1
|
Zhang Z, Zhang Y, Li Y, Jiang S, Xu F, Li K, Chang L, Gao H, Kukic P, Carmichael P, Liddell M, Li J, Zhang Q, Lyu Z, Peng S, Zuo T, Tulum L, Xu P. Quantitative phosphoproteomics reveal cellular responses from caffeine, coumarin and quercetin in treated HepG2 cells. Toxicol Appl Pharmacol 2022; 449:116110. [DOI: 10.1016/j.taap.2022.116110] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2022] [Revised: 05/30/2022] [Accepted: 06/02/2022] [Indexed: 11/15/2022]
|
2
|
Tan YM, Barton HA, Boobis A, Brunner R, Clewell H, Cope R, Dawson J, Domoradzki J, Egeghy P, Gulati P, Ingle B, Kleinstreuer N, Lowe K, Lowit A, Mendez E, Miller D, Minucci J, Nguyen J, Paini A, Perron M, Phillips K, Qian H, Ramanarayanan T, Sewell F, Villanueva P, Wambaugh J, Embry M. Opportunities and challenges related to saturation of toxicokinetic processes: Implications for risk assessment. Regul Toxicol Pharmacol 2021; 127:105070. [PMID: 34718074 DOI: 10.1016/j.yrtph.2021.105070] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Revised: 10/18/2021] [Accepted: 10/25/2021] [Indexed: 02/08/2023]
Abstract
Top dose selection for repeated dose animal studies has generally focused on identification of apical endpoints, use of the limit dose, or determination of a maximum tolerated dose (MTD). The intent is to optimize the ability of toxicity tests performed in a small number of animals to detect effects for hazard identification. An alternative approach, the kinetically derived maximum dose (KMD), has been proposed as a mechanism to integrate toxicokinetic (TK) data into the dose selection process. The approach refers to the dose above which the systemic exposures depart from being proportional to external doses. This non-linear external-internal dose relationship arises from saturation or limitation of TK process(es), such as absorption or metabolism. The importance of TK information is widely acknowledged when assessing human health risks arising from exposures to environmental chemicals, as TK determines the amount of chemical at potential sites of toxicological responses. However, there have been differing opinions and interpretations within the scientific and regulatory communities related to the validity and application of the KMD concept. A multi-stakeholder working group, led by the Health and Environmental Sciences Institute (HESI), was formed to provide an opportunity for impacted stakeholders to address commonly raised scientific and technical issues related to this topic and, more specifically, a weight of evidence approach is recommended to inform design and dose selection for repeated dose animal studies. Commonly raised challenges related to the use of TK data for dose selection are discussed, recommendations are provided, and illustrative case examples are provided to address these challenges or refute misconceptions.
Collapse
Affiliation(s)
- Yu-Mei Tan
- U.S. Environmental Protection Agency, Office of Pesticide Programs, Durham, NC, USA
| | | | | | - Rachel Brunner
- U.S. Environmental Protection Agency, Office of Pesticide Programs, Durham, NC, USA
| | | | - Rhian Cope
- Australian Pesticides and Veterinary Medicines Authority, Sydney, NSW, Australia
| | - Jeffrey Dawson
- U.S. Environmental Protection Agency, Office of Chemical Safety and Pollution Prevention, Washington, DC, USA
| | | | - Peter Egeghy
- U.S. Environmental Protection Agency, Office of Research & Development, Durham, NC, USA
| | - Pankaj Gulati
- Australian Pesticides and Veterinary Medicines Authority, Sydney, NSW, Australia
| | - Brandall Ingle
- U.S. Environmental Protection Agency, Office of Pesticide Programs, Durham, NC, USA
| | - Nicole Kleinstreuer
- National Toxicology Program, Interagency Center for the Evaluation of Alternative Toxicological Methods, Research Triangle Park, NC, USA
| | - Kelly Lowe
- U.S. Environmental Protection Agency, Office of Pesticide Programs, Washington, DC, USA
| | - Anna Lowit
- U.S. Environmental Protection Agency, Office of Pesticide Programs, Washington, DC, USA
| | - Elizabeth Mendez
- U.S. Environmental Protection Agency, Office of Pesticide Programs, Washington, DC, USA
| | - David Miller
- U.S. Environmental Protection Agency, Office of Pesticide Programs, Washington, DC, USA
| | - Jeffrey Minucci
- U.S. Environmental Protection Agency, Office of Research & Development, Durham, NC, USA
| | - James Nguyen
- U.S. Environmental Protection Agency, Office of Pesticide Programs, Washington, DC, USA
| | - Alicia Paini
- European Commission, Joint Research Centre, Ispra, Italy
| | - Monique Perron
- U.S. Environmental Protection Agency, Office of Pesticide Programs, Washington, DC, USA
| | - Katherine Phillips
- U.S. Environmental Protection Agency, Office of Research & Development, Durham, NC, USA
| | - Hua Qian
- ExxonMobil Biomedical Sciences, Inc., Annandale, NJ, USA
| | | | - Fiona Sewell
- National Centre for the Replacement, Refinement, and Reduction of Animals in Research, London, UK
| | - Philip Villanueva
- U.S. Environmental Protection Agency, Office of Pesticide Programs, Washington, DC, USA
| | - John Wambaugh
- U.S. Environmental Protection Agency, Office of Research & Development, Durham, NC, USA
| | - Michelle Embry
- Health and Environmental Sciences Institute, Washington DC, USA.
| |
Collapse
|
3
|
Efremenko A, Balbuena P, Clewell RA, Black M, Pluta L, Andersen ME, Gentry PR, Yager JW, Clewell HJ. Time-dependent genomic response in primary human uroepithelial cells exposed to arsenite for up to 60 days. Toxicology 2021; 461:152893. [PMID: 34425169 DOI: 10.1016/j.tox.2021.152893] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 07/02/2021] [Accepted: 08/05/2021] [Indexed: 11/16/2022]
Abstract
Evidence from both in vivo and in vitro studies suggests that gene expression changes from long-term exposure to arsenite evolve markedly over time, including reversals in the direction of expression change in key regulatory genes. In this study, human uroepithelial cells from the ureter segments of 4 kidney-donors were continuously treated in culture with arsenite at concentrations of 0.1 or 1 μM for 60 days. Gene expression at 10, 20, 30, 40, and 60 days was determined using Affymetrix human genome microarrays and signal pathway analysis was performed using GeneGo Metacore. Arsenic treated cells continued to proliferate for the full 60-day period, whereas untreated cells ceased proliferating after approximately 30 days. A peak in the number of gene changes in the treated cells compared to untreated controls was observed between 30 and 40 days of exposure, with substantially fewer changes at 10 and 60 days, suggesting remodeling of the cells over time. Consistent with this possibility, the direction of expression change for a number of key genes was reversed between 20 and 30 days, including CFOS and MDM2. While the progression of gene changes was different for each subject, a common pattern was observed in arsenic treated cells over time, with early upregulation of oxidative stress responses (HMOX1, NQ01, TXN, TXNRD1) and down-regulation of immune/inflammatory responses (IKKα). At around 30 days, there was a transition to increased inflammatory and proliferative signaling (AKT, CFOS), evidence of epithelial-to-mesenchymal transition (EMT), and alterations in DNA damage responses (MDM2, ATM). A common element in the changing response of cells to arsenite over time appears to involve up-regulation of MDM2 by inflammatory signaling (through AP-1 and NF-κB), leading to inhibition of P53 function.
Collapse
Affiliation(s)
- Alina Efremenko
- The Hamner Institutes for Health Sciences, RTP, NC, United States
| | | | | | - Michael Black
- The Hamner Institutes for Health Sciences, RTP, NC, United States
| | - Linda Pluta
- The Hamner Institutes for Health Sciences, RTP, NC, United States
| | | | | | - Janice W Yager
- Ramboll US Corporation, Emeryville, CA, United States(1)
| | - Harvey J Clewell
- The Hamner Institutes for Health Sciences, RTP, NC, United States.
| |
Collapse
|
4
|
Influence of Kv11.1 (hERG1) K + channel expression on DNA damage induced by the genotoxic agent methyl methanesulfonate. Pflugers Arch 2021; 473:197-217. [PMID: 33452554 DOI: 10.1007/s00424-021-02517-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Revised: 12/22/2020] [Accepted: 01/05/2021] [Indexed: 10/22/2022]
Abstract
Besides their crucial role in cell electrogenesis and maintenance of basal membrane potential, the voltage-dependent K+ channel Kv11.1/hERG1 shows an essential impact in cell proliferation and other processes linked to the maintenance of tumour phenotype. To check the possible influence of channel expression on DNA damage responses, HEK293 cells, treated with the genotoxic agent methyl methanesulfonate (MMS), were compared with those of a HEK-derived cell line (H36), permanently transfected with the Kv11.1-encoding gene, and with a third cell line (T2) obtained under identical conditions as H36, by permanent transfection of another unrelated plasma membrane protein encoding gene. In addition, to gain some insights about the canonical/conduction-dependent channel mechanisms that might be involved, the specific erg channel inhibitor E4031 was used as a tool. Our results indicate that the expression of Kv11.1 does not influence MMS-induced changes in cell cycle progression, because no differences were found between H36 and T2 cells. However, the canonical ion conduction function of the channel appeared to be associated with decreased cell viability at low/medium MMS concentrations. Moreover, direct DNA damage measurements, using the comet assay, demonstrated for the first time that Kv11.1 conduction activity was able to modify MMS-induced DNA damage, decreasing it particularly at high MMS concentration, in a way related to PARP1 gene expression. Finally, our data suggest that the canonical Kv11.1 effects may be relevant for tumour cell responses to anti-tumour therapies.
Collapse
|
5
|
Pink M, Verma N, Schmitz-Spanke S. Benchmark dose analyses of toxic endpoints in lung cells provide sensitivity and toxicity ranking across metal oxide nanoparticles and give insights into the mode of action. Toxicol Lett 2020; 331:218-226. [PMID: 32562635 DOI: 10.1016/j.toxlet.2020.06.012] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2019] [Revised: 06/10/2020] [Accepted: 06/16/2020] [Indexed: 10/24/2022]
Abstract
INTRODUCTION The benchmark dose (BMD) is a dose that produces a predetermined change in the response rate of an adverse effect. This approach is increasingly utilized to analyze quantitative dose-response relationships. To proof this concept, statistical analysis was compared with the BMD approach in order to rank the sensitivity as well as the toxicity and to describe the mode of action. METHODS Bronchial (BEAS-2B) and alveolar epithelial cells (A549) were exposed to a wide concentration range (0.4-100 μg/mL) of five metal oxide nanoparticles (CeO2, CuO, TiO2, ZnO, ZrO2). Eight toxicity endpoints were determined representing integrity of lysosomal and cell membrane, oxidative stress level, glutathione based detoxification (glutathione S-transferase), oxidative metabolism (cytochrome P450), alteration of the mitochondrial membrane potential, alteration of phase II antioxidative enzyme (NAD(P)H:quinone oxidoreductase), and de novo DNA synthesis. RESULTS Based on the BMD calculated for the most sensitive test, the toxicity decreased in the following order: ZnO > CuO > TiO2>ZrO2>CeO2 in BEAS-2B. Both statistical evaluation methods revealed a higher sensitivity of BEAS-2B cells. The BMD-derived mode of action for CuO confirmed the existing hypotheses and provided insights into less known mechanisms. CONCLUSION The findings proofed that BMD analysis is an effective tool to evaluate different aspects of risk assessment.
Collapse
Affiliation(s)
- Mario Pink
- Institute and Outpatient Clinic of Occupational, Social and Environmental Medicine, University of Erlangen-Nuremberg, Henkestr. 9-11, 91054 Erlangen, Germany; Department of Chemical and Product Safety, German Federal Institute for Risk Assessment (BfR), Max-Dohrn-Str. 8-10, 10589 Berlin, Germany.
| | - Nisha Verma
- Institute and Outpatient Clinic of Occupational, Social and Environmental Medicine, University of Erlangen-Nuremberg, Henkestr. 9-11, 91054 Erlangen, Germany.
| | - Simone Schmitz-Spanke
- Institute and Outpatient Clinic of Occupational, Social and Environmental Medicine, University of Erlangen-Nuremberg, Henkestr. 9-11, 91054 Erlangen, Germany.
| |
Collapse
|
6
|
Hatherell S, Baltazar MT, Reynolds J, Carmichael PL, Dent M, Li H, Ryder S, White A, Walker P, Middleton AM. Identifying and Characterizing Stress Pathways of Concern for Consumer Safety in Next-Generation Risk Assessment. Toxicol Sci 2020; 176:11-33. [PMID: 32374857 PMCID: PMC7357173 DOI: 10.1093/toxsci/kfaa054] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Many substances for which consumer safety risk assessments need to be conducted are not associated with specific toxicity modes of action, but rather exhibit nonspecific toxicity leading to cell stress. In this work, a cellular stress panel is described, consisting of 36 biomarkers representing mitochondrial toxicity, cell stress, and cell health, measured predominantly using high content imaging. To evaluate the panel, data were generated for 13 substances at exposures consistent with typical use-case scenarios. These included some that have been shown to cause adverse effects in a proportion of exposed humans and have a toxicological mode-of-action associated with cellular stress (eg, doxorubicin, troglitazone, and diclofenac), and some that are not associated with adverse effects due to cellular stress at human-relevant exposures (eg, caffeine, niacinamide, and phenoxyethanol). For each substance, concentration response data were generated for each biomarker at 3 timepoints. A Bayesian model was then developed to quantify the evidence for a biological response, and if present, a credibility range for the estimated point of departure (PoD) was determined. PoDs were compared with the plasma Cmax associated with the typical substance exposures, and indicated a clear differentiation between "low" risk and "high" risk chemical exposure scenarios. Developing robust methods to characterize the in vitro bioactivity of xenobiotics is an important part of non-animal safety assessment. The results presented in this work show that the cellular stress panel can be used, together with other new approach methodologies, to identify chemical exposures that are protective of consumer health.
Collapse
Affiliation(s)
- Sarah Hatherell
- Unilever Safety and Environmental Assurance Centre, Colworth Science Park, Sharnbrook, Bedfordshire MK44 1LQ, UK
| | - Maria T Baltazar
- Unilever Safety and Environmental Assurance Centre, Colworth Science Park, Sharnbrook, Bedfordshire MK44 1LQ, UK
| | - Joe Reynolds
- Unilever Safety and Environmental Assurance Centre, Colworth Science Park, Sharnbrook, Bedfordshire MK44 1LQ, UK
| | - Paul L Carmichael
- Unilever Safety and Environmental Assurance Centre, Colworth Science Park, Sharnbrook, Bedfordshire MK44 1LQ, UK
| | - Matthew Dent
- Unilever Safety and Environmental Assurance Centre, Colworth Science Park, Sharnbrook, Bedfordshire MK44 1LQ, UK
| | - Hequn Li
- Unilever Safety and Environmental Assurance Centre, Colworth Science Park, Sharnbrook, Bedfordshire MK44 1LQ, UK
| | | | - Andrew White
- Unilever Safety and Environmental Assurance Centre, Colworth Science Park, Sharnbrook, Bedfordshire MK44 1LQ, UK
| | - Paul Walker
- Cyprotex Discovery Ltd, Macclesfield, Cheshire SK10 4TG, UK
| | - Alistair M Middleton
- Unilever Safety and Environmental Assurance Centre, Colworth Science Park, Sharnbrook, Bedfordshire MK44 1LQ, UK
| |
Collapse
|
7
|
Lin N, Zhou X, Geng X, Drewell C, Hübner J, Li Z, Zhang Y, Xue M, Marx U, Li B. Repeated dose multi-drug testing using a microfluidic chip-based coculture of human liver and kidney proximal tubules equivalents. Sci Rep 2020; 10:8879. [PMID: 32483208 PMCID: PMC7264205 DOI: 10.1038/s41598-020-65817-0] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2019] [Accepted: 04/15/2020] [Indexed: 11/28/2022] Open
Abstract
A microfluidic multi-organ chip emulates the tissue culture microenvironment, enables interconnection of organ equivalents and overcomes interspecies differences, making this technology a promising and powerful tool for preclinical drug screening. In this study, we established a microfluidic chip-based model that enabled non-contact cocultivation of liver spheroids and renal proximal tubule barriers in a connecting media circuit over 16 days. Meanwhile, a 14-day repeated-dose systemic administration of cyclosporine A (CsA) alone or in combination with rifampicin was performed. Toxicity profiles of the two different doses of CsA on different target organs could be discriminated and that concomitant treatment with rifampicin from day6 onwards decreased the CsA concentration and attenuated the toxicity compared with that after treatment with CsA for 14 consecutive days. The latter is manifested with the changes in cytotoxicity, cell viability and apoptosis, gene expression of metabolic enzymes and transporters, and noninvasive toxicity biomarkers. The on chip coculture of the liver and the proximal tubulus equivalents showed its potential as an effective and translational tool for repeated dose multi-drug toxicity screening in the preclinical stage of drug development.
Collapse
Affiliation(s)
- Ni Lin
- Key Laboratory of Beijing for Safety Evaluation of Drugs, National Center for Safety Evaluation of Drugs, National Institutes for Food and Drug Control, A8 Hongda Middle Street, Beijing Economic-Technological Development Area, Beijing, 100176, P. R. China.,Department of Pharmacology, Beijing Laboratory for Biomedical Detection Technology and Instrument, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China.,Beijing Institute for Drug Control, 25 Science Park Road, Changping District, Beijing, 102206, China
| | - Xiaobing Zhou
- Key Laboratory of Beijing for Safety Evaluation of Drugs, National Center for Safety Evaluation of Drugs, National Institutes for Food and Drug Control, A8 Hongda Middle Street, Beijing Economic-Technological Development Area, Beijing, 100176, P. R. China
| | - Xingchao Geng
- Key Laboratory of Beijing for Safety Evaluation of Drugs, National Center for Safety Evaluation of Drugs, National Institutes for Food and Drug Control, A8 Hongda Middle Street, Beijing Economic-Technological Development Area, Beijing, 100176, P. R. China
| | - Christopher Drewell
- Technische Universitaet Berlin, Institute of Biotechnology, Department Medical Biotechnology, Gustav-Meyer-Allee 25, 13355, Berlin, Germany
| | - Juliane Hübner
- Technische Universitaet Berlin, Institute of Biotechnology, Department Medical Biotechnology, Gustav-Meyer-Allee 25, 13355, Berlin, Germany
| | - Zuogang Li
- Key Laboratory of Beijing for Safety Evaluation of Drugs, National Center for Safety Evaluation of Drugs, National Institutes for Food and Drug Control, A8 Hongda Middle Street, Beijing Economic-Technological Development Area, Beijing, 100176, P. R. China
| | - Yingli Zhang
- Key Laboratory of Beijing for Safety Evaluation of Drugs, National Center for Safety Evaluation of Drugs, National Institutes for Food and Drug Control, A8 Hongda Middle Street, Beijing Economic-Technological Development Area, Beijing, 100176, P. R. China
| | - Ming Xue
- Department of Pharmacology, Beijing Laboratory for Biomedical Detection Technology and Instrument, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China.
| | - Uwe Marx
- TissUse GmbH, Oudenarder Strasse 16, 13347, Berlin, Germany.
| | - Bo Li
- National Institutes for Food and Drug Control, 31 Hua Tuo road, Daxing district, Beijing, 102629, China.
| |
Collapse
|
8
|
Klapacz J, Gollapudi BB. Considerations for the Use of Mutation as a Regulatory Endpoint in Risk Assessment. ENVIRONMENTAL AND MOLECULAR MUTAGENESIS 2020; 61:84-93. [PMID: 31301246 DOI: 10.1002/em.22318] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/30/2019] [Revised: 07/08/2019] [Accepted: 07/10/2019] [Indexed: 06/10/2023]
Abstract
Assessment of a chemical's potential to cause permanent changes in the genetic code has been a common practice in the industry and regulatory settings for decades. Furthermore, the genetic toxicity battery of tests has typically been employed during the earliest stages of the research and development programs of new product development. A positive outcome from such battery has a major impact on the chemical's utility, industrial hygiene, product stewardship practices, and product life cycle analysis, among many other decisions that need to be taken by the industry, even before the registration of a chemical is undertaken. Under the prevailing regulatory paradigm, the dichotomous (yes/no) evaluation of the chemical's genotoxic potential leads to a conservative, linear no-threshold (LNT) risk assessment, unless compelling and undeniable data to the contrary can be provided to satisfy regulators, typically in a number of different global jurisdictions. With the current advent of predictive methods, new testing paradigms, mode-of-action/adverse outcome pathways, and quantitative risk assessment approaches, various stakeholders are starting to employ these state-of-the-science methodologies to further the conversation on decision making and advance the regulatory paradigm beyond the dominant LNT status quo. This commentary describes these novel methodologies, relevant biological responses, and how these can affect internal and regulatory risk assessment approaches. Environ. Mol. Mutagen. 61:84-93, 2020. © 2019 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Joanna Klapacz
- Toxicology and Environmental Research and Consulting, The Dow Chemical Company, Midland, Michigan
| | | |
Collapse
|
9
|
Krewski D, Andersen ME, Tyshenko MG, Krishnan K, Hartung T, Boekelheide K, Wambaugh JF, Jones D, Whelan M, Thomas R, Yauk C, Barton-Maclaren T, Cote I. Toxicity testing in the 21st century: progress in the past decade and future perspectives. Arch Toxicol 2019; 94:1-58. [DOI: 10.1007/s00204-019-02613-4] [Citation(s) in RCA: 124] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2019] [Accepted: 11/05/2019] [Indexed: 12/19/2022]
|
10
|
Hasselgren C, Ahlberg E, Akahori Y, Amberg A, Anger LT, Atienzar F, Auerbach S, Beilke L, Bellion P, Benigni R, Bercu J, Booth ED, Bower D, Brigo A, Cammerer Z, Cronin MTD, Crooks I, Cross KP, Custer L, Dobo K, Doktorova T, Faulkner D, Ford KA, Fortin MC, Frericks M, Gad-McDonald SE, Gellatly N, Gerets H, Gervais V, Glowienke S, Van Gompel J, Harvey JS, Hillegass J, Honma M, Hsieh JH, Hsu CW, Barton-Maclaren TS, Johnson C, Jolly R, Jones D, Kemper R, Kenyon MO, Kruhlak NL, Kulkarni SA, Kümmerer K, Leavitt P, Masten S, Miller S, Moudgal C, Muster W, Paulino A, Lo Piparo E, Powley M, Quigley DP, Reddy MV, Richarz AN, Schilter B, Snyder RD, Stavitskaya L, Stidl R, Szabo DT, Teasdale A, Tice RR, Trejo-Martin A, Vuorinen A, Wall BA, Watts P, White AT, Wichard J, Witt KL, Woolley A, Woolley D, Zwickl C, Myatt GJ. Genetic toxicology in silico protocol. Regul Toxicol Pharmacol 2019; 107:104403. [PMID: 31195068 PMCID: PMC7485926 DOI: 10.1016/j.yrtph.2019.104403] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Revised: 05/20/2019] [Accepted: 06/05/2019] [Indexed: 01/23/2023]
Abstract
In silico toxicology (IST) approaches to rapidly assess chemical hazard, and usage of such methods is increasing in all applications but especially for regulatory submissions, such as for assessing chemicals under REACH as well as the ICH M7 guideline for drug impurities. There are a number of obstacles to performing an IST assessment, including uncertainty in how such an assessment and associated expert review should be performed or what is fit for purpose, as well as a lack of confidence that the results will be accepted by colleagues, collaborators and regulatory authorities. To address this, a project to develop a series of IST protocols for different hazard endpoints has been initiated and this paper describes the genetic toxicity in silico (GIST) protocol. The protocol outlines a hazard assessment framework including key effects/mechanisms and their relationships to endpoints such as gene mutation and clastogenicity. IST models and data are reviewed that support the assessment of these effects/mechanisms along with defined approaches for combining the information and evaluating the confidence in the assessment. This protocol has been developed through a consortium of toxicologists, computational scientists, and regulatory scientists across several industries to support the implementation and acceptance of in silico approaches.
Collapse
Affiliation(s)
| | - Ernst Ahlberg
- Predictive Compound ADME & Safety, Drug Safety & Metabolism, AstraZeneca IMED Biotech Unit, Mölndal, Sweden
| | - Yumi Akahori
- Chemicals Evaluation and Research Institute, 1-4-25 Kouraku, Bunkyo-ku, Tokyo, 112-0004, Japan
| | - Alexander Amberg
- Sanofi, R&D Preclinical Safety Frankfurt, Industriepark Hoechst, D-65926, Frankfurt am Main, Germany
| | - Lennart T Anger
- Sanofi, R&D Preclinical Safety Frankfurt, Industriepark Hoechst, D-65926, Frankfurt am Main, Germany
| | - Franck Atienzar
- UCB BioPharma SPRL, Chemin du Foriest, B-1420 Braine-l'Alleud, Belgium
| | - Scott Auerbach
- The National Institute of Environmental Health Sciences, Division of the National Toxicology Program, Research Triangle Park, NC, 27709, USA
| | - Lisa Beilke
- Toxicology Solutions Inc., San Diego, CA, USA
| | | | | | - Joel Bercu
- Gilead Sciences, 333 Lakeside Drive, Foster City, CA, USA
| | - Ewan D Booth
- Syngenta, Product Safety Department, Jealott's Hill International Research Centre, Bracknell, Berkshire, RG42 6EY, UK
| | - Dave Bower
- Leadscope, Inc, 1393 Dublin Rd, Columbus, OH, 43215, USA
| | - Alessandro Brigo
- Roche Pharmaceutical Research & Early Development, Pharmaceutical Sciences, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Grenzacherstrasse 124, 4070, Basel, Switzerland
| | - Zoryana Cammerer
- Janssen Research & Development, 1400 McKean Road, Spring House, PA, 19477, USA
| | - Mark T D Cronin
- School of Pharmacy and Biomolecular Sciences, Liverpool John Moores University, Liverpool, L3 3AF, UK
| | - Ian Crooks
- British American Tobacco, Research and Development, Regents Park Road, Southampton, Hampshire, SO15 8TL, UK
| | - Kevin P Cross
- Leadscope, Inc, 1393 Dublin Rd, Columbus, OH, 43215, USA
| | - Laura Custer
- Bristol-Myers Squibb, Drug Safety Evaluation, 1 Squibb Dr, New Brunswick, NJ, 08903, USA
| | - Krista Dobo
- Pfizer Global Research & Development, 558 Eastern Point Road, Groton, CT, 06340, USA
| | - Tatyana Doktorova
- Douglas Connect GmbH, Technology Park Basel, Hochbergerstrasse 60C, CH-4057, Basel / Basel-Stadt, Switzerland
| | - David Faulkner
- Lawrence Berkeley National Laboratory, One Cyclotron Road, MS 70A-1161A, Berkeley, CA, 947020, USA
| | - Kevin A Ford
- Global Blood Therapeutics, 171 Oyster Point Boulevard, South San Francisco, CA, 94080, USA
| | - Marie C Fortin
- Jazz Pharmaceuticals, Inc., 200 Princeton South Corporate Center, Suite 180, Ewing, NJ, 08628, USA; Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy, Rutgers University, 170 Frelinghuysen Rd, Piscataway, NJ, 08855, USA
| | | | | | - Nichola Gellatly
- National Centre for the Replacement, Refinement and Reduction of Animals in Research (NC3Rs), Gibbs Building, 215 Euston Road, London, NW1 2BE, UK
| | - Helga Gerets
- UCB BioPharma SPRL, Chemin du Foriest, B-1420, Braine-l'Alleud, Belgium
| | | | - Susanne Glowienke
- Novartis Pharma AG, Pre-Clinical Safety, Werk Klybeck, CH, 4057, Basel, Switzerland
| | - Jacky Van Gompel
- Janssen Pharmaceutical Companies of Johnson & Johnson, 2340, Beerse, Belgium
| | - James S Harvey
- GlaxoSmithKline Pre-Clinical Development, Park Road, Ware, Hertfordshire, SG12 0DP, UK
| | - Jedd Hillegass
- Bristol-Myers Squibb, Drug Safety Evaluation, 1 Squibb Dr, New Brunswick, NJ, 08903, USA
| | - Masamitsu Honma
- Division of Genetics and Mutagenesis, National Institute of Health Sciences, Kanagawa, 210-9501, Japan
| | - Jui-Hua Hsieh
- Kelly Government Solutions, Research Triangle Park, NC, 27709, USA
| | - Chia-Wen Hsu
- FDA Center for Drug Evaluation and Research, Silver Spring, MD, USA
| | | | | | - Robert Jolly
- Toxicology Division, Eli Lilly and Company, Indianapolis, IN, USA
| | - David Jones
- Medicines and Healthcare Products Regulatory Agency, 10 South Colonnade, Canary Wharf, London, E14 4PU, UK
| | - Ray Kemper
- Vertex Pharmaceuticals Inc., Predictive and Investigative Safety Assessment, 50 Northern Ave, Boston, MA, USA
| | - Michelle O Kenyon
- Pfizer Global Research & Development, 558 Eastern Point Road, Groton, CT, 06340, USA
| | - Naomi L Kruhlak
- FDA Center for Drug Evaluation and Research, Silver Spring, MD, USA
| | - Sunil A Kulkarni
- Existing Substances Risk Assessment Bureau, Health Canada, Ottawa, ON, K1A 0K9, Canada
| | - Klaus Kümmerer
- Institute for Sustainable and Environmental Chemistry, Leuphana University Lüneburg, Scharnhorststraße 1/C13.311b, 21335, Lüneburg, Germany
| | - Penny Leavitt
- Bristol-Myers Squibb, Drug Safety Evaluation, 1 Squibb Dr, New Brunswick, NJ, 08903, USA
| | - Scott Masten
- The National Institute of Environmental Health Sciences, Division of the National Toxicology Program, Research Triangle Park, NC, 27709, USA
| | - Scott Miller
- Leadscope, Inc, 1393 Dublin Rd, Columbus, OH, 43215, USA
| | | | - Wolfgang Muster
- Roche Pharmaceutical Research & Early Development, Pharmaceutical Sciences, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Grenzacherstrasse 124, 4070, Basel, Switzerland
| | | | | | - Mark Powley
- Merck Research Laboratories, West Point, PA, 19486, USA
| | | | | | | | | | - Ronald D Snyder
- RDS Consulting Services, 2936 Wooded Vista Ct, Mason, OH, 45040, USA
| | | | | | | | | | | | | | | | - Brian A Wall
- Colgate-Palmolive Company, Piscataway, NJ, 08854, USA
| | - Pete Watts
- Bibra, Cantium House, Railway Approach, Wallington, Surrey, SM6 0DZ, UK
| | - Angela T White
- GlaxoSmithKline Pre-Clinical Development, Park Road, Ware, Hertfordshire, SG12 0DP, UK
| | - Joerg Wichard
- Bayer AG, Pharmaceuticals Division, Investigational Toxicology, Muellerstr. 178, D-13353, Berlin, Germany
| | - Kristine L Witt
- The National Institute of Environmental Health Sciences, Division of the National Toxicology Program, Research Triangle Park, NC, 27709, USA
| | - Adam Woolley
- ForthTox Limited, PO Box 13550, Linlithgow, EH49 7YU, UK
| | - David Woolley
- ForthTox Limited, PO Box 13550, Linlithgow, EH49 7YU, UK
| | - Craig Zwickl
- Transendix LLC, 1407 Moores Manor, Indianapolis, IN, 46229, USA
| | - Glenn J Myatt
- Leadscope, Inc, 1393 Dublin Rd, Columbus, OH, 43215, USA
| |
Collapse
|
11
|
Addressing systematic inconsistencies between in vitro and in vivo transcriptomic mode of action signatures. Toxicol In Vitro 2019; 58:1-12. [DOI: 10.1016/j.tiv.2019.02.014] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2018] [Revised: 01/14/2019] [Accepted: 02/14/2019] [Indexed: 12/26/2022]
|
12
|
Bariar B, Vestal CG, Deem B, Goodenow D, Ughetta M, Engledove RW, Sahyouni M, Richardson C. Bioflavonoids promote stable translocations between MLL-AF9 breakpoint cluster regions independent of normal chromosomal context: Model system to screen environmental risks. ENVIRONMENTAL AND MOLECULAR MUTAGENESIS 2019; 60:154-167. [PMID: 30387535 PMCID: PMC6363851 DOI: 10.1002/em.22245] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/07/2018] [Revised: 07/26/2018] [Accepted: 08/06/2018] [Indexed: 06/08/2023]
Abstract
Infant acute leukemias are aggressive and characterized by rapid onset after birth. The majority harbor translocations involving the MLL gene with AF9 as one of its most common fusion partners. MLL and AF9 loci contain breakpoint cluster regions (bcrs) with sequences hypothesized to be targets of topoisomerase II inhibitors that promote translocation formation. Overlap of MLL bcr sequences associated with both infant acute leukemia and therapy-related leukemia following exposure to the topoisomerase II inhibitor etoposide led to the hypothesis that exposure during pregnancy to biochemically similar compounds may promote infant acute leukemia. We established a reporter system to systematically quantitate and stratify the potential for such compounds to promote chromosomal translocations between the MLL and AF9 bcrs analogous to those in infant leukemia. We show bioflavonoids genistein and quercetin most biochemically similar to etoposide have a strong association with MLL-AF9 bcr translocations, while kaempferol, fisetin, flavone, and myricetin have a weak but consistent association, and other compounds have a minimal association in both embryonic stem (ES) and hematopoietic stem cell (HSC) populations. The frequency of translocations induced by bioflavonoids at later stages of myelopoiesis is significantly reduced by more than one log. The MLL and AF9 bcrs are sensitive to these agents and recombinogenic independent of their native context suggesting bcr sequences themselves are drivers of illegitimate DNA repair reactions and translocations, not generation of functional oncogenic fusions. This system provides for rapid systematic screening of relative risk, dose dependence, and combinatorial impact of multitudes of dietary and environmental exposures on MLL-AF9 translocations. Environ. Mol. Mutagen. 60: 154-167, 2019. © 2018 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Bhawana Bariar
- University of North Carolina at Charlotte, Dept of Biological Sciences, 9201 University City Boulevard, Charlotte NC, 28223
| | - C. Greer Vestal
- University of North Carolina at Charlotte, Dept of Biological Sciences, 9201 University City Boulevard, Charlotte NC, 28223
| | - Bradley Deem
- University of North Carolina at Charlotte, Dept of Biological Sciences, 9201 University City Boulevard, Charlotte NC, 28223
| | - Donna Goodenow
- University of North Carolina at Charlotte, Dept of Biological Sciences, 9201 University City Boulevard, Charlotte NC, 28223
| | - Mimi Ughetta
- University of North Carolina at Charlotte, Dept of Biological Sciences, 9201 University City Boulevard, Charlotte NC, 28223
| | - R. Warren Engledove
- University of North Carolina at Charlotte, Dept of Biological Sciences, 9201 University City Boulevard, Charlotte NC, 28223
| | - Mark Sahyouni
- University of North Carolina at Charlotte, Dept of Biological Sciences, 9201 University City Boulevard, Charlotte NC, 28223
| | - Christine Richardson
- University of North Carolina at Charlotte, Dept of Biological Sciences, 9201 University City Boulevard, Charlotte NC, 28223
| |
Collapse
|
13
|
Dose-dependence of chemical carcinogenicity: Biological mechanisms for thresholds and implications for risk assessment. Chem Biol Interact 2019; 301:112-127. [DOI: 10.1016/j.cbi.2019.01.025] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2018] [Revised: 01/11/2019] [Accepted: 01/25/2019] [Indexed: 12/19/2022]
|
14
|
Haider S, Black MB, Parks BB, Foley B, Wetmore BA, Andersen ME, Clewell RA, Mansouri K, McMullen PD. A Qualitative Modeling Approach for Whole Genome Prediction Using High-Throughput Toxicogenomics Data and Pathway-Based Validation. Front Pharmacol 2018; 9:1072. [PMID: 30333746 PMCID: PMC6176017 DOI: 10.3389/fphar.2018.01072] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2018] [Accepted: 09/05/2018] [Indexed: 01/05/2023] Open
Abstract
Efficient high-throughput transcriptomics (HTT) tools promise inexpensive, rapid assessment of possible biological consequences of human and environmental exposures to tens of thousands of chemicals in commerce. HTT systems have used relatively small sets of gene expression measurements coupled with mathematical prediction methods to estimate genome-wide gene expression and are often trained and validated using pharmaceutical compounds. It is unclear whether these training sets are suitable for general toxicity testing applications and the more diverse chemical space represented by commercial chemicals and environmental contaminants. In this work, we built predictive computational models that inferred whole genome transcriptional profiles from a smaller sample of surrogate genes. The model was trained and validated using a large scale toxicogenomics database with gene expression data from exposure to heterogeneous chemicals from a wide range of classes (the Open TG-GATEs data base). The method of predictor selection was designed to allow high fidelity gene prediction from any pre-existing gene expression data set, regardless of animal species or data measurement platform. Predictive qualitative models were developed with this TG-GATES data that contained gene expression data of human primary hepatocytes with over 941 samples covering 158 compounds. A sequential forward search-based greedy algorithm, combining different fitting approaches and machine learning techniques, was used to find an optimal set of surrogate genes that predicted differential expression changes of the remaining genome. We then used pathway enrichment of up-regulated and down-regulated genes to assess the ability of a limited gene set to determine relevant patterns of tissue response. In addition, we compared prediction performance using the surrogate genes found from our greedy algorithm (referred to as the SV2000) with the landmark genes provided by existing technologies such as L1000 (Genometry) and S1500 (Tox21), finding better predictive performance for the SV2000. The ability of these predictive algorithms to predict pathway level responses is a positive step toward incorporating mode of action (MOA) analysis into the high throughput prioritization and testing of the large number of chemicals in need of safety evaluation.
Collapse
Affiliation(s)
- Saad Haider
- ScitoVation, Research Triangle Park, NC, United States
| | | | | | - Briana Foley
- ScitoVation, Research Triangle Park, NC, United States
| | | | | | | | | | | |
Collapse
|
15
|
Zhang Q, Li J, Middleton A, Bhattacharya S, Conolly RB. Bridging the Data Gap From in vitro Toxicity Testing to Chemical Safety Assessment Through Computational Modeling. Front Public Health 2018; 6:261. [PMID: 30255008 PMCID: PMC6141783 DOI: 10.3389/fpubh.2018.00261] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2018] [Accepted: 08/21/2018] [Indexed: 12/18/2022] Open
Abstract
Chemical toxicity testing is moving steadily toward a human cell and organoid-based in vitro approach for reasons including scientific relevancy, efficiency, cost, and ethical rightfulness. Inferring human health risk from chemical exposure based on in vitro testing data is a challenging task, facing various data gaps along the way. This review identifies these gaps and makes a case for the in silico approach of computational dose-response and extrapolation modeling to address many of the challenges. Mathematical models that can mechanistically describe chemical toxicokinetics (TK) and toxicodynamics (TD), for both in vitro and in vivo conditions, are the founding pieces in this regard. Identifying toxicity pathways and in vitro point of departure (PoD) associated with adverse health outcomes requires an understanding of the molecular key events in the interacting transcriptome, proteome, and metabolome. Such an understanding will in turn help determine the sets of sensitive biomarkers to be measured in vitro and the scope of toxicity pathways to be modeled in silico. In vitro data reporting both pathway perturbation and chemical biokinetics in the culture medium serve to calibrate the toxicity pathway and virtual tissue models, which can then help predict PoDs in response to chemical dosimetry experienced by cells in vivo. Two types of in vitro to in vivo extrapolation (IVIVE) are needed. (1) For toxic effects involving systemic regulations, such as endocrine disruption, organism-level adverse outcome pathway (AOP) models are needed to extrapolate in vitro toxicity pathway perturbation to in vivo PoD. (2) Physiologically-based toxicokinetic (PBTK) modeling is needed to extrapolate in vitro PoD dose metrics into external doses for expected exposure scenarios. Linked PBTK and TD models can explore the parameter space to recapitulate human population variability in response to chemical insults. While challenges remain for applying these modeling tools to support in vitro toxicity testing, they open the door toward population-stratified and personalized risk assessment.
Collapse
Affiliation(s)
- Qiang Zhang
- Department of Environmental Health, Rollins School of Public Health, Emory University, Atlanta, GA, United States
| | - Jin Li
- Unilever, Safety and Environmental Assurance Centre, Colworth Science Park, Sharnbrook, United Kingdom
| | - Alistair Middleton
- Unilever, Safety and Environmental Assurance Centre, Colworth Science Park, Sharnbrook, United Kingdom
| | - Sudin Bhattacharya
- Biomedical Engineering, Michigan State University, East Lansing, MI, United States
| | - Rory B Conolly
- Integrated Systems Toxicology Division, National Health and Environmental Effects Research Laboratory, United States Environmental Protection Agency, Durham, NC, United States
| |
Collapse
|
16
|
Marcus JM, Burke RT, Doak AE, Park S, Orth JD. Loss of p53 expression in cancer cells alters cell cycle response after inhibition of exportin-1 but does not prevent cell death. Cell Cycle 2018; 17:1329-1344. [PMID: 30037299 PMCID: PMC6110605 DOI: 10.1080/15384101.2018.1480224] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Accepted: 05/17/2018] [Indexed: 01/07/2023] Open
Abstract
The tumor suppressor protein p53 is central to the cellular stress response and may be a predictive biomarker for cancer treatments. Upon stress, wildtype p53 accumulates in the nucleus where it enforces cellular responses, including cell cycle arrest and cell death. p53 is so dominant in its effects, that p53 enforcement - or - restoration therapy is being studied for anti-cancer therapy. Two mechanistically distinct small molecules that act via p53 are the selective inhibitor of nuclear export, selinexor, and MDM2 inhibitor, nutlin-3a. Here, individual cells are studied to define cell cycle response signatures, which captures the variability of responses and includes the impact of loss of p53 expression on cell fates. The individual responses are then used to build the population level response. Matched cell lines with and without p53 expression indicate that while loss-of-function results in altered cell cycle signatures to selinexor treatment, it does not diminish overall cell loss. On the contrary, response to single-agent nutlin-3a shows a strong p53-dependence. Upon treatment with both selinexor and nutlin-3a there are combination effects in at least some cell lines - even when p53 is absent. Collectively, the findings indicate that p53 does act downstream of selinexor and nutlin-3a, and that p53 expression is dispensable for selinexor to cause cell death, but nutlin-3a response is more p53-dependent. Thus, TP53 disruption and lack of expression may not predict poor cell response to selinexor, and selinexor's mechanism of action potentially provides for strong efficacy regardless of p53 function.
Collapse
Affiliation(s)
- Joshua M. Marcus
- Department of Molecular, Cellular, and Developmental Biology, University of Colorado Boulder, Boulder, CO, USA
- Department of Pharmacology and Toxicology, University of Alabama, Birmingham, AL, USA
| | - Russell T. Burke
- Department of Molecular, Cellular, and Developmental Biology, University of Colorado Boulder, Boulder, CO, USA
| | - Andrea E. Doak
- Department of Molecular, Cellular, and Developmental Biology, University of Colorado Boulder, Boulder, CO, USA
- Division of Medical Oncology, University of Colorado School of Medicine, Aurora, CO, USA
| | - Soyeon Park
- Department of Molecular, Cellular, and Developmental Biology, University of Colorado Boulder, Boulder, CO, USA
| | - James D. Orth
- Department of Molecular, Cellular, and Developmental Biology, University of Colorado Boulder, Boulder, CO, USA
| |
Collapse
|
17
|
Averbeck D, Salomaa S, Bouffler S, Ottolenghi A, Smyth V, Sabatier L. Progress in low dose health risk research. MUTATION RESEARCH-REVIEWS IN MUTATION RESEARCH 2018; 776:46-69. [DOI: 10.1016/j.mrrev.2018.04.001] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/15/2018] [Revised: 04/11/2018] [Accepted: 04/12/2018] [Indexed: 12/11/2022]
|
18
|
Di Paolo C, Müller Y, Thalmann B, Hollert H, Seiler TB. p53 induction and cell viability modulation by genotoxic individual chemicals and mixtures. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2018; 25:4012-4022. [PMID: 28303539 DOI: 10.1007/s11356-017-8790-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/10/2016] [Accepted: 03/08/2017] [Indexed: 06/06/2023]
Abstract
The binding of the p53 tumor suppression protein to DNA response elements after genotoxic stress can be quantified by cell-based reporter gene assays as a DNA damage endpoint. Currently, bioassay evaluation of environmental samples requires further knowledge on p53 induction by chemical mixtures and on cytotoxicity interference with p53 induction analysis for proper interpretation of results. We investigated the effects of genotoxic pharmaceuticals (actinomycin D, cyclophosphamide) and nitroaromatic compounds (4-nitroquinoline 1-oxide, 3-nitrobenzanthrone) on p53 induction and cell viability using a reporter gene and a colorimetric assay, respectively. Individual exposures were conducted in the absence or presence of metabolic activation system, while binary and tertiary mixtures were tested in its absence only. Cell viability reduction tended to present direct correlation with p53 induction, and induction peaks occurred mainly at chemical concentrations causing cell viability below 80%. Mixtures presented in general good agreement between predicted and measured p53 induction factors at lower concentrations, while higher chemical concentrations gave lower values than expected. Cytotoxicity evaluation supported the selection of concentration ranges for the p53 assay and the interpretation of its results. The often used 80% viability threshold as a basis to select the maximum test concentration for cell-based assays was not adequate for p53 induction assessment. Instead, concentrations causing up to 50% cell viability reduction should be evaluated in order to identify the lowest observed effect concentration and peak values following meaningful p53 induction.
Collapse
Affiliation(s)
- Carolina Di Paolo
- Department of Ecosystem Analysis, Institute for Environmental Research, Aachen Biology and Biotechnology (ABBt), RWTH Aachen University, Aachen, Germany.
| | - Yvonne Müller
- Department of Ecosystem Analysis, Institute for Environmental Research, Aachen Biology and Biotechnology (ABBt), RWTH Aachen University, Aachen, Germany
| | - Beat Thalmann
- Department of Ecosystem Analysis, Institute for Environmental Research, Aachen Biology and Biotechnology (ABBt), RWTH Aachen University, Aachen, Germany
| | - Henner Hollert
- Department of Ecosystem Analysis, Institute for Environmental Research, Aachen Biology and Biotechnology (ABBt), RWTH Aachen University, Aachen, Germany
- College of Resources and Environmental Science, Chongqing University, 1 Tiansheng Road, Beibei, Chongqing, 400715, China
- College of Environmental Science and Engineering and State Key Laboratory of Pollution Control and Resource Reuse, Tongji University, 1239 Siping Road, Shanghai, China
- State Key Laboratory of Pollution Control and Resource Reuse, School of the Environment, Nanjing University, Nanjing, China
| | - Thomas-Benjamin Seiler
- Department of Ecosystem Analysis, Institute for Environmental Research, Aachen Biology and Biotechnology (ABBt), RWTH Aachen University, Aachen, Germany
| |
Collapse
|
19
|
Integration of the TGx-28.65 genomic biomarker with the flow cytometry micronucleus test to assess the genotoxicity of disperse orange and 1,2,4-benzenetriol in human TK6 cells. Mutat Res 2017; 806:51-62. [PMID: 29017062 DOI: 10.1016/j.mrfmmm.2017.09.002] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2017] [Revised: 07/21/2017] [Accepted: 09/10/2017] [Indexed: 12/13/2022]
Abstract
In vitro gene expression signatures to predict toxicological responses can provide mechanistic context for regulatory testing. We previously developed the TGx-28.65 genomic biomarker from a database of gene expression profiles derived from human TK6 cells exposed to 28 well-known compounds. The biomarker comprises 65 genes that can classify chemicals as DNA damaging or non-DNA damaging. In this study, we applied the TGx-28.65 genomic biomarker in parallel with the in vitro micronucleus (MN) assay to determine if two chemicals of regulatory interest at Health Canada, disperse orange (DO: the orange azo dye 3-[[4-[(4-Nitrophenyl)azo]phenyl] benzylamino]propanenitrile) and 1,2,4-benzenetriol (BT: a metabolite of benzene) are genotoxic or non-genotoxic. Both chemicals caused dose-dependent declines in relative survival and increases in apoptosis. A strong significant increase in MN induction was observed for all concentrations of BT; the top two concentrations of DO also caused a statistically significant increase in MN, but these increases were <2-fold above controls. TGx-28.65 analysis classified BT as genotoxic at all three concentrations and DO as genotoxic at the mid and high concentrations. Thus, although DO only caused a small increase in MN, this response was sufficient to induce a cellular DNA damage response. Benchmark dose modeling confirmed that BT is much more potent than DO. The results strongly suggest that follow-up work is required to assess whether DO and BT are also genotoxic in vivo. This is particularly important for DO, which may require metabolic activation by bacterial gut flora to fully induce its genotoxic potential. Our previously published data and this proof of concept study suggest that the TGx-28.65 genomic biomarker has the potential to add significant value to existing approaches used to assess genotoxicity.
Collapse
|
20
|
Witt KL, Hsieh JH, Smith-Roe SL, Xia M, Huang R, Auerbach SS, Hur J, Tice RR. Assessment of the DNA damaging potential of environmental chemicals using a quantitative high-throughput screening approach to measure p53 activation. ENVIRONMENTAL AND MOLECULAR MUTAGENESIS 2017; 58:494-507. [PMID: 28714573 PMCID: PMC5555817 DOI: 10.1002/em.22112] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/03/2017] [Revised: 06/16/2017] [Accepted: 06/19/2017] [Indexed: 05/08/2023]
Abstract
Genotoxicity potential is a critical component of any comprehensive toxicological profile. Compounds that induce DNA or chromosomal damage often activate p53, a transcription factor essential to cell cycle regulation. Thus, within the US Tox21 Program, we screened a library of ∼10,000 (∼8,300 unique) environmental compounds and drugs for activation of the p53-signaling pathway using a quantitative high-throughput screening assay employing HCT-116 cells (p53+/+ ) containing a stably integrated β-lactamase reporter gene under control of the p53 response element (p53RE). Cells were exposed (-S9) for 16 hr at 15 concentrations (generally 1.2 nM to 92 μM) three times, independently. Excluding compounds that failed analytical chemistry analysis or were suspected of inducing assay interference, 365 (4.7%) of 7,849 unique compounds were concluded to activate p53. As part of an in-depth characterization of our results, we first compared them with results from traditional in vitro genotoxicity assays (bacterial mutation, chromosomal aberration); ∼15% of known, direct-acting genotoxicants in our library activated the p53RE. Mining the Comparative Toxicogenomics Database revealed that these p53 actives were significantly associated with increased expression of p53 downstream genes involved in DNA damage responses. Furthermore, 53 chemical substructures associated with genotoxicity were enriched in certain classes of p53 actives, for example, anthracyclines (antineoplastics) and vinca alkaloids (tubulin disruptors). Interestingly, the tubulin disruptors manifested unusual nonmonotonic concentration response curves suggesting activity through a unique p53 regulatory mechanism. Through the analysis of our results, we aim to define a role for this assay as one component of a comprehensive toxicological characterization of large compound libraries. Environ. Mol. Mutagen. 58:494-507, 2017. © 2017 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Kristine L. Witt
- National Institute of Environmental Health Sciences, Division of the National Toxicology Program, Research Triangle Park, NC
- Corresponding author: Kristine L. Witt, NIEHS/DNTP, 919-541-2761,
| | | | - Stephanie L. Smith-Roe
- National Institute of Environmental Health Sciences, Division of the National Toxicology Program, Research Triangle Park, NC
| | - Menghang Xia
- National Institutes of Health Center for Advancing Translational Sciences, Bethesda, MD
| | - Ruili Huang
- National Institutes of Health Center for Advancing Translational Sciences, Bethesda, MD
| | - Scott S. Auerbach
- National Institute of Environmental Health Sciences, Division of the National Toxicology Program, Research Triangle Park, NC
| | - Junguk Hur
- Department of Biomedical Sciences, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, ND
| | - Raymond R. Tice
- National Institute of Environmental Health Sciences, Division of the National Toxicology Program, Research Triangle Park, NC
| |
Collapse
|
21
|
Burke RT, Marcus JM, Orth JD. Inhibition of exportin-1 function results in rapid cell cycle-associated DNA damage in cancer cells. Oncotarget 2017; 8:39460-39475. [PMID: 28467801 PMCID: PMC5503625 DOI: 10.18632/oncotarget.17063] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2016] [Accepted: 03/08/2017] [Indexed: 01/05/2023] Open
Abstract
Selective inhibitors of nuclear export (SINE) are small molecules in development as anti-cancer agents. The first-in-class SINE, selinexor, is in clinical trials for blood and solid cancers. Selinexor forms a covalent bond with exportin-1 at cysteine-528, and blocks its ability to export cargos. Previous work has shown strong cell cycle effects and drug-induced cell death across many different cancer-derived cell lines. Here, we report strong cell cycle-associated DNA double-stranded break formation upon the treatment of cancer cells with SINE. In multiple cell models, selinexor treatment results in the formation of clustered DNA damage foci in 30-40% of cells within 8 hours that is dependent upon cysteine-528. DNA damage strongly correlates with G1/S-phase and decreased DNA replication. Live cell microscopy reveals an association between DNA damage and cell fate. Cells that form damage in G1-phase more often die or arrest, while those damaged in S/G2-phase frequently progress to cell division. Up to half of all treated cells form damage foci, and most cells that die after being damaged, were damaged in G1-phase. By comparison, non-transformed cell lines show strong cell cycle effects but little DNA damage and less death than cancer cells. Significant drug combination effects occur when selinexor is paired with different classes of agents that either cause DNA damage or that diminish DNA damage repair. These data present a novel effect of exportin-1 inhibition and provide a strong rationale for multiple combination treatments of selinexor with agents that are currently in use for the treatment of different solid cancers.
Collapse
Affiliation(s)
- Russell T. Burke
- Department of Molecular, Cellular and Developmental Biology, University of Colorado Boulder, Boulder, CO, USA
| | - Joshua M. Marcus
- Department of Molecular, Cellular and Developmental Biology, University of Colorado Boulder, Boulder, CO, USA
- Current/Present address: Cell, Molecular and Developmental Biology, Graduate Biomedical Sciences, University of Alabama Birmingham, Birmingham, AL, USA
| | - James D. Orth
- Department of Molecular, Cellular and Developmental Biology, University of Colorado Boulder, Boulder, CO, USA
| |
Collapse
|
22
|
Klaus V, Bastek H, Damme K, Collins LB, Frötschl R, Benda N, Lutter D, Ellinger-Ziegelbauer H, Swenberg JA, Dietrich DR, Stemmer K. Time-matched analysis of DNA adduct formation and early gene expression as predictive tool for renal carcinogenesis in methylazoxymethanol acetate treated Eker rats. Arch Toxicol 2017; 91:3427-3438. [PMID: 28349193 DOI: 10.1007/s00204-017-1953-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2016] [Accepted: 03/01/2017] [Indexed: 11/28/2022]
Abstract
Genotoxic carcinogens pose great hazard to human health. Uncertainty of current risk assessment strategies and long latency periods between first carcinogen exposure and diagnosis of tumors have raised interest in predictive biomarkers. Initial DNA adduct formation is a necessary step for genotoxin induced carcinogenesis. However, as DNA adducts not always translate into tumorigenesis, their predictive value is limited. Here we hypothesize that the combined analysis of pro-mutagenic DNA adducts along with time-matched gene expression changes could serve as a superior prediction tool for genotoxic carcinogenesis. Eker rats, heterozygous for the tuberous sclerosis (Tsc2) tumor suppressor gene and thus highly susceptible towards genotoxic renal carcinogens, were continuously treated with the DNA alkylating carcinogen methylazoxymethanol acetate (MAMAc). Two weeks of MAMAc treatment resulted in a time-dependent increase of O6-methylguanine and N7-methylguanine adducts in the kidney cortex, which was however not reflected by significant expression changes of cyto-protective genes involved in DNA repair, cell cycle arrest or apoptosis. Instead, we found a transcriptional regulation of genes involved in the tumor-related MAPK, FoxO and TGF-beta pathways. Continuous MAMAc treatment for up to 6 months resulted in a mild but significant increase of cancerous lesions. In summary, the combined analysis of DNA adducts and early gene expression changes could serve as a suitable predictive tool for genotoxicant-induced carcinogenesis.
Collapse
Affiliation(s)
- Valentina Klaus
- Computational Discovery Research, Institute for Diabetes and Obesity, Helmholtz Diabetes Center & German Center for Diabetes Research (DZD), Helmholtz Zentrum München, Neuherberg, Germany
| | - Heinke Bastek
- Human and Environmental Toxicology, University of Konstanz, Konstanz, Germany
| | - Katja Damme
- Department of Environmental Sciences and Engineering, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Leonard B Collins
- Department of Environmental Sciences and Engineering, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Roland Frötschl
- Federal Institute for Drugs and Medical Devices, Bonn, Germany
| | - Norbert Benda
- Federal Institute for Drugs and Medical Devices, Bonn, Germany
| | - Dominik Lutter
- Computational Discovery Research, Institute for Diabetes and Obesity, Helmholtz Diabetes Center & German Center for Diabetes Research (DZD), Helmholtz Zentrum München, Neuherberg, Germany
| | | | - James A Swenberg
- Department of Environmental Sciences and Engineering, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Daniel R Dietrich
- Human and Environmental Toxicology, University of Konstanz, Konstanz, Germany
| | - Kerstin Stemmer
- Metabolism and Cancer, Institute for Diabetes and Obesity, Helmholtz Diabetes Center & German Center for Diabetes Research (DZD), Helmholtz Zentrum München, Neuherberg, Germany.
| |
Collapse
|
23
|
Sun B, Ross SM, Rowley S, Adeleye Y, Clewell RA. Contribution of ATM and ATR kinase pathways to p53-mediated response in etoposide and methyl methanesulfonate induced DNA damage. ENVIRONMENTAL AND MOLECULAR MUTAGENESIS 2017; 58:72-83. [PMID: 28195382 DOI: 10.1002/em.22070] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/09/2016] [Revised: 12/06/2016] [Accepted: 12/07/2016] [Indexed: 06/06/2023]
Abstract
p53 is a key integrator of cellular response to DNA damage, supporting post-translational repair and driving transcription-mediated responses including cell cycle arrest, apoptosis, and repair. DNA damage sensing kinases recognize different types of DNA damage and initiate specific responses through various post-translational modifications of p53. This study evaluated chemical specificity of the p53 pathway response by manipulating p53 or its upstream kinases and assessing the effect on DNA damage and cellular responses to prototype chemicals: etoposide (ETP, topoisomerase II inhibitor) and methyl methane sulfonate (MMS, alkylating agent). p53-deficient cells demonstrated reduced accumulation of the p53 target proteins MDM2, p21, and Wip1; reduced apoptotic response; and increased DNA damage (p-H2AX and micronuclei) with both chemicals. However, p53 was not essential for cell cycle arrest in HT1080 or HCT116 cells. The two chemicals induced different patterns of kinase activation, particularly in terms of Chk 1, Chk 2, p38, and ERK 1/2. However, inhibition of the ATM pathway showed a greater effect on p53 activtation, apoptosis, and accumulation of DNA damage than ATR-Chk 1 or the MAP kinases regardless of the chemical used. These results indicate that ATM is the predominant upstream kinase responsible for activation of the p53-mediated DNA damage response for both MMS and ETP, though the downstream kinase response is markedly different. Environ. Mol. Mutagen. 58:72-83, 2017. © 2017 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Bin Sun
- The Hamner Institutes for Health Sciences, Research Triangle Park, North Carolina, 27709
| | - Susan M Ross
- ScitoVation, LLC, Research Triangle Park, NC, 27709
| | - Sean Rowley
- ScitoVation, LLC, Research Triangle Park, NC, 27709
| | - Yeyejide Adeleye
- Unilever, Safety and Environmental Assurance Centre, Colworth Science Park, Sharnbrook, Bedfordshire, MK44 1LQ, United Kingdom
| | | |
Collapse
|
24
|
McCann J, McCann T. 2015 Lush Science Prize. Altern Lab Anim 2016; 44:461-468. [PMID: 27805829 DOI: 10.1177/026119291604400510] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The Lush Prize supports animal-free testing by rewarding the most effective projects and individuals who have been working toward the goal of replacing animals in product or ingredient safety testing. Prizes are awarded for developments in five strategic areas: Science; Lobbying; Training; Public Awareness; and Young Researchers. Should there be a major breakthrough in 21st century toxicology, a Black Box Prize equivalent to the entire annual fund of £250,000 is awarded. A Background Paper is prepared each year, prior to the judging process, to provide the panel with a brief overview of current developments in the field of Replacement alternatives, particularly those relevant to the concept of toxicity pathways. The Background Paper includes information on recent work by the relevant scientific institutions and projects in this area, including AXLR8, the OECD, The Hamner Institutes, the Human Toxome Project, EURL ECVAM, ICCVAM, the US Tox21 Programme, the ToxCast programme, and the Human Toxicology Project Consortium. Recent developments in toxicity pathway research are also assessed by reviewing the relevant literature (including conference proceedings), and the abstracts and papers receiving the highest score are presented to the judges for consideration.
Collapse
|
25
|
McMullen PD, Pendse S, Adeleye Y, Carmichael PL, Andersen ME, Clewell RA. Using Transcriptomics to Evaluate Thresholds in Genotoxicity Dose–Response. TOXICOGENOMICS IN PREDICTIVE CARCINOGENICITY 2016. [DOI: 10.1039/9781782624059-00185] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Several genotoxic chemicals have been reported to produce threshold-shaped dose–response curves for mutation and genotoxicity assays, both in vivo and in vitro, challenging the current default practice for risk assessment of genotoxic chemicals, which assumes a linear dose–response below the lowest tested dose. Statistical methods cannot determine whether a biological threshold exists with sufficient confidence to overturn this assumption of linearity. Indeed, to truly define the shape of the dose–response curves, we must look to the underlying biology and develop targeted experiments to identify and measure the key processes governing the response of the cell to DNA damage. This chapter describes a series of studies aimed at defining the key transcriptional responses. Two approaches were taken to evaluate transcriptional responses preventing micronucleus induction: (1) comparison of gene signatures for several prototype compounds at a single chemical dose that led to a similar activation of the p53-DNA damage pathway (i.e. 1.5-fold increase in total p53); and (2) evaluation of a subset of chemicals with in-depth dose–response studies. The goal of these efforts was to determine the transcriptional pathways responsible for maintaining homeostasis at low levels of DNA damage, i.e., the biological underpinning of threshold-shaped dose–response curves for mutagenicity.
Collapse
Affiliation(s)
| | - Salil Pendse
- The Hamner Institutes for Health Sciences Research Triangle Park NC USA
| | | | | | | | | |
Collapse
|
26
|
Zhang Q, Bhattacharya S, Pi J, Clewell RA, Carmichael PL, Andersen ME. Adaptive Posttranslational Control in Cellular Stress Response Pathways and Its Relationship to Toxicity Testing and Safety Assessment. Toxicol Sci 2016; 147:302-16. [PMID: 26408567 DOI: 10.1093/toxsci/kfv130] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Although transcriptional induction of stress genes constitutes a major cellular defense program against a variety of stressors, posttranslational control directly regulating the activities of preexisting stress proteins provides a faster-acting alternative response. We propose that posttranslational control is a general adaptive mechanism operating in many stress pathways. Here with the aid of computational models, we first show that posttranslational control fulfills two roles: (1) handling small, transient stresses quickly and (2) stabilizing the negative feedback transcriptional network. We then review the posttranslational control pathways for major stress responses-oxidative stress, metal stress, hyperosmotic stress, DNA damage, heat shock, and hypoxia. Posttranslational regulation of stress protein activities occurs by reversible covalent modifications, allosteric or non-allosteric enzymatic regulations, and physically induced protein structural changes. Acting in feedback or feedforward networks, posttranslational control may establish a threshold level of cellular stress. Sub-threshold stresses are handled adequately by posttranslational control without invoking gene transcription. With supra-threshold stress levels, cellular homeostasis cannot be maintained and transcriptional induction of stress genes and other gene programs, eg, those regulating cell metabolism, proliferation, and apoptosis, takes place. The loss of homeostasis with consequent changes in cellular function may lead to adverse cellular outcomes. Overall, posttranslational and transcriptional control pathways constitute a stratified cellular defense system, handling stresses coherently across time and intensity. As cell-based assays become a focus for chemical testing anchored on toxicity pathways, examination of proteomic and metabolomic changes as a result of posttranslational control occurring in the absence of transcriptomic alterations deserves more attention.
Collapse
Affiliation(s)
- Qiang Zhang
- *Institute for Chemical Safety Sciences, The Hamner Institutes for Health Sciences, Research Triangle Park, North Carolina 27709; School of Public Health, China Medical University, Shenyang, China; and Unilever, Safety and Environmental Assurance Centre, Colworth Science Park, Sharnbrook, Bedfordshire, UK
| | - Sudin Bhattacharya
- *Institute for Chemical Safety Sciences, The Hamner Institutes for Health Sciences, Research Triangle Park, North Carolina 27709; School of Public Health, China Medical University, Shenyang, China; and Unilever, Safety and Environmental Assurance Centre, Colworth Science Park, Sharnbrook, Bedfordshire, UK
| | - Jingbo Pi
- *Institute for Chemical Safety Sciences, The Hamner Institutes for Health Sciences, Research Triangle Park, North Carolina 27709; School of Public Health, China Medical University, Shenyang, China; and Unilever, Safety and Environmental Assurance Centre, Colworth Science Park, Sharnbrook, Bedfordshire, UK
| | - Rebecca A Clewell
- *Institute for Chemical Safety Sciences, The Hamner Institutes for Health Sciences, Research Triangle Park, North Carolina 27709; School of Public Health, China Medical University, Shenyang, China; and Unilever, Safety and Environmental Assurance Centre, Colworth Science Park, Sharnbrook, Bedfordshire, UK
| | - Paul L Carmichael
- *Institute for Chemical Safety Sciences, The Hamner Institutes for Health Sciences, Research Triangle Park, North Carolina 27709; School of Public Health, China Medical University, Shenyang, China; and Unilever, Safety and Environmental Assurance Centre, Colworth Science Park, Sharnbrook, Bedfordshire, UK
| | - Melvin E Andersen
- *Institute for Chemical Safety Sciences, The Hamner Institutes for Health Sciences, Research Triangle Park, North Carolina 27709; School of Public Health, China Medical University, Shenyang, China; and Unilever, Safety and Environmental Assurance Centre, Colworth Science Park, Sharnbrook, Bedfordshire, UK
| |
Collapse
|
27
|
Clewell RA, Andersen ME. Approaches for characterizing threshold dose-response relationships for DNA-damage pathways involved in carcinogenicity in vivo and micronuclei formation in vitro. Mutagenesis 2016; 31:333-40. [PMID: 26846943 DOI: 10.1093/mutage/gev078] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Assessing the shape of dose-response curves for DNA-damage in cellular systems and for the consequences of DNA damage in intact animals remains a controversial topic. This overview looks at aspects of the pharmacokinetics (PK) and pharmacodynamics (PD) of cellular DNA-damage/repair and their role in defining the shape of dose-response curves using an in vivo example with formaldehyde and in vitro examples for micronuclei (MN) formation with several test compounds. Formaldehyde is both strongly mutagenic and an endogenous metabolite in cells. With increasing inhaled concentrations, there were transitions in gene changes, from activation of selective stress pathway genes at low concentrations, to activation of pathways for cell-cycle control, p53-DNA damage, and stem cell niche pathways at higher exposures. These gene expression changes were more consistent with dose-dependent transitions in the PD responses to formaldehyde in epithelial cells in the intact rat rather than the low-dose linear extrapolation methods currently used for carcinogens. However, more complete PD explanations of non-linear dose response for creation of fixed damage in cells require detailed examination of cellular responses in vitro using measures of DNA damage and repair that are not easily accessible in the intact animal. In the second section of the article, we illustrate an approach from our laboratory that develops fit-for-purpose, in vitro assays and evaluates the PD of DNA damage and repair through studies using prototypical DNA-damaging agents. Examination of a broad range of responses in these cells showed that transcriptional upregulation of cell cycle control and DNA repair pathways only occurred at doses higher than those causing overt damage fixed damage-measured as MN formation. Lower levels of damage appear to be handled by post-translational repair process using pre-existing proteins. In depth evaluation of the PD properties of one such post-translational process (formation of DNA repair centers; DRCs) has indicated that the formation of DRCs and their ability to complete repair before replication are consistent with threshold behaviours for mutagenesis and, by extension, with chemical carcinogenesis.
Collapse
Affiliation(s)
- Rebecca A Clewell
- The Hamner Institutes for Health Sciences, Research Triangle Park, NC 27709-2137 , USA
| | - Melvin E Andersen
- The Hamner Institutes for Health Sciences, Research Triangle Park, NC 27709-2137 , USA
| |
Collapse
|
28
|
Yuan H, Zhang Q, Guo J, Zhang T, Zhao J, Li J, White A, Carmichael PL, Westmoreland C, Peng S. A PGC-1α-Mediated Transcriptional Network Maintains Mitochondrial Redox and Bioenergetic Homeostasis against Doxorubicin-Induced Toxicity in Human Cardiomyocytes: Implementation of TT21C. Toxicol Sci 2016; 150:400-17. [PMID: 26781513 DOI: 10.1093/toxsci/kfw006] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
Chemical toxicity testing is fast moving in a direction that relies increasingly on cell-basedin vitroassays anchored on toxicity pathways according to the toxicity testing in the 21st century vision. Identifying points of departure (POD) via these assays and revealing their mechanistic underpinnings via computational modeling of the relevant pathways are critical and challenging steps. Here we used doxorubicin (DOX) as a prototype chemical to study mitochondrial toxicity in human AC16 cells. Mitochondrial toxicity has been linked to cardiovascular risk of DOX, which has limited its clinical use as an antitumor drug. Ourin vitrostudy revealed a well-defined POD concentration of DOX below which adaptive induction of proliferator-activated receptor-γ coactivator-1α (PGC-1α) -mediated mitochondrial genes, including NRF-1, MnSOD, UCP2, and COX1, concurred with negligible changes in mitochondrial superoxide and cytotoxicity. At higher DOX concentrations adversity became significant with elevated superoxide and suppressed ATP levels. A computational model was formulated to simulate the PGC-1α-mediated transcriptional network comprising multiple negative feedback loops that underlie redox and bioenergetics homeostasis in the mitochondrion. The model recapitulated the transition phase from adaptive to adverse responses, supporting the notion that saturated induction of PGC-1α-mediated gene network underpins POD. The model further predicts (follow-up experiments verified) that silencing PGC-1α compromises the adaptive function of the transcriptional network, leading to disruption of mitochondria and cytotoxicity at lower DOX concentrations. In summary, our study demonstrates that combining pathway-focusedin vitroassays and computational simulation of relevant biochemical network is synergistic for understanding dose-response behaviors in the low-dose region and identifying POD.
Collapse
Affiliation(s)
- Haitao Yuan
- *Evaluation and Research Centre for Toxicology, Institute of Disease Control and Prevention, Academy of Military Medical Sciences, Beijing 100071, China;
| | - Qiang Zhang
- Department of Environmental Health, Rollins School of Public Health, Emory University, Atlanta, GA 30322, USA; and
| | - Jiabin Guo
- *Evaluation and Research Centre for Toxicology, Institute of Disease Control and Prevention, Academy of Military Medical Sciences, Beijing 100071, China
| | - Tingfen Zhang
- *Evaluation and Research Centre for Toxicology, Institute of Disease Control and Prevention, Academy of Military Medical Sciences, Beijing 100071, China
| | - Jun Zhao
- *Evaluation and Research Centre for Toxicology, Institute of Disease Control and Prevention, Academy of Military Medical Sciences, Beijing 100071, China
| | - Jin Li
- Unilever Safety and Environmental Assurance Center, Colworth Science Park, Sharnbrook, Bedfordshire MK44 1LQ, UK
| | - Andrew White
- Unilever Safety and Environmental Assurance Center, Colworth Science Park, Sharnbrook, Bedfordshire MK44 1LQ, UK
| | - Paul L Carmichael
- Unilever Safety and Environmental Assurance Center, Colworth Science Park, Sharnbrook, Bedfordshire MK44 1LQ, UK
| | - Carl Westmoreland
- Unilever Safety and Environmental Assurance Center, Colworth Science Park, Sharnbrook, Bedfordshire MK44 1LQ, UK
| | - Shuangqing Peng
- *Evaluation and Research Centre for Toxicology, Institute of Disease Control and Prevention, Academy of Military Medical Sciences, Beijing 100071, China;
| |
Collapse
|
29
|
Grafström RC, Nymark P, Hongisto V, Spjuth O, Ceder R, Willighagen E, Hardy B, Kaski S, Kohonen P. Toward the Replacement of Animal Experiments through the Bioinformatics-driven Analysis of 'Omics' Data from Human Cell Cultures. Altern Lab Anim 2016; 43:325-32. [PMID: 26551289 DOI: 10.1177/026119291504300506] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
This paper outlines the work for which Roland Grafström and Pekka Kohonen were awarded the 2014 Lush Science Prize. The research activities of the Grafström laboratory have, for many years, covered cancer biology studies, as well as the development and application of toxicity-predictive in vitro models to determine chemical safety. Through the integration of in silico analyses of diverse types of genomics data (transcriptomic and proteomic), their efforts have proved to fit well into the recently-developed Adverse Outcome Pathway paradigm. Genomics analysis within state-of-the-art cancer biology research and Toxicology in the 21st Century concepts share many technological tools. A key category within the Three Rs paradigm is the Replacement of animals in toxicity testing with alternative methods, such as bioinformatics-driven analyses of data obtained from human cell cultures exposed to diverse toxicants. This work was recently expanded within the pan-European SEURAT-1 project (Safety Evaluation Ultimately Replacing Animal Testing), to replace repeat-dose toxicity testing with data-rich analyses of sophisticated cell culture models. The aims and objectives of the SEURAT project have been to guide the application, analysis, interpretation and storage of 'omics' technology-derived data within the service-oriented sub-project, ToxBank. Particularly addressing the Lush Science Prize focus on the relevance of toxicity pathways, a 'data warehouse' that is under continuous expansion, coupled with the development of novel data storage and management methods for toxicology, serve to address data integration across multiple 'omics' technologies. The prize winners' guiding principles and concepts for modern knowledge management of toxicological data are summarised. The translation of basic discovery results ranged from chemical-testing and material-testing data, to information relevant to human health and environmental safety.
Collapse
Affiliation(s)
- Roland C Grafström
- Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Penny Nymark
- Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Vesa Hongisto
- Toxicology Department, Misvik Biology Corporation, Turku, Finland
| | - Ola Spjuth
- Department of Medical Epidemiology and Biostatistics, Swedish e-Science Research Centre, Karolinska Institutet, Stockholm, Sweden and Department of Pharmaceutical Biosciences and Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Rebecca Ceder
- Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Egon Willighagen
- Department of Bioinformatics-BiGCat, Maastricht University, Maastricht, The Netherlands
| | - Barry Hardy
- Douglas Connect GmbH, Zeiningen, Switzerland
| | - Samuel Kaski
- Helsinki Institute for Information Technology, Aalto University, Department of Computer Science, Helsinki, Finland
| | - Pekka Kohonen
- Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
30
|
Clewell RA, McMullen PD, Adeleye Y, Carmichael PL, Andersen ME. Pathway Based Toxicology and Fit-for-Purpose Assays. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2016; 856:205-230. [DOI: 10.1007/978-3-319-33826-2_8] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
|
31
|
Klapacz J, Pottenger LH, Engelward BP, Heinen CD, Johnson GE, Clewell RA, Carmichael PL, Adeleye Y, Andersen ME. Contributions of DNA repair and damage response pathways to the non-linear genotoxic responses of alkylating agents. MUTATION RESEARCH-REVIEWS IN MUTATION RESEARCH 2015; 767:77-91. [PMID: 27036068 DOI: 10.1016/j.mrrev.2015.11.001] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/16/2015] [Revised: 11/26/2015] [Accepted: 11/27/2015] [Indexed: 11/27/2022]
Abstract
From a risk assessment perspective, DNA-reactive agents are conventionally assumed to have genotoxic risks at all exposure levels, thus applying a linear extrapolation for low-dose responses. New approaches discussed here, including more diverse and sensitive methods for assessing DNA damage and DNA repair, strongly support the existence of measurable regions where genotoxic responses with increasing doses are insignificant relative to control. Model monofunctional alkylating agents have in vitro and in vivo datasets amenable to determination of points of departure (PoDs) for genotoxic effects. A session at the 2013 Society of Toxicology meeting provided an opportunity to survey the progress in understanding the biological basis of empirically-observed PoDs for DNA alkylating agents. Together with the literature published since, this review discusses cellular pathways activated by endogenous and exogenous alkylation DNA damage. Cells have evolved conserved processes that monitor and counteract a spontaneous steady-state level of DNA damage. The ubiquitous network of DNA repair pathways serves as the first line of defense for clearing of the DNA damage and preventing mutation. Other biological pathways discussed here that are activated by genotoxic stress include post-translational activation of cell cycle networks and transcriptional networks for apoptosis/cell death. The interactions of various DNA repair and DNA damage response pathways provide biological bases for the observed PoD behaviors seen with genotoxic compounds. Thus, after formation of DNA adducts, the activation of cellular pathways can lead to the avoidance of a mutagenic outcome. The understanding of the cellular mechanisms acting within the low-dose region will serve to better characterize risks from exposures to DNA-reactive agents at environmentally-relevant concentrations.
Collapse
Affiliation(s)
- Joanna Klapacz
- Toxicology & Environmental Research and Consulting, The Dow Chemical Company, Midland, MI 48674, USA.
| | - Lynn H Pottenger
- Toxicology & Environmental Research and Consulting, The Dow Chemical Company, Midland, MI 48674, USA; Current Address: Olin Corporation, Midland, MI 48674, USA
| | - Bevin P Engelward
- Department of Biological Engineering, MA Institute of Technology, Cambridge, MA 02139, USA
| | - Christopher D Heinen
- Center for Molecular Medicine, Neag Comprehensive Cancer Center, University of CT Health Center, Farmington, CT 06030, USA
| | - George E Johnson
- Institute of Life Science, College of Medicine, Swansea University, SA2 8PP, UK
| | - Rebecca A Clewell
- Hamner Institutes for Health Sciences, Research Triangle Park, NC 27709, USA
| | - Paul L Carmichael
- Unilever, Safety & Environmental Assurance Centre, Colworth Science Park, Sharnbrook, Bedfordshire MK44 1LQ, UK
| | - Yeyejide Adeleye
- Unilever, Safety & Environmental Assurance Centre, Colworth Science Park, Sharnbrook, Bedfordshire MK44 1LQ, UK
| | - Melvin E Andersen
- Hamner Institutes for Health Sciences, Research Triangle Park, NC 27709, USA
| |
Collapse
|
32
|
Marcus JM, Burke RT, DeSisto JA, Landesman Y, Orth JD. Longitudinal tracking of single live cancer cells to understand cell cycle effects of the nuclear export inhibitor, selinexor. Sci Rep 2015; 5:14391. [PMID: 26399741 PMCID: PMC4585873 DOI: 10.1038/srep14391] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2015] [Accepted: 08/28/2015] [Indexed: 12/22/2022] Open
Abstract
Longitudinal tracking is a powerful approach to understand the biology of single cells. In cancer therapy, outcome is determined at the molecular and cellular scale, yet relationships between cellular response and cell fate are often unknown. The selective inhibitor of nuclear export, selinexor, is in development for the treatment of various cancers. Selinexor covalently binds exportin-1, causing nuclear sequestration of cargo proteins, including key regulators of the cell cycle and apoptosis. The cell cycle effects of selinexor and the relationships between cell cycle effects and cell fates, has not been described for individual cells. Using fluorescent cell cycle indicators we report the majority of cell death after selinexor treatment occurs from a protracted G1-phase and early S-phase. G1- or early S-phase treated cells show the strongest response and either die or arrest, while those treated in late S- or G2-phase progress to mitosis and divide. Importantly, the progeny of cell divisions also die or arrest, mostly in the next G1-phase. Cells that survive selinexor are negative for multiple proliferation biomarkers, indicating a penetrant, arrested state. Selinexor acts quickly, shows strong cell cycle selectivity, and is highly effective at arresting cell growth and inducing death in cancer-derived cells.
Collapse
Affiliation(s)
- Joshua M Marcus
- Department of Molecular, Cellular, and Developmental Biology, GOLD A240B, 347 UCB, University of Colorado-Boulder, Boulder, CO 80309 USA
| | - Russell T Burke
- Department of Molecular, Cellular, and Developmental Biology, GOLD A240B, 347 UCB, University of Colorado-Boulder, Boulder, CO 80309 USA
| | - John A DeSisto
- Department of Molecular, Cellular, and Developmental Biology, GOLD A240B, 347 UCB, University of Colorado-Boulder, Boulder, CO 80309 USA
| | - Yosef Landesman
- Karyopharm Therapeutics, Inc., 85 Wells Ave., Newton, MA 02459
| | - James D Orth
- Department of Molecular, Cellular, and Developmental Biology, GOLD A240B, 347 UCB, University of Colorado-Boulder, Boulder, CO 80309 USA
| |
Collapse
|
33
|
Adeleye Y, Andersen M, Clewell R, Davies M, Dent M, Edwards S, Fowler P, Malcomber S, Nicol B, Scott A, Scott S, Sun B, Westmoreland C, White A, Zhang Q, Carmichael PL. Implementing Toxicity Testing in the 21st Century (TT21C): Making safety decisions using toxicity pathways, and progress in a prototype risk assessment. Toxicology 2015; 332:102-11. [DOI: 10.1016/j.tox.2014.02.007] [Citation(s) in RCA: 98] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2013] [Revised: 01/31/2014] [Accepted: 02/04/2014] [Indexed: 02/07/2023]
|
34
|
Deng Z, Yu L, Cao W, Zheng W, Chen T. Rational Design of Ruthenium Complexes Containing 2,6-Bis(benzimidazolyl)pyridine Derivatives with Radiosensitization Activity by Enhancing p53 Activation. ChemMedChem 2015; 10:991-8. [DOI: 10.1002/cmdc.201500127] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2015] [Indexed: 01/07/2023]
|
35
|
Bouhifd M, Andersen ME, Baghdikian C, Boekelheide K, Crofton KM, Fornace AJ, Kleensang A, Li H, Livi C, Maertens A, McMullen PD, Rosenberg M, Thomas R, Vantangoli M, Yager JD, Zhao L, Hartung T. The human toxome project. ALTEX 2015; 32:112-24. [PMID: 25742299 PMCID: PMC4778566 DOI: 10.14573/altex.1502091] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/09/2015] [Accepted: 03/02/2015] [Indexed: 12/26/2022]
Abstract
The Human Toxome Project, funded as an NIH Transformative Research grant 2011-2016, is focused on developing the concepts and the means for deducing, validating and sharing molecular pathways of toxicity (PoT). Using the test case of estrogenic endocrine disruption, the responses of MCF-7 human breast cancer cells are being phenotyped by transcriptomics and mass-spectroscopy-based metabolomics. The bioinformatics tools for PoT deduction represent a core deliverable. A number of challenges for quality and standardization of cell systems, omics technologies and bioinformatics are being addressed. In parallel, concepts for annotation, validation and sharing of PoT information, as well as their link to adverse outcomes, are being developed. A reasonably comprehensive public database of PoT, the Human Toxome Knowledge-base, could become a point of reference for toxicological research and regulatory test strategies.
Collapse
Affiliation(s)
- Mounir Bouhifd
- Johns Hopkins Bloomberg School of Public Health, Center for Alternatives to Animal Testing, Baltimore, MD, USA
| | | | - Christina Baghdikian
- ASPPH Fellow, National Center for Computational Toxicology, US EPA, Research Triangle Park, NC, USA
| | - Kim Boekelheide
- Brown University, Pathology & Laboratory Medicine, Providence, RI, USA
| | - Kevin M. Crofton
- US EPA, National Center for Computational Toxicology, Research Triangle Park, NC, USA
| | | | - Andre Kleensang
- Johns Hopkins Bloomberg School of Public Health, Center for Alternatives to Animal Testing, Baltimore, MD, USA
| | - Henghong Li
- Georgetown University Medical Center, Washington, DC, USA
| | | | - Alexandra Maertens
- Johns Hopkins Bloomberg School of Public Health, Center for Alternatives to Animal Testing, Baltimore, MD, USA
| | | | | | - Russell Thomas
- US EPA, National Center for Computational Toxicology, Research Triangle Park, NC, USA
| | | | - James D. Yager
- Johns Hopkins Bloomberg School of Public Health, Department of Environmental Health Sciences, Baltimore, MD, USA
| | - Liang Zhao
- Johns Hopkins Bloomberg School of Public Health, Center for Alternatives to Animal Testing, Baltimore, MD, USA
| | - Thomas Hartung
- Johns Hopkins Bloomberg School of Public Health, Center for Alternatives to Animal Testing, Baltimore, MD, USA
- University of Konstanz, Center for Alternatives to Animal Testing Europe, Konstanz, Germany
| |
Collapse
|
36
|
Guérard M, Baum M, Bitsch A, Eisenbrand G, Elhajouji A, Epe B, Habermeyer M, Kaina B, Martus H, Pfuhler S, Schmitz C, Sutter A, Thomas A, Ziemann C, Froetschl R. Assessment of mechanisms driving non-linear dose–response relationships in genotoxicity testing. MUTATION RESEARCH-REVIEWS IN MUTATION RESEARCH 2015; 763:181-201. [DOI: 10.1016/j.mrrev.2014.11.001] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/01/2014] [Revised: 10/31/2014] [Accepted: 11/01/2014] [Indexed: 01/15/2023]
|
37
|
Li F, Cao L, Li X, Li N, Wang Z, Wu H. Affinities of organophosphate flame retardants to tumor suppressor gene p53: An integrated in vitro and in silico study. Toxicol Lett 2015; 232:533-41. [DOI: 10.1016/j.toxlet.2014.12.006] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2014] [Revised: 12/01/2014] [Accepted: 12/07/2014] [Indexed: 11/16/2022]
|