1
|
Premraj A, Aleyas AG, Nautiyal B, Rasool TJ. First report of a chemokine from camelids: Dromedary CXCL8 is induced by poxvirus and heavy metal toxicity. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2024; 161:105261. [PMID: 39241936 DOI: 10.1016/j.dci.2024.105261] [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: 06/14/2024] [Revised: 08/27/2024] [Accepted: 09/02/2024] [Indexed: 09/09/2024]
Abstract
Low molecular weight proteins, known as chemokines, facilitate the migration and localization of immune cells to the site of infection and injury. One of the first chemokines identified, CXCL8 functions as a key neutrophil activator, recruiting neutrophils to sites of inflammation. Several viral infections, including zoonotic coronaviruses and poxviruses, have been reported to induce the expression of CXCL8. Dromedary camels are known to harbor several potentially zoonotic pathogens, but critical immune molecules such as chemokines remain unidentified. We report here the identification of CXCL8 from the dromedary camel - the first chemokine identified from camelids. The complete dromedary CXCL8 cDNA sequence as well as the corresponding gene sequence from dromedary and two New World camelids - alpaca and llama were cloned. CXCL8 mRNA expression was relatively higher in PBMC, spleen, lung, intestine, and liver. Poly(I:C) and lipopolysaccharide stimulated CXCL8 expression in vitro, while interferon treatment inhibited it. In vitro infection with potentially zoonotic camelpox virus induced the expression of CXCL8 in camel kidney cells. Toxicological studies on camelids have been limited, and no biomarkers have been identified. Hence, we also evaluated CXCL8 mRNA expression as a potential biomarker to assess heavy metal toxicity in camel kidney cells in vitro. CXCL8 expression was increased after in vitro exposure to heavy metal compounds of cobalt and cadmium, suggesting potential utility as a biomarker for renal toxicity in camels. The results of our study demonstrate that camel CXCL8 plays a significant role in immunomodulatory and induced toxicity responses in dromedary camels.
Collapse
Affiliation(s)
- Avinash Premraj
- Camel Biotechnology Center, Presidential Camels & Camel Racing Affairs Centre, Department of the President's Affairs, PO Box 17292, Al Ain, United Arab Emirates
| | - Abi George Aleyas
- Camel Biotechnology Center, Presidential Camels & Camel Racing Affairs Centre, Department of the President's Affairs, PO Box 17292, Al Ain, United Arab Emirates
| | - Binita Nautiyal
- Camel Biotechnology Center, Presidential Camels & Camel Racing Affairs Centre, Department of the President's Affairs, PO Box 17292, Al Ain, United Arab Emirates
| | - Thaha Jamal Rasool
- Camel Biotechnology Center, Presidential Camels & Camel Racing Affairs Centre, Department of the President's Affairs, PO Box 17292, Al Ain, United Arab Emirates.
| |
Collapse
|
2
|
Kang S, Chen EC, Cifuentes H, Co JY, Cole G, Graham J, Hsia R, Kiyota T, Klein JA, Kroll KT, Nieves Lopez LM, Norona LM, Peiris H, Potla R, Romero-Lopez M, Roth JG, Tseng M, Fullerton AM, Homan KA. Complex in vitromodels positioned for impact to drug testing in pharma: a review. Biofabrication 2024; 16:042006. [PMID: 39189069 DOI: 10.1088/1758-5090/ad6933] [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: 12/22/2023] [Accepted: 07/30/2024] [Indexed: 08/28/2024]
Abstract
Recent years have seen the creation and popularization of various complexin vitromodels (CIVMs), such as organoids and organs-on-chip, as a technology with the potential to reduce animal usage in pharma while also enhancing our ability to create safe and efficacious drugs for patients. Public awareness of CIVMs has increased, in part, due to the recent passage of the FDA Modernization Act 2.0. This visibility is expected to spur deeper investment in and adoption of such models. Thus, end-users and model developers alike require a framework to both understand the readiness of current models to enter the drug development process, and to assess upcoming models for the same. This review presents such a framework for model selection based on comparative -omics data (which we term model-omics), and metrics for qualification of specific test assays that a model may support that we term context-of-use (COU) assays. We surveyed existing healthy tissue models and assays for ten drug development-critical organs of the body, and provide evaluations of readiness and suggestions for improving model-omics and COU assays for each. In whole, this review comes from a pharma perspective, and seeks to provide an evaluation of where CIVMs are poised for maximum impact in the drug development process, and a roadmap for realizing that potential.
Collapse
Affiliation(s)
- Serah Kang
- Complex in vitro Systems Group, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, United States of America
| | - Eugene C Chen
- Department of Drug Metabolism and Pharmacokinetics, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, United States of America
| | - Helen Cifuentes
- Complex in vitro Systems Group, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, United States of America
| | - Julia Y Co
- Complex in vitro Systems Group, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, United States of America
| | - Gabrielle Cole
- Investigative Toxicology, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, United States of America
| | - Jessica Graham
- Product Quality & Occupational Toxicology, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, United States of Americaica
| | - Rebecca Hsia
- Complex in vitro Systems Group, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, United States of America
| | - Tomomi Kiyota
- Investigative Toxicology, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, United States of America
| | - Jessica A Klein
- Complex in vitro Systems Group, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, United States of America
| | - Katharina T Kroll
- Complex in vitro Systems Group, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, United States of America
| | - Lenitza M Nieves Lopez
- Complex in vitro Systems Group, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, United States of America
| | - Leah M Norona
- Investigative Toxicology, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, United States of America
| | - Heshan Peiris
- Human Genetics, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, United States of America
| | - Ratnakar Potla
- Complex in vitro Systems Group, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, United States of America
| | - Monica Romero-Lopez
- Complex in vitro Systems Group, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, United States of America
| | - Julien G Roth
- Complex in vitro Systems Group, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, United States of America
| | - Min Tseng
- Investigative Toxicology, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, United States of America
| | - Aaron M Fullerton
- Investigative Toxicology, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, United States of America
| | - Kimberly A Homan
- Complex in vitro Systems Group, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, United States of America
| |
Collapse
|
3
|
Yu P, Zhu H, Bosholm CC, Beiner D, Duan Z, Shetty AK, Mou SS, Kramer PA, Barroso LF, Liu H, Cheng K, Ihnat M, Gorris MA, Aloi JA, Woldemichael JA, Bleyer A, Zhang Y. Precision nephrotoxicity testing using 3D in vitro models. Cell Biosci 2023; 13:231. [PMID: 38129901 PMCID: PMC10740310 DOI: 10.1186/s13578-023-01187-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Accepted: 12/15/2023] [Indexed: 12/23/2023] Open
Abstract
Nephrotoxicity is a significant concern during the development of new drugs or when assessing the safety of chemicals in consumer products. Traditional methods for testing nephrotoxicity involve animal models or 2D in vitro cell cultures, the latter of which lack the complexity and functionality of the human kidney. 3D in vitro models are created by culturing human primary kidney cells derived from urine in a 3D microenvironment that mimics the fluid shear stresses of the kidney. Thus, 3D in vitro models provide more accurate and reliable predictions of human nephrotoxicity compared to existing 2D models. In this review, we focus on precision nephrotoxicity testing using 3D in vitro models with human autologous urine-derived kidney cells as a promising approach for evaluating drug safety.
Collapse
Affiliation(s)
- Pengfei Yu
- Wake Forest Institute for Regenerative Medicine, Wake Forest University Health Sciences, Winston-Salem, NC, USA
- The Fourth Department of Liver Disease, Beijing You An Hospital, Capital Medical University, Beijing, China
| | - Hainan Zhu
- Wake Forest Institute for Regenerative Medicine, Wake Forest University Health Sciences, Winston-Salem, NC, USA
| | - Carol Christine Bosholm
- Wake Forest Institute for Regenerative Medicine, Wake Forest University Health Sciences, Winston-Salem, NC, USA
| | - Daniella Beiner
- Wake Forest Institute for Regenerative Medicine, Wake Forest University Health Sciences, Winston-Salem, NC, USA
| | - Zhongping Duan
- The Fourth Department of Liver Disease, Beijing You An Hospital, Capital Medical University, Beijing, China
| | - Avinash K Shetty
- Department of Pediatrics, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Steve S Mou
- Department of Anesthesiology and Pediatrics, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Philip Adam Kramer
- Department of Internal Medicine, Section on Gerontology and Geriatrics, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Luis F Barroso
- Internal Medicine/Infectious Diseases, Wake Forest University Health Sciences, Winston-Salem, NC, USA
| | - Hongbing Liu
- Department of Pediatrics and The Tulane Hypertension and Renal Center of Excellence, Tulane University School of Medicine, Tulane Avenue, New Orleans, LA, USA
| | - Kun Cheng
- Division of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Missouri-Kansas City, 2464 Charlotte Street, Kansas City, MO, 64108, USA
| | - Michael Ihnat
- Department of Pharmaceutical Sciences, University of Oklahoma College of Pharmacy, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Matthew A Gorris
- Division of Endocrinology and Metabolism at Wake Forest Baptist Health, Winston-Salem, NC, USA
| | - Joseph A Aloi
- Division of Endocrinology and Metabolism at Wake Forest Baptist Health, Winston-Salem, NC, USA
| | - Jobira A Woldemichael
- Division of Nephrology, Wake Forest University Health Sciences, Winston-Salem, NC, USA
| | - Anthony Bleyer
- Division of Nephrology, Wake Forest University Health Sciences, Winston-Salem, NC, USA
| | - Yuanyuan Zhang
- Wake Forest Institute for Regenerative Medicine, Wake Forest University Health Sciences, Winston-Salem, NC, USA.
| |
Collapse
|
4
|
Hsiao HY, Yen TH, Wu FY, Cheng CM, Liu JW, Fan YT, Huang JJ, Nien CY. Delivery and Transcriptome Assessment of an In Vitro Three-Dimensional Proximal Tubule Model Established by Human Kidney 2 Cells in Clinical Gelatin Sponges. Int J Mol Sci 2023; 24:15547. [PMID: 37958530 PMCID: PMC10650118 DOI: 10.3390/ijms242115547] [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: 09/21/2023] [Revised: 10/10/2023] [Accepted: 10/14/2023] [Indexed: 11/15/2023] Open
Abstract
The high prevalence of kidney diseases and the low identification rate of drug nephrotoxicity in preclinical studies reinforce the need for representative yet feasible renal models. Although in vitro cell-based models utilizing renal proximal tubules are widely used for kidney research, many proximal tubule cell (PTC) lines have been indicated to be less sensitive to nephrotoxins, mainly due to altered expression of transporters under a two-dimensional culture (2D) environment. Here, we selected HK-2 cells to establish a simplified three-dimensional (3D) model using gelatin sponges as scaffolds. In addition to cell viability and morphology, we conducted a comprehensive transcriptome comparison and correlation analysis of 2D and 3D cultured HK-2 cells to native human PTCs. Our 3D model displayed stable and long-term growth with a tubule-like morphology and demonstrated a more comparable gene expression profile to native human PTCs compared to the 2D model. Many missing or low expressions of major genes involved in PTC transport and metabolic processes were restored, which is crucial for successful nephrotoxicity prediction. Consequently, we established a cost-effective yet more representative model for in vivo PTC studies and presented a comprehensive transcriptome analysis for the systematic characterization of PTC lines.
Collapse
Affiliation(s)
- Hui-Yi Hsiao
- Department of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan;
- Center for Tissue Engineering, Linkuo Chang Gung Memorial Hospital, Taoyuan 33305, Taiwan;
| | - Tzung-Hai Yen
- Department of Nephrology, Clinical Poison Center, Linkuo Chang Gung Memorial Hospital, Taoyuan 33305, Taiwan;
- Department of Nephrology, College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan
| | - Fang-Yu Wu
- Department of Life Science, National Central University, Taoyuan 32001, Taiwan; (F.-Y.W.); (Y.-T.F.)
| | - Chao-Min Cheng
- Institute of Biomedical Engineering, National Tsing Hua University, Hsinchu 300193, Taiwan;
| | - Jia-Wei Liu
- Center for Tissue Engineering, Linkuo Chang Gung Memorial Hospital, Taoyuan 33305, Taiwan;
| | - Yu-Ting Fan
- Department of Life Science, National Central University, Taoyuan 32001, Taiwan; (F.-Y.W.); (Y.-T.F.)
| | - Jung-Ju Huang
- Division of Reconstructive Microsurgery, Department of Plastic and Reconstructive Surgery, Linkou Chang Gung Memorial Hospital, Taoyuan 33305, Taiwan;
| | - Chung-Yi Nien
- Department of Life Science, National Central University, Taoyuan 32001, Taiwan; (F.-Y.W.); (Y.-T.F.)
| |
Collapse
|
5
|
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
|
6
|
Piossek F, Beneke S, Schlichenmaier N, Mucic G, Drewitz S, Dietrich DR. Physiological oxygen and co-culture with human fibroblasts facilitate in vivo-like properties in human renal proximal tubular epithelial cells. Chem Biol Interact 2022; 361:109959. [DOI: 10.1016/j.cbi.2022.109959] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Revised: 04/08/2022] [Accepted: 04/12/2022] [Indexed: 11/28/2022]
|
7
|
Lawrence M, Elhendawi M, Morlock M, Liu W, Liu S, Palakkan A, Seidl L, Hohenstein P, Sjögren A, Davies J. Human iPSC-derived renal organoids engineered to report oxidative stress can predict drug-induced toxicity. iScience 2022; 25:103884. [PMID: 35243244 PMCID: PMC8861638 DOI: 10.1016/j.isci.2022.103884] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Revised: 05/10/2021] [Accepted: 02/03/2022] [Indexed: 01/08/2023] Open
Abstract
Advances in regenerative medicine have led to the construction of many types of organoids, which reproduce important aspects of endogenous organs but may be limited or disorganized in nature. While their usefulness for restoring function remains unclear, they have undoubted usefulness in research, diagnostics, and toxicology. In toxicology, there is an urgent need for better models for human kidneys. We used human iPS-cell (hiPSC)-derived renal organoids to identify HMOX1 as a useful marker of toxic stress via the oxidative stress pathway, and then constructed an HMOX1 reporter in hiPSCs. We used two forms of hiPSC-derived HMOX1-reporter renal organoids to probe their ability to detect nephrotoxicants in a panel of blind-coded compounds. Our results highlight the potential usefulness, and some limitations, of HMOX1-reporter renal organoids as screening tools. The results may guide development of similar stress-reporting organoid assays for other stem-cell-derived organs and tissues.
Collapse
Affiliation(s)
- M.L. Lawrence
- Deanery of Biomedical Sciences, University of Edinburgh, Hugh Robson Building, George Square, Edinburgh, EH8 9XD UK
| | - M. Elhendawi
- Deanery of Biomedical Sciences, University of Edinburgh, Hugh Robson Building, George Square, Edinburgh, EH8 9XD UK
- Clinical Pathology Department, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| | - M. Morlock
- R&D Graduate, R&D, AstraZeneca, Gothenburg, Sweden
| | - W. Liu
- SynthSys Centre for Synthetic and Systems Biology, UK Centre for Mammalian Synthetic Biology, School of Biological Sciences, University of Edinburgh, C.H Waddington Building, Max Born Crescent, Edinburgh, EH9 3BF, UK
| | - S. Liu
- Deanery of Biomedical Sciences, University of Edinburgh, Hugh Robson Building, George Square, Edinburgh, EH8 9XD UK
| | - A. Palakkan
- Deanery of Biomedical Sciences, University of Edinburgh, Hugh Robson Building, George Square, Edinburgh, EH8 9XD UK
| | - L.F. Seidl
- Deanery of Biomedical Sciences, University of Edinburgh, Hugh Robson Building, George Square, Edinburgh, EH8 9XD UK
| | - P. Hohenstein
- Leiden University Medical Center, Leiden University, Leiden, the Netherlands
- The Roslin Institute, The University of Edinburgh, Midlothian, UK
| | - A.K. Sjögren
- CVRM Safety, Clinical Pharmacology and Safety Science, R&D, AstraZeneca, Gothenburg, Sweden
| | - J.A. Davies
- Deanery of Biomedical Sciences, University of Edinburgh, Hugh Robson Building, George Square, Edinburgh, EH8 9XD UK
| |
Collapse
|
8
|
Bejoy J, Qian ES, Woodard LE. Tissue Culture Models of AKI: From Tubule Cells to Human Kidney Organoids. J Am Soc Nephrol 2022; 33:487-501. [PMID: 35031569 PMCID: PMC8975068 DOI: 10.1681/asn.2021050693] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
Abstract
AKI affects approximately 13.3 million people around the world each year, causing CKD and/or mortality. The mammalian kidney cannot generate new nephrons after postnatal renal damage and regenerative therapies for AKI are not available. Human kidney tissue culture systems can complement animal models of AKI and/or address some of their limitations. Donor-derived somatic cells, such as renal tubule epithelial cells or cell lines (RPTEC/hTERT, ciPTEC, HK-2, Nki-2, and CIHP-1), have been used for decades to permit drug toxicity screening and studies into potential AKI mechanisms. However, tubule cell lines do not fully recapitulate tubular epithelial cell properties in situ when grown under classic tissue culture conditions. Improving tissue culture models of AKI would increase our understanding of the mechanisms, leading to new therapeutics. Human pluripotent stem cells (hPSCs) can be differentiated into kidney organoids and various renal cell types. Injury to human kidney organoids results in renal cell-type crosstalk and upregulation of kidney injury biomarkers that are difficult to induce in primary tubule cell cultures. However, current protocols produce kidney organoids that are not mature and contain off-target cell types. Promising bioengineering techniques, such as bioprinting and "kidney-on-a-chip" methods, as applied to kidney nephrotoxicity modeling advantages and limitations are discussed. This review explores the mechanisms and detection of AKI in tissue culture, with an emphasis on bioengineered approaches such as human kidney organoid models.
Collapse
Affiliation(s)
- Julie Bejoy
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Eddie S. Qian
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Lauren E. Woodard
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee,Department of Veterans Affairs, Tennessee Valley Healthcare System, Nashville, Tennessee,Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee
| |
Collapse
|
9
|
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: 13.0] [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
|
10
|
Irvine AR, van Berlo D, Shekhani R, Masereeuw R. A systematic review of in vitro models of drug-induced kidney injury. CURRENT OPINION IN TOXICOLOGY 2021. [DOI: 10.1016/j.cotox.2021.06.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
11
|
Abstract
Drug induced kidney injury is one of the leading causes of failure of drug development programs in the clinic. Early prediction of renal toxicity potential of drugs is crucial to the success of drug candidates in the clinic. The dynamic nature of the functioning of the kidney and the presence of drug uptake proteins introduce additional challenges in the prediction of renal injury caused by drugs. Renal injury due to drugs can be caused by a wide variety of mechanisms and can be broadly classified as toxic or obstructive. Several biomarkers are available for in vitro and in vivo detection of renal injury. In vitro static and dynamic (microfluidic) cellular models and preclinical models can provide valuable information regarding the toxicity potential of drugs. Differences in pharmacology and subsequent disconnect in biomarker response, differences in the expression of transporter and enzyme proteins between in vitro to in vivo systems and between preclinical species and humans are some of the limitations of current experimental models. The progress in microfluidic (kidney-on-chip) platforms in combination with the ability of 3-dimensional cell culture can help in addressing some of these issues in the future. Finally, newer in silico and computational techniques like physiologically based pharmacokinetic modeling and machine learning have demonstrated potential in assisting prediction of drug induced kidney injury.
Collapse
Affiliation(s)
- Priyanka Kulkarni
- Department of Drug Metabolism and Pharmacokinetics, Millennium Pharmaceuticals, a fully owned subsidiary of Takeda Pharmaceuticals, Cambridge, MA, USA
| |
Collapse
|
12
|
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: 3.8] [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
|
13
|
Hussain F, Basu S, Heng JJH, Loo LH, Zink D. Predicting direct hepatocyte toxicity in humans by combining high-throughput imaging of HepaRG cells and machine learning-based phenotypic profiling. Arch Toxicol 2020; 94:2749-2767. [PMID: 32533217 DOI: 10.1007/s00204-020-02778-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2019] [Accepted: 05/05/2020] [Indexed: 02/07/2023]
Abstract
Accurate prediction of drug- and chemical-induced hepatotoxicity remains to be a problem for pharmaceutical companies as well as other industries and regulators. The goal of the current study was to develop an in vitro/in silico method for the rapid and accurate prediction of drug- and chemical-induced hepatocyte injury in humans. HepaRG cells were employed for high-throughput imaging in combination with phenotypic profiling. A reference set of 69 drugs and chemicals was screened at a range of 7 concentrations, and the cellular response values were used for training a supervised classifier and for determining assay performance by using tenfold cross-validation. The results showed that the best performing phenotypic features were related to nuclear translocation of RELA (RELA proto-oncogene, NF-kB subunit; also known as NF-kappa B p65), DNA organization, and the F-actin cytoskeleton. Using a subset of 30 phenotypic features, direct hepatocyte toxicity in humans could be predicted with a test sensitivity, specificity and balanced accuracy of 73%, 92%, and 83%, respectively. The method was applied to another set of 26 drugs and chemicals with unclear annotation and their hepatocyte toxicity in humans was predicted. The results also revealed that the identified discriminative phenotypic changes were related to cell death and cellular senescence. Whereas cell death-related endpoints are widely applied in in vitro toxicology, cellular senescence-related endpoints are not, although cellular senescence can be induced by various drugs and other small molecule compounds and plays an important role in liver injury and disease. These findings show how phenotypic profiling can reveal unexpected chemical-induced mechanisms in toxicology.
Collapse
Affiliation(s)
- Faezah Hussain
- NanoBio Lab and Institute of Bioengineering and Nanotechnology (IBN), 31 Biopolis Way, The Nanos, Singapore, 138669, Singapore
| | - Sreetama Basu
- Bioinformatics Institute, 30 Biopolis Street, #07-01 Matrix, Singapore, 138671, Singapore
| | - Javen Jun Hao Heng
- NanoBio Lab and Institute of Bioengineering and Nanotechnology (IBN), 31 Biopolis Way, The Nanos, Singapore, 138669, Singapore
| | - Lit-Hsin Loo
- Bioinformatics Institute, 30 Biopolis Street, #07-01 Matrix, Singapore, 138671, Singapore. .,Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, 16 Medical Drive, Singapore, 117600, Singapore.
| | - Daniele Zink
- NanoBio Lab and Institute of Bioengineering and Nanotechnology (IBN), 31 Biopolis Way, The Nanos, Singapore, 138669, Singapore.
| |
Collapse
|
14
|
Mossoba ME, Vohra SN, Bigley E, Sprando J, Wiesenfeld PL. Genetically Engineered Human Kidney Cells for Real-Time Cytotoxicity Testing In Vitro. Mol Biotechnol 2020; 62:252-259. [PMID: 32146690 DOI: 10.1007/s12033-020-00245-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Classic toxicology studies often utilize in vivo animal models. Newer approaches employing in vitro organ-specific cellular models have been developed in recent years to help accelerate the speed and reduce the cost of traditional toxicology testing. Toward the goal of supporting in vitro cellular model research with a regulatory application in mind, we have developed a 'designer' human kidney cell line called HK2-Vi that can fluorescently measure the cytotoxicity of potential toxins on proximal tubule cell viability in a direct exposure in vitro model. HK2-Vi was designed to be a reagent-less kinetic assay that can yield data on short- or long-term cell viability after toxin exposure. To generate HK2-Vi, we used monocistronic lentiviral transduction methods to genetically engineer a human kidney cell line called HK-2 to stably co-express two transgenes. The first is Perceval HR, which encodes a fluorescent biosensor of both cytosolic ATP and ADP and the second is pHRed, which encodes a biosensor of cytosolic pH. Relative levels of cellular ATP and ADP effectively serve as a reliable and robust indicator of cell viability. Because the fluorescence Perceval HR is pH-dependent, we co-expressed the pHRed genetic biosensor to correct for variations in pH if necessary. Heterogenous populations of transduced renal cells were enriched by flow cytometry before monoclonal cellular populations were isolated by cell culture methods. A single clonal population of co-transduced cells expressing both Perceval HR and pHRed was selected to be HK2-Vi. This established cell line can now serve as a tool for in vitro toxicology testing and the methods described herein serve as a model for developing designer cell lines derived from other organs.
Collapse
Affiliation(s)
- Miriam E Mossoba
- Neurotoxicology and In Vitro Toxicology Branch (NIVTB), Division of Applied Regulatory Toxicology (DART), Office of Applied Research and Safety Assessment (OARSA), Center for Food Safety and Applied Nutrition (CFSAN), Food and Drug Administration (FDA), Laurel, MD, USA.
| | - Sanah N Vohra
- Neurotoxicology and In Vitro Toxicology Branch (NIVTB), Division of Applied Regulatory Toxicology (DART), Office of Applied Research and Safety Assessment (OARSA), Center for Food Safety and Applied Nutrition (CFSAN), Food and Drug Administration (FDA), Laurel, MD, USA
| | - Elmer Bigley
- Immunobiology Branch (IB), Division of Virulence Assessment (DVA), Office of Applied Research and Safety Assessment (OARSA), Center for Food Safety and Applied Nutrition (CFSAN), Food and Drug Administration (FDA), Laurel, MD, USA
| | - Jessica Sprando
- Neurotoxicology and In Vitro Toxicology Branch (NIVTB), Division of Applied Regulatory Toxicology (DART), Office of Applied Research and Safety Assessment (OARSA), Center for Food Safety and Applied Nutrition (CFSAN), Food and Drug Administration (FDA), Laurel, MD, USA
| | - Paddy L Wiesenfeld
- Neurotoxicology and In Vitro Toxicology Branch (NIVTB), Division of Applied Regulatory Toxicology (DART), Office of Applied Research and Safety Assessment (OARSA), Center for Food Safety and Applied Nutrition (CFSAN), Food and Drug Administration (FDA), Laurel, MD, USA
| |
Collapse
|
15
|
Zink D, Chuah JKC, Ying JY. Assessing Toxicity with Human Cell-Based In Vitro Methods. Trends Mol Med 2020; 26:570-582. [PMID: 32470384 DOI: 10.1016/j.molmed.2020.01.008] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2019] [Revised: 01/02/2020] [Accepted: 01/21/2020] [Indexed: 01/01/2023]
Abstract
In toxicology, there is a strong push towards replacing animal experiments with alternative methods, which include cell-based in vitro methods for the assessment of adverse health effects in humans. High-throughput methods are of central interest due to the large and steadily growing numbers of compounds that require assessment. Tremendous progress has been made during the last decade in developing and applying such methods. Innovative technologies for addressing complex biological interactions include induced pluripotent stem cell- and organoid-based approaches, organotypic coculture systems, and microfluidic 'multiorgan' chips. Combining in vitro methods with bioinformatics and in silico modeling generates new powerful tools for toxicity assessment, and the rapid progress in the field is expected to continue.
Collapse
Affiliation(s)
- Daniele Zink
- NanoBio Lab, Agency for Science, Technology and Research (A*STAR), 31 Biopolis Way, The Nanos, #09-01, Singapore 138669, Singapore; Innovations in Food and Chemical Safety Programme, A*STAR, Singapore.
| | - Jacqueline Kai Chin Chuah
- NanoBio Lab, Agency for Science, Technology and Research (A*STAR), 31 Biopolis Way, The Nanos, #09-01, Singapore 138669, Singapore; Cellbae Pte Ltd, 31 Biopolis Way, The Nanos, Singapore 138669, Singapore
| | - Jackie Y Ying
- NanoBio Lab, Agency for Science, Technology and Research (A*STAR), 31 Biopolis Way, The Nanos, #09-01, Singapore 138669, Singapore.
| |
Collapse
|
16
|
Qiu X, Miao Y, Geng X, Zhou X, Li B. Evaluation of biomarkers for in vitro prediction of drug-induced nephrotoxicity in RPTEC/TERT1 cells. Toxicol Res (Camb) 2020; 9:91-100. [PMID: 32440340 DOI: 10.1093/toxres/tfaa005] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2019] [Revised: 02/04/2020] [Accepted: 02/18/2020] [Indexed: 12/16/2022] Open
Abstract
There have been intensive efforts to identify in vivo biomarkers that can be used to monitor drug-induced kidney damage before significant impairment occurs. Kidney injury molecule-1, neutrophil gelatinase-associated lipocalin, clusterin, β2-microglobulin and cystatin C (CysC) have been validated as clinical or preclinical biomarkers in urinary and plasma predictive of acute and chronic kidney injuries and diseases. A high-throughput in vitro assay predictive of nephrotoxicity could potentially be implemented in early drug discovery stage to reduce attrition at later stages of drug development. To assess the potential of these known in vivo biomarkers for in vitro evaluation of drug-induced nephrotoxicity, we selected four nephrotoxic agents (cisplatin, cyclosporin, aristolochic acid I and gentamicin) and detected their effects on the protein levels of nephrotoxic biomarkers in RPTEC/TERT1 cells. The protein levels of clusterin, CysC, GSTπ and TIMP-1 significantly increased in the conditioned media of RPTEC/TERT1 cells treated with cisplatin, cyclosporin, aristolochic acid I and gentamicin. The messenger RNA levels of clusterin, CysC, GSTπ and TIMP-1 also increased in RPTEC/TERT1 cells treated with cisplatin, cyclosporin, aristolochic acid I and gentamicin, indicating that drug-induced upregulation involves transcriptional activation. Taken together, the results clearly demonstrate that among the known in vivo nephrotoxic biomarkers, clusterin, CysC, GSTπ and TIMP-1 can be effectively used as in vitro biomarkers for drug-induced nephrotoxicity in RPTEC/TERT1 cells.
Collapse
Affiliation(s)
- Xuan Qiu
- Chinese Academy of Medical Sciences & Peking Union Medical College, No. 9, Dongdan Santiao, Dongcheng District, Beijing 100730, China.,National Center for Safety Evaluation of Drugs, National Institutes for Food and Drug Control, A8 Hongda Middle Street, Beijing Economic-Technological Development Area, Beijing 100176, China
| | - Yufa Miao
- National Center for Safety Evaluation of Drugs, National Institutes for Food and Drug Control, A8 Hongda Middle Street, Beijing Economic-Technological Development Area, Beijing 100176, China
| | - Xingchao Geng
- National Center for Safety Evaluation of Drugs, National Institutes for Food and Drug Control, A8 Hongda Middle Street, Beijing Economic-Technological Development Area, Beijing 100176, China
| | - Xiaobing Zhou
- National Center for Safety Evaluation of Drugs, National Institutes for Food and Drug Control, A8 Hongda Middle Street, Beijing Economic-Technological Development Area, Beijing 100176, China
| | - Bo Li
- Chinese Academy of Medical Sciences & Peking Union Medical College, No. 9, Dongdan Santiao, Dongcheng District, Beijing 100730, China.,National Center for Safety Evaluation of Drugs, National Institutes for Food and Drug Control, A8 Hongda Middle Street, Beijing Economic-Technological Development Area, Beijing 100176, China
| |
Collapse
|
17
|
Acetoacetate enhances oxidative metabolism and response to toxicants of cultured kidney cells. Toxicol Lett 2020; 323:19-24. [DOI: 10.1016/j.toxlet.2020.01.015] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2019] [Revised: 01/16/2020] [Accepted: 01/17/2020] [Indexed: 01/16/2023]
|
18
|
In vitro prediction of organ toxicity: the challenges of scaling and secondary mechanisms of toxicity. Arch Toxicol 2020; 94:353-356. [PMID: 32067068 PMCID: PMC8211595 DOI: 10.1007/s00204-020-02669-7] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2020] [Accepted: 01/27/2020] [Indexed: 12/31/2022]
|
19
|
Phillips JA, Grandhi TSP, Davis M, Gautier JC, Hariparsad N, Keller D, Sura R, Van Vleet TR. A pharmaceutical industry perspective on microphysiological kidney systems for evaluation of safety for new therapies. LAB ON A CHIP 2020; 20:468-476. [PMID: 31989145 DOI: 10.1039/c9lc00925f] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
The human kidney contains approximately one million nephrons. As the functional unit of the kidney, the nephron affords an opportunity to approximate the kidney at a microphysiological scale. Recent emergence of physiologically accurate human tissue models has radically advanced the possibilities of mimicking organ biology and multi-organ combinations in vitro. Anatomically, the nephron is one of the most complex, sequentially integrated microfluidic units in the body making the miniaturized microfluidic systems excellent candidates for capturing the kidney biology in vitro. While these models are promising, there are a number of considerations for practical implementation into a drug development paradigm. Opportunities for pharmaceutical industry applications of new MPS models often start with drug safety testing. As such, the intent of this article is to focus on safety and ADME applications. This article reviews biological functions of the kidney and options for characterizing known roles in nephrotoxicity. The concept of "context-of-use" is introduced as a framework for describing and verifying the specific features of an MPS platform for use in drug development. Overall, we present a perspective on key attributes of microphysiological kidney models, which the pharmaceutical industry could leverage to improve confident safety and ADME evaluations of experimental therapies.
Collapse
Affiliation(s)
| | - Taraka Sai Pavan Grandhi
- The Genomics Institute of the Novartis Research Foundation, 10675 John J Hopkins Drive, San Diego, CA 92121, USA
| | - Myrtle Davis
- Bristol-Myers Squibb Company, Province Line Road, Princeton, New Jersey 08648, USA
| | | | | | - Douglas Keller
- Sanofi US, 55 Corporate Drive, Bridgewater, NJ 08807, USA
| | - Radhakrishna Sura
- Preclinical Safety, AbbVie, 1 Waukegan Rd, N Chicago, IL 60064, USA.
| | - Terry R Van Vleet
- Preclinical Safety, AbbVie, 1 Waukegan Rd, N Chicago, IL 60064, USA.
| |
Collapse
|
20
|
Abstract
Drug attrition related to kidney toxicity remains a challenge in drug discovery and development. In vitro models established over the past 2 decades to supplement in vivo studies have improved the throughput capacity of toxicity evaluation, but usually suffer from low predictive value. To achieve a paradigm shift in the prediction of drug-induced kidney toxicity, two aspects are fundamental: increased physiological relevance of the kidney model, and use of appropriate toxicity end points. Recent studies have suggested that increasing the physiological relevance of kidney models can improve their sensitivity to drug-induced damage. Here, we discuss how advanced culture models, including modified cell lines, induced pluripotent stem cells, kidney organoid cultures, and microfluidic devices enhance in vivo similarity. To this end, culture models aim to increase the proximal tubule epithelial phenotype, reconstitute multiple tissue compartments and extracellular matrix, allow exposure to fluid shear stress, and enable interaction between multiple cell types. Applying computation-aided end points and novel biomarkers to advanced culture models will further improve sensitivity and clinical relevance of in vitro drug-induced toxicity prediction. Implemented at the right stage of drug discovery and development and coupled to high-content evaluation techniques, these models have the potential to reduce attrition and aid the selection of candidate drugs with an appropriate safety profile.
Collapse
Affiliation(s)
- Tom T G Nieskens
- CVRMSafety, Drug Safety and Metabolism, IMED Biotech Unit, AstraZeneca, Gothenburg, Sweden
| | - Anna-Karin Sjögren
- CVRMSafety, Drug Safety and Metabolism, IMED Biotech Unit, AstraZeneca, Gothenburg, Sweden.
| |
Collapse
|
21
|
Titmarsh DM, Nurcombe V, Cheung C, Cool SM. Vascular Cells and Tissue Constructs Derived from Human Pluripotent Stem Cells for Toxicological Screening. Stem Cells Dev 2019; 28:1347-1364. [PMID: 31397206 DOI: 10.1089/scd.2018.0246] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The ability of human stem cells to generate somatic cell lineages makes them ideal candidates for use in toxicological testing and eventually, preclinical drug development. Such resources would support an evolution away from human primary cells or research animal models, which suffer from variability and poor predictability, toward off-the-shelf assays of chemical toxicity and drug efficacy using human cells and tissues. To this end, we generated vascular cell populations (smooth muscle cells and endothelial cells) from human pluripotent stem cells (hPSCs), arranged them into 3D co-cultures within supportive gel matrices, and directed their propensity for self-organization resembling microvasculature. The resulting vascular cell populations and co-cultured constructs were then arrayed in high throughput and used for screening a library of environmental and clinical chemical agents for immunological and toxicological responses. The screen effectively stratified the chemicals into various levels of toxicity, with both cell type-specific and co-culture-dependent responses observed. Thus, hPSC-derived vascular cells and constructs could be progressed further toward use in toxicant and drug screening.
Collapse
Affiliation(s)
- Drew M Titmarsh
- Institute of Medical Biology and Agency for Science Technology and Research (A*STAR), Singapore, Singapore
| | - Victor Nurcombe
- Institute of Medical Biology and Agency for Science Technology and Research (A*STAR), Singapore, Singapore.,Lee Kong Chian School of Medicine, Nanyang Technological University-Imperial College London, Singapore, Singapore
| | - Christine Cheung
- Lee Kong Chian School of Medicine, Nanyang Technological University-Imperial College London, Singapore, Singapore.,Institute of Molecular and Cell Biology, Agency for Science Technology and Research (A*STAR), Singapore, Singapore
| | - Simon M Cool
- Institute of Medical Biology and Agency for Science Technology and Research (A*STAR), Singapore, Singapore.,Department of Orthopaedic Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| |
Collapse
|
22
|
Sachinidis A, Albrecht W, Nell P, Cherianidou A, Hewitt NJ, Edlund K, Hengstler JG. Road Map for Development of Stem Cell-Based Alternative Test Methods. Trends Mol Med 2019; 25:470-481. [PMID: 31130451 DOI: 10.1016/j.molmed.2019.04.003] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2019] [Revised: 03/28/2019] [Accepted: 04/02/2019] [Indexed: 12/12/2022]
Abstract
Much progress has been made in establishing strategies for differentiation of induced human pluripotent stem cells (hiPSCs). However, differentiated hiPSCs are not yet routinely used for prediction of toxicity. Here, limiting factors are summarised and possibilities for improvement are discussed, with a focus on hepatocytes, cardiomyocytes, tubular epithelial cells, and developmental toxicity. Moreover, we make recommendations for further fine-tuning of differentiation protocols for hiPSCs to hepatocyte-like cells by comparing individual steps of currently available protocols to the mechanisms occurring during embryonic development. A road map is proposed to facilitate test system development, including a description of the most useful performance metrics.
Collapse
Affiliation(s)
- Agapios Sachinidis
- Institute of Neurophysiology and Centre for Molecular Medicine Cologne (CMMC), University of Cologne (UKK), Cologne, Germany.
| | - Wiebke Albrecht
- Leibniz Research Centre for Working Environment and Human Factors, Technical University of Dortmund (IfADo), 44139 Dortmund, Germany
| | - Patrick Nell
- Leibniz Research Centre for Working Environment and Human Factors, Technical University of Dortmund (IfADo), 44139 Dortmund, Germany
| | - Anna Cherianidou
- Institute of Neurophysiology and Centre for Molecular Medicine Cologne (CMMC), University of Cologne (UKK), Cologne, Germany
| | | | - Karolina Edlund
- Leibniz Research Centre for Working Environment and Human Factors, Technical University of Dortmund (IfADo), 44139 Dortmund, Germany
| | - Jan G Hengstler
- Leibniz Research Centre for Working Environment and Human Factors, Technical University of Dortmund (IfADo), 44139 Dortmund, Germany.
| |
Collapse
|
23
|
Ojeda AS, Ford SD, Gallucci RM, Ihnat MA, Philp RP. Geochemical characterization and renal cell toxicity of water-soluble extracts from U.S. Gulf Coast lignite. ENVIRONMENTAL GEOCHEMISTRY AND HEALTH 2019; 41:1037-1053. [PMID: 30276587 DOI: 10.1007/s10653-018-0196-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/18/2018] [Accepted: 09/21/2018] [Indexed: 06/08/2023]
Abstract
An assortment of organic material can leach from lignite (low-rank coal) in water, and the water-soluble fraction from lignite has been associated with adverse health effects in areas of the Balkans. Recent efforts have been made to evaluate this hypothesis in other areas where lignite is in contact with groundwater like in the U.S. Gulf Coast region. In this study, five Gulf Coast lignite samples were extracted with water, and the water-soluble portion of the coal was then characterized by total organic carbon, UV-Vis spectroscopy, and gas chromatography/mass spectrometry. Additionally, human kidney cells (HK-2) were exposed to water-soluble extracts of Gulf Coast lignite to assess toxicity. Cell viability was measured, and a dose-response curve was used to generate IC50 values that ranged from 490 to 3000 ppm. The most toxic extract (Dolet Hills) was from Louisiana where lignite-derived organic material has been previously linked to high incidence of renal pelvic cancer. Concentrations of nephrotoxic metals (As, Cd, Co, Cu, Hg, Pb, V, Zn) were screened and were below those considered toxic to renal cells. We conclude that leachates from lignite do indeed have toxic affects on cultured human renal cells. Although the IC50 values are higher than the concentration of organic matter in the local groundwater, typically < 5 ppm, the effects of long-term low-level exposure is not known.
Collapse
Affiliation(s)
- A S Ojeda
- Department of Geology and Geophysics, The University of Oklahoma, Norman, OK, 73019, USA.
| | - S D Ford
- Department of Pharmaceutical Sciences, College of Pharmacy, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA
| | - R M Gallucci
- Department of Pharmaceutical Sciences, College of Pharmacy, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA
| | - M A Ihnat
- Department of Pharmaceutical Sciences, College of Pharmacy, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA
| | - R P Philp
- Department of Geology and Geophysics, The University of Oklahoma, Norman, OK, 73019, USA
| |
Collapse
|
24
|
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: 1.6] [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
|
25
|
Zink D. Comment on Sjögren et al. (2018) A novel multi-parametric high-content screening assay in ciPTEC-OAT1 to predict drug-induced nephrotoxicity in drug discovery. Arch Toxicol 92(10):3175-3190. Arch Toxicol 2018; 93:221-223. [PMID: 30328497 DOI: 10.1007/s00204-018-2327-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2018] [Accepted: 10/08/2018] [Indexed: 11/27/2022]
Abstract
Rapid progress is made in the development of high-content screening assays for the prediction of nephrotoxicity. The findings from different laboratories are consistent with respect to endpoints and concentration ranges screened. Discrepancies regarding compound annotation and the predictive performance analysis are discussed.
Collapse
Affiliation(s)
- Daniele Zink
- Institute of Bioengineering and Nanotechnology, 31 Biopolis Way, The Nanos, 138669, Singapore, Singapore.
| |
Collapse
|
26
|
Bajaj P, Rodrigues AD, Steppan CM, Engle SJ, Mathialagan S, Schroeter T. Human Pluripotent Stem Cell–Derived Kidney Model for Nephrotoxicity Studies. Drug Metab Dispos 2018; 46:1703-1711. [DOI: 10.1124/dmd.118.082727] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Accepted: 08/28/2018] [Indexed: 12/29/2022] Open
|
27
|
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: 4.7] [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
|
28
|
Qiu X, Zhou X, Miao Y, Li B. An in vitro method for nephrotoxicity evaluation using HK-2 human kidney epithelial cells combined with biomarkers of nephrotoxicity. Toxicol Res (Camb) 2018; 7:1205-1213. [PMID: 30510689 DOI: 10.1039/c8tx00095f] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2018] [Accepted: 08/06/2018] [Indexed: 02/02/2023] Open
Abstract
The kidney is one of the major target organs for drug-induced toxicity. During drug development, the traditional markers of nephrotoxicity indicate only severe and late damage, which leads to high costs. The new biomarkers are needed for a more sensitive and reliable evaluation of nephrotoxicity, especially for the regulatory accepted and validated in vitro model. We developed an in vitro model based on the HK-2 cell using the biomarkers of nephrotoxicity as endpoints for the evaluation of nephrotoxicity. The predictive performance of the biomarkers including LDH, GGT, KIM-1, clusterin, CysC, NGAL, TIMP-1, GSTπ and osteopontin was evaluated with 22 well characterized compounds. The area under the curve (AUC) values of KIM-1, clusterin, CysC and osteopontin ranged between 0.79 and 0.84. The combination of clusterin, KIM-1 and/or osteopontin improved the AUC value (ranging between 0.88 and 0.95) compared to one biomarker. Taken together, these results suggest that the model based on the HK-2 cell using clusterin, osteopontin, CysC and KIM-1 as endpoints would allow the prediction of nephrotoxicity at early preclinical stages.
Collapse
Affiliation(s)
- Xuan Qiu
- Chinese Academy of Medical Sciences & Peking Union Medical College , No. 9 , Dongdan Santiao , Dongcheng District , Beijing 100730 , China . .,National Center for Safety Evaluation of Drugs , National Institutes for Food and Drug Control , A8 Hongda Middle Street , Beijing Economic-Technological Development Area , Beijing 100176 , China
| | - Xiaobing Zhou
- National Center for Safety Evaluation of Drugs , National Institutes for Food and Drug Control , A8 Hongda Middle Street , Beijing Economic-Technological Development Area , Beijing 100176 , China
| | - Yufa Miao
- National Center for Safety Evaluation of Drugs , National Institutes for Food and Drug Control , A8 Hongda Middle Street , Beijing Economic-Technological Development Area , Beijing 100176 , China
| | - Bo Li
- Chinese Academy of Medical Sciences & Peking Union Medical College , No. 9 , Dongdan Santiao , Dongcheng District , Beijing 100730 , China . .,National Center for Safety Evaluation of Drugs , National Institutes for Food and Drug Control , A8 Hongda Middle Street , Beijing Economic-Technological Development Area , Beijing 100176 , China
| |
Collapse
|
29
|
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: 9.2] [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
|
30
|
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: 3.3] [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
|
31
|
Soo JYC, Jansen J, Masereeuw R, Little MH. Advances in predictive in vitro models of drug-induced nephrotoxicity. Nat Rev Nephrol 2018; 14:378-393. [PMID: 29626199 PMCID: PMC6013592 DOI: 10.1038/s41581-018-0003-9] [Citation(s) in RCA: 123] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
In vitro screens for nephrotoxicity are currently poorly predictive of toxicity in humans. Although the functional proteins that are expressed by nephron tubules and mediate drug susceptibility are well known, current in vitro cellular models poorly replicate both the morphology and the function of kidney tubules and therefore fail to demonstrate injury responses to drugs that would be nephrotoxic in vivo. Advances in protocols to enable the directed differentiation of pluripotent stem cells into multiple renal cell types and the development of microfluidic and 3D culture systems have opened a range of potential new platforms for evaluating drug nephrotoxicity. Many of the new in vitro culture systems have been characterized by the expression and function of transporters, enzymes, and other functional proteins that are expressed by the kidney and have been implicated in drug-induced renal injury. In vitro platforms that express these proteins and exhibit molecular biomarkers that have been used as readouts of injury demonstrate improved functional maturity compared with static 2D cultures and represent an opportunity to model injury to renal cell types that have hitherto received little attention. As nephrotoxicity screening platforms become more physiologically relevant, they will facilitate the development of safer drugs and improved clinical management of nephrotoxicants.
Collapse
Affiliation(s)
- Joanne Y-C Soo
- Department of Paediatrics, The University of Melbourne, Parkville, Victoria, Australia
- Murdoch Children's Research Institute, Parkville, Victoria, Australia
| | - Jitske Jansen
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, Netherlands
| | - Rosalinde Masereeuw
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, Netherlands
| | - Melissa H Little
- Department of Paediatrics, The University of Melbourne, Parkville, Victoria, Australia.
- Murdoch Children's Research Institute, Parkville, Victoria, Australia.
- Department of Anatomy and Neuroscience, The University of Melbourne, Parkville, Victoria, Australia.
| |
Collapse
|
32
|
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: 1.3] [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]
|
33
|
Abstract
The Lush Science Prize 2016 was awarded to Daniele Zink and Lit-Hsin Loo for the interdisciplinary and collaborative work between their research groups in developing alternative methods for the prediction of nephrotoxicity in humans. The collaboration has led to the establishment of a series of pioneering alternative methods for nephrotoxicity prediction, which includes: predictive gene expression markers based on pro-inflammatory responses; predictive in vitro cellular models based on pluripotent stem cell-derived proximal tubular-like cells; and predictive cellular phenotypic markers based on chromatin and cytoskeletal changes. A high-throughput method was established for chemical testing, which is currently being used to predict the potential human nephrotoxicity of ToxCast compounds in collaboration with the US Environmental Protection Agency. Similar high-throughput imaging-based methodologies are currently being developed and adapted by the Zink and Loo groups, to include other human organs and cell types. The ultimate goal is to develop a portfolio of methods accepted for the accurate prediction of human organ-specific toxicity and the consequent replacement of animal experiments.
Collapse
Affiliation(s)
- Lit-Hsin Loo
- Bioinformatics Institute (BII), Singapore and Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Daniele Zink
- Institute of Bioengineering and Nanotechnology (IBN), Singapore
| |
Collapse
|
34
|
Sakolish C, Mahler GJ. A novel microfluidic device to model the human proximal tubule and glomerulus. RSC Adv 2017. [DOI: 10.1039/c6ra25641d] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
A novel multi-channel microfluidic device to model human glomerular filtration and proximal tubular re-uptake.
Collapse
|
35
|
Satarug S, Vesey DA, Gobe GC. Kidney Cadmium Toxicity, Diabetes and High Blood Pressure: The Perfect Storm. TOHOKU J EXP MED 2017; 241:65-87. [DOI: 10.1620/tjem.241.65] [Citation(s) in RCA: 69] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Affiliation(s)
- Soisungwan Satarug
- Centre for Kidney Disease Research, Translational Research Institute and The University of Queensland Diamantina Institute
| | - David A. Vesey
- Centre for Kidney Disease Research, Translational Research Institute and The University of Queensland Diamantina Institute
| | - Glenda C. Gobe
- Centre for Kidney Disease Research, Translational Research Institute and The University of Queensland Diamantina Institute
| |
Collapse
|
36
|
Chuah JKC, Zink D. Stem cell-derived kidney cells and organoids: Recent breakthroughs and emerging applications. Biotechnol Adv 2016; 35:150-167. [PMID: 28017905 DOI: 10.1016/j.biotechadv.2016.12.001] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2016] [Revised: 12/12/2016] [Accepted: 12/17/2016] [Indexed: 02/09/2023]
Abstract
The global rise in the numbers of kidney patients and the shortage in transplantable organs have led to an increasing interest in kidney-specific regenerative therapies, renal disease modelling and bioartificial kidneys. Sources for large quantities of high-quality renal cells and tissues would be required, also for applications in in vitro platforms for compound safety and efficacy screening. Stem cell-based approaches for the generation of renal-like cells and tissues would be most attractive, but such methods were not available until recently. This situation has drastically changed since 2013, and various protocols for the generation of renal-like cells and precursors from pluripotent stem cells (PSC) have been established. The most recent breakthroughs were related to the establishment of various protocols for the generation of PSC-derived kidney organoids. In combination with recent advances in genome editing, bioprinting and the establishment of predictive renal screening platforms this results in exciting new possibilities. This review will give a comprehensive overview over current PSC-based protocols for the generation of renal-like cells, precursors and organoids, and their current and potential applications in regenerative medicine, compound screening, disease modelling and bioartificial organs.
Collapse
Affiliation(s)
- Jacqueline Kai Chin Chuah
- Institute of Bioengineering and Nanotechnology, Agency for Science, Technology and Research (A*STAR), 31 Biopolis Way, The Nanos, Singapore 138669, Singapore.
| | - Daniele Zink
- Institute of Bioengineering and Nanotechnology, Agency for Science, Technology and Research (A*STAR), 31 Biopolis Way, The Nanos, Singapore 138669, Singapore.
| |
Collapse
|
37
|
Ramm S, Adler M, Vaidya VS. A High-Throughput Screening Assay to Identify Kidney Toxic Compounds. ACTA ACUST UNITED AC 2016; 69:9.10.1-9.10.26. [PMID: 27479365 DOI: 10.1002/cptx.12] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Kidney toxicity due to drugs and chemicals poses a significant health burden for patients and a financial risk for pharmaceutical companies. However, currently no sensitive and high-throughput in vitro method exists for predictive nephrotoxicity assessment. Primary human proximal tubular epithelial cells (HPTECs) possess characteristics of differentiated epithelial cells, making them a desirable model to use in in vitro screening systems. Additionally, heme oxygenase 1 (HO-1) protein expression is upregulated as a protective mechanism during kidney toxicant-induced oxidative stress or inflammation in HPTECs and can therefore be used as a biomarker for nephrotoxicity. In this article, we describe two different methods to screen for HO-1 increase: A homogeneous time resolved fluorescence (HTRF) assay and an immunofluorescence assay. The latter provides lower throughput but higher sensitivity due to the combination of two readouts, HO-1 intensity and cell number. The methods described in the protocol are amendable for other cell types as well. © 2016 by John Wiley & Sons, Inc.
Collapse
Affiliation(s)
- Susanne Ramm
- Laboratory of Systems Pharmacology, Harvard Program in Therapeutic Sciences, Harvard Medical School, Boston, Massachusetts.,Renal Division, Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts
| | - Melanie Adler
- Laboratory of Systems Pharmacology, Harvard Program in Therapeutic Sciences, Harvard Medical School, Boston, Massachusetts.,Renal Division, Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts
| | - Vishal S Vaidya
- Laboratory of Systems Pharmacology, Harvard Program in Therapeutic Sciences, Harvard Medical School, Boston, Massachusetts.,Renal Division, Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts.,Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, Massachusetts
| |
Collapse
|
38
|
Li Y, Deng H, Ju L, Zhang X, Zhang Z, Yang Z, Wang L, Hou Z, Zhang Y. Screening and validation for plasma biomarkers of nephrotoxicity based on metabolomics in male rats. Toxicol Res (Camb) 2016; 5:259-267. [PMID: 30090342 PMCID: PMC6062367 DOI: 10.1039/c5tx00171d] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2015] [Accepted: 11/02/2015] [Indexed: 12/16/2022] Open
Abstract
Currently, drug-induced nephrotoxicity is widespread and seriously affects human health. However, the conventional indexes of renal function lack sensitivity, leading to a delay in the detection of nephrotoxicity. Therefore, we need to identify more sensitive indexes for evaluating nephrotoxicity. In this study, we used gentamicin (100 mg kg-1), etimicin (100 mg kg-1) and amphotericin B (4 mg kg-1) to establish renal injury models in rats, and we collected information using ultra-performance liquid chromatography quadrupole time-of-flight mass spectrometry in the screening stage. Thirteen nephrotoxicity metabolites were selected after multivariate statistical and integration analyses. Then, we conducted trend analysis to select 5 nephrotoxicity biomarkers [thymidine, LysoPC(16:1), LysoPC(18:4), LysoPC(20:5), and LysoPC(22:5)] whose content changed consistently at different timepoints after drug administration. To verify the sensitivity and specificity of these biomarkers for nephrotoxicity, receiver operating characteristic (ROC) and support vector machine (SVM) analyses were applied. The area under the curve of the 5 biomarkers were 0.806-0.901 at the 95% confidence interval according to the ROC analysis. We used the SVM classified model to verify these biomarkers, and the prediction rate was 95.83%. Therefore, the 5 biomarkers have strong sensitivity and high accuracy; these biomarkers are more sensitive indexes for evaluating renal function to identify nephrotoxicity and initiate prompt treatment.
Collapse
Affiliation(s)
- Yubo Li
- Tianjin State Key Laboratory of Modern Chinese Medicine , School of Traditional Chinese Materia Medica , Tianjin University of Traditional Chinese Medicine , 312 Anshan west Road , Tianjin 300193 , China
| | - Haoyue Deng
- Tianjin State Key Laboratory of Modern Chinese Medicine , School of Traditional Chinese Materia Medica , Tianjin University of Traditional Chinese Medicine , 312 Anshan west Road , Tianjin 300193 , China
| | - Liang Ju
- Tianjin State Key Laboratory of Modern Chinese Medicine , School of Traditional Chinese Materia Medica , Tianjin University of Traditional Chinese Medicine , 312 Anshan west Road , Tianjin 300193 , China
| | - Xiuxiu Zhang
- Tianjin State Key Laboratory of Modern Chinese Medicine , School of Traditional Chinese Materia Medica , Tianjin University of Traditional Chinese Medicine , 312 Anshan west Road , Tianjin 300193 , China
| | - Zhenzhu Zhang
- Tianjin State Key Laboratory of Modern Chinese Medicine , School of Traditional Chinese Materia Medica , Tianjin University of Traditional Chinese Medicine , 312 Anshan west Road , Tianjin 300193 , China
| | - Zhen Yang
- Tianjin State Key Laboratory of Modern Chinese Medicine , School of Traditional Chinese Materia Medica , Tianjin University of Traditional Chinese Medicine , 312 Anshan west Road , Tianjin 300193 , China
| | - Lei Wang
- Tianjin State Key Laboratory of Modern Chinese Medicine , School of Traditional Chinese Materia Medica , Tianjin University of Traditional Chinese Medicine , 312 Anshan west Road , Tianjin 300193 , China
| | - Zhiguo Hou
- Tianjin State Key Laboratory of Modern Chinese Medicine , School of Traditional Chinese Materia Medica , Tianjin University of Traditional Chinese Medicine , 312 Anshan west Road , Tianjin 300193 , China
| | - Yanjun Zhang
- Tianjin State Key Laboratory of Modern Chinese Medicine , Tianjin University of Traditional Chinese Medicine , 312 Anshan west Road , Tianjin 300193 , China . ; ; Tel: +86-22-59596221
| |
Collapse
|
39
|
High-throughput imaging-based nephrotoxicity prediction for xenobiotics with diverse chemical structures. Arch Toxicol 2015; 90:2793-2808. [PMID: 26612367 PMCID: PMC5065616 DOI: 10.1007/s00204-015-1638-y] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2015] [Accepted: 11/09/2015] [Indexed: 02/06/2023]
Abstract
The kidney is a major target for xenobiotics, which include drugs, industrial chemicals, environmental toxicants and other compounds. Accurate methods for screening large numbers of potentially nephrotoxic xenobiotics with diverse chemical structures are currently not available. Here, we describe an approach for nephrotoxicity prediction that combines high-throughput imaging of cultured human renal proximal tubular cells (PTCs), quantitative phenotypic profiling, and machine learning methods. We automatically quantified 129 image-based phenotypic features, and identified chromatin and cytoskeletal features that can predict the human in vivo PTC toxicity of 44 reference compounds with ~82 % (primary PTCs) or 89 % (immortalized PTCs) test balanced accuracies. Surprisingly, our results also revealed that a DNA damage response is commonly induced by different PTC toxicants that have diverse chemical structures and injury mechanisms. Together, our results show that human nephrotoxicity can be predicted with high efficiency and accuracy by combining cell-based and computational methods that are suitable for automation.
Collapse
|
40
|
Mollet BB, Bogaerts ILJ, van Almen GC, Dankers PYW. A bioartificial environment for kidney epithelial cells based on a supramolecular polymer basement membrane mimic and an organotypical culture system. J Tissue Eng Regen Med 2015; 11:1820-1834. [PMID: 28586546 DOI: 10.1002/term.2080] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2014] [Revised: 05/18/2015] [Accepted: 06/23/2015] [Indexed: 01/27/2023]
Abstract
Renal applications in healthcare, such as renal replacement therapies and nephrotoxicity tests, could potentially benefit from bioartificial kidney membranes with fully differentiated and functional human tubular epithelial cells. A replacement of the natural environment of these cells is required to maintain and study cell functionality cell differentiation in vitro. Our approach was based on synthetic supramolecular biomaterials to mimic the natural basement membrane (BM) on which these cells grow and a bioreactor to provide the desired organotypical culture parameters. The BM mimics were constructed from ureidopyrimidinone (UPy)-functionalized polymer and bioactive peptides by electrospinning. The resultant membranes were shown to have a hierarchical fibrous BM-like structure consisting of self-assembled nanofibres within the electrospun microfibres. Human kidney-2 (HK-2) epithelial cells were cultured on the BM mimics under organotypical conditions in a custom-built bioreactor. The bioreactor facilitated in situ monitoring and functionality testing of the cultures. Cell viability and the integrity of the epithelial cell barrier were demonstrated inside the bioreactor by microscopy and transmembrane leakage of fluorescently labelled inulin, respectively. Furthermore, HK-2 cells maintained a polarized cell layer and showed modulation of both gene expression of membrane transporter proteins and metabolic activity of brush border enzymes when subjected to a continuous flow of culture medium inside the new bioreactor for 21 days. These results demonstrated that both the culture and study of renal epithelial cells was facilitated by the bioartificial in vitro environment that is formed by synthetic supramolecular BM mimics in our custom-built bioreactor. Copyright © 2015 John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Björne B Mollet
- Department of Biomedical Engineering, Laboratory of Chemical Biology, Eindhoven University of Technology, The Netherlands.,Institute for Complex Molecular Systems, Eindhoven University of Technology, The Netherlands
| | - Iven L J Bogaerts
- Department of Biomedical Engineering, Laboratory of Chemical Biology, Eindhoven University of Technology, The Netherlands.,Institute for Complex Molecular Systems, Eindhoven University of Technology, The Netherlands
| | - Geert C van Almen
- Department of Biomedical Engineering, Laboratory of Chemical Biology, Eindhoven University of Technology, The Netherlands.,Institute for Complex Molecular Systems, Eindhoven University of Technology, The Netherlands
| | - Patricia Y W Dankers
- Department of Biomedical Engineering, Laboratory of Chemical Biology, Eindhoven University of Technology, The Netherlands.,Institute for Complex Molecular Systems, Eindhoven University of Technology, The Netherlands
| |
Collapse
|
41
|
Adler M, Ramm S, Hafner M, Muhlich JL, Gottwald EM, Weber E, Jaklic A, Ajay AK, Svoboda D, Auerbach S, Kelly EJ, Himmelfarb J, Vaidya VS. A Quantitative Approach to Screen for Nephrotoxic Compounds In Vitro. J Am Soc Nephrol 2015; 27:1015-28. [PMID: 26260164 DOI: 10.1681/asn.2015010060] [Citation(s) in RCA: 84] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2015] [Accepted: 06/16/2015] [Indexed: 12/16/2022] Open
Abstract
Nephrotoxicity due to drugs and environmental chemicals accounts for significant patient mortality and morbidity, but there is no high throughput in vitro method for predictive nephrotoxicity assessment. We show that primary human proximal tubular epithelial cells (HPTECs) possess characteristics of differentiated epithelial cells rendering them desirable to use in such in vitro systems. To identify a reliable biomarker of nephrotoxicity, we conducted multiplexed gene expression profiling of HPTECs after exposure to six different concentrations of nine human nephrotoxicants. Only overexpression of the gene encoding heme oxygenase-1 (HO-1) significantly correlated with increasing dose for six of the compounds, and significant HO-1 protein deregulation was confirmed with each of the nine nephrotoxicants. Translatability of HO-1 increase across species and platforms was demonstrated by computationally mining two large rat toxicogenomic databases for kidney tubular toxicity and by observing a significant increase in HO-1 after toxicity using an ex vivo three-dimensional microphysiologic system (kidney-on-a-chip). The predictive potential of HO-1 was tested using an additional panel of 39 mechanistically distinct nephrotoxic compounds. Although HO-1 performed better (area under the curve receiver-operator characteristic curve [AUC-ROC]=0.89) than traditional endpoints of cell viability (AUC-ROC for ATP=0.78; AUC-ROC for cell count=0.88), the combination of HO-1 and cell count further improved the predictive ability (AUC-ROC=0.92). We also developed and optimized a homogenous time-resolved fluorescence assay to allow high throughput quantitative screening of nephrotoxic compounds using HO-1 as a sensitive biomarker. This cell-based approach may facilitate rapid assessment of potential nephrotoxic therapeutics and environmental chemicals.
Collapse
Affiliation(s)
- Melanie Adler
- Laboratory of Systems Pharmacology, Harvard Program in Therapeutic Sciences, Harvard Medical School, Boston, Massachusetts; Renal Division, Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts
| | - Susanne Ramm
- Laboratory of Systems Pharmacology, Harvard Program in Therapeutic Sciences, Harvard Medical School, Boston, Massachusetts
| | - Marc Hafner
- Laboratory of Systems Pharmacology, Harvard Program in Therapeutic Sciences, Harvard Medical School, Boston, Massachusetts
| | - Jeremy L Muhlich
- Laboratory of Systems Pharmacology, Harvard Program in Therapeutic Sciences, Harvard Medical School, Boston, Massachusetts
| | - Esther Maria Gottwald
- Laboratory of Systems Pharmacology, Harvard Program in Therapeutic Sciences, Harvard Medical School, Boston, Massachusetts
| | - Elijah Weber
- Department of Pharmaceutics, University of Washington, Seattle, Washington
| | - Alenka Jaklic
- Department of Pharmaceutics, University of Washington, Seattle, Washington
| | - Amrendra Kumar Ajay
- Renal Division, Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts
| | | | - Scott Auerbach
- National Toxicology Program, National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina
| | - Edward J Kelly
- Department of Pharmaceutics, University of Washington, Seattle, Washington
| | - Jonathan Himmelfarb
- Kidney Research Institute, Department of Medicine, University of Washington, Seattle, Washington; and
| | - Vishal S Vaidya
- Laboratory of Systems Pharmacology, Harvard Program in Therapeutic Sciences, Harvard Medical School, Boston, Massachusetts; Renal Division, Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts; Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, Massachusetts
| |
Collapse
|
42
|
Prediction of drug-induced nephrotoxicity and injury mechanisms with human induced pluripotent stem cell-derived cells and machine learning methods. Sci Rep 2015. [PMID: 26212763 PMCID: PMC4515747 DOI: 10.1038/srep12337] [Citation(s) in RCA: 68] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The renal proximal tubule is a main target for drug-induced toxicity. The prediction of proximal tubular toxicity during drug development remains difficult. Any in vitro methods based on induced pluripotent stem cell-derived renal cells had not been developed, so far. Here, we developed a rapid 1-step protocol for the differentiation of human induced pluripotent stem cells (hiPSC) into proximal tubular-like cells. These proximal tubular-like cells had a purity of >90% after 8 days of differentiation and could be directly applied for compound screening. The nephrotoxicity prediction performance of the cells was determined by evaluating their responses to 30 compounds. The results were automatically determined using a machine learning algorithm called random forest. In this way, proximal tubular toxicity in humans could be predicted with 99.8% training accuracy and 87.0% test accuracy. Further, we studied the underlying mechanisms of injury and drug-induced cellular pathways in these hiPSC-derived renal cells, and the results were in agreement with human and animal data. Our methods will enable the development of personalized or disease-specific hiPSC-based renal in vitro models for compound screening and nephrotoxicity prediction.
Collapse
|
43
|
Simon-Friedt BR, Wilson MJ, Blake DA, Yu H, Eriksson Y, Wickliffe JK. The RPTEC/TERT1 Cell Line as an Improved Tool for In Vitro Nephrotoxicity Assessments. Biol Trace Elem Res 2015; 166:66-71. [PMID: 25893367 PMCID: PMC4470802 DOI: 10.1007/s12011-015-0339-y] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2015] [Accepted: 04/08/2015] [Indexed: 12/15/2022]
Abstract
In earlier studies, we have characterized a newly developed cell line derived from the renal proximal tubule epithelial cells (RPTEC) of a healthy human male donor in order to provide an improved in vitro model with which to investigate human diseases, such as cancer, that may be promoted by toxicant exposure. The RPTEC/TERT1 cell line has been immortalized using the human telomerase reverse transcriptase (hTERT) catalytic subunit and does not exhibit chromosomal abnormalities (Evercyte Laboratories). We have previously conducted single-compound and binary mixture experiments with the common environmental carcinogens, cadmium (Cd), and benzo[a]pyrene (B[a]P). Cells exhibited cytotoxic and compound-specific responses to low concentrations of B[a]P and Cd. We detected responses after exposure consistent with what is known regarding these cells in a normal, healthy kidney including significant gene expression changes, BPDE-DNA adducts in the presence of B[a]P, and indications of oxidative stress in the presence of Cd. The RPTEC/TERT1 cell line was also amenable to co-exposure studies due to its sensitivity and compound-specific properties. Here, we review our earlier work, compare our findings with commonly used renal cell lines, and suggest directions for future experiments. We conclude that the RPTEC/TERT1 cell line can provide a useful tool for future toxicological and mixture studies.
Collapse
Affiliation(s)
- Bridget R. Simon-Friedt
- Graduate Program in Biomedical Sciences, Tulane University School of Medicine, New Orleans, LA 70112
- Department of Global Environmental Health Sciences, Tulane University School of Public Health and Tropical Medicine, New Orleans, LA 70112
| | - Mark J Wilson
- Department of Global Environmental Health Sciences, Tulane University School of Public Health and Tropical Medicine, New Orleans, LA 70112
| | - Diane A. Blake
- Graduate Program in Biomedical Sciences, Tulane University School of Medicine, New Orleans, LA 70112
- Department of Biochemistry and Molecular Biology, Tulane University School of Medicine, New Orleans, LA 70112
| | - Haini Yu
- Department of Biochemistry and Molecular Biology, Tulane University School of Medicine, New Orleans, LA 70112
| | - Yasmin Eriksson
- Emerging Scholars Environmental Health Sciences Academy, Tulane University, Department of Global Environmental Health Sciences, Tulane University School of Public Health and Tropical Medicine, New Orleans, LA, 70112
| | - Jeffrey K. Wickliffe
- Graduate Program in Biomedical Sciences, Tulane University School of Medicine, New Orleans, LA 70112
- Department of Global Environmental Health Sciences, Tulane University School of Public Health and Tropical Medicine, New Orleans, LA 70112
- Emerging Scholars Environmental Health Sciences Academy, Tulane University, Department of Global Environmental Health Sciences, Tulane University School of Public Health and Tropical Medicine, New Orleans, LA, 70112
| |
Collapse
|
44
|
Su R, Li Y, Zink D, Loo LH. Supervised prediction of drug-induced nephrotoxicity based on interleukin-6 and -8 expression levels. BMC Bioinformatics 2014; 15 Suppl 16:S16. [PMID: 25521947 PMCID: PMC4290648 DOI: 10.1186/1471-2105-15-s16-s16] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Background Drug-induced nephrotoxicity causes acute kidney injury and chronic kidney diseases, and is a major reason for late-stage failures in the clinical trials of new drugs. Therefore, early, pre-clinical prediction of nephrotoxicity could help to prioritize drug candidates for further evaluations, and increase the success rates of clinical trials. Recently, an in vitro model for predicting renal-proximal-tubular-cell (PTC) toxicity based on the expression levels of two inflammatory markers, interleukin (IL)-6 and -8, has been described. However, this and other existing models usually use linear and manually determined thresholds to predict nephrotoxicity. Automated machine learning algorithms may improve these models, and produce more accurate and unbiased predictions. Results Here, we report a systematic comparison of the performances of four supervised classifiers, namely random forest, support vector machine, k-nearest-neighbor and naive Bayes classifiers, in predicting PTC toxicity based on IL-6 and -8 expression levels. Using a dataset of human primary PTCs treated with 41 well-characterized compounds that are toxic or not toxic to PTC, we found that random forest classifiers have the highest cross-validated classification performance (mean balanced accuracy = 87.8%, sensitivity = 89.4%, and specificity = 85.9%). Furthermore, we also found that IL-8 is more predictive than IL-6, but a combination of both markers gives higher classification accuracy. Finally, we also show that random forest classifiers trained automatically on the whole dataset have higher mean balanced accuracy than a previous threshold-based classifier constructed for the same dataset (99.3% vs. 80.7%). Conclusions Our results suggest that a random forest classifier can be used to automatically predict drug-induced PTC toxicity based on the expression levels of IL-6 and -8.
Collapse
|
45
|
Huang JX, Blaskovich MA, Cooper MA. Cell- and biomarker-based assays for predicting nephrotoxicity. Expert Opin Drug Metab Toxicol 2014; 10:1621-35. [DOI: 10.1517/17425255.2014.967681] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
46
|
Umeh VN, Ilodigwe EE, Ajaghaku DL, Erhirhie EO, Moke GE, Akah PA. Wound-healing Activity of the Aqueous Leaf Extract and Fractions of Ficus exasperata (Moraceae) and its Safety Evaluation on Albino Rats. J Tradit Complement Med 2014; 4:246-52. [PMID: 25379466 PMCID: PMC4220502 DOI: 10.4103/2225-4110.139105] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
Ficus exasperata have been reported to have wide applications in the treatment of many human diseases. However, its traditional use in the treatment of wounds has not been validated by any scientific study. Also, its safety in the management of chronic disease conditions requires attention. We evaluated the wound-healing activity of the aqueous extract and fractions of F. exasperata, as well as its safety after subchronic oral administration. Similar percentage of wound contraction was observed with 5% w/w extract ointment application and administration of cicatrin powder (standard) on the 4(th) day, while better contraction than the standard was recorded with higher concentrations of the extract ointment. Of all the fractions tested, significant (P < 0.05) contraction was only noticed in chloroform fraction, though lower than that of the aqueous extract. The extract also showed concentration-dependent inhibition of all the tested microbial isolates. Extract administered up to 5000 mg/kg (single dose administration) did not cause any mortality after 24 h. Mortality was, however, recorded at 4000 mg/kg within the first 20 days of subchronic administration of the extract. Significant (P < 0.05) increases in alanine aminotransaminase (ALT), aspartate aminotransaminase (AST), and in particular, alkaline phosphatase (ALP) were observed at different doses and time periods. Pathological and histological changes were noticed in the liver and kidney on the 91(st) day of the study with 4000 mg/kg of the extract. Except for the significant (P < 0.05) reduction in WBC on the 91(st) day, no other significant (P < 0.05) changes were observed in other hematological parameters. The aqueous extract demonstrated better wound-healing activity than its fractions; however, the extract may not be safe at higher doses for subchronic oral administration, as may be the case in the management of chronic disease conditions.
Collapse
Affiliation(s)
- Victoria Nonyelum Umeh
- Department of Pharmacology and Toxicology, Nnamdi Azikiwe University, Awka, Anambra State, Nigeria
| | - Emmanuel Emeka Ilodigwe
- Department of Pharmacology and Toxicology, Nnamdi Azikiwe University, Awka, Anambra State, Nigeria
| | - Daniel Lotanna Ajaghaku
- Department of Pharmacology and Toxicology, Nnamdi Azikiwe University, Awka, Anambra State, Nigeria
| | - Earnest Oghenesuvwe Erhirhie
- Department of Pharmacology and Therapeutics, Faculty of Basic Medical Sciences, Delta State University, Abraka, Delta State, Nigeria
| | - Goodies Emuesiri Moke
- Department of Pharmacology and Therapeutics, Faculty of Basic Medical Sciences, Delta State University, Abraka, Delta State, Nigeria
| | - Peter Achunike Akah
- Department of Pharmacology and Toxicology, University of Nigeria, Nsukka, Nigeria
| |
Collapse
|
47
|
Tiong HY, Huang P, Xiong S, Li Y, Vathsala A, Zink D. Drug-induced nephrotoxicity: clinical impact and preclinical in vitro models. Mol Pharm 2014; 11:1933-48. [PMID: 24502545 DOI: 10.1021/mp400720w] [Citation(s) in RCA: 116] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The kidney is a major target for drug-induced toxicity. Drug-induced nephrotoxicity remains a major problem in the clinical setting, where the use of nephrotoxic drugs is often unavoidable. This leads frequently to acute kidney injury, and current problems are discussed. One strategy to avoid such problems would be the development of drugs with decreased nephrotoxic potential. However, the prediction of nephrotoxicity during preclinical drug development is difficult and nephrotoxicity is typically detected only late. Also, the nephrotoxic potential of newly approved drugs is often underestimated. Regulatory approved or validated in vitro models for the prediction of nephrotoxicity are currently not available. Here, we will review current approaches on the development of such models. This includes a discussion of three-dimensional and microfluidic models and recently developed stem cell based approaches. Most in vitro models have been tested with a limited number of compounds and are of unclear predictivity. However, some studies have tested larger numbers of compounds and the predictivity of the respective in vitro model had been determined. The results showed that high predictivity can be obtained by using primary or stem cell derived human renal cells in combination with appropriate end points.
Collapse
Affiliation(s)
- Ho Yee Tiong
- Yong Loo Lin School of Medicine, National University Health System , 1E Kent Ridge Road, NUHS Tower Block, Singapore 119228, Singapore
| | | | | | | | | | | |
Collapse
|
48
|
Li Y, Kandasamy K, Chuah JKC, Lam YN, Toh WS, Oo ZY, Zink D. Identification of Nephrotoxic Compounds with Embryonic Stem-Cell-Derived Human Renal Proximal Tubular-Like Cells. Mol Pharm 2014; 11:1982-90. [DOI: 10.1021/mp400637s] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Yao Li
- Institute of Bioengineering
and Nanotechnology, 31
Biopolis Way, The Nanos, Singapore 138669, Singapore
| | - Karthikeyan Kandasamy
- Institute of Bioengineering
and Nanotechnology, 31
Biopolis Way, The Nanos, Singapore 138669, Singapore
| | - Jacqueline Kai Chin Chuah
- Institute of Bioengineering
and Nanotechnology, 31
Biopolis Way, The Nanos, Singapore 138669, Singapore
| | - Yue Ning Lam
- Institute of Bioengineering
and Nanotechnology, 31
Biopolis Way, The Nanos, Singapore 138669, Singapore
| | - Wei Seong Toh
- Institute of Bioengineering
and Nanotechnology, 31
Biopolis Way, The Nanos, Singapore 138669, Singapore
| | - Zay Yar Oo
- Institute of Bioengineering
and Nanotechnology, 31
Biopolis Way, The Nanos, Singapore 138669, Singapore
| | - Daniele Zink
- Institute of Bioengineering
and Nanotechnology, 31
Biopolis Way, The Nanos, Singapore 138669, Singapore
| |
Collapse
|