generated from the training set, nine candidate internal Takeda compounds were screened where PTEC toxicity was identified as one of the findings in preclinical animal studies. The model correctly classified four of six true positives and two of three true negatives, showing validation of the in vitro model for detection of tubular toxicants. This work thus shows the potential application of freshly isolated primary hPTECs using translational biomarkers in assessment of tubular toxicity within the drug discovery pipeline.
Collapse
Affiliation(s)
- Piyush Bajaj
- Drug Safety Research and Evaluation, Takeda Pharmaceutical International Co., Cambridge, MA USA
| | | | | | - Tomoya Yukawa
- Drug Safety Research and Evaluation, Takeda Pharmaceutical International Co., Cambridge, MA USA
| | - Akio Imanishi
- Drug Safety Research and Evaluation, Takeda Pharmaceutical International Co., Kanagawa, Japan
| | - Yuichi Takai
- Drug Safety Research and Evaluation, Takeda Pharmaceutical International Co., Kanagawa, Japan
| | | | - Matthew P Wagoner
- Drug Safety Research and Evaluation, Takeda Pharmaceutical International Co., Cambridge, MA USA.
| |
Collapse
|
29
|
Chapron A, Chapron BD, Hailey DW, Chang SY, Imaoka T, Thummel KE, Kelly E, Himmelfarb J, Shen D, Yeung CK. An Improved Vascularized, Dual-Channel Microphysiological System Facilitates Modeling of Proximal Tubular Solute Secretion. ACS Pharmacol Transl Sci 2020; 3:496-508. [PMID: 32566915 PMCID: PMC7296546 DOI: 10.1021/acsptsci.9b00078] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Indexed: 11/30/2022]
Abstract
A vascularized human proximal tubule model in a dual-channel microphysiological system (VPT-MPS) was developed, representing an advance over previous, single-cell-type kidney microphysiological systems. Human proximal tubule epithelial cells (PTECs) and human umbilical vein endothelial cells (HUVECs) were cocultured in side-by-side channels. Over 24 h of coculturing, PTECs maintained polarized expression of Na+/K+ ATPase, tight junctions (ZO-1), and OAT1. HUVECs showed the absence of ZO-1 but expressed endothelial cell marker (CD-31). In time-lapse imaging studies, fluorescein isothiocyanate (FITC)-dextran passed freely from the HUVEC vessel into the supporting extracellular matrix, confirming the leakiness of the endothelium (at 80 min, matrix/intravessel fluorescence ratio = 0.2). Dextran-associated fluorescence accumulated in the matrix adjacent to the basolateral aspect of the PTEC tubule with minimal passage of the compound into the tubule lumen observed (at 80 min, tubule lumen/matrix fluorescence ratio = 0.01). This demonstrates that the proximal tubule compartment is the rate-limiting step in the secretion of compounds in VPT-MPS. In kinetic studies with radiolabeled markers, p-aminohippuric acid (PAH) exhibited greater output into the tubule lumen than did paracellular markers mannitol and FITC-dextran (tubule outflow/vessel outflow concentration ratio of 7.7% vs 0.5 and 0.4%, respectively). A trend toward reduced PAH secretion by 45% was observed upon coadministration of probenecid. This signifies functional expression of renal transporters in PTECs that normally mediate the renal secretion of PAH. The VPT-MPS holds the promise of providing an in vitro platform for evaluating the renal secretion of new drug candidates and investigating the dysregulation of tubular drug secretion in chronic kidney disease.
Collapse
Affiliation(s)
- Alenka Chapron
- Department
of Pharmaceutics, School of Pharmacy, University
of Washington, Seattle, Washington 98195, United States
| | - Brian D. Chapron
- Department
of Pharmaceutics, School of Pharmacy, University
of Washington, Seattle, Washington 98195, United States
| | - Dale W. Hailey
- Lynn
and Mike Garvey Imaging Core, Institute
for Stem Cell and Regenerative Medicine, Seattle, Washington 98109, United States
- Department
of Pathology, School of Medicine, University
of Washington, Seattle, Washington 98195, United States
| | - Shih-Yu Chang
- Department
of Pharmacy, School of Pharmacy, University
of Washington, Seattle Washington 98195, United States
| | - Tomoki Imaoka
- Department
of Pharmaceutics, School of Pharmacy, University
of Washington, Seattle, Washington 98195, United States
| | - Kenneth E. Thummel
- Department
of Pharmaceutics, School of Pharmacy, University
of Washington, Seattle, Washington 98195, United States
| | - Edward Kelly
- Department
of Pharmaceutics, School of Pharmacy, University
of Washington, Seattle, Washington 98195, United States
| | - Jonathan Himmelfarb
- Kidney
Research Institute, Division of Nephrology, Department of Medicine, University of Washington, Seattle, Washington 98195, United States
| | - Danny Shen
- Department
of Pharmaceutics, School of Pharmacy, University
of Washington, Seattle, Washington 98195, United States
| | - Catherine K. Yeung
- Department
of Pharmacy, School of Pharmacy, University
of Washington, Seattle Washington 98195, United States
- Kidney
Research Institute, Division of Nephrology, Department of Medicine, University of Washington, Seattle, Washington 98195, United States
| |
Collapse
|
30
|
Evaluation of Renal Anionic Secretion Following Living-donor and Deceased-donor Renal Transplantation: A Clinical Pharmacokinetic Study of Cefoxitin Microdosing. Transplant Direct 2020; 6:e561. [PMID: 33062845 PMCID: PMC7531749 DOI: 10.1097/txd.0000000000001001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Accepted: 03/19/2020] [Indexed: 11/25/2022] Open
Abstract
Renal transplantation is the treatment of choice for patients with end-stage renal disease. Because kidneys are the primary excretory organs for various drugs/drug metabolites, changes in renal graft function would significantly alter the clearance and exposure of renally secreted drugs. Renal allografts from living and deceased donors normally undergo numerous insults, including injuries associated with prolonged cold ischemic time, reperfusion, and nephrotoxicity due to calcineurin inhibitors. These physiologic and pharmacologic stresses can alter the expression and functional capacity of renal organic anionic transporters (OATs). Methods The objectives of this study were to assess the longitudinal changes in renal anionic secretion in kidney transplant patients, to study the effect of prolonged cold ischemic time on OAT secretion in kidney transplant patients (living- versus deceased-donor recipients), and to compare OAT secretory capacity of renal transplant recipients with healthy volunteers. Cefoxitin was used as a probe drug to assess OAT secretion. Cefoxitin pharmacokinetics was studied in 15 de novo renal transplant recipients following intravenous administration of 200 mg cefoxitin within 14 d and beyond 90 d posttransplantation. Results No longitudinal changes in real OAT secretion in early posttransplant period were observed, and there were no differences in renal OAT secretion between living- and deceased-donor renal transplant recipients. Overall, cefoxitin exposure was 2.6-fold higher and half-life increased by 2.2-fold in renal transplant recipients when compared with historical healthy controls. Conclusions These results suggest that OAT system is functioning well, but renal transplant recipients would need significantly lower dosage of drugs that are primarily secreted via the OAT system compared with normal subjects.
Collapse
|
31
|
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
|
32
|
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
|
33
|
Sánchez-Romero N, Martínez-Gimeno L, Caetano-Pinto P, Saez B, Sánchez-Zalabardo JM, Masereeuw R, Giménez I. A simple method for the isolation and detailed characterization of primary human proximal tubule cells for renal replacement therapy. Int J Artif Organs 2019; 43:45-57. [DOI: 10.1177/0391398819866458] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The main physiological functions of renal proximal tubule cells in vivo are reabsorption of essential nutrients from the glomerular filtrate and secretion of waste products and xenobiotics into urine. Currently, there are several established cell lines of human origin available as in vitro models of proximal tubule. However, these cells appeared to be limited in their biological relevance, because essential characteristics of the original tissue are lost once the cells are cultured. As a consequence of these limitations, primary human proximal tubule cells constitute a suitable and a biologically more relevant in vitro model to study this specific segment of the nephron and therefore, these cells can play an important role in renal regenerative medicine applications. Here, we describe a protocol to isolate proximal tubule cells from human nephrectomies. We explain the steps performed for an in-depth characterization of the cells, including the study of markers from others segments of the nephron, with the goal to determine the purity of the culture and the stability of proteins, enzymes, and transporters along time. The human proximal tubule cells isolated and used throughout this study showed many proximal tubule characteristics, including monolayer organization, cell polarization with the expression of tight junctions and primary cilia, expression of proximal tubule–specific proteins, such as megalin and sodium/glucose cotransporter 2, among others. The cells also expressed enzymatic activity for dipeptidyl peptidase IV, as well as for gamma glutamyl transferase 1, and expressed transporter activity for organic anion transporter 1, P-glycoprotein, multidrug resistance proteins, and breast cancer resistance protein. In conclusion, characterization of our cells confirmed presence of putative proximal tubule markers and the functional expression of multiple endogenous organic ion transporters mimicking renal reabsorption and excretion. These findings can constitute a valuable tool in the development of bioartificial kidney devices.
Collapse
Affiliation(s)
- Natalia Sánchez-Romero
- Instituto Aragonés de Ciencias de la Salud (IACS), Zaragoza, Spain
- Instituto de Investigación Sanitaria Aragón (IIS Aragón), Zaragoza, Spain
| | - Laura Martínez-Gimeno
- Instituto Aragonés de Ciencias de la Salud (IACS), Zaragoza, Spain
- Instituto de Investigación Sanitaria Aragón (IIS Aragón), Zaragoza, Spain
| | - Pedro Caetano-Pinto
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, Utrecht, The Netherlands
- Mechanistic Safety and ADME Sciences, Drug Safety and Metabolism, IMED Biotech Unit, AstraZeneca, Cambridge, UK
| | - Berta Saez
- Hospital Clínico Universitario Lozano Blesa, Zaragoza, Spain
- Universidad San Jorge, Zaragoza, Spain
| | | | - Rosalinde Masereeuw
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, Utrecht, The Netherlands
| | - Ignacio Giménez
- Instituto Aragonés de Ciencias de la Salud (IACS), Zaragoza, Spain
- Instituto de Investigación Sanitaria Aragón (IIS Aragón), Zaragoza, Spain
- Department of Pharmacology and Physiology, University of Zaragoza, Zaragoza, Spain
| |
Collapse
|
34
|
Rat Kidney Slices for Evaluation of Apical Membrane Transporters in Proximal Tubular Cells. J Pharm Sci 2019; 108:2798-2804. [DOI: 10.1016/j.xphs.2019.03.031] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2018] [Revised: 03/27/2019] [Accepted: 03/28/2019] [Indexed: 11/22/2022]
|
35
|
Diekjürgen D, Grainger DW. A murine ex vivo 3D kidney proximal tubule model predicts clinical drug-induced nephrotoxicity. Arch Toxicol 2019; 93:1349-1364. [PMID: 30863989 DOI: 10.1007/s00204-019-02430-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2018] [Accepted: 03/05/2019] [Indexed: 12/18/2022]
Abstract
Drug attrition and clinical product withdrawals due to nephrotoxicity remain major challenges for pharmaceutical drug development pipelines. Currently, no reliable high-throughput in vitro screening models are available that provide reliable, predictive toxicology data for clinical nephrotoxicity. Drug screens to predict toxicity and pharmacology assessments are compromised by standard two-dimensional (2D) cell monoculture models. Here we extend a previously reported murine three-dimensional (3D) kidney-derived intact proximal tubule model to provide ex vivo drug toxicity data that reliably compare to clinical experiences and improve nephrotoxicity predictions over current 2D cell assays. Proximal tubule cytotoxicity was monitored by ATP depletion for 12 compounds (acarbose, acetylsalicylic acid, captopril, cimetidine, cidofovir, cisplatin, doxorubicin, gentamicin, polymyxin B, polymyxin B nonapeptide, probenecid and vancomycin) in 3D proximal tubule ex vivo assays. Drug concentration-response curves (1-1000 µM) and IC50, lowest effective concentration (LEC) and AUC values were compared to clinical therapeutic exposure levels (Cmax). The 100-fold Cmax threshold demonstrated best sensitivity (96.9%) and specificity (87.5%) for this assay, with high positive (93.9%) and negative (93.3%) predictive values for nephrotoxicity. IC50 values were superior to LEC. Results also support the model's capability to predict substrate-inhibitor/competitor interactions, yielding toxicity results similar to those reported in vivo. These 3D proximal tubule-based drug screens provide more reliable nephrotoxicity predictions, and more insight into complex mechanisms implicated in nephrotoxicity than current standard 2D cell assays. This new approach for rapid drug toxicity testing produces more reliable clinical comparisons than current 2D cell culture screening techniques.
Collapse
Affiliation(s)
- Dorina Diekjürgen
- Department of Pharmaceutics and Pharmaceutical Chemistry, University of Utah, Salt Lake City, UT, 84112-5820, USA
| | - David W Grainger
- Department of Pharmaceutics and Pharmaceutical Chemistry, University of Utah, Salt Lake City, UT, 84112-5820, USA. .,Department of Biomedical Engineering, University of Utah, Salt Lake City, UT, 84112-5820, USA.
| |
Collapse
|
36
|
Shen H, Scialis RJ, Lehman-McKeeman L. Xenobiotic Transporters in the Kidney: Function and Role in Toxicity. Semin Nephrol 2019; 39:159-175. [DOI: 10.1016/j.semnephrol.2018.12.010] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
|
37
|
Ferreras L, Moles A, Situmorang GR, El Masri R, Wilson IL, Cooke K, Thompson E, Kusche-Gullberg M, Vivès RR, Sheerin NS, Ali S. Heparan sulfate in chronic kidney diseases: Exploring the role of 3-O-sulfation. Biochim Biophys Acta Gen Subj 2019; 1863:839-848. [PMID: 30794825 DOI: 10.1016/j.bbagen.2019.02.009] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2018] [Revised: 02/07/2019] [Accepted: 02/16/2019] [Indexed: 01/03/2023]
Abstract
One of the main feature of chronic kidney disease is the development of renal fibrosis. Heparan Sulfate (HS) is involved in disease development by modifying the function of growth factors and cytokines and creating chemokine gradients. In this context, we aimed to understand the function of HS sulfation in renal fibrosis. Using a mouse model of renal fibrosis, we found that total HS 2-O-sulfation was increased in damaged kidneys, whilst, tubular staining of HS 3-O-sulfation was decreased. The expression of HS modifying enzymes significantly correlated with the development of fibrosis with HS3ST1 demonstrating the strongest correlation. The pro-fibrotic factors TGFβ1 and TGFβ2/IL1β significantly downregulated HS3ST1 expression in both renal epithelial cells and renal fibroblasts. To determine the implication of HS3ST1 in growth factor binding and signalling, we generated an in vitro model of renal epithelial cells overexpressing HS3ST1 (HKC8-HS3ST1). Heparin Binding EGF like growth factor (HB-EGF) induced rapid, transient STAT3 phosphorylation in control HKC8 cells. In contrast, a prolonged response was demonstrated in HKC8-HS3ST1 cells. Finally, we showed that both HS 3-O-sulfation and HB-EGF tubular staining were decreased with the development of fibrosis. Taken together, these data suggest that HS 3-O-sulfation is modified in fibrosis and highlight HS3ST1 as an attractive biomarker of fibrosis progression with a potential role in HB-EGF signalling.
Collapse
Affiliation(s)
- Laura Ferreras
- Institute of Cellular Medicine, Faculty of Medical Sciences, Newcastle University, NE2 4HH, UK
| | - Anna Moles
- Institute of Cellular Medicine, Faculty of Medical Sciences, Newcastle University, NE2 4HH, UK
| | - Gerhard R Situmorang
- Institute of Cellular Medicine, Faculty of Medical Sciences, Newcastle University, NE2 4HH, UK
| | - Rana El Masri
- Univ. Grenoble Alpes, CNRS, CEA, IBS, Grenoble, France
| | - Imogen L Wilson
- Institute of Cellular Medicine, Faculty of Medical Sciences, Newcastle University, NE2 4HH, UK
| | - Katie Cooke
- Institute of Cellular Medicine, Faculty of Medical Sciences, Newcastle University, NE2 4HH, UK
| | - Emily Thompson
- Institute of Cellular Medicine, Faculty of Medical Sciences, Newcastle University, NE2 4HH, UK
| | - Marion Kusche-Gullberg
- University of Bergen, Department of Biomedicine, Jonas Lies vei 91, N-5009 Bergen, Norway
| | | | - Neil S Sheerin
- Institute of Cellular Medicine, Faculty of Medical Sciences, Newcastle University, NE2 4HH, UK; Newcastle upon Tyne Hospitals, NHS Foundation Trust, NIHR Newcastle Biomedical Research Centre, United Kingdom
| | - Simi Ali
- Institute of Cellular Medicine, Faculty of Medical Sciences, Newcastle University, NE2 4HH, UK.
| |
Collapse
|
38
|
Establishment of renal proximal tubule cell lines derived from the kidney of p53 knockout mice. Cytotechnology 2019; 71:45-56. [PMID: 30603921 DOI: 10.1007/s10616-018-0261-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2018] [Accepted: 09/19/2018] [Indexed: 12/14/2022] Open
Abstract
The human cell line HK-2 is most commonly used as a model of renal proximal tubular epithelial cells (PTECs) for various studies despite the absence or low expression of transporters characteristic of parental PTECs. In an effort to develop reliable PTEC models, several human cell lines have been newly established over the last decade. In contrast, reliable mouse PTEC models are still unavailable. In this study, we established immortalized renal cortex tubule cell lines derived from p53 knockout mice and evaluated various PTEC characteristics toward the development of reliable mouse PTEC models. Here, we focus on MuRTE61, one of 13 newly established clonal cell lines. Albumin uptake in MuRTE61 cells was verified by incubation with fluorescent dye-labeled albumin. RT-PCR confirmed the expression of efflux transporter genes characteristic of PTECs in the MuRTE61 cells. MuRTE61 cells exhibited high sensitivity to treatment with cisplatin, a nephrotoxic agent, accompanied by upregulated expression of the uptake transporter Slc22a2 gene. Furthermore, MuRTE61 cells consistently formed spheroids with a lumen and apicobasal polarity in three-dimensional Matrigel cultures. Apical brush border microvilli were also observed in the spheroids by transmission electron microscopy. These data validate that MuRTE61 can be characterized as a reliable mouse PTEC line. In future, detailed analysis of reliable mouse and human PTEC lines will provide an accurate extrapolation of results of experiments using mice and humans, and may help resolve apparent inconsistencies between mouse and human nephrotoxicity.
Collapse
|
39
|
Legallais C, Kim D, Mihaila SM, Mihajlovic M, Figliuzzi M, Bonandrini B, Salerno S, Yousef Yengej FA, Rookmaaker MB, Sanchez Romero N, Sainz-Arnal P, Pereira U, Pasqua M, Gerritsen KGF, Verhaar MC, Remuzzi A, Baptista PM, De Bartolo L, Masereeuw R, Stamatialis D. Bioengineering Organs for Blood Detoxification. Adv Healthc Mater 2018; 7:e1800430. [PMID: 30230709 DOI: 10.1002/adhm.201800430] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2018] [Revised: 08/23/2018] [Indexed: 12/11/2022]
Abstract
For patients with severe kidney or liver failure the best solution is currently organ transplantation. However, not all patients are eligible for transplantation and due to limited organ availability, most patients are currently treated with therapies using artificial kidney and artificial liver devices. These therapies, despite their relative success in preserving the patients' life, have important limitations since they can only replace part of the natural kidney or liver functions. As blood detoxification (and other functions) in these highly perfused organs is achieved by specialized cells, it seems relevant to review the approaches leading to bioengineered organs fulfilling most of the native organ functions. There, the culture of cells of specific phenotypes on adapted scaffolds that can be perfused takes place. In this review paper, first the functions of kidney and liver organs are briefly described. Then artificial kidney/liver devices, bioartificial kidney devices, and bioartificial liver devices are focused on, as well as biohybrid constructs obtained by decellularization and recellularization of animal organs. For all organs, a thorough overview of the literature is given and the perspectives for their application in the clinic are discussed.
Collapse
Affiliation(s)
- Cécile Legallais
- UMR CNRS 7338 Biomechanics & Bioengineering; Université de technologie de Compiègne; Sorbonne Universités; 60203 Compiègne France
| | - Dooli Kim
- (Bio)artificial organs; Department of Biomaterials Science and Technology; Faculty of Science and Technology; TechMed Institute; University of Twente; P.O. Box 217 7500 AE Enschede The Netherlands
| | - Sylvia M. Mihaila
- Division of Pharmacology; Utrecht Institute for Pharmaceutical Sciences; Utrecht University; Universiteitsweg 99 3584 CG Utrecht The Netherlands
- Department of Nephrology and Hypertension; University Medical Center Utrecht and Regenerative Medicine Utrecht; Utrecht University; Heidelberglaan 100 3584 CX Utrecht The Netherlands
| | - Milos Mihajlovic
- Division of Pharmacology; Utrecht Institute for Pharmaceutical Sciences; Utrecht University; Universiteitsweg 99 3584 CG Utrecht The Netherlands
| | - Marina Figliuzzi
- IRCCS-Istituto di Ricerche Farmacologiche Mario Negri; via Stezzano 87 24126 Bergamo Italy
| | - Barbara Bonandrini
- Department of Chemistry; Materials and Chemical Engineering “Giulio Natta”; Politecnico di Milano; Piazza Leonardo da Vinci 32 20133 Milan Italy
| | - Simona Salerno
- Institute on Membrane Technology; National Research Council of Italy; ITM-CNR; Via Pietro BUCCI, Cubo 17C - 87036 Rende Italy
| | - Fjodor A. Yousef Yengej
- Department of Nephrology and Hypertension; University Medical Center Utrecht and Regenerative Medicine Utrecht; Utrecht University; Heidelberglaan 100 3584 CX Utrecht The Netherlands
| | - Maarten B. Rookmaaker
- Department of Nephrology and Hypertension; University Medical Center Utrecht and Regenerative Medicine Utrecht; Utrecht University; Heidelberglaan 100 3584 CX Utrecht The Netherlands
| | | | - Pilar Sainz-Arnal
- Instituto de Investigación Sanitaria de Aragón (IIS Aragon); 50009 Zaragoza Spain
- Instituto Aragonés de Ciencias de la Salud (IACS); 50009 Zaragoza Spain
| | - Ulysse Pereira
- UMR CNRS 7338 Biomechanics & Bioengineering; Université de technologie de Compiègne; Sorbonne Universités; 60203 Compiègne France
| | - Mattia Pasqua
- UMR CNRS 7338 Biomechanics & Bioengineering; Université de technologie de Compiègne; Sorbonne Universités; 60203 Compiègne France
| | - Karin G. F. Gerritsen
- Department of Nephrology and Hypertension; University Medical Center Utrecht and Regenerative Medicine Utrecht; Utrecht University; Heidelberglaan 100 3584 CX Utrecht The Netherlands
| | - Marianne C. Verhaar
- Department of Nephrology and Hypertension; University Medical Center Utrecht and Regenerative Medicine Utrecht; Utrecht University; Heidelberglaan 100 3584 CX Utrecht The Netherlands
| | - Andrea Remuzzi
- IRCCS-Istituto di Ricerche Farmacologiche Mario Negri; via Stezzano 87 24126 Bergamo Italy
- Department of Management; Information and Production Engineering; University of Bergamo; viale Marconi 5 24044 Dalmine Italy
| | - Pedro M. Baptista
- Instituto de Investigación Sanitaria de Aragón (IIS Aragon); 50009 Zaragoza Spain
- Department of Management; Information and Production Engineering; University of Bergamo; viale Marconi 5 24044 Dalmine Italy
- Centro de Investigación Biomédica en Red en el Área Temática de Enfermedades Hepáticas (CIBERehd); 28029 Barcelona Spain
- Fundación ARAID; 50009 Zaragoza Spain
- Instituto de Investigación Sanitaria de la Fundación Jiménez Díaz; 28040 Madrid Spain. Department of Biomedical and Aerospace Engineering; Universidad Carlos III de Madrid; 28911 Madrid Spain
| | - Loredana De Bartolo
- Institute on Membrane Technology; National Research Council of Italy; ITM-CNR; Via Pietro BUCCI, Cubo 17C - 87036 Rende Italy
| | - Rosalinde Masereeuw
- Division of Pharmacology; Utrecht Institute for Pharmaceutical Sciences; Utrecht University; Universiteitsweg 99 3584 CG Utrecht The Netherlands
| | - Dimitrios Stamatialis
- (Bio)artificial organs; Department of Biomaterials Science and Technology; Faculty of Science and Technology; TechMed Institute; University of Twente; P.O. Box 217 7500 AE Enschede The Netherlands
| |
Collapse
|
40
|
A novel multi-parametric high content screening assay in ciPTEC-OAT1 to predict drug-induced nephrotoxicity during drug discovery. Arch Toxicol 2018; 92:3175-3190. [PMID: 30155723 DOI: 10.1007/s00204-018-2284-y] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2018] [Accepted: 08/06/2018] [Indexed: 12/11/2022]
Abstract
Drug-induced nephrotoxicity is a major concern in the clinic and hampers the use of available treatments as well as the development of innovative medicines. It is typically discovered late during drug development, which reflects a lack of in vitro nephrotoxicity assays available that can be employed readily in early drug discovery, to identify and hence steer away from the risk. Here, we report the development of a high content screening assay in ciPTEC-OAT1, a proximal tubular cell line that expresses several relevant renal transporters, using five fluorescent dyes to quantify cell health parameters. We used a validation set of 62 drugs, tested across a relevant concentration range compared to their exposure in humans, to develop a model that integrates multi-parametric data and drug exposure information, which identified most proximal tubular toxic drugs tested (sensitivity 75%) without any false positives (specificity 100%). Due to the relatively high throughput (straight-forward assay protocol, 96-well format, cost-effective) the assay is compatible with the needs in the early drug discovery setting to enable identification, quantification and subsequent mitigation of the risk for nephrotoxicity.
Collapse
|
41
|
Caetano-Pinto P, Stahl SH. Perspective on the Application of Microphysiological Systems to Drug Transporter Studies. Drug Metab Dispos 2018; 46:1647-1657. [DOI: 10.1124/dmd.118.082750] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2018] [Accepted: 08/20/2018] [Indexed: 12/11/2022] Open
|
42
|
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
|
43
|
Vriend J, Nieskens TTG, Vormann MK, van den Berge BT, van den Heuvel A, Russel FGM, Suter-Dick L, Lanz HL, Vulto P, Masereeuw R, Wilmer MJ. Screening of Drug-Transporter Interactions in a 3D Microfluidic Renal Proximal Tubule on a Chip. AAPS JOURNAL 2018; 20:87. [PMID: 30051196 DOI: 10.1208/s12248-018-0247-0] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/03/2018] [Accepted: 07/13/2018] [Indexed: 01/08/2023]
Abstract
Drug-transporter interactions could impact renal drug clearance and should ideally be detected in early stages of drug development to avoid toxicity-related withdrawals in later stages. This requires reliable and robust assays for which current high-throughput screenings have, however, poor predictability. Kidney-on-a-chip platforms have the potential to improve predictability, but often lack compatibility with high-content detection platforms. Here, we combined conditionally immortalized proximal tubule epithelial cells overexpressing organic anion transporter 1 (ciPTEC-OAT1) with the microfluidic titer plate OrganoPlate to develop a screenings assay for renal drug-transporter interactions. In this platform, apical localization of F-actin and intracellular tight-junction protein zonula occludens-1 (ZO-1) indicated appropriate cell polarization. Gene expression levels of the drug transporters organic anion transporter 1 (OAT1; SLC22A6), organic cation transporter 2 (OCT2; SLC22A2), P-glycoprotein (P-gp; ABCB1), and multidrug resistance-associated protein 2 and 4 (MRP2/4; ABCC2/4) were similar levels to 2D static cultures. Functionality of the efflux transporters P-gp and MRP2/4 was studied as proof-of-concept for 3D assays using calcein-AM and 5-chloromethylfluorescein-diacetate (CMFDA), respectively. Confocal imaging demonstrated a 4.4 ± 0.2-fold increase in calcein accumulation upon P-gp inhibition using PSC833. For MRP2/4, a 3.0 ± 0.2-fold increased accumulation of glutathione-methylfluorescein (GS-MF) was observed upon inhibition with a combination of PSC833, MK571, and KO143. Semi-quantitative image processing methods for P-gp and MRP2/4 was demonstrated with corresponding Z'-factors of 0.1 ± 0.3 and 0.4 ± 0.1, respectively. In conclusion, we demonstrate a 3D microfluidic PTEC model valuable for screening of drug-transporter interactions that further allows multiplexing of endpoint read-outs for drug-transporter interactions and toxicity.
Collapse
Affiliation(s)
- Jelle Vriend
- Department of Pharmacology and Toxicology (149), Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, P.O. Box 9101, 6500 HB, Nijmegen, The Netherlands
| | - Tom T G Nieskens
- Department of Pharmacology and Toxicology (149), Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, P.O. Box 9101, 6500 HB, Nijmegen, The Netherlands
| | | | - Bartholomeus T van den Berge
- Department of Pharmacology and Toxicology (149), Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, P.O. Box 9101, 6500 HB, Nijmegen, The Netherlands
| | | | - Frans G M Russel
- Department of Pharmacology and Toxicology (149), Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, P.O. Box 9101, 6500 HB, Nijmegen, The Netherlands
| | - Laura Suter-Dick
- School of Life Sciences, University of Applied Sciences Northwestern Switzerland, Muttenz, Switzerland
| | | | | | - Rosalinde Masereeuw
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht, The Netherlands
| | - Martijn J Wilmer
- Department of Pharmacology and Toxicology (149), Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, P.O. Box 9101, 6500 HB, Nijmegen, The Netherlands.
| |
Collapse
|
44
|
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
|
45
|
Fedecostante M, Westphal KGC, Buono MF, Sanchez Romero N, Wilmer MJ, Kerkering J, Baptista PM, Hoenderop JG, Masereeuw R. Recellularized Native Kidney Scaffolds as a Novel Tool in Nephrotoxicity Screening. Drug Metab Dispos 2018; 46:1338-1350. [PMID: 29980578 DOI: 10.1124/dmd.118.080721] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2018] [Accepted: 06/28/2018] [Indexed: 12/15/2022] Open
Abstract
Drug-induced kidney injury in medicinal compound development accounts for over 20% of clinical trial failures and involves damage to different nephron segments, mostly the proximal tubule. Yet, currently applied cell models fail to reliably predict nephrotoxicity; neither are such models easy to establish. Here, we developed a novel three-dimensional (3D) nephrotoxicity platform on the basis of decellularized rat kidney scaffolds (DS) recellularized with conditionally immortalized human renal proximal tubule epithelial cells overexpressing the organic anion transporter 1 (ciPTEC-OAT1). A 5-day SDS-based decellularization protocol was used to generate DS, of which 100-μm slices were cut and used for cell seeding. After 8 days of culturing, recellularized scaffolds (RS) demonstrated 3D-tubule formation along with tubular epithelial characteristics, including drug transporter function. Exposure of RS to cisplatin (CDDP), tenofovir (TFV), or cyclosporin A (CsA) as prototypical nephrotoxic drugs revealed concentration-dependent reduction in cell viability, as assessed by PrestoBlue and Live/Dead staining assays. This was most probably attributable to specific uptake of CDDP by the organic cation transporter 2 (OCT2), TFV through organic anion transporter 1 (OAT1), and CsA competing for P-glycoprotein-mediated efflux. Compared with 2D cultures, RS showed an increased sensitivity to cisplatin and tenofovir toxicity after 24-hour exposure (9 and 2.2 fold, respectively). In conclusion, we developed a physiologically relevant 3D nephrotoxicity screening platform that could be a novel tool in drug development.
Collapse
Affiliation(s)
- Michele Fedecostante
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht, The Netherlands (M.F., K.G.C.W., M.F.B., N.S.R., R.M.); Aragon's Health Science Institutes (IACS), Zaragoza, Spain (N.S.M.); Departments of Pharmacology and Toxicology (M.J.W., J.K.) and Physiology (J.G.H.), Radboud Institute for Molecular Life Sciences, Radboud university medical center, Nijmegen, The Netherlands; Aragon Health Research Institute (IIS Aragon), Zaragoza, Spain (P.M.B.); Liver and Digestive Diseases Networking Biomedical Research Centre (CIBERehd), Madrid, Spain (P.M.B.); Jiménez Díaz Foundation Health Research Institute, Madrid, Spain (P.M.B.); and Department of Biomedical and Aerospace Engineering, Carlos III University of Madrid, Spain (P.M.B.)
| | - Koen G C Westphal
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht, The Netherlands (M.F., K.G.C.W., M.F.B., N.S.R., R.M.); Aragon's Health Science Institutes (IACS), Zaragoza, Spain (N.S.M.); Departments of Pharmacology and Toxicology (M.J.W., J.K.) and Physiology (J.G.H.), Radboud Institute for Molecular Life Sciences, Radboud university medical center, Nijmegen, The Netherlands; Aragon Health Research Institute (IIS Aragon), Zaragoza, Spain (P.M.B.); Liver and Digestive Diseases Networking Biomedical Research Centre (CIBERehd), Madrid, Spain (P.M.B.); Jiménez Díaz Foundation Health Research Institute, Madrid, Spain (P.M.B.); and Department of Biomedical and Aerospace Engineering, Carlos III University of Madrid, Spain (P.M.B.)
| | - Michele F Buono
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht, The Netherlands (M.F., K.G.C.W., M.F.B., N.S.R., R.M.); Aragon's Health Science Institutes (IACS), Zaragoza, Spain (N.S.M.); Departments of Pharmacology and Toxicology (M.J.W., J.K.) and Physiology (J.G.H.), Radboud Institute for Molecular Life Sciences, Radboud university medical center, Nijmegen, The Netherlands; Aragon Health Research Institute (IIS Aragon), Zaragoza, Spain (P.M.B.); Liver and Digestive Diseases Networking Biomedical Research Centre (CIBERehd), Madrid, Spain (P.M.B.); Jiménez Díaz Foundation Health Research Institute, Madrid, Spain (P.M.B.); and Department of Biomedical and Aerospace Engineering, Carlos III University of Madrid, Spain (P.M.B.)
| | - Natalia Sanchez Romero
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht, The Netherlands (M.F., K.G.C.W., M.F.B., N.S.R., R.M.); Aragon's Health Science Institutes (IACS), Zaragoza, Spain (N.S.M.); Departments of Pharmacology and Toxicology (M.J.W., J.K.) and Physiology (J.G.H.), Radboud Institute for Molecular Life Sciences, Radboud university medical center, Nijmegen, The Netherlands; Aragon Health Research Institute (IIS Aragon), Zaragoza, Spain (P.M.B.); Liver and Digestive Diseases Networking Biomedical Research Centre (CIBERehd), Madrid, Spain (P.M.B.); Jiménez Díaz Foundation Health Research Institute, Madrid, Spain (P.M.B.); and Department of Biomedical and Aerospace Engineering, Carlos III University of Madrid, Spain (P.M.B.)
| | - Martijn J Wilmer
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht, The Netherlands (M.F., K.G.C.W., M.F.B., N.S.R., R.M.); Aragon's Health Science Institutes (IACS), Zaragoza, Spain (N.S.M.); Departments of Pharmacology and Toxicology (M.J.W., J.K.) and Physiology (J.G.H.), Radboud Institute for Molecular Life Sciences, Radboud university medical center, Nijmegen, The Netherlands; Aragon Health Research Institute (IIS Aragon), Zaragoza, Spain (P.M.B.); Liver and Digestive Diseases Networking Biomedical Research Centre (CIBERehd), Madrid, Spain (P.M.B.); Jiménez Díaz Foundation Health Research Institute, Madrid, Spain (P.M.B.); and Department of Biomedical and Aerospace Engineering, Carlos III University of Madrid, Spain (P.M.B.)
| | - Janis Kerkering
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht, The Netherlands (M.F., K.G.C.W., M.F.B., N.S.R., R.M.); Aragon's Health Science Institutes (IACS), Zaragoza, Spain (N.S.M.); Departments of Pharmacology and Toxicology (M.J.W., J.K.) and Physiology (J.G.H.), Radboud Institute for Molecular Life Sciences, Radboud university medical center, Nijmegen, The Netherlands; Aragon Health Research Institute (IIS Aragon), Zaragoza, Spain (P.M.B.); Liver and Digestive Diseases Networking Biomedical Research Centre (CIBERehd), Madrid, Spain (P.M.B.); Jiménez Díaz Foundation Health Research Institute, Madrid, Spain (P.M.B.); and Department of Biomedical and Aerospace Engineering, Carlos III University of Madrid, Spain (P.M.B.)
| | - Pedro Miguel Baptista
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht, The Netherlands (M.F., K.G.C.W., M.F.B., N.S.R., R.M.); Aragon's Health Science Institutes (IACS), Zaragoza, Spain (N.S.M.); Departments of Pharmacology and Toxicology (M.J.W., J.K.) and Physiology (J.G.H.), Radboud Institute for Molecular Life Sciences, Radboud university medical center, Nijmegen, The Netherlands; Aragon Health Research Institute (IIS Aragon), Zaragoza, Spain (P.M.B.); Liver and Digestive Diseases Networking Biomedical Research Centre (CIBERehd), Madrid, Spain (P.M.B.); Jiménez Díaz Foundation Health Research Institute, Madrid, Spain (P.M.B.); and Department of Biomedical and Aerospace Engineering, Carlos III University of Madrid, Spain (P.M.B.)
| | - Joost G Hoenderop
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht, The Netherlands (M.F., K.G.C.W., M.F.B., N.S.R., R.M.); Aragon's Health Science Institutes (IACS), Zaragoza, Spain (N.S.M.); Departments of Pharmacology and Toxicology (M.J.W., J.K.) and Physiology (J.G.H.), Radboud Institute for Molecular Life Sciences, Radboud university medical center, Nijmegen, The Netherlands; Aragon Health Research Institute (IIS Aragon), Zaragoza, Spain (P.M.B.); Liver and Digestive Diseases Networking Biomedical Research Centre (CIBERehd), Madrid, Spain (P.M.B.); Jiménez Díaz Foundation Health Research Institute, Madrid, Spain (P.M.B.); and Department of Biomedical and Aerospace Engineering, Carlos III University of Madrid, Spain (P.M.B.)
| | - Rosalinde Masereeuw
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht, The Netherlands (M.F., K.G.C.W., M.F.B., N.S.R., R.M.); Aragon's Health Science Institutes (IACS), Zaragoza, Spain (N.S.M.); Departments of Pharmacology and Toxicology (M.J.W., J.K.) and Physiology (J.G.H.), Radboud Institute for Molecular Life Sciences, Radboud university medical center, Nijmegen, The Netherlands; Aragon Health Research Institute (IIS Aragon), Zaragoza, Spain (P.M.B.); Liver and Digestive Diseases Networking Biomedical Research Centre (CIBERehd), Madrid, Spain (P.M.B.); Jiménez Díaz Foundation Health Research Institute, Madrid, Spain (P.M.B.); and Department of Biomedical and Aerospace Engineering, Carlos III University of Madrid, Spain (P.M.B.)
| |
Collapse
|
46
|
Kim YK, Nam SA, Yang CW. Applications of kidney organoids derived from human pluripotent stem cells. Korean J Intern Med 2018; 33:649-659. [PMID: 29961307 PMCID: PMC6030416 DOI: 10.3904/kjim.2018.198] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/11/2018] [Accepted: 06/18/2018] [Indexed: 12/11/2022] Open
Abstract
The establishment of protocols to differentiate kidney organoids from human pluripotent stem cells provides potential applications of kidney organoids in regenerative medicine. Modeling of renal diseases, drug screening, nephrotoxicity testing of compounds, and regenerative therapy are attractive applications. Although much progress still remains to be made in the development of kidney organoids, recent advances in clustered regularly interspaced short palindromic repeat (CRISPR)-CRISPR-associated system 9 (Cas9) genome editing and three-dimensional bioprinting technologies have contributed to the application of kidney organoids in clinical fields. In this section, we review recent advances in the applications of kidney organoids to kidney disease modelling, drug screening, nephrotoxicity testing, and regenerative therapy.
Collapse
Affiliation(s)
- Yong Kyun Kim
- Cell Death Disease Research Center, The Catholic University of Korea, Seoul, Korea
- Department of Internal Medicine, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Sun Ah Nam
- Cell Death Disease Research Center, The Catholic University of Korea, Seoul, Korea
- Department of Internal Medicine, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Chul Woo Yang
- Department of Internal Medicine, College of Medicine, The Catholic University of Korea, Seoul, Korea
- Convergent Research Consortium for Immunologic Disease, and Division of Nephrology, College of Medicine, The Catholic University of Korea, Seoul, Korea
- Correspondence to Chul Woo Yang, M.D. Convergent Research Consortium for Immunologic Disease and Department of Internal Medicine, College of Medicine, Seoul St. Mary's Hospital, The Catholic University of Korea, 222 Banpo-daero, Seocho-gu, Seoul 06591, Korea Tel: +82-2-2258-6037 Fax: +82-2-22258-6917 E-mail:
| |
Collapse
|
47
|
Ellens H, Meng Z, Le Marchand SJ, Bentz J. Mechanistic kinetic modeling generates system-independent P-glycoprotein mediated transport elementary rate constants for inhibition and, in combination with 3D SIM microscopy, elucidates the importance of microvilli morphology on P-glycoprotein mediated efflux activity. Expert Opin Drug Metab Toxicol 2018; 14:571-584. [PMID: 29788828 DOI: 10.1080/17425255.2018.1480720] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/16/2022]
Abstract
INTRODUCTION In vitro transporter kinetics are typically analyzed by steady-state Michaelis-Menten approximations. However, no clear evidence exists that these approximations, applied to multiple transporters in biological membranes, yield system-independent mechanistic parameters needed for reliable in vivo hypothesis generation and testing. Areas covered: The classical mass action model has been developed for P-glycoprotein (P-gp) mediated transport across confluent polarized cell monolayers. Numerical integration of the mass action equations for transport using a stable global optimization program yields fitted elementary rate constants that are system-independent. The efflux active P-gp was defined by the rate at which P-gp delivers drugs to the apical chamber, since as much as 90% of drugs effluxed by P-gp partition back into nearby microvilli prior to reaching the apical chamber. The efflux active P-gp concentration was 10-fold smaller than the total expressed P-gp for Caco-2 cells, due to their microvilli membrane morphology. The mechanistic insights from this analysis are readily extrapolated to P-gp mediated transport in vivo. Expert opinion: In vitro system-independent elementary rate constants for transporters are essential for the generation and validation of robust mechanistic PBPK models. Our modeling approach and programs have broad application potential. They can be used for any drug transporter with minor adaptations.
Collapse
Affiliation(s)
- Harma Ellens
- a Department of Biology , Drexel University , Philadelphia , PA , USA
| | - Zhou Meng
- a Department of Biology , Drexel University , Philadelphia , PA , USA
| | | | - Joe Bentz
- a Department of Biology , Drexel University , Philadelphia , PA , USA
| |
Collapse
|
48
|
Drug transporter expression profiling in a three-dimensional kidney proximal tubule in vitro nephrotoxicity model. Pflugers Arch 2018; 470:1311-1323. [DOI: 10.1007/s00424-018-2150-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2018] [Revised: 04/20/2018] [Accepted: 04/27/2018] [Indexed: 01/09/2023]
|
49
|
El-Kattan AF, Varma MVS. Navigating Transporter Sciences in Pharmacokinetics Characterization Using the Extended Clearance Classification System. Drug Metab Dispos 2018; 46:729-739. [DOI: 10.1124/dmd.117.080044] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2017] [Accepted: 02/22/2018] [Indexed: 12/12/2022] Open
|
50
|
Sager G, Smaglyukova N, Fuskevaag OM. The role of OAT2 (SLC22A7) in the cyclic nucleotide biokinetics of human erythrocytes. J Cell Physiol 2018; 233:5972-5980. [PMID: 29244191 PMCID: PMC5947735 DOI: 10.1002/jcp.26409] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2017] [Accepted: 12/07/2017] [Indexed: 01/25/2023]
Abstract
The present study was conducted to characterise the transporter(s) responsible for the uptake of cyclic nucleotides to human erythrocytes. Western blotting showed that hRBC expressed OAT2 (SLC22A7), but detection of OAT1 (SLC22A6), or OAT3 (SLC22A8) was not possible. Intact hRBC were employed to clarify the simultaneous cyclic nucleotide egression and uptake. Both these opposing processes were studied. The Km‐values for high affinity efflux was 3.5 ± 0.1 and 39.4 ± 5.7 μM for cGMP and cAMP, respectively. The respective values for low affinity efflux were 212 ± 11 and 339 ± 42 μM. The uptake was characterised with apparently low affinity and similar Km‐values for cGMP (2.2 mM) and cAMP (0.89 mM). Using an iterative approach in order to balance uptake with efflux, the predicted real Km‐values for uptake were 100–200 μM for cGMP and 50–150 μM for cAMP. The established OAT2‐substrate indomethacin showed a competitive interaction with cyclic nucleotide uptake. Creatinine, also an OAT2 substrate, showed saturable uptake with a Km of 854 ± 98 μM. Unexpectedly, co‐incubation with cyclic nucleotides showed an uncompetitive inhibition. The observed Km‐values were 399 ± 44 and 259 ± 30 μM for creatinine, in the presence of cGMP and cAMP, respectively. Finally, the OAT1‐substrate para‐aminohippurate (PAH) showed some uptake (Km‐value of 2.0 ± 0.4 mM) but did not interact with cyclic nucleotide or indomethacin transport.
Collapse
Affiliation(s)
- Georg Sager
- Faculty of Health Science, Department of Medical Biology, Research Group of Experimental and Clinical Pharmacology, University of Tromsø - The Arctic University of Norway, Tromsø, Norway.,Laboratory Medicine, University Hospital of North Norway, Tromsø, Norway
| | - Natalia Smaglyukova
- Faculty of Health Science, Department of Medical Biology, Research Group of Experimental and Clinical Pharmacology, University of Tromsø - The Arctic University of Norway, Tromsø, Norway
| | - Ole-Martin Fuskevaag
- Faculty of Health Science, Department of Medical Biology, Research Group of Experimental and Clinical Pharmacology, University of Tromsø - The Arctic University of Norway, Tromsø, Norway.,Laboratory Medicine, University Hospital of North Norway, Tromsø, Norway
| |
Collapse
|