1
|
López-Pascual E, Rienda I, Perez-Rojas J, Rapisarda A, Garcia-Llorens G, Jover R, Castell JV. Drug-Induced Fatty Liver Disease (DIFLD): A Comprehensive Analysis of Clinical, Biochemical, and Histopathological Data for Mechanisms Identification and Consistency with Current Adverse Outcome Pathways. Int J Mol Sci 2024; 25:5203. [PMID: 38791241 PMCID: PMC11121209 DOI: 10.3390/ijms25105203] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Revised: 05/02/2024] [Accepted: 05/06/2024] [Indexed: 05/26/2024] Open
Abstract
Drug induced fatty liver disease (DIFLD) is a form of drug-induced liver injury (DILI), which can also be included in the more general metabolic dysfunction-associated steatotic liver disease (MASLD), which specifically refers to the accumulation of fat in the liver unrelated to alcohol intake. A bi-directional relationship between DILI and MASLD is likely to exist: while certain drugs can cause MASLD by acting as pro-steatogenic factors, MASLD may make hepatocytes more vulnerable to drugs. Having a pre-existing MASLD significantly heightens the likelihood of experiencing DILI from certain medications. Thus, the prevalence of steatosis within DILI may be biased by pre-existing MASLD, and it can be concluded that the genuine true incidence of DIFLD in the general population remains unknown. In certain individuals, drug-induced steatosis is often accompanied by concomitant injury mechanisms such as oxidative stress, cell death, and inflammation, which leads to the development of drug-induced steatohepatitis (DISH). DISH is much more severe from the clinical point of view, has worse prognosis and outcome, and resembles MASH (metabolic-associated steatohepatitis), as it is associated with inflammation and sometimes with fibrosis. A literature review of clinical case reports allowed us to examine and evaluate the clinical features of DIFLD and their association with specific drugs, enabling us to propose a classification of DIFLD drugs based on clinical outcomes and pathological severity: Group 1, drugs with low intrinsic toxicity (e.g., ibuprofen, naproxen, acetaminophen, irinotecan, methotrexate, and tamoxifen), but expected to promote/aggravate steatosis in patients with pre-existing MASLD; Group 2, drugs associated with steatosis and only occasionally with steatohepatitis (e.g., amiodarone, valproic acid, and tetracycline); and Group 3, drugs with a great tendency to transit to steatohepatitis and further to fibrosis. Different mechanisms may be in play when identifying drug mode of action: (1) inhibition of mitochondrial fatty acid β-oxidation; (2) inhibition of fatty acid transport across mitochondrial membranes; (3) increased de novo lipid synthesis; (4) reduction in lipid export by the inhibition of microsomal triglyceride transfer protein; (5) induction of mitochondrial permeability transition pore opening; (6) dissipation of the mitochondrial transmembrane potential; (7) impairment of the mitochondrial respiratory chain/oxidative phosphorylation; (8) mitochondrial DNA damage, degradation and depletion; and (9) nuclear receptors (NRs)/transcriptomic alterations. Currently, the majority of, if not all, adverse outcome pathways (AOPs) for steatosis in AOP-Wiki highlight the interaction with NRs or transcription factors as the key molecular initiating event (MIE). This perspective suggests that chemical-induced steatosis typically results from the interplay between a chemical and a NR or transcription factors, implying that this interaction represents the primary and pivotal MIE. However, upon conducting this exhaustive literature review, it became evident that the current AOPs tend to overly emphasize this interaction as the sole MIE. Some studies indeed support the involvement of NRs in steatosis, but others demonstrate that such NR interactions alone do not necessarily lead to steatosis. This view, ignoring other mitochondrial-related injury mechanisms, falls short in encapsulating the intricate biological mechanisms involved in chemically induced liver steatosis, necessitating their consideration as part of the AOP's map road as well.
Collapse
Affiliation(s)
- Ernesto López-Pascual
- Department of Biochemistry and Molecular Biology, University of Valencia, 46010 Valencia, Spain
- Joint Research Unit in Experimental Hepatology, Health Research Institute La Fe, 46026 Valencia, Spain
| | - Ivan Rienda
- Pathology Department, Hospital Universitario y Politécnico La Fe, 46026 Valencia, Spain
| | - Judith Perez-Rojas
- Pathology Department, Hospital Universitario y Politécnico La Fe, 46026 Valencia, Spain
| | - Anna Rapisarda
- Department of Biochemistry and Molecular Biology, University of Valencia, 46010 Valencia, Spain
- Joint Research Unit in Experimental Hepatology, Health Research Institute La Fe, 46026 Valencia, Spain
| | - Guillem Garcia-Llorens
- Joint Research Unit in Experimental Hepatology, Health Research Institute La Fe, 46026 Valencia, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Ramiro Jover
- Department of Biochemistry and Molecular Biology, University of Valencia, 46010 Valencia, Spain
- Joint Research Unit in Experimental Hepatology, Health Research Institute La Fe, 46026 Valencia, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - José V. Castell
- Department of Biochemistry and Molecular Biology, University of Valencia, 46010 Valencia, Spain
- Joint Research Unit in Experimental Hepatology, Health Research Institute La Fe, 46026 Valencia, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Instituto de Salud Carlos III, 28029 Madrid, Spain
| |
Collapse
|
2
|
Harris KL, Harris KJ, Banks LD, Adunyah SE, Ramesh A. Acceleration of benzo(a)pyrene-induced colon carcinogenesis by Western diet in a rat model of colon cancer. Curr Res Toxicol 2024; 6:100162. [PMID: 38496007 PMCID: PMC10943645 DOI: 10.1016/j.crtox.2024.100162] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2024] [Revised: 03/02/2024] [Accepted: 03/06/2024] [Indexed: 03/19/2024] Open
Abstract
Colorectal cancer (CRC) is the third leading cause of cancer-related mortalities in the USA and around 52,550 people were expected to die from this disease by December 2023. The objective of this study was to investigate the effect of diet type on benzo(a)pyrene [B(a)P]-induced colon cancer in an adult male rat model, the Polyposis In the Rat Colon (PIRC) kindred type. Groups of PIRC rats (n = 10) were fed with AIN-76A regular diet (RD) or Western diet (WD) and received 25, 50 and 100 µg B(a)P/kg body wt. via oral gavage for 60 days. Rats fed diets alone, but no B(a)P, served as controls. After exposure, rats were euthanized; colon and liver samples were analyzed for activation of drug metabolizing enzymes (DMEs) CYP1A1, CYP1B1, SULT and GST. Plasma and tissue samples were analyzed by reverse phase-HPLC for B(a)P metabolites. In addition to these studies, DNA isolated from colon and liver tissues was analyzed for B(a)P-induced DNA adducts by the 32P-postlabeling method using a thin-layer chromatography system. Western diet consumption resulted in a marked increase in DME expression and B(a)P metabolite concentrations in rats that were administered 100 µg/kg B(a)P + WD (p < 0.05) compared to other treatment groups. Our findings demonstrate that WD accelerates the development of colon tumors induced by B(a)P through enhanced biotransformation, and the products of this process (metabolites) were found to bind with DNA and form B(a)P-DNA adducts, which may have given rise to colon polyps characterized by gain in tumor number, sizes, and dysplasia.
Collapse
Affiliation(s)
- Kelly L Harris
- Department of Biochemistry, Cancer Biology, Neuroscience & Pharmacology, Meharry Medical College, 1005 D.B. Todd Blvd., Nashville, TN 37208, United States
| | - Kenneth J Harris
- Department of Biochemistry, Cancer Biology, Neuroscience & Pharmacology, Meharry Medical College, 1005 D.B. Todd Blvd., Nashville, TN 37208, United States
| | - Leah D Banks
- Department of Biochemistry, Cancer Biology, Neuroscience & Pharmacology, Meharry Medical College, 1005 D.B. Todd Blvd., Nashville, TN 37208, United States
| | - Samuel E Adunyah
- Department of Biochemistry, Cancer Biology, Neuroscience & Pharmacology, Meharry Medical College, 1005 D.B. Todd Blvd., Nashville, TN 37208, United States
| | - Aramandla Ramesh
- Department of Biochemistry, Cancer Biology, Neuroscience & Pharmacology, Meharry Medical College, 1005 D.B. Todd Blvd., Nashville, TN 37208, United States
| |
Collapse
|
3
|
Olsvik PA, Meier S, Zhang X, Goksøyr A, Karlsen OA, Yadetie F. Environmentally realistic concentrations of chlorinated, brominated, and fluorinated persistent organic pollutants induce the unfolded protein response as a shared stress pathway in the liver of Atlantic cod (Gadus morhua). J Appl Toxicol 2023; 43:1859-1871. [PMID: 37528559 DOI: 10.1002/jat.4519] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Revised: 06/23/2023] [Accepted: 07/05/2023] [Indexed: 08/03/2023]
Abstract
In the North Sea and North Atlantic coastal areas, fish experience relatively high background levels of persistent organic pollutants. This study aimed to compare the mode of action of environmentally relevant concentrations of mixtures of halogenated compounds in Atlantic cod. Juvenile male cod with mean weight of 840 g were exposed by gavage to dietary mixtures of chlorinated (PCBs, DDT analogs, chlordane, lindane, and toxaphene), brominated (PBDEs), and fluorinated (PFOS) compounds for 4 weeks. One group received a combined mixture of all three compound groups. The results showed that the accumulated levels of chemicals in cod liver after 4 weeks of exposure reflected concentrations found in wild fish in this region. Pathway analysis revealed that the treatment effects by each of the three groups of chemicals (chlorinated, brominated, and fluorinated) converged on activation of the unfolded protein response (UPR). Upstream regulator analysis predicted that almost all the key transcription factors (XBP1, ERN1, ATF4, EIF2AK3, and NFE2L2) regulating the UPR were significantly activated. No additive effect was observed in cod co-treated with all three compound groups. In conclusion, the genome-wide transcriptomic study suggests that the UPR pathway is a sensitive common target of halogenated organic environmental pollutants in fish.
Collapse
Affiliation(s)
- Pål A Olsvik
- Faculty of Biosciences and Aquaculture, Nord University, Bodø, Norway
- Institute of Marine Research, Bergen, Norway
| | | | - Xiaokang Zhang
- Computational Biology Unit, Department of Informatics, University of Bergen, Bergen, Norway
| | - Anders Goksøyr
- Department of Biological Sciences, University of Bergen, Bergen, Norway
| | - Odd Andre Karlsen
- Department of Biological Sciences, University of Bergen, Bergen, Norway
| | - Fekadu Yadetie
- Department of Biological Sciences, University of Bergen, Bergen, Norway
| |
Collapse
|
4
|
Carta G, van der Stel W, Scuric EWJ, Capinha L, Delp J, Bennekou SH, Forsby A, Walker P, Leist M, van de Water B, Jennings P. Transcriptional landscape of mitochondrial electron transport chain inhibition in renal cells. Cell Biol Toxicol 2023; 39:3031-3059. [PMID: 37353587 PMCID: PMC10693540 DOI: 10.1007/s10565-023-09816-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Accepted: 05/23/2023] [Indexed: 06/25/2023]
Abstract
Analysis of the transcriptomic alterations upon chemical challenge, provides in depth mechanistic information on the compound's toxic mode of action, by revealing specific pathway activation and other transcriptional modulations. Mapping changes in cellular behaviour to chemical insult, facilitates the characterisation of chemical hazard. In this study, we assessed the transcriptional landscape of mitochondrial impairment through the inhibition of the electron transport chain (ETC) in a human renal proximal tubular cell line (RPTEC/TERT1). We identified the unfolded protein response pathway (UPR), particularly the PERK/ATF4 branch as a common cellular response across ETC I, II and III inhibitions. This finding and the specific genes elaborated may aid the identification of mitochondrial liabilities of chemicals in both legacy data and prospective transcriptomic studies.
Collapse
Affiliation(s)
- Giada Carta
- Division of Molecular and Computational Toxicology, Vrije University Amsterdam, Amsterdam, the Netherlands.
| | - Wanda van der Stel
- Division of Drug Discovery and Safety, Leiden Academic Centre of Drug Research, Leiden University, Leiden, the Netherlands
| | - Emma W J Scuric
- Division of Molecular and Computational Toxicology, Vrije University Amsterdam, Amsterdam, the Netherlands
| | - Liliana Capinha
- Division of Molecular and Computational Toxicology, Vrije University Amsterdam, Amsterdam, the Netherlands
| | - Johannes Delp
- In Vitro Toxicology and Biomedicine, Department inaugurated by the Doerenkamp‑Zbinden Foundation, University of Konstanz, Konstanz, Germany
| | | | - Anna Forsby
- Department of Biochemistry and Biophysics, Stockholm University, Stockholm, Sweden
| | - Paul Walker
- Cyprotex Discovery Ltd., Alderley Park, Macclesfield, Cheshire, UK
| | - Marcel Leist
- In Vitro Toxicology and Biomedicine, Department inaugurated by the Doerenkamp‑Zbinden Foundation, University of Konstanz, Konstanz, Germany
| | - Bob van de Water
- Division of Drug Discovery and Safety, Leiden Academic Centre of Drug Research, Leiden University, Leiden, the Netherlands
| | - Paul Jennings
- Division of Molecular and Computational Toxicology, Vrije University Amsterdam, Amsterdam, the Netherlands
| |
Collapse
|
5
|
Jennings P, Carta G, Singh P, da Costa Pereira D, Feher A, Dinnyes A, Exner TE, Wilmes A. Capturing time-dependent activation of genes and stress-response pathways using transcriptomics in iPSC-derived renal proximal tubule cells. Cell Biol Toxicol 2023; 39:1773-1793. [PMID: 36586010 PMCID: PMC10425493 DOI: 10.1007/s10565-022-09783-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Accepted: 12/06/2022] [Indexed: 01/01/2023]
Abstract
Transcriptomic analysis is a powerful method in the utilization of New Approach Methods (NAMs) for identifying mechanisms of toxicity and application to hazard characterization. With this regard, mapping toxicological events to time of exposure would be helpful to characterize early events. Here, we investigated time-dependent changes in gene expression levels in iPSC-derived renal proximal tubular-like cells (PTL) treated with five diverse compounds using TempO-Seq transcriptomics with the aims to evaluate the application of PTL for toxicity prediction and to report on temporal effects for the activation of cellular stress response pathways. PTL were treated with either 50 μM amiodarone, 10 μM sodium arsenate, 5 nM rotenone, or 300 nM tunicamycin over a temporal time course between 1 and 24 h. The TGFβ-type I receptor kinase inhibitor GW788388 (1 μM) was used as a negative control. Pathway analysis revealed the induction of key stress-response pathways, including Nrf2 oxidative stress response, unfolding protein response, and metal stress response. Early response genes per pathway were identified much earlier than 24 h and included HMOX1, ATF3, DDIT3, and several MT1 isotypes. GW788388 did not induce any genes within the stress response pathways above, but showed deregulation of genes involved in TGFβ inhibition, including downregulation of CYP24A1 and SERPINE1 and upregulation of WT1. This study highlights the application of iPSC-derived renal cells for prediction of cellular toxicity and sheds new light on the temporal and early effects of key genes that are involved in cellular stress response pathways.
Collapse
Affiliation(s)
- Paul Jennings
- Division of Molecular and Computational Toxicology, Chemistry and Pharmaceutical Sciences, AIMMS, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Giada Carta
- Division of Molecular and Computational Toxicology, Chemistry and Pharmaceutical Sciences, AIMMS, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Pranika Singh
- Edelweiss Connect GmbH, Technology Park Basel, Hochbergerstrasse 60C, 4057, Basel, Switzerland
- Division of Molecular and Systems Toxicology, Department of Pharmaceutical Sciences, University of Basel, Klingelbergstrasse 50, 4056, Basel, Switzerland
| | - Daniel da Costa Pereira
- Division of Molecular and Computational Toxicology, Chemistry and Pharmaceutical Sciences, AIMMS, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Anita Feher
- BioTalentum Ltd, Aulich Lajos Street 26, Gödöllő, 2100, Hungary
| | - Andras Dinnyes
- BioTalentum Ltd, Aulich Lajos Street 26, Gödöllő, 2100, Hungary
- HCEMM-USZ Stem Cell Research Group, Hungarian Centre of Excellence for Molecular Medicine, Szeged, 6723, Hungary
| | - Thomas E Exner
- Seven Past Nine d.o.o., Hribljane 10, 1380, Cerknica, Slovenia
| | - Anja Wilmes
- Division of Molecular and Computational Toxicology, Chemistry and Pharmaceutical Sciences, AIMMS, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands.
| |
Collapse
|
6
|
Jin Y, Qi G, Feng M, Yu D. The path via pathway-based approaches towards safety assessment: A concise review. Toxicol Appl Pharmacol 2022; 452:116195. [PMID: 35977605 DOI: 10.1016/j.taap.2022.116195] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Revised: 07/26/2022] [Accepted: 08/10/2022] [Indexed: 10/15/2022]
Abstract
For decades, chemical safety assessment has been proposed to shift from animal testing to in vitro testing systems in response to the call for the 3R. In Europe, the answer was to combine various information sources in integrated testing strategies (ITS); In the US, it was in 2007 when the landmark report by the National Research Council put forward a vision of in vitro toxicity testing paradigm. Since then, efforts to develop pathway-based assessment framework have been on the track. In 2010, systems biology brought out a conceptual framework called adverse outcome pathway (AOP), which took one step further from toxicity pathway to regulatory toxicology. Computational modeling, high-throughput screening, high-content omics have all been approached to facilitate this progress. This paper briefly reviewed the achievement of pathway-based chemical assessment since 2007, discussed potential pitfalls and challenges that mechanism-driven chemical assessment may undergo, and presented future perspectives of safety assessment that is to be based on computational system biology.
Collapse
Affiliation(s)
- Yuan Jin
- School of Public Health, Qingdao University, Qingdao, China
| | - Guangshuai Qi
- School of Public Health, Qingdao University, Qingdao, China
| | - Meiyao Feng
- Department of Environmental Health, Qingdao Municipal Center for Disease Control and Prevention, Qingdao Institute of Preventive Medicine, Qingdao, China
| | - Dianke Yu
- School of Public Health, Qingdao University, Qingdao, China..
| |
Collapse
|
7
|
Avilés-Ramírez C, Moreno-Godínez ME, Bonner MR, Parra-Rojas I, Flores-Alfaro E, Ramírez M, Huerta-Beristain G, Ramírez-Vargas MA. Effects of exposure to environmental pollutants on mitochondrial DNA copy number: a meta-analysis. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2022; 29:43588-43606. [PMID: 35399130 DOI: 10.1007/s11356-022-19967-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Accepted: 03/25/2022] [Indexed: 06/14/2023]
Abstract
Exposure to environmental pollutants has been associated with alteration on relative levels of mitochondrial DNA copy number (mtDNAcn). However, the results obtained from epidemiological studies are inconsistent. This meta-analysis aimed to evaluate whether environmental pollutant exposure can modify the relative levels of mtDNAcn in humans. We performed a literature search using PubMed, Scopus, and Web of Science databases. We selected and reviewed original articles performed in humans that analyzed the relationship between environmental pollutant exposure and the relative levels of mtDNAcn; the selection of the included studies was based on inclusion and exclusion criteria. Only twenty-two studies fulfilled our inclusion criteria. A total of 6011 study participants were included in this systematic review and meta-analysis. We grouped the included studies into four main categories according to the type of environmental pollutant: (1) heavy metals, (2) polycyclic aromatic hydrocarbons (PAHs), (3) particulate matter (PM), and (4) cigarette smoking. Inconclusive results were observed in all categories; the pooled analysis shows a marginal increase of relative levels of mtDNAcn in response to environmental pollutant exposure. The trial sequential analysis and rate confidence in body evidence showed the need to perform new studies. Therefore, a large-scale cohort and mechanistic studies in this area are required to probe the possible use of relative levels of mtDNAcn as biomarkers linked to environmental pollution exposure.
Collapse
Affiliation(s)
- Cristian Avilés-Ramírez
- Facultad de Ciencias Químico-Biológicas, Laboratorio de Toxicología Y Salud Ambiental, Universidad Autónoma De Guerrero, Av. Lázaro Cárdenas s/n, 39089, Chilpancingo, GRO, México
| | - Ma Elena Moreno-Godínez
- Facultad de Ciencias Químico-Biológicas, Laboratorio de Toxicología Y Salud Ambiental, Universidad Autónoma De Guerrero, Av. Lázaro Cárdenas s/n, 39089, Chilpancingo, GRO, México
| | - Matthew R Bonner
- Department of Epidemiology and Environmental Health, School of Public Health and Health Professions, University at Buffalo, Buffalo, NY, USA
| | - Isela Parra-Rojas
- Facultad de Ciencias Químico-Biológicas, Laboratorio de Investigación en Obesidad Y Diabetes, Universidad Autónoma de Guerrero, Chilpancingo, Guerrero, México
| | - Eugenia Flores-Alfaro
- Facultad de Ciencias Químico-Biológicas, Laboratorio de Epidemiología Clínica Y Molecular, Universidad Autónoma de Guerrero, Chilpancingo, Guerrero, México
| | - Mónica Ramírez
- Facultad de Ciencias Químico-Biológicas, CONACyT, Universidad Autónoma de Guerrero, Chilpancingo, Guerrero, México
| | - Gerardo Huerta-Beristain
- Facultad de Ciencias Químico-Biológicas, Laboratorio de Toxicología Y Salud Ambiental, Universidad Autónoma De Guerrero, Av. Lázaro Cárdenas s/n, 39089, Chilpancingo, GRO, México
| | - Marco Antonio Ramírez-Vargas
- Facultad de Ciencias Químico-Biológicas, Laboratorio de Toxicología Y Salud Ambiental, Universidad Autónoma De Guerrero, Av. Lázaro Cárdenas s/n, 39089, Chilpancingo, GRO, México.
| |
Collapse
|
8
|
Šimečková P, Pěnčíková K, Kováč O, Slavík J, Pařenicová M, Vondráček J, Machala M. In vitro profiling of toxic effects of environmental polycyclic aromatic hydrocarbons on nuclear receptor signaling, disruption of endogenous metabolism and induction of cellular stress. THE SCIENCE OF THE TOTAL ENVIRONMENT 2022; 815:151967. [PMID: 34843781 DOI: 10.1016/j.scitotenv.2021.151967] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Revised: 11/03/2021] [Accepted: 11/22/2021] [Indexed: 06/13/2023]
Abstract
Polycyclic aromatic hydrocarbons (PAHs) may interact with multiple intracellular receptors and related signaling pathways. We comprehensively evaluated the toxicity profiles of six environmentally relevant PAHs differing in structure, genotoxicity and their ability to activate the aryl hydrocarbon receptor (AhR). We focused particularly on their impact on intracellular hormone-, xenobiotic- and lipid-sensing receptors, as well as on cellular stress markers, combining a battery of human reporter gene assays and qRT-PCR evaluation of endogenous gene expression in human hepatocyte-like HepaRG cells, with LC/MS-MS analysis of cellular sphingolipids. The effects of PAHs included: activation of estrogen receptor α (in case of fluoranthene (Fla), pyrene (Pyr), benz[a]anthracene (BaA), benzo[a]pyrene (BaP)), suppression of androgen receptor activity (Fla, BaA, BaP and benzo[k]fluoranthene (BkF)), enhancement of dexamethasone-induced glucocorticoid receptor activity (chrysene (Chry), BaA, and BaP), and potentiation of triiodothyronine-induced thyroid receptor α activity (all tested PAHs). PAHs also induced transcription of endogenous gene targets of constitutive androstane receptor (Fla, Pyr), or repression of target genes of pregnane X receptor and peroxisome proliferator-activated receptor α (in case of the AhR-activating PAHs - Chry, BaA, BaP, and BkF) in HepaRG cells. In the same cell model, the AhR agonists reduced the expression of glucose metabolism genes (PCK1, G6PC and PDK4), and they up-regulated levels of glucosylceramides, together with a concomitant induction of expression of UGCG, glucosylceramide synthesis enzyme. Finally, both BaP and BkF were found to induce expression of early stress and genotoxicity markers: ATF3, EGR1, GDF15, CDKN1A/p21, and GADD45A mRNAs, while BaP alone increased levels of IL-6 mRNA. Overall, whereas low-molecular-weight PAHs exerted significant effects on nuclear receptors (with CYP2B6 induction observed already at nanomolar concentrations), the AhR activation by 4-ring and 5-ring PAHs appeared to be a key mechanism underlying their impact on nuclear receptor signaling, endogenous metabolism and induction of early stress and genotoxicity markers.
Collapse
Affiliation(s)
- Pavlína Šimečková
- Department of Pharmacology and Toxicology, Veterinary Research Institute, 62100 Brno, Czech Republic
| | - Kateřina Pěnčíková
- Department of Pharmacology and Toxicology, Veterinary Research Institute, 62100 Brno, Czech Republic
| | - Ondrej Kováč
- Department of Pharmacology and Toxicology, Veterinary Research Institute, 62100 Brno, Czech Republic
| | - Josef Slavík
- Department of Pharmacology and Toxicology, Veterinary Research Institute, 62100 Brno, Czech Republic
| | - Martina Pařenicová
- Department of Pharmacology and Toxicology, Veterinary Research Institute, 62100 Brno, Czech Republic
| | - Jan Vondráček
- Department of Cytokinetics, Institute of Biophysics of the Czech Academy of Sciences, 61265 Brno, Czech Republic
| | - Miroslav Machala
- Department of Pharmacology and Toxicology, Veterinary Research Institute, 62100 Brno, Czech Republic.
| |
Collapse
|
9
|
PBX1 attenuates 6-OHDA-induced oxidative stress and apoptosis and affects PINK1/PARKIN expression in dopaminergic neurons via FOXA1. Cytotechnology 2022; 74:217-229. [PMID: 35464170 PMCID: PMC8975925 DOI: 10.1007/s10616-021-00518-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Accepted: 12/31/2021] [Indexed: 11/03/2022] Open
Abstract
PBX1 expression has been found to be significantly reduced in nigrostriatal neurons of PD patients, but the effect of PBX1 on ROS and apoptosis in nigrostriatal dopamine neurons is not yet known. This paper aimed to explore whether PBX1 could be involved in the development of PD. The construction of the in vitro PD model was followed by the determination of PBX1 expression. Then, PBX1 was overexpressed to observe the changes in the cell viability, TH expression, oxidative stress and apoptosis of the model. The mitochondrial membrane potential analysis and detection of PINK1/parkin expression were also conducted. To observe whether FOXA1 was involved in the mechanism, its expression was measured, and its association with PBX1 was determined. Subsequently, FOXA1 was silenced to observe whether PBX1 did effects on dopaminergic neuron via FOXA1. PBX1 attenuates 6-OHDA-induced dopaminergic neuronal cell injury and oxidative stress, and apoptosis. Its overexpression ameliorates mitochondrial dysfunction in dopaminergic neurons and upregulates the expression of PINK1/parkin. PBX1 could combine with FOXA1 and affects 6-OHDA-induced dopaminergic neuronal damage and regulates PINK1/PARKIN expression via FOXA1. To conclude, PBX1 attenuates 6-OHDA-induced oxidative stress and apoptosis in dopaminergic neurons, and affected PINK1/PARKIN expression via FOXA1, which indicates the great potential of the PBX1 in prevention against PD.
Collapse
|
10
|
Drosophila Keap1 xenobiotic response factor regulates developmental transcription through binding to chromatin. Dev Biol 2022; 481:139-147. [PMID: 34662537 PMCID: PMC9502878 DOI: 10.1016/j.ydbio.2021.10.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2021] [Revised: 10/01/2021] [Accepted: 10/08/2021] [Indexed: 01/03/2023]
Abstract
The Keap1-Nrf2 complex is a central regulator that mediates transcriptional responses to xenobiotic stimuli and is highly related with multiple human diseases. The molecular mechanisms and biological functions of Keap1 and Nrf2 are not fully understood. The Drosophila Keap1 homolog (dKeap1) is conserved with mammalian Keap1 except that dKeap1 contains a 156 aa C-terminal tail (CTD). A dKeap1 truncation with the CTD removed (dKeap1-ΔCTD) shows abolished nuclear localization and chromatin-binding. Expression of dKeap1-ΔCTD in the dKeap1 null background significantly rescues this mutant to the adult stage, but the files showed partial lethality, sterility and defects in adipose tissue. In the rescued flies, expression levels of ecdysone-response genes, ecdysone-synthetic genes and adipogenesis genes were down-regulated in specific tissues, indicating that the chromatin-binding of dKeap1 mediates the activation of these developmental genes. At the same time, dKeap1-ΔCTD can still suppress the basal expression of detoxifying genes and mediate the activation of these genes in response to xenobiotic stimuli, suggesting that the chromatin-binding of dKeap1 is not required for the regulation of detoxifying genes. These results support a model in which dKeap1 on one hand functions as an inhibitor for the Nrf2-mediated transcription in the xenobiotic response pathway and on the other hand functions as a chromatin-binding transcription activator in the developmental pathway. Our study reveals a novel mechanism whereby Keap1-Nrf2 xenobiotic response signaling regulates development using a mechanism independent of redox signaling.
Collapse
|
11
|
Chambers B, Shah I. Evaluating adaptive stress response gene signatures using transcriptomics. COMPUTATIONAL TOXICOLOGY (AMSTERDAM, NETHERLANDS) 2021; 20:1-9. [PMID: 37829472 PMCID: PMC10569130 DOI: 10.1016/j.comtox.2021.100179] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/14/2023]
Abstract
Stress response pathways (SRPs) mitigate the cellular effects of chemicals, but excessive perturbation can lead to adverse outcomes. Here, we investigated a computational approach to evaluate SRP activity from transcriptomic data using gene set enrichment analysis (GSEA). We extracted published gene signatures for DNA damage response (DDR), unfolded protein response (UPR), heat shock response (HSR), response to hypoxia (HPX), metal-associated response (MTL), and oxidative stress response (OSR) from the Molecular Signatures Database (MSigDB). Next, we used a gene-frequency approach to build consensus SRP signatures of varying lengths from 50 to 477 genes. We then prepared a reference dataset from perturbagens associated with SRPs from the literature with their transcriptomic profiles retrieved from public repositories. Lastly, we used receiver-operator characteristic analysis to evaluate the GSEA scores from matching transcriptomic reference profiles to SRP signatures. Our consensus signatures performed better than or as well as published signatures for 4 out of the 6 SRPs, with the best consensus signature area under the curve (% performance relative to median of published signatures) of 1.00 for DDR (109%), 0.86 for UPR (169%), 0.99 for HTS (103%), 1.00 for HPX (104%), 0.74 for MTL (150%) and 0.83 for OSR (148%). The best matches between transcriptomic profiles and SRP signatures correctly classified perturbagens in 78% and 88% of the cases by first and second rank, respectively. We believe this approach can characterize SRP activity for new chemicals using transcriptomics with further evaluation.
Collapse
Affiliation(s)
- Bryant Chambers
- Center for Computational Toxicology and Exposure, Office of Research and Development, U.S. Environmental Protection Agency, Research Triangle Park, North Carolina, USA
| | - Imran Shah
- Center for Computational Toxicology and Exposure, Office of Research and Development, U.S. Environmental Protection Agency, Research Triangle Park, North Carolina, USA
| |
Collapse
|
12
|
Singh P, Chandrasekaran V, Hardy B, Wilmes A, Jennings P, Exner TE. Temporal transcriptomic alterations of cadmium exposed human iPSC-derived renal proximal tubule-like cells. Toxicol In Vitro 2021; 76:105229. [PMID: 34352368 DOI: 10.1016/j.tiv.2021.105229] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Revised: 06/25/2021] [Accepted: 07/26/2021] [Indexed: 12/12/2022]
Abstract
Cadmium is a well-studied environmental pollutant where the kidney and particularly the proximal tubule cells are especially sensitive as they are exposed to higher concentrations of cadmium than other tissues. Here we investigated the temporal transcriptomic alterations (TempO-Seq) of human induced pluripotent stem cell (iPSC)-derived renal proximal tubule-like (PTL) cells exposed to 5 μM cadmium chloride for 1, 2, 4, 8, 12, 16, 20, 24, 72 and 168 h. There was an early activation (within 4 h) of the metal and oxidative stress responses (metal-responsive transcription factor-1 (MTF1) and nuclear factor erythroid-2-related factor 2 (Nrf2) genes). The Nrf2 response returned to baseline within 24 h. The Activator Protein 1 (AP-1) regulated genes HSPA6 and FOSL-1 followed the Nrf2 time course. While the MTF1 genes also spiked at 4 h, they remained strongly elevated over the entire exposure period. The data and cell culture model utilised will be useful in further research aimed at the refinement of safe human exposure limits for cadmium, other metals and their mixtures.
Collapse
Affiliation(s)
- Pranika Singh
- Edelweiss Connect GmbH, Technology Park Basel, Hochbergerstrasse 60C, 4057 Basel, Switzerland; Division of Molecular and Systems Toxicology, Department of Pharmaceutical Sciences, University of Basel, Klingelbergstrasse 50, 4056 Basel, Switzerland
| | - Vidya Chandrasekaran
- Division of Molecular and Computational Toxicology, Chemistry and Pharmaceutical Sciences, AIMMS, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | - Barry Hardy
- Edelweiss Connect GmbH, Technology Park Basel, Hochbergerstrasse 60C, 4057 Basel, Switzerland
| | - Anja Wilmes
- Division of Molecular and Computational Toxicology, Chemistry and Pharmaceutical Sciences, AIMMS, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | - Paul Jennings
- Division of Molecular and Computational Toxicology, Chemistry and Pharmaceutical Sciences, AIMMS, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands.
| | - Thomas E Exner
- Seven Past Nine d.o.o., Hribljane 10, 1380 Cerknica, Slovenia.
| |
Collapse
|
13
|
Craven CB, Blackstock LKJ, Xie J, Li J, Yuan CG, Li XF. Analytical discovery of water disinfection byproducts of toxicological relevance: highlighting halobenzoquinones. CAN J CHEM 2021. [DOI: 10.1139/cjc-2021-0036] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Analytical advancement enables discoveries in water research, and challenges in the identification and determination of a wide range of trace level toxic compounds in water drives the development of new analytical platforms and tools. The identification of toxic disinfection byproducts (DBPs) in disinfected drinking water is an excellent example. Water disinfection is necessary to protect the public from waterborne disease. However, an unintentional consequence is the formation of DBPs resulting from reactions of disinfectants with natural organic matter in source water. To date, regulated DBPs do not account for the increased bladder cancer risk estimated in epidemiological studies. The majority of halogenated DBPs remain unidentified and the discovery of unknown DBPs of toxicological relevance continues to be a major focus of current water research. This review will highlight halobenzoquinones as a class of DBPs that serves as an example of analytical development and toxicological studies. We will feature recent trends and gaps in analytical technologies for identification of unknown DBPs and bioassays for evaluation of the toxicological effects of specific DBPs and their mixtures.
Collapse
Affiliation(s)
- Caley B. Craven
- Division of Analytical and Environmental Toxicology, Department of Laboratory Medicine and Pathology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB T6G 2G3, Canada
- Department of Chemistry, Faculty of Science, University of Alberta, Edmonton, AB T6G 2G3, Canada
| | - Lindsay K. Jmaiff Blackstock
- Division of Analytical and Environmental Toxicology, Department of Laboratory Medicine and Pathology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB T6G 2G3, Canada
| | - Jiaojiao Xie
- Division of Analytical and Environmental Toxicology, Department of Laboratory Medicine and Pathology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB T6G 2G3, Canada
- Department of Environmental Science and Engineering, North China Electric Power University, Baoding, 071000, PR China
| | - Jinhua Li
- Division of Analytical and Environmental Toxicology, Department of Laboratory Medicine and Pathology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB T6G 2G3, Canada
| | - Chun-Gang Yuan
- Department of Environmental Science and Engineering, North China Electric Power University, Baoding, 071000, PR China
| | - Xing-Fang Li
- Division of Analytical and Environmental Toxicology, Department of Laboratory Medicine and Pathology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB T6G 2G3, Canada
| |
Collapse
|
14
|
Cervantes PW, Corton JC. A Gene Expression Biomarker Predicts Heat Shock Factor 1 Activation in a Gene Expression Compendium. Chem Res Toxicol 2021; 34:1721-1737. [PMID: 34170685 DOI: 10.1021/acs.chemrestox.0c00510] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
The United States Environmental Protection Agency (US EPA) recently developed a tiered testing strategy to use advances in high-throughput transcriptomics (HTTr) testing to identify molecular targets of thousands of environmental chemicals that can be linked to adverse outcomes. Here, we describe a method that uses a gene expression biomarker to predict chemical activation of heat shock factor 1 (HSF1), a transcription factor critical for proteome maintenance. The HSF1 biomarker was built from transcript profiles derived from A375 cells exposed to a HSF1-activating heat shock protein (HSP) 90 inhibitor in the presence or absence of HSF1 expression. The resultant 44 identified genes included those that (1) are dependent on HSF1 for regulation, (2) have direct interactions with HSF1 assessed by ChIP-Seq, and (3) are in the molecular chaperone family. To test for accuracy, the biomarker was compared in a pairwise manner to gene lists derived from treatments with known HSF1 activity (HSP and proteasomal inhibitors) using the correlation-based Running Fisher test; the balanced accuracy for prediction was 96%. A microarray compendium consisting of 12,092 microarray comparisons from human cells exposed to 2670 individual chemicals was screened using our approach; 112 and 19 chemicals were identified as putative HSF1 activators or suppressors, respectively, and most appear to be novel modulators. A large percentage of the chemical treatments that induced HSF1 also induced oxidant-activated NRF2 (∼46%). For five compounds or mixtures, we found that NRF2 activation occurred at lower concentrations or at earlier times than HSF1 activation, supporting the concept of a tiered cellular protection system dependent on the level of chemical-induced stress. The approach described here could be used to identify environmentally relevant chemical HSF1 activators in HTTr data sets.
Collapse
Affiliation(s)
- Patrick W Cervantes
- Center for Computational Toxicology and Exposure, United States Environmental Protection Agency, Research Triangle Park, North Carolina 27711, United States.,Department of Medical Microbiology and Immunology, University of Wisconsin-Madison, Madison 53706, Wisconsin, United States
| | - J Christopher Corton
- Center for Computational Toxicology and Exposure, United States Environmental Protection Agency, Research Triangle Park, North Carolina 27711, United States
| |
Collapse
|
15
|
Bao S, Lin J, Xie M, Wang C, Nie X. Simvastatin affects Nrf2/MAPK signaling pathway and hepatic histological structure change in Gambusia affinis. CHEMOSPHERE 2021; 269:128725. [PMID: 33153852 DOI: 10.1016/j.chemosphere.2020.128725] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/23/2020] [Revised: 10/20/2020] [Accepted: 10/21/2020] [Indexed: 06/11/2023]
Abstract
Simvastatin (SV) is a typical lipid-lowering agent detected widely in waters, so its latent toxic effects to fish are deserved of concern. The purposes of this study aim at revealing the responses of antioxidant system in mosquitofish (Gambusia affinis) under SV exposure. Transcriptional expressions of oxidative stress-related key transcriptional factor Nrf2 and its downstream genes in mosquitofish were determined under SV exposure for different time. Partly related enzymatic activities, Nrf2 and MAPK protein expressions were also addressed in the same conditions, and histological changes in liver tissues were investigated too. Results showed that Nrf2 mRNA increased with the rising SV concentrations at 3 d and 7 d, displaying typical dose-dependent relationship, and Nrf2 protein by WB showed consistency with transcriptional changes to some degree. Comparatively, responses of gene expressions were more sensitive than enzymatic changes. The histological changes in the mosquitofish liver exposed to SV for 7 d indicated the potential adverse effects of statins. This work demonstrated that SV in aquatic environment could affect the transcriptional expression of antioxidant system, partly related enzymatic activity, and hepatic structure in the mosquitofish, revealing its potential risk on non-target organisms and environmental safety.
Collapse
Affiliation(s)
- Shuang Bao
- Department of Ecology/Hydrobiology Research Institute, Jinan University, Guangzhou, 510632, China
| | - Jiawei Lin
- Department of Ecology/Hydrobiology Research Institute, Jinan University, Guangzhou, 510632, China
| | - Meinan Xie
- Department of Ecology/Hydrobiology Research Institute, Jinan University, Guangzhou, 510632, China
| | - Chao Wang
- Department of Ecology/Hydrobiology Research Institute, Jinan University, Guangzhou, 510632, China
| | - Xiangping Nie
- Department of Ecology/Hydrobiology Research Institute, Jinan University, Guangzhou, 510632, China; Key Laboratory of Eutrophication and Red Tide Prevention of Guangdong Higher Education Institutes, Jinan University, Guangzhou, 510632, China.
| |
Collapse
|
16
|
Wellens S, Dehouck L, Chandrasekaran V, Singh P, Loiola RA, Sevin E, Exner T, Jennings P, Gosselet F, Culot M. Evaluation of a human iPSC-derived BBB model for repeated dose toxicity testing with cyclosporine A as model compound. Toxicol In Vitro 2021; 73:105112. [PMID: 33631201 DOI: 10.1016/j.tiv.2021.105112] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Revised: 01/25/2021] [Accepted: 02/09/2021] [Indexed: 12/25/2022]
Abstract
The blood-brain barrier (BBB) is a highly restrictive barrier that preserves central nervous system homeostasis and ensures optimal brain functioning. Using BBB cell assays makes it possible to investigate whether a compound is likely to compromise BBBs functionality, thereby probably resulting in neurotoxicity. Recently, several protocols to obtain human brain-like endothelial cells (BLECs) from induced pluripotent stem cells (iPSCs) have been reported. Within the framework of the European MSCA-ITN in3 project, we explored the possibility to use an iPSC-derived BBB model to assess the effects of repeated dose treatment with chemicals, using Cyclosporine A (CsA) as a model compound. The BLECs were found to exhibit important BBB characteristics up to 15 days after the end of the differentiation and could be used to assess the effects of repeated dose treatment. Although BLECs were still undergoing transcriptional changes over time, a targeted transcriptome analysis (TempO-Seq) indicated a time and concentration dependent activation of ATF4, XBP1, Nrf2 and p53 stress response pathways under CsA treatment. Taken together, these results demonstrate that this iPSC-derived BBB model and iPSC-derived models in general hold great potential to study the effects of repeated dose exposure with chemicals, allowing personalized and patient-specific studies in the future.
Collapse
Affiliation(s)
- Sara Wellens
- University of Artois, UR 2465, Laboratoire de la Barrière Hémato-Encéphalique (LBHE), Faculté des sciences Jean Perrin, Rue Jean Souvraz SP18, F-62300 Lens, France
| | - Lucie Dehouck
- University of Artois, UR 2465, Laboratoire de la Barrière Hémato-Encéphalique (LBHE), Faculté des sciences Jean Perrin, Rue Jean Souvraz SP18, F-62300 Lens, France
| | - Vidya Chandrasekaran
- Division of Molecular and Computational Toxicology, Department of Chemistry and Pharmaceutical Sciences, Amsterdam Institute for Molecules, Medicines and Systems, Vrije Universiteit Amsterdam, De Boelelaan 1108, 1081HZ Amsterdam, the Netherlands
| | - Pranika Singh
- Edelweiss Connect GmbH, Technology Park Basel, Hochbergerstrasse 60C, 4057 Basel, Switzerland; Division of Molecular and Systems Toxicology, Department of Pharmaceutical Sciences, University of Basel, Klingelbergstrasse 50, 4056 Basel, Switzerland
| | - Rodrigo Azevedo Loiola
- University of Artois, UR 2465, Laboratoire de la Barrière Hémato-Encéphalique (LBHE), Faculté des sciences Jean Perrin, Rue Jean Souvraz SP18, F-62300 Lens, France
| | - Emmanuel Sevin
- University of Artois, UR 2465, Laboratoire de la Barrière Hémato-Encéphalique (LBHE), Faculté des sciences Jean Perrin, Rue Jean Souvraz SP18, F-62300 Lens, France
| | - Thomas Exner
- Edelweiss Connect GmbH, Technology Park Basel, Hochbergerstrasse 60C, 4057 Basel, Switzerland
| | - Paul Jennings
- Division of Molecular and Computational Toxicology, Department of Chemistry and Pharmaceutical Sciences, Amsterdam Institute for Molecules, Medicines and Systems, Vrije Universiteit Amsterdam, De Boelelaan 1108, 1081HZ Amsterdam, the Netherlands
| | - Fabien Gosselet
- University of Artois, UR 2465, Laboratoire de la Barrière Hémato-Encéphalique (LBHE), Faculté des sciences Jean Perrin, Rue Jean Souvraz SP18, F-62300 Lens, France
| | - Maxime Culot
- University of Artois, UR 2465, Laboratoire de la Barrière Hémato-Encéphalique (LBHE), Faculté des sciences Jean Perrin, Rue Jean Souvraz SP18, F-62300 Lens, France.
| |
Collapse
|
17
|
Bao S, He C, Ku P, Xie M, Lin J, Lu S, Nie X. Effects of triclosan on the RedoximiRs/Sirtuin/Nrf2/ARE signaling pathway in mosquitofish (Gambusia affinis). AQUATIC TOXICOLOGY (AMSTERDAM, NETHERLANDS) 2021; 230:105679. [PMID: 33227666 DOI: 10.1016/j.aquatox.2020.105679] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Revised: 11/05/2020] [Accepted: 11/07/2020] [Indexed: 06/11/2023]
Abstract
Triclosan (TCS) has been widely used in daily life for its broad-spectrum antibacterial property and subsequently detected frequently in aquatic waterborne. Environmental relevant concentrations of TCS in water (ng-μg/L) may pose potential unexpected impact on non-target aquatic organisms. In the present work, we investigated the transcriptional responses of Nrf2 as well as its downstream genes, sirtuins and redox-sensitive microRNAs (RedoximiRs) in livers of the small freshwater fish mosquitofish (Gambusia affinis) which were exposed to environmental relevant concentrations of TCS (0.05 μg/L, 0.5 μg/L and 5 μg/L for 24 h and 168 h). Results showed there were similar up-regulations in Nrf2 and its target genes (e. g. NQO1, CAT and SOD) at transcriptional, enzymatic and protein levels, reflecting oxidative stress of TCS to mosquitofish. Meanwhile, up-regulations of Sirt1, Sirt2 and down-regulations of miR-34b, miR-200b-5p and miR-21 could modulate antioxidant system via the Nrf2/ARE signaling pathway by the post-transcriptional regulations. Some oxidative stress-related biomarkers displayed in concentration-dependent manners (e. g. NQO1 mRNA, CAT mRNA) and/or time-dependent manners (e. g. GSH contents). This study indicated that the RedoximiRs/Sirtuin/Nrf2/ARE signaling pathway played a crucial role in mosquitofish exposed to TCS, and there might be potentially profound effects for TCS on the aquatic ecological safety.
Collapse
Affiliation(s)
- Shuang Bao
- Department of Ecology/Hydrobiology Research Institute, Jinan University, Guangzhou, 510632, China
| | - Cuiping He
- Department of Ecology/Hydrobiology Research Institute, Jinan University, Guangzhou, 510632, China
| | - Peijia Ku
- Department of Biology, University of North Carolina at Greensboro, Greensboro, NC 27402, United States
| | - Meinan Xie
- Department of Ecology/Hydrobiology Research Institute, Jinan University, Guangzhou, 510632, China
| | - Jiawei Lin
- Department of Ecology/Hydrobiology Research Institute, Jinan University, Guangzhou, 510632, China
| | - Songhui Lu
- Research Center of Harmful Algae and Marine Biology, Jinan University, Guangzhou, 510632, China.
| | - Xiangping Nie
- Department of Ecology/Hydrobiology Research Institute, Jinan University, Guangzhou, 510632, China.
| |
Collapse
|
18
|
Kövesi B, Kulcsár S, Zándoki E, Szabó-Fodor J, Mézes M, Balogh K, Ancsin Z, Pelyhe C. Short-term effects of deoxynivalenol, T-2 toxin, fumonisin B1 or ochratoxin on lipid peroxidation and glutathione redox system and its regulatory genes in common carp (Cyprinus carpio L.) liver. FISH PHYSIOLOGY AND BIOCHEMISTRY 2020; 46:1921-1932. [PMID: 32617788 PMCID: PMC7584534 DOI: 10.1007/s10695-020-00845-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Accepted: 06/23/2020] [Indexed: 05/09/2023]
Abstract
The effects of a single oral dose of 1.82 mg kg-1 bw of T-2 and HT-2 toxin (T-2), 1.75 mg kg-1 bw deoxynivalenol (DON) and 15-acetyl DON, 1.96 mg kg-1 bw fumonisin B1 (FB1) or 1.85 mg kg-1 bw ochratoxin A (OTA) were investigated in common carp juveniles on lipid peroxidation, the parameters of the glutathione redox system including the expression of their encoding genes in a short-term (24 h) experiment. Markers of the initiation phase of lipid peroxidation, conjugated dienes, and trienes, were slightly affected by DON and OTA treatment at 16-h sampling. The termination marker, malondialdehyde, concentration increased only as an effect of FB1. Glutathione content and glutathione peroxidase activity showed significantly higher levels in the T-2 and FB1 groups at 8 h, and in the DON and FB1 groups at 16 h. The expression of glutathione peroxidase genes (gpx4a, gpx4b) showed a dual response. Downregulation of gpxa was observed at 8 h, as the effect of DON, FB1, and OTA, but an upregulation in the T-2 group. At 16 h gpx4a upregulated as an effect of DON, T-2, and FB1, and at 24 h in the DON and T-2 groups. Expression of gpx4b downregulated at 8 h, except in the T-2 group, and upregulation observed as an effect of T-2 at 24 h. The lack of an increase in the expression of nrf2, except as the effect of DON at 8 h, and a decrease in the keap1 expression suggests that the antioxidant defence system was activated at gene and protein levels through Keap1-Nrf2 independent pathways.
Collapse
Affiliation(s)
- Benjámin Kövesi
- Department of Nutrition, Szent István University, Gödöllő, H-2103, Hungary
| | - Szabina Kulcsár
- Department of Nutrition, Szent István University, Gödöllő, H-2103, Hungary
| | - Erika Zándoki
- Hungarian Academy of Sciences-Kaposvár University- Szent István University, Mycotoxins in the Food Chain Research Group, Kaposvár, H-7400, Hungary
| | - Judit Szabó-Fodor
- Hungarian Academy of Sciences-Kaposvár University- Szent István University, Mycotoxins in the Food Chain Research Group, Kaposvár, H-7400, Hungary
| | - Miklós Mézes
- Department of Nutrition, Szent István University, Gödöllő, H-2103, Hungary.
- Hungarian Academy of Sciences-Kaposvár University- Szent István University, Mycotoxins in the Food Chain Research Group, Kaposvár, H-7400, Hungary.
| | - Krisztián Balogh
- Department of Nutrition, Szent István University, Gödöllő, H-2103, Hungary
- Hungarian Academy of Sciences-Kaposvár University- Szent István University, Mycotoxins in the Food Chain Research Group, Kaposvár, H-7400, Hungary
| | - Zsolt Ancsin
- Department of Nutrition, Szent István University, Gödöllő, H-2103, Hungary
| | - Csilla Pelyhe
- Department of Nutrition, Szent István University, Gödöllő, H-2103, Hungary
| |
Collapse
|
19
|
Damiano S, Iovane V, Squillacioti C, Mirabella N, Prisco F, Ariano A, Amenta M, Giordano A, Florio S, Ciarcia R. Red orange and lemon extract prevents the renal toxicity induced by ochratoxin A in rats. J Cell Physiol 2020; 235:5386-5393. [PMID: 31898818 DOI: 10.1002/jcp.29425] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Accepted: 12/19/2019] [Indexed: 12/11/2022]
Abstract
In this work, we investigated the effects of red orange and lemon extract (RLE) on ochratoxin A (OTA)-induced nephrotoxicity. In particular, we analyzed the change in renal function and oxidative stress in Sprague-Dawley rats treated with OTA (0.5 mg/kg body weight, b.w.) and with RLE (90 mg/kg b.w.) by oral administration. After OTA treatment, we found alterations of biochemical and oxidative stress parameters in the kidney, related to a severe decrease of glomerular filtration rate. The RLE treatment normalized the activity of antioxidant enzymes and prevented the glomerular hyperfiltration. Histopathological examinations revealed glomerular damages and kidney cortex fibrosis in OTA-rats, while we observed less severe fibrosis in OTA plus RLE group. Then, we demonstrated that oxidative stress could be the cause of OTA renal injury and that RLE reduces this effect.
Collapse
Affiliation(s)
- Sara Damiano
- Department of Veterinary Medicine and Animal Productions, University of Naples, Napoli, Italy
| | | | - Caterina Squillacioti
- Department of Veterinary Medicine and Animal Productions, University of Naples, Napoli, Italy
| | - Nicola Mirabella
- Department of Veterinary Medicine and Animal Productions, University of Naples, Napoli, Italy
| | - Francesco Prisco
- Department of Veterinary Medicine and Animal Productions, University of Naples, Napoli, Italy
| | - Andrea Ariano
- Department of Veterinary Medicine and Animal Productions, University of Naples, Napoli, Italy
| | - Margherita Amenta
- Council for Agricultural Research and Economics, Research Centre for Olive, Citrus and Tree Fruit, Acireale, Italy
| | - Antonio Giordano
- Department of Medical Biotechnologies, University of Siena, Siena, Italy.,Sbarro Institute for Cancer Research and Molecular Medicine, Center of Biotechnology, College of Science and Technology, Temple University, Philadelphia, Pennsylvania
| | - Salvatore Florio
- Department of Veterinary Medicine and Animal Productions, University of Naples, Napoli, Italy
| | - Roberto Ciarcia
- Department of Veterinary Medicine and Animal Productions, University of Naples, Napoli, Italy
| |
Collapse
|
20
|
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: 24.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2019] [Accepted: 11/05/2019] [Indexed: 12/19/2022]
|
21
|
Josyula N, Andersen ME, Kaminski NE, Dere E, Zacharewski TR, Bhattacharya S. Gene co-regulation and co-expression in the aryl hydrocarbon receptor-mediated transcriptional regulatory network in the mouse liver. Arch Toxicol 2019; 94:113-126. [PMID: 31728591 DOI: 10.1007/s00204-019-02620-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2019] [Accepted: 11/06/2019] [Indexed: 01/16/2023]
Abstract
Four decades after its discovery, the aryl hydrocarbon receptor (AHR), a ligand-inducible transcription factor (TF) activated by the persistent environmental contaminant 2,3,7,8-Tetrachlorodibenzo-p-dioxin (TCDD), remains an enigmatic molecule with a controversial endogenous role. Here, we have assembled a global map of the AHR gene regulatory network in female C57BL/6 mice orally gavaged with 30 µg/kg of TCDD from a combination of previously published gene expression and genome-wide TF-binding data sets. Using Kohonen self-organizing maps and subspace clustering, we show that genes co-regulated by common upstream TFs in the AHR network exhibit a pattern of co-expression. Directly bound, indirectly bound, and non-genomic AHR target genes exhibit distinct expression patterns, with the directly bound targets associated with highest median expression. Interestingly, among the directly bound AHR target genes, the expression level increases with the number of AHR-binding sites in the proximal promoter regions. Finally, we show that co-regulated genes in the AHR network activate distinct groups of downstream biological processes. Although the specific findings described here are restricted to hepatic effects under short-term TCDD exposure, this work describes a generalizable approach to the reconstruction and analysis of transcriptional regulatory cascades underlying cellular stress response, revealing network hierarchy and the nature of information flow from the initial signaling events to phenotypic outcomes. Such reconstructed networks can form the basis of a new generation of quantitative adverse outcome pathways.
Collapse
Affiliation(s)
- Navya Josyula
- Biomedical and Translational Informatics Program, Geisinger Health System, Rockville, MD, 20850, USA
| | | | - Norbert E Kaminski
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, MI, 48824, USA.,Institute for Integrative Toxicology, Michigan State University, East Lansing, MI, 48824, USA.,Center for Research on Ingredient Safety, Michigan State University, East Lansing, MI, 48824, USA
| | - Edward Dere
- Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, MI, 48824, USA.,Genentech, South San Francisco, CA, 94080, USA
| | - Timothy R Zacharewski
- Institute for Integrative Toxicology, Michigan State University, East Lansing, MI, 48824, USA.,Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, MI, 48824, USA
| | - Sudin Bhattacharya
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, MI, 48824, USA. .,Institute for Integrative Toxicology, Michigan State University, East Lansing, MI, 48824, USA. .,Department of Biomedical Engineering, Michigan State University, East Lansing, MI, 48824-1226, USA. .,Center for Research on Ingredient Safety, Michigan State University, East Lansing, MI, 48824, USA. .,Institute for Quantitative Health Science and Engineering, Michigan State University, East Lansing, MI, 48824, USA.
| |
Collapse
|
22
|
Rivetti C, Allen TEH, Brown JB, Butler E, Carmichael PL, Colbourne JK, Dent M, Falciani F, Gunnarsson L, Gutsell S, Harrill JA, Hodges G, Jennings P, Judson R, Kienzler A, Margiotta-Casaluci L, Muller I, Owen SF, Rendal C, Russell PJ, Scott S, Sewell F, Shah I, Sorrel I, Viant MR, Westmoreland C, White A, Campos B. Vision of a near future: Bridging the human health-environment divide. Toward an integrated strategy to understand mechanisms across species for chemical safety assessment. Toxicol In Vitro 2019; 62:104692. [PMID: 31669395 DOI: 10.1016/j.tiv.2019.104692] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2019] [Revised: 09/25/2019] [Accepted: 10/14/2019] [Indexed: 12/31/2022]
Abstract
There is a growing recognition that application of mechanistic approaches to understand cross-species shared molecular targets and pathway conservation in the context of hazard characterization, provide significant opportunities in risk assessment (RA) for both human health and environmental safety. Specifically, it has been recognized that a more comprehensive and reliable understanding of similarities and differences in biological pathways across a variety of species will better enable cross-species extrapolation of potential adverse toxicological effects. Ultimately, this would also advance the generation and use of mechanistic data for both human health and environmental RA. A workshop brought together representatives from industry, academia and government to discuss how to improve the use of existing data, and to generate new NAMs data to derive better mechanistic understanding between humans and environmentally-relevant species, ultimately resulting in holistic chemical safety decisions. Thanks to a thorough dialogue among all participants, key challenges, current gaps and research needs were identified, and potential solutions proposed. This discussion highlighted the common objective to progress toward more predictive, mechanistically based, data-driven and animal-free chemical safety assessments. Overall, the participants recognized that there is no single approach which would provide all the answers for bridging the gap between mechanism-based human health and environmental RA, but acknowledged we now have the incentive, tools and data availability to address this concept, maximizing the potential for improvements in both human health and environmental RA.
Collapse
Affiliation(s)
- Claudia Rivetti
- Unilever, Safety and Environmental Assurance Centre, Colworth Science Park, Sharnbrook, Bedfordshire MK44 1LQ, United Kingdom
| | - Timothy E H Allen
- Centre for Molecular Informatics, Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge CB2 1EW, United Kingdom
| | - James B Brown
- Department of Genome Dynamics Lawrence Berkeley National Laboratory, University of California Berkeley, Berkeley, California 94720, USA
| | - Emma Butler
- Unilever, Safety and Environmental Assurance Centre, Colworth Science Park, Sharnbrook, Bedfordshire MK44 1LQ, United Kingdom
| | - Paul L Carmichael
- Unilever, Safety and Environmental Assurance Centre, Colworth Science Park, Sharnbrook, Bedfordshire MK44 1LQ, United Kingdom
| | - John K Colbourne
- School of Biosciences, University of Birmingham, Birmingham B15 2TT, United Kingdom
| | - Matthew Dent
- Unilever, Safety and Environmental Assurance Centre, Colworth Science Park, Sharnbrook, Bedfordshire MK44 1LQ, United Kingdom
| | - Francesco Falciani
- Institute for Integrative Biology, University of Liverpool, L69 7ZB Liverpool, United Kingdom
| | - Lina Gunnarsson
- Biosciences, College of Life and Environmental Sciences, University of Exeter, Geoffrey Pope, Stocker Road, Exeter, Devon EX4 4QD, United Kingdom
| | - Steve Gutsell
- Unilever, Safety and Environmental Assurance Centre, Colworth Science Park, Sharnbrook, Bedfordshire MK44 1LQ, United Kingdom
| | - Joshua A Harrill
- National Center for Computational Toxicology, Office of Research & Development, U.S. Environmental Protection Agency, Mail Code B205-01, Research Triangle Park, Durham, North Carolina 27711, USA
| | - Geoff Hodges
- Unilever, Safety and Environmental Assurance Centre, Colworth Science Park, Sharnbrook, Bedfordshire MK44 1LQ, United Kingdom
| | - Paul Jennings
- Division of Molecular and Computational Toxicology, Amsterdam Institute for Molecules, Medicines and Systems, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | - Richard Judson
- National Center for Computational Toxicology, Office of Research & Development, U.S. Environmental Protection Agency, Mail Code B205-01, Research Triangle Park, Durham, North Carolina 27711, USA
| | - Aude Kienzler
- European Commission, Joint Research Centre (JRC), Ispra, VA, Italy
| | | | - Iris Muller
- Unilever, Safety and Environmental Assurance Centre, Colworth Science Park, Sharnbrook, Bedfordshire MK44 1LQ, United Kingdom
| | - Stewart F Owen
- AstraZeneca, Alderley Park, Macclesfield, Cheshire SK10 4TF, United Kingdom
| | - Cecilie Rendal
- Unilever, Safety and Environmental Assurance Centre, Colworth Science Park, Sharnbrook, Bedfordshire MK44 1LQ, United Kingdom
| | - Paul J Russell
- Unilever, Safety and Environmental Assurance Centre, Colworth Science Park, Sharnbrook, Bedfordshire MK44 1LQ, United Kingdom
| | - Sharon Scott
- Unilever, Safety and Environmental Assurance Centre, Colworth Science Park, Sharnbrook, Bedfordshire MK44 1LQ, United Kingdom
| | - Fiona Sewell
- NC3Rs, Gibbs Building, 215 Euston Road, London NW1 2BE, United Kingdom
| | - Imran Shah
- National Center for Computational Toxicology, Office of Research & Development, U.S. Environmental Protection Agency, Mail Code B205-01, Research Triangle Park, Durham, North Carolina 27711, USA
| | - Ian Sorrel
- Unilever, Safety and Environmental Assurance Centre, Colworth Science Park, Sharnbrook, Bedfordshire MK44 1LQ, United Kingdom
| | - Mark R Viant
- School of Biosciences, University of Birmingham, Birmingham B15 2TT, United Kingdom
| | - Carl Westmoreland
- Unilever, Safety and Environmental Assurance Centre, Colworth Science Park, Sharnbrook, Bedfordshire MK44 1LQ, United Kingdom
| | - Andrew White
- Unilever, Safety and Environmental Assurance Centre, Colworth Science Park, Sharnbrook, Bedfordshire MK44 1LQ, United Kingdom
| | - Bruno Campos
- Unilever, Safety and Environmental Assurance Centre, Colworth Science Park, Sharnbrook, Bedfordshire MK44 1LQ, United Kingdom.
| |
Collapse
|
23
|
Li HH, Yauk CL, Chen R, Hyduke DR, Williams A, Frötschl R, Ellinger-Ziegelbauer H, Pettit S, Aubrecht J, Fornace AJ. TGx-DDI, a Transcriptomic Biomarker for Genotoxicity Hazard Assessment of Pharmaceuticals and Environmental Chemicals. Front Big Data 2019; 2:36. [PMID: 33693359 PMCID: PMC7931968 DOI: 10.3389/fdata.2019.00036] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2019] [Accepted: 09/17/2019] [Indexed: 01/27/2023] Open
Abstract
Genotoxicity testing is an essential component of the safety assessment paradigm required by regulatory agencies world-wide for analysis of drug candidates, and environmental and industrial chemicals. Current genotoxicity testing batteries feature a high incidence of irrelevant positive findings—particularly for in vitro chromosomal damage (CD) assays. The risk management of compounds with positive in vitro findings is a major challenge and requires complex, time consuming, and costly follow-up strategies including animal testing. Thus, regulators are urgently in need of new testing approaches to meet legislated mandates. Using machine learning, we identified a set of transcripts that responds predictably to DNA-damage in human cells that we refer to as the TGx-DDI biomarker, which was originally referred to as TGx-28.65. We proposed to use this biomarker in conjunction with current genotoxicity testing batteries to differentiate compounds with irrelevant “false” positive findings in the in vitro CD assays from true DNA damaging agents (i.e., for de-risking agents that are clastogenic in vitro but not in vivo). We validated the performance of the TGx-DDI biomarker to identify true DNA damaging agents, assessed intra- and inter- laboratory reproducibility, and cross-platform performance. Recently, to augment the application of this biomarker, we developed a high-throughput cell-based genotoxicity testing system using the NanoString nCounter® technology. Here, we review the status of TGx-DDI development, its integration in the genotoxicity testing paradigm, and progress to date in its qualification at the US Food and Drug Administration (FDA) as a drug development tool. If successfully validated and implemented, the TGx-DDI biomarker assay is expected to significantly augment the current strategy for the assessment of genotoxic hazards for drugs and chemicals.
Collapse
Affiliation(s)
- Heng-Hong Li
- Department of Oncology, Department of Biochemistry and Molecular & Cellular Biology, Georgetown University Medical Center, Washington, DC, United States
| | - Carole L Yauk
- Environmental Health Science and Research Bureau, Health Canada, Ottawa, ON, Canada
| | - Renxiang Chen
- Department of Oncology, Department of Biochemistry and Molecular & Cellular Biology, Georgetown University Medical Center, Washington, DC, United States.,Amelia Technologies LLC, Rockville, MD, United States
| | - Daniel R Hyduke
- Department of Oncology, Department of Biochemistry and Molecular & Cellular Biology, Georgetown University Medical Center, Washington, DC, United States
| | - Andrew Williams
- Environmental Health Science and Research Bureau, Health Canada, Ottawa, ON, Canada
| | - Roland Frötschl
- Federal Institute for Drugs and Medical Devices, Bonn, Germany
| | | | - Syril Pettit
- Health and Environmental Sciences Institute, Washington, DC, United States
| | - Jiri Aubrecht
- Department of Oncology, Department of Biochemistry and Molecular & Cellular Biology, Georgetown University Medical Center, Washington, DC, United States
| | - Albert J Fornace
- Department of Oncology, Department of Biochemistry and Molecular & Cellular Biology, Georgetown University Medical Center, Washington, DC, United States
| |
Collapse
|
24
|
High-throughput confocal imaging of differentiated 3D liver-like spheroid cellular stress response reporters for identification of drug-induced liver injury liability. Arch Toxicol 2019; 93:2895-2911. [DOI: 10.1007/s00204-019-02552-0] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2018] [Accepted: 08/22/2019] [Indexed: 12/27/2022]
|
25
|
Qi L, Luo Q, Zhang Y, Jia F, Zhao Y, Wang F. Advances in Toxicological Research of the Anticancer Drug Cisplatin. Chem Res Toxicol 2019; 32:1469-1486. [PMID: 31353895 DOI: 10.1021/acs.chemrestox.9b00204] [Citation(s) in RCA: 219] [Impact Index Per Article: 43.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Cisplatin is one of the most widely used chemotherapeutic agents for various solid tumors in the clinic due to its high efficacy and broad spectrum. The antineoplastic activity of cisplatin is mainly due to its ability to cross-link with DNA, thus blocking transcription and replication. Unfortunately, the clinical use of cisplatin is limited by its severe, dose-dependent toxic side effects. There are approximately 40 specific toxicities of cisplatin, among which nephrotoxicity is the most common one. Other common side effects include ototoxicity, neurotoxicity, gastrointestinal toxicity, hematological toxicity, cardiotoxicity, and hepatotoxicity. These side effects together reduce the life quality of patients and require lowering the dosage of the drug, even stopping administration, thus weakening the treatment effect. Few effective measures exist clinically against these side effects because the exact mechanisms of various side effects from cisplatin remain still unclear. Therefore, substantial effort has been made to explore the complicated biochemical processes involved in the toxicology of cisplatin, aiming to identify effective ways to reduce or eradicate its toxicity. This review summarizes and reviews the updated advances in the toxicological research of cisplatin. We anticipate to provide insights into the understanding of the mechanisms underlying the side effects of cisplatin and designing comprehensive therapeutic strategies involving cisplatin.
Collapse
Affiliation(s)
- Luyu Qi
- Beijing National Laboratory for Molecular Sciences; National Centre for Mass Spectrometry in Beijing; CAS Key Laboratory of Analytical Chemistry for Living Biosystems , Institute of Chemistry, Chinese Academy of Sciences , Beijing 100190 , P.R. China.,University of Chinese Academy of Sciences , Beijing 100049 , P.R. China
| | - Qun Luo
- Beijing National Laboratory for Molecular Sciences; National Centre for Mass Spectrometry in Beijing; CAS Key Laboratory of Analytical Chemistry for Living Biosystems , Institute of Chemistry, Chinese Academy of Sciences , Beijing 100190 , P.R. China.,University of Chinese Academy of Sciences , Beijing 100049 , P.R. China
| | - Yanyan Zhang
- Beijing National Laboratory for Molecular Sciences; National Centre for Mass Spectrometry in Beijing; CAS Key Laboratory of Analytical Chemistry for Living Biosystems , Institute of Chemistry, Chinese Academy of Sciences , Beijing 100190 , P.R. China
| | - Feifei Jia
- Beijing National Laboratory for Molecular Sciences; National Centre for Mass Spectrometry in Beijing; CAS Key Laboratory of Analytical Chemistry for Living Biosystems , Institute of Chemistry, Chinese Academy of Sciences , Beijing 100190 , P.R. China
| | - Yao Zhao
- Beijing National Laboratory for Molecular Sciences; National Centre for Mass Spectrometry in Beijing; CAS Key Laboratory of Analytical Chemistry for Living Biosystems , Institute of Chemistry, Chinese Academy of Sciences , Beijing 100190 , P.R. China
| | - Fuyi Wang
- Beijing National Laboratory for Molecular Sciences; National Centre for Mass Spectrometry in Beijing; CAS Key Laboratory of Analytical Chemistry for Living Biosystems , Institute of Chemistry, Chinese Academy of Sciences , Beijing 100190 , P.R. China.,University of Chinese Academy of Sciences , Beijing 100049 , P.R. China.,Basic Medical College , Shandong University of Chinese Traditional Medicine , Jinan 250355 , P.R. China
| |
Collapse
|
26
|
Zgheib E, Gao W, Limonciel A, Aladjov H, Yang H, Tebby C, Gayraud G, Jennings P, Sachana M, Beltman JB, Bois FY. Application of three approaches for quantitative AOP development to renal toxicity. ACTA ACUST UNITED AC 2019. [DOI: 10.1016/j.comtox.2019.02.001] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|
27
|
Zhernovkov V, Santra T, Cassidy H, Rukhlenko O, Matallanas D, Krstic A, Kolch W, Lobaskin V, Kholodenko BN. An integrative computational approach for a prioritization of key transcription regulators associated with nanomaterial-induced toxicity. Toxicol Sci 2019; 171:303-314. [PMID: 31271423 DOI: 10.1093/toxsci/kfz151] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2018] [Revised: 06/27/2019] [Accepted: 06/28/2019] [Indexed: 12/19/2022] Open
Abstract
A rapid increase of new nanomaterial products poses new challenges for their risk assessment. Current traditional methods for estimating potential adverse health effect of nanomaterials (NMs) are complex, time consuming and expensive. In order to develop new prediction tests for nanotoxicity evaluation, a systems biology approach and data from high-throughput omics experiments can be used. We present a computational approach that combines reverse engineering techniques, network analysis and pathway enrichment analysis for inferring the transcriptional regulation landscape and its functional interpretation. To illustrate this approach, we used published transcriptomic data derived from mice lung tissue exposed to carbon nanotubes (NM-401 and NRCWE-26). Because fibrosis is the most common adverse effect of these NMs, we included in our analysis the data for bleomycin (BLM) treatment, which is a well-known fibrosis inducer. We inferred gene regulatory networks for each NM and BLM to capture functional hierarchical regulatory structures between genes and their regulators. Despite the different nature of the lung injury caused by nanoparticles and BLM, we identified several conserved core regulators for all agents. We reason that these regulators can be considered as early predictors of toxic responses after NMs exposure. This integrative approach, which refines traditional methods of transcriptomic analysis, can be useful for prioritization of potential core regulators and generation of new hypothesis about mechanisms of nanoparticles toxicity.
Collapse
Affiliation(s)
- Vadim Zhernovkov
- Systems Biology Ireland, University College Dublin, Dublin 4, Ireland
| | - Tapesh Santra
- Systems Biology Ireland, University College Dublin, Dublin 4, Ireland
| | - Hilary Cassidy
- Systems Biology Ireland, University College Dublin, Dublin 4, Ireland
| | - Oleksii Rukhlenko
- Systems Biology Ireland, University College Dublin, Dublin 4, Ireland
| | - David Matallanas
- Systems Biology Ireland, University College Dublin, Dublin 4, Ireland.,School of Medicine and Medical Science, University College Dublin, Belfield, Dublin 4, Ireland
| | - Aleksandar Krstic
- Systems Biology Ireland, University College Dublin, Dublin 4, Ireland
| | - Walter Kolch
- Systems Biology Ireland, University College Dublin, Dublin 4, Ireland.,School of Medicine and Medical Science, University College Dublin, Belfield, Dublin 4, Ireland.,Conway Institute of Biomolecular & Biomedical Research, University College Dublin, Ireland
| | | | - Boris N Kholodenko
- Systems Biology Ireland, University College Dublin, Dublin 4, Ireland.,School of Medicine and Medical Science, University College Dublin, Belfield, Dublin 4, Ireland.,Conway Institute of Biomolecular & Biomedical Research, University College Dublin, Ireland.,Department of Pharmacology, Yale University School of Medicine, New Haven CT, USA
| |
Collapse
|
28
|
Guéguen Y, Bontemps A, Ebrahimian TG. Adaptive responses to low doses of radiation or chemicals: their cellular and molecular mechanisms. Cell Mol Life Sci 2019; 76:1255-1273. [PMID: 30535789 PMCID: PMC11105647 DOI: 10.1007/s00018-018-2987-5] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2018] [Revised: 11/09/2018] [Accepted: 12/03/2018] [Indexed: 12/17/2022]
Abstract
This article reviews the current knowledge on the mechanisms of adaptive response to low doses of ionizing radiation or chemical exposure. A better knowledge of these mechanisms is needed to improve our understanding of health risks at low levels of environmental or occupational exposure and their involvement in cancer or non-cancer diseases. This response is orchestrated through a multifaceted cellular program involving the concerted action of diverse stress response pathways. These evolutionary highly conserved defense mechanisms determine the cellular response to chemical and physical aggression. They include DNA damage repair (p53, ATM, PARP pathways), antioxidant response (Nrf2 pathway), immune/inflammatory response (NF-κB pathway), cell survival/death pathway (apoptosis), endoplasmic response to stress (UPR response), and other cytoprotective processes including autophagy, cell cycle regulation, and the unfolded protein response. The coordinated action of these processes induced by low-dose radiation or chemicals produces biological effects that are currently estimated with the linear non-threshold model. These effects are controversial. They are difficult to detect because of their low magnitude, the scarcity of events in humans, and the difficulty of corroborating associations over the long term. Improving our understanding of these biological consequences should help humans and their environment by enabling better risk estimates, the revision of radiation protection standards, and possible therapeutic advances.
Collapse
Affiliation(s)
- Yann Guéguen
- Institut de Radioprotection et de Sureté Nucléaire (IRSN), PSE-SANTE, SESANE, LRTOX, B.P. no 17, 92262, Fontenay-aux-Roses Cedex, France.
- Institut de Radioprotection et de Sureté Nucléaire (IRSN), PSE-SANTE, SESANE, LRSI, Fontenay-aux-Roses, France.
| | - Alice Bontemps
- Institut de Radioprotection et de Sureté Nucléaire (IRSN), PSE-SANTE, SESANE, LRTOX, B.P. no 17, 92262, Fontenay-aux-Roses Cedex, France
| | - Teni G Ebrahimian
- Institut de Radioprotection et de Sureté Nucléaire (IRSN), PSE-SANTE, SESANE, LRTOX, B.P. no 17, 92262, Fontenay-aux-Roses Cedex, France
| |
Collapse
|
29
|
Wang Z, Xu X, He B, Guo J, Zhao B, Zhang Y, Zhou Z, Zhou X, Zhang R, Abliz Z. The impact of chronic environmental metal and benzene exposure on human urinary metabolome among Chinese children and the elderly population. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2019; 169:232-239. [PMID: 30448706 DOI: 10.1016/j.ecoenv.2018.11.016] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/25/2018] [Revised: 11/01/2018] [Accepted: 11/05/2018] [Indexed: 06/09/2023]
Abstract
The health effects of metals and benzene exposure have been extensively investigated; however, information on the impact of chronic environmental metal and benzene exposure on human urinary metabolome is limited. In this study, a total of 566 participants, including 352 elderly and 214 children, were split into the "exposed" and "control" groups. The urine samples of all the participants were collected and stored at - 80 °C until analysis. The urinary levels of 17 metals and S-phenylmercapturic acid (S-PMA) were determined by the ICP-MS and LC-MS/MS methods to comprehensively assess the personal metal and benzene exposure levels, respectively. Then, the individual levels of metal and benzene exposure were correlated to the metabolic consequences of ambient pollutant exposure, which were previously observed in our metabolomics study. As a result, multiple metals, including Cd, Co, Cr, Cu, Fe, Hg, Li, Mo, Ni, Pb, Se, and Zn, exhibited a significant linear dose-dependent association with one or more urinary metabolites, including two amino acids (pyroglutamic acid and 3-methylhistidine), three organic acids (azelaic acid, decenedioic acid, and hydroxytetradecanedioic acid), ten medium-chainacylcarnitines (heptenedioylcarnitine, octenedioylcarnitine, nonenedioylcarnitine, decenedioylglucuronide, 3-hydroxydecanoylcarnitine, dodecanedioylcarnitine, nonanoylcarnitine, decadienylcarnitine, hydroxydodecenoylcarnitine, dodecadienylcarnitine, and dodecenoylcarnitine), and one glucuronide conjugate (decenedioylglucuronide). These observations indicate that the increased environmental metal exposure has caused various oxidative stress-related effects, including the depletion of antioxidants, accelerated muscle proteolysis, elevated activity of UGTs, increased lipid peroxidation, and the disorder of mitochondrial lipid metabolism among exposed children and the elderly. The current study provides new insights into the biological effects induced by metal exposure in the environment.
Collapse
Affiliation(s)
- Zhonghua Wang
- Center for Imaging and Systems Biology, College of Life and Environmental Sciences, Minzu University of China, 27 Zhongguancun South Avenue, Beijing 100081, China
| | - Xiaoyu Xu
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of MateriaMedica, Chinese Academy of Medical Sciences and Peking Union Medical College, 1 Xiannongtan Street, Beijing 100050, China
| | - Bingshu He
- Center for Imaging and Systems Biology, College of Life and Environmental Sciences, Minzu University of China, 27 Zhongguancun South Avenue, Beijing 100081, China
| | - Jinghua Guo
- Analytical and Testing Center, Beijing Normal University, Beijing 100875, China
| | - Baoxin Zhao
- Taiyuan Centre for Disease Control and Prevention, Taiyuan 030012, China
| | - Yanping Zhang
- Taiyuan Centre for Disease Control and Prevention, Taiyuan 030012, China
| | - Zhi Zhou
- Center for Imaging and Systems Biology, College of Life and Environmental Sciences, Minzu University of China, 27 Zhongguancun South Avenue, Beijing 100081, China
| | - Xia Zhou
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of MateriaMedica, Chinese Academy of Medical Sciences and Peking Union Medical College, 1 Xiannongtan Street, Beijing 100050, China
| | - Ruiping Zhang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of MateriaMedica, Chinese Academy of Medical Sciences and Peking Union Medical College, 1 Xiannongtan Street, Beijing 100050, China.
| | - Zeper Abliz
- Center for Imaging and Systems Biology, College of Life and Environmental Sciences, Minzu University of China, 27 Zhongguancun South Avenue, Beijing 100081, China; State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of MateriaMedica, Chinese Academy of Medical Sciences and Peking Union Medical College, 1 Xiannongtan Street, Beijing 100050, China.
| |
Collapse
|
30
|
Pelyhe C, Kövesi B, Szabó-Fodor J, Zándoki E, Erdélyi M, Kovács B, Mézes M, Balogh K. Age-dependent effects of short-term exposure of T-2 toxin or deoxynivalenol on lipid peroxidation and glutathione redox system in broiler chickens. WORLD MYCOTOXIN J 2018. [DOI: 10.3920/wmj2018.2325] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Purpose of this study was to investigate the age-dependent, short-term effects of T-2 toxin (5.77 mg T-2 and 1.40 mg HT-2 toxin/kg feed) or deoxynivalenol (DON) (4.86 mg DON and 1.39 mg 15-acetyl-DON/kg feed) in one and three weeks old broiler chicken to observe the changes in parameters of lipid peroxidation, glutathione redox system, and expression of genes related to glutathione redox system in the first 24 h of mycotoxin exposure. Glutathione-redox system responsed to T-2 toxin exposure in both age groups for T-2 toxin in the first 8 h of exposure, while a reactivation was observed in the 3-week-old group after 20 h, although lipid peroxidation did not change significantly. DON did not alter these parameters, only at gene expression level. Gene expression of phospholipid hydroperoxide glutathione peroxidase (GPX4) showed minor, but significant, changes in both age- and mycotoxin exposure groups. Glutathione reductase (GSR) showed a dual response for the mycotoxin exposure, which was not consequent in either age groups, or treatments. Glutathione synthetase (GSS) showed a decreasing tendency in the younger animals while in the older group elevating tendency was observed as effect of both mycotoxins. Time, treatment and their combined effect also showed relation with the changes in the parameters.
Collapse
Affiliation(s)
- Cs. Pelyhe
- Hungarian Academy of Sciences, Kaposvár University – Szent István University ‘MTA-KE-SZIE Mycotoxins in the Food Chain’ Research Group, Guba S. u. 40, Kaposvár 7400, Hungary
| | - B. Kövesi
- Department of Nutrition, Szent István University, Páter K. u. 1, Gödöllő 2103, Hungary
| | - J. Szabó-Fodor
- Hungarian Academy of Sciences, Kaposvár University – Szent István University ‘MTA-KE-SZIE Mycotoxins in the Food Chain’ Research Group, Guba S. u. 40, Kaposvár 7400, Hungary
| | - E. Zándoki
- Hungarian Academy of Sciences, Kaposvár University – Szent István University ‘MTA-KE-SZIE Mycotoxins in the Food Chain’ Research Group, Guba S. u. 40, Kaposvár 7400, Hungary
| | - M. Erdélyi
- Department of Nutrition, Szent István University, Páter K. u. 1, Gödöllő 2103, Hungary
| | - B. Kovács
- Department of Aquaculture, Szent István University, Páter K. u. 1, Gödöllő 2103, Hungary
| | - M. Mézes
- Department of Nutrition, Szent István University, Páter K. u. 1, Gödöllő 2103, Hungary
| | - K. Balogh
- Department of Nutrition, Szent István University, Páter K. u. 1, Gödöllő 2103, Hungary
| |
Collapse
|
31
|
Souza T, Trairatphisan P, Piñero J, Furlong LI, Saez-Rodriguez J, Kleinjans J, Jennen D. Embracing the Dark Side: Computational Approaches to Unveil the Functionality of Genes Lacking Biological Annotation in Drug-Induced Liver Injury. Front Genet 2018; 9:527. [PMID: 30515189 PMCID: PMC6255978 DOI: 10.3389/fgene.2018.00527] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2018] [Accepted: 10/19/2018] [Indexed: 12/03/2022] Open
Abstract
In toxicogenomics, functional annotation is an important step to gain additional insights into genes with aberrant expression that drive pathophysiological mechanisms. Nevertheless, there exists a gap on annotation of these genes which often hampers the interpretation of results and limits their applicability in translational medicine. In this study, we evaluated the coverage of functional annotations of differentially expressed genes (DEGs) induced by 10 selected compounds from the TG-GATEs database identified as high- or no-risk in causing drug-induced liver injury (most-DILI or no-DILI, respectively) using in vitro human data. Functional roles of DEGs not present in the most common biological annotation databases – termed “dark genes” – were unveiled via literature mining and via the identification of shared regulatory transcription factors or signaling pathways. Our results demonstrated that there were approximately 13% of dark genes induced by these compounds in vitro and we were able to obtain additional relevant information for up to 76% of those. Using interactome data from several sources, we have uncovered genes such as LRBA, and WDR26 as highly connected in the protein network that play roles in drug response. Genes such as MALAT1, H19, and MIR29C – whose links to hepatotoxicity have been confirmed – were identified as markers for the most-DILI group and appeared as top hits across all literature-based mining methods. Furthermore, we investigated the potential impact of dark genes on liver toxicity by identifying their rat orthologs in combination with their correlation to drug-induced liver pathologies observed in vivo following chemical exposure. We identified a set of important regulatory transcription factors of dark genes for all most-DILI compounds including E2F1 and JUND with supporting evidences in literature and we found Magee1 correlated with chemically induced bile duct hyperplasia and adverse responses at 29 days in rats in vivo. In conclusion, in this study we show the potential role of these poorly annotated genes in mechanisms underlying hepatotoxicity and offer a number of computational approaches that may help to minimize current gaps in gene annotation and highlight their values as potential biomarkers in toxicological studies.
Collapse
Affiliation(s)
- Terezinha Souza
- Department of Toxicogenomics, GROW School for Oncology and Developmental Biology, Maastricht University, Maastricht, Netherlands
| | - Panuwat Trairatphisan
- Joint Research Center for Computational Biomedicine (JRC-COMBINE), Faculty of Medicine, RWTH Aachen University, Aachen, Germany
| | - Janet Piñero
- Integrative Biomedical Informatics Group, Research Programme on Biomedical Informatics (GRIB), Department of Experimental and Health Sciences (DCEXS), Hospital del Mar Medical Research Institute (IMIM), Universitat Pompeu Fabra, Barcelona, Spain
| | - Laura I Furlong
- Integrative Biomedical Informatics Group, Research Programme on Biomedical Informatics (GRIB), Department of Experimental and Health Sciences (DCEXS), Hospital del Mar Medical Research Institute (IMIM), Universitat Pompeu Fabra, Barcelona, Spain
| | - Julio Saez-Rodriguez
- Joint Research Center for Computational Biomedicine (JRC-COMBINE), Faculty of Medicine, RWTH Aachen University, Aachen, Germany.,European Bioinformatics Institute, European Molecular Biology Laboratory (EMBL-EBI), Cambridge, United Kingdom
| | - Jos Kleinjans
- Department of Toxicogenomics, GROW School for Oncology and Developmental Biology, Maastricht University, Maastricht, Netherlands
| | - Danyel Jennen
- Department of Toxicogenomics, GROW School for Oncology and Developmental Biology, Maastricht University, Maastricht, Netherlands
| |
Collapse
|
32
|
Limonciel A, van Breda SG, Jiang X, Tredwell GD, Wilmes A, Aschauer L, Siskos AP, Sachinidis A, Keun HC, Kopp-Schneider A, de Kok TM, Kleinjans JCS, Jennings P. Persistence of Epigenomic Effects After Recovery From Repeated Treatment With Two Nephrocarcinogens. Front Genet 2018; 9:558. [PMID: 30559759 PMCID: PMC6286959 DOI: 10.3389/fgene.2018.00558] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2018] [Accepted: 10/31/2018] [Indexed: 11/13/2022] Open
Abstract
The discovery of the epigenetic regulation of transcription has provided a new source of mechanistic understanding to long lasting effects of chemicals. However, this information is still seldom exploited in a toxicological context and studies of chemical effect after washout remain rare. Here we studied the effects of two nephrocarcinogens on the human proximal tubule cell line RPTEC/TERT1 using high-content mRNA microarrays coupled with miRNA, histone acetylation (HA) and DNA methylation (DM) arrays and metabolomics during a 5-day repeat-dose exposure and 3 days after washout. The mycotoxin ochratoxin A (OTA) was chosen as a model compound for its known impact on HA and DM. The foremost effect observed was the modulation of thousands of mRNAs and histones by OTA during and after exposure. In comparison, the oxidant potassium bromate (KBrO3) had a milder impact on gene expression and epigenetics. However, there was no strong correlation between epigenetic modifications and mRNA changes with OTA while with KBrO3 the gene expression data correlated better with HA for both up- and down-regulated genes. Even when focusing on the genes with persistent epigenetic modifications after washout, only half were coupled to matching changes in gene expression induced by OTA, suggesting that while OTA causes a major effect on the two epigenetic mechanisms studied, these alone cannot explain its impact on gene expression. Mechanistic analysis confirmed the known activation of Nrf2 and p53 by KBrO3, while OTA inhibited most of the same genes, and genes involved in the unfolded protein response. A few miRNAs could be linked to these effects of OTA, albeit without clear contribution of epigenetics to the modulation of the pathways at large. Metabolomics revealed disturbances in amino acid balance, energy catabolism, nucleotide metabolism and polyamine metabolism with both chemicals. In conclusion, the large impact of OTA on transcription was confirmed at the mRNA level but also with two high-content epigenomic methodologies. Transcriptomic data confirmed the previously reported activation (by KBrO3) and inhibition (by OTA) of protective pathways. However, the integration of omic datasets suggested that HA and DM were not driving forces in the gene expression changes induced by either chemical.
Collapse
Affiliation(s)
- Alice Limonciel
- Division of Molecular and Computational Toxicology, Amsterdam Institute for Molecules, Medicines and Systems, Vrije Universiteit Amsterdam, Amsterdam, Netherlands.,Division of Physiology, Department of Physiology and Medical Physics, Medical University of Innsbruck, Innsbruck, Austria
| | - Simone G van Breda
- Department of Toxicogenomics, GROW-School for Oncology and Development Biology, Maastricht University Medical Center, Maastricht, Netherlands
| | - Xiaoqi Jiang
- Division of Biostatistics, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Gregory D Tredwell
- Division of Cancer, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, Hammersmith Hospital, London, United Kingdom.,Department of Applied Mathematics, Research School of Physics and Engineering, Australian National University, Canberra, ACT, Australia
| | - Anja Wilmes
- Division of Molecular and Computational Toxicology, Amsterdam Institute for Molecules, Medicines and Systems, Vrije Universiteit Amsterdam, Amsterdam, Netherlands.,Division of Physiology, Department of Physiology and Medical Physics, Medical University of Innsbruck, Innsbruck, Austria
| | - Lydia Aschauer
- Division of Physiology, Department of Physiology and Medical Physics, Medical University of Innsbruck, Innsbruck, Austria.,Brookes Innovation Hub, Orbit Discovery, Oxford, United Kingdom
| | - Alexandros P Siskos
- Division of Cancer, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, Hammersmith Hospital, London, United Kingdom
| | - Agapios Sachinidis
- Institute of Neurophysiology and Center for Molecular Medicine Cologne (CMMC), University of Cologne (UKK), Cologne, Germany
| | - Hector C Keun
- Division of Cancer, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, Hammersmith Hospital, London, United Kingdom
| | | | - Theo M de Kok
- Department of Toxicogenomics, GROW-School for Oncology and Development Biology, Maastricht University Medical Center, Maastricht, Netherlands
| | - Jos C S Kleinjans
- Department of Toxicogenomics, GROW-School for Oncology and Development Biology, Maastricht University Medical Center, Maastricht, Netherlands
| | - Paul Jennings
- Division of Molecular and Computational Toxicology, Amsterdam Institute for Molecules, Medicines and Systems, Vrije Universiteit Amsterdam, Amsterdam, Netherlands.,Division of Physiology, Department of Physiology and Medical Physics, Medical University of Innsbruck, Innsbruck, Austria
| |
Collapse
|
33
|
A systematic analysis of Nrf2 pathway activation dynamics during repeated xenobiotic exposure. Arch Toxicol 2018; 93:435-451. [PMID: 30456486 DOI: 10.1007/s00204-018-2353-2] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2018] [Accepted: 11/08/2018] [Indexed: 11/27/2022]
Abstract
Oxidative stress leads to the activation of the Nuclear factor-erythroid-2-related factor 2 (Nrf2) pathway. While most studies have focused on the activation of the Nrf2 pathway after single chemical treatment, little is known about the dynamic regulation of the Nrf2 pathway in the context of repeated exposure scenarios. Here we employed single cell live imaging to quantitatively monitor the dynamics of the Nrf2 pathway during repeated exposure, making advantage of two HepG2 fluorescent protein reporter cell lines, expressing GFP tagged Nrf2 or sulfiredoxin 1 (Srxn1), a direct downstream target of Nrf2. High throughput live confocal imaging was used to measure the temporal dynamics of these two components of the Nrf2 pathway after repeated exposure to an extensive concentration range of diethyl maleate (DEM) and tert-butylhydroquinone (tBHQ). Single treatment with DEM or tBHQ induced Nrf2 and Srxn1 over time in a concentration-dependent manner. The Nrf2 response to a second treatment was lower than the response to the first exposure with the same concentration, indicating that the response is adaptive. Moreover, a limited fraction of individual cells committed themselves into the Nrf2 response during the second treatment. Despite the suppression of the Nrf2 pathway, the second treatment resulted in a three-fold higher Srxn1-GFP response compared to the first treatment, with all cells participating in the response. While after the first treatment Srxn1-GFP response was linearly related to Nrf2-GFP nuclear translocation, such a linear relationship was less clear for the second exposure. siRNA-mediated knockdown demonstrated that the second response is dependent on the activity of Nrf2. Several other, clinically relevant, compounds (i.e., sulphorophane, nitrofurantoin and CDDO-Me) also enhanced the induction of Srxn1-GFP upon two consecutive repeated exposure. Together the data indicate that adaptation towards pro-oxidants lowers the Nrf2 activation capacity, but simultaneously primes cells for the enhancement of an antioxidant response which depends on factors other than just Nrf2. These data provide further insight in the overall dynamics of stress pathway activation after repeated exposure and underscore the complexity of responses that may govern repeated dose toxicity.
Collapse
|
34
|
Zgheib E, Limonciel A, Jiang X, Wilmes A, Wink S, van de Water B, Kopp-Schneider A, Bois FY, Jennings P. Investigation of Nrf2, AhR and ATF4 Activation in Toxicogenomic Databases. Front Genet 2018; 9:429. [PMID: 30333853 PMCID: PMC6176024 DOI: 10.3389/fgene.2018.00429] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Accepted: 09/11/2018] [Indexed: 12/14/2022] Open
Abstract
Toxicological responses to chemical insult are largely regulated by transcriptionally activated pathways that may be independent, correlated and partially or fully overlapping. Investigating the dynamics of the interactions between stress responsive transcription factors from toxicogenomic data and defining the signature of each of them is an additional step toward a system level understanding of perturbation driven mechanisms. To this end, we investigated the segregation of the genes belonging to the three following transcriptionally regulated pathways: the AhR pathway, the Nrf2 pathway and the ATF4 pathway. Toxicogenomic datasets from three projects (carcinoGENOMICS, Predict-IV and TG-GATEs) obtained in various experimental conditions (in human and rat in vitro liver and kidney models and rat in vivo, with bolus administration and with repeated doses) were combined and consolidated where overlaps between datasets existed. A bioinformatic analysis was performed to refine pathways' signatures and to create chemical activation capacity scores to classify chemicals by their potency and selectivity of activation of each pathway. With some refinement such an approach may improve chemical safety classification and allow biological read across on a pathway level.
Collapse
Affiliation(s)
- Elias Zgheib
- Laboratoire de Biomécanique et Bio-ingénierie, Sorbonne Universités - Université de Technologie de Compiègne, Compiègne, France
| | - Alice Limonciel
- Division of Molecular and Computational Toxicology, Amsterdam Institute for Molecules, Medicines and Systems, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Xiaoqi Jiang
- Division of Biostatistics, German Cancer Research Center, Heidelberg, Germany
| | - Anja Wilmes
- Division of Molecular and Computational Toxicology, Amsterdam Institute for Molecules, Medicines and Systems, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Steven Wink
- Division of Drug Discovery and Safety, Leiden Cell Observatory High Content Imaging Screening Facility, Leiden Academic Center for Drug Research, Leiden University, Leiden, Netherlands
| | - Bob van de Water
- Division of Drug Discovery and Safety, Leiden Cell Observatory High Content Imaging Screening Facility, Leiden Academic Center for Drug Research, Leiden University, Leiden, Netherlands
| | | | - Frederic Y Bois
- Models for Ecotoxicology and Toxicology Unit (DRC/VIVA/METO), Institut National de l'Environnement Industriel et des Risques, Verneuil-en-Halatte, France
| | - Paul Jennings
- Division of Molecular and Computational Toxicology, Amsterdam Institute for Molecules, Medicines and Systems, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| |
Collapse
|
35
|
Lacher SE, Levings DC, Freeman S, Slattery M. Identification of a functional antioxidant response element at the HIF1A locus. Redox Biol 2018; 19:401-411. [PMID: 30241031 PMCID: PMC6146589 DOI: 10.1016/j.redox.2018.08.014] [Citation(s) in RCA: 74] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2018] [Revised: 08/21/2018] [Accepted: 08/22/2018] [Indexed: 12/12/2022] Open
Abstract
Reactive oxygen species (ROS), which are a byproduct of oxidative metabolism, serve as signaling molecules in a number of physiological settings. However, if their levels are not tightly maintained, excess ROS lead to potentially cytotoxic oxidative stress. Accordingly, several transcriptional regulatory networks have evolved to include components that are highly ROS-responsive. Depending on the context, these regulatory networks can leverage ROS to respond to nutrient conditions, metabolism, or other physiological signals, or to respond to oxidative stress. However, ROS signaling is complex, so regulatory interactions between various ROS-responsive transcription factors are still being mapped out. Here we show that the transcription factor NRF2, a key regulator of the adaptive response to oxidative stress, directly regulates expression of HIF1A, which encodes HIF1α, a key transcriptional regulator of the adaptive response to hypoxia. We used an integrative genomics approach to identify HIF1A as a ROS-responsive transcript and we found an NRF2-bound antioxidant response element (ARE) approximately 30 kilobases upstream of HIF1A. This ARE sequence is deeply conserved, and we verified that it is directly bound and activated by NRF2. In addition, we found that HIF1A is upregulated in breast and bladder tumors with high NRF2 activity. Taken together, our results demonstrate that NRF2 targets a functional ARE at the HIF1A locus, and reveal a direct regulatory connection between two important oxygen responsive transcription factors.
Collapse
Affiliation(s)
- Sarah E Lacher
- Department of Biomedical Sciences, University of Minnesota Medical School, 1035 University Drive, SMed 255, Duluth, MN 55812, United States
| | - Daniel C Levings
- Department of Biomedical Sciences, University of Minnesota Medical School, 1035 University Drive, SMed 255, Duluth, MN 55812, United States
| | - Samuel Freeman
- Department of Biomedical Sciences, University of Minnesota Medical School, 1035 University Drive, SMed 255, Duluth, MN 55812, United States
| | - Matthew Slattery
- Department of Biomedical Sciences, University of Minnesota Medical School, 1035 University Drive, SMed 255, Duluth, MN 55812, United States.
| |
Collapse
|
36
|
Alexander-Dann B, Pruteanu LL, Oerton E, Sharma N, Berindan-Neagoe I, Módos D, Bender A. Developments in toxicogenomics: understanding and predicting compound-induced toxicity from gene expression data. Mol Omics 2018; 14:218-236. [PMID: 29917034 PMCID: PMC6080592 DOI: 10.1039/c8mo00042e] [Citation(s) in RCA: 65] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2018] [Accepted: 05/08/2018] [Indexed: 12/12/2022]
Abstract
The toxicogenomics field aims to understand and predict toxicity by using 'omics' data in order to study systems-level responses to compound treatments. In recent years there has been a rapid increase in publicly available toxicological and 'omics' data, particularly gene expression data, and a corresponding development of methods for its analysis. In this review, we summarize recent progress relating to the analysis of RNA-Seq and microarray data, review relevant databases, and highlight recent applications of toxicogenomics data for understanding and predicting compound toxicity. These include the analysis of differentially expressed genes and their enrichment, signature matching, methods based on interaction networks, and the analysis of co-expression networks. In the future, these state-of-the-art methods will likely be combined with new technologies, such as whole human body models, to produce a comprehensive systems-level understanding of toxicity that reduces the necessity of in vivo toxicity assessment in animal models.
Collapse
Affiliation(s)
- Benjamin Alexander-Dann
- University of Cambridge
, Centre for Molecular Informatics
, Department of Chemistry
,
Lensfield Road
, Cambridge CB2 1EW
, UK
.
;
| | - Lavinia Lorena Pruteanu
- University of Cambridge
, Centre for Molecular Informatics
, Department of Chemistry
,
Lensfield Road
, Cambridge CB2 1EW
, UK
.
;
- Babeş-Bolyai University
, Institute for Doctoral Studies
,
1 Kogălniceanu Street
, Cluj-Napoca 400084
, Romania
- University of Medicine and Pharmacy “Iuliu Haţieganu”
, MedFuture Research Centre for Advanced Medicine
,
23 Marinescu Street/4-6 Pasteur Street
, Cluj-Napoca 400337
, Romania
| | - Erin Oerton
- University of Cambridge
, Centre for Molecular Informatics
, Department of Chemistry
,
Lensfield Road
, Cambridge CB2 1EW
, UK
.
;
| | - Nitin Sharma
- University of Cambridge
, Centre for Molecular Informatics
, Department of Chemistry
,
Lensfield Road
, Cambridge CB2 1EW
, UK
.
;
| | - Ioana Berindan-Neagoe
- University of Medicine and Pharmacy “Iuliu Haţieganu”
, MedFuture Research Centre for Advanced Medicine
,
23 Marinescu Street/4-6 Pasteur Street
, Cluj-Napoca 400337
, Romania
- University of Medicine and Pharmacy “Iuliu Haţieganu”
, Research Center for Functional Genomics
, Biomedicine and Translational Medicine
,
23 Marinescu Street
, Cluj-Napoca 400337
, Romania
- The Oncology Institute “Prof. Dr Ion Chiricuţă”
, Department of Functional Genomics and Experimental Pathology
,
34-36 Republicii Street
, Cluj-Napoca 400015
, Romania
| | - Dezső Módos
- University of Cambridge
, Centre for Molecular Informatics
, Department of Chemistry
,
Lensfield Road
, Cambridge CB2 1EW
, UK
.
;
| | - Andreas Bender
- University of Cambridge
, Centre for Molecular Informatics
, Department of Chemistry
,
Lensfield Road
, Cambridge CB2 1EW
, UK
.
;
| |
Collapse
|
37
|
Limonciel A, Ates G, Carta G, Wilmes A, Watzele M, Shepard PJ, VanSteenhouse HC, Seligmann B, Yeakley JM, van de Water B, Vinken M, Jennings P. Comparison of base-line and chemical-induced transcriptomic responses in HepaRG and RPTEC/TERT1 cells using TempO-Seq. Arch Toxicol 2018; 92:2517-2531. [PMID: 30008028 PMCID: PMC6063331 DOI: 10.1007/s00204-018-2256-2] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2017] [Accepted: 07/04/2018] [Indexed: 12/21/2022]
Abstract
The utilisation of genome-wide transcriptomics has played a pivotal role in advancing the field of toxicology, allowing the mapping of transcriptional signatures to chemical exposures. These activities have uncovered several transcriptionally regulated pathways that can be utilised for assessing the perturbation impact of a chemical and also the identification of toxic mode of action. However, current transcriptomic platforms are not very amenable to high-throughput workflows due to, high cost, complexities in sample preparation and relatively complex bioinformatic analysis. Thus, transcriptomic investigations are usually limited in dose and time dimensions and are, therefore, not optimal for implementation in risk assessment workflows. In this study, we investigated a new cost-effective, transcriptomic assay, TempO-Seq, which alleviates the aforementioned limitations. This technique was evaluated in a 6-compound screen, utilising differentiated kidney (RPTEC/TERT1) and liver (HepaRG) cells and compared to non-transcriptomic label-free sensitive endpoints of chemical-induced disturbances, namely phase contrast morphology, xCELLigence and glycolysis. Non-proliferating cell monolayers were exposed to six sub-lethal concentrations of each compound for 24 h. The results show that utilising a 2839 gene panel, it is possible to discriminate basal tissue-specific signatures, generate dose-response relationships and to discriminate compound-specific and cell type-specific responses. This study also reiterates previous findings that chemical-induced transcriptomic alterations occur prior to cytotoxicity and that transcriptomics provides in depth mechanistic information of the effects of chemicals on cellular transcriptional responses. TempO-Seq is a robust transcriptomic platform that is well suited for in vitro toxicity experiments.
Collapse
Affiliation(s)
- Alice Limonciel
- Division of Molecular and Computational Toxicology, Amsterdam Institute for Molecules, Medicines and Systems, Vrije Universiteit Amsterdam, De Boelelaan 1108, 1081 HZ, Amsterdam, The Netherlands
- Division of Physiology, Department of Physiology and Medical Physics, Medical University of Innsbruck, 6020, Innsbruck, Austria
| | - Gamze Ates
- Department of In Vitro Toxicology and Dermato-Cosmetology, Vrije Universiteit Brussel, Laarbeeklaan 103, 1090, Brussels, Belgium
| | - Giada Carta
- Division of Molecular and Computational Toxicology, Amsterdam Institute for Molecules, Medicines and Systems, Vrije Universiteit Amsterdam, De Boelelaan 1108, 1081 HZ, Amsterdam, The Netherlands
- Division of Physiology, Department of Physiology and Medical Physics, Medical University of Innsbruck, 6020, Innsbruck, Austria
| | - Anja Wilmes
- Division of Molecular and Computational Toxicology, Amsterdam Institute for Molecules, Medicines and Systems, Vrije Universiteit Amsterdam, De Boelelaan 1108, 1081 HZ, Amsterdam, The Netherlands
- Division of Physiology, Department of Physiology and Medical Physics, Medical University of Innsbruck, 6020, Innsbruck, Austria
| | - Manfred Watzele
- Roche Diagnostics GmbH, Nonnenwald 2, 82377, Penzberg, Germany
| | - Peter J Shepard
- BioSpyder Technologies, Inc., 5922 Farnsworth Ct Ste 102, Carlsbad, CA, 92008, USA
| | | | - Bruce Seligmann
- BioSpyder Technologies, Inc., 5922 Farnsworth Ct Ste 102, Carlsbad, CA, 92008, USA
| | - Joanne M Yeakley
- BioSpyder Technologies, Inc., 5922 Farnsworth Ct Ste 102, Carlsbad, CA, 92008, USA
| | - Bob van de Water
- Division of Toxicology, Leiden Academic Center for Drug Research, Leiden University, PO Box 9503, 2300 RA, Leiden, The Netherlands
| | - Mathieu Vinken
- Department of In Vitro Toxicology and Dermato-Cosmetology, Vrije Universiteit Brussel, Laarbeeklaan 103, 1090, Brussels, Belgium.
| | - Paul Jennings
- Division of Molecular and Computational Toxicology, Amsterdam Institute for Molecules, Medicines and Systems, Vrije Universiteit Amsterdam, De Boelelaan 1108, 1081 HZ, Amsterdam, The Netherlands.
- Division of Physiology, Department of Physiology and Medical Physics, Medical University of Innsbruck, 6020, Innsbruck, Austria.
| |
Collapse
|
38
|
Li J, Moe B, Liu Y, Li XF. Halobenzoquinone-Induced Alteration of Gene Expression Associated with Oxidative Stress Signaling Pathways. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2018; 52:6576-6584. [PMID: 29737854 DOI: 10.1021/acs.est.7b06428] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/08/2023]
Abstract
Halobenzoquinones (HBQs) are emerging disinfection byproducts (DBPs) that effectively induce reactive oxygen species and oxidative damage in vitro. However, the impacts of HBQs on oxidative-stress-related gene expression have not been investigated. In this study, we examined alterations in the expression of 44 genes related to oxidative-stress-induced signaling pathways in human uroepithelial cells (SV-HUC-1) upon exposure to six HBQs. The results show the structure-dependent effects of HBQs on the studied gene expression. After 2 h of exposure, the expression levels of 9 to 28 genes were altered, while after 8 h of exposure, the expression levels of 29 to 31 genes were altered. Four genes ( HMOX1, NQO1, PTGS2, and TXNRD1) were significantly upregulated by all six HBQs at both exposure time points. Ingenuity pathway analysis revealed that the Nrf2 pathway was significantly responsive to HBQ exposure. Other canonical pathways responsive to HBQ exposure included GSH redox reductions, superoxide radical degradation, and xenobiotic metabolism signaling. This study has demonstrated that HBQs significantly alter the gene expression of oxidative-stress-related signaling pathways and contributes to the understanding of HBQ-DBP-associated toxicity.
Collapse
Affiliation(s)
- Jinhua Li
- Department of Health Toxicology, School of Public Health , Jilin University , Changchun , Jilin , China 130021
- Division of Analytical and Environmental Toxicology, Department of Laboratory Medicine and Pathology, Faculty of Medicine and Dentistry , University of Alberta , Edmonton , Alberta , Canada T6G 2G3
| | - Birget Moe
- Division of Analytical and Environmental Toxicology, Department of Laboratory Medicine and Pathology, Faculty of Medicine and Dentistry , University of Alberta , Edmonton , Alberta , Canada T6G 2G3
- Alberta Centre for Toxicology, Department of Physiology and Pharmacology, Faculty of Medicine , University of Calgary , Calgary , Alberta , Canada T2N 4N1
| | - Yanming Liu
- Division of Analytical and Environmental Toxicology, Department of Laboratory Medicine and Pathology, Faculty of Medicine and Dentistry , University of Alberta , Edmonton , Alberta , Canada T6G 2G3
| | - Xing-Fang Li
- Division of Analytical and Environmental Toxicology, Department of Laboratory Medicine and Pathology, Faculty of Medicine and Dentistry , University of Alberta , Edmonton , Alberta , Canada T6G 2G3
| |
Collapse
|
39
|
Andersen ME, Pendse SN, Black MB, McMullen PD. Application of transcriptomic data, visualization tools and bioinformatics resources for informing mode of action. CURRENT OPINION IN TOXICOLOGY 2018. [DOI: 10.1016/j.cotox.2018.05.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
|
40
|
Guo C, Buckley A, Marczylo T, Seiffert J, Römer I, Warren J, Hodgson A, Chung KF, Gant TW, Smith R, Leonard MO. The small airway epithelium as a target for the adverse pulmonary effects of silver nanoparticle inhalation. Nanotoxicology 2018; 12:539-553. [PMID: 29750584 DOI: 10.1080/17435390.2018.1465140] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/16/2022]
Abstract
Experimental modeling to identify specific inhalation hazards for nanomaterials has in the main focused on in vivo approaches. However, these models suffer from uncertainties surrounding species-specific differences and cellular targets for biologic response. In terms of pulmonary exposure, approaches which combine 'inhalation-like' nanoparticulate aerosol deposition with relevant human cell and tissue air-liquid interface cultures are considered an important complement to in vivo work. In this study, we utilized such a model system to build on previous results from in vivo exposures, which highlighted the small airway epithelium as a target for silver nanoparticle (AgNP) deposition. RNA-SEQ was used to characterize alterations in mRNA and miRNA within the lung. Organotypic-reconstituted 3D human primary small airway epithelial cell cultures (SmallAir) were exposed to the same spark-generated AgNP and at the same dose used in vivo, in an aerosol-exposure air-liquid interface (AE-ALI) system. Adverse effects were characterized using lactate, LDH release and alterations in mRNA and miRNA. Modest toxicological effects were paralleled by significant regulation in gene expression, reflective mainly of specific inflammatory events. Importantly, there was a level of concordance between gene expression changes observed in vitro and in vivo. We also observed a significant correlation between AgNP and mass equivalent silver ion (Ag+) induced transcriptional changes in SmallAir cultures. In addition to key mechanistic information relevant for our understanding of the potential health risks associated with AgNP inhalation exposure, this work further highlights the small airway epithelium as an important target for adverse effects.
Collapse
Affiliation(s)
- Chang Guo
- a Centre for Radiation, Chemical and Environmental Hazards , Public Health England , Oxfordshire , UK.,b The National Institute for Health Research Health Protection Research Unit (NIHR HPRU) in Health Impact of Environmental Hazards at King's College London in partnership with Public Health England (PHE) in collaboration with Imperial College London , London , UK
| | - Alison Buckley
- a Centre for Radiation, Chemical and Environmental Hazards , Public Health England , Oxfordshire , UK.,b The National Institute for Health Research Health Protection Research Unit (NIHR HPRU) in Health Impact of Environmental Hazards at King's College London in partnership with Public Health England (PHE) in collaboration with Imperial College London , London , UK
| | - Tim Marczylo
- a Centre for Radiation, Chemical and Environmental Hazards , Public Health England , Oxfordshire , UK.,b The National Institute for Health Research Health Protection Research Unit (NIHR HPRU) in Health Impact of Environmental Hazards at King's College London in partnership with Public Health England (PHE) in collaboration with Imperial College London , London , UK
| | - Joanna Seiffert
- c Airways Disease, National Heart & Lung Institute, Imperial College , London , UK
| | - Isabella Römer
- a Centre for Radiation, Chemical and Environmental Hazards , Public Health England , Oxfordshire , UK.,b The National Institute for Health Research Health Protection Research Unit (NIHR HPRU) in Health Impact of Environmental Hazards at King's College London in partnership with Public Health England (PHE) in collaboration with Imperial College London , London , UK
| | - James Warren
- a Centre for Radiation, Chemical and Environmental Hazards , Public Health England , Oxfordshire , UK
| | - Alan Hodgson
- a Centre for Radiation, Chemical and Environmental Hazards , Public Health England , Oxfordshire , UK
| | - Kian Fan Chung
- c Airways Disease, National Heart & Lung Institute, Imperial College , London , UK
| | - Timothy W Gant
- a Centre for Radiation, Chemical and Environmental Hazards , Public Health England , Oxfordshire , UK.,b The National Institute for Health Research Health Protection Research Unit (NIHR HPRU) in Health Impact of Environmental Hazards at King's College London in partnership with Public Health England (PHE) in collaboration with Imperial College London , London , UK
| | - Rachel Smith
- a Centre for Radiation, Chemical and Environmental Hazards , Public Health England , Oxfordshire , UK.,b The National Institute for Health Research Health Protection Research Unit (NIHR HPRU) in Health Impact of Environmental Hazards at King's College London in partnership with Public Health England (PHE) in collaboration with Imperial College London , London , UK
| | - Martin O Leonard
- a Centre for Radiation, Chemical and Environmental Hazards , Public Health England , Oxfordshire , UK.,b The National Institute for Health Research Health Protection Research Unit (NIHR HPRU) in Health Impact of Environmental Hazards at King's College London in partnership with Public Health England (PHE) in collaboration with Imperial College London , London , UK
| |
Collapse
|
41
|
Zhang Y, den Braver-Sewradj SP, den Braver MW, Hiemstra S, Vermeulen NPE, van de Water B, Commandeur JNM, Vos JC. Glutathione S-Transferase P1 Protects Against Amodiaquine Quinoneimines-Induced Cytotoxicity but Does Not Prevent Activation of Endoplasmic Reticulum Stress in HepG2 Cells. Front Pharmacol 2018; 9:388. [PMID: 29720942 PMCID: PMC5915463 DOI: 10.3389/fphar.2018.00388] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2018] [Accepted: 04/04/2018] [Indexed: 12/15/2022] Open
Abstract
Formation of the reactive amodiaquine quinoneimine (AQ-QI) and N-desethylamodiaquine quinoneimine (DEAQ-QI) plays an important role in the toxicity of the anti-malaria drug amodiaquine (AQ). Glutathione conjugation protects against AQ-induced toxicity and GSTP1 is able to conjugate its quinoneimine metabolites AQ-QI and DEA-QI with glutathione. In this study, HepG2 cells transiently transfected with the human GSTP1 construct were utilized to investigate the protective effect of GSTP1 in a cellular context. HepG2 cells were exposed to synthesized QIs, which bypasses the need for intracellular bioactivation of AQ or DEAQ. Exposure was accompanied by decreased cell viability, increased caspase 3 activity, and decreased intracellular GSH levels. Using high-content imaging-based BAC-GFP reporters, it was shown that AQ-QI and DEAQ-QI specifically activated the endoplasmic reticulum (ER) stress response. In contrast, oxidative stress, DNA damage, or inflammatory stress responses were not activated. Overexpression of GSTP1 resulted in a two-fold increase in GSH-conjugation of the QIs, attenuated QI-induced cytotoxicity especially under GSH-depletion condition, abolished QIs-induced apoptosis but did not significantly inhibit the activation of the ER stress response. In conclusion, these results indicate a protective role of GSTP1 by increasing enzymatic detoxification of AQ-QI and DEAQ-QI and suggest a second protective mechanism by interfering with ER stress induced apoptosis.
Collapse
Affiliation(s)
- Yongjie Zhang
- Division of Molecular Toxicology, Department of Chemistry and Pharmaceutical Sciences, Amsterdam Institute for Molecules, Medicines and Systems, Vrije Universiteit Amsterdam, Amsterdam, Netherlands.,Clinical Pharmacokinetics Research Laboratory, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Shalenie P den Braver-Sewradj
- Division of Molecular Toxicology, Department of Chemistry and Pharmaceutical Sciences, Amsterdam Institute for Molecules, Medicines and Systems, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Michiel W den Braver
- Division of Molecular Toxicology, Department of Chemistry and Pharmaceutical Sciences, Amsterdam Institute for Molecules, Medicines and Systems, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Steven Hiemstra
- Division of Drug Discovery and Safety, Leiden Academic Centre for Drug Research, Leiden University, Leiden, Netherlands
| | - Nico P E Vermeulen
- Division of Molecular Toxicology, Department of Chemistry and Pharmaceutical Sciences, Amsterdam Institute for Molecules, Medicines and Systems, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Bob van de Water
- Division of Drug Discovery and Safety, Leiden Academic Centre for Drug Research, Leiden University, Leiden, Netherlands
| | - Jan N M Commandeur
- Division of Molecular Toxicology, Department of Chemistry and Pharmaceutical Sciences, Amsterdam Institute for Molecules, Medicines and Systems, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - J C Vos
- Division of Molecular Toxicology, Department of Chemistry and Pharmaceutical Sciences, Amsterdam Institute for Molecules, Medicines and Systems, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| |
Collapse
|
42
|
Choudhuri S, Patton GW, Chanderbhan RF, Mattia A, Klaassen CD. From Classical Toxicology to Tox21: Some Critical Conceptual and Technological Advances in the Molecular Understanding of the Toxic Response Beginning From the Last Quarter of the 20th Century. Toxicol Sci 2018; 161:5-22. [PMID: 28973688 PMCID: PMC5837539 DOI: 10.1093/toxsci/kfx186] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Toxicology has made steady advances over the last 60+ years in understanding the mechanisms of toxicity at an increasingly finer level of cellular organization. Traditionally, toxicological studies have used animal models. However, the general adoption of the principles of 3R (Replace, Reduce, Refine) provided the impetus for the development of in vitro models in toxicity testing. The present commentary is an attempt to briefly discuss the transformation in toxicology that began around 1980. Many genes important in cellular protection and metabolism of toxicants were cloned and characterized in the 80s, and gene expression studies became feasible, too. The development of transgenic and knockout mice provided valuable animal models to investigate the role of specific genes in producing toxic effects of chemicals or protecting the organism from the toxic effects of chemicals. Further developments in toxicology came from the incorporation of the tools of "omics" (genomics, proteomics, metabolomics, interactomics), epigenetics, systems biology, computational biology, and in vitro biology. Collectively, the advances in toxicology made during the last 30-40 years are expected to provide more innovative and efficient approaches to risk assessment. A goal of experimental toxicology going forward is to reduce animal use and yet be able to conduct appropriate risk assessments and make sound regulatory decisions using alternative methods of toxicity testing. In that respect, Tox21 has provided a big picture framework for the future. Currently, regulatory decisions involving drugs, biologics, food additives, and similar compounds still utilize data from animal testing and human clinical trials. In contrast, the prioritization of environmental chemicals for further study can be made using in vitro screening and computational tools.
Collapse
Affiliation(s)
- Supratim Choudhuri
- Office of Food Additive Safety, Center for Food Safety and Applied Nutrition, U.S. Food and Drug Administration, College Park, Maryland
| | - Geoffrey W Patton
- Department of Environmental and Occupational Health Sciences, School of Public Health, University of Washington, Seattle, Washington
| | - Ronald F Chanderbhan
- Office of Food Additive Safety, Center for Food Safety and Applied Nutrition, U.S. Food and Drug Administration, College Park, Maryland
| | - Antonia Mattia
- Office of Food Additive Safety, Center for Food Safety and Applied Nutrition, U.S. Food and Drug Administration, College Park, Maryland
| | - Curtis D Klaassen
- Department of Environmental and Occupational Health Sciences, School of Public Health, University of Washington, Seattle, Washington
| |
Collapse
|
43
|
Systems Microscopy Approaches in Unraveling and Predicting Drug-Induced Liver Injury (DILI). METHODS IN PHARMACOLOGY AND TOXICOLOGY 2018. [DOI: 10.1007/978-1-4939-7677-5_29] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
44
|
Leist M, Ghallab A, Graepel R, Marchan R, Hassan R, Bennekou SH, Limonciel A, Vinken M, Schildknecht S, Waldmann T, Danen E, van Ravenzwaay B, Kamp H, Gardner I, Godoy P, Bois FY, Braeuning A, Reif R, Oesch F, Drasdo D, Höhme S, Schwarz M, Hartung T, Braunbeck T, Beltman J, Vrieling H, Sanz F, Forsby A, Gadaleta D, Fisher C, Kelm J, Fluri D, Ecker G, Zdrazil B, Terron A, Jennings P, van der Burg B, Dooley S, Meijer AH, Willighagen E, Martens M, Evelo C, Mombelli E, Taboureau O, Mantovani A, Hardy B, Koch B, Escher S, van Thriel C, Cadenas C, Kroese D, van de Water B, Hengstler JG. Adverse outcome pathways: opportunities, limitations and open questions. Arch Toxicol 2017; 91:3477-3505. [DOI: 10.1007/s00204-017-2045-3] [Citation(s) in RCA: 238] [Impact Index Per Article: 34.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2017] [Accepted: 08/21/2017] [Indexed: 12/18/2022]
|
45
|
Sun W, Min B, Du D, Yang F, Meng J, Wang W, Zhao J, Tan X, Li Z, Sun J. miR-181c protects CsA-induced renal damage and fibrosis through inhibiting EMT. FEBS Lett 2017; 591:3588-3599. [PMID: 28976551 DOI: 10.1002/1873-3468.12872] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2017] [Revised: 09/17/2017] [Accepted: 09/26/2017] [Indexed: 01/09/2023]
Abstract
Cyclosporine A (CsA), a widely used immunosuppressive drug in organ transplantation and autoimmune disorders, frequently induces renal damage and fibrosis. Recent evidence has implicated epithelial-mesenchymal transition (EMT) in CsA-induced nephrotoxicity. Microarray analysis disclosed miR-181c as the microRNA most dramatically repressed by CsA. Downregulation of miR-181c expression at the transcriptional level by CsA is dependent on the transcription factor Nrf2. miR-181c mimics or inhibitors attenuate or aggravate CsA-induced EMT gene changes, respectively. Importantly, in Nrf2-/- mice, CsA-induced renal damage, fibrosis, and EMT gene changes are restored by miR-181c mimics. Mechanistically, we identified Notch2 as a potential target of miR-181c. Collectively, our data support the notion that miR-181c may serve as an important factor for protecting renal tissues from CsA-induced nephrotoxicity.
Collapse
Affiliation(s)
- Wenjuan Sun
- Nephrology Division, Tangdu Hospital, The Fourth Military Medical University, Xi'an, China
| | - Binying Min
- Department of Ultrasound, Tangdu Hospital, The Fourth Military Medical University, Xi'an, China
| | - Dewei Du
- Nephrology Division, Tangdu Hospital, The Fourth Military Medical University, Xi'an, China
| | - Feng Yang
- Nephrology Division, Tangdu Hospital, The Fourth Military Medical University, Xi'an, China
| | - Junping Meng
- Nephrology Division, Tangdu Hospital, The Fourth Military Medical University, Xi'an, China
| | - Wen Wang
- Nephrology Division, Tangdu Hospital, The Fourth Military Medical University, Xi'an, China
| | - Jie Zhao
- Nephrology Division, Tangdu Hospital, The Fourth Military Medical University, Xi'an, China
| | - Xiaomeng Tan
- Nephrology Division, Tangdu Hospital, The Fourth Military Medical University, Xi'an, China
| | - Zhanting Li
- Nephrology Division, Tangdu Hospital, The Fourth Military Medical University, Xi'an, China
| | - Jifeng Sun
- Nephrology Division, Tangdu Hospital, The Fourth Military Medical University, Xi'an, China
| |
Collapse
|
46
|
Souza TM, Kleinjans JCS, Jennen DGJ. Dose and Time Dependencies in Stress Pathway Responses during Chemical Exposure: Novel Insights from Gene Regulatory Networks. Front Genet 2017; 8:142. [PMID: 29085386 PMCID: PMC5649202 DOI: 10.3389/fgene.2017.00142] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2017] [Accepted: 09/21/2017] [Indexed: 11/19/2022] Open
Abstract
Perturbation of biological networks is often observed during exposure to xenobiotics, and the identification of disturbed processes, their dynamic traits, and dose–response relationships are some of the current challenges for elucidating the mechanisms determining adverse outcomes. In this scenario, reverse engineering of gene regulatory networks (GRNs) from expression data may provide a system-level snapshot embedded within accurate molecular events. Here, we investigate the composition of GRNs inferred from groups of chemicals with two distinct outcomes, namely carcinogenicity [azathioprine (AZA) and cyclophosphamide (CYC)] and drug-induced liver injury (DILI; diclofenac, nitrofurantoin, and propylthiouracil), and a non-carcinogenic/non-DILI group (aspirin, diazepam, and omeprazole). For this, we analyzed publicly available exposed in vitro human data, taking into account dose and time dependencies. Dose–Time Network Identification (DTNI) was applied to gene sets from exposed primary human hepatocytes using four stress pathways, namely endoplasmic reticulum (ER), NF-κB, NRF2, and TP53. Inferred GRNs suggested case specificity, varying in interactions, starting nodes, and target genes across groups. DILI and carcinogenic compounds were shown to directly affect all pathway-based GRNs, while non-DILI/non-carcinogenic chemicals only affected NF-κB. NF-κB-based GRNs clearly illustrated group-specific disturbances, with the cancer-related casein kinase CSNK2A1 being a target gene only in the carcinogenic group, and opposite regulation of NF-κB subunits being observed in DILI and non-DILI/non-carcinogenic groups. Target genes in NRF2-based GRNs shared by DILI and carcinogenic compounds suggested markers of hepatotoxicity. Finally, we indicate several of these group-specific interactions as potentially novel. In summary, our reversed-engineered GRNs are capable of revealing dose dependent, chemical-specific mechanisms of action in stress-related biological networks.
Collapse
Affiliation(s)
- Terezinha M Souza
- Department of Toxicogenomics, GROW School for Oncology and Developmental Biology, Maastricht University, Maastricht, Netherlands
| | - Jos C S Kleinjans
- Department of Toxicogenomics, GROW School for Oncology and Developmental Biology, Maastricht University, Maastricht, Netherlands
| | - Danyel G J Jennen
- Department of Toxicogenomics, GROW School for Oncology and Developmental Biology, Maastricht University, Maastricht, Netherlands
| |
Collapse
|
47
|
Pisani C, Rascol E, Dorandeu C, Charnay C, Guari Y, Chopineau J, Devoisselle JM, Prat O. Biocompatibility assessment of functionalized magnetic mesoporous silica nanoparticles in human HepaRG cells. Nanotoxicology 2017; 11:871-890. [PMID: 28937306 DOI: 10.1080/17435390.2017.1378749] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Magnetic mesoporous silica nanoparticles (M-MSNs) are a promising class of nanoparticles for drug delivery. However, a deep understanding of the toxicological mechanisms of action of these nanocarriers is essential, especially in the liver. The potential toxicity on HepaRG cells of pristine, pegylated (PEG), and lipid (DMPC) M-MSNs were compared. Based on MTT assay and real-time cell impedance, none of these NPs presented an extensive toxicity on hepatic cells. However, we observed by transmission electron microscopy (TEM) that the DMPC and pristine M-MSNs were greatly internalized. In comparison, PEG M-MSNs showed a slower cellular uptake. Whole gene expression profiling revealed the M-MSNs molecular modes of action in a time- and dose-dependent manner. The lowest dose tested (1.6 µg/cm2) induced no molecular effect and was defined as 'No Observed Transcriptional Effect level.' The dose 16 µg/cm2 revealed nascent but transient effects. At the highest dose (80 µg/cm2), adverse effects have clearly arisen and increased over time. The limit of biocompatibility for HepaRG cells could be set at 16 µg/cm2 for these NPs. Thanks to a comparative pathway-driven analysis, we highlighted the sequence of events that leads to the disruption of hepatobiliary system, elicited by the three types of M-MSNs, at the highest dose. The Adverse Outcome Pathway of hepatic cholestasis was implicated. Toxicogenomics applied to cell cultures is an effective tool to characterize and compare the modes of action of many substances. We propose this strategy as an asset for upstream selection of the safest nanocarriers in the framework of regulation for nanobiosafety.
Collapse
Affiliation(s)
- Cédric Pisani
- a MACS, UMR 5253 CNRS-ENSCM-UM , Institut Charles Gerhardt de Montpellier , Montpellier , France.,b Direction de la Recherche Fondamentale-BIAM , CEA , Bagnols-sur-Cèze , France
| | - Estelle Rascol
- a MACS, UMR 5253 CNRS-ENSCM-UM , Institut Charles Gerhardt de Montpellier , Montpellier , France
| | - Christophe Dorandeu
- a MACS, UMR 5253 CNRS-ENSCM-UM , Institut Charles Gerhardt de Montpellier , Montpellier , France
| | - Clarence Charnay
- c IMNO, UMR 5253 CNRS-ENSCM-UM , Institut Charles Gerhardt de Montpellier , Montpellier , France
| | - Yannick Guari
- c IMNO, UMR 5253 CNRS-ENSCM-UM , Institut Charles Gerhardt de Montpellier , Montpellier , France
| | - Joël Chopineau
- a MACS, UMR 5253 CNRS-ENSCM-UM , Institut Charles Gerhardt de Montpellier , Montpellier , France
| | - Jean-Marie Devoisselle
- a MACS, UMR 5253 CNRS-ENSCM-UM , Institut Charles Gerhardt de Montpellier , Montpellier , France
| | - Odette Prat
- b Direction de la Recherche Fondamentale-BIAM , CEA , Bagnols-sur-Cèze , France
| |
Collapse
|
48
|
Perez-Leal O, Barrero CA, Merali S. Pharmacological stimulation of nuclear factor (erythroid-derived 2)-like 2 translation activates antioxidant responses. J Biol Chem 2017; 292:14108-14121. [PMID: 28684421 DOI: 10.1074/jbc.m116.770925] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2016] [Revised: 07/05/2017] [Indexed: 12/30/2022] Open
Abstract
Nuclear factor (erythroid-derived 2)-like 2 (Nrf2) is the master regulator of the antioxidant response, and its function is tightly regulated at the transcriptional, translational, and post-translational levels. It is well-known that Nrf2 is regulated at the protein level by proteasomal degradation via Kelch-like ECH-associated protein 1 (Keap1), but how Nrf2 is regulated at the translational level is less clear. Here, we show that pharmacological stimulation increases Nrf2 levels by overcoming basal translational repression. We developed a novel reporter assay that enabled identification of natural compounds that induce Nrf2 translation by a mechanism independent of Keap1-mediated degradation. Apigenin, resveratrol, and piceatannol all induced Nrf2 translation. More importantly, the pharmacologically induced Nrf2 overcomes Keap1 regulation, translocates to the nucleus, and activates the antioxidant response. We conclude that translational regulation controls physiological levels of Nrf2, and this can be modulated by apigenin, resveratrol, and piceatannol. Also, targeting this mechanism with novel compounds could provide new insights into prevention and treatment of multiple diseases in which oxidative stress plays a significant role.
Collapse
Affiliation(s)
- Oscar Perez-Leal
- From the Department of Pharmaceutical Sciences, Moulder Center for Drug Discovery Research, Temple University School of Pharmacy, Philadelphia, Pennsylvania 19140.
| | - Carlos Alberto Barrero
- From the Department of Pharmaceutical Sciences, Moulder Center for Drug Discovery Research, Temple University School of Pharmacy, Philadelphia, Pennsylvania 19140
| | - Salim Merali
- From the Department of Pharmaceutical Sciences, Moulder Center for Drug Discovery Research, Temple University School of Pharmacy, Philadelphia, Pennsylvania 19140.
| |
Collapse
|
49
|
Meldrum K, Gant TW, Leonard MO. Diesel exhaust particulate associated chemicals attenuate expression of CXCL10 in human primary bronchial epithelial cells. Toxicol In Vitro 2017; 45:409-416. [PMID: 28655636 DOI: 10.1016/j.tiv.2017.06.023] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2017] [Revised: 06/19/2017] [Accepted: 06/22/2017] [Indexed: 12/21/2022]
Abstract
Air pollution affects a large proportion of the population particularly in urban areas, with diesel particulates recognised as particular causes for concern in respiratory conditions such as asthma. In this study we examined the response of human primary airway epithelial cells to diesel particulate chemical extracts (DE) and characterised gene expression alterations using RNA-SEQ. Using the antagonist CH223191, DE induced CYP1A1 and attenuation of CXCL10 among other genes were observed to be aryl hydrocarbon receptor dependent. Basal and toll like receptor dependent protein levels for CXCL10 were markedly reduced. Investigation of similar regulation in plasmacytoid dendritic GEN2.2 cells did not show DE dependent regulation of CXCL10. Instillation of DE into mice to recapitulate airway epithelial exposure to chemical extracts in an in vivo setting failed to demonstrate a reduction in CXCL10. There was however an increase in the Th2 type epithelial cell derived inflammatory mediators TSLP and SERPINB2. We also observed an increased macrophages and a decrease in the proportion of lymphocytes in bronchoalveolar lavage fluid. CXCL10 can play a role in allergic airway disease through recruitment of Th1 type CD4+ T-cells, which can act to counterbalance Th2 type allergic responses. Modulation of such chemokines within the airway epithelium may represent a mechanism through which pollutant material can modify respiratory conditions such as allergic asthma.
Collapse
Affiliation(s)
- Kirsty Meldrum
- Centre for Radiation, Chemical and Environmental Hazards, Public Health England, Chilton, Didcot OX11 0RQ, UK; The National Institute for Health Research Health Protection Research Unit (NIHR HPRU) in Health Impact of Environmental Hazards at King's College London in partnership with Public Health England (PHE) in collaboration with Imperial College London, UK
| | - Timothy W Gant
- Centre for Radiation, Chemical and Environmental Hazards, Public Health England, Chilton, Didcot OX11 0RQ, UK; The National Institute for Health Research Health Protection Research Unit (NIHR HPRU) in Health Impact of Environmental Hazards at King's College London in partnership with Public Health England (PHE) in collaboration with Imperial College London, UK
| | - Martin O Leonard
- Centre for Radiation, Chemical and Environmental Hazards, Public Health England, Chilton, Didcot OX11 0RQ, UK; The National Institute for Health Research Health Protection Research Unit (NIHR HPRU) in Health Impact of Environmental Hazards at King's College London in partnership with Public Health England (PHE) in collaboration with Imperial College London, UK.
| |
Collapse
|
50
|
Gola JM, Strzałka-Mrozik B, Kruszniewska-Rajs C, Adamska J, Gagoś M, Mazurek U. Differential expression of microRNAs and their predicted targets in renal cells exposed to amphotericin B and its complex with copper (II) ions. Toxicol Mech Methods 2017; 27:537-543. [PMID: 28534445 DOI: 10.1080/15376516.2017.1333554] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
MicroRNAs (miRNAs) have been reported to regulate essential biological processes, and their expression was shown to be affected by pathological processes and drug-induced toxicity. Amphotericin B (AmB) can cause liver and kidney injury, but a recently developed complex of AmB with copper (II) ions (AmB-Cu2+) exhibits a lower toxicity to human renal cells while retaining a high antifungal activity. The aim of our study was to assess AmB-Cu2+-induced changes in the miRNA profile of renal cells and examine which biological processes are significantly affected by AmB-Cu2+. We also aimed to predict whether differentially expressed miRNAs would influence observed changes in the mRNA profile. miRNA and mRNA profiles in normal human renal proximal tubule epithelial cells (RPTEC) treated with AmB-Cu2+ or AmB were appointed with the use of microarray technology. For differentially expressed mRNAs, the PANTHER overrepresentation binomial test was performed. miRNA target interactions (MTIs) were predicted using the miRTar tool. The mRNA profile was much more strongly affected than the miRNA profile, in both AmB-Cu2+- and AmB-treated cells. AmB-Cu2+ influenced both the miRNA and mRNA profiles much more strongly than AmB. The most affected biological processes were intracellular signal transduction (AmB-Cu2+) and signal transduction (AmB). Only a few interactions between differentiating miRNAs and mRNAs were found. Changes in the profiles of genes involved in signal transduction and intracellular signal transduction may not result from interactions with differentially expressed miRNAs. Changes in the miRNA profile suggest the possible influence of tested drugs on the regulation of fibrosis via a miRNA-dependent mechanism.
Collapse
Affiliation(s)
- Joanna Magdalena Gola
- a Department of Molecular Biology , School of Pharmacy with the Division of Laboratory Medicine in Sosnowiec, Medical University of Silesia , Sosnowiec , Poland
| | - Barbara Strzałka-Mrozik
- a Department of Molecular Biology , School of Pharmacy with the Division of Laboratory Medicine in Sosnowiec, Medical University of Silesia , Sosnowiec , Poland
| | - Celina Kruszniewska-Rajs
- a Department of Molecular Biology , School of Pharmacy with the Division of Laboratory Medicine in Sosnowiec, Medical University of Silesia , Sosnowiec , Poland
| | - Jolanta Adamska
- a Department of Molecular Biology , School of Pharmacy with the Division of Laboratory Medicine in Sosnowiec, Medical University of Silesia , Sosnowiec , Poland
| | - Mariusz Gagoś
- b Department of Cell Biology , Institute of Biology and Biotechnology, Maria Curie-Skłodowska University , Lublin , Poland
| | - Urszula Mazurek
- a Department of Molecular Biology , School of Pharmacy with the Division of Laboratory Medicine in Sosnowiec, Medical University of Silesia , Sosnowiec , Poland
| |
Collapse
|