Re-evaluation of the risks to public health related to the presence of bisphenol A (BPA) in foodstuffs - Genotoxicity

Last updated: 02 August 2022

Re-evaluation of the risks to public health related to the presence of bisphenol A (BPA) in foodstuffs - Genotoxicity

Genotoxicity - Background

Previous 2015 EFSA conclusion

1.     In the 2015 EFSA opinion on BPA (EFSA CEF Panel, 2015), the CEF Panel concluded that BPA is not mutagenic (in bacteria or mammalian cells), or clastogenic (micronuclei and chromosomal aberrations). The potential of BPA to produce aneuploidy in vitro was not expressed in vivo. The positive findings in the post labelling assays in vitro and in vivo were judged unlikely to be of concern, given the lack of mutagenicity and clastogenicity of BPA in vitro and in vivo.

Current new data examined, literature search timeline and screening methodology

2.      For the health outcome category (HOC) genotoxicity, the time span of the literature search was extended until 21 July 2021 and the studies assessed in the 2015 EFSA opinion were re-considered.

3.      The methods that were used for data collection through literature searches were conducted in the following bibliographic databases: PubMed, Web of Science and Core Collection.

4.     For the additional time span considered in the literature search, the screening question was: ‘Is the paper reporting information about exposure to BPA and genotoxicity?’

5.     For screening the additional genotoxicity studies, the categorisation was made into different subgroups of genotoxicity endpoints (genotoxicity, epigenetics, oxidative stress). An additional screening of the relevance of the studies was done by experts in this field following the full-text screening.

6.     A specific internal validity approach was applied and a specific Weight of Evidence (WoE) approach was applied, as described in detail in Annex A. The CEP Panel examined whether new data from the published literature could provide new evidence on the potential genotoxicity of BPA. the references from the previous CEF Panel opinion (EFSA CEF Panel, 2015) have also been included in the current assessment using the same appraisal criteria applied to the newly published data and considering the EFSA Scientific Committee guidance documents on genotoxicity published after 2015 (EFSA Scientific Committee, 2017, 2021).

Re-evaluation of the risks to public health related to the presence of bisphenol A (BPA) in foodstuffs - Genotoxicity

Methods for assessing genotoxicity

Methods for assessing genotoxicity

7.     The evaluation of data quality for hazard/risk assessment includes the evaluation of reliability and relevance (Klimisch et al., 1997; OECD, 2005; ECHA, 2011; EFSA Scientific Committee, 2017c; EFSA Scientific Committee, 2021).

8.     In the assessment of genotoxicity studies, the data quality has been evaluated based on reliability and relevance. Reliability has been assessed using a scoring system based on criteria published by Klimisch et al. (1997).

9.     In a second step, the relevance (high, limited, low) of the study results was assessed based on reliability of the study and other aspects, e.g. genetic endpoint, purity of test substance, route of administration and status of validation of the assay.

10.     Genotoxicity studies evaluated as of high or limited relevance have been considered in a WoE approach as described in Annex A. Genotoxicity studies evaluated as of low relevance have not been further considered in the assessment.

The different steps of the evaluation of reliability and relevance are described in Annex A.

Method for uncertainty analysis for genotoxicity
 

11.    Details on how the uncertainty analysis was carried out as well as the results discussion can be found in Annex A.

Genotoxicity studies considered for this assessment

12.    Publication of 88 in vitro and in vivo studies were retrieved from the literature search:

• in vitro and in vivo studies (15 publications) considered in the Scientific opinion on the risks to public health related to the presence of BPA in foodstuffs (EFSA CEF Panel, 2015) (Annex A).

13.    In vitro and in vivo studies were grouped based on the genotoxicity endpoint investigated:

• gene mutations (e.g. bacterial reverse mutation assay);

• chromosomal damage (CA and micronucleus assays);

• DNA damage (comet assay).

14.    These studies were summarized in synoptic tables (Annex A), evaluated for reliability and relevance and grouped into lines of evidence in a WoE approach (Annex A).

15.    Studies not investigating classical genotoxicity endpoints (e.g. γH2AX, oxidative DNA damage, DNA binding, ROS generation) and studies in humans are considered in the Mode of Action MoA and as supportive evidence.

Re-evaluation of the risks to public health related to the presence of bisphenol A (BPA) in foodstuffs - Genotoxicity

Weight of evidence

Gene mutations in vitro and in vivo

In vitro gene mutation

16.    Of the six available studies of the mutagenicity of BPA in bacteria, only one describes the application of the Ames test in a comprehensive battery of Salmonella Typhimurium strains (TA1535, TA97, TA98, 11407 TA100 and TA102) at a range of concentrations up to 5000 μg/plate. It reports negative results both in the presence and absence of metabolic activation (Xin et al., 2015).

17.    Three studies reported negative results in TA98 and TA100 (Masuda et al., 2005; Fic et al., 2013; Zemheri and Uguz, 2016). A study shows negative results in TA98, TA100 and TA102 strains (Tiwari et al., 2012). The sixth used the bacterial SOS/umuC assay with a range of concentrations from 1 to 1000 μg/L in presence and absence of S9 mix. It also reported negative results (Balabanič et al., 2021). The CEP Panel concluded that BPA does not induce gene mutations in bacteria.

Summaries of studies

18.    Summary of Xin et al study 2015: The study evaluated the cytotoxic, genotoxic and clastogenic activity of BPA (purity 99%) in Chinese hamster ovary cells (CHO) cells and its mutagenicity in the Ames test. The battery of assays applied in CHO cells included the MTT assay for the evaluation of cytotoxicity, and the comet, micronucleus and chromosome aberration tests. In the Ames test, BPA (10-5000 μg/plate) was uniformly negative in all Salmonella Typhimurium strains (TA1535, TA97, TA98, TA100 and TA102), with and without metabolic activation. Exposure of CHO cells to four BPA doses (40, 80, 100 and 120 μM) for 12 and 24 h resulted in a significant decrease in cell viability (at 80 μM and above), which however remained above 50% in all cases; a concentration-related increase of DNA damage was observed in comet assay [increased Olive tail moment (OTM), tail length and % tail DNA, statistically significant at all doses] after 12 and 24 h exposure to BPA; after 24 h treatment, an increase in micronuclei (MN) (statistically significant at 100 and 120 μM) and structural chromosomal aberrations (chromatid breaks and chromosome fragments, statistically significant at 80 μM and above) was also observed.

19.    Summary of Masuda et al., 2005: The study evaluated the mutagenicity of BPA in Ames test in the presence or absence of S9-mix. BPA (Tokyo Kasei Kogyo Co., Ltd) was tested on S. Typhimurium strains TA98 and TA100 at the single dose of 0.1 μmole/plate (100 μL of 1 mM solution). No mutagenic effect was observed.

20.    Summary of Fic et al., 2013: In this study the mutagenic and genotoxic potential of eight BPA (purity >99%) structural analogues [BPF, BPAF, bisphenol Z (BPZ), BPS, bis(4-hydroxy-3-methylphenyl)propane (DMBPA), 4,4’-sulfonylbis(2-methylphenol) (DMBPS), [sulphonylbis(benzene-4,1-diyloxy)]diethanol (BP-1), and 4,4’-sulphanediyldiphenol (BP-2)] were investigated using the Ames and comet assay. None of these bisphenols were mutagenic in Salmonella Typhimurium strains TA98 and TA100 either in the presence or absence of external S9-mediated metabolic activation (Aroclor 1254-induced male rat liver). Potential genotoxicity of bisphenols was determined in the HepG2 human hepatoma cell line following 4-h and 24-h exposure to non-cytotoxic concentrations 0.1 μmol/L to 10 μmol/L. In the comet assay, BPA and its analogue BPS induced significant DNA damage only after the 24-h exposure, while analogues DMBPS, BP-1, and BP-2 induced a transient increase in DNA strand breaks observed only after the 4-h exposure. BPF, BPAF, BPZ, and DMBPA did not induce DNA damage.

21.    Summary of Zemheri and Uguz, 2016: The study evaluated the mutagenicity of BPA (Merck) in a limited Ames test, using two tester strains (TA98 and TA100) and four dose levels (0.1, 1, 10 and 100 μg/plate). The results were negative, with and without metabolic activation.

22.    Summary of Tiwari et al., 2012: The study evaluated the mutagenicity of BPA in Ames test. BPA (purity 99%) was tested at concentrations from 6.25 to 200 μg/plate on different strains of S. Typhimurium (TA 98, TA 100 and TA 102). The mutagenic response was not observed in any of the tester strains at the various concentration of BPA in absence of S9 fractions. A slight increase in the numbers of revertants was observed in the presence of S9 fractions from the 6.25 - 25 μg/plate of BPA in each strain, but the increase was statistically significant only in strain TA 102 at 25 μg/plate.

23.    Summary of Balabanič et al., 2021: The study evaluated cytotoxic and genotoxic effects of some endocrine disrupting chemicals (EDCs), including BPA, which have been previously identified in effluents from two paper mills. BPA (Sigma-Aldrich) tested at concentrations of 1, 10, 100, 1000 μg/L with the bacterial SOS/umuC assay in S. Typhimurium TA1535/pSK1002 strain did not induce toxic nor genotoxic effects in the presence or absence of S9 metabolic activation. The compound was also assessed in HepG2 cells with MTT assay for cell viability and with comet assay at 1, 10, 100 and 1000 μg/L for 4 and 24 h. No significant reduction of the viability. A statistically significant concentration-dependent increase of DNA damage, expressed as percent of DNA in tail, was reported starting from 10 μg/L.

In vivo gene mutation

24.    No studies on gene mutation assays in mammalian cells following the OECD guidelines were available.

Induction of chromosomal aberrations/micronuclei in vitro and in vivo

In vitro chromosomal aberrations/micronuclei

25.    Fifteen in vitro studies of micronuclei (MN) and structural chromosomal aberrations (CA) induction in different cell lines were available for evaluation. Of these, nine were further considered in the assessment, classified as having high (1 study) or limited relevance (8 studies).

26.    All showed positive results in both blood cells and established cell lines. In the single study classified as of high relevance, a concentration-dependent increase of MN frequency over a wide range of concentrations (1.5 to 37 μg/ml corresponding to 6.6 μM and 162 μM) was observed in the AHH-1 human lymphoblastoid cell line (Johnson and Parry, 2008). Positive CA results were also reported from cultures of human peripheral lymphocytes in two studies with limited relevance (Santovito et al., 2018; Di Pietro et al., 2020). In one of these (Santovito et al., 2018), MN frequency was also measured. A study of MN in bovine peripheral blood lymphocytes also reported positive findings (Šutiaková et al., 2014).

27.    In murine macrophage RAW264.7 cells, positive MN results were associated with an increase in reactive oxygen species (ROS), and a decreased level of antioxidant enzymes (GPx, SOD and CAT. Concomitant phosphorylation of P53 and release of cytochrome C from mitochondria were detected along with increased apoptosis. Pretreatment with N-acetylcysteine (NAC) reduced BPA-induced cytotoxicity, apoptosis and genotoxicity (MN frequency was reduced by 30%). These results indicate that the toxic effect of BPA in macrophages was mainly through the oxidative stress-associated mitochondrial apoptotic pathway (Huang FM et al., 2018).

28.    Finally, two studies in the Chinese hamster ovary (CHO) and V79 cell lines reported positive results (Xin et al., 2015; Yu et al., 2020). Xin and co-workers reported a concentration dependent increase of both MN and CAs in CHO cells in the absence of metabolic activation. In contrast, the BPA-induced increase in MN frequency in V79, reported by Yu and colleagues, apparently required CYP1A1 and CYP1B1 expression.

29.    Overall, the significant increases of chromatid and chromosome breaks observed in several studies in vitro indicated that BPA has clastogenic activity also at non-cytotoxic concentrations. Two reports indicated that oxidative stress is implicated in the observed induction of chromosomal damage. In addition, Johnson and Parry (2008) reported the formation of aberrant mitotic spindles, with multiple poles, in cells treated with BPA.

30.    In conclusion, the in vitro studies on CA and MN induced by BPA indicated that both clastogenic[1] and aneugenic[2] mechanisms may operate.

Summary of studies

31.    John and Parry 2008: In this mechanistic study the aneugenicity of two known spindle poisons model compounds, namely rotenone and BPA, has been investigated following low dose-exposure to mammalian cells, using the cytokinesis blocked micronucleus assay (CBMA)  and immunofluorescence methods to visualize modifications of the microtubule organizing centres (MTOCs) of the mitotic spindles. For induction of MN BPA (Sigma-Aldrich) was added over a range of narrowed low concentrations (1.5, 3.1, 6.2, 7.7, 9.2, 10.8, 12.3, 18.5, 24.6, and 37.0 μg/ml) to cultures of human (AHH-1) lymphoblastoid cell line for a complete cell cycle (22-26 h dependent upon any cell cycle delay) in the presence of cytochalasin-B. A minimum of five separate experiments were performed. A concentration-related and statistically significant increase of binucleate-micronucleated cells from 12.3 μg/mL was reported with a clear threshold for induction of MN (NOEL at 10.80 μg/mL and LOEL at 12.3 μg/mL). A NOEL and LOEL for percentage of binucleate cells was also observed at 9.2 μg/mL and 10.8 μg/mL BPA respectively. For mechanistic evaluation of the aneugenic effects of BPA, fluorescently labelled antibodies were used to visualize microtubules (α-tubulin) and MTOCs (γ-tubulin) in V79 culture. BPA in this case was added to V79 cells growing on sterile glass microscope slides placed in Petri dishes at concentrations 4.2, 4.9, 5.6, 7.0, 8.4, 9.8, 11.2 and 14 μg/mL for 20 h (i.e. one cell cycle for V79). Similarly for induction of aberrations in the mitotic machinery a NOEL was observed at 7.0 μg/mL and a LOEL at 8.4 μg/mL BPA in V79 cells. Aberrant mitotic divisions, in the form of multiple spindle poles were detected and it was suggested by the study authors to be the mechanism for the production of chromosome loss into MN.

32.    Santovito et al., 2018: In this study the possible induction of chromosomal damage by BPA (Sigma-Aldrich) was tested in human peripheral blood lymphocytes cultures applying the CA assay and the micronucleus test (MN). Cell cultures were exposed to a range of concentrations from 0.01 to 0.20 μg/mL, (including the reference dose established by United States Environmental Protection Agency (US EPA) (0.05 μg/mL), the tolerable daily intake established by European Union (0.01 μg/mL) and the highest concentration of unconjugated BPA found in human serum (0.02 μg/mL)) for 24 h for the chromosomal aberration test and for 48 for the micronucleus test. A statistically significant increase of cells with structural chromosomal aberrations, with a prevalence of chromatid breaks, was reported starting from 0.05 μg/mL; no numerical aberration was observed. A concentration related increase in MN frequency was detected starting from 0.02 μg/mL in which a four-fold increase with respect to the control level was observed.

33.    Di Pietro et al., 2020: The study investigated the effects of BPA exposure on cell proliferation, cell cycle progression and DNA damage in human peripheral blood mononuclear cells (PBMC) and the BPA-induced neurotoxicity in rats exposed to environmental relevant doses of BPA during development. Human PBMC from five unrelated healthy donors (adult males and females) were cultured and treated with BPA (Merck) from 5 nM to 200 μM. The treatment with BPA of unstimulated resting PBMC did not affect cell proliferation (determined by the colorimetric MTT) at all the concentrations tested except for 200 μM for which a marked inhibition of cell proliferation was observed at 24 and 48 h after the treatment. By contrast, in PHA-stimulated cells, BPA caused a pronounced increase of cell growth starting from 10 nM to 100 nM and a concentration-dependent decrease of cell proliferation from 25 to 200 μM. The cell cycle was analyzed by flow cytometry. BPA at 50 nM increased the percentage of cells in S phase of the cell cycle at 24 h and this effect was higher at 48 h with an increase of about 17% of cells in the S phase compared with the control. At 100 μM, BPA induced a significant increase of the percentage of cells in the G0/G1 phase, suggesting that BPA affected cell growth in a non-monotonic way. BPA-treatment at 25, 50 and 100 nM for 48 h induced a significant increase (p < 0.001) of both the percentage of aberrant cells (about 20% at 100 nM) and structural aberrations (about 27% at 100 nM) including chromatid and chromosome breaks, rings and fragments. BPA also increased significantly the percentage of highly fragmented metaphases (shattered cells). In PHA-stimulated PBMC treated with BPA (50 nM) for 24 h, γH2AX was significantly increased in CD3+ T lymphocytes and was also detected in a higher proportion of CD8+ T lymphocytes than the CD4+ T lymphocytes and a slight percentage of γH2AX was reported among the B cells. The treatment of PHA-stimulated PBMC with BPA (50 nM) induced p21/Waf1 and PARP1 protein expressions approximately within the same time interval. These findings suggest that BPA could affect the p53-p21/ Waf1 checkpoint and PARP1 levels resulting in DNA damage repair defects. BPA (50 nM) for 24 h modulated the expression of ER-α and ER-β in both sexes inducing or inhibiting its expression in males and in females with effects similar to the variations induced by pharmacological concentrations of E2 (100 nM). The study investigated also the BPA-induced neurotoxicity in terms of DNA damage. After the coupling period, three females/group received BPA (0.1 mg/L), or vehicle (ethanol 0.1 mL/L) in the drinking water during gestation, lactation and weaning of their offspring. Five females and three males pups from BPA-exposed mothers and five females and three males newborns from vehicle-treated dams were then sacrificed at PND 17. BPA was shown to induce ɣH2AX phosphorylation in cells possessing immune function in the CNS, such as microglia and astrocytes of rat hippocampus. In BPA-exposed rats a marked decreasing trend of ERα expression was found therefore proposing a role for this receptor in the effects induced by BPA.

34.    Šutiaková et al., 2014: The study evaluated the genotoxic and cytotoxic effects of BPA (Sigma-Aldrich) on bovine peripheral lymphocytes in vitro. Lymphocyte cultures from two animals were exposed to four different concentrations of BPA (1×10−4, 1×10−5, 1×10−6 and 1×10−7 mol. L−1) 24 h after stimulation by L-phytohemagglutinin, and incubated for total 72 h. Micronucleus frequency was determined using the cytokinesis block method, adding 6 μg/mL cytochalasin B at 44 h. A significant increase in the number of MN (p= 0.018) was observed at the highest concentration of BPA; at lower concentrations micronucleus frequency was not significantly different from vehicle (DMSO) control. The nuclear division index (NDI) was not affected by BPA treatment at any concentration level.

35.    Huang FM et al., 2018: The study reported positive results for induction of DNA strand breaks (evaluated by comet assay) and MN frequency in murine macrophage RAW264.7 cells. Cell cultures were treated at 0, 3, 10, 30, and 50 μM of BPA (Sigma-Aldrich) dissolved in DMSO for 24 h. Concentration-dependent increase of tail length, based on the analysis of 50 cells/slide, and of MN frequency by the evaluation of 1000 binucleated cells per concentration were observed. No positive controls were used. The genotoxic effects were observed starting from 10 μM and were associated with an increase of reactive oxygen species (ROS), measured by Dichlorofluorescein Diacetate Assay (DCFH-DA) and a decrease of antioxidant enzymes, including GPx, SOD and CAT. Concomitant phosphorylation of P53 and release of cyto C from mitochondria into cytosol were reported. A reduced expression of antiapoptotic proteins BCL2 and BCL-XL significant from 10 and 3 μM respectively and an increase of the expression of proapoptotic proteins BAX, BID, and BAD beginning at 10, 10 and 30 μM respectively were observed in a concentration-dependent manner. Increased level of the apoptosis-inducing factor (AIF) in the nucleus and a decrease in the mitochondria was detected. Expression of pro-caspase-3 and pro-caspase-9 is reduced by BPA in a concentration-dependent manner and PARP-1 cleavage was induced by BPA. Pre-treatment of the cell cultures with N-acetylcysteine (NAC), a cysteine precursor of the antioxidant glutathione, at the concentration of 10 μM for 30 min reduced BPA-induced cytotoxicity, apoptosis, and genotoxicity. The results of this study indicates that the toxic effects induced by BPA in macrophages was mainly through oxidative stress-associated mitochondrial apoptotic pathway.

35.    Xin et al 2015: See summary in the in vitro gene mutation section.

36.    Yu et al 2020: In this study, induction of MN and double-strand DNA breaks by BPA, BPF, and BPS were investigated in Chinese hamster V79-derived cell lines expressing various human CYP enzymes and a human hepatoma (C3A) (metabolism-proficient) cell line. In a first step a prediction of BPA, BPF, and BPS as potential substrates for several human CYP enzymes, which are commonly involved in the metabolic activation of compounds, was conducted by molecular docking. The results of the analysis showed a similar affinity of the compound with all the enzymes tested: CYP1A1, 1A2, 1B1, 2B6, 2E1, and 3A4. BPA (99.6% analytical purity) tested at 40, 80 and 160 μM for 9 h, followed by 15 h of recovery induced a concentration related increase of MN frequency in V79-hCYP1A1. In V79-hCYP1B1 cells MN were observed only at the two highest concentrations. No induction of MN was reported in V79-Mz, V79-hCYP1A2, V79-hCYP2E1, or V79-hCYP3A4-hOR cells. A consistency with the results of the molecular.

In vivo chromosomal aberrations/micronuclei

37.    Eleven in vivo studies addressing BPA-induced MN and structural CA after oral exposure were evaluated. After a screening for the reliability and relevance of the results, six studies from four publications, all ranked as of limited relevance, were selected for further consideration (Table 1). Of these, three studies were considered positive for the induction of MN and CA in the same publication (Tiwari et al., 2012) or of MN (Panpatil et al., 2020 ) in rats following daily oral BPA administrations for 6 and 28 days, respectively. Tiwari et al. (2012) applied a range of doses from 2.4 μg up to 50 mg/kg bw per day. In a separate publication, the same authors (Tiwari and Vanage, 2017) reported that these experimental conditions were associated with the induction of lipid peroxidation (malonaldehyde, MDA) and oxidative stress (decreased SOD, CAT, GSH) in rat bone marrow and peripheral blood lymphocytes. In Panpatil et al. (2020) the dose range was much lower (50 and 100 μg/kg bw per day). A fourth study tested positive in the mouse bone marrow MN test after the administration of a daily dose of 50 mg/kg bw for 28 days in presence of high level of cytotoxicity (Fawzy et al., 2018). A study by Naik and Vijayalaxmi (2009) reported negative findings in the mouse bone marrow MN test and CAs following a single dose in the range 10 to 100 mg/kg bw.

38.    Overall, the available data provided evidence of chromosomal damage after multiple oral administrations but not after single oral administration of BPA.

Table 1. Summary table of test results of MN and CAs in vivo studies.

Test System

Dose

Results

Reference

MN and CA in bone marrow
Swiss albino mice

6 animals /group

10, 50 and 100 mg/kg bw, single dose by gavage; 10 mg/kg for 5 days (50mg by gavage

Negative

No significant decrease of PCE/NCE ratio but significant increase of gaps and C mitoses.

Naik and Vijayalaxmi, 2009

MN in bone marrow
Holtzman rats

10 animals /group

2.4µg, 10 µg, 5 mg snf 50 mg/kg bw per day orally for 6 days

Positive

Dose related increase of CA and MN PCE starting from 10 µg

Tiwari et al., 2012

MN in bone marrow
Male Swiss albino mice
10 animals /group

50 mg/kg bw per day orally for 28 days

Positive

Significant reduction in the ratio of PCE/NCE

Fawzy et al., 2018

MN in bone marrow Male Wistar rats

6 animals / group

50 and 100 µg/kg/bw per day orally for 28 days

Positive

Dose related increase of MDA in blood and of urinary 8OHdG

Panpatil et al., 2020.

Source:  Re-evaluation of the risks to public health related to the presence of bisphenol A (BPA) in foodstuffs, EFSA, (2021)

Summary of studies

40.    Tiwari et al., 2012: This study was aimed to assess potential genotoxic effects of BPA (Sigma-Aldrich) in rats (five males and five females per group) following oral administration of test compound once a day for 6 consecutive days at dose-levels of 2.4 μg, 10 μg, 5 mg and 50 mg/kg bw by measuring induction of MN and structural chromosome aberrations in bone marrow cells and primary DNA damage in blood lymphocytes using single cell gel electrophoresis (comet assay). Furthermore, plasma concentrations of 8-hydroxydeoxyguanosine (8-OHdG), lipid peroxidation and glutathione activity were evaluated to assess potential induction of oxidative DNA damage. Results obtained for genotoxicity endpoints show marked dose-related increases of both MN and structural chromosome aberrations in bone marrow cells of male and female rats exposed to BPA. The observed increases achieved statistical significance at dose-levels as low as 10 μg/kg bw per day. Similarly, primary DNA damage evaluated by comet assay, in isolated peripheral blood lymphocytes showed marked and dose-related increases that were statistically significant at dose-levels as low as 10 μg/kg bw per day. Significant increase in plasma concentration of 8-OHdG was detected only at 50 mg/kg bw. A dose-related increase of malonaldehyde and decrease of glutathione were observed in liver.

41.    Panpatil et al., 2020: The study evaluated the protective action of turmeric acid on the genotoxic effects of BPA in Wistar rats. Six groups of six animals were administered with BPA (Sigma-Aldrich) at 0, 50 and 100 μg/kg by oral gavage for a period of 4 weeks: three groups were fed with a normal diet, the others with a diet containing 3% turmeric. At the end of the experiment the animals were sacrificed. Urine was collected 24 h before the sacrifice. 8-OHdG was measured in urine using an ELISA kit. DNA damage by comet assay was evaluated in blood, liver and kidney: 50 cells per slide were counted twice. Micronucleus assay was applied in bone marrow: 2000 PCE were evaluated. A weak but statistically significant and dose related increase of tail length was observed in liver. In kidney an increase of DNA damage was observed only at the dose of 50 μg/kg. A dose related increase of 8-OHdG in urine and of the concentration of MDA in blood serum was observed. A dose related increase of MNPCE was reported associated with a low decrease of the PCE/NCE ratio. A significant decrease of the genotoxic effects was observed in animal fed with diet with turmeric.

42.    Tiwari and Vanage, 2013: This study investigated the induction by BPA of dominant lethal mutations in the different stages of spermatogenesis in the rat. Furthermore, the induction of DNA damage by BPA in epididymal sperm was investigated. Holtzman male rats (7 per group) were treated by oral gavage with BPA (Sigma Chemical Co.) dissolved in ethyl alcohol and diluted in sesame oil, at dose-levels of 10 μg/kg bw and 5 mg/kg bw once a day for 6 consecutive days. Negative controls were treated with vehicle. Each treated male was mated with two females per week over a period of eight weeks. The mated females were then sacrificed on the day 15th of their gestation and uterine content examined. DNA damage in epididymal sperm was evaluated by alkaline comet assay in sperm samples from treated males (4 animals per group) sacrificed after completion of the mating phase. In the dominant lethal study, a significant decrease in total implants/female and live implants/female, with a concurrent significant increase in the number of resorbed embryos per female, was observed during the fourth week and sixth week in females mated with males treated with 5 mg BPA/kg bw, suggesting the induction of post-implantation loss due to dominant lethal mutations in mid-spermatids and spermatocytes. No significant change was observed in the pre-implantation and post-implantation losses in pregnant female mated with males exposed to 10 μg/kg bw of BPA. In the comet assay with epididymal sperm, a significant increase in comet parameters (tail length, tail moment and % tail DNA) was observed in rats treated with 5 mg/kg bw compared with control.

43.    Fawzy et al., 2018: The study was conducted to evaluate the protective action of pumpkin seed oil (PSO) against adverse effects induced by BPA. BPA (Sigma-Aldrich) was administered orally to male Swiss albino mice at 50 mg/kg bw once a day for 28 days. PSO was administered at 1 mL/kg bw either before, with or after treatment of BPA, for 28 days. Seven groups of animals (n = 10) were treated: group 1 (control); group 2 (vehicle); group 3 (PSO); group 4 (BPA); group 5 (PSO before BPA); group 6 (PSO with BPA) and group 7 (PSO after BPA). DNA damage was evaluated by comet assay in liver and testes. Fifty randomly selected nuclei per experimental group were analysed. MN frequencies were evaluated in bone marrow. Two thousand polychromatic erythrocytes (PCE) were scored per animal. A significant (p<0.05) increase of tail DNA % in liver and testes of BPA-treated group with respect to controls (19.93 ± 0.68 vs 13.15 ± 0.22 and 23.56 ± 0.45 vs 15.00 ± 0.50) was observed. A significant increase of MNPCEs (66.40 ± 9.94 vs 10.40 ± 2.96) and a decrease in the ratio of PCE/NCE were also detected. The histopathological examination revealed hepatocyte vacuolar degeneration with many necrotic cells. A defective spermatogenesis was also observed characterized by severe necrosis and loss of the spermatogonial layers with multiple spermatid giant cells formation in most of the seminiferous tubules and a congestion of the interstitial blood vessels. The treatment with PSO reduced the genotoxic effects induced by BPA. PSO before BPA treatment was the best regimen in the alleviation of the adverse effects.

44.    Naik and Vijayalaxmi, 2009: This study evaluated potential genotoxic effects of BPA by induction of chromosomal aberrations and MN in bone marrow cells of Swiss albino mice. To assess for potential interference of BPA with mitotic spindle apparatus, induction of c-mitoses was also performed. BPA (Loba Chemie, Mumbai, India) was administered orally in a 2% acacia gum suspension at dose-levels of 10, 50 and 100 mg/kg bw to groups of three male and three female mice, as single acute dose. Cumulative dose-level experiments were also performed at the lowest (10 mg/kg bw) dose-level for five consecutive days. In single treatment schedule, sampling of bone marrow was performed at 6, 24, 48 and 72 h from beginning of treatment for both micronucleus and chromosome aberration assays. In cumulative treatment schedule, bone marrow was sampled in both assays 24 h after the last administration of BPA. For induction of c-mitoses, the same dose levels used for micronucleus and chromosome aberration assays were applied as single dose and sampling of bone marrow was performed at 2, 6, 12, 24, 48 and 72 h. Results showed that no significant increases of chromosomal aberrations or MN were induced at any dose-level and sampling time used. Conversely, significant increases in the frequencies of gaps were observed in all dose-levels assayed at the 48 and 72 h sampling time and at the two higher dose-levels (50 and 100 mg/kg bw) at the 24 h sampling time. The significant increases of achromatic lesions (gaps) are not considered relevant for clastogenicity. In addition, BPA also induced c-mitotic effects through increases of mitotic indices and decrease in anaphase for both higher dose-levels at 24, 48 and 72 h sampling times.

Comet Assay

In vitro comet assay

45.    Twenty-two in vitro studies using a comet assay in different cell lines were available for evaluation. Twelve were classified as of limited relevance and further considered in the assessment. Most cell lines used in these studies were of human origin from blood, mammary gland and prostate. Rodent cell lines from rat, mouse and hamster and one cell line from monkey were also considered.

46.    Eleven of the 12 studies reported positive results. Three studies on HepG2 cell line yielded both positive (Li XH et al., 2017); Balabanič et al., 2021) and negative (Fic et al., 2013) results. In a non-tumorigenic human prostatic cell line, BPA induced a significant increase in DNA strand breaks paralleled by a decrease in total GSH, antioxidant capacity, glutathione peroxidase 1 (GPx1) and SOD activity and an increase in glutathione reductase (Kose et al., 2020). Positive results were also reported in CHO cells (Xin et al., 2015). Positive results were reported from two studies in which human PBMC were analysed by both alkaline and neutral comet assays (Mokra et al., 2017). Evidence of oxidative damage to DNA bases was provided by the addition of endonuclease III (Nth) and 8-oxoguanine DNA glycosylase (hOGG1) DNA repair enzymes (Mokra et al., 2018). DNA strand breaks induction by BPA was associated with increased ROS, MDA and reduced SOD activity in HepG2 (Li XH et al., 2017). In murine macrophage RAW264.7 cells, positive DNA strand breaks were associated with an increase in ROS and decreased level of antioxidant enzymes (Huang FM et al., 2018). In Marc-145 rhesus monkey embryo renal epithelial cells, DNA strand breaks induction was associated with increased ROS and TBARS and decrease in GSH and SOD activity (Yuan et al., 2019).

47.    DNA strand breaks induction in mouse embryonic fibroblast cell line (NIH3T3) is associated with elevated ROS and a modest increase in DNA 8-hydroxy-2′-deoxyguanosine (8-OHdG) at the highest concentration tested (Chen et al., 2016). In rat INS-1 insulinoma cells DNA strand breaks and ROS level increased in parallel along with the induction of DNA damage-associated proteins (p53 and p-Chk2). At the highest concentration of 100 μM, pre-treatment with NAC reduced the number of induced DNA strand breaks by two-fold (Xin et al., 2014). Finally, ER-positive MCF-7 cells were more sensitive than ER-negative MDA-MB-231 cells to BPA-induced DNA damage, as measured by comet assay (Iso et al., 2006).

48.    The available in vitro studies provided evidence that BPA induces DNA strand breaks most likely related to the induction of oxidative stress.

Summary of studies

49.    Li XH et al., 2017: The study investigated the cytotoxic effects and oxidative stress induced by BPA (Sigma-Aldrich) alone and in combination with dibutyl phthalate (DBP) or cadmium (Cd) in vitro in HepG2 cells. The cell cultures were exposed for a period of 6 h to a range of concentrations of the single substances ensuring a cell viability above 50%. BPA tested from 10-8 to 10-4 mol/L for 6 hours induced a concentration dependent increase of reactive oxygen species (ROS), measured by DCFH-DA, and malondialdehyde (MDA) level and a decreased activity of SOD. An increase of DNA strand breaks (up to eight- fold with respect to the control value) applying the comet assay, was detected after BPA treatment at 10-8, 10-7, 10-6 mol/L for 24 h without a clear concentration response. The co-exposure treatments (BPA and DBP or BPA and Cd) showed higher ROS and MDA levels and lower SOD activity than the mono-exposure treatments. The combined treatments with BPA and Cd had stronger DNA damage effect.

50.    Balabanič et al., 2021: See summary in the in vitro gene mutation section.

51.    Fic et al., 2013: See summary in the in vitro gene mutation section.

52.    Kose et al., 2020: This study investigated the relative toxicity, potential oxidative stress and genotoxicity induced by BPA (>99% purity), BPS and BPF on the RWPE-1 non-tumorigenic prostatic cell line. RWPE-1 cells were incubated with BPA at concentrations of 50–600 μM for 24 h exposure. The IC20 and IC50 values, concentrations that causes 20 and 50% of cell viability loss, after a 24 exposure to BPA were 45 and 113.7 μM. BPA induced significant decreases in the activities of glutathione peroxidase (GPx1) and SOD, an increase in glutathione reductase and total GSH and a decrease in total antioxidant capacity. At a single concentration (IC20), BPA produced significantly higher levels of DNA damage vs the control both in the standard (2.5-fold increase) and Fpg-modified comet assays. No changes in the mRNA levels of p53 and the OGG1, Ape-1, DNA polymerase β base excision repair (BER) proteins were induced by BPA. The single exception was a small decrease in the expression levels of MYH expression.

53.    Xin et al study 2015: See summary in the in vitro gene mutation section.

54.    Mokra et al., 2017: The study reported concentration-related induction of DNA single and double strand breaks (detected with alkaline and neutral comet assay) by BPA (Sigma-Aldrich) and its analogues, BPS, BPF and BPAF in human peripheral blood mononuclear cells (PBMC) treated in the concentrations ranging from 0.01 to 10 μg/mL after 1 and 4 h treatment. No significant decrease of cell viability, evaluated using calcein-AM/PI stains, was observed at the concentrations tested for DNA damage. After 1 h incubation, BPA caused statistically significant increase in DNA strand breaks at 0.1 mg/mL. The highest effects were induced by BPA and BPAF, which produced single strand breaks starting from 0.01 μg/mL, while BPS caused the lowest effect at 10 μg/mL after 4 h of exposure. Statistically significant increases of DNA double strand breaks were induced by BPA at concentrations of 1 μg/mL and 10 μg/mL after 1 h incubation and at 0.1 μg/mL and 1 μg/mL after 4 h incubation. The strongest effect was observed with BPAF. DNA repair was also evaluated at different times (30, 60 and 120 min) after the treatment with BPA at 10 μg/mL. A significant decrease of the DNA damage was observed at 60 min, but the repair was not complete after 120 min.

55.    Mokra et al., 2018: The study reported that BPA (Sigma-Aldrich) and its analogues, BPS, BPF and BPAF caused oxidative DNA damage to purine and pyrimidines in human peripheral blood mononuclear cells (PBMC) treated at concentrations of 0.01, 0.1 and 1 μg/mL for 4 h and 0.001, 0.01 and 0.1 μg/mL for 48 h. BPA was dissolved in ethanol. No significant decrease of cell viability, evaluated using calcein-AM/PI stains, was observed at the concentrations tested. DNA damage was detected with alkaline comet assay coupled with repair enzyme endonuclease III (Nth) and 8-oxoguanine DNA glycosylase (hOGG1). Statistically significant and concentration related oxidative damage to purines (from 0.01 μg/mL) and to pyrimidines (from 0.1 μg/mL) was reported after 4 h treatment. After 48 h treatment significant damage to purine was observed from 0.001 μg/mL and to pyrimidines from 0.01 μg/mL. Statistically significant differences for DNA damage between 4 h and 48 h exposure at the highest concentrations tested (0.01 and 0.1 μg/mL).

56.    Huang FM et al., 2018: See summary in the in vitro chromosomal aberrations/micronuclei section.

57.    Yuan et al., 2019: In this study, markers of oxidative stress and DNA damage were evaluated in Marc-145 rhesus monkey embryo renal epithelial cells exposed to BPA (Sigma-Aldrich, purity > 99%) in the range 10-1, 10-2, 10-3, 10-4, 10-5 and 10-6 M (24 hr exposure). The results showed that BPA induced a concentration-dependent decrease in cell viability (from 20% at the lowest concentration up to almost 80% at the highest concentration), in SOD activity and GSH level. Concomitant concentration-dependent increases in apoptosis, lactate dehydrogenase (LDH) activity, ROS and thiobarbituric acid reactive substances content were observed. BPA also induced a concentration-dependent increase in DNA strand breaks by comet assay in the range of concentrations measured ( 10-3 -to 10-6 M).

58.    Chen et al., 2016: The study investigated the cytotoxic and genotoxic effects induced by BPA alone and in combination with cadmium (Cd) in vitro in mouse embryonic fibroblast cell line (NIH3T3). The treatment of the cell cultures with BPA (Sigma-Aldrich) at 2, 10 and 50 μM was shown to induce, only at the highest concentration tested, a decrease in the cell viability and an increase of the oxidative damage as reactive oxygen species (ROS), measured by DCFH-DA and as 8-OHdG. Significant increase of DNA strand breaks was also detected as tail DNA% and tail moment by comet assay. Higher number of γH2AX foci detected through the use of immunofluorescence and increased γH2AX expression evaluated by western blot in BPA treated cells are indicative of DNA double strand breaks. In addition, 50 μM BPA treatment did significantly decrease the percentage of cells in G1 phase and increased the percentage of cells in G2 phase but not in S phase. Pre-treatment of cells with Cd was observed to aggravate BPA- induced cytotoxicity, and increase ROS production, DNA damage, G2 phase arrest, total TUNEL positive cells and cleaved-PARP expression levels.

59.    Xin et al., 2014: The aim of this study was to assess how BPA can influence the function of pancreatic islets. To measure DNA damage, rat INS-1 insulinoma cells were exposed to different concentrations of BPA (Sigma-Aldrich, 99% purity) (0, 25, 50, 100 μM for 24 h) and analysed by the single-cell gel electrophoresis (comet assay). To investigate the possible mechanism of DNA damage induced by BPA, p53 and p-Chk2 levels were also analysed by western blotting together with measurements of intracellular ROS and glutathione (GSH). The results show that BPA caused an increase in DNA strand-breaks at 50 and 100 μM (as measured by tail moment, tail length and tail DNA %). The authors state that these experimental conditions did not cause any significant toxicity (90% survival; no data provided). Pre-treatment with NAC decreased to half the number of DNA strand breaks induced at the highest dose. A significant increase in intracellular ROS, which was decreased by NAC pre-treatment, was also observed. A significant reduction in the level of GSH levels was observed at all BPA concentrations. Finally, expression of DNA damage-associated proteins (p53 and p-Chk2) was significantly increased by BPA exposure (all concentrations).

60.    Iso et al., 2006: In this study the effects of BPA and 17β-oestradiol (E2) on DNA damage was analysed in ER-positive MCF-7 cells by comet assay. One thousand higher concentrations of BPA (Wako Pure Chemicals Industries, Ltd.) were needed to induce the same levels of effects of E2. Levels of γH2AX foci measured by immunofluorescence microscopy were increased after treatment with E2 or BPA. Foci of γH2AX co-localized with the Bloom helicase, an enzyme involved in the repair of DSBs. In comparison with MCF-7 cells, DNA damage was not as severe in the ER-negative MDA-MB-231 cells. In addition, the ER antagonist ICI182780 blocked E2 and BPA genotoxic effects on MCF-7 cells. These results together suggest that BPA causes genotoxicity ER dependently in the same way as E2.

In vivo comet assay

61.    In the current assessment only 5 of 21 in vivo comet assay studies of DNA strand breaks induction by BPA were classified as of high (one study) or limited relevance and have been considered for evaluation. Among the five oral studies selected, three were positive and two were negative. A single study of high relevance reported negative results in multiple mouse organs (liver, kidney, testes, urinary bladder, colon and lungs) after single treatment at three doses up to the MTD of 500 mg/kg bw (Sharma et al., 2018). Negative results were also reported in rats exposed to 200 mg/kg bw per day orally for 10 days (De Flora et al., 2011). In contrast, dose-related increases in DNA strand breaks were reported at doses greater than 10 μg/kg bw in rats treated for 6 days with a range of doses between 2.4 μg and 50 mg/kg bw per day (Tiwari et al., 2012). A weak and dose-dependent increase in liver DNA strand breaks was observed at 50 and 100 mg/kg bw per day, whereas the increase in kidney was limited to 50 μg/kg bw (Panpatil et al., 2020). Finally, in a study on BPA neurotoxicity, a significant increase of strand breaks in brain cells was observed after treatment in a range of doses from 0.5 to 5000 μg/kg bw per day for 8 weeks (Zhou YX et al., 2017).

62.    Overall, the comet assays provided only limited evidence of DNA damage following multiple administrations of BPA, but not following single dose administrations.

Table 3. Summary table of test results of Comet in vivo studies.

Test system

Dose

Results

Reference

Comet assay in liver, kidney testes, urinary bladder, colon and lungs

CD-1 male mice
5 animals/group

125, 250 and 500 (MTD) mg/kg bw
Single dose by gavage

Negative

Sharma et al., 2018

Comet assay in liver, kidney, testes, urinary bladder, colon and lungs

Spraguely Dawley rats

8 animals/group

200 mg/kg bw per day orally for 10 days

Negative

De Flora et al., 2011

Holtzman rats
10 animals/group

2.4 µg, 10 µg, 5mg and 50 mg/kg per day orally for 6 days

Positive
Dose-related increase starting from 10 µg/kg

Tiwari et al., 2012

Comet assay in liver and kidney

 Male Wistar rats (WNIN) 6 animals/ group

50 100 µg/kg orally for 4 weeks

Positive

Weak dose-related in liver, only at 50 µg/kg in kidney

Panpatil et al., 2020

Comet assay in brain cells

KM male mice

11 animals/group

0.5, 50 and 5000 µg/kg bw per day

Orally for 8 weeks

Positive

Zhou YX et al., 2017

Source:  Re-evaluation of the risks to public health related to the presence of bisphenol A (BPA) in foodstuffs, EFSA, (2021)

Summary of studies

63.    Sharma et al., 2018: The in vivo genotoxic potential of BPA in mouse organs was investigated using the alkaline comet assay. Male CD-1 mice (5 per group) were administered by gavage with BPA (Sigma-Aldrich) suspensions in corn oil prepared by ultrasonication at three dose levels (125, 250 and 500 mg/kg bw), twice 24 h apart. Ethyl methane sulphonate, given once by gavage at 300 mg/kg bw, served as positive control. Animals were sacrificed 3 h after the last treatment and DNA damage investigated by a commercial kit for comet assay in liver, kidney, testes, urinary bladder, colon and lungs cells. For each mouse, 200 cells were analysed (100 per gel) using an automatic comet assay scoring imaging system. Median values for each tissue from each animal were used, and the mean of the median values was evaluated in a statistical analysis. The results of comet assay did not show BPA related effects in any tissue, except for the testes, in which an increased level of DNA strand breaks (p < 0.01 compared with control group) was observed at the lowest dose; however, no dose response relationship was observed as the effects at the medium and highest doses were at the same level as the control group. A modified alkaline comet assay was conducted on human sperm cells treated with BPA 0, 1, 1.5, 2 and 3 μmol/L for 1h. BPA 3 μmol/L reduced cell viability to 60%, therefore it was the highest concentration tested. Ethyl methanesulfonate (EMS) was used as positive control. In total, 600 cells were scored for each concentration. No increase in % tail DNA was observed compared with the negative control.

64.    De Flora et al., 2011: The ability of BPA to form DNA adducts was investigated in two human prostatic cell lines: PNT1a non tumorigenic epithelial cells and PC3 cells androgen-independent prostate cancer cells originated from bone metastasis of prostatic carcinoma. PNT1a and PC3 cells were treated with BPA (Sigma-Aldrich), dissolved in ethanol at a concentration corresponding to the IC50 (200 μM for PNT1a and 250 μM for PC3) for 24 h. PNT1a cells were also treated at a concentration of 1 nM, for 2 months. Significant levels of DNA adducts were detected by 32P-postlabeling technique in prostate cell lines treated with high-concentration of BPA for 24 h (4.2-fold increase over controls) in PNT1a cells and a 2.7-fold increase over controls in PC3 cells) and in a lower extent in PNT1a cells treated at low-concentration for 2 months.

65.    Tiwari et al., 2012: See summary in the in vitro gene mutation section.

66.    Panpatil et al., 2020: See summary in the in vivo chromosomal aberrations/micronuclei section.

67.    Zhou YX et al., 2017: The study investigated the neurotoxicity of low-dose exposure to BPA in a mouse model, examining brain cell damage and the effects of learning and memory ability after 8 weeks exposure to BPA at 0.5, 50 and 5000 μg/kg bw (daily dose, by gavage). The comet assay was used to detect brain cell damage. At the end of treatment 11 mice per group were sacrificed and brain processed for comet assay. Forty cells from each brain were analyzed. Based on tail DNA percentage, the damage level was divided into five grades, from 0 (undamaged) to 4 (maximum damage). The results obtained indicated that with increasing exposure concentrations the fraction of damaged cells (all types) increased significantly from 23.0% in the control group to 47.3%, 66.6% and 72.5% in the low-, medium and high exposed groups, respectively. Also, the severity of DNA damage, expressed as arbitrary units (AUs), increased with AUs of 0.28 in the control to AUs of 0.59, 0.96 and 1.28 in the low-, medium and high-exposed groups, respectively.

Other studies

Induction of γH2AX foci

68.    Several studies have investigated the induction of γH2AX foci (generally regarded as a marker of DNA DSBs) following BPA treatment (Iso et al., 2006; Pfeifer et al., 2015; George and Rupasinghe, 2018; Kim et al., 2018b; Mahemuti et al., 2018; Hercog et al., 2019; Hercog et al., 2020; Nair et al., 2020; Yin et al., 2020; Escarda-Castro et al., 2021; Yuan et al., 2021).

69.     Iso et al. (2006) reported increased levels of γH2AX foci after treatment with 17β-E2 or BPA in ER- positive MCF-7 cells (1000x higher concentrations of BPA were needed to induce the same levels of effects as E2). Induction was less severe in ER-negative MDA-MB-231 cells and the ER antagonist ICI182780 blocked BPA-induced γH2AX focus formation in MCF-7 cells. Taken together, these findings indicate that BPA-induced genotoxicity is ER-dependent.

70.    The effects of low-dose BPA were studied in the ERα-negative MCF10A and in 184A1 normal breast epithelial cell lines and the ERα-positive MCF7 and MDA-MB-231 human breast epithelial adenocarcinomas. Low doses (10 and 100 nM) induced DSBs as measured by γH2AX foci in all cell lines and increased the level of c-Myc and of the cell-cycle regulatory proteins cyclins D1 and E and E2F1. Silencing c-Myc reduced BPA-induced γ-H2AX foci and abolished BPA-mediated mitochondrial ROS production. BPA also induced proliferation in ERα-negative mammary cells. The authors conclude that low-dose BPA exerts a c-Myc–dependent genotoxicity and mitogenicity in ERα-negative mammary cells (Pfeifer et al., 2015).

Summary of studies (in order of mention)

71.    Iso et al., 2006: See summary in the in vitro comet assay section.

72.    Pfeifer et al., 2015: The objective of this study was to investigate the effects of low-dose BPA (Sigma-Aldrich) in mammary gland cells. The human cell lines used in the study are the ERα-negative immortalized benign and normal breast epithelial cell lines (MCF10A and 184A1, respectively) and the ERα-positive MCF7 and MDA-MB-231 cell lines originate from human breast epithelial adenocarcinomas. Low concentrations BPA (10 and 100 nM) induced double strand breaks (DSBs) as measured by γH2AX foci in all cell lines. Both MCF10A and MCF7 cells had also a greater number of ATM-pS1981–positive nuclei after 24 h treatment compared with the control. Low-concentration BPA significantly increased the level of c-Myc protein and other cell-cycle regulatory proteins (cyclin D1, cyclin E and E2F1) and induced proliferation in parallel in ERα-negative 184A1 mammary cells. Silencing c-Myc reduced BPA-mediated increase of γH2AX suggesting that c-Myc plays an essential role in BPA-induced DNA damage. The increased level of DNA double strand breaks induced by BPA exposure in 184A1 cells was also confirmed in a neutral comet assay and was found to be reduced by c-Myc silencing. Similarly, silencing c-Myc abolished BPA-mediated ROS production, which was localized to mitochondria. The authors concluded that low-concentration BPA exerted a c-Myc–dependent genotoxic and mitogenic effects on ERα-negative mammary cells (results reported as tail moment only and a single BPA concentration analysed).

73.    George and Rupasinghe, 2018: This study investigated the relative toxicity of BPA (Sigma-Aldrich) and BPS on human bronchial epithelial cells (BEAS-2B). The tested endpoints included cytotoxicity, induction of ROS, DNA fragmentation, γH2AX foci and DNA tail damage. To evaluate mechanism of cell death the DDR and activation of caspase-3 were also investigated. In all the assays a single concentration and a single time of exposure were used (200 μM BPA for 24 h). According to the authors this concentration caused 50% of cell viability loss (IC50). However, the data reported indicate high levels of toxicity (90%), with all the results being unreliable at this level of toxicity.

74.    Kim et al., 2018: BPA (> 99% purity, Sigma-Aldrich) promoted cell proliferation in undifferentiated and differentiated human hepatocyte cell lines (HepG2 and NKNT-3, respectively) at submicromolar concentrations (0.3-5 μM for 24 h). The proliferative effects of BPA disappeared at concentrations higher than 5 μM (cell viability decreased at concentrations higher than 10 μM). Exposure to BPA in the submicromolar range induced DNA damage in both cell lines as shown by a dose-dependent increase in phosphorylation of histone H2AX (γH2AX), p53 activation and induction of cyclin B1. Increased levels of γH2AX were also observed in liver tissue of juvenile rats (PND 9) orally exposed to a relatively low dose of BPA (0.5 mg/kg for 90 days). At a higher BPA dose (250 mg/kg) no increase in hepatocyte proliferation or cyclin B1 was observed. BPA promoted ROS generation as measured by DCF-DA-enhanced fluorescence in HepG2 cells. Increased levels of ROS were suggested to play a role in BPA-induced proliferation and DNA damage as shown by the partial reversion of both processes upon pre-treatment with NAC.

75.    Mahemuti et al., 2018: The aim of this study was to investigate the key molecular pathways involved in the developmental effects of BPA on human fetal lung and their potential implications in the link between pre-natal exposure to BPA and increased sensitivity to childhood respiratory diseases. Global gene expression profiles and pathway analysis was performed in cultured HFLF exposed to non-cytotoxic concentrations of BPA (0.01, 1 and 100 μM BPA for 24 h, 99% purity, Sigma-Aldrich). Molecular pathways and gene networks were affected by 100, but not 0.01 and 1 μM BPA. These changes were confirmed at both gene and protein levels. The pathways affected by BPA included the cell cycle control of chromosome replication and a decreased DDR. BPA increased DNA DSBs as shown by phosphorylation of H2AX and activated ATM signalling (increased phosphorylation of p53). This resulted in increased cell cycle arrest at G1 phase, senescence and autophagy, and decreased cell proliferation in HFLF. Finally, BPA increased cellular ROS level and activated Nrf2-regulated stress response and xenobiotic detoxification pathways. The authors suggest that pre-natal exposure to BPA may affect fetal lung development and maturation, thereby affecting susceptibility to childhood respiratory diseases.

76.    Hercog et al., 2019: With the aim of comparing the toxicological profiles of possibly safer analogues of BPA, the authors investigated the cytotoxic/genotoxic effects of BPS, BPF and BPAF and their mixtures in human hepatocellular carcinoma HepG2 cells. Single exposure to BPA (99% analytical purity, Sigma-Aldrich) did not induce any significant changes in cell viability at the tested concentrations (2.5, 5, 10, 20 μg/mL for 24 or 72 h). Induction of a significant increase in DNA double strand breaks, as determined by γH2AX assay, was observed only at the highest dose (20 μg/mL for 72 h). BPA (tested at the 10 μg/mL concentration) induced changes in the expression of some genes involved in the xenobiotic metabolism (CYP1A1, UGT1A1, but not GST1), response to oxidative stress (GCLC but not GPX1, GSR, SOD1, CAT), while no changes were observed in any of the genes involved in the DDR (TP53, MDM2, CDKN1A, GADD45A, CHK1, ERCC4). Similar results were obtained when cells were exposed to BPA as a single compound or in mixtures with its analogues at concentrations relevant for human exposure (10 ng/mL). The relevance of these changes is of uncertain biological significance.

77.    Hercog et al., 2020: In a follow-up study by Hercog et al. (2020) the genotoxic effects induced by co-exposure of the cyanotoxin cylindrospermopsin (CYN)(0.5 μg/mL) and BPA (Sigma-Aldrich), BPS and BPF(10 μg/mL, 24 and 72 h exposure) were investigated on HepG2 cells using the same techniques and experimental conditions of Hercog et al. (2019). The results obtained with BPA confirm the previously published observations, but the relevance of these changes remains of uncertain biological significance.

78.    Nair et al., 2020: The effects of BPA (Sigma-Aldrich) as a single agent, or in combination with 4-tert-octylphenol (OP) and hexabromocyclododecane (HBCD), were studied in the HME1 mammary epithelial cells and in the MCF7 breast cancer cell line. Following a 2-month exposure to a low non-toxic BPA concentration (0.0043 nM), increased levels of DNA damage were evidenced by upregulation in both cell lines of phosphorylated DNA damage markers (γ-H2AX, pCHK1, pCHK2, p-P53). Disruption of the cell cycle was observed both after short exposures (24 h and 48 h, G2/M arrest) as well as after the 2-month exposure treatment (G1 and S phase increases). BPA increased cellular invasiveness through collagen. Methylation changes were investigated by Methylation Specific Multiplex-Ligation Dependent Probe Amplification (MS-MLPA) using a panel of 24 tumour suppressor genes (all hypomethylated) and identified hypermethylation of TIMP3, CHFR, ESR1, IGSF4 in MCF7 cells and CDH13 and GSTP1 genes in HME1 cells. Finally, BPA induced phosphorylation of six protein kinases in HME1 cells (EGFR, CREB, STAT6, c-Jun, STAT3, HSP60) and increased levels of several other proteins involved in potential oncogenic pathways (HSP27, AMPKα1, FAK, p53, GSK-3α/β, and P70S6).

79.    Yin et al., 2020: The scope of the study was developing a novel in vitro three-dimensional testicular cell co-culture mouse model that enables the classification of reproductive toxic substances. BPA (99%, Sigma-Aldrich) as well as BPS, TBBPA, and BPAF were used as model compounds. A concentration-dependent increase in BPA toxicity was found in the range 2.5 - 400 μM following 24, 48 and 72 h exposures. The large variations in the number of gH2AX foci observed at 72 h make the relevance of these results questionable. No increase in gH2AX used as marker of DNA damage was found up to a dose of 100 mM (70% cell viability).

80.    Escarda-Castro et al., 2021: The ability of BPA to induce genotoxic and epigenetic changes was investigated before and during cardiomyocyte differentiation in H9c2 rat myoblasts exposed to 10 and 30 μM BPA (92% and 73% of cell viability, respectively). Exposure to BPA (no information on purity or the supplier company) before differentiation repressed the expression of the Hand2 and Gata4 heart transcription factors and three genes belonging to the myosin heavy chain family (Myh1, Myh3, and Myh8), whereas exposure after the 5 days of differentiation reduced the expression of cardiac-specific Tnnt2, Myom2, Sln, and Atp2a1 genes. BPA did not induce ROS and did not increase DNA 8-oxodG levels (as measured by immunostaining) in either myoblasts or cardiomyocytes. After BPA exposure the percentage of DNA repair foci formed by co-localization of the γH2AX and 53BP1 proteins increased in a concentration-dependent manner in myoblasts (from 44% in the control group to 61% and 86% at 10 and 30 μM BPA, respectively), with no increase in MN. Repair foci also increased in cardiomyocytes (from 45% in the control group to 59% and 72% at 10 and 30 μM BPA, respectively). A small increase (up to 13%) in MN was also reported only in cardiomyocytes treated with 10 μM BPA. A decrease in the epigenetic markers H3K9ac and H3K27ac was also reported. The authors concluded from these in vitro data that BPA interferes with the process of cardiomyocyte differentiation. However, the reliability and significance of the data on BPA-induced DNA damage is questioned by several negative factors (high background levels of DNA repair foci, lack of information on methods for micronucleus assays and the small increase of MN over high background).

81.    Yuan et al., 2021: This study investigated the combinatorial toxicity of BPA (≥ 99.8% purity), decabrominated diphenyl ether and acrylamide to HepG2 cells. Increased number of γH2AX foci were induced in HepG2 by a 24h exposure to a single BPA dose that induced 25% toxicity. The majority of the data (ROS measurements, Ca2+ flux, DNA damage, Caspase-3 and decreased mitochondrial membrane potential) refers to additive/synergistic effects induced by varying combinations of contaminants. The authors conclude that BPA induced an increase in γH2AX fluorescence and in the number of γH2AX foci/nucleus. However, this conclusion is not fully supported by the data presented.

Changes in gene expression and DNA methylation

82.    Changes in DNA methylation have been investigated in several studies (De Felice et al, 2015; Porreca et al., 2016; Karmakar et al., 2017; Karaman et al., 2019).

83.    No specific discussion on DNA repair or DDR genes is reported in these publications.

84.    None of the information present in these studies is relevant for the clarification of the genotoxic potential of BPA.

Studies in humans

85.    Overall, human studies are not considered to provide additional relevant information for the evaluation of BPA genotoxicity

[1] A clastogen is a mutagenic agent that disturbs normal DNA related processes or directly causes DNA strand breakages, thus causing the deletion, insertion, or rearrangement of entire chromosome sections. These processes are a form of mutagenesis which if left unrepaired, or improperly repaired, can lead to cancer.

[2] An aneugen is a substance that causes a daughter cell to have an abnormal number of chromosomes or aneuploidy.

Re-evaluation of the risks to public health related to the presence of bisphenol A (BPA) in foodstuffs - Genotoxicity

Mode of action

86.    BPA did not induce gene mutations in bacteria. All the available in vitro studies on chromosomal damage, classified as of high or limited relevance, reported positive results such as increase of CA or MN frequency, in different cellular systems. The increases of BPA-induced chromatid and chromosome breaks observed in some studies (Xin et al., 2015; Santovito et al., 2018; Di Pietro et al., 2020) in association with the induction of DNA strand breaks, detected by comet assay (Xin et al., 2015) are consistent with a clastogenic activity. Moreover, the potential of BPA to affect the spindle integrity and interfere with the chromosome segregation machinery was demonstrated in some reliable studies. Johnson and Parry (2008) reported the formation of aberrant mitotic spindles, with multiple poles, in V79 cells treated with BPA. Altered cytoskeleton organization, with multipolar spindles, failure of microtubule attachment to the kinetochore with the concomitant activation of SAC and chromosome misalignment, were also observed in HeLa cells (Kim et al., 2019). Studies on spindle morphology of mouse (Yang et al., 2020) and bovine (Campen et al., 2018) oocytes during in vitro maturation reported a pattern of alterations similar to that observed in permanent cell lines, namely shorter and multipolar spindles, with altered kinetochore-microtubule attachment and chromosome misalignment at M II.

87.    The conclusion, based on these in vitro studies, is that BPA may act by both clastogenic and aneugenic mechanisms.

88.    The large majority (11 out of 12) of the in vitro studies on comet assay, classified as of limited relevance, reported BPA-induced increases of DNA strand breaks. In some studies, the increase of DNA damage was associated with a parallel increase of ROS and MDA and decrease in antioxidant capacity and in total GSH (Xin et al., 2014; Li XH et al., 2017; Huang FM et al., 2018; Yuan et al., 2019; Kose et al., 2020). A study in macrophages reported also a release of cytochrome c from mitochondria along with increased apoptosis with the indication that the DNA strand breaks could be mainly through the oxidative stress-associated mitochondrial apoptotic pathway (Huang FM et al., 2018). In a study on human PBMC, the application of comet assay with the addition of endonuclease III (Nth) and 8-oxoguanine DNA glycosylase (hOGG1) DNA repair enzymes allowed the detection of oxidative damage to DNA bases (Mokra et al., 2018). Further indication of the role of oxidative damage in induction of DNA strand breaks was provided by the protective effects on DNA damage induced by the pre-treatment with NAC (Xin et al., 2014; Huang FM et al., 2018).

89.    In conclusion, the evidence of DNA strand breaks in vitro is in agreement with the ability of BPA to induce clastogenic damage. In addition, the studies on comet assay provide consistent evidence that BPA induces DNA strand breaks most probably related to the induction of oxidative stress.

90.    The available in vivo studies for BPA-induced chromosomal damage in somatic cells reported mixed results. No increase of CA and MN frequency was reported after a single administration of BPA to mice in a range of doses inducing toxicity at the bone marrow level (Naik and Vijayalaxmi, 2009). In contrast, in another study in mice, increased MN frequency was detected in the presence of high bone marrow toxicity (Fawzy et al., 2018). Positive results were observed in two rat studies (Tiwari et al., 2012; Panpatil et al., 2020) after repeated dose administration, possibly associated with lipid peroxidation and oxidative stress in the first study. No induction of hyperploidy or polyploidy was observed in these studies.

91.    These results indicate that the in vivo induction of chromosomal damage requires specific conditions such as repeated exposure to BPA.

92.    Induction of DNA strand breaks, detected by comet assay in vivo, was observed only after repeated exposure for extensive periods of time up to 8 weeks (Tiwari et al., 2012; Zhou YX et al., 2017; Panpatil et al., 2020). Only one study of high relevance was available on single administration of BPA reporting negative results in multiple mouse organs in a range of doses up to the MTD of 500 mg/kg bw (Sharma et al., 2018). An indication of a possible role of the oxidative stress in inducing DNA strand breaks by BPA was provided by the results of several studies (Abdel-Rahman et al., 2018; Fawzy et al., 2018; Kazmi et al., 2018; Majid et al., 2019; Mohammed et al., 2020) showing the protective effects of natural extracts with antioxidant properties. However, these studies were evaluated as low relevance.

93.    Finally, studies on germ cells, carried out by four laboratories in the framework of a collaborative project on aneugenic chemicals, did not provide any evidence of increased frequency of aneuploidy in mouse oocytes and zygotes and in sperm cells following exposure to low BPA doses (Pacchierotti et al., 2008).

94.    BPA is genotoxic in vitro inducing chromosomal damage and DNA breaks. However, in vivo the evidence of genotoxic properties of BPA is contradictory. This might depend on multiple mechanisms of action described or proposed for BPA. A major difficulty in the interpretation of these contradictory results is the lack of knowledge on the role of BPA metabolism that could be operational in genotoxic activity. Indeed, the role of the proposed DNA adducts has not been clarified. Other uncertainties include the role of ER receptors in the oxidative stress induced by BPA.

Summary of studies

95.    Xin et al., 2015: summary in the in vitro gene mutation section.

96.    Santovito et al., 2018: summary in the in vitro chromosomal aberrations/micronuclei section.

97.    Di Pietro et al., 2020: summary in the in vitro chromosomal aberrations/micronuclei section.

98.    Johnson and Parry (2008): summary in the in vitro chromosomal aberrations/micronuclei section.
99.    Kim et al., 2019: In vitro effects of BPA (Sigma-Aldrich) on mitotic progression were examined in HeLa cells exposed to 100 nM BPA for 5 h. Proteins involved in mitotic processes were detected by western blot, live cell imaging, and immunofluorescence staining. Under the applied treatment conditions, BPA was shown to disturb spindle microtubule attachment to the kinetochore, with the concomitant activation of spindle assembly checkpoint (SAC). Spindle attachment failure was attributed to BPA interference with proper localization of microtubule associated proteins, such as HURP to the proximal ends of spindle microtubules, Kif2a to the minus ends of spindle microtubules, and TPX2 on the mitotic spindle. BPA also caused centriole overduplication, with the formation of multipolar spindle.

100.    Yang et al., 2020: The effect(s) of exposure to BPA (Sigma-Aldrich) on assembled spindle stability in ovulated oocytes were studied. Mature M II oocytes, recovered from the oviducts of superovulated B6D2F1 mice, were cultured for 4 h in the presence of increasing concentrations (5, 25, and 50 μg/mL) of BPA. After treatment oocytes were analyzed by immunofluorescence and live cell imaging to investigate the effect of BPA on spindle dynamics. BPA disrupted spindle organization in a dose-dependent manner, resulting in significantly shorter spindles with unfocused poles and chromosomes congressed in an abnormally elongated metaphase-like configuration, with increased erroneous kinetochore-microtubule interactions.

101.    Campen et al., 2018: The aim of the study was to compare the effects of in vitro exposure to either BPA (Sigma-Aldrich) or BPS on meiotic progression, spindle morphology and chromosome alignment in the bovine oocyte. Bovine ovaries were sourced from an abattoir. Groups of 5–20 cumulus–oocyte complexes (COCs) extracted from the bovine ovaries were treated with BPA or BPS at 10 concentrations between 1 fM and 50 μM and underwent to in vitro maturation for 24 h, then the oocytes were extracted. For BPA experiments, a total of 939 oocytes were analyzed for meiotic stage (including 250 vehicle-only control oocytes), of which a total of 767 were at metaphase II (MII) (including 211 MII oocytes in the control) and were included for analysis of spindle and chromosome configuration. Immunocytochemistry was used to label the chromatin, actin and microtubules in the fixed oocytes. The meiotic stage was assessed using immunofluorescence, and the MII oocytes were further assessed for spindle morphology and chromosome alignment (in all MII oocytes regardless of spindle morphology). No difference in the proportion of bovine oocytes that reached MII was observed for BPA treatment. Significant effect on spindle morphology (p < 0.0001) was induced by BPA treatment at very low concentration (1 fM). Fewer oocytes with bipolar spindles were seen following exposure to BPA at concentrations of 1 fM, 10 fM, 100 fM, 10 pM, 1 nM, 10 nM, 100 nM and 50 μM, compared with the control. There was no effect of BPA on spindle morphology at concentrations of 1 or 100 pM. Increased chromosome misalignments were observed at BPA concentrations of 10 fM, 10 nM and 50 μM of BPA, no effect was detected at any other concentration. The study presents limitations: in the ovaries the effects were evaluated in a specific period of development (namely, the 24 h window of oocyte maturation), without considering potential prior historical exposures in vivo.

102.    Xin et al., 2014 : summary in the in vitro comet assay section.
103.    Li XH et al., 2017: summary in the in vitro comet assay section.
104.    Huang FM et al., 2018: summary in the in vitro chromosomal aberrations/micronuclei section.

105.    Yuan et al., 2019: summary in the in vitro comet assay section.

106.    Kose et al., 2020: summary in the in vitro comet assay section.

107.    Mokra et al., 2018: summary in the in vitro comet assay section.

108.    Naik and Vijayalaxmi, 2009: summary in the in vitro chromosomal aberrations/micronuclei section.

109.    Fawzy et al., 2018: summary in the in vitro chromosomal aberrations/micronuclei section.

110.    Tiwari et al., 2012: Summary in the in vitro gene mutation section.

111.    Panpatil et al., 2020: summary in the in vivo chromosomal aberrations/micronuclei section.

112.    Zhou YX et al., 2017: summary in the in vitro comet assay section.

113.    Sharma et al., 2018: summary in the in vivo comet assay section.

114.    Abdel-Rahman et al., 2018: The study evaluated the protective action of lycopene (LYC), an antioxidant agent, on the toxic effects of BPA (Sigma-Aldrich). Four groups of seven Wistar rats were treated daily for 30 days via gavage: the first group (controls) received corn oil, the second group was given lycopene at a dose of 10 mg/kg bw, the third group was given BPA at 10 mg/kg bw, the fourth group was administrated both BPA and LYC at the 10 mg/kg. Rats were sacrificed immediately after the last administration. Liver was frozen at -80 °C. Single-cell suspensions for comet assay were prepared from frozen livers. No positive controls were used. The comet method applied was not reported. A significant (p < 0.05) increase of tail DNA % in liver of BPA-treated group with respect to controls (25.05 vs 6.68) was observed. Higher activities (p < 0.05) of liver enzymes (serum ALT, alkaline phosphatase (ALP) and GGT and lower levels of total protein and albumin than control rats were detected in serum. Antioxidant enzymes (GPx, SOD and CYPR450 activities) significantly (p < 0.05) decreased while MDA level significantly increased in liver of BPA treated animals. Caspase-3 protein in liver of BPA-treated rats is overexpressed. Histopathological analyses showed deleterious hepatic changes ranging from hepatocytes’ vacuolization and eccentric nuclei to focal necrosis and fibrosis. LYC administration reduced the cytotoxic effects of BPA on hepatic tissue, through improving the liver function biomarkers and oxidant-antioxidant state as well as DNA damage around the control values.

115.    Kazmi et al., 2018: The study evaluated the protective role of Quercus dilatate extracts against BPA (no information on purity) induced hepatotoxicity. Ten groups of SD rats (7 animals/group) were considered, including untreated control group and a group receiving the vehicle. The distilled water-acetone (QDDAE) and methanol-ethyl acetate (QDMEtE) extracts were administered in high (300 mg/kg bw) or low (150 mg/kg bw) doses to SD rats, intraperitoneally injected with BPA (25 mg/kg bw). A group of rats was treated only with BPA. Rats were sacrificed after 4 weeks of treatment and blood and liver were collected. The comet method applied was not sufficiently detailed. An increase of DNA strand breaks in hepatocytes was reported for animals treated with BPA alone. However, the results reported using the different parameters (tail length, % of DNA in tail, tail moment) are not consistent. The % of DNA in tail is 28.35 ± 1.2 in BPA treated animals vs 0.01 ± 0.005 in controls. The value of % of DNA in tail in controls is extremely low with respect to the data reported in the scientific literature. Significant reduction in haemoglobin level, red blood cells and platelet count, whereas elevated levels of white blood cells and erythrocyte sedimentation rate (ESR) were observed in the BPA treated group. Administration of BPA significantly (p < 0.05) decreased the endogenous antioxidant enzyme (CAT,GPx, superoxide dismutase (SOD) and GSH) levels compared with control group. In addition, in the BPA treated group, H2O2, nitrite and TBARS levels in the hepatic tissue were found to be higher when compared with controls. Histopathological examination of BPA treated animals revealed intense hepatic cytoplasm inflammation, centrilobular necrosis, cellular hypertrophy, fatty degeneration, vacuolization, steatosis and distortion of portal vein. A dose dependent hepatoprotective activity was exhibited by both the extracts of Quercus dilatate in different extent for the parameters analysed.

116.    Majid et al., 2019: The study evaluated the protective role of sweet potato (Ipomoea batatas L. Lam.) against BPA-induced testicular toxicity. Sixteen groups of seven Male SD rats were established, including controls, animals treated with the vehicle, with ethyl acetate and methanol extracts from tuber and aerial part of Ipomoea batatas, with BPA (Merck KGaA) and with BPA and different extracts of Ipomea batatas. The BPA group received 50 mg/kg bw dissolved in 10% DMSO, injected intraperitoneal on alternate days for 21 days. The rats were sacrificed 24 h after the last treatment. Comet assay was applied to evaluate the DNA damage. An average 50–100 cells were analysed in each sample for comet parameters (head length, comet length, tail moment, tail length, and amount of DNA in head) of gonadal cell’s nuclei. A statistically significant increase of % DNA in tail (3 folds with respect to the control value) was reported in the group of rats treated with BPA. Endogenous antioxidant enzymes were measured in supernatant from the testicular homogenates: BPA decreased the levels of peroxidases (POD), CAT, SOD. BPA induced also gonadotoxicity measured as size and weight of testes and epididymis, concentration and quality of sperms. The treatment with extracts of Ipomea batatas significantly reduced the gonadotoxicity induced by BPA, the DNA damage and restored the levels of antioxidant enzymes.

117.    Mohammed et al., 2020: The study evaluated the protective role of ginger extract (GE) against BPA-induced toxic effects on thyroid. Four groups of 20 male albino rats were treated orally with BPA (Sigma–Aldrich), GE or both once a day for 35 days as follow: Control group: 0.1 ml/rat of corn oil; BPA group: 200 mg/kg bw per day (1/20 of the oral LD50); GE group: ginger extract 250 mg/kg bw; BPA + GE group: ginger extract followed by BPA after 1 h with the same doses as the other groups. The animals were sacrificed 24 h after the last administration. DNA damage was evaluated by comet assay. A statistically significant increase of DNA damage expressed as tail % DNA, tail length and tail moment were shown in thyroid follicular cells of animals treated with BPA. A concurrent increase of MDA and a decrease of GSH, and SOD were also observed. Adverse effects on the thyroid gland were reported with a significant decrease in serum levels of T3 and T4 accompanied by a significantly increase in serum TSH level. A decrease of Nrf-2 mRNA relative expression and protein concentration and of HO-1 mRNA expression in the BPA-induced thyroid injured rats were also described. The histopathological analysis revealed an alteration of the thyroid gland follicles most of which containing scanty colloid secretion and some others atrophied. The treatment with GE significantly reduced the genotoxic damage and the alteration of thyroid hormones regulating genes.

118.    Pacchierotti et al., 2008: The study evaluated the potential aneugenic effects of BPA on mouse male and female germ cells and bone marrow cells following acute, subacute or subchronic oral exposure. For experiments with acute and subacute exposure, female C57BL/6 mice were treated by gavage with BPA (from Sigma-Aldrich) dissolved in corn oil once with 0.2 and 20 mg/kg bw, or with seven daily administrations of 0.04 mg/kg bw. In subchronic experiments, mice received BPA in drinking water at 0.5 mg/L for 7 weeks. The dose levels tested for subacute effects in bone marrow and male germ cells were 0.002, 0.02 and 0.2 mg/kg bw for 6 days. For the assessment of aneugenicity in female germ cells, M II oocytes were harvested 17 h after induced superovulation, and cytogenetically analyzed after C-banding. The percentages of metaphase I-arrested oocytes, polyploid oocytes and oocytes that had undergone Premature Centromere Separation (PCS) or Premature Anaphase II (PA) were calculated. To evaluate the aneugenic effects of BPA upon the second meiotic division, zygote metaphases were prepared from superovulated females mated with untreated C57Bl/6 males. Zygote metaphases were prepared, C-banded and cytogenetically analyzed for the occurrence of polyploidy and hyperploidy. Experiments on male germ cells were performed with 102/ElxC3H/El)F1 males. Epididymal sperms were collected and hybridized with fluocrochrome-labelled DNA probes for chromosomes 8, X and Y and 10,000 sperm per animal were analyzed to evaluate the incidence of hyperhaploid (X88, Y88, XY8) and diploid (XY88, XX88, YY88) sperm cells. Micronucleus test was performed with four groups of five (102/ElxC3H/El)F1 male mice treated with 0, 0.002, 0.02 or 0.2 mg/kg BPA by gavage on 2 consecutive days and sacrificed 24 h after the second administration. In total, 2000 PCE from two slides were scored per animal for the presence of MN. No significant induction of hyperploidy or polyploidy was observed in oocytes and zygotes at any treatment condition. The only detectable effect was a significant increase of M II oocytes with prematurely separated chromatids after chronic exposure; this effect, however, had no consequence upon the fidelity of chromosome segregation, as demonstrated by the normal chromosome constitution of zygotes under the same exposure condition. Similarly, with male mice no induction of hyperploidy and polyploidy was shown in epididymal sperm after six daily oral BPA doses, and no induction of MN in PCE.

Re-evaluation of the risks to public health related to the presence of bisphenol A (BPA) in foodstuffs - Genotoxicity

Conclusion on hazard identification for genotoxicity effects of BPA

119.    In 2015, the CEF Panel concluded that: The available data support that BPA is not mutagenic (in bacteria or mammalian cells), or clastogenic (MN and CAs). The potential of BPA to produce aneuploidy in vitro was not expressed in vivo. The positive finding in the post labelling assays in vitro and in vivo is unlikely to be of concern, given the lack of mutagenicity and clastogenicity of BPA in vitro and in vivo.

120.    Based on the scientific literature considered in the previous EFSA opinions and published thereafter until 21 July 2021, the CEP Panel concluded that:

•BPA does not induce gene mutations in bacteria;

• BPA induces DNA strand breaks, clastogenic and aneugenic effects in mammalian cells in vitro;

• oxidative stress related mechanism(s) are likely to be involved in the DNA damaging and clastogenic activity elicited by BPA in vitro;

  • there is some evidence for DNA and chromosomal damaging activities of BPA in vivo following repeated administrations, but not following single administrations;
  • the available studies do not provide evidence of aneugenicity of BPA in germ cells in vivo.

121.    In contrast with consistent positive in vitro findings, the in vivo findings in several studies with high/limited reliability were inconsistent. The CEP Panel concluded that the evidence does not support an in vivo genotoxic hazard posed by BPA through direct interaction with DNA.

Re-evaluation of the risks to public health related to the presence of bisphenol A (BPA) in foodstuffs - Genotoxicity

Uncertainty analysis for the genotoxicity assessment

122.    It was concluded that it is Unlikely to Very Unlikely (5 – 30% probability) that BPA presents a genotoxic hazard, the causes of which include a direct mechanism (combining subquestion 1 and 2 (Annex A). Accordingly, it was concluded that it is Likely to Very Likely (70 - 95% probability) that BPA either presents a genotoxic hazard only through indirect mechanism(s) or is not genotoxic. The likelihood terms used in these conclusions are taken from the approximate probability scale, which is recommended by EFSA (EFSA Scientific Committee, 2018) for harmonised use in EFSA assessments.

 123.    EFSA Scientific Committee (2017) has advised that, where the overall evaluation of genotoxicity for a substance leaves no concerns for genotoxicity, HBGVs may be established. However, if concerns for genotoxicity remain, establishing a HBGV is not considered appropriate and a Margin of Exposure (MoE) approach should be followed.

124.    Considering the WoE for probabilities closer to either 70% or 95% that BPA does not present a genotoxic hazard by a direct mechanism, the CEP Panel concluded that probabilities close to 95% are more strongly supported by the evidence than probabilities close to 70% and, therefore, the balance of evidence allows a HBGV to be established.

Re-evaluation of the risks to public health related to the presence of bisphenol A (BPA) in foodstuffs - Genotoxicity

Overall conclusions on genotoxicity

125.    The analysis of the available literature data indicate that BPA does not induce gene mutations in bacteria. BPA induces DNA strand breaks, clastogenic and aneugenic effects in mammalian cells in vitro. Oxidative stress-related mechanism(s) are likely to be involved in this DNA damaging and clastogenic activity.

 126.    In contrast with consistent positive in vitro findings, the in vivo findings in several studies with high/limited reliability were inconsistent. The CEP Panel concluded that the evidence does not support an in vivo genotoxic hazard posed by BPA through direct interaction with DNA.

127.    The CEP Panel concluded that it is unlikely to very unlikely that BPA presents a genotoxic hazard, the causes of which include a direct mechanism, and that the balance of evidence allows a HBGV to be established.

 Questions for the Committee

128.    Members are asked to consider the following questions.

1)    Do Members have any comments on the approach taken by the EFSA panel to assess genotoxicity? Including the weight of evidence and uncertainty analyses?

2)    Do Members have any comments on the overall conclusions reached by EFSA?

Re-evaluation of the risks to public health related to the presence of bisphenol A (BPA) in foodstuffs - Genotoxicity

Genotox-references and abbreviations

References

Abdel-Rahman, H.G., Abdelrazek, H., Zeidan, D.W., Mohamed, R.M. and Abdelazim, A.M., 2018. Lycopene: hepatoprotective and antioxidant effects toward bisphenol A-induced toxicity in female Wistar rats. Oxidative medicine and cellular longevity, 2018.

Balabanič, D., Filipič, M., Klemenčič, A.K. and Žegura, B., 2021. Genotoxic activity of endocrine disrupting compounds commonly present in paper mill effluents. Science of The Total Environment, p.148489.

EFSA Panel on Food Contact Materials, Enzymes, Flavourings and Processing Aids (CEF), 2015. Scientific opinion on the risks to public health related to the presence of bisphenol A (BPA) in foodstuffs. EFSA Journal, 13(1), p.3978.

Campen, K.A., Kucharczyk, K.M., Bogin, B., Ehrlich, J.M. and Combelles, C.M., 2018. Spindle abnormalities and chromosome misalignment in bovine oocytes after exposure to low doses of bisphenol A or bisphenol S. Human Reproduction, 33(5), pp.895-904.

Chen, Z.Y., Liu, C., Lu, Y.H., Yang, L.L., Li, M., He, M.D., Chen, C.H., Zhang, L., Yu, Z.P. and Zhou, Z., 2016. Cadmium exposure enhances bisphenol A-induced genotoxicity through 8-oxoguanine-DNA glycosylase-1 OGG1 inhibition in NIH3T3 fibroblast cells. Cellular Physiology and Biochemistry, 39(3), pp.961-974.

De Felice, B., Manfellotto, F., Palumbo, A., Troisi, J., Zullo, F., Di Carlo, C., Sardo, A.D.S., De Stefano, N., Ferbo, U., Guida, M. and Guida, M., 2015. Genome–wide microRNA expression profiling in placentas from pregnant women exposed to BPA. BMC medical genomics, 8(1), pp.1-13.

De Flora, S., Micale, R.T., La Maestra, S., Izzotti, A., D’Agostini, F., Camoirano, A., Davoli, S.A., Troglio, M.G., Rizzi, F., Davalli, P. and Bettuzzi, S., 2011. Upregulation of clusterin in prostate and DNA damage in spermatozoa from bisphenol A–treated rats and formation of DNA adducts in cultured human prostatic cells. Toxicological sciences, 122(1), pp.45-51.

Di Pietro, P., D’Auria, R., Viggiano, A., Ciaglia, E., Meccariello, R., Russo, R.D., Puca, A.A., Vecchione, C., Nori, S.L. and Santoro, A., 2020. Bisphenol A induces DNA damage in cells exerting immune surveillance functions at peripheral and central level. Chemosphere, 254, p.126819.

ECHA (European Chemicals Agency), 2011. Guidance on information requirements and chemical safety assessment Chapter R. 4: Evaluation of available information.

EFSA Panel on Food Contact Materials, Enzymes, Flavourings and Processing Aids (CEF), 2015. Scientific opinion on the risks to public health related to the presence of bisphenol A (BPA) in foodstuffs. EFSA Journal, 13(1), p.3978.
Scientific Opinion on the risks to public health related to the presence of bisphenol A (BPA) in foodstuffs - - 2015 - EFSA Journal - Wiley Online Library

EFSA Scientific Committee, Hardy A, Benford D, Halldorsson T, Jeger MJ, Knutsen KH, More S, Mortensen A, Naegeli H, Noteborn H, Ockleford C, Ricci A, Rychen G, Silano V, Solecki R, Turck D, Aerts M, Bodin L, Davis A, Edler L, Gundert-Remy U, Sand S, Slob W, Bottex B, Cortiñas Abrahantes J, Court Marques D, Kass G and Schlatter J, 2017a. Update: use of the benchmark dose approach in risk assessment. EFSA Journal 2017;15(1):4658, 41 pp. Update: use of the benchmark dose approach in risk assessment - - 2017 - EFSA Journal - Wiley Online Library

EFSA Scientific Committee, Hardy A, Benford D, Halldorsson T, Jeger M, Knutsen HK, More S, Naegeli H, Noteborn H, Ockleford C, Ricci A, Rychen G, Silano V, Solecki R, Turck D, Younes M, Aquilina G, Crebelli R, Gürtler R, Hirsch-Ernst K, Mosesso P, Nielsen E, van Benthem J, Carfì M, Georgiadis N, Maurici D, Parra Morte J and Schlatter J, 2017. Clarification of some aspects related to genotoxicity assessment. EFSA Journal 2017;15(12):5113, 25 pp. Clarification of some aspects related to genotoxicity assessment | EFSA (europa.eu)

Escarda-Castro, E., Herráez, M.P. and Lombó, M., 2021. Effects of bisphenol A exposure during cardiac cell differentiation. Environmental Pollution, p.117567.

Fawzy, E.I., El Makawy, A.I., El-Bamby, M.M. and Elhamalawy, H.O., 2018. Improved effect of pumpkin seed oil against the bisphenol-A adverse effects in male mice. Toxicology reports, 5, pp.857-863.

Fic, A., Sollner Dolenc, M., Filipič, M. and Peterlin Mašić, L., 2013. Mutagenicity and DNA damage of bisphenol A and its structural analogues in HepG2 cells., Archives of Industrial Hygiene & Toxicology 64(2), pp.189-199.

George, V.C. and Rupasinghe, H.V., 2018. DNA damaging and apoptotic potentials of Bisphenol A and Bisphenol S in human bronchial epithelial cells. Environmental toxicology and pharmacology, 60, pp.52-57.

Hercog, K., Maisanaba, S., Filipič, M., Sollner-Dolenc, M., Kač, L. and Žegura, B., 2019. Genotoxic activity of bisphenol A and its analogues bisphenol S, bisphenol F and bisphenol AF and their mixtures in human hepatocellular carcinoma (HepG2) cells. Science of the total environment, 687, pp.267-276.

Hercog, K., Štern, A., Maisanaba, S., Filipič, M. and Žegura, B., 2020. Plastics in cyanobacterial blooms—genotoxic effects of binary mixtures of cylindrospermopsin and bisphenols in HepG2 cells. Toxins, 12(4), p.219.

Huang, Fu-Mei, Yu-Chao Chang, Shiuan-Shinn Lee, Yung-Chyuan Ho, Ming-Ling Yang, Hui-Wen Lin, and Yu-Hsiang Kuan. "Bisphenol A exhibits cytotoxic or genotoxic potential via oxidative stress-associated mitochondrial apoptotic pathway in murine macrophages." Food and Chemical Toxicology 122 (2018): 215-224.

Iso, T., Watanabe, T., Iwamoto, T., Shimamoto, A. and Furuichi, Y., 2006. DNA damage caused by bisphenol A and estradiol through estrogenic activity. Biological and Pharmaceutical Bulletin, 29(2), pp.206-210.

Johnson, G.E. and Parry, E.M., 2008. Mechanistic investigations of low dose exposures to the genotoxic compounds bisphenol-A and rotenone. Mutation Research/Genetic Toxicology and Environmental Mutagenesis, 651(1-2), pp.56-63.

Karaman FE, Caglayan M, Sancar-Bas S, Ozal-Coskun C, Arda-Pirincci P and Ozden S, 2019. Global and region-specific post-transcriptional and post-translational modifications of bisphenol A in human prostate cancer cells. Environmental Pollution, 255

Karmakar, P.C., Kang, H.G., Kim, Y.H., Jung, S.E., Rahman, M.S., Lee, H.S., Kim, Y.H., Pang, M.G. and Ryu, B.Y., 2017. Bisphenol A affects on the functional properties and proteome of testicular germ cells and spermatogonial stem cells in vitro culture model. Scientific reports, 7(1), pp.1-14.

Kazmi, S.T.B., Majid, M., Maryam, S., Rahat, A., Ahmed, M., Khan, M.R. and ul Haq, I., 2018. Quercus dilatata Lindl. ex Royle ameliorates BPA induced hepatotoxicity in Sprague Dawley rats. Biomedicine & Pharmacotherapy, 102, pp.728-738.

Klimisch, H.J., Andreae, M. and Tillmann, U., 1997. A Systematic Approach for Evaluating the Quality of Experimental Toxicological and Ecotoxicological Data Regulatory Toxicology and Pharmacology.

Kim, S., Choi, E., Kim, M., Jeong, J.S., Kang, K.W., Jee, S., Lim, K.M. and Lee, Y.S., 2018. Submicromolar bisphenol A induces proliferation and DNA damage in human hepatocyte cell lines in vitro and in juvenile rats in vivo. Food and Chemical Toxicology, 111, pp.125-132.

Kim, S., Gwon, D., Kim, J.A., Choi, H. and Jang, C.Y., 2019. Bisphenol A disrupts mitotic progression via disturbing spindle attachment to kinetochore and centriole duplication in cancer cell lines. Toxicology in Vitro, 59, pp.115-125.

Kose, O., Rachidi, W., Beal, D., Erkekoglu, P., FayyadKazan, H. and Kocer Gumusel, B., 2020. The effects of different bisphenol derivatives on oxidative stress, DNA damage and DNA repair in RWPE1 cells: A comparative study. Journal of Applied Toxicology, 40(5), pp.643-654.

Li, X., Yin, P. and Zhao, L., 2017. Effects of individual and combined toxicity of bisphenol A, dibutyl phthalate and cadmium on oxidative stress and genotoxicity in HepG 2 cells. Food and Chemical Toxicology, 105, pp.73-81.

Mahemuti, L., Chen, Q., Coughlan, M.C., Qiao, C., Chepelev, N.L., Florian, M., Dong, D., Woodworth, R.G., Yan, J., Cao, X.L. and Scoggan, K.A., 2018. Bisphenol A induces DSB-ATM-p53 signaling leading to cell cycle arrest, senescence, autophagy, stress response, and estrogen release in human fetal lung fibroblasts. Archives of toxicology, 92(4), pp.1453-1469.

Majid, M., Ijaz, F., Baig, M.W., Nasir, B., Khan, M.R. and Haq, I.U., 2019. Scientific validation of ethnomedicinal use of Ipomoea batatas L. Lam. as aphrodisiac and gonadoprotective agent against bisphenol A induced testicular toxicity in male Sprague Dawley rats. BioMed research international, 2019.

Masuda, S., Terashima, Y., Sano, A., Kuruto, R., Sugiyama, Y., Shimoi, K., Tanji, K., Yoshioka, H., Terao, Y. and Kinae, N., 2005. Changes in the mutagenic and estrogenic activities of bisphenol A upon treatment with nitrite. Mutation Research/Genetic Toxicology and Environmental Mutagenesis, 585(1-2), pp.137-146.

Mohammed, E.T., Hashem, K.S., Ahmed, A.E., Aly, M.T., Aleya, L. and Abdel-Daim, M.M., 2020. Ginger extract ameliorates bisphenol A (BPA)-induced disruption in thyroid hormones synthesis and metabolism: involvement of Nrf-2/HO-1 pathway. Science of the Total Environment, 703, p.134664.

Mokra, K., Kuźmińska-Surowaniec, A., Woźniak, K. and Michałowicz, J., 2017. Evaluation of DNA-damaging potential of bisphenol A and its selected analogs in human peripheral blood mononuclear cells (in vitro study). Food and chemical toxicology, 100, pp.62-69.

Mokra, K., Woźniak, K., Bukowska, B., Sicińska, P. and Michałowicz, J., 2018. Low-concentration exposure to BPA, BPF and BPAF induces oxidative DNA bases lesions in human peripheral blood mononuclear cells. Chemosphere, 201, pp.119-126.

Naik, P. and Vijayalaxmi, K.K., 2009. Cytogenetic evaluation for genotoxicity of bisphenol-A in bone marrow cells of Swiss albino mice. Mutation Research/Genetic Toxicology and Environmental Mutagenesis, 676(1-2), pp.106-112.

Nair, V.A., Valo, S., Peltomäki, P., Bajbouj, K. and Abdel-Rahman, W.M., 2020. Oncogenic potential of Bisphenol A and common environmental contaminants in human mammary epithelial cells. International journal of molecular sciences, 21(10), p.3735.

OECD (Organisation for Economic Co-operation and Development), 2005. Manual for the investigation of 14112 HPV chemicals. Chapter 3.1 Guidance for Determining the Quality of Data for the SIDS Dossier 14113 (Reliability, Relevance and Adequacy) (Last updated: December 2005). Available online: Microsoft Word - section_31_Jan06.doc (oecd.org)

Pacchierotti, F., Ranaldi, R., Eichenlaub-Ritter, U., Attia, S. and Adler, I.D., 2008. Evaluation of aneugenic effects of bisphenol A in somatic and germ cells of the mouse. Mutation Research/Genetic Toxicology and Environmental Mutagenesis, 651(1-2), pp.64-70.

Panpatil, V.V., Kumari, D., Chatterjee, A., Kumar, S., Bhaskar, V., Polasa, K. and Ghosh, S., 2020. Protective Effect of Turmeric against Bisphenol-A Induced Genotoxicity in Rats. Journal of nutritional science and vitaminology, 66(Supplement), pp.S336-S342.

Pfeifer, D., Chung, Y.M. and Hu, M.C., 2015. Effects of low-dose bisphenol A on DNA damage and proliferation of breast cells: the role of c-Myc. Environmental health perspectives, 123(12), pp.1271-1279.

Porreca, I., Ulloa Severino, L., D’Angelo, F., Cuomo, D., Ceccarelli, M., Altucci, L., Amendola, E., Nebbioso, A., Mallardo, M., De Felice, M. and Ambrosino, C., 2016. “Stockpile” of slight transcriptomic changes determines the indirect genotoxicity of low-dose BPA in thyroid cells. PloS one, 11(3), p.e0151618.

Santovito, A., Cannarsa, E., Schleicherova, D. and Cervella, P., 2018. Clastogenic effects of bisphenol A on human cultured lymphocytes. Human & experimental toxicology, 37(1), pp.69-77.

Sharma, A.K., Boberg, J. and Dybdahl, M., 2018. DNA damage in mouse organs and in human sperm cells by bisphenol A. Toxicological & Environmental Chemistry, 100(4), pp.465-478.

Šutiaková, I., Kovalkovičová, N. and Šutiak, V., 2014. Micronucleus assay in bovine lymphocytes after exposure to bisphenol A in vitro. In Vitro Cellular & Developmental Biology-Animal, 50(6), pp.502-506.

Tiwari, D., Kamble, J., Chilgunde, S., Patil, P., Maru, G., Kawle, D., Bhartiya, U., Joseph, L. and Vanage, G., 2012. Clastogenic and mutagenic effects of bisphenol A: an endocrine disruptor. Mutation Research/Genetic Toxicology and Environmental Mutagenesis, 743(1-2), pp.83-90.

Tiwari, D. and Vanage, G., 2013. Mutagenic effect of Bisphenol A on adult rat male germ cells and their fertility. Reproductive Toxicology, 40, pp.60-68.

Xin, L., Lin, Y., Wang, A., Zhu, W., Liang, Y., Su, X., Hong, C., Wan, J., Wang, Y. and Tian, H., 2015. Cytogenetic evaluation for the genotoxicity of bisphenol-A in Chinese hamster ovary cells. Environmental toxicology and pharmacology, 40(2), pp.524-529.

Xin, F., Jiang, L., Liu, X., Geng, C., Wang, W., Zhong, L., Yang, G. and Chen, M., 2014. Bisphenol A induces oxidative stress-associated DNA damage in INS-1 cells. Mutation Research/Genetic Toxicology and Environmental Mutagenesis, 769, pp.29-33.

Yang L, Baumann C, De La Fuente R and Viveiros MM, 2020. Mechanisms underlying disruption of oocyte 14988 spindle stability by bisphenol compounds. Reproduction, 159(4), 383—396.

Yin, L., Siracusa, J.S., Measel, E., Guan, X., Edenfield, C., Liang, S. and Yu, X., 2020. High-content image-based single-cell phenotypic analysis for the testicular toxicity prediction induced by bisphenol A and its analogs bisphenol S, Bisphenol AF, and tetrabromobisphenol a in a three-dimensional testicular cell co-culture model. Toxicological Sciences, 173(2), pp.313-335.

Yu, H., Chen, Z., Hu, K., Yang, Z., Song, M., Li, Z. and Liu, Y., 2020. Potent Clastogenicity of Bisphenol Compounds in Mammalian Cells—Human CYP1A1 Being a Major Activating Enzyme. Environmental Science & Technology, 54(23), pp.15267-15276.

Yuan, J., Kong, Y., Ommati, M.M., Tang, Z., Li, H., Li, L., Zhao, C., Shi, Z. and Wang, J., 2019. Bisphenol A-induced apoptosis, oxidative stress and DNA damage in cultured rhesus monkey embryo renal epithelial Marc-145 cells. Chemosphere, 234, pp.682-689.

Yuan, J., Che, S., Zhang, L., Li, X., Yang, J., Sun, X. and Ruan, Z., 2021. Assessing the combinatorial cytotoxicity of the exogenous contamination with BDE-209, bisphenol A, and acrylamide via high-content analysis. Chemosphere, 284, p.131346.

Zhou, Y., Wang, Z., Xia, M., Zhuang, S., Gong, X., Pan, J., Li, C., Fan, R., Pang, Q. and Lu, S., 2017. Neurotoxicity of low bisphenol A (BPA) exposure for young male mice: Implications for children exposed to environmental levels of BPA. Environmental pollution, 229, pp.40-48.

Zemheri F and Uguz C, 2016. Determining mutagenic effect of nonylphenol and bisphenol A by using Ames/Salmonella/microsome test. Journal of Applied Biological Sciences, 10(3), pp.09-12.

Abbreviations

BP-1

Sulphonylbis(benzene-4,1-diyloxy)]diethanol

BP-2

4,4’-Sulphanediyldiphenol

BPA

Bisphenol A

BPAF

Bisphenol AF

BW

birth weight

CA

chromosomal aberrations

Cd

cadmium

CBMA

cytokinesis blocked micronucleus assay

CHO cells

chinese hamster ovary cells

DCFH-DA

Dichlorofluorescein Diacetate Assay

DBP

dibutyl phthalate

E2

Oestradiol

ECHA

European Chemicals Agency

HBCD

hexabromocyclododecane

HOC

health outcome category

LYC

lycopene

MN

micronuclei

MTOCs

microtubule organizing centres

MoA

mode of action

NAC

N-acetylcysteine

NDI

nuclear division index

OTM

olive tail moment

OP

4-tert-octylphenol

8-OHdG

8-hydroxydeoxyguanosine

SAC

spindle assembly checkpoint

SD

Sprague Dawley

PBMC

peripheral blood mononuclear cells

PSO

pumpkin seed oil

ROS

reactive oxygen species

WG

working group

WoE

weight of evidence

Re-evaluation of the risks to public health related to the presence of bisphenol A (BPA) in foodstuffs - Genotoxicity

Annex A evaluation of reliability of results of genotoxicity studies – general considerations

1.    Reliability is defined as “evaluating the inherent quality of a test report or publication relating to preferably standardized methodology and the way that the experimental procedure and results are described to give evidence of the clarity and plausibility of the findings” (Klimisch et al., 1997).

2.    In assigning the reliability score, the compliance with the Organization for European Economic Cooperation and Development (OECD) Test Guidelines (TGs) or standardized methodology and the completeness of the reporting as detailed below were considered.

3.    The reliability scores were:

1) reliable without restriction : This includes studies or data from the literature or reports which were carried out or generated according to generally valid and/or internationally accepted testing guidelines (preferably performed according to Good Laboratory Practice (GLP)) or in which the test parameters documented are based on a specific (national) testing guideline (preferably performed according to GLP) or in which all parameters described are closely related/comparable to a guideline method.

2) reliable with restrictions: This includes studies or data from the literature or reports (mostly not performed according to GLP), in which the test parameters documented do not totally comply with the specific testing guideline, but are sufficient to accept the data or in which investigations are described which cannot be subsumed under a testing guideline, but which are nevertheless well documented and scientifically acceptable.

3) insufficient reliability: testing guideline, but are sufficient to accept the data or in which investigations are described which cannot be subsumed under a testing guideline, but which are nevertheless well documented and scientifically acceptable.

4) reliability cannot be evaluated: This includes studies or data from the literature, that do not give sufficient experimental details and that are only listed in short abstracts or secondary literature (books, reviews, etc.).

5) reliability not evaluated, since the study is not relevant and/or not required for the risk assessment (in case the study is reported for reasons of transparency only): The study is not relevant and/or not useful for the risk assessment.

Re-evaluation of the risks to public health related to the presence of bisphenol A (BPA) in foodstuffs - Genotoxicity

WoE approach

7.    The WoE approach applied to the evaluation of genotoxicity data is based on EFSA Scientific Committee recommendations (EFSA Scientific Committee, 2011, 2017). As recommended by the EFSA Scientific Committee (EFSA Scientific Committee, 2011, 2017), a documented WoE approach for the evaluation and interpretation of genotoxicity data’ has been applied, taking into account not only the quality and availability of the data on genotoxicity itself, but also all other relevant data that may be available. The main steps of the WoE approach applied in the genotoxicity assessment of BPA are described below.

Assembling of the evidence into lines of evidence of similar type

8.    In a first step, the CEP Panel evaluated all available in vitro and in vivo studies addressing the three main endpoints of genotoxicity: gene mutations, structural and numerical chromosomal aberrations (CA) in addition to DNA damage endpoint (evaluated by Comet assay). The study results addressing each of these endpoints were grouped into lines of evidence. Only the studies of high and limited relevance were included.

9.    Studies investigating the BPA MoA were considered, e.g. DNA oxidation, ROS (when genotoxicity was also investigated in the same study), DNA binding, interference with proteins involved in chromosome segregation during cell division, modulation of expression of genes involved in DNA repair or in chromosome segregation and markers of DNA double strand breaks (DSBs) (e.g. γH2AX). Evidence from the mechanistic studies may support the lines of evidence for the genotoxicity endpoints

Weighting of the evidence

10.    A quantitative method to weight the evidence was not considered appropriate due to the quantity and heterogeneity of the evidence to be integrated. A qualitative method based on expert judgment was applied. All studies evaluated for reliability and relevance (as described above) were listed in tables below). The evaluation of the studies of high and limited relevance was described in the opinion, including the conclusion for each line of evidence. The consistency of the evidence was assessed and presented in the opinion.

Integrating all the evidence

11.   Integrating evidence from the MoA with lines of evidence from genotoxicity endpoints allows a reduction in the uncertainty on the potential genotoxicity. In case genotoxic effects were observed, evidence from the MoA may allow clarification if the genotoxicity is due to a direct or indirect mechanism.

Re-evaluation of the risks to public health related to the presence of bisphenol A (BPA) in foodstuffs - Genotoxicity

Evaluation of relevance of results of genotoxicity studies -general considerations

 4.    The relevance of the study (high, limited or low) is based both on its reliability and on the relevance of the test results.

5.    The relevance of the test results was mainly, but not exclusively, based on:

  • Genetic endpoint (high relevance for gene mutations, structural and numerical chromosomal alterations as well as results obtained in an in vivo comet assay, which belongs to the assays recommended by the EFSA Scientific Committee (2011) for the follow-up of a positive in vitro result; lower relevance for other genotoxic effects). Other test systems although potentially considered of limited or low relevance may provide useful supporting information.
  • Route of administration (e.g. oral vs. intravenous, intraperitoneal injection, subcutaneous injection, inhalation exposure) in case of in vivo studies.
  • Status of validation (e.g. for which an OECD TG exists or is in the course of development, internationally recommended protocol, validation at national level only, no validation)
  • Reliability and relevance of the test system/test design irrespectively of whether a study has been conducted in compliance with GLP or not.
  • Information on BPA purity grade and/or the supplier. If only the supplier was available, the company’s website was consulted to retrieve the purity grade, or the authors were contacted to ask for it. If none of the two information were reported or obtained, the relevance was considered low and the study was excluded from the WoE assessment.

6.    Studies for which the relevance of the result was judged to be low were not considered further.

Re-evaluation of the risks to public health related to the presence of bisphenol A (BPA) in foodstuffs - Genotoxicity

Uncertainty analysis for genotoxicity including results

12.    The purpose of the uncertainty analysis for genotoxicity was to assess the degree of certainty for the conclusion on whether BPA presents a genotoxic hazard by a direct mechanism (direct interaction with DNA), taking into account the available evidence and also the associated uncertainties. This overall question was divided into two sub-questions, which were assessed by three WG members with specialist expertise in genotoxicity assessment:

Sub-question 1: What is your probability (%) that there is a genotoxic hazard in humans from BPA?

Sub-question 2: If there would be a genotoxic hazard in humans from BPA, what is your probability that its causes include a direct mechanism?

13.    When assessing the two sub-questions, the experts considered all the data they had reviewed for the genotoxicity assessment, including results from in vitro studies and animal models, taking into account their relevance to humans; the available data from human studies were considered not relevant.
14.    The experts’ judgements were elicited by the structured procedure described below:

15.    The word ‘include’ in sub-question 2 was introduced to accommodate the possibility that both direct and indirect mechanisms could operate together.

16.     The experts were provided with guidance on how to assess and express their probability judgements for the two questions. They were asked to consider all the data they had reviewed for the genotoxicity assessment, including results from in vitro studies and animal models, taking into account their relevance to humans; the available human data were considered not relevant.

17.    The three experts first worked on the questions independently, based on the evidence they had already reviewed and evaluated for the opinion, and recorded their probabilities and the reasoning for their judgements in an excel template similar to that which was used for Question 1 in the uncertainty analysis for non-genotoxic endpoints. This was followed by a facilitated meeting, where the three experts presented their judgements and reasoning and discussed  them together with the WG Chair. After the meeting, the three experts were invited to review and, if they wished, revise their judgements and reasoning in the light of the discussion.

18.    Each expert’s revised probabilities for the two sub-questions were multiplied to provide a probability for the overall question. This is appropriate because the second question is conditional on the first. The first sub-question provides a probability for BPA presenting a genotoxic hazard; the second question provides a conditional probability that, if BPA presents a genotoxic hazard, there is a direct mechanism. So the product of these is a probability that both are true: that BPA does present a genotoxic hazard and that there is a direct mechanism. As the experts’ probabilities were approximate (ranges), the calculation is done by interval arithmetic and the resulting probabilities are also approximate.

19.    The three experts presented and discussed their revised judgements and reasoning in a facilitated meeting with the full WG. The WG discussed the results of the calculations combining the experts’ probabilities for the two questions and expressed the conclusion of the WG both as a probability range and using verbal likelihood terms from the approximate probability scale, which is recommended by EFSA (EFSA Scientific Committee, 2018) for harmonised use in EFSA assessments. Finally, the WG discussed the implications of their conclusion for whether a TDI could be set for BPA or whether a Margin of Exposure approach was required.

20.    Table 1 shows the revised judgements provided by the three experts together after sharing and discussing their initial judgements and reasoning. The third row of Table 1 shows their probabilities for the overall question, which were obtained by multiplying each expert’s probabilities for the two sub- questions. These are their probabilities that BPA does present a genotoxic hazard and that there is a direct mechanism. The bottom row of Table 1 shows the complement of the probabilities in the third row, obtained by subtracting each probability from 100%.  These are the experts’ probabilities for the opposite outcome: that BPA does not present a genotoxic hazard by a direct mechanism. The fifth column of Table 1 shows the ‘envelope’ of the probabilities for the three experts, obtained by taking the lowest and highest probabilities in each row. These express the range of opinion across the three experts.

Table 1 Results of the uncertainty analysis for the genotoxicity assessment.

 

Expert A

Expert B

Expert C

Envelope of three experts

Assessment (rounded values)*

Experts’ probabilities that BPA presents a genotoxic hazard in humans (sub-questions 1)

70-90%

66-90%

70-90%

66-90%

66-90%

 

Experts’ probabilities that, if BPA is genotoxic, there is a direct mechanism (sub-question 2)

10-33%

10-33%

20-30%

10-33%

10-33%

 

Calculated probabilities that BPA is genotoxic by a direct mechanism ((sub-question 1) x (sub-question 2)

7-29.7%

6.6-29.7%

14.27%

6.6-29.7%

5-30%

 

Calculated probabilities that BPA is not genotoxic by a direct mechanism (100% minus row above)

70.3%-93%

70.3%-93.4%

73-86%

70.3-93.4

70-95%

 

                 

*The calculated probabilities were rounded to the nearest 5%. The experts probabilities of 33% and 66% were not changed because they correspond approximately to a 1 in 3 chance and a 2 in 3 chance, respectively.

Source:  Re-evaluation of the risks to public health related to the presence of bisphenol A (BPA) in foodstuffs, EFSA, (2021).

21.    The results in Table 1 and the reasoning of the three experts were presented and discussed in detail at a facilitated meeting with the full WG. It was agreed to take the envelope of the 3 experts’ results as the consensus of the WG, taking account of the available evidence and associated uncertainties. The WG also agreed that their consensus probability that BPA is genotoxic by a direct mechanism should be rounded to 5 – 30%, as shown in the right-hand column of Table 1, to take account that it is based on expert judgement and avoid the implied precision of the calculated values. Similarly, the WG rounded their consensus probability that BPA is not genotoxic by a direct mechanism to 70 – 95%.

22.    The width of the consensus probability range for BPA not being genotoxic by a direct mechanism, reflects the uncertainty of the three experts and the other WG members about the judgements on sub- questions 1 and 2. The WG discussed in more detail which lines of evidence tended to support probabilities in the lower end of this range, and which tended to support the upper end of the range  (Table 2).

Table 2. Summary of lines of evidence supporting either lower or higher probabilities that BPA does not present a genotoxic hazard by a direct mechanism, within the range assessed by the WG  (70-95%).

Evidence supporting probabilities closer to 95 %

  • Consistent negative Ames tests
  • Indications of carcinogenic effects of BPA do not indicate direct genotoxic mechanism because only at very low doses and not higher doses (non monotonic), only after development exposure (up to weaning) and only in one target tissue
  • Reactive non-conjugated metabolites of BPA are observed in animals but not in humans
  • Effects only from repeated exposure, so might be secondly
  • Evidence for several indirect mechanisms

 

Evidence supporting probabilities closer to 70%

  • Presence of uncharacterised DNA adducts
  • Mutational spectrum from whole genome assessment

 

Source:  Re-evaluation of the risks to public health related to the presence of bisphenol A (BPA) in foodstuffs, EFSA, (2021).

23.    It was concluded that it is Unlikely to Very Unlikely (5 – 30% probability) that BPA presents a genotoxic hazard, the causes of which include a direct mechanism (combining subquestion 1 and 2, see third row of Table 1). Accordingly, it was concluded that it is Likely to Very Likely (70 - 95% probability) that BPA either presents a genotoxic hazard only through indirect mechanism(s) or is not genotoxic. The likelihood terms used in these conclusions are taken from the approximate probability scale, which is recommended by EFSA (Table 2 in EFSA Scientific Committee, 2018) for harmonised use in EFSA assessments.

24.    EFSA Scientific Committee (2017) has advised that, where the overall evaluation of genotoxicity for a substance leaves no concerns for genotoxicity, HBGVs may be established. However, if concerns for genotoxicity remain, establishing a HBGV is not considered appropriate and a Margin of Exposure (MoE) approach should be followed.

25.    Considering the WoE for probabilities closer to either 70% or 95% that BPA does not present a genotoxic hazard by a direct mechanism (Table 2), the CEP Panel concluded that probabilities close to 95% are more strongly supported by the evidence than probabilities close to 70% and, therefore, the balance of evidence allows a HBGV to be established.

Re-evaluation of the risks to public health related to the presence of bisphenol A (BPA) in foodstuffs - Genotoxicity

Weight of evidence studies

26.    The following are tables summarising new in vitro and in vivo genotoxicity studies on BPA identified in the literature (2013 –2021) and studies considered in the ‘Scientific Opinion on the risks to public health related to the presence of bisphenol A (BPA) in foodstuffs’ (EFSA CEF Panel, 2015). Key : *Indicates that more than one assay is reported/indicates when papers belong to more than one table. **Indicates that both in vitro and in vivo assays are reported in the same paper.

27.    The studies have been evaluated based on the criteria described above in Annex A.

Bacterial reverse mutation assay

Table 1. Bacterial reverse mutation assay (OECD TG 471 was considered for the evaluation of reliability.

Test system/Test

object

Exposure

conditions

(concentration/

duration/metabolic

activation)

Information on the

characteristics of

the test substance

Results

Reliability/

Comments

Relevance of the

result

Reference

Bacterial reverse

mutation assay

Salmonella

Typhimurium strains

TA 98 and TA 100
 

In vivo micronucleus

assay (Table 7)**

BPA 1–10

μmoles/plate with or

without S9; 3

replicates

BPA (Tokyo Kasei

Kogyo Co., Ltd)

Purity 99% not

reported in the study

but available in the

website of the

company

Negative

Reliability: 2

Only 2 strains

Data on negative

controls subtracted

(but not shown)

No positive control

Limited

Masuda et al.,

20051**

Bacterial reverse

mutation assay

Salmonella

Typhimurium strains

TA98, TA100, TA102
 

In vivo chromosomal

aberration (Table 6)
micronucleus assay

(Table 7)

comet assay (Table

8)**

BPA 0, 6.25, 12.5, 25,

50, 100, 150 and 200

μg/plate for 48 h; with

or without S9; preincubation

method

BPA, purity 99%

(Sigma Chemical

Company)

Negative

Reliability: 2

Only 3 strains used

Limited

Tiwari et al., 20121**

Bacterial reverse

mutation assay

Salmonella

Typhimurium strains

TA98 and TA 100

In vitro comet assay

(Table 5)*

BPA 0, 4, 20, 100, 500

μg/plate for 48 h

(TA100) and 72 h

(TA98); 3 replicates;

with or without S9

BPA, purity >99%

(Sigma-Aldrich)

Negative

Reliability: 2

Only 2 strains

Limited

Fic et al., 20131*

Bacterial reverse

mutation assay

Salmonella

Typhimurium strains

TA1535, TA97, TA98,

TA100 and TA102

In vitro chromosomal

aberration (Table 3)

micronucleus assay

(Table 4)

comet assay (Table 5)

in CHO cells*

BPA 10–5000

μg/plate; 48 h

incubation; with or

without S9; preincubation

method in

triplicates; 3

independent

experiments

BPA (purity 99%)2,

was purchased from

Tianjin Guangfu Fine

Chemical Research

Institute (Tianjin,

China)

Negative

Reliability: 1

High

Xin et al., 2015*

Bacterial reverse

mutation assay

Salmonella

Typhimurium strains

TA98 and TA100

BPA 0.1, 1, 10 and

100 μg/plate with or

without S9; plate

incorporation assay in

triplicates; 2

independent

experiments

BPA (Merck)

Purity >97% not

reported in the study

but available on the

website of the

company

Negative

Reliability: 2

Only 2 bacterial strains

used

Limited

Zemheri and Uguz,

2016

SOS/umuC assay in

Salmonella

Typhimurium TA1535

pSK1002

In vitro comet assay

(Table 5)*

BPA 0, 1, 10, 100,

1000 μg/L, without or

with metabolic

activation (S9)

BPA (Sigma-Aldrich)

Purity >97% not

reported in the study

but available on the

website of the

company

Negative

Reliability: 2

Non-standard test

applied as a

preliminary analysis of

toxicity and

mutagenicity

Limited

Balabanič et al., 2021*

1Studies considered in the Scientific Opinion on the Risks to Public Health Related to the Presence of Bisphenol A (BPA) in 2Foodstuffs (EFSA CEF Panel, 2015) Information on BPA purity provided by the study authors on 11 October 2021, upon EFSA request
Source:  Re-evaluation of the risks to public health related to the presence of bisphenol A (BPA) in foodstuffs, EFSA, (2021).

In vitro gene mutation in mammalian cells

Table 2: In vitro gene mutation in mammalian cells.

Test system/Test

object

Exposure

conditions

(concentration/

duration/metabolic

activation)

Information on the

characteristics of

the test substance

Results

Reliability/

Comments

Relevance of the

result

Reference

Analysis of mutational

spectra in immortalised

human embryonic kidney

cells HEK 293T using

whole genome

sequencing (WGS)

 

DNA double strand

breaks as measured

using γH2AX

immunofluorescence

staining

100 μM for 24 h

exposure and WGS of

clonally expanded cells

populations

 

No metabolic

Activation

 

Cell viability analysed

in HEK 293T cells,

treated for 24 h with

0.1, 1 and 100 μM

BPA; cells were

stained with crystal

violet and results

reported as colony

area percentage

BPA from TCI (B04

94) purity ≥ 99% not

reported in the study

but available on the

website of the

company

Positive

 

Increased levels of

single base

substitutions, doublestrand

breaks and

small

insertions/deletions in

BPA-treated HEK 293T

cells in comparison

with DMSO-treated

controls

 

Single base

substitutions (C>A

transversions) in BPAtreated

cells

preferentially occur at

guanines

Mutations at A:T bp

were also reported

 

Colony formation

assay:

concentration dependent

decrease

in % colony area

 

Concentration dependent

increase in

DNA double strand

breaks as increased

number of nuclei with

> 5 γH2AX foci

Reliability: 2

Although there is no

TG for this type of

study, the research

was adequately

conducted and

reported

However, there is

uncertainty in the

level of toxicity of the

BPA treatment

Limited

Hu et al., 2021

Source:  Re-evaluation of the risks to public health related to the presence of bisphenol A (BPA) in foodstuffs, EFSA, (2021).

In vitro chromosomal aberrations test

Table 3: In vitro chromosomal aberrations test (OECD TG 473 was considered for the evaluation of reliability).

Test system/Test

object

Exposure

conditions

(concentration/

duration/metabolic

activation)

Information on the

characteristics of

the test substance

Results

Reliability/

Comments

Relevance of the

result

Reference

Chromosomal

aberrations and

SCE assays
 

CHO-K1 cell line

Cytotoxicity: cellcycle

delay

‘recognised by the

metaphases

without differently

staining sister

chromatids’
 

In vitro comet

assay (Table 5)*

BPA 0, 0.1 to 0.6 mM for 3 h

followed by 27 h recovery
 

100 metaphases
 

SCE: 50 metaphases
 

Without metabolic activation

BPA, purity >

99% (Tokyo

Kasei Kogyo

Co., Ltd)

Positive
 

Only in presence of severe

Cytotoxicity
 

Increased CA (0.5, 0.55, 0.6

mM, % of differently staining

sister chromatids 29%, 11%,

and 0%, respectively)
 

Increased endoreduplications

(0.45 and 0.55

mM)
 

Increased frequency of cmitosis-

like figures (above

0.3 mM)
 

Increased SCE (0.4 and 0.5

mM)

Reliability: 3
 

Only short-term

treatment; high

level of

cytotoxicity
 

The recovery time

exceeded the

recommended

(18–21 h)
 

Cells recovered in

the presence of

BrdU

Low

Tayama et al., 20081*

Chromosomal

aberration assay

CHO cells

Cytotoxicity: MTT

assay

Bacterial reverse

mutation assay

(Table 1)
 

In vitro

micronucleus assay

(Table 4),

comet assay (Table

5)*

BPA 0, 80, 100 and 120 μM

for 24 h

500 metaphases/group;

without metabolic activation

MTT assay: BPA 0, 40, 80,

100 and 120 μM for 12 and 24

h

BPA (purity

99%)2, was

purchased from

Tianjin Guangfu

Fine Chemical

Research

Institute

(Tianjin, China)

Positive
 

Increase of structural

chromosomal aberrations

from 80 μM, with significant

decrease in cell viability (but

not lower than 50%)

MTT assay: increase of cell

proliferation at 40 μM;

cytotoxicity from 80 μM

Reliability: 2
 

No short-term

Treatment
 

No positive

control

Limted

Xin et al., 2015*

Chromosomal

aberration assay in:

- MCF-7 human

breast cancer line;

- human

amniocytes from

male

[oestrogen

receptors (ER)

negative] and from

female (ER

positive)

Cytotoxicity: MTT

test

BPA 0, 0.4, 1, 4, 40 and 100

μg/mL for 48 h; 200 cells

analysed for each treatment

(less at highest concentrations

in amniocytes for high

toxicity)

Without metabolic activation

MTT test: BPA 0, 0.4, 1, 4, 40,

100 and 400 μg/mL for 48 h.

BPA, no

information on

purity or the

supplier

company

Positive
 

Increase of cells with

chromosome aberrations

(from 1 μg/mL) in all cell

types; the increase in cells

with aberrations was not

clearly concentration related

and decreased at the highest

concentrations, possible due

to cytotoxicity that was not

concurrently evaluated; no

clear association with ER

expression

In a preliminary evaluation of

cytotoxicity by the MTT test,

the IC50 of BPA was 100, 40

and 4 μg/mL in MCF-7 and

ER-negative (male) and ERpositive

(female) amniocytes,

respectively

Reliability: 2
 

Cells scored less

than

recommended in

OECD TG 473

No short-term

treatment

No positive

control

No concurrent

control of toxicity

Low
 

No information on

source and purity

of BPA

Aghajanpour-Mir et al.,

2016

Chromosomal

aberration assay

Human peripheral

blood lymphocytes

from 5 female

subjects

In vitro

micronucleus assay

(Table 4)*

BPA 0, 0.20, 0.10, 0.05, 0.02

and 0.01 μg/mL for 24 h

1000 metaphases

(200/subject)/concentration

Without metabolic activation

BPA (Sigma-

Aldrich) purity

≥97% not

reported in the

study but

available on the

website of the

company

Positive
 

Increase from 0.05 μg/mL

(prevalence of chromatid

breaks)
 

No numerical aberrations

Reliability: 2

No short-term

treatment

Limited

Santovito et al., 2018*

Chromosomal

Aberrations

Mouse embryonic

fibroblasts (MEF)

 

In vitro comet

assay (Table 5)*

BPA 150 μM for 24 h

or co-exposure with

camptothecin (CPT)

 

25 metaphases/treatment

were analysed

Without metabolic activation

BPA (Sigma-

Aldrich) purity

≥97% not reported in the

study but

available on the

website of the

company

Negative

No significant increase in CA

frequency

Cytotoxicity of BPA alone was

not measured but the

authors refer to 150 μM as

concentration with minimal

toxic effect from a previous

publication

Reliability: 3

 

Single

concentration

tested; low

number of

metaphases

analysed

No short-term

treatment

Low

Sonavane et al., 2018*

Chromosomal

aberrations

Human peripheral

blood mononuclear

cells (PBMC)

Cell proliferation:

MTT test

Cell-cycle analysis:

FACS

γH2AX:

western blot and

FACS analysis

BPA 0, 25, 50, 100 nM, cells

stimulated with PHA for 16h

and then treated with BPA for

48 h

30

metaphases/treatment/subject

(5 donors)
 

MTT test: BPA 0, 5, 10, 25,

50, 100, 200 nM and BPA 25,

50, 100, 200 μM, cells were

treated with or without PHA

for 16 h and then treated with

BPA for 24 and 48 h

γH2AX: cells treated with PHA

and then with BPA 50 nM for

24 h or 48 h (western blot) or

only for 24 h (FACS analysis

analysing T and B

lymphocytes)

Without metabolic activation

BPA (Merck)

Purity ≥97%

not reported in

the study but

available on the

website of the

company

Positive
 

Increased number of

aberrant cells, structural

chromosomal aberrations

and highly fragmented

metaphases
 

MTT test:

- unstimulated PBMCs:

decreased cell proliferation

only at 200 μM at both 24

and 48 h
 

PHA stimulated PBMCs:

- increased cell proliferation

from 10 nM to 100 nM;

- concentration-dependent

decreased cell proliferation

from 25 to 200 μM

Effect on cell proliferation

confirmed using cell-cycle

analysis

γH2AX (western blot):

- increase of protein

phosphorylation only at 24 h

(BPA 50 nM)

γH2AX (FACS): increase in CD3+ and in CD4+

T cells

Reliability: 2
 

No positive

Control
 

No short-term

treatment

Limited

Di Pietro et al., 2020

Chromosomal

aberrations assay

in human

peripheral blood

lymphocytes

BPA 0, 5, 10, 20 and 50

μg/mL for 24 and 48 h

Mitomycin C (MMC) at 0.10

μg/mL ‘was added to the

negative and a positive

controls and to each

concentration and chemical

groups as well’

Without metabolic activation

BPA, no

information on

purity or the

supplier

company

No data on chromosome

aberrations were reported

Reliability: 3
 

MMC added to all

Treatments
 

No mitogenic

Stimulation
 

No short-term

treatment

Low
 

No information on

BPA purity

Özgür et al., 2021

Source:  Re-evaluation of the risks to public health related to the presence of bisphenol A (BPA) in foodstuffs, EFSA, (2021).

In vitro mammalian cell micronucleus test

Table 4: In vitro mammalian cell micronucleus test (OECD TG 487 was considered for the evaluation of reliability).

Test system/Test

object

Exposure

conditions

(concentration/

duration/metabolic

activation)

Information on the

characteristics of

the test substance

Results

Reliability/

Comments

Relevance of the

result

Reference

Cytokinesis block

micronucleus assay

(CBMN)

AHH-1 cell line (human

lymphoblastoid cells)

Effects on mitotic spindle

using staining: brilliant

blue and safranin O; α-

and ƴ-tubulin

immunofluorescence

staining

BPA 0, 1.5, 3.1, 6.2,

7.7, 9.2, 10.8, 12.3,

18.5, 24.6, 37 μg/mL for

a complete cell cycle

(22–26 h),

Five experiments:

average of 8082 cells

scored for each

treatment

Effects on mitotic

spindle: BPA 0, 4.2–14

μg/mL for 20 h (one cell

cycle); 100 cells

undergoing mitosis

scored in each

experiment, 3

experiments

Without metabolic

activation

BPA (Sigma-Aldrich)

purity ≥97% not

reported in the study

but available on the

website of the

company

Positive
 

increased BNMN cells

from 12.3 μg/m
l

Aberrant mitotic

divisions (multiple

spindle poles)

Reliability: 1
 

BN cells % as

parameter of

cytotoxicity

High number of

analysed binucleated

cells

High

Johnson and Parry,

20081

Micronucleus test in:

- human umbilical

vascular endothelial cells

(HUVEC);

- human colon

adenocarcinoma (HT29)

cell line

Immunofluorescence

analysis of cytoskeleton

organisation of HUVEC

 

cells with anti-α-tubulin

and anti-γ-tubulin

Apoptosis using TUNEL

assay and cell viability

using CellTiter-Blue assay

BPA 0, 44 nM and 4.4

μM, (i.e. 10 ng/mL and

1 μg/mL) for 72 h

BPA 10 ng/mL and 1

μg/ml for 24 or 72 h

CellTiter-Blue assay:

BPA 10 ng/mL and 1

μg/mL for 24, 48 or 72

h

 

Without metabolic

activation

BPA, no information

on purity or the

supplier company

Positive in HUVEC

cells: slight increase

of MN frequency

Negative in HT29

cells

Multipolar spindles

and microtubule

misalignment

associated with BPA

exposure

 

No effects on cell

viability, proliferation

and apoptosis in both

cell lines

Reliability: 2

 

No analysis of cell

proliferation; no

positive control; no

short-term treatment

Low

 

No information on

source and purity of

BPA

Ribeiro-Varandas et

al., 2013

Cytokinesis block

micronucleus assay;

bovine peripheral blood

lymphocytes;

cell proliferation: nuclear

division index (NDI)

BPA 1×10−4, 1×10−5,

1×10−6 and 1×10−7

mol/L for 48 h

 

Without metabolic

activation

BPA (Sigma-Aldrich)

Purity ≥97% not

reported in the study

but available on the

website of the

company

Positive

 

concentration-related

increase in MN

frequency,

statistically

significant at the

highest

concentration; no

effect on NDI at any

concentration

Reliability: 2

 

No short-term

treatment; bovine

lymphocytes are not

commonly used in the

micronucleus test, and

their use has not been

validate.

However the

study appears to be

adequately performed

and reported

Limited

Šutiaková et al.,

2014

Micronucleus assay

CHO cells

Cytotoxicity: MTT test

Bacterial reverse

mutation assay (Table 1)

In vitro chromosomal

aberration (Table 3)

comet assay (Table 5)*

BPA 0, 80, 100 and 120

μM for 24 h, without

cytochalasin B; 1000

cells were scored for

each sample; 3

independent

experiments

Without metabolic

activation

MTT test:

- BPA 0, 40, 80, 100 and

120 μM for 12 and 24 h

BPA (purity 99%)2,

was purchased from

Tianjin Guangfu Fine

Chemical Research

Institute (Tianjin,

China)

Positive

 

increase in MN

frequency at 100 and

120 μM

MTT assay:

concentration-related

decrease in cell

viability from 100 μM

Reliability: 2

 

No short-term

Treatment

 

No positive control

Limited

Xin et al., 2015*

Cytokinesis-blocked

micronucleus assay in

murine macrophage

RAW264.7 cells

1000 binucleated

cells/concentration

Cell viability: MTT test

 

In vitro comet assay

(Table 5)*

BPA 0, 3, 10, 30, or 50

μM for 24 h

BPA 10 μM tested for

MN assay and cell

viability, in the presence

or absence of pretreatment

with N-acetyl-

L-cysteine (NAC) at the concentration of 10 μM

for 30 min

Without metabolic

activation

MTT test: BPA 0, 3, 10,

30, or 50 μM for 12 or

24 h

BPA (Sigma-Aldrich)

Purity ≥97% not

reported in the study

but available on the

website of the

company

Positive

 

Concentration dependent

increase

in MN frequency

from 10 μM

In the presence of

NAC, MN frequencyand cytotoxicity were

statistically

significantly reduced

(see also data on

ROS in Table 5)

MTT test:

concentration- and

time-dependent

decrease of cell

viability

Reliability: 2

 

No short-term

treatments; no

positive controls;

no data on cell

proliferation

Limited

Huang et al., 2018*

Cytokinesis block

micronucleus assay

Human peripheral blood

lymphocytes

from 5 female subjects

1000 binucleated

lymphocytes/subject

(5000 binucleated cells

per concentration)

In vitro chromosomal

aberrations assay (Table

3)*

BPA 0, 0.20, 0.10, 0.05,

0.02 and 0.01 μg/mL for

48 h

 

Without metabolic

activation

BPA (Sigma-Aldrich)

Purity ≥97% not

reported in the study

but available on the

website of the

company

Positive

 

Increase in MN

frequency from 0.02

μg/mL. At 0.2 μg/mL

4-fold increase with

respect to the vehicle

control (DMSO) level

No significant

reduction of the CBPI

value

Reliability: 2

 

No short-term

treatment

Limited

Santovito et al.,

2018*

Mitotic abnormalities and

micronuclei evaluated in

DAPI stained cells:

- Hep-2 cells (human

epithelial cells from

laryngeal carcinoma);

- MRC-5 cells (human

lung fibroblasts)

Cell viability using

CellTiter-Blue assay, after

48 h exposure

In vitro comet assay

(Table 5)*

BPA 0.44 nM, 4.4 nM,

4.4 μM (0.1 ng/mL, 1

ng/mL, 1 μg/mL) for 48

h; 1000 cells scored for

each treatment

BPA (Sigma) purity

≥97% not reported in

the study but

available on the

website of the

company

Positive

 

Slight (two-fold)

increase in MN

frequency from BPA

4.4 nM in both cell

lines

Mitotic index:

- in Hep-2 cells, no

effects;

- in MRC-5 cells,

statistically

significant increase

Cytotoxicity: no

effects on cell

viability

Reliability: 3

 

No short-term

treatment

Proliferation of the cell

population not

determined; extremely

low % of mitosis is

indicative of a very

low rate of cell

division, which is not

appropriate to

measure MN formation

 

Protocol of MN assay

not reported; no

positive control

Low

Ramos et al., 2019*

Micronucleus assay in

Chinese hamster V79-

derived cell lines

expressing various

human CYP enzymes

Micronucleus assay in

C3A cells (human

hepatoma cell line,

endogenously express

various CYP enzymes,

including CYP1A1, 1A2,

1B1, 2E1, 3A4, and phase

II metabolic enzymes,

such as UGTs and SULTs)

2000 cells analysed for

each treatment

Cytotoxicity: CCK-8 Assay

γ-H2AX in V79-Mz, V79-

hCYP1A1 cells and in C3A

cells; analysis using In-

Cell Western Blot

Immunofluorescence

staining of CENP-B of MN

induced in C3A cells

1) Micronucleus assay in

V79-derived cell lines:

- BPA 0, 40, 80, 160 μM

for 9 h + 15 h;

(recovery period);

- 2000 cells analysed for

each treatment

2) Micronucleus assay

in:

- V79-Mz, V79-

hCYP1A1 cells: BPA 0 to

80 μM for 24 h + 0 h ;

with or without ABT;

- C3A cells: BPA 0 to 80

μM for 72 h + 0 h; with

or without ABT or 7-HF

3) Micronucleus assay in

C3A cells: BPA 0 to 5

μM for 72 h + 0 h, with

or without KET or PCP

(phase II enzyme

inhibitors), an inhibitor

of UGT1 and SULT1,

respectively

Immunofluorescence

staining of CENP-B was

applied

Cytotoxicity performed

for each test using the

same testing conditions

of the MN assay or of

γH2AX analysis

γH2AX: BPA 0, 10, 20, 40, 80,

160 μM for 9 h; ABT (1-

aminobenzotriazole a

CYP inhibitor) or 7-HF (a

selective CYP1A1

inhibitor) were added

from 2 h ahead of test

compound exposure to

the end of cell culture

 

BPA 0, 10, 20, 40, 80,

160 μM for 9 h; ABT (1-

aminobenzotriazole a

CYP inhibitor) or 7-HF (a

selective CYP1A1

inhibitor) were added

from 2 h ahead of test

compound exposure to

the end of cell culture

BPA (99.6%),

AccuStandard Inc.

1) Micronucleus

assay (9 h + 15 h):

- Negative in V79-

Mz;

- Positive in V79-

hCYP1A1 cells and in

V79-hCYP1B1 cells;

- Cytotoxicity:

statistically

significant decrease

at the highest

concentrations

2) Micronucleus

assay (24 h + 0 h):

- Negative in V79-

Mz;

- Positive in V79-

hCYP1A1 cells, effect

abrogated by ABT

2)Micronucleus assay

(72 h + 0 h):

- Positive in C3A

cells, effect

abrogated by ABT or

7-HF;

- Cytotoxicity:

statistically

significant decrease

at the highest

concentrations

3)Micronucleus assay

in C3A cells (72 h +

0 h):

- Positive

- Effects enhanced

by KET or PCP;

statistically

significant increase of

MN negative for

CENP-B staining,

(clastogenic

mechanism)

Cytotoxicity:

statistically

significant increase in

cell viability from 2.5

μM

γH2AX:

- increase in V79-Mz,

in V79-hCYP1A1 cells

and in C3A cells

(concentration

dependent); effect

reduced by ABT or 7-

HF - Effects enhanced

by KET or PCP;

statistically

significant increase of

MN negative for

CENP-B staining,

(clastogenic

mechanism)

Cytotoxicity:

statistically

significant increase in

cell viability from 2.5

μM

γH2AX:

- increase in V79-Mz,

in V79-hCYP1A1 cells

and in C3A cells

(concentration

dependent); effect

reduced by ABT or 7-

HF

Reliability: 2

Micronucleus method

poorly described

No short-term

treatment

Limited

Yu et al., 2020

Source:  Re-evaluation of the risks to public health related to the presence of bisphenol A (BPA) in foodstuffs, EFSA, (2021).

In vitro DNA damage (comet assay)

Table 5: In vitro DNA damage (comet assay).

Test system/Test

object

Exposure

conditions

(concentration/

duration/metabolic

activation)

Information on the

characteristics of

the test substance

Results

Reliability/

Comments

Relevance of the

result

Reference

Alkaline comet assay

MCF-7 (oestrogen

receptor (ER) positive)

and MDA-MB-231 (ER

negative)

γH2AX foci using

immunofluorescence in

MCF-7 cells

MCF-7 cells exposure:

- BPA 0, 0.1 10, 100 μM

for 3 h;

- BPA 100 μM for 1, 3,

24 h

MDA-MB-231 cells

exposure:

- BPA 100 μM for 3, 24

h;

30 cells analysed (10

cells/slide)

Immunofluorescence in

MCF-7 cells: BPA 10 μM

for 3 h

Without metabolic

activation

BPA (Wako Pure Chemicals Industries,

Ltd) purity ≥99% not reported in the

study but available on the website of

the company

Positive

 

MCF-7:

increased comet tail

length after 3 h at

10, 100 μM and

after all exposure

times at 100 μM

MDA-MB-231:

increased comet tail

length after 3 and

24 h exposure times

at 100 μM

No toxicity in comet

assays

Induction of γH2AX

foci in MCF-7 cells

(10 μM)

ER-positive

MCF-7 cells are

more sensitive than

ER-negative

MDA-MB-231 cells to

BPA-induced DNA

damage

Reliability: 2

 

Only 30 cells were

Analysed

 

No positive control

Limited

Iso et al., 20061

Alkaline comet assay in

CHO-K1 cell line

In vitro chromosomal

aberrations (Table 3)*

BPA 0, 0.2, 0.3, 0.4, 0.5,

0.6, 0.7 mM; 1 h

exposure

Positive control: H2O2

200 cells were scored

Quantification of DNA

damage: a score of 0–3

(mean score value =

mean comet points/cell,

comet points)

Cell viability (trypan

blue)

Without metabolic

activation

BPA, purity > 99% (Tokyo Kasei Kogyo

Co., Ltd)

Positive

Increased DNA

strand breaks

only at the highest

concentration tested

(0.7 mM)

Reliability: 3

 

Non-standard

method of DNA

damage

quantification

Data of

cytotoxicity not

clearly reported

Low

Tayama et al.,

20081*

Alkaline comet assay

HepG2 cells

Cell viability: MTT test

Bacterial reverse

mutation assay (Table

1)*

BPA 0, 0.1, 1.0 and 10.0

μM for 4 and 24 h; 50

nuclei scored/treatment;

at least 2 independent

experiments; positive

control: benzo[a]pyrene

 

MTT test: 12.5, 25, 50,

100 μM for 24 h

BPA, purity >99% (Sigma-Aldrich)

Negative after 4 h

of exposure

 

Equivocal after 24

h exposure (no

concentration related

effect)

 

No cytotoxicity was

observed

Reliability: 2

 

Only 50 nuclei

scored

Limited

Fic et al., 20131*

Comet assay in

rat INS-1 insulinoma

cells

Cell viability: Hoechst

staining kit and trypan

blue (apoptotic cells

detection)

Expression of nuclear

p53 and p-Chk2 (T68)

proteins: western

blotting

Intracellular (ROS):

DCFH-DA

Glutathione (GSH):

detection with ophthalaldehyde

(OPT)

BPA 0, 25, 50, 100 μM

for 24 h; or pretreatment

with or

without NAC (10 mM) for

1 h then BPA (100 μM)

was added for 24 h;

Without metabolic

activation

50 cells/slide were

analysed; 3 experiments

ROS and GSH analysis:

BPA 0, 25, 50, 100 μM

for 24 h

 

ROS measurements also

in cells pre-treated with

NAC and exposed to 100

μM BPA

BPA, purity 99% (Sigma-Aldrich)

Positive

concentration related

increase in

tail DNA %, tail

moment and tail

length at 50 and

100 μM

 

Significant decrease

in tail DNA % in

cells pre-treated

with NAC

 

No apoptotic cells

and 90% cell

survival were used

in comet assays

(results are not

shown)

 

Increase of

expression of DNA

damage-associated

proteins: p53 (from

50 μM) and p-Chk2

(at 100 μM)

Levels of p53 are

reduced by NAC

pre-treatment

Intracellular ROS:

increase at 50 and

100 μM

Decrease of ROS

upon NAC pretreatment

 

GSH: concentration related

decrease

 

Reliability: 2

 

No positive

control;

results on

cytotoxicity

assessment are

not reported

Limited

Xin et al., 2014

Alkaline comet assay in

CHO cells

Cytotoxicity: MTT assay

Bacterial reverse

mutation assay (Table 1)

In vitro chromosomal

aberration (Table 3)

micronucleus assays

(Table 4)*

BPA 0, 40, 80, 100 and

120 μM for 12 and 24 h;

100 cells were

analysed/sample.

Without metabolic

activation

MTT assay: BPA 0, 40,

80, 100 and 120 μM for

12 and 24 h

BPA (purity 99%)2, was purchased

from Tianjin Guangfu Fine Chemical

Research Institute (Tianjin, China)

Positive

Concentration related

increase in

(%) tail DNA from

80 μM with 12 h

treatment, and at all

tested

concentrations after

24 h

MTT assay:

decrease in cell

viability (but less

than 50%) from 80

μM after 12 and 24

h

Reliability: 2

 

No positive control

Limited

Xin et al., 2015*

Alkaline comet assay

NIH3T3 cells (mouse

embryonic fibroblast cell

line)

At least 100

nucleoids/sample

Cytotoxicity: CCK-8

assay and LDH release

Intracellular ROS: DCFHDA

8-OHdG: EpiQuick 8-

OHdG DNA damage

quantification direct kit

γH2AX:

immunofluorescence and

western blot

BPA 0, 2, 10 and 50 μM

(0.4–11 μg/mL) for 24 h

CCK-8 and LDH assays,

ROS, 8-OHdG, γH2AX

analysis: BPA 0, 2, 10

and 50 μM for 24 h

At least 100 nucleoids of

each sample were

obtained in 3

independent experiments

without metabolic

activation

BPA (Sigma-Aldrich) purity >97% not

reported in the study but available on

the website of the company

Positive

 

increase tail DNA%

at 50 μM

Cytotoxicity: 80%

cell survival at 50

μM

γH2AX, ROS and 8-

OHdG: increase at

50 μM

Reliability: 2

 

No positive control

Limited

Chen et al., 2016

Alkaline comet assay in

FRTL-5 rat immortalised

thyrocyte cell line

Cell proliferation

(population doubling)

Transcriptome analysis

(microarray)

Intracellular ROS:

H2DCFDA

BPA 10−9 M for 6h, 48h,

96 h; 100 cells for each

condition

Transcriptome analysis

and intracellular ROS:

cells exposed for 1, 3,

and 7 days to 10−9 M

BPA

Without metabolic

activation

BPA (Sigma-Aldrich), purity ≥97% not

reported in the study but available on

the website of the company

Comet assay on BPA

alone: Negative

Intracellular ROS:

statistically

significant increase

after 1 and 3 days

exposure

Transcriptome

analysis: decreased

expression of genes

involved in DNA

replication,

recombination and

repair (confirmed by

RT-PCR) (after 3

and 7 days BPA

exposure)

Reliability: 3

 

Comet assay:

- one low

concentration

tested;

- no positive

control

Small effects on

transcription

Large variations in

DNA strand breaks

in the comet

assay

Low

Porreca et al.,

2016

Comet assay

MCF-7 cells (from

human breast

adenocarcinoma)

Cell viability: CCK-8

assay

Cell membrane damage:

LDH

ROS

BPA 0, 1, 10, 25, 50 μM;

24 h

Positive control: tBHP

(tert-butyl

hydroperoxide); 300 cells

from each sample were

analysed

Without metabolic

activation

CCK-8 assay: 0, 0.01,

0.1, 1, 10, 25, 50, 100

μM for 24 h

LDH: 0, 1, 10, 25, 50,

100 μM for 24 h

ROS: 0, 0.01, 0.1, 1, 10,

25, 50 μM for 24 h

BPA, purity > 98% (Tokyo Chemical

Industry)

Positive

 

Concentration dependent

increase in % tail

DNA from 10 μM

Cell viability:

at 1 μM

increase in cell

viability; inhibition of

cell viability at

concentrations from

10 μM (70%) to 100

μM (80%)

Cell membrane

damage:

increase in LDH

release in a

concentration dependent

manner

from 10 μM

ROS formation:

concentration dependent

increase

in ROS levels

No measurement at

50 μM, because of

excessive cell death

(90%)

Reliability: 3

 

Excessive toxicity

at the analysed

positive

concentrations

 

Results of positive

control are not

reported

 

Comet methods

are not described

in detail

Low

Lei et al., 2017

Alkaline comet assay

HepG2 cells

Cytotoxicity: MTT assay

Oxidative stress:

intracellular ROS: DCFHDA

in the same cells,

also MDA and SOD

BPA from 10–8 to 10–6

mol/L (0.02–22.8 μg/mL)

for 24 h

 

MTT: BPA from 10–8 to

10–4 mol/L for 24 h

ROS, MDA and SOD

analysis: BPA from 10–8

to 10–4 mol/L for 6 h

Positive control: H2O2

BPA purity > 99.8% (Sigma-Aldrich)

Positive

 

Concentration related

increase of

tail DNA (%)

MTT: concentration related

increase of

cytotoxicity;

increase of ROS and

MDA; decrease of

SOD

Reliability: 2

 

No sufficient

details on the

comet method

 

(e.g. number of

cells analysed is

not specified)

Limited

Li et al., 2017

Alkaline and neutral

comet assay

Human PBMC (3 donors)

450 cells/concentration

Cytotoxicity using flow

cytometry

Alkaline comet assay:

- BPA 0.1, 1 and 10

μg/mL for 1 h;

- 0.01, 0.1, 1 and 10

μg/mL for 4 h

Neutral comet assay:

- BPA 0.1, 1 and 10

μg/mL for 1 h

DNA repair: BPA at 10

μg/mL

Without metabolic

activation

BPA, 99–99.5% purity (Sigma-Aldrich)

Positive

 

Both alkaline and

neutral comet

DNA repair of DNA

breaks:

decrease at 60 min,

but the repair was

not complete after

120 min

Reliability: 2

 

unusual software

for comet analysis

 

No positive control

Limited

Mokra et al.,

2017

Alkaline comet assay

and modified

comet assay with Fpg

enzyme in human

peripheral blood

lymphocytes

1 h exposure to BPA:

0.001 mM, 0.1 mM, 2.5

mM

Three experiments

BPA (Sigma-Aldrich)

Purity ≥97% not reported in the study

but available on the website of the

company

Positive

 

Increase of % tail

DNA, only at the

first 2

concentrations

tested

With Fpg a higher

increase of % tail

DNA was observed

at all

concentrations, but

not concentration

related

Reliability: 3

 

Inadequate

response of

positive control;

the use of

hydrogen peroxide

as positive control

is not adequate

for the comet +

Fpg

Number of cells

scored in not

specified

Low

Durovcova et al.,

2018

Comet assay in human

sperm cells

 

Cell viability measured

with a Nucleocounter NC

3000

In vivo comet assay

(Table 8)**

BPA 0, 1, 1.5, 2 and 3

μmol/L for 1 h

 

Without metabolic

activation

Each concentration was

scored in 3 independent

experiments

and 2 replicates of each

experiment

600 cells were

scored/concentration

Cell viability: BPA from 0

to 5 μmol/L

BPA (purity >99%, Sigma-Aldrich)

Negative

No differences in %

tail DNA between control samples and

BPA-treated cells at

all concentrations

tested

Cell viability assay:

concentrationdependent

decrease

in cell viability from

3 μmol/L (reduced

cell viability to 60%)

Reliability: 3

 

Test not validated

and not adequate for cryopreserved

samples

Low

Sharma et al.,

2018**

Comet assay in human

bronchial epithelial

BEAS-2B cells

Cytotoxicity: MTS assay

after 24 h treatment

γ-H2AX foci using

immunofluorescence

Intracellular ROS: DCF

proteins involved in the

DNA damage response

(p-ATM, p-ATR, p-Chk1,

p-p53) using western

blot

BEAS-2B cells were

exposed to BPA 200 μM

for 24 h

MTS assay: 12.5 to 200

μM; tests performed in

triplicates and for at

least 3 independent

times

Without metabolic

activation

BPA (Sigma-Aldrich) purity ≥97% not

reported in the study but available on

the website of the company

Increase of DNA

damage, but no

quantitative data are

reported

MTS assay:

- concentrationdependent

cytotoxic

effect;

- cytotoxicity at 200

μM: 84.7 ± 2.1%;

γ-H2AX: BPAinduced

phosphorylation

BPA-induced also

phosphorylation of

ATM/ATR complex

and triggered Chk1

and p53 proteins

Statistically

significant increase

of ROS

Reliability: 3

 

Only one

concentration

tested, which

resulted in high

cytotoxicity

Comet assay

results not

reported in detail,

(no quantitative

data)

 

No positive control

Low

George and

Rupasinghe,

2018

Comet assay in

TM3 murine Leydig cells

Cell viability: MTT assay

Real-time cell growth

kinetics [cellular index

(CI)]

Cell-cycle analysis (PI,

FACS analysis)

Morphological analysis of

cell death: chromatin

staining with the

Hoechst 33342 dye

BPA 0, 1, 10 and 100 μM

for 3 h;

cell viability analysed

with trypan blue

exclusion method;

Positive control:

doxorubicin;

250 nucleoids were

analysed in each

repetition (3

experiments)

Without metabolic

activation

BPA concentrations for

MTT assay and real-time

cell growth kinetics: 0,

0.5, 1, 5, 10, 50, 100,

250, 500 μM

MTT assay exposure: 24

or 48 h

Real-time cell growth

kinetics: measurement

every 30 min for 96 h

Cell-cycle analysis,

chromatin staining: BPA

0, 1, 10 and 100 μM for

24 or 48 h

BPA (Sigma-Aldrich) purity ≥97% not

reported in the study but available on

the website of the company

Negative

 

No increase in

damage index (DI)

Cell viability was

evaluated using

trypan blue

exclusion method,

and only treatments

with an index

greater than 80%

were considered

(results not shown)

Cell viability:

statistically

significant and

concentrationrelated

decrease

from 5 and from 50

μM after 24 and 48

h exposure,

respectively

CI: TM3 cells

exhibited a decrease

in their CI after 34 h

of exposure at

concentrations from

10 μM

BPA 100, 250 and

500 μM decreased

CI within a few

hours of exposure

Cell-cycle analysis:

BPA 100 μM induced

an increase in the

sub-G1 phase cell

population

 

No other effects

induced in the

distribution of TM3

cells in the G0 + G1,

S, and G2 + M

phases

Morphological

analysis of cell

death: increase in

chromatin staining

upon exposure to

BPA 100 μM for 24

or 48 h

Reliability: 3

 

Results are

reported as

damage index

(not a standard

parameter)

Low

Gonçalves et al.,

2018

Alkaline comet assay

with repair enzymes

[with DNA glycosylases,

i.e. endonuclease III

(Nth) and human 8-

oxoguanine DNA

glycosylase (hOGG1)]

Oxidised purines and

pyrimidines

Human PBMC

300 comets from 2

independent

experiments

Cell viability: flow

cytometry

BPA 0, 0.01, 0.1 and 1

μg/mL for 4 h

and 0, 0.001, 0.01 and

0.1 μg/mL for 48 h

Positive control: H2O2

(2 blood donors)

Without metabolic

activation

BPA, 99–99.5% purity (Sigma-Aldrich)

Positive

 

After 4 h incubation:

- statistically

significant and

concentrationdependent

oxidative

damage to purines

(from 0.01 μg/mL)

and to pyrimidines

(from 0.1 μg/mL)

After 48 h

incubation:

- concentrationdependent

oxidative

DNA damage to

purines (from 0.001

μg/mL) and to

pyrimidines from

(0.01 μg/mL)

Statistically

significant

differences for DNA

damage between 4

h and 48 h exposure

 

at the highest

concentrations

tested (0.01 and 0.1

μg/mL)

Cell viability: no

significant changes

Reliability: 2

 

No appropriate

positive control

unusual software

for comet analysis

Limited

Mokra et al.,

2018

Alkaline comet assay

(CometChip platform) in

mouse embryonic

fibroblasts (MEF)

Analysis of γH2AX

(immunofluorescence)

In vitro chromosomal

aberrations test (Table

3)*

BPA 150 μM for 24 and

48 h (24 h for γH2AX),

or co-exposure with

camptothecin (CPT)

Data of 4 replicates,

each with 1500 ± 300

comets

Without metabolic

activation

BPA (Sigma-Aldrich) purity ≥97% not

reported in the study but available on

the website of the company

Negative

 

No significant

increase in the %

tail DNA

No significant

increase in the

percentage of

γH2AX-positive

nuclei

Reliability: 3

 

No positive

controls, no

sufficient details

on the methods

applied; single

concentration;

cytotoxicity not

evaluated

Low

Sonavane et al.,

2018*

Comet assay in murine

macrophage RAW264.7

cells

Cell viability: MTT assay

Intracellular ROS level:

semiquantitative DCFHDA

fluorescence assay

Assessment of the

antioxidative enzymes

activities: CAT, SOD, and

GPx

In vitro micronucleus

assay (Table 4)*

BPA 0, 3, 10, 30, or 50

μM for 24 h; no positive

control; a minimum of 50

cells/slide were analysed

MTT assay:

BPA 0, 3, 10, 30, or 50

μM for 12 or 24 h

DCFH-DA assay and

assessment of

antioxidative enzymes

activities:

- BPA 0, 3, 10, 30, or 50

μM for 24 h

Without metabolic

activation

BPA (Sigma-Aldrich) purity ≥97% not

reported in the study but available on

the website of the company

Positive

 

Increase in tail

moment and tail

length in a

concentrationdependent

manner

starting from 10 μM

of BPA

Cytotoxicity:

concentration- and

time-dependent

decrease of cell

viability

BPA-induced ROS

generation and

reduced

antioxidative

enzyme activities

from 10 μM

Reliability: 2

 

No positive control

Limited

Huang et al.,

2018*

Comet assay and comet

modified with FpG

In cryopreserved:

- Hep-2 cells (human

epithelial cells from

laryngeal carcinoma);

- MRC-5 cells (DNA

damage responsive cell

line, human lung

fibroblasts)

Cell viability: CellTiter-

Blue assay

In vitro micronucleus

assay (Table 4)*

BPA 0.44 nM, 4.4 nM,

4.4 μM for 48 h;

Hep-2 cells: 300 cells

analysed for each

treatment

MRC-5 cells: 100 cells

analysed for each

treatment

Cell viability: BPA 0.44

nM, 4.4 nM, 4.4 μM, 48 h

exposure in both Hep-2

and MRC-5 cells

BPA (Sigma) purity ≥97% not reported

in the study but available on the

website of the company

Inconclusive

Reliability: 3

 

Comet assay is

not validated and

recommended for

testing

cryopreserved cell

samples

No positive control

Low

Ramos et al.,

2019*

Comet assay in sperm

cells from Sprague

Dawley rats

Analysis: ROS, LPO, SOD

In vivo comet assay

(Table 8)**

BPA 0, 1, 10, and 100

μg/L for 2 h

No positive control

Without metabolic

activation

BPA (99% purity) Santa Cruz

Biotechnology

Positive

Increase of tail

DNA% only at 100

μg/L

BPA increased SOD,

ROS, TBARS

[thiobarbituric acid

reactive substances

(TBARS) as an index

of LPO] only at 100

μg/L

Reliability: 3

 

The study was

performed

following a nonstandard,

neutral

protocol and

unusual evaluation

of comets based

on the analysis of

microphotographs.

No positive control

Low

Ullah et al.,

2019**

Comet assay in Marc-

145 cells (rhesus

monkey embryo renal

epithelial cells)

Cytotoxicity: MTT and

LDH assays

Intracellular ROS

levels: DCFH-DA

Lipid peroxidation: - TBARS;

- SOD activity and GSH

content

BPA 10–6 to 10–3 M for 24

h; 50 cells from each of

6 independent

experiments were

analysed

MTT assay: BPA 10–6 to

10–1 M for 24 h;

DCFH-DA and TBARS

assays: BPA 10–6 to 10–3

M for 24 h; SOD activity and GSH

content: BPA 10–6 to 10–3

M for 24 h

Without metabolic

activation

BPA (purity > 99%) Sigma-Aldrich

Positive

 

Increase in % tail

DNA, tail length and

tail moment (10–6 -

10–3 M);

Cytotoxicity:

concentrationrelated

increase;

excess of toxicity at

10–3 and 10–4 M BPADCFH-DA, TBARS

assays:

- concentrationrelated

increase of

ROS and lipid

peroxidation;

- SOD activity and

GSH content:

concentrationrelated

decrease

Reliability: 2

 

No positive control

Limited

Yuan et al., 2019

Alkaline comet assay

and Fpg modified comet

assay

RWPE-1 cells [human

papilloma virus 18

(HPV18) immortalised,

non-tumorigenic

prostatic cell line]

Cell viability: modified

MTT assay and trypan

blue exclusion

Enzymatic and nonenzymatic

antioxidants:

analysis of GPx, GR,

SOD, GSH and TAOC

levels

BPA 0, 45 μM (IC20) for

24 h

450 comets

analysed/treatment;

experiments in triplicates

Cell viability: 0, 50, 100,

200, 300, 600 μM for 24

h

Enzymatic and nonenzymatic

antioxidants:

BPA 0, 45 μM (IC20) for

24 h

Without metabolic

activation

BPA (>99% pure)

Positive

Comet assay:

increase (2.5-fold)

in tail intensity (at

IC20 BPA)

Fpg modified comet:

increase in tail

intensity

Cell viability:

decrease in cell

viability (IC20 45 μM)

Enzymatic and nonenzymatic

antioxidants:

decrease in:

- GPx1 and SOD

activity (29% and

24% respectively);

- TAOC levels

(20%);

increase in:

- GR activity (4.5-

fold);

- total GSH level

(30%)

Reliability: 2

 

One concentration

tested

No positive control

No metabolic

activation

Limited

Kose et al., 2020

Comet assay in HepG2

cells (human

hepatocellular carcinoma

cell line)

Cell viability: MTT test

SOS/umuC assay (Table

1)*

BPA 0, 1, 10, 100 and

1000 μg/L, for 4 and 24

h; 3 independent

experiments; 50 nuclei

analysed/treatment

MTT test: BPA 0, 1, 10,

100 and 1000 μg/L, for

24 h

BPA (Sigma-Aldrich) purity >97% not

reported in the study but available on

the website of the company

Positive

 

increase of % tail

DNA from 10 μg/L

at both 4 h and 24 h

exposure

MTT test: no effects

on cell viability

Reliability: 2

 

Low number of

nuclei analysed

Limited

Balabanič et al.,

2021*

Source:  Re-evaluation of the risks to public health related to the presence of bisphenol A (BPA) in foodstuffs, EFSA, (2021).

In vivo chromosomal aberrations assay

Table 6: In vivo chromosomal aberrations assay (OECD TG 475 was considered for the evaluation of the reliability).

Test system/Test

object

Exposure

conditions

(concentration/

duration/metabolic

activation)

Information on the

characteristics of

the test substance

Results

Reliability/

Comments

Relevance of the

result

Reference

Chromosomal

aberration assay in

bone marrow

Swiss albino mice

Six animals (3 females

and 3 males)/group

(control and BPAtreated

animals)

100 metaphases were

scored per animal

Mitotic effects

In vivo micronucleus

assay (Table 7)*

BPA 0, 10, 50 and 100

mg/kg bw; 2% gum

acacia was used as

the suspending

medium for BPA

Single oral dose

administered by

gavage

Sampling of bone

marrow at 6, 24, 48

and 72 h

Cumulative dose level:

10 mg/kg bw for 5

consecutive days

Sampling of the bone

marrow 24 h after the

last administration of

BPA

BPA, purity 98%

(Loba Chemie,

Mumbai, India)

Negative

No significant increase

of structural

chromosomal

aberrations

Significant increases in

the frequencies of

gaps at all doses at 48

and 72 h sampling

time and at 50 and

100 mg/kg bw at the

24 h sampling time

C-mitotic effects

through increases of

mitotic indices and

decrease in anaphase

for both higher dose

level at 24, 48 and 72

h sampling times

Reliability: 2

 

Low number of

animals/sex, but in

total 6 animals/group

Low number of

metaphases scored,

treatment with

colchicine shorter (1.5

h) than recommended

(5–6 h)

Limited

Naik and Vijayalaxmi,

20091*

Chromosomal

aberration in bone

marrow

 

Holtzman rats

Ten animals (5

females and 5

males)/group (control

and BPA-treated

animals)

Analysis of 100

metaphases per

animal

In vivo micronucleus

assay (Table 7)* and

comet assay (Table

8)*

Bacterial reverse

mutation assay (Table 1)**

 

BPA 0, 2.4 μg, 10 μg,

5 mg and 50 mg/kg

bw administered orally

once a day for 6

consecutive days; BPA

dissolved in distilled

ethyl alcohol and

diluted with sesame oil

Sampling of the bone

marrow 24 h after the

last administration of

BPA

BPA, 99% purity

(Sigma Chemical

Company)

Positive

 

Dose-related increase

of structural

chromosomal

aberrations starting

from 10 μg

Reliability: 2

 

Mitotic index as a

measure of

cytotoxicity not

determined

Limited

Tiwari et al.,

20121*,**

Source:  Re-evaluation of the risks to public health related to the presence of bisphenol A (BPA) in foodstuffs, EFSA, (2021).

In vivo micronucleus assay

Table 7: In vivo micronucleus assay
(OECD TG 474 was considered for the evaluation of the reliability).

Test system/Test

object

Exposure

conditions

(concentration/

duration/metabolic

activation)

Information on the

characteristics of

the test substance

Results

Reliability/

Comments

Relevance of the

result

Reference

Micronucleus assay

Male ICR mice

Peripheral blood

reticulocytes

(1000/animal

analysed, 5 mice per

group)

Bacterial reverse

mutation assay (Table

1)**

228 mg/kg bw of BPA

dissolved in DMSO,

once by gavage;

controls received

vehicle alone

Peripheral blood

collected at 24, 48

and 72 h after

administration

BPA purity >99%

(Tokyo Kasei Kogyo

Co., Ltd)

Inconclusive

 

(negative with no

demonstration of bone

marrow exposure)

No increase of

micronucleated

reticulocytes at any

sampling time

Cytotoxicity was not

evaluated

Reliability: 2

 

Single dose tested,

although relatively high;

1000 scored

reticulocytes/animal

instead of 2000 as in

OECD TG 474 (1997)

No positive control

Low

Masuda et al.,

20051**

Micronucleus assay in

bone marrow

Male mice

(102/ElxC3H/El)F1 (5

animals per group)

BPA 0, 0.002, 0.02

and 0.2 mg/kg bw

oral gavage on 2 days

Cells collected 24 h

after last

administration

2000 polychromatic

erythrocytes (PCE)

were scored per

animal

BPA (Sigma-Aldrich)

purity >97% not

reported in the study

but available on the

website of the

company

Inconclusive

 

(negative with no

demonstration of bone

marrow exposure)

No induction of

micronuclei in the bone

marrow polychromatic

erythrocytes

Reliability: 2

 

No positive control;

very low doses applied

Low

Pacchierotti et al.,

20081

Cytogenetic analyses

of oocytes and

zygotes in female

C57Bl/6 mice

Assessment of meiotic

delay in

spermatocytes by

BrdU incorporation

and aneuploidy in

epididymal sperm by multicolor FISH in

male

102/ElxC3H/El)F1

mice (5 mice per

dose)

Acute exposure: 0.2

or 20 mg/kg

Sub-acute exposure:

0.04 mg/kg for 7 days

by gavage

Sub-chronic exposure:

0.5 mg/L for 7 weeks

in drinking water 0.2 mg/kg bw starting

on day 8 after BrdU,

for 6 consecutive days

BPA 0, 0.002, 0.02

and 0.2 mg/kg for 6

consecutive days

BPA (Sigma-Aldrich)

Negative

 

No significant induction

of hyperploidy or

polyploidy in oocytes

and zygotes in any

treatment condition

 

No delay of meiotic

divisions

No induction of

hyperploidy or

polyploidy in epididymal

sperms

Reliability: 2

 

This study was

adequately planned,

performed and

reported, even though

specific guidelines for

the effects in germ cells

are not available

No positive control

 

Very low doses for the

analysis of sperm

aneuploidy

Limited

Micronucleus in bone

marrow

Swiss albino mice

Six animals (3 females

and 3 males)/group

(control and BPAtreated

animals);

2000 PCE/animal

In vivo chromosomal

aberration (Table 6)*

BPA 0, 10, 50 and 100

mg/kg bw; 2% gum

acacia was used as

the suspending

medium for BPA

Single oral dose

administered by

gavage sampling of

bone marrow at 6, 24,

48 and 72 h

Cumulative dose

level: 10 mg/kg bw

for 5 consecutive days

Sampling of the bone

marrow 24 h after the

last administration of

BPA

BPA purity 98%

(Loba Chemie,

Mumbai, India)

Negative

No significant decrease

of PCE/NCE ratio

Significant increase of

gaps and C-mitoses

Reliability: 2

 

Low number of

animals/sex in each

group, but in total 6

animals/group

Limited

Naik and Vijayalaxmi,

20091*

Micronucleus in bone

marrow

Male Sprague Dawley

rats

8 rats/group (control

and BPA-treated

animals)

In vivo comet assay

(Table 8)*

BPA 0, 200 mg/kg bw

per day for 10 days

Orally via drinking

water

Bone marrow

processed at the end

of treatment

BPA (Sigma-Aldrich)

purity >97% not

reported in the study

but available on the

website of the

company

Inconclusive

(negative with no

demonstration of bone

marrow exposure)

No data on bone

marrow toxicity are

reported

Reliability: 2

 

Exposure of the bone

marrow not

demonstrated

Single dose tested

No positive control

Low

De Flora et al.,

20111*

Micronucleus in bone

marrow

Holtzman rats

Ten animals (5

females and 5

males)/group (control

and BPA-treated

animals)

In vivo chromosomal

aberration (Table 6)*

Comet assay (Table

8)*

Bacterial reverse

mutation assay (Table

1)**

BPA 0, 2.4 μg, 10 μg,

5 mg and 50 mg/kg

bw per day

administered orally for

6 consecutive days

Sampling of the bone

marrow 24 h after the

last administration of

BPA

Analysis of 2000 PCE

BPA, 99% purity

(Sigma Chemical

Company)

Positive

 

Dose-related increase of

MN-PCE starting from

10 μg/kg bw per day

Reliability: 2

 

Inappropriate staining

Limited

Tiwari et al.,

20121*,**

Micronucleus test in

peripheral blood

reticulocytes and in

bone marrow of

Pzh:Sfis female mice

No. of animals/group:

9 in control, 6 in BPA

5 mg/kg bw, 8 in BPA

10 mg/kg bw, 6 in

BPA 20 mg/kg bw;

1000 reticulocytes or

PCE were scored

In vivo comet assay

(Table 8)*

BPA 5, 10, or 20

mg/kg bw per day for

2 weeks in drinking

water

Animals were

sacrificed 24 h after

the end

of treatment

Blood was collected at

1 and 2 weeks of

exposure

BPA, no information

on purity or the

supplier company

Positive in

reticulocytes at 10 and

20 mg/kg bw after 2

weeks of exposure

 

Negative in

reticulocytes after 1

week of treatment

 

Negative in bone

marrow

Reliability: 2

 

No criteria for scoring

micronuclei were

described

No positive control

Low

 

No information on

source and purity of

BPA

Gajowik et al., 2013*

Micronucleus test in

bone marrow cells

Adult male Wistar

albino rats

Ten animals per group

Oral administration of

5 μg, 50 μg and 100

μg BPA/100 g bw

once a day for 90

days, sacrifice and

sampling of bone

marrow on the 91th

day

BPA (<99% pure)

purchased from

Sigma-Aldrich, diluted

in olive oil

Positive

 

Increases (2–3-fold at

the highest dose) in the

frequency of

micronuclei in

polychromatic

erythrocytes and

normochromatic

erythrocytes

Statistical significance of

the difference with

negative controls not

determined

No decrease in PCE/NCE

ratio

Reliability: 3

 

Major limitation in data

presentation and

analysis: low number of

scored cells per animal

lack of historical control

data

Low

Srivastava and Gupta,

2016 [

Micronucleus test in

bone marrow

Male Swiss albino

mice, 10

animals/group;

analysis of 2000

PCE/animal

In vivo comet assay

(Table 8)*

50 mg/kg bw, orally

once a day for 28

days

Sampling of the bone

marrow at the end of

treatment

BPA, purity ≥ 99%,

(Sigma-Aldrich)

Positive

 

Increase in the mean

values of MNPCEs

(66.40 ± 9.94 vs 10.40

± 2.96)

Cytotoxic (reduction in

the ratio of PCE/NCE

compared to control)

Reliability: 2

 

No positive control only

one dose

Limited

Fawzy et al., 2018*

Micronucleus test in

bone marrow

Male Wistar rats; 6

animals/group

Analysis of 2000 PCE

for MN scoring and of

200 cells for PCE/NCE

Ratio

 

Lipid peroxidation:

serum level of

malondialdehyde

(MDA)

(8-OHdG) in urine

In vivo comet assay

(Table 8)*

0, 50 and 100 μg/kg

bw per day, 4 weeks,

by gavage

Sampling at the end

of treatment

BPA (Sigma-Aldrich)

purity >97% not

reported in the study

but available on the

website of the

company

Positive

Significant dose-related

increase (up to 3-fold)

in the mean values of

MNPCEs compared with

control

Cytotoxic (a weak

statistically significant

decrease in PCE/NCE

ratio); dose-related

increase of MDA in

blood and of urinary 8-

OHdG levels

Reliability: 2

 

No positive control only

2 doses

Limited

Panpatil et al., 2020*

Source:  Re-evaluation of the risks to public health related to the presence of bisphenol A (BPA) in foodstuffs, EFSA, (2021).

In vivo DNA damage

Table 8: In vivo DNA damage (comet assay, OECD TG 489 was considered for the evaluation of the reliability).

Test system/Test

object

Exposure

conditions

(concentration/

duration/metabolic

activation)

Information on the

characteristics of

the test substance

Results

Reliability/

Comments

Relevance of the

result

Reference

Comet assay in

peripheral blood

lymphocytes

Sprague Dawley rats

8 rats/group (control

and BPA-treated

animals); 100 nuclei

were scored

In vivo micronucleus

assay (Table 7)*

200 mg/kg bw for 10

consecutive days,

orally via drinking water

Sampling at the end of

treatment

BPA (Sigma-Aldrich)

purity >97% not

reported in the study

but available on the

website of the

company

Negative

Reliability: 2

 

Tail moment, used as

only parameter to

report the results for

the comet assay, is

not recommended by

the Comet

international

Committee; single

dose tested;

no positive control

Limited

De Flora et al.,

20111*

Comet assay in

peripheral whole blood

cells of Wistar rats (6

animals/group BPAtreated

animals; 5

animals in the control

group; 3 animals in the

positive control group)

0, 125 and 250 mg/kg

bw; oral administration

(gavage) for 4 weeks

Positive control: MMS

(i.p., sampling after 24 h);

50 cells were analysed on

each replicated slide

BPA purity > 99%

(Merkolab Chemistry)

Positive

 

Increase of both tail length

and tail moment at 250

mg/kg bw

Reliability: 3

 

Inappropriate

presentation and

evaluation of results

Group mean tail

length and tail

moment values,

rather than the

means of animal

median values

(OECD TG 489)

Sampling time, and

frequency of

administrations not

stated

Low

Ulutaş et al., 20111

Comet assay in blood

lymphocytes

Holtzman rats

Ten animals (5 females

and 5 males)/group

(control and BPAtreated

animals);

analysis of 50

nucleoids/animal

Plasma concentrations

of 8-

hydroxydeoxyguanosine

(8-OHdG), lipid

peroxidation (MDA) and

glutathione activity

In vivo micronucleus

assay (Table 7)*

chromosomal

aberrations assay

(Table 6)*

Bacterial reverse

mutation assay (Table

1)**

2.4 μg, 10 μg, 5 mg and

50 mg/kg bw per day

administered once a day

for 6 consecutive days

Sampling 24 h after the

last administration of BPA

BPA, 99% purity

(Sigma Chemical Co.)

Positive

 

Dose-related increase

starting from 10 μg/kg bw

per day

Significant increase in

plasma concentration of 8-

OHdG only at 50 mg/kg

bw per day

Dose-related increase of

MDA and decrease of

glutathione in liver

Inconsistent results of 8-

OHdG with comet assay

Reliability: 2

 

Inappropriate

sampling time

Low number of

nucleoids scored

Limited

Tiwari et al.,

20121*,**

Comet assay in bone

marrow, spleen, liver

and kidney and germ

cells

Male Pzh:SFIS mice; 5

animals/group; 100

cells were analysed

0, 5, 10, 20 or 40 mg/kg

bw

Orally in drinking water

Daily for 2 weeks

Animals were sacrificed 24

h after the last treatment

BPA, no information

on purity or the

supplier company

Positive

 

Increases of DNA tail

moment in bone marrow,

spleen, kidney and lung

cells at any dose level

without a clear dose response

No increase of tail moment

was detected in liver cells

In sperm cells increase of

tail moment: at all doses

24 h after the end of

exposure; at the 2 highest

doses 5 weeks after the

end of treatment

Reliability: 3

 

No information on

purity; drinking

water consumption

(containing BPA) not

measured,

inadequate sampling

time, poor study

report; tail moment,

used as only

parameter to report

the results for the

comet assay, is not

recommended by the

Comet International

Committees

Low

Dobrzyńska and

Radzikowska,

20131

Alkaline comet assay in

epididymal sperm of

Holtzman rats

In vivo dominant lethal

mutations in male rats

(Table 9)*

Oral gavage of 10 μg/kg

bw and 5 mg/kg bw BPA

dissolved in ethyl alcohol

and diluted in sesame oil,

for 6 consecutive day

BPA 99% purity

(Sigma Chemical Co.)

Positive

 

Significant increase in the

sperm DNA damage at 5

mg/kg bw

Reliability: 3

 

Comet assay is not

considered

appropriate to

measure DNA strand

breaks in mature

germ cells due to the

high and variable

background levels in

DNA damage in this

cell type (OECD TG

489); moreover, the

sampling time, i.e. 8

weeks after last

treatment, is

inappropriate for in

vivo comet assay

Low

Tiwari and Vanage,

20131*

Comet assay in lung,

spleen, kidneys, liver

and bone marrow of

Pzh:Sfis female mice

No. of animals/group

9 in control, 6 in BPA 5

mg/kg bw, 8 in BPA 10

mg/kg bw; 6 in BPA 20

mg/kg bw

100 nucleoids

scored/animal

In vivo micronucleus

assay (Table 7)*

BPA 5, 10, or 20 mg/kg

bw/day for 2 weeks in

drinking water

Sampling 24 h after the

end of treatment

BPA, no information

on purity or the

supplier company

Positive in lung at 5 and

10 mg/kg

Negative in spleen,

kidneys, liver and bone

marrow

Reliability: 2

 

Inappropriate

sampling time,

tail moment, used as

only parameter to

report the results for

the comet assay, is

not recommended by

the Comet

International

Committees

Low

 

No information on

source and purity

of BPA

Gajowik et al.,

2013*

Alkaline comet assay in

brain cells of KM male

mice; (11

animals/group); 200

cells for each group

analysed

BPA 0.5, 50 and 5000

μg/kg bw (daily dose,

diluted in tea oil, by

gavage) for 8 weeks

After 8 weeks of

exposure, mice were

sacrificed and the

brain samples were

immediately removed

The tail DNA%, tail length

and tail moment were

measured using CASP

comet analysis software

Based on the DNA

percentage of the tail

intensity, the damage

level was divided into 5

grades

Arbitrary units computed

with the score of DNA

damage in analysed cells

were used to express the

DNA damage

BPA from Sigma-

Aldrich (HPLC grade)

purity >97% not

reported in the study

but available on the

website of the

company

Positive

 

Significant increase of

damaged cells from 23.0%

in the control group to

47.3%, 66.6% and 72.5%

in the low-, medium and

high-exposed groups

Severity of DNA damage,

expressed as arbitrary

units (AUs), increased with

AUs of 0.28 in the control

to AUs of 0.59, 0.96 and

1.28 in the low, medium

and highly exposed

groups, respectively

Reliability: 2

 

DNA damage was

evaluated using

arbitrary units and

considering the

distribution of DNA

damage in the cell

population analysed

(n = 440), rather

than using median

animals data as the

statistical unit, as

recommended in

OECD TG 489

Limited

Zhou et al., 2017

Comet assay in liver

female Wistar rats; (7

animals/group)

Serum biochemical

analysis: ALT, ALP, TP,

Alb, GGT, TC,

Triglycerides, HDL; LDL

Hepatic antioxidants

and lipid peroxidation

level: GPx, SOD, MDA

CYPR450 (ELISA)

Histopathology

Immunohistochemical

evaluation of caspase-3

7 animals/group: control

(corn oil)

BPA 10 mg/kg bw; daily

administration via gavage

for 30 days

Sampling at the end of

treatment

BPA (Sigma-Aldrich)

purity >97% not

reported in the study

but available on the

website of the

company

Positive:

increase of tail

DNA %

BPA-induced:

- increase of ALT, ALP,

GGT, TC, LDL, MDA,

caspase-3;

- decrease of Alb, TP, GPx,

SOD, CCYPR450

Histopathological analyses

showed deleterious

hepatic changes ranging

from hepatocytes’

vacuolisation and eccentric

nuclei to focal necrosis and

fibrosis

 

Reliability: 3

 

Use of frozen

tissues; without a

positive control; a

single dose applied;

toxic effects in liver

Low

Abdel-Rahman et

al., 2018

Comet assay in liver of

Sprague Dawley rats of

either sex;

7 animals/group

Serum analysis: ALT,

ALP, AST, bilirubin

Analysis of antioxidant

effects: CAT, POD,

SOD, GSH

Lipid peroxidation

assay, hydrogen

peroxide assay, nitrite

assay

Liver histopathology

BPA, 25 mg/kg by i.p.

negative control group;

vehicle control group

(10% DMSO in olive oil)

Sampling: 4 weeks after

the treatment

BPA, no information

on purity or the

supplier company

Positive

increase of tail DNA %

28.35 ± 1.2 vs 0.01 ±

0.005

BPA-induced:

- increase of WBC, ALT,

AST, ALP, bilirubin, H2O2,

nitrite

- decrease of RBC,

platelets, Hb, albumin,

CAT, POD, SOD, GSH,

‘Histopathological

examination of BPAtreated

animals revealedintense hepatic cytoplasm

inflammation, centrilobular

necrosis, cellular

hypertrophy, fatty

degeneration,

vacuolisation, steatosis

and distortion of portal

vein’

Reliability: 3

 

Limitations:

- a single

administration by i.p.

and comet, analysis

after 4 weeks;

- unusual software

used for the comet

analysis;

- the results reported

using the different

parameters (tail

length, % of DNA in

tail, tail moment) are

not consistent; - the value of % of

DNA in tail in

controls is extremely

low with respect to

the data reported in

the scientific

literature;

- high liver toxicity

Low

 

A single

administration by

i.p.

No information on

source and purity

of BPA

Kazmi et al., 2018

Comet assay in liver of

Male Swiss albino mice

(10 animals/group);

images of 50 randomly

selected nuclei/

experimental group

Analysis of liver toxicity

markers (AST and ALT)

and liver histopathology

BPA dissolved in ethanol

and diluted in corn oil by

gavage at 50 mg/kg bw,

once a day for 28

successive days

BPA (≥ 99 %) Sigma-

Aldrich

Positive

 

Mean tail length, tail

moment and % tail DNA

were significantly

increased (p < 0.05) in

liver of BPA-treated mice

Increase of AST, ALT,

marked histopathological

alteration in liver of BPAtreated

animals

‘congestion of the hepatic

blood vessels as well as

marked vacuolar

degeneration of the

hepatocytes with many

necrotic cells’

Reliability: 3

 

Major deviation from

OECD TG 489:

-too low number of

analysed cells per

animal

-aggregated mean

data analysed

(instead of animal

median)

-no positive control

- too high liver

toxicity associated

with treatment

Low

Elhamalawy et al.,

2018

Alkaline comet assay in

liver, kidney, testes,

urinary bladder, colon

and lungs cells

CD-1 male mice (5

mice/group)

In vitro comet assay

(Table 5)**

Gavage 0, 125, 250 and

500 mg/kg bw BPA

(maximum tolerated dose)

as suspensions in corn oil

prepared by

ultrasonication

2 doses (24 h apart)

Animals were sacrificed 3

h after 2nd dose

200 cells analysed/mice

(100 cells per gel and 2

gels per mouse)

BPA (purity >99%,

Sigma-Aldrich)

Negative

 

None of the tissues

showed an effect of BPA

except in testicular

cells, in which an

increased level of DNA

strand breaks (p < 0.01

compared with control

group) was observed at

the lowest dose only

Reliability: 1

 

This study basically

followed the OECD

TG 489

High

Sharma et al.,

2018**

Comet assay in liver

and testes of male

Swiss albino mice

Male Swiss albino mice,

10 animals/group

50 nuclei/group were

analysed

In vivo micronucleus

assay (Table 7)*

50 mg/kg bw, orally once

a day for 28 days

Sampling at the end of

treatment

BPA, purity ≥ 99%

(Sigma-Aldrich)

Positive

 

Increase (p ≤ 0.05) in the

mean values of tail length,

percentage of tail DNA and

Olive tail moment in liver

and testes

Histopathological

examination hepatocyte

vacuolar degeneration with

many necrotic cells

Defective spermatogenesis

characterised by severe

necrosis and loss of the

spermatogonial layers with

multiple spermatid giant

cells formation in most of

the seminiferous tubules

and a congestion of the

interstitial blood vessels

Reliability: 3

 

No positive control,

low number of

nucleoids analysed,

toxic effects

observed in liver and

testes, a single dose

applied

The standard alkaline

comet assay applied

is not considered

appropriate to

measure DNA strand

breaks in mature

germ cells

Low

Fawzy et al., 2018*

Comet assay in heart of

Wistar rats; 20

animals/group

BPA dissolved in corn oil

30 mg/kg bw per day

injected subcutaneously

(SC) 6 days/week for 4

weeks

Sacrifice at the end of

treatment

BPA Sigma-Aldrich;

purity >97% not

reported in the study

but available on the

website of the

company

Positive

 

Increase tail DNA % (6.88

vs 1.67)

Histopathological changes:

focal disruption of

cardiomyocytes with some

nuclear changes, such as

karyolysis and pyknosis

and sarcoplasmic

vacuolisation

The mitochondria

appeared swollen and

deranged with different

sizes and shapes

Reliability: 3

 

Single dose; no

positive control;

inadequate cell

preparation for

comet assay; high

toxicity

Low

 

route of

administration:

subcutaneous

Amin et al., 2019

Comet assay in testes

of Sprague Dawley

rats; 7 rats/group

Histopathology

Antioxidant enzymes:

CAT, SOD, GSH, POD,

NO

BPA (50 mg/kg bw)

injected intraperitoneal on

alternate days for 21 days

Sacrifice 24 h after the

end of treatment

BPA analytical grade

(Merck KGaA); purity

>97% not reported in

the study but

available on the

website of the

company

Positive

 

Histopathology: ‘BPA

caused significant damage

and abrasions to

seminiferous tubules with

low cellular density’

BPA-induced:

- decrease of body weight,

epididymis and testes

weight, testosterone, FSH,

LH, CAT, SOD, GSH, POD;

- decrease of sperm count,

viability, motility

- increase of estradiol

Reliability: 3

 

Single dose; no

positive controls; an

unusual software for

the comet analysis

used; the comet

presented in the

microphotographs

are of low quality

The standard alkaline

comet assay applied

is not considered

appropriate to

measure DNA strand

breaks in mature

germ cells

Low

 

BPA was

administered by

i.p.

Majid et al., 2019

Comet assay (neutral)

on spermatozoa of

Sprague Dawley rats

(7 per group)

100 scored cells per

animal

In vitro comet assay

(Table 5)**

Animals treated by

gavage with 5, 25 and 50

mg BPA/kg bw per day for

28 days and sacrificed on

day 29th, control received

the vehicle alone (0.1%

ethanol)

BPA (99% purity)

from Santa Cruz

Biotechnology

Positive

 

Both tail moment and %

tail DNA were significantly

(p < 0.05) increased in the

BPA 50 mg/kg bw per day

group compared to vehicle

controls, while no

significant difference with

controls was observed in

the BPA 5 and 25 mg/kg

bw per day groups

Reliability: 3

 

The study was

performed following

a non-standard,

neutral protocol and

unusual evaluation

of comets based

on the analysis of

microphotographs

No detailed

information on data

analysis is provided

(e.g. the use of

median vs mean as

individual animal

descriptor)

No positive control

Low

Ullah et al., 2019**

Comet assay in testes

of offspring of BPA treated

mice (pregnant Kumming mice, 20 in

each group)

Animals were randomly

divided into 7 groups. One

group served as controlthe others received BPA in

drinking water at 0.05,

0.5, 5, 10, 20 or 50 mg/kg

bw per day, for 40 days

from gestation day 0 to

lactation day 21. F1 male

mice were sacrificed at

weaning (post-natal day

21) and DNA damage in

testes evaluated by comet

assay

BPA (purity 99%,

Sigma)

Positive

 

The results obtained

showed significantly

increased Olive tail

moment (OTM) in testes

cells of F1 animals treated

with 5, 10, 20 and 50

mg/kg bw per day,

compared with the control

group (p < 0.05).

Reliability: 3

 

The results obtained

showed significantly

increased Olive tail

moment (OTM) in testes

cells of F1 animals treated

with 5, 10, 20 and 50

mg/kg bw per day,

compared with the control

group (p < 0.05).

Low

Zhang et al., 2019

Alkaline comet assay in

thyroid tissue

Male albino rats

20 rats/group

Biochemical

investigation of MPO

activity, GSH, SOD

activity and MDA

BPA dissolved in corn oil

200 mg/kg bw per day

(1/20 of the oral LD50) for

35 days

Sacrifice 24 h after the

last administration

BPA (99.5% purity)

was obtained from

Sigma-Aldrich Co.

Positive

 

% tail DNA 4 times

increase compared with

control level

The histopathological

examinations of thyroid

gland showed severe

congestion of interstitial

blood capillaries, severe

lymphocytic infiltration

associated with variablesized

follicles, most of

which contain scanty

colloid secretion, and

some are atrophied in BPA

group

Significant induction of

MPO activity and MDA

concentration associated

with significant decreases

of SOD activity and GSH

concentration in the

thyroid gland of BPA group

Reliability: 3

 

Only one dose level

No positive control

Comet method

poorly described

The

microphotographs of

comets are of low

quality

High toxicity

Low

Mohammed et al.,

2020

Alkaline comet assay in

testes

Male juvenile Sprague

Dawley (SD) rats (7

animals/group)

Sperm DNA damage

was evaluated by the

comet and Halo assays

using duplicate slides;

apoptosis in testes cells

was quantified using

TUNEL assay, and

testicular levels of 8-

OHdG were determined

by

immunohistochemistry

Gavage 8 weeks

BPA (100 mg/kg bw per

day) daily/5 days per

week by gavage for 8

consecutive weeks

Animals were sacrificed

after 8 weeks

BPA (Sigma-Aldrich)

Purity >99% not

reported in the study

but available on the

website of the

company

Negative

All comet assay

parameters (tail length,

Olive tail moment and %

DNA in the tail) and the

nuclear diffusion factor in

Halo assay, were slightly

but not significantly

increased in testes cells of

BPA-treated rats compared

with controls

TUNEL-positive cells and

per cent of 8-OHdG

positive areas in testicular

tissue were also slightly

but non-significantly

increased in BPA-treated

rats

Reliability: 3

The standard alkaline

comet assay applied

(OECD TG 489) is

not considered

appropriate to

measure DNA strand

breaks in mature

germ cells

Other test methods

(Halo and

immunohistochemical

determination of 8-

OHdG) are not

standardised and/or

validated for

regulatory use

For all end-points,

only a single dose

was tested

Sampling time not

specified

No positive control

Low

Sahu et al., 2020

Comet assay on whole

brain cells from KM

mice of F1 and F2 (8

male and 8 female)

Pregnant mice (F0) were

orally dosed with BPA

dissolved in tea oil at 0.5,

50, 5000 μg/kg bw per

day from gestational day

1 until weaning (post-natal day 21). Then, the

first generation (F1) of

mice were used to

generate the F2

DNA damage in brain cells

was evaluated by comet

assay in mice from both

F1 and F2

BPA (purity: 98 %)

Sigma-Aldrich

Equivocal

DNA damage, expressed

as arbitrary units, was

slightly (less than twofold)

increased in the F1male mice at the lowest

dose and in females at the

intermediate dose. No

effect of BPA exposure

was observed in the F2

mice

Reliability: 3

The study protocol is

only shortly

described

 

The presentation and

interpretation of the

results is inadequate

No positive control

Low

Zhang et al., 2020

Comet assay in blood

liver and kidney

Male Wistar rats

(WNIN)

6 animals/group

50 nuclei/slides were

scored

Lipid peroxidation:

serum level of

malondialdehyde (MDA)

8-Hydroxy-2-

deoxyguanosine (8-

OHdG) in urine

collected 24 h before

the sacrifice

In vivo micronucleus

assay (Table 7)*

0, 50, and 100 μg/kg, per

oral (gavage) for a period

of 4 weeks

Sampling at the end of

treatment

BPA, (Sigma-Aldrich)

purity >97% not

reported in the study

but available on the

website of the

company

Positive

 

A weak but statistically

significant and doserelated

increase of tail

length in liver

In kidney increase of DNA

damage observed only at

the dose of 50 μg/kg

Comet parameters are not

reported for blood cells

Dose-related increase of

MDA in serum and of 8-

OHdG levels in urine

Reliability: 2

 

Low number of

nucleoids analysed

No positive controls

Limited

Panpatil et al.,

2020*

Evaluation of sperm

DNA damage by

alkaline comet and DNA

ladder assays

Male Sprague Dawley

rats (groups of 7

animals) ROS, Catalase, POD

and SOD, GSH, Lipid

peroxidation, TBARS,

hydrogen peroxide,

nitrite assay, AOPP

BPA diluted in 10% DMSO

was injected

intraperitoneally at 25

mg/kg bw on alternate

days for 30 days

BPA, no information

on purity or the

supplier company

Positive

 

Significant (p < 0.01)

increase of all comet

parameters in BPA-treated

animals compared with

vehicle controls

Electrophoresis on agarose

gel showed extensive DNA fragmentation in testes of

BPA-treated rats

Significant increase in ROS

level and decreased levels

of CAT, GSH SOD and POD

in the testis of BPA-treated

group

Reliability: 3

 

The standard alkaline

comet assay applied

(OECD TG 489) is

not considered

appropriate to

measure DNA strand

breaks in mature

germ cells

 

The comet protocol

is shortly described,

with no information

on the number of

analysed sperm cells

per animal; sampling

time not specified;

cytotoxicity not

evaluated; no

positive control

The DNA ladder

assay is a

biochemical method

not validated for

genotoxicity

assessment

Low

 

For insufficient

reliability and lack

of information on

test item purity

Zahra et al., 2020

Source:  Re-evaluation of the risks to public health related to the presence of bisphenol A (BPA) in foodstuffs, EFSA, (2021).

In vivo dominant lethal assay

Table 9: In vivo dominant lethal assay (OECD TG 478 was considered for the evaluation of the reliability).

Test system/Test

object

Exposure

conditions

(concentration/

duration/metabolic activation)

Information on the

characteristics of

the test substance

Results

Reliability/

Comments

Relevance of the

result

Reference

Dominant lethal test

with male Holtzman

rats (7 per group)

Each treated male was

mated with 2 females

per week over a

period of 8 weeks; the

mated females were

sacrificed on 15th day

of gestation and

uterine content

examined

In vivo comet assay in

rat epididymal sperm

(Table 8)*

Rats treated by oral

gavage with BPA

dissolved in ethyl

alcohol and diluted in

sesame oil, at dose

levels of 10 μg/kg bw

and 5 mg/kg bw once

a day for 6

consecutive days

Negative controls were

treated with vehicle

BPA 99% purity

(Sigma Chemical Co.)

Positive

 

Significant decrease in

total implants/female

and live

implants/female, in

females mated with

males treated with 5.0

mg BPA/kg bw the

fourth week and sixth

week after treatment

Reliability: 2

 

No positive control

No negative historical

control

Limited study design,

with less analysable

total implants and

resorptions than

recommended (OECD

TG 478)

Limited

Tiwari and Vanage,

20131*

Source:  Re-evaluation of the risks to public health related to the presence of bisphenol A (BPA) in foodstuffs, EFSA, (2021).

Re-evaluation of the risks to public health related to the presence of bisphenol A (BPA) in foodstuffs - Genotoxicity

Genotoxicity Annex A - references and abbreviations

References

Abdel-Rahman HG, Abdelrazek HMA, Zeidan DW, Mohamed RM and Abdelazim AM, 2018. Lycopene: hepatoprotective and antioxidant effects toward bisphenol A-induced toxicity in female Wistar rats. Oxidative Medicine and Cellular Longevity, 2018.

Aghajanpour-Mir SM, Zabihi E, Akhavan-Niaki H, Keyhani E, Bagherizadeh I, Biglari S and Behjati F, 2016. The genotoxic and cytotoxic effects of bisphenol-A (BPA) in MCF-7 cell line and amniocytes. International Journal of Molecular and Cellular Medicine, 5(1), 19–29.

Amin DM, 2019. Role of copeptin as a novel biomarker of bisphenol A toxic effects on cardiac tissues: Biochemical, histological, immunohistological, and genotoxic study. Environmental Science and Pollution Research International, 26(35), 36037–36047.

Balabanič D, Filipič M, Krivograd Klemenčič A and Žegura B, 2021. Genotoxic activity of endocrine disrupting compounds commonly present in paper mill effluents. Science of the Total Environment, 794, 148489.

Chen ZY, Liu C, Lu YH, Yang LL, Li M, He MD, Chen CH, Zhang L, Yu ZP and Zhou Z, 2016. Cadmium exposure enhances bisphenol A-induced genotoxicity through 8-oxoguanine-DNA glycosylase-1 OGG1 inhibition in NIH3T3 fibroblast cells. Cellular Physiology and Biochemistry, 39(3), 961–974.

De Flora S, Micale RT, La Maestra S, Izzotti A, D’Agostini F, Camoirano A, Davoli SA, Troglio MG, Rizzi F, Davalli P and Bettuzzi S, 2011. Upregulation of clusterin in prostate and DNA damage in spermatozoa from bisphenol A-treated rats and formation of DNA adducts in cultured human prostatic cells. Toxicological Sciences, 122(1), 45–51.

Di Pietro P, D’Auria R, Viggiano A, Ciaglia E, Meccariello R, Russo RD, Puca AA, Vecchione C, Nori SL and Santoro A, 2020. Bisphenol A induces DNA damage in cells exerting immune surveillance functions at peripheral and central level. Chemosphere, 254, 126819.

Dobrzyńska MM and Radzikowska J, 2013. Genotoxicity and reproductive toxicity of bisphenol A and Xray/ bisphenol A combination in male mice. Drug and Chemical Toxicology, 36(1), 19–26.

Durovcova I, Spackova J, Puskar M, Galova E and Sevcovicova A, 2018. Bisphenol A as an environmental pollutant with dual genotoxic and DNA-protective effects. Neuro Endocrinology Letters, 39(4), 294– 298.

EFSA CEF Panel, 2015. (EFSA Panel on Food Contact Materials, Enzymes, Flavourings and Processing Aids), 2015. Scientific opinion on the risks to public health related to the presence of bisphenol A (BPA) in foodstuffs. EFSA Journal 2015;13(1):3978.

Elhamalawy OH, Eissa FI, El Makawy AI and El-Bamby MM, 2018. Bisphenol-A hepatotoxicity and the protective role of sesame oil in male mice. Jordan Journal of Biological Sciences, 11(4), 461–467.

Fawzy EI, El Makawy AI, El-Bamby MM and Elhamalawy HO, 2018. Improved effect of pumpkin seed oil against the bisphenol-A adverse effects in male mice. Toxicology Reports, 5, 857–863.

Fic A, Žegura B, Sollner Dolenc M, Filipič M and Peterlin Mašič L, 2013. Mutagenicity and DNA damage of bisphenol A and its structural analogues in HepG2 cells. Arhiv za Higijenu Rada i Toksikologiju, 64(2), 189–200.

Gajowik A, Radzikowska J and Dobrzyńska MM, 2013. Genotoxic effects of bisphenol A on somatic cells of female mice, alone and in combination with X-rays. Mutation Research. Genetic Toxicology and Environmental Mutagenesis, 757(2), 120–124.

George VC and Rupasinghe HPV, 2018. DNA damaging and apoptotic potentials of bisphenol A and bisphenol S in human bronchial epithelial cells. Environmental Toxicology and Pharmacology, 60, 52–57.

Gonçalves GD, Semprebon SC, Biazi BI, Mantovani MS and Fernandes GSA, 2018. Bisphenol A reduces testosterone production in TM3 Leydig cells independently of its effects on cell death and mitochondrial membrane potential. Reproductive Toxicology, 76, 26–34.

Hu X, Biswas A, Sharma A, Sarkodie H, Tran I, Pal I and De S, 2021. Mutational signatures associated with exposure to carcinogenic microplastic compounds bisphenol A and styrene oxide. NAR Cancer, 3(1), zcab004. doi:10.1093/narcan/zcab004 [RefID 295-G].

Huang FM, Chang YC, Lee SS, Ho YC, Yang ML, Lin HW and Kuan YH, 2018. Bisphenol A exhibits cytotoxic or genotoxic potential via oxidative stress-associated mitochondrial apoptotic pathway in murine macrophages. Food and Chemical Toxicology, 215–224.

Iso T, Watanabe T, Iwamoto T, Shimamoto A and Furuichi Y, 2006. DNA damage caused by bisphenol A and oestradiol through estrogenic activity. Biological and Pharmaceutical Bulletin, 29(2), 206–210.

Johnson GE and Parry EM, 2008. Mechanistic investigations of low dose exposures to the genotoxic compounds bisphenol-A and rotenone. Mutation Research, 651(1–2), 56–63.

Kazmi STB, Majid M, Maryam S, Rahat A, Ahmed M, Khan MR and Haq IU, 2018. BPA induced hepatotoxicity in Sprague Dawley rats. Biomedicine and Pharmacotherapy, 102, 728–738.

Kose O, Rachidi W, Beal D, Erkekoglu P, Fayyad-Kazan H and Kocer Gumusel B, 2020. The effects of different bisphenol derivatives on oxidative stress, DNA damage and DNA repair in RWPE-1 cells: A comparative study. Journal of Applied Toxicology, 40(5), 643–654.

Lei BL, Xu J, Peng W, Wen Y, Zeng XY, Yu ZQ, Wang YP and Chen T, 2017. In vitro profiling of toxicity and endocrine disrupting effects of bisphenol analogues by employing MCF-7 cells and two-hybrid yeast bioassay. Environmental Toxicology, 32(1), 278–289.

Li XH, Yin PH and Zhao L, 2017. Effects of individual and combined toxicity of bisphenol A, dibutyl phthalate and cadmium on oxidative stress and genotoxicity in HepG 2 cells. Food and Chemical Toxicology, 105, 73–81.

Majid M, Ijaz F, Baig MW, Nasir B, Khan MR and Haq IU, 2019. Scientific validation of ethnomedicinal.

use of Ipomoea batatas L. Lam. as aphrodisiac and gonadoprotective agent against bisphenol A induced testicular toxicity in male Sprague Dawley rats. BioMed Research International, 2019, 8939854.

Masuda S, Terashima Y, Sano A, Kuruto R, Sugiyama Y, Shimoi K, Tanji K, Yoshioka H, Terao Y and Kinae N, 2005. Changes in the mutagenic and estrogenic activities of bisphenol A upon treatment with nitrite. Mutation Research, 585(1–2), 137–146.

Mohammed ET, Hashem KS, Ahmed AE, Aly MT, Aleya L and Abdel-Daim MM, 2020. Ginger extract ameliorates bisphenol A (BPA)-induced disruption in thyroid hormones synthesis and metabolism: Involvement of Nrf-2/HO-1 pathway. Science of the Total Environment, 703, 134664.

Mokra K, Kuźmińska-Surowaniec A, Woźniak K and Michałowicz J, 2017. Evaluation of DNA-damaging potential of bisphenol A and its selected analogs in human peripheral blood mononuclear cells (in vitro study). Food and Chemical Toxicology, 100, 62–69.

Mokra K, Woźniak K, Bukowska B, Sicińska P and Michałowicz J, 2018. Low-concentration exposure to BPA, BPF and BPAF induces oxidative DNA bases lesions in human peripheral blood mononuclear cells. Chemosphere, 201, 119–126.

Naik P and Vijayalaxmi KK, 2009. Cytogenetic evaluation for genotoxicity of bisphenol-A in bone marrow cells of Swiss albino mice. Mutation Research, 676(1–2), 106–112.

Özgür M, Gül Yılmaz ŞG, Uçar A and Yılmaz S, 2021. Cytotoxic effects of bisphenol A as an endocrine disruptor on human lymphocytes. Iranian Journal of Toxicology, 15(2), 115–120.

Pacchierotti F, Ranaldi R, Eichenlaub-Ritter U, Attia S and Adler ID, 2008. Evaluation of aneugenic effects of bisphenol A in somatic and germ cells of the mouse. Mutation Research, 651(1–2), 64–70.

Panpatil VV, Kumari D, Chatterjee A, Kumar S, Bhaskar V, Polasa K and Ghosh S, 2020. Protective effect of turmeric against bisphenol-A induced genotoxicity in rats. Journal of Nutritional Science and Vitaminology, 66(Supplement), S336–S342.

Porreca I, Ulloa Severino L, D’Angelo F, Cuomo D, Ceccarelli M, Altucci L, Amendola E, Nebbioso A, Mallardo M, De Felice M and Ambrosino C, 2016. “Stockpile” of slight transcriptomic changes determines the indirect genotoxicity of low-dose BPA in thyroid cells. PLoS ONE, 11(3), e0151618.

Ramos C, Ladeira C, Zeferino S, Dias A, Faria I, Cristovam E, Gomes M and Ribeiro E, 2019. Cytotoxic and genotoxic effects of environmental relevant concentrations of bisphenol A and interactions withdoxorubicin. Mutation Research. Genetic Toxicology and Environmental Mutagenesis, 838, 28–36.

Ribeiro-Varandas E, Viegas W, Sofia Pereira HS and Delgado M, 2013. Bisphenol A at concentrations found in human serum induces aneugenic effects in endothelial cells. Mutation Research, 751(1), 27–33.

Sahu C, Charaya A, Singla S, Dwivedi DK and Jena G, 2020. Zinc deficient diet increases the toxicity of bisphenol A in rat testis. Journal of Biochemical and Molecular Toxicology, 34(10), e22549.

Santovito A, Cannarsa E, Schleicherova D and Cervella P, 2018. Clastogenic effects of bisphenol A on human cultured lymphocytes. Human and Experimental Toxicology, 37(1), 69–77.

Sharma AK, Boberg J and Dybdahl M, 2018. DNA damage in mouse organs and in human sperm cells by bisphenol A. Toxicological and Environmental Chemistry, 100(4), 465–478.

Sonavane M, Sykora P, Andrews JF, Sobol RW and Gassman NR, 2018. Camptothecin efficacy to poison top1 is altered by bisphenol A in mouse embryonic fibroblasts. Chemical Research in Toxicology, 31(6), 510–519.

Srivastava S and Gupta P, 2016. Genotoxic and infertility effects of bisphenol A on Wistar albino rats. International Journal of Pharmaceutical Sciences Review and Research, 41(1), 126–131.

Šutiaková I, Kovalkovičová N and Šutiak V, 2014. Micronucleus assay in bovine lymphocytes after exposure to bisphenol A in vitro. In Vitro Cellular and Developmental Biology. Animal, 50(6), 502– 506.

Tayama S, Nakagawa Y and Tayama K, 2008. Genotoxic effects of environmental estrogen-like compounds in CHO-K1 cells. Mutation Research, 649(1–2), 114–125.

Tiwari D and Vanage G, 2013. Mutagenic effect of bisphenol A on adult rat male germ cells and their fertility. Reproductive Toxicology, 40, 60–68.

Tiwari D, Kamble J, Chilgunde S, Patil P, Maru G, Kawle D, Bhartiya U, Joseph L and Vanage G, 2012. Clastogenic and mutagenic effects of bisphenol A: An endocrine disruptor. Mutation Research, 743(1–2), 83–90.

Ullah A, Pirzada M, Jahan S, Ullah H and Khan MJ, 2019. Bisphenol A analogues bisphenol B, bisphenol F, and bisphenol S induce oxidative stress, disrupt daily sperm production, and damage DNA in rat spermatozoa: A comparative in vitro and in vivo study. Toxicology and Industrial Health, 35(4), 294– 303.

Ulutaş OK, Yıldız N, Durmaz E, Ahbab MA, Barlas N and Çok İ, 2011. An in vivo assessment of the genotoxic potential of bisphenol A and 4-tert-octylphenol in rats. Archives of Toxicology, 85(8), 995– 1001.

Xin F, Jiang LP, Liu XF, Geng CY, Wang WB, Zhong LF, Yang G and Chen M, 2014. Bisphenol A induces oxidative stress-associated DNA damage in INS-1 cells. Mutation Research. Genetic Toxicology and Environmental Mutagenesis, 769, 29–33.

Xin LL, Lin Y, Wang AQ, Zhu W, Liang Y, Su XJ, Hong CJ, Wan JM, Wang YR and Tian HL, 2015. Cytogenetic evaluation for the genotoxicity of bisphenol-A in Chinese hamster ovary cells. Environmental Toxicology and Pharmacology, 40(2), 524–529.

Yu H, Chen Z, Hu K, Yang Z, Song M, Li Z and Liu Y, 2020. Potent clastogenicity of bisphenol compounds in mammalian cells – human CYP1A1 being a major activating enzyme. Environmental Science and Technology, 54(23), 15267–15276.

Yuan J, Kong Y, Ommati MM, Tang Z, Li H, Li L, Zhao C, Shi Z and Wang J, 2019. Bisphenol A-induced apoptosis, oxidative stress and DNA damage in cultured rhesus monkey embryo renal epithelial Marc- 145 cells. Chemosphere, 234, 682–689.

Zahra Z, Khan MR, Majid M, Maryam S and Sajid M, 2020. Gonadoprotective ability of Vincetoxicum arnottianum extract against bisphenol A-induced testicular toxicity and hormonal imbalance in male Sprague Dawley rats. Andrologia, 52(6), e13590.

Zemheri F and Uguz C, 2016. Determining mutagenic effect of nonylphenol and bisphenol A by using Ames/Salmonella/microsome test. Journal of Applied Biological Sciences, 10(3), 9–12.

Zhang S, Bao J, Gong X, Shi W and Zhong X, 2019. Hazards of bisphenol A—blocks RNA splicing leading to abnormal testicular development in offspring male mice. Chemosphere, 230, 432–439.

Zhang H, Wang Z, Meng L, Kuang H, Liu J, Lv X, Pang Q and Fan R, 2020. Maternal exposure to environmental bisphenol A impairs the neurons in hippocampus across generations. Toxicology, 432, 152393.

Zhou YX, Wang ZY, Xia MH, Zhuang SY, Gong XB, Pan JW, Li CH, Fan RF, Pang QH and Lu SY, 2017. Neurotoxicity of low bisphenol A (BPA) exposure for young male mice: implications for children exposed to environmental levels of BPA. Environmental Pollution, 229, 40–48.

Abbreviations

7-HF

7-hydroxyflavone

8-OHdG

8-hydroxy-2′-deoxyguanosine

ABT   

1-Aminobenzotriazole

ALP

Alkaline phosphatase

ALT

Alanine aminotransferase

AOPP

Advanced oxidation protein products

ATM

Ataxia-telangiectasia mutated

AU

Arbitrary units

BPA

Bisphenol A

BrdU

5-bromo-2-deoxyuridine

Bw

Body weight

CA

Chromosomal aberrations

CAT

Catalase

ChE

Cholinesterase

CHO cells

Chinese hamster ovary cells

CI

Cellular index

CYPR450

Cytochrome P450 reductase

DCF

Dichlorofluorescein

DMSO

Dimethylsulphoxide

EM

Electron microscopy

ER

Oestrogen receptor

FACS

Fluorescence activated cell sorting

FISH

Fluorescence in situ hybridisation

Fpg

Formamide pyrimidine glycosylase

GGT

Gamma glutamyl transferase

GPx

Glutathione peroxidase

GR

Glutathione reductase

GSH

Reduced glutathione

Hb

Haemoglobin

HDL

High-density lipoprotein cholesterol

HPLC

High performance liquid chromatography

HUVEC

Human umbilical vascular endothelial cells

i.p.

Intraperitoneal

KET

Ketoconazole

LDH

Lactate dehydrogenase

LDL

Low density lipoprotein cholesterol

MDA

Malondialdehyde

MMC

Mitomycin

MMS

Methyl methane sulfonate

MPO

Myeloperoxidase

NAC

N-Acetyl-L-cysteine

NO

Nitric oxide

OTM

Olive tail moment

PBMC

Human peripheral blood mononuclear cells

PCE

Polychromatic erythrocytes

PCP

Pentachlorophenol

PHA

Phytohemagglutinin

POD

Peroxidase

RBC

Red blood cells

ROS

Reactive oxygen species

RT-PCR

Real time polymerase chain reaction

SCE

Sister chromatid exchange

SD

Sprague Dawley

SOD

Superoxide dismutase

TBARS

Thiobarbituric acid reactive substances

TC

Total cholesterol

TG

Test guideline

TP

Total protein

TUNEL

Terminal deoxynucleotidyl transferase dUTP nick end labelling

WBC

White blood cells

WGS

Whole genome sequencing