Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2020 Jan 3.
Published in final edited form as: Environ Sci Technol. 2018 May 17;52(11):6565–6575. doi: 10.1021/acs.est.7b06389

Genotoxicity Assessment of Drinking Water Disinfection Byproducts by DNA Damage and Repair Pathway Profiling Analysis

Jiaqi Lan †,, Sheikh Mokhlesur Rahman , Na Gou , Tao Jiang , Micheal J Plewa §, Akram Alshawabkeh , April Z Gu †,‖,*
PMCID: PMC6941474  NIHMSID: NIHMS995886  PMID: 29660283

Abstract

Genotoxicity is considered a major concern for drinking water disinfection byproducts (DBPs). Of over 700 DBPs identified to date, only a small number has been assessed with limited information for DBP genotoxicity mechanism(s). In this study, we evaluated genotoxicity of 20 regulated and unregulated DBPs applying a quantitative toxicogenomics approach. We used GFP-fused yeast strains that examine protein expression profiling of 38 proteins indicative of all known DNA damage and repair pathways. The toxicogenomics assay detected genotoxicity potential of these DBPs that is consistent with conventional genotoxicity assays end points. Furthermore, the high-resolution, real-time pathway activation and protein expression profiling, in combination with clustering analysis, revealed molecular level details in the genotoxicity mechanisms among different DBPs and enabled classification of DBPs based on their distinct DNA damage effects and repair mechanisms. Oxidative DNA damage and base alkylation were confirmed to be the main molecular mechanisms of DBP genotoxicity. Initial exploration of QSAR modeling using moleular genotoxicity end points (PELI) suggested that genotoxicity of DBPs in this study was correlated with topological and quantum chemical descriptors. This study presents a toxicogenomics-based assay for fast and efficient mechanistic genotoxicity screening and assessment of a large number of DBPs. The results help to fill in the knowledge gap in the understanding of the molecular mechanisms of DBP genotoxicity.

Graphical Abstract

graphic file with name nihms-995886-f0007.jpg

INTRODUCTION

Drinking water disinfection byproducts (DBPs) are formed during the reaction of disinfectants (such as chlorine, chlorine dioxide, chloramine, UV, and ozone) with naturally occurring organic matter (NOM) and other contaminants present in water.1 DBPs therefore widely exist in drinking water at sub-μg/L (ppb) to low-to-mid-μg/L levels. Currently, there are over 700 DBPs reported in drinking water, and new DBPs continue to be discovered.13

Great knowledge gaps exist for toxicological information and health impacts of DBPs. Humans are exposed to DBPs through multiple routes, including ingestion (the common route studied), inhalation, and dermal exposures.3 Literature review indicates that only approximately 15% (~100) of identified DBPs have been assessed with in vitro bioassays and a few with chronic in vivo studies.24 Potential health risks of DBPs have been reported, including cancer and other adverse reproductive effects, such as early term miscarriage and birth defects.57 An association of specific cancers and exposure to disinfected water has emerged by epidemiological research.5,8,9 Several toxicity mechanisms for DBPs have been implicated, including genotoxicity, oxidative stress, disruption of folate metabolism, and cell cycle disruption.5,8,9 Genotoxicity is of particular importance because of its link to mutagenicity, carcinogenicity, as well as cancer.7,10

The genotoxicity of evaluated DBPs seems to be dependent on their structure and substituents. For example, among the halogenated DBPs, iodinated DBPs were observed to be more toxic than their brominated and chlorinated analogues.11,12 Nitrogen-containing DBPs were more genotoxic than the DBPs that do not contain nitrogen.3,13 Genotoxicity mechanisms of DBPs are also strongly related to their structures. For example, for halogenated DBPs, oxidative stress-induced DNA damage1421 and DNA alkylation2225 are two major mechanisms. Halonitriles may also induce genomic damage by cell cycle disruption and the induction of hyperploidy.26 Nitrosamines may act as alkylating agents after metabolism,27 and formaldehyde can form DNA—protein cross-links.24 The genotoxicity and mechanisms of most DBPs remain unknown.

The standard and most reliable genotoxicity tests are in vivo assays; however, they are resource-intensive and time-consuming and therefore cannot meet the demand for evaluating a large number of potential genotoxic DBPs.28,29 In vitro genotoxicity assays, including Ames, comet, and micronucleus tests, require relatively shorter testing time (several days) but often yield inconsistent or false results compared to in vivo outcomes7,30,31 due to the inherent limitations of the target and DNA damage effects they can detect.7,30 In recent years, high-throughput genotoxicity assessment has been reported where the activation of single or selected biomarkers indicative of DNA damage recognition and repair are used to indicate potential genotoxicity.7,3237 Our group has recently developed and validated a new quantitative toxicogenomics-based assay based on real time protein expression profiling of known DNA damage and repair pathways ensemble.3841 Compared to other biomarker-based tests, our assay derives quantitative end points that correlate with conventional genotoxicity end points and promises to be a cost-effective and mechanistic genotoxicity assessment assay.40,41

In this study, we employed the newly developed quantitative toxicogenomics genotoxicity assay to perform a mechanistic genotoxicity assessment and profiling of 20 DBPs representing nine different chemical classes of DBPs. The results provide new genotoxicity information and insights of underlying DNA damaging mechanisms at the molecular level for these 20 DBPs.

The high-resolution protein expression profiles of DNA damage and repair pathways also enabled DBP classification and further exploration of association between DBP chemical structure and the genotoxicity mechanisms.

MATERIALS AND METHODS

Chemicals.

Twenty DBPs were selected that belong to nine different chemical classes (Table 1, manufacturer information in Table S1). Each DBP was evaluated across a 6-log subcytotoxic concentration range (Table S1). The maximum noncytotoxic concentration was predetermined (>95% cell survival tested by growth inhibition in yeast for 24 h, Figure S1).

Table 1.

Summary of PELI-Based Molecular End Points (PELImax, PELI1.5, and Geno-TEQ1.5 (MMC as reference compound) with R2 Indicating the Fitness of the Data to Four Parameter Logistic Models (in Table S1) for 20 DBPs Tested in This Study and Phenotypic End Points from the Literature40,a

class chemical PELI1.5 (mM) geno-TEQ1.5 (in reference to MMC) Ames (mutants/μM)5,21,58,59 GP (μM)5,12,b TD50 (mg kg−1 day−1, mice)60 carcinogenicity5,60
HAAs/iodo-acids chloroacetic acid 8.826 × 10−7 14.820 27 411
bromoacetic acid 4.028 × 10−4 0.032 5465 17 NA NA
iodoacetic acid 3.240 × 10−5 0.404 14129 8.7 NA NA
trichloroacetic acid (TCA) 4.123 × 10−3 0.003 584 +
oxyhalides sodium bromate 5.451 × 10−6 2.400 NA 41 +
sodium chlorite 1.648 × 10−6 7.937 26.5 NA
trihalomethanes (THMs) bromodichloromethane 5.245 × 10−5 0.249 0.6254 47.7 +
chlorodibromomethane 3.254 × 10−5 0.402 288.6 139 +
halonitromethanes trichloronitromethane 4.015 × 10−7 32.578 40.5 93.4
nitrosamines N-nitrosodimethylamine (NDMA) 7.065 × 10−8 185.138 533c 220 0.189 +
haloamides chloroacetamide 2.071 × 10−6 6.316 NA 1380 NA NA
2-bromoacetamide 1.051 × 10−6 12.445 NA 36.8 NA NA
2,2-dichloroacetamide 1.703 × 10−6 7.681 NA NA NA
halonitriles (HANs) dichloroacetonitrile 5.664 × 10−7 23.093 + 2750 NA NA
dibromoacetonitrile 7.155 × 10−4 0.018 + 47.1 NA +
chloroacetonitrile NA NA + 601 NA +
iodoacetonitrile 2.384 × 10−6 5.487 NA 37.1 NA NA
aldehydes trichloroacetaldehyde 7.709 × 10−6 1.697 + 99 +
formaldehyde 4.296 × 10−6 3.045 + NA 43.9 +
haloquinones 2,6-dichloro-1,4-benzoquinone 7.259 × 10−5 0.180 + NA NA NA
a

NA: PELI1.5 and geno-TEQ11.5 were not determined for chloroacetonitrile with PELImax less than 1.5 based on concentration response curve.

b

GP: the genotoxic potency derived from comet assay in CHO cells, which is the concentration at the midpoint of the concentration–response curve.5,12

c

CYP used in Ames test of his reversion for mutagenicity.61

Yeast Whole Cell Array and Real Time Protein Expression Analysis upon DBP Exposure.

The whole cell assay library consists of 38 in-frame GFP fusion proteins (Table S2) of Saccharomyces cerevisiae (Invitrogen, no. 95702, ATCC 201388) constructed by oligonucleotide-directed homologous recombination to tag each open reading frame (ORF) with Aequrea victoria GFP (S65T) in its chromosomal location at the 3’ end,42 covering all seven known DNA damage repair pathways. The library expresses full-length, chromosomally tagged green fluorescent protein fusion proteins,42 which makes the GFP signal reflect protein expression directly.

Details of the proteomics assay for using GFP-tagged yeast cells were described in our previous reports.3841 Briefly, the yeast strains were grown in clear bottom black 384-well plates (Costar) with Synthetic Dextrose base (SD medium) that contains –His Dropout ( DO) supplement (Clontech, CA, US) for 4–6 h at 30 °C to reach early exponential growth. Then, 10 μL DBP sample aliquots in PBS or vehicle control (PBS only) were added to each well to obtain the target concentrations (Table S1). The plates were then placed in a Microplate Reader (Synergy H1Multi-Mode, Biotech, Winooski, VT) for absorbance (OD600 for cell growth), and GFP signal (filters with 485 nm excitation and 535 nm emission for protein expression) measurements were taken every 5 min for 2 h after double orbital shaking (425 cpm) for 1 min. All tests were performed in the dark in triplicate. Considering that all of the DBPs were tested at concentrations much lower than their solubility, evaporation and loss of the volatile DBPs during the 2 h assay was not considered in this study.43,44

Protein Expression Profiling Data Processing and Quantitative Molecular End Point Derivation.

Temporal protein expression profiling data of the yeast library were processed as described previously.39,41,45 Temporal OD and GFP raw data are first corrected by background OD and GFP signal of blank medium control with or without chemical. The protein expression level P for each protein biomarker (ORF) i, in treatment x, and at time point t is normalized by cell density as

Pi,x,t=GFPi,x,tcorrectedODi,x,tcorrected (1)

where GFPi,x,t-corrected is the GFP reading of protein i in treatment x at time t corrected by the GFP reading in the blank medium control at time t; ODi,x,t-corrected is the OD reading of protein i in treatment x at time t corrected by the OD reading in the blank medium control at time t.

The altered protein expression in relative to untreated control (without chemical) for a given protein ORFi in treatment x at time t due to chemical exposure, also referred as induction factor I, is calculated as

Ii,x,t=Pi,x,tPi,untreated,t (2)

where Pi,x,t = (GFPcorrected/ODcorrected)treatment,x is the altered protein expression GFP level for protein (ORF) i for treatment x at time t in the treated experimental condition with chemical exposure; Pi,untreated,t = (GFPcorrected/ ODcorrected)untreated control is the altered protein expression GFP level for protein (ORF) i for treatment x at time t in the untreated control without chemical exposure. P values of both treated experiments and untreated controls are normalized and scaled against internal control (housekeeping protein PGK146).

For the chemical-induced protein expression level changes of a treatment to be quantified, the protein effect level index (PELI) was derived as a quantitative molecular end point.3841 The accumulative altered protein expression change over the 2 h exposure period for a given protein (ORF) i was calculated as

PELIORF,i=t=0tIupregulateddtexposure time (3)

where t is the exposure time. For upregulated protein, Iupregulated = I, when I ≥ 1; for proteins that showed downregulation, Iupregulated = 1 when I < 1.

The pathway activation response is calculated by integrating the protein expression changes for all of the proteins (ORFs) in a pathway as

PELIpathway j=i=1nwi×PELIORFin (4)

where n is the number of ORFs in one particular pathway, and wi is the weight factor of ORFi. For this study, we assigned a value of 1 for all of the weight factors.

Similar to PELIpathway, the overall protein expression effect level for the DNA damage and repair pathway ensemble is calculated as PELIgeno with all of the PELIpathway in the pathway ensemble library as

PELIgeno=j=1NWj×PELIpathway jN (5)

where N is the number of pathways in this geno-sensor library, Wj is the weight factor of pathway j, and the value is assigned as 1 for this study.

For each DBP, six PELIgeno values are evaluated by mean ± SD. The PELIgeno-based concentration—response pattern was modeled using a four parameter logistic (4PL) nonlinear regression model (the fitted curves). End point PELImax was derived based on the PELIgeno concentration–response curve using 4PL model fitting.40,47 End point PELI1.5 was derived based on the concentration–response curves, which was defined as the corresponding concentration that causes the PELI value to reach 1.5, similar to the approach that has been applied for the umuC genotoxicity assay by Escher et al.48 and our previous study.40,49 Additionally, genotoxicity for each chemical with PELI1.5 was also expressed as toxic equivalents as

geno-TEQ1.5=PELI1.5reference compoundPELI1.5sample (6)

where mytomycin C (MMC) is used as reference compound.50 PELI1.5MMC = 2.15 X 10−3 mM based on our previous study.40,41

DNA Damage Alkaline Comet Assay in Human A549 Cells for Phenotypic Confirmation.

The alkaline comet assay in human A549 cells5153 upon exposure to the DBPs at selected concentrations (details in Table S1) or 1% FBS-F12 medium only (as untreated control) for 24 h was carried out using Trevigen Inc. CometAssay 96 slides (www.trevigen.com). All the procedures were performed in the dark in triplicate. Each treatment (25 cells) was measured by the software CASP (University of Wroclaw, Institute of Theoretical Physics) randomly, and the damage was valued as % tail DNA (mean ± SD).12

Physicochemical Descriptors.

Quantitative structure–activity relationship (QSAR) analyses were performed to obtain insights into the physiochemical characteristics of DBPs that impact their genotoxicity. Various descriptors are used (Table S3) to support the QSAR analyses and they include the PaDEL descriptor software used for topological descriptors such as autocorrelation descriptors AATSC4c and AATSC3v, electrotopological state atom-type descriptor minsCl, and extended topochemical atom descriptor ETA_Eta_L.54 The US-EPA EPI suite was used for log Kow; Gaussian03 (using Hartree–Fock 3-21G) was used to calculate quantum chemical descriptors such as Ehomo (the energy of the highest occupied molecular orbital), Elumo (the energy of the lowest unoccupied molecular orbital), and G (Gibbs free energy), and the numbers of freely rotatable bonds, H acceptors, and H donors, polar surface area, and molecular weight were collected from the PubChem Web site (http://www.ncbi.nlm.nih.gov/pccompound).

Clustering Analysis.

Hierarchical clustering (HCL) was performed to cluster all 20 DBPs across six concentrations (120 samples in total) based on their protein expression profiles by software suit MeV (MutiExperiment Viewer) v4.8.55 The relationships were elucidated using the order of average linkage clustering based on Pearson correlation.

RESULTS AND DISCUSSION

DNA Damage Mechanisms Revealed by Concentration-Dependent, Chemical-Specific Temporal Differential Protein Expression Profiles among DBPs.

The temporal altered protein expression profiles (Figure 1 and Figure S2) indicative of DNA damage and repair pathway activities were distinctive for each of the 20 DBPs tested in this study, suggesting compound-specific cellular responses resulted from their different DNA-damaging mechanisms. These chemical-specific response patterns were also concentration-dependent, showing generally an increase in magnitude of altered protein expression as concentration increases (Figure 1A). For some DBPs tested, such as bromoacetic acid and dibromoacetonitrile (Figure S2), the highest exposure concentration led to decreases in the magnitude of upregulation or even a shift from up- to downregulation for most of the tested proteins. Consistent with our previous reports,38,40 this was likely caused by the transition from a mode-of-action specific effect to subcytotoxic nonspecific cellular responses.

Figure 1.

Figure 1.

Temporal protein expression profiles of 38 biomarkers indicative of different DNA damage repair pathways upon exposure to trichloroacetic acid (A, a haloacetic acid40) and chloroacetonitrile (B, a halonitrile) across six concentrations. The mean natural log of the induction factor (ln I, n = 3) indicates the magnitude of altered protein expression (represented by a green–black–red color scale at the bottom. The red spectrum colors indicate upregulation, and the green spectrum colors indicate downregulation. Values beyond ±1.5 are shown as ±1.5. X-axis top: concentrations for each chemical; X-axis bottom: testing time in minutes. The first data point shown is at 20 min after exposure due to data smoothing with moving average of every five data points. Y-axis left: clusters of proteins by DNA damage repair pathways and list of proteins (ORFs) tested; Y-axis right: description of DNA damage repair pathway abbreviations.

Correlation between Molecular End Points and Conventional Genotoxicity/Carcinogenicity End Points.

The molecular quantifier PELIgeno exhibited a concentration response for all 20 DBPs tested (Figure 2). Our previous studies have demonstrated that quantitative genotoxicity molecular end point PELIgeno derived from the yeast assay could statistically correlate to conventionally accepted genotoxicity assays for genotoxins, known genotoxic positive and negative chemicals.40,41 Consistent with previous studies, a statistically significant strong correlation (rP = 0.5136, P = 0.0205) was observed between molecular genotoxicity end point PELIgeno and the phenotpyic DNA damage end point % tail DNA from comet assay we performed in human A549 cells (Figure 3, comet assay details in Figure S3).

Figure 2.

Figure 2.

Concentration-response curves of the 20 DBPs tested based on PELIgeno values: (A) haloacetic acids/iodo-acids and oxyhalides; (B) trihalomethanes, halonitromethanes, and NDMA; (C) haloamides and aldehydes; (D) halonitriles and 2,6-dichloro-1,4-benzoquinone. Data points with an error bar represent the PELIgeno value determined at each concentration. R2 values indicative of fitness are listed in Table S1. Genotoxicity positive is defined as having a PELImax value (determined via model fitting concentration–response curves) greater than 1.5 (the dashed line).40 X-axis: concentration for chemicals studied (mM). Y-axis: PELIgeno. Mean ± SD, n = 3. Note that data for five DBPs (trichloroacetic acid, NDMA, bromodichloromethane, chlorodibromomethane, and formaldehyde) were reported previously.40

Figure 3.

Figure 3.

Correlation of molecular end point PELIgeno derived from our GFP-fused yeast assay with phenotypic end point of DNA damage measured by % tail DNA tested by alkaline comet assay in human A549 cell line for selected concentrations (Table S1). X-axis: 24 h DNA damage measured by % tail DNA in human A549 cells (details in Figure S3); Y-axis: PELIgeno, the integrated quantifier of altered protein expression levels of 38 protein biomarkers indicative of DNA damage repair responses. Mean ± SD, n = 3. rP indicates the Pearson correlation coefficient of PELIgeno to DNA damage comet assay phenotypic end points (% tail DNA).

We also compared our quantitative genotoxicity molecular end point PELI1.5 with end points from different conventional genotoxicity assays (Table 1). We examined the correlation between the derived molecular end point PELI1.5 with other in vitro genotoxicity assay results including Ames assay in bacteria and comet assay in CHO cells. The results indicated that the molecular end point PELI1.5 correlated with both genotoxic potency (GP) of comet assay (CHO cell, rP = –0.5568, P = 0.0945, n = 10 in Figure 4A) and TD50 in vivo for carcinogenic potency (mice, rP = 0.8186, P = 0.0464, n = 6 in Figure 4B). No significant correlation was found between yeast genotoxicity end points and Ames test (his reversion), rP = 0.4472, P = 0.2666, n = 8; data not shown).

Figure 4.

Figure 4.

Correlation of molecular end point PELI1.5 with phenotypic end points: genotoxic potency from comet assay in CHO cells (A, n = 10) and carcinogenic potency from a 2-year carcinogenesis test in mice (B, n = 6) (data collected from references as shown in Table 1). X-axis: PELI1.5 determined via model fitting concentration–response curves (lg(PELI1.5), mM); Y-axis: genotoxic potency (lg(GP), μM, A) and carcinogenic potency (lg(TD50), mM kg−1 day−1, B). rP indicates the Pearson correlation coefficient of PELI1.5 to phenotypic end points.

These results confirmed that our assay, consisting of biomarkers-ensemble indicative of DNA damage and repair pathway activities, could capture various DNA damage potentials and therefore reliably predict DNA damage-related carcinogenicity. The results also indicate the conservation of DNA repair response among species. The quantitative correlation between the toxicogenomic assay-derived end points and conventional end points suggest that it can possibly be incorporated into a toxicity and risk assessment framework. Our results are in general agreement with results from different genotoxicity assays reported in the literarure. Where inconsistencies were noted, these could be attributed to varying detection targets and inherent limitations of each specific assay as discussed previously.40

Note that although no extra metabolic activation (e.g., liver extract S9) was used for genotoxicity evaluation of DBPs in this study, detectable molecular genotoxicity was observed for the known metabolically activated genotoxicant NDMA.27 Several cytochrome P-448 monooxygenase enzymes in yeast (including S. cerevisiae of this study) can perform Phase I metabolism on some compounds in a manner analogous to mammalian microsomal enzymes, although less efficiently.38,56,57 The enzymatic capability of yeast may explain the genotoxicity observed in this study for NDMA without extra metabolic activation.

DNA Damaging Pathway Activation Profiling Revealed Distinct Genotoxicity Mechanisms among DBPs.

As shown in Figure 5, the activation of biomarkers indicative of specific DNA damage and repair pathways revealed insights into the underlying mechanism(s) involved in the genotoxicity of studied DBPs;40,41 16 out of 20 DBPs in this study induced oxidative DNA damage indicated by OGG1 upregulation (indicated by “oxidation” in Figure 5) of the base excision repair system (BER), which was consistent with their strong oxidizing ability.17,19,21,62 Activation of BER via other base damages, including base alkylation and deamination, as well as single strand break, was also widely observed for various DBPs at multiple concentrations, which is also consistent with their alkylating potential.22,23 Strong activation of nucleotide excision repair (NER) was observed for NDMA and formaldehyde, which was consistent with the DNA single or double strand breaks caused by NDMA63 and DNA-protein crosslinks led by formaldehyde,24 respectively. The similarity of DNA damage repair pathway activation revealed by our assay to previously reported genotoxicity mechanisms suggest that the information obtained from our assay may provide insights into potential genotoxicity mechanisms of those DBPs that have not been well studied. For example, the pathway activation of oxidation in BER confirmed the oxidative damage effect of 2,6-dichloro-1,4-benzoquinone, an emerging halobenzoquinone being studied in recent years. It was reported to induce cellular ROS, oxidative DNA adduct 8-OHdG, and activation of the Nrf2/ARE pathway (associated with oxidative stress) with an effect on intracellular antioxidant systems including GSH/GSSG and antioxidant enzymes.20,21,43,64

Figure 5.

Figure 5.

DNA damage repair pathway response profiles reveal distinct potential DNA damage mechanisms among different DBPs across 6-log concentrations (five DBPs were reported previously40). The mean natural log value of PELIpathway indicates the magnitude of pathway responses (represented by a black–red color scale at left; values over 1.5 are shown in the same color as 1.5). X-axis top: pathways of DNA damage repair (see Figure 1 and Table S2 for details). Y-axis left: DBPs tested in this study; Y-axis right: concentrations from lowest to highest from top to bottom (see concentrations in Table S1). Aberrations for DNA repair pathways: DDS, DNA damage signaling; TLS, translesion synthesis; DRR, direct reversal repair; BER, base excision repair; NER, nucleotide excision repair; MMR, mismatch repair; DSB, double strand break.

Examination and comparison of DNA damage and repair pathway activation profiles among DBPs suggest that genotoxicity of DBPs may be structure-dependent. For example, dichloroacetonitrile, dibromoacetonitrile, and two haloaldehydes analyzed in this study induced a wide range of pathway activations likely reflecting their strong oxidative damage to DNA structure. However, some DBPs within the same chemical class demonstrated different pathway activation patterns and magnitude. For example, chloroacetonitrile demonstrated little pathway activation compared to the other three halonitriles tested in this study. Our analyses revealed high-resolution molecular details of DNA damage effects of DBPs tested in this study.

Chemical Clustering of DBPs Based on DNA Damage Repair Pathway Protein Expression Profiles.

The high-resolution molecular genotoxicity profiling of all known DNA damage repair pathways could serve as fingerprints for DBP clustering analysis and classification. We performed hierarchical clustering using average linkage clustering and Pearson correlation as shown in Figure 6. For most DBPs, the profiles of the same DBP at varying concentrations generally clustered together as a result of the chemical-specific DNA-damaging mechanism(s). However, some DBPs (e.g., formaldehyde and iodoacetonitrile) showed concentration-sensitive DNA-damaging profiles at varying concentrations.

Figure 6.

Figure 6.

Hierarchical clustering (HCL) analysis diagram based on protein expression profiles of the 20 DBPs across six concentrations in this study (average linkage clustering, Pearson correlation). Rows represent individual experimental samples. Columns represent protein expression profiles. The mean magnitude of altered protein expression (ln I) is represented by a green-black-red color spectrum. Red spectrum colors indicate upregulation; green spectrum colors indicate downregulation. Values beyond ±1.5 are shown in the same color as ±1.5. Numbers 1–6 represent concentrations from lowest to highest (see concentrations in Table S1). X-axis top: cluster roots of protein biomarkers used in this study; X-axis bottom: DNA damage and repair pathways with color codes; Y-axis right: cluster roots and list of chemicals tested with color codes for chemical classes.

The clustering analysis revealed clear clusters of DBPs that shared high similarity in their DNA damage effects profiles, such as the clusters of chloroacetamide and dichloroacetamide, formaldehyde and iodoacetic acid, and that of trichloroacetic acid (TCA) and chloroacetonitrile. The results indicated that DBPs from the same chemical structural class may not share similar genotoxicity mechanisms as often expected. For example, bromodichloromethane—chlorodibromomethane exhibited distant profiles and so were the two aldehydes (trichloroacetaldehyde—formaldehyde). Therefore, gene or protein profiling fingerprints provided by the toxicogenomics assay can better distinguish chemicals based on their molecular toxicity mechanisms.

QSAR Physicochemical Descriptors versus Genotoxicity to Assess Mechanisms.

Quantitative structure-activity relationship (QSAR) has been employed as a diagnostic tool to assess the molecular initiating events of DBPs in the interaction with DNA.4,65 Here, we explored the QSAR modeling for predicting molecular end points PELImax and PELI1.5. For different classes of DBPs, their genotoxicity mechanism(s) may be reflected by different physicochemical properties and thus may lead to different relationships with different descriptor(s). Although only a few DBPs were tested for each chemical class in this study, linear regression could still be useful to identify potential toxicity mechanism(s) (descriptors in Table 2, linear regression in Figure S4). For example, for the four HAAs, AATSC3v (an autocorrelation descriptor of average centered Broto-Moreau autocorrelation weighted by van der Waals volumes), and Elomo (the energy of the lowest unoccupied molecular orbital) seemed to be suitable descriptors for PELImax modeling with R2 = 0.9991 (P = 0.0297). Log Kow, and Elomo could be used for their PELI1.5 prediction with R2 = 0.9183 (P=0.2858), suggesting that the electron-donating and -accepting ability (reflected by Elomo)66 and topological property (reflected by AATSC3v) may correlate to HAA genotoxicity. For the four halonitriles tested, PELImax could be predicted by AATSC3v with R2 = 0.7686 (P = 0.1233), and PELI1.5 could be predicted by AATSC3v with R2 = 0.9894 (P = 0.1379), suggesting that the genotoxicity of halonitriles may be mainly related to their topological property. For the three haloamides tested, PELImax could be predicted by ATS3p (an autocorrelation descriptor of centered Broto-Moreau autocorrelation weighted by polarizabilities) with R2 = 0.9345 (P=0.1645), and PELI1.5 could be predicted by G (Gibbs free energy) with R2 = 0.9943 (P= 0.0482). The correlation of Elomo to toxicity of haloacetic acids is consistent with other published studies4,16,25,67

Table 2.

Results of the Correlation between PELI-based and Physicochemical Descriptors

PELImax
lg(PELI1.5)
descriptor(s) r (p value)a descriptor(s) r (p value)
HAAs Elomo −0.9783 (0.0262) log Kow 0.7265 (0.2735)
AATSC3v −0.4196 (0.5804) Elomo −0.6547 (0.3453)
HANs AATSC3v 0.8767 (0.1233) AATSC3v −0.9766 (0.1379)
haloamides ATS3p −0.9667 (0.1648) G 0.9971 (0.0482)
a

r indicates the correlation for the single descriptor. The fitness of the regression of multiple descriptors is shown in Figure S4.

The QSAR modeling exercise of the DBPs suggested that chemical properties of the molecules, for example, topological and quantum chemical properties, correlate with the genotoxicity of DBPs. In addition, the correlation of different descriptors implicated different mechanism(s). For example, electron-donating and -accepting ability may be involved for DBPs in certain chemical classes.

In this study, we applied a quantitative toxicogenomics-based assay for relatively fast (2 h assay time length compared with days required as in the comet assay and micronucleus test), efficient, and mechanistic genotoxicity analyses for 20 DBPs in various chemical classes. The results provided new insights and fundamental knowledge of DNA-damage-related genotoxicity of studied DBPs and contributed to filling of the existing knowledge gap in DBP genotoxicity. PELI-based quantitative end points showed correlation with conventional genotoxicity and carcinogenicity end points and therefore can be potentially incorporated into DBP risk assessment. The pathway activation and clustering analysis based on the high-resolution protein expression profiles enabled DBP classification based on their molecular initiating patterns. The analyses also provide evidence for a structure–genotoxicity relationship of DBPs. Initial exploration of QSAR modeling using molecular genotoxicity end points (PELI) suggested that genotoxicity of DBPs in this study correlated with topological and quantum chemical descriptors. Establishment of such a unified DBPgenotoxicity database will allow identification and prediction of genotoxicity and mechanism analysis for new DBPs in water samples.4 The assay can be further applied to evaluate effects of the DBP mixtures as they present in drinking water. This study demonstrated that the quantitative toxicogenomics-based genotoxicity assay can serve as an alternative and complementary method for genotoxicity screening and evaluation of environmental water pollutants such as DBPs and provide timely guidance for drinking water safety and public health research.

Supplementary Material

Supplementary Data

ACKNOWLEDGMENTS

This study was supported by the United States National Science Foundation (NSF, CBET-1437257, CBET-1810769, IIS-1546428), National Institute of Environmental Health Sciences (NIEHS, PROTECT 3P42ES017109, and CRECE 1P50ES026049), and Environmental Protection Agency (EPA, CRECE 83615501).

Footnotes

Supporting Information

The Supporting Information is available free of charge on the ACS Publications website at DOI:10.1021/acs.est.7b06389.

Details for the 20 DBPs and their concentration range in the study, 24 h cytotoxicity of chemicals in yeast cells for concentration range selection, yeast library used in this study, physicochemical descriptors of the 20 DBPs in this study, real-time protein expression profiles of all other 18 DBPs tested and mitomycin C as reference compound in this study, results of the Comet assay in human A549 cells, and linear regression of physicochemical descriptors for the mechanistic study (PDF)

The authors declare no competing financial interest.

REFERENCES

  • (1).Richardson SD; Postigo C Formation of DBPs: state of the science In Recent Advances in Disinfection By-Products; Karanfil T, Mitch B,Westerhoff P, Xie Y, Eds.; ACS Publications: Washington, DC, 2015; pp 189–214. [Google Scholar]
  • (2).Plewa MJ; Richardson SD Disinfection By-Products in Drinking Water, Recycled Water and Wastewater: Formation, Detection, Toxicity and Health Effects: Preface. J. Environ. Sci 2017, 58 (Supplement C), 1. [DOI] [PubMed] [Google Scholar]
  • (3).Richardson S; Postigo C Drinking water disinfection by-products 2012, 1–45. [Google Scholar]
  • (4).Stalter D; O’Malley E; von Gunten U; Escher BI Fingerprinting the reactive toxicity pathways of 50 drinking water disinfection by-products. Water Res. 2016, 91, 19–30. [DOI] [PubMed] [Google Scholar]
  • (5).Richardson SD; Plewa MJ; Wagner ED; Schoeny R; DeMarini DM Occurrence, genotoxicity, and carcinogenicity of regulated and emerging disinfection by-products in drinking water: A review and roadmap for research. Mutat. Res., Rev. Mutat. Res 2007, 636 (1–3), 178–242. [DOI] [PubMed] [Google Scholar]
  • (6).Singh N; Manshian B; Jenkins GJS; Griffiths SM; Williams PM; Maffeis TGG; Wright CJ; Doak SH NanoGenotoxicology: The DNA damaging potential of engineered nanomaterials. Biomaterials 2009, 30 (23–24), 3891–3914. [DOI] [PubMed] [Google Scholar]
  • (7).Ahn JM; Hwang ET; Youn CH; Banu DL; Kim BC; Niazi JH; Gu MB Prediction and classification of the modes of genotoxic actions using bacterial biosensors specific for DNA damages. Biosens. Bioelectron 2009, 25 (4), 767–772. [DOI] [PubMed] [Google Scholar]
  • (8).Bull RJ; Reckhow DA; Rotello V; Bull OM; Kim J Use of toxicological and chemical models to prioritize DBP research; American Water Works Association: Denver, CO, 2006. [Google Scholar]
  • (9).Nieuwenhuijsen MJ; Grellier J; Smith R; Iszatt N; Bennett J; Best N; Toledano M The epidemiology and possible mechanisms of disinfection by-products in drinking water. Philos. Trans. R. Soc., A 2009, 367 (1904), 4043–76. [DOI] [PubMed] [Google Scholar]
  • (10).Reifferscheid G; Buchinger S Cell-Based Genotoxicity Testing In Whole Cell Sensing System II; Springer, 2009; pp 85–111. [Google Scholar]
  • (11).Plewa MJ; Wagner ED; Richardson SD; Thruston AD; Woo Y-T; McKague AB Chemical and biological characterization of newly discovered iodoacid drinking water disinfection byproducts. Environ. Sci. Technol 2004, 38 (18), 4713–4722. [DOI] [PubMed] [Google Scholar]
  • (12).Plewa MJ; Wagner ED Mammalian Cell Cytotoxicity and Genotoxicity of Disinfection By-Products; Water Research Foundation, 2009. [Google Scholar]
  • (13).Plewa MJ; Wagner ED; Muellner MG; Hsu K-M; Richardson SD Comparative mammalian cell toxicity of N-DBPs and C-DBPs In Occurrence, Formation, Health Effects and Control of Disinfection By-Products in Drinking Water; Karanfil T, Krasner SW, Westerhoff P, Xie Y, Eds.; ACS Publications: Washington, D.C, 2008; Vol. 995, pp 36–50. [Google Scholar]
  • (14).Pals JA; Ang JK; Wagner ED; Plewa MJ Biological mechanism for the toxicity of haloacetic acid drinking water disinfection byproducts. Environ. Sci. Technol 2011, 45 (13), 5791–5797. [DOI] [PubMed] [Google Scholar]
  • (15).Dad A; Jeong CH; Pals JA; Wagner ED; Plewa MJ Pyruvate remediation of cell stress and genotoxicity induced by haloacetic acid drinking water disinfection by-products. Environmental and molecular mutagenesis 2013, 54 (8), 629–637. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (16).Pals J; Attene-Ramos MS; Xia M; Wagner ED; Plewa MJ Human cell toxicogenomic analysis linking reactive oxygen species to the toxicity of monohaloacetic acid drinking water disinfection byproducts. Environ. Sci. Technol 2013, 47 (21), 12514–12523. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (17).Liviac D; Creus A; Marcos R Genotoxicity analysis of two halonitromethanes, a novel group of disinfection by-products (DBPs), in human cells treated in vitro. Environ. Res 2009, 109 (3), 232–238. [DOI] [PubMed] [Google Scholar]
  • (18).Plewa MJ; Wagner ED; Jazwierska P; Richardson SD; Chen PH; McKague AB Halonitromethane drinking water disinfection byproducts: chemical characterization and mammalian cell cytotoxicity and genotoxicity. Environ. Sci. Technol 2004, 38 (1), 62–68. [DOI] [PubMed] [Google Scholar]
  • (19).Ballmaier D; Epe B DNA damage by bromate: mechanism and consequences. Toxicology 2006, 221 (2), 166–171. [DOI] [PubMed] [Google Scholar]
  • (20).Procházka E; Escher BI; Plewa MJ; Leusch FD In vitro cytotoxicity and adaptive stress responses to selected haloacetic acid and halobenzoquinone water disinfection byproducts. Chem. Res. Toxicol 2015, 28 (10), 2059–2068. [DOI] [PubMed] [Google Scholar]
  • (21).Li J; Wang W; Moe B; Wang H; Li X-F Chemical and Toxicological Characterization of Halobenzoquinones, an Emerging Class of Disinfection Byproducts. Chem. Res. Toxicol 2015, 28 (3), 306–318. [DOI] [PubMed] [Google Scholar]
  • (22).Daniel FB; Schenck KM; Mattox JK; Lin EL; Haas DL; Pereira MA Genotoxic properties of haloacetonitriles: drinking water by-products of chlorine disinfection. Toxicol. Sci 1986, 6 (3), 447–53. [DOI] [PubMed] [Google Scholar]
  • (23).Muellner MG; Wagner ED; McCalla K; Richardson SD; Woo YT; Plewa MJ Haloacetonitriles vs. regulated haloacetic acids: are nitrogen-containing DBPs more toxic? Environ. Sci. Technol 2007, 41 (2), 645–51. [DOI] [PubMed] [Google Scholar]
  • (24).Vock E; Lutz W; Ilinskaya O; Vamvakas S Discrimination between genotoxicity and cytotoxicity for the induction of DNA double-strand breaks in cells treated with aldehydes and diepoxides. Mutat. Res., Genet. Toxicol. Environ. Mutagen 1999, 441 (1), 85–93. [DOI] [PubMed] [Google Scholar]
  • (25).Pals JA; Wagner ED; Plewa MJ Energy of the Lowest Unoccupied Molecular Orbital, Thiol Reactivity, and Toxicity of Three Monobrominated Water Disinfection Byproducts. Environ. Sci. Technol 2016, 50 (6), 3215–3221. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (26).Komaki Y; Mariñas BJ; Plewa MJ Toxicity of drinking water disinfection byproducts: cell cycle alterations induced by the monohaloacetonitriles. Environ. Sci. Technol 2014, 48 (19), 11662–11669. [DOI] [PubMed] [Google Scholar]
  • (27).Swenberg JA; Hoel DG; Magee PN Mechanistic and statistical insight into the large carcinogenesis bioassays on N-nitrosodiethylamine and N-nitrosodimethylamine. Cancer Res. 1991, 51 (23 Part 2), 6409–6414. [PubMed] [Google Scholar]
  • (28).Shukla SJ; Huang R; Austin CP; Xia M The future of toxicity testing: a focus on in vitro methods using a quantitative high-throughput screening platform. Drug Discovery Today 2010, 15 (23–24), 997–1007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (29).Johnson TD Report calls for examination of chemical safety: National coalition notes difficulty determining exposures. Nation’s Health 2011, 41 (6), 9–9. [Google Scholar]
  • (30).Knight AW; Little S; Houck K; Dix D; Judson R; Richard A; McCarroll N; Akerman G; Yang C; Birrell L Evaluation of high-throughput genotoxicity assays used in profiling the US EPA ToxCast chemicals. Regul. Toxicol. Pharmacol 2009, 55 (2), 188–199. [DOI] [PubMed] [Google Scholar]
  • (31).Walmsley RM; Billinton N How accurate is in vitro prediction of carcinogenicity? British journal of pharmacology 2011, 162 (6), 1250–1258. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (32).Muellner MG; Attene-Ramos MS; Hudson ME; Wagner ED; Plewa MJ Human cell toxicogenomic analysis of bromoacetic acid: A regulated drinking water disinfection by-product. Environ. Mol. Mutagen 2010, 51 (3), 205–214. [DOI] [PubMed] [Google Scholar]
  • (33).Attene-Ramos MS; Wagner ED; Plewa MJ Comparative human cell toxicogenomic analysis of monohaloacetic acid drinking water disinfection byproducts. Environ. Sci. Technol 2010, 44 (19), 7206–7212. [DOI] [PubMed] [Google Scholar]
  • (34).Mahadevan B; Snyder RD; Waters MD; Benz RD; Kemper RA; Tice RR; Richard AM, Genetic Toxicology in the 21st Century: Reflections and Future Directions. Environmental and molecular mutagenesis 2011, 52 (5), 339–354. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (35).Walmsley R; Billinton N; Heyer W Green fluorescent protein as a reporter for the DNA damage-induced gene RAD54 in Saccharomyces cerevisiae. Yeast 1997, 13 (16), 1535–1545. [DOI] [PubMed] [Google Scholar]
  • (36).Cahill P; Knight A; Billinton N; Barker M; Walsh L; Keenan P; Williams C; Tweats D; Walmsley R The Green-Screen® genotoxicity assay: a screening validation programme. Mutagenesis 2004, 19 (2), 105–119. [DOI] [PubMed] [Google Scholar]
  • (37).Attene-Ramos MS; Huang R; Sam M; Witt KL; Richard A; Tice RR; Simeonov A; Austin CP; Xia M Profiling of the Tox21 chemical collection for mitochondrial function to identify compounds that acutely decrease mitochondrial membrane potential. Environmental Health Perspectives (Online) 2015, 123 (1), 49. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (38).O’Connor STF; Lan J; North M; Loguinov A; Zhang L; Smith MT; Gu AZ; Vulpe C Genome-wide functional and stress response profiling reveals toxic mechanism and genes required for tolerance to benzo [a] pyrene in S. cerevisiae. Front. Genet 2013, 3, 316. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (39).Lan J; Hu M; Gao C; Alshawabkeh A; Gu AZ Toxicity Assessment of 4-Methyl-1-cyclohexanemethanol and Its Metabolites in Response to a Recent Chemical Spill in West Virginia, USA. Environ. Sci. Technol 2015, 49 (10), 6284–6293. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (40).Lan J; Gou N; Rahman SM; Gao C; He M; Gu AZ A quantitative toxicogenomics assay for high-throughput and mechanistic genotoxicity assessment and screening of environmental pollutants. Environ. Sci. Technol 2016, 50 (6), 3202–3214. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (41).Lan J; Gou N; Gao C; He M; Gu A Comparative and Mechanistic Genotoxicity Assessment of Nanomaterials via A Quantitative Toxicogenomics Approach Across Multiple Species. Environ. Sci. Technol 2014, 48 (21), 12937–45. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (42).Huh WK; Falvo JV; Gerke LC; Carroll AS; Howson RW ; Weissman JS; O’Shea EK. Global analysis of protein localization in budding yeast. Nature 2003, 425 (6959), 686–91. [DOI] [PubMed] [Google Scholar]
  • (43).Zuo Y-T; Hu Y; Lu W-W; Cao J-J; Wang F; Han X; Lu W-Q; Liu A-L Toxicity of 2,6-dichloro-1,4-benzoquinone and five regulated drinking water disinfection by-products for the Caenorhabditis elegans nematode. J. Hazard. Mater 2017, 321, 456–463. [DOI] [PubMed] [Google Scholar]
  • (44).Zhang S; Miao D; Tan L; Liu A; Lu W Comparative cytotoxic and genotoxic potential of 13 drinking water disinfection byproducts using a microplate-based cytotoxicity assay and a developed SOS/umu assay. Mutagenesis 2015, 31 (1), 35–41. [DOI] [PubMed] [Google Scholar]
  • (45).O’Connor STF; Lan J; North M; Loguinov A; Zhang L; Smith MT; Gu AZ; Vulpe C Genome-wide functional and stress response profiling reveals toxic mechanism and genes required for tolerance to benzo [a] pyrene in S. cerevisiae. Front. Genet 2013, 3, 1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (46).Nakatani Y; Yamada R; Ogino C; Kondo A Synergetic effect of yeast cell-surface expression of cellulase and expansin-like protein on direct ethanol production from cellulose. Microb. Cell Fact 2013, 12 (1), 66. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (47).Gou N; Gu AZ A New Transcriptional Effect Level Index (TELI) for Toxicogenomics-based Toxicity Assessment. Environ. Sci. Technol 2011, 45 (12), 5410–5417. [DOI] [PubMed] [Google Scholar]
  • (48).Escher BI; Bramaz N; Mueller JF; Quayle P; Rutishauser S; Vermeirssen EL Toxic equivalent concentrations (TEQs) for baseline toxicity and specific modes of action as a tool to improve interpretation of ecotoxicity testing of environmental samples. J. Environ. Monit 2008, 10 (5), 612–21. [DOI] [PubMed] [Google Scholar]
  • (49).Gou N; Yuan S; Lan J; Gao C; Alshawabkeh AN; Gu AZ A quantitative toxicogenomics assay reveals the evolution and nature of toxicity during the transformation of environmental pollutants. Environ. Sci. Technol 2014, 48 (15), 8855–63. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (50).Salamone M; Heddle J; Stuart E; Katz M Towards an improved micronucleus test Studies on 3 model agents, mitomycin C, cyclophosphamide and dimethylbenzanthracene. Mutation Research/fundamental & Molecular Mechanisms of Mutagenesis 1980, 74 (5), 347–56. [DOI] [PubMed] [Google Scholar]
  • (51).Weber S; Hebestreit M; Wilms T; Conroy LL; Rodrigo G Comet assay and air–liquid interface exposure system: a new combination to evaluate genotoxic effects of cigarette whole smoke in human lung cell lines. Toxicol. In Vitro 2013, 27 (6), 1987–1991. [DOI] [PubMed] [Google Scholar]
  • (52).Godschalk RW .; Ersson C; Riso P; Porrini M; Langie SA; van Schooten F-J; Azqueta A; Collins AR; Jones GD; Kwok RW. DNA-repair measurements by use of the modified comet assay: an inter-laboratory comparison within the European Comet Assay Validation Group (ECVAG). Mutat. Res. Genet. Toxicol. Environ. Mutagen 2013, 757 (1), 60–67. [DOI] [PubMed] [Google Scholar]
  • (53).Bausinger J; Schütz P; Piberger AL; Speit G Further characterization of benzo [a] pyrene diol-epoxide (BPDE)-induced comet assay effects. Mutagenesis 2016, 31 (2), 161–169. [DOI] [PubMed] [Google Scholar]
  • (54).Yap CW PaDEL-descriptor: an open source software to calculate molecular descriptors and fingerprints. J. Comput. Chem 2011, 32 (7), 1466–74. [DOI] [PubMed] [Google Scholar]
  • (55).Saeed AI; Bhagabati NK; Braisted JC; Liang W; Sharov V; Howe EA; Li J; Thiagarajan M; White JA ; Quackenbush J TM4Microarray Software Suite. Methods Enzymol. 2006, 411, 134–193. [DOI] [PubMed] [Google Scholar]
  • (56).King DJ; Wiseman A; Wilkie D Studies on the genetic regulation of cytochrome P-450 production in Saccharomyces cerevisiae. Mol. Gen. Genet 1983, 192 (3), 466–70. [DOI] [PubMed] [Google Scholar]
  • (57).Kappeli O Cytochromes P-450 of yeasts. Microbiol Rev. 1986, 50 (3), 244–258. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (58).Muller-Pillet V; Joyeux M; Ambroise D; Hartemann P Genotoxic activity of five haloacetonitriles: comparative investigations in the single cell gel electrophoresis (comet) assay and the ames-fluctuation test. Environ. Mol. Mutagen 2000, 36 (1), 52–8. [DOI] [PubMed] [Google Scholar]
  • (59).Kundu B; Richardson SD; Swartz PD; Matthews PP; Richard AM; Demarini DM Mutagenicity in Salmonella of halonitromethanes: a recently recognized class of disinfection byproducts in drinking water. Mutat. Res. Genet. Toxicol. Environ. Mutagen 2004, 562 (1–2), 39–65. [DOI] [PubMed] [Google Scholar]
  • (60).Gold LS; Slone TH; Bernstein L Summary of Carcinogenic Potency and Positivity for 492 Rodent Carcinogens in the Carcinogenic Potency Database. Environ. Health Persp 1989, 79, 259–272. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (61).Fujita K-i.; Kamataki T. Role of human cytochrome P450 (CYP) in the metabolic activation of N-alkylnitrosamines: application of genetically engineered Salmonella typhimurium YG7108 expressing each form of CYP together with human NADPH-cytochrome P450 reductase. Mutat. Res., Fundam. Mol. Meek. Mutagen 2001, 483 (1–2), 35–41. [DOI] [PubMed] [Google Scholar]
  • (62).Pals J Mechanisms of monohalogenated acetic acid induced genomic DNA damage; University of Illinois at Urbana-Champaign, 2014. [Google Scholar]
  • (63).Arranz N; Haza AI; García A; Rafter J; Morales P Protective effect of vitamin C towards N-nitrosamine-induced DNA damage in the single-cell gel electrophoresis (SCGE)/HepG2 assay. Toxicol. In Vitro 2007, 21 (7), 1311–1317. [DOI] [PubMed] [Google Scholar]
  • (64).Li J; Moe B; Vemula S; Wang W; Li X-F Emerging Disinfection Byproducts, Halobenzoquinones: Effects of Isomeric Structure and Halogen Substitution on Cytotoxicity, Formation of Reactive Oxygen Species, and Genotoxicity. Environ. Sci. Technol 2016, 50 (13), 6744–6752. [DOI] [PubMed] [Google Scholar]
  • (65).Kundu B; Richardson SD; Granville CA; Shaughnessy DT; Hanley NM; Swartz PD; Richard AM; DeMarini DM Comparative mutagenicity of halomethanes and halonitromethanes in Salmonella TA100: structure–activity analysis and mutation spectra. Mutat. Res., Fundam. Mol. Meek. Mutagen 2004, 554 (1), 335–350. [DOI] [PubMed] [Google Scholar]
  • (66).Wang H; Wang X; Wang H; Wang L; Liu A DFT study of new bipyrazole derivatives and their potential activity as corrosion inhibitors. J. Mol. Model 2006, 13 (1), 147–153. [DOI] [PubMed] [Google Scholar]
  • (67).Richard AM; Hunter ES Quantitative structure-activity relationships for the developmental toxicity of haloacetic acids in mammalian whole embryo culture. Teratology 1996, 53 (6), 352–360. [DOI] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Supplementary Materials

Supplementary Data

RESOURCES