Food and Life
Korean Society for Food Science of Animal Resources
Research Article

Assessing the genotoxicity of oral zinc oxide nanoparticle administration in male rats using micronuclei and comet assay

Amal G. Ramadan1https://orcid.org/0000-0002-7122-4074, Ahmed A. M. Yassein1https://orcid.org/0000-0002-1592-1334, Eissa A. Eissa1https://orcid.org/0000-0002-3730-0627, Gamal M. Hassan1,*https://orcid.org/0000-0003-2575-6886
1Department of Genetics, Faculty of Agriculture, Fayoum University, Fayoum 63514, Egypt
*Corresponding author : Gamal M. Hassan. Department of Genetics, Faculty of Agriculture, Fayoum University, Fayoum 63514, Egypt. Tel: +20-1002332803, E-mail: gmh01@fayoum.edu.eg

© Copyright 2022 Korean Society for Food Science of Animal Resources. This is an Open-Access article distributed under the terms of the Creative Commons Attribution Non-Commercial License (http://creativecommons.org/licenses/by-nc/4.0/) which permits unrestricted non-commercial use, distribution, and reproduction in any medium, provided the original work is properly cited.

Received: Aug 21, 2022; Revised: Sep 14, 2022; Accepted: Sep 20, 2022

Published Online: Nov 30, 2022

Abstract

Zinc oxide nanoparticles (ZnO-NPs) are regularly utilized in the food and fertilizers industries. In our investigation, rats received oral administration of ZnO NPs with a particle size of 30±5 nm once daily at doses of 100, 200, 300, 400, and 600 mg/kg for ten weeks in order to assess the genotoxic effect. Impacts on hematological markers, genotoxic impact, and growth were investigated. The findings showed that ZnO-NPs significantly reduced body weight gain, red blood cell count (RBC), hemoglobin concentration (Hb), hematocrit value (HCT), and platelet count (PLT), while increasing white blood cell (WBC), mean capsular volume (MCV), mean capsular hemoglobin (MCH), and mean capsular hemoglobin concentration (MCHC) in the treated rats. Our results for the comet assay and micronuclei test show a dosage-dependent increase in DNA fragmentation, which was supported by an increase in the percentage of DNA that is tailed, the length and intensity of DNA tails, and the tail moment, especially at the dose of 600 mg/kg. According to the findings, the frequency of micronucleated cells has increased.

Keywords: rats; zinc oxide nanoparticle; hematology; micronuclei; comet assay

Introduction

Small substances with at least one dimension between one and one hundred nanometers are known as nanoparticles (NPs). Due to their tiny size and large surface area, nanoparticles play a key role in all aspects of modern life (Zayed and Luaibi, 2018). By shrinking matter to a scale of 1 to 100 nm, nanotechnology is a significant area of innovation in the industry (Minetto et al., 2016). The food sector, drug delivery, diagnosis, cosmetics, and several sunscreens are just a few industries where nanoparticles (NPs) are used (Al-Suhaibani and El-Morshedi, 2014). ZnO nanoparticles are exposed to the human body more and more as their use increases. Inhalation, cutaneous contact, and ingestion are the three main ways that ZnO nanoparticles are exposed to the body (Iavicoli et al., 2017). ZnO nanoparticles can enter the circulatory system through a variety of routes after exposure, and through it, they can spread throughout the entire body (Yeh et al., 2012). When ZnO nanoparticles enter the body, they are of a nanosize that quickly allows them to penetrate cells, where they are internalized as either free Zn2+ ions or nanoparticles by the cells (Liu et al., 2017).

Micronuclei have been scored extensively to discover potential genotoxic substances since they are good indicators of genotoxic exposure in both humans and animals (Terradas et al., 2010). A crucial in vivo cytogenetic screening method for identifying newly produced structural chromosomal damage in bone marrow cells is the micronucleus test (Schmid, 1975). Chromosome abnormalities in rat bone marrow and the micronucleus test have both been used extensively to clarify the connection between food and mutagenesis. Micronuclei can be utilized as a mutation index because they are a sign of permanent DNA loss (McKelvey et al., 1993). At a dose of 2,000 mg/kg, Srivastav et al. (2016) found that Wistar rats had decreased red cell counts, liver lesions, and hepatocyte inflammation. According to Chupani et al. (2017), diet-borne ZnO NPs can modify the molecular structure and morphology of the blood, intestine, liver, and kidneys. These NPs may display unpredictable genotoxic features through direct interaction with genetic material or by indirect DNA damage brought on by reactive oxygen species due to their tiny size and increased surface area combined with physiochemical properties like charged surfaces (Kisin et al., 2007). Chromosome abnormalities in rat bone marrow and the micronucleus test have both been used extensively to clarify the connection between food and mutagenesis. Micronuclei can be utilized as a mutation index because they are a sign of permanent DNA loss (McKelvey et al., 1993). Evaluation of nanoparticle toxicity is necessary in order to understand the genotoxic potential of ZnO NPs in an animal model due to the rapid development of nanotechnology and rising exposure to nanoparticles. The current study was carried out to investigate the acute oral toxicity of ZnO-NPs at doses 100, 200, 300, 400, and 600 mg/kg. Hematology assays and the genototoxic effects of ZnO NPs using micronucleus and comet assay.

Materials and Methods

Chemicals

Zinc oxide nanoparticles (ZnO-NPs) were obtained from NanoTech Company, Egypt. According to the information provided by manufacturer the particles size of ZnO nanoparticles is 30±5 nm. Chemicals used for the quantitative determination of various biochemical and hematological parameters were purchased from Bio Diagnostic company and Human company (Egypt).

Preparation of ZnONPs suspension

The ZnO NPs participles (30±5 nm) were dispersed in distilled water (10 mg/mL) and the suspension was sonicated at 230 V for 20 minutes using ultrasonic cleaner sonicator (Branson ultrasonic, Danbury, CT, USA) at room temperature. The suspension was stirred on vortex agitator immediately before administration in different dosages (100, 200, 300, 400, and 600 mg/kg).

Characterization of zinc oxide nanoparticles

After vigorous sonication, the solution distribution of the nanoparticles was dropped onto a copper grid that had been coated with carbon to examine the diameter and shapes of the particles. The grid was then observed using a JEOL JEM 1010 Transmission Electron Microscope after being air - dried at room temperature. Using a transmission electron microscope (TEM) with a 200 kV accelerating voltage, the morphological structure of zinc oxide nanoparticles (ZnO-NPs) was analyzed. The crystal structures measured 30±5 nm and closely resemble a sphere (Fig. 1).

fl-2022-3-79-g1
Fig. 1. Electron micrograph of spherical ZnO-NPs nanoparticles with a size of less than 30 nm.
Download Original Figure
Animals and their housing

Thirty male Sprague-Dawley rats, 8–10 weeks old and weighing 140–160 g, were purchased from Rapitco Farm Company in Giza, Egypt. Five animals per cage were kept in standard plastic cages under controlled environmental conditions at a temperature of 25±2°C with 12-hour cycles of light and darkness.

Experimental design

Six groups of thirty rats were created after the adaptation phase (Five rats per group). Rats in Group 1 (G1), which was designated as the control group, consumed a regular synthetic meal and had unlimited access to water, while those in the other five groups received oral gavage administration of zinc oxide nanoparticles at varying concentrations during a 10-week period. The rats in group 2 (G2) were received 100 mg/kg ZnO NPs suspension orally once daily, Group 3 (G3) 200 mg/kg ZnO-NPs suspension orally once daily, Group 4 (G4) 300 mg/kg ZnONPs suspension orally once daily, Group 5 (G5) 400 mg/kg ZnONPs suspension orally once daily, and Group 6 (G6) 600 mg/kg ZnONPs suspension orally once daily.

Hematological examination

For the hematological study, blood samples were taken from the retinal vein of each rat in each group. A Clindiag Hematology Analyzer (HA-22/Vet, Ninove, Belgium) was used to obtain a complete blood picture for each group. Red blood cell count (RBC), hematocrit (HCT), hemoglobin (Hb) concentration, and red cell indices were included in the analysis. Also included were mean corpuscular volume (MCV), mean corpuscular hemoglobin (MCH), mean corpuscular hemoglobin concentration (MCHC), white blood cell count (WBC), and their differential (lymphocytes, granulocytes, and monocytes), as well as platelet count (PLT).

Genotoxicity assay
Micronuclei assay

According to the method published by Schmid (1975), the micronuclei (MN) test and the scoring of micronucleated polychromatic erythrocytes (MnPCEs) were performed. The ratio of polychromatic erythrocytes (PCEs) / normochromatic erythrocytes (NCEs) was calculated using a minimum of 2000 PCEs per animal (NCEs). To estimate the MnPCEs, the coded slides were examined under 100 oil immersions.

Comet assay

The single cell electrophoresis (comet assay) which developed by Singh et al. (1988) was used in cytogenetic assays. It was combined with the simplicity of biochemical methods for identifying DNA single strand breaks (frank strand breaks and incomplete excision repair sites), alkali-labile sites, and crosslinking in the single cell gel electrophoresis (SCGE). Comet assay was performed in Animal Reproduction Research Institute (ARRI), in Giza, Egypt.

Statistical analysis

SPSS-PC software (1999; Chicago, Il, USA) was used for the statistical analysis of experimental data for the quantitative variables, namely body weight growth and MnPCEs. Probability values with a p-value of 0.05 (p<0.05) were deemed statistically significant.

Results and Discussion

Animal observation

The observation during this study showed that no serve toxicity signs such as diarrhea or hair loss. Furthermore, no mortality was observed related to different doses of ZnO-NPs administration (100, 200, 300, 400, and 600 mg/kg body weight). Also no behavioral changes were observed related to be orally administered to rats. This finding was agreed with the results of (Ben-Slama et al., 2015).

ZnO-NPs effect on rats body weight gain

Table 1 displays the impact of various ZnO-NP concentrations on rat body weight gain throughout the duration of the entire experiment. The average rate of rat body weight gain significantly decreased when compared to the control group, according to the data (p<0.05). The findings of the present investigation regarding the decrees in the body weight gain were consistent with those of Hong et al. (2014), who found that rats given ZnO NPs by gavage at doses of 0, 100, 200, and 400 mg/kg/day saw a drop in body weight due to a decrease in food intake. It was reported that a high dose of ZnO NPs in the diet could have toxicological effects, but Wang et al. (2016) showed that 50 and 500 mg/kg nano-ZnOs demonstrated increases in body weight while at 5,000 mg/kg showed decreases in body weight. This suggests that the decrease in body weight at 5,000 mg/kg ZnO NPs may help explain the increase in the relative weights of the pancreas, brain, and lung. Decrease of body weight may be attributed to anabolic metabolism in body of treated animals, or as a result of antidigestion effect, or due to the loose of appetite in treated animals as a result of nanoparticles administration (Shirvani et al., 2014).

Table 1. Effects of different concentrations of nano-ZnOs on the body weight gain of male rats for ten weeks
Groups Dose (mg/kg) Initial weight Final weight Weight gain
G1 0 161.80±3.55a 241.60±9.35a 79.80±8.02a
G2 100 171.40±5.02a 161.60±8.07ab 19.00±6.42b
G3 200 169.25±9.11a 170.00±13.49ab 20.80±3.81b
G4 300 171.83±4.88a 151.40±8.90b 12.20±4.15b
G5 400 165.20±6.09a 160.40±10.89ab 17.00±5.71b
G6 600 169.80±2.71a 185.40±9.38ab 25.20±7.09b

Data represent the means±SE of 5 animals per group. The difference between mean values with different superscripts in the same column is statistically significant.

Download Excel Table
Effects of ZnO-NPs on hematological parameters of male rats

According to the findings showed in Table 2, treated rats' hemoglobin content and PLT did not differ significantly from the control groups (p<0.05). The findings showed that all rat groups that received ZnO NPs during the experiment experienced significantly lower hematocrit percentages and RBCs than the control group. However, after 10 weeks, the treatment groups' rats had significantly higher levels of WBCs, MCV values, MCH, and MCHC than the control groups' rats. Hematological studies showed that ZnO-NPs can penetrate and translocate within living creatures, which is important for assessing the toxicity of ZnO-NPs (Yang et al., 2016).

Table 2. Effects of different concentrations of nano-ZnOs on hematological parameters of male rats
Parameters Groups
G1 G2 G3 G4 G5 G6
WBC (109/L) 4.27±0.07ab 4.65±0.14ab 4.40±0.35ab 4.66±0.46ab 4.60±0.03ab 4.62±0.18ab
RBCs (1012/L) 7.42±0.15a 7.00±0.14b 6.74±0.11b 6.57±0.17b 6.82±0.13b 6.84±0.08b
Hematocrit (%) 36.73±1.26ab 35.04±0.44b 34.98±0.45b 34.84±0.71b 35.00±0.56b 34.24±0.96b
Hemoglobin (g/dL) 14.35±0.63a 13.84±0.18a 13.64±0.18a 13.84±0.39a 13.94±0.31a 13.96±0.33a
MCV (fL=10–15) 48.25±0.47a 49.74±0.48ab 51.60±0.67b 50.00±0.001ab 51.75±0.85b 50.75±1.25b
MCH (Pg=10–12) 18.91±0.50a 19.85±0.52ab 20.65±0.26b 19.70±0.16ab 20.57±0.24b 20.52±0.31b
MCHC (g/dL) 38.64±0.62a 39.54±0.73ab 39.38±0.62ab 39.94±0.45ab 39.94±0.45ab 41.06±0.24b
PLT (109/L) 567.20±8.45a 519.60±25.70a 498±28.37a 528±16.71a 551.60±41.01a 497±9.57a

The abbreviations RBC, WBC, MCV, MCH, and MCHC stand for red blood cells, white blood cells, and mean corpuscular volume, hemoglobin, and concentration, respectively. Data are the means and standard errors of five animals per group. At p<0.05, the difference between mean values with different superscripts in the same row is statistically significant.

WBC, white blood cell; RBC, red blood cell count; MCV, mean capsular volume; MCHC, mean capsular hemoglobin concentration; PLT, platelet count.

Download Excel Table

According to research by Dhawan and Sharma (2010), when given orally and exposed acutely, NPs of various materials are more hazardous than their microparticle counterparts. According to Sano et al. (2006), a significant decline in lymphocyte percentage, WBC, and MCHC may be the result of blood leakage from vessel walls brought on by high dosages of ZnO-NPs. According to Ben-Slama et al. (2015), platelet levels decreased, which is consistent with our findings. They did, however, demonstrate an increase in RBC, which is in contradiction to our findings. According to research by Wang et al. (2008), an increase in the number of red blood cells may cause a rise in blood viscosity. Additionally, in line with our research, Somayeh and Mohammad (2014) found that the toxicity of zinc oxide nanoparticles causes an increase in white cell count. According to Liu et al. (2015) zinc oxide nanoparticles can lead to increase in white and RBC, a decrease in lymphocyte count, and an increase in neutrophil count. By utilizing the same doses and routes administration of ZnO NPs but with various administration times up to 21 days, Espanani et al. (2015) demonstrated in this period a decrease in the counts of platelet and lymphocyte with an increase in white cell count and no effect on the red cell count. Depending on the administered dose, sub-chronic use of zinc oxide nanoparticles was reported to cause toxic symptoms in the lymphatic system and in the blood cell count (Elshama et al., 2017).

Bone marrow micronuclei assay

Table 3 displays the mean of the micronuclei frequency after treatment with various concentrations of ZnO NPs and the corresponding controls. The PCEs were painted a light blue to grey colour, whereas the NCEs were painted a light pink to light yellow colour (Fig. 2). The results demonstrate that as the dose of ZnO NPs was increased, the frequencies of micronucleated erythrocytes in the bone marrow of treated rats increase as well. By increasing the dose of ZnO NPs, the percentage of PCEs/NCEs was decreased. According to the findings obtained, 10 weeks of administration with 600 mg/kg body weight ZnO-NPs triggered the greatest increase in bone marrow cytotoxicity (PCE / NCE ratio). Extranuclear entities known as micronuclei (MN) are made up of broken-down chromosomal fragments and/or complete chromosomes that were not absorbed into the nucleus after cell division. A buildup of DNA damage, chromosomal abnormalities, and deficiencies in the cell repair mechanism can cause MN (Fenech et al., 2011). Tripathi et al. (2012) evaluated the correlation between the numbers of immature erythrocytes (PCEs) and the mature erythrocytes (NCEs), and they discovered that when the normal bone marrow cell proliferation is hampered by any toxic agent, a decline in the PCE/NCE ratio could be seen.

Table 3. Effects of different concentrations of nano-ZnOs on micronuclei assay
Groups Doses (mg/kg) No. of PCEs Mn/PCEs% PCEs/NCEs%
G1 0 2,000 0.15 17.99
G2 100 2,000 0.25 12.11
G3 200 2,000 0.45 12.38
G4 300 2,000 0.5 11.88
G5 400 2,000 0.6 11.38
G6 600 2,000 0.8 11.70

PCEs, polychromatic erythrocytes; NCE, normochromatic erythrocyte.

Download Excel Table
fl-2022-3-79-g2
Fig. 2. Nano-ZnOs caused the formation of micronuclei (MN) in rat bone marrow cells (Giemsa stain (10%) for staining). PCE, polychromatic erythrocyte; NCE, normochromatic erythrocyte; MNPCE, micronucleated polychromatic erythrocyte (magnification 1,000×).
Download Original Figure

According to Suzuki et al. (1989), PCE counts in peripheral blood were the most widely used and practical way to monitor erythropoiesis. They also noted that examination of erythropoietic cytotoxicity was a significant component of safety assessment in novel drug development. Fewer immature erythrocytes (PCE) than mature or NCE were thought to be a marker of cytotoxicity caused by mutagens (Kirsch-Volders et al., 2003). Some micronucleus test guidelines advise using the P/N ratio to estimate a compound's toxicity to bone marrow cells. It has been demonstrated that the P/N ratio changes when stronger pharmacologic doses are given or when bone marrow cells are extracted at later sampling intervals (Heddle et al., 1984). A decline in the P/N ratio could be brought on by either a decline in PCE, an increase in NCE, or imbalanced alterations in the populations of both cell types.

Alkaline comet assay

The findings in Tables 4 and 5 demonstrated that ZnO-NPs increased the tail length, tail intensity, tail migration, and tail moments in the kidneys and liver, respectively. According to the results in Table 4, all treatment groups of rats had their liver DNA's tail length increases as a result of the administration of nano-ZnOs; the tail lengths of the rats in group (G6) and control group (G1) were 10.57 μm and 4.94 μm, respectively. According to the results illustrated in Table 5, kidney cell tail length, DNA content, tail moment, and olive tail moment were all dose-dependently increased by ZnO-NP compared to control. These results reveal that genotoxicity was the cause of the DNA damage that was seen, and that the DNA damage caused by ZnO-NPs in the rat liver and kidneys might be detected using the comet assay. Photomicrographs of the DNA damage (comet assay) in rat liver and kidney cells were shown in Figs. 3 and 4, respectively. The single-cell gel electrophoresis assay, also referred to as the comet assay, is a technique for quantifying DNA strand breaks in eukaryotic cells (Tice et al., 2000). Based on the length of the genetic material's movement (tail length) in the anode-directed direction during the comet assay, the quantity of DNA breakage in a cell was calculated (Singh et al., 1988). Furthermore, it has been demonstrated that the frequency of DNA strand breaks is inversely correlated with the proportion of DNA in the tail (tail intensity) (Olive et al., 1990). The computerized image analysis system calculates the tail moment, a straightforward descriptor by taking into account both the migration tail length and the percentage of DNA that travelled in the tail (Villarini et al., 1998). DNA damage results from the lengthening of DNA tails at all ZnONP doses over the course of 70 days, as is seen from DNA fragmentation. This genotoxicity may be caused by ZnONPs' oxidative and nitrosative actions (Kumar and Dhawan, 2013; Sharma et al., 2012).

Table 4. Measurements of DNA damage in liver cells from control and ZnONP-treated rats
Treatment Dose (mg/kg) Tail length Tail in DNA Tail moment Olive tail moment
G1 0 4.94 8.87 0.46 1.23
G2 100 7.43 9.421 0.56 1.36
G3 200 6.22 13.21 0.98 1.39
G4 300 10.44 16.79 2.097 2.09
G5 400 9.39 14.48 1.90 2.02
G6 600 10.67 18.89 2.44 2.39
Download Excel Table
Table 5. Measurements of DNA damage in kidney cells from control and ZnONP-treated rats
Treatment Dose (mg/kg) Tail length Tail in DNA Tail moment Olive tail moment
G1 0 8.84 12.77 0.80 1.04
G2 100 9.69 14.48 1.72 1.64
G3 200 9.39 15.53 1.90 2.03
G4 300 10.55 17.19 1.93 2.08
G5 400 10.67 18.89 2.44 2.39
G6 600 11.97 24.75 3.63 2.97
Download Excel Table
fl-2022-3-79-g3
Fig. 3. Photographs of representative DNA damage (comet assay) in rats liver cells consumption of different concentrations of ZnONPs (SYBR Green stain for staining). (G1) Normal cell; (G2 and G3) little DNA damage; (G4) moderate DNA damage; (G5) extensive DNA damage; (G6) completely damaged DNA (Magnification 1,000×).
Download Original Figure
fl-2022-3-79-g4
Fig. 4. Photographs of representative DNA damage (comet assay) in rat kidney cells consumption of different concentrations of ZnONPs (SYBR Green stain for staining). (G1) Normal cell; (G2 and G3) little DNA damage; (G4) moderate DNA damage; (G5) extensive DNA damage; (G6) completely damaged DNA (Magnification 1,000×).
Download Original Figure

Conclusion

A conclusion that can be made from in vivo genotoxicity experiments is that ZnO NPs with particle sizes of 30 nm for ten weeks are capable of causing genotoxicity and cytotoxicity in rat bone marrow cells, liver, and kidney. The results of present study may increase the alarms about the probable risk on human health that might be related with plentiful applications of ZnO NPs.

Conflicts of Interest

The authors declare no potential conflict of interest.

Acknowledgments

Not applicable.

Ethics Approval

Following approval from the Institutional Animal Ethical Committee for Fayoum University, all experiments were done following general international guidelines on the use of living laboratory animals in scientific research. The work has been carried out in accordance with EU Directive 2010/63/EU for animal experiments (Fu-IACUC, Permission number 6200, 2011).

Author Contributions

Conceptualization: Ramadan AG, Hassan GM.

Data curation: Ramadan AG, Yassein AAM, Hassan GM.

Formal analysis: Ramadan AG, Yassein AAM.

Validation: Hassan GM.

Investigation: Ramadan AG, Yassein AAM, Hassan GM.

Writing-original draft: Ramadan AG, Hassan GM.

Writing-review&editing: Ramadan AG, Yassein AAM, Eissa EA, Hassan GM.

References

1.

Al-Suhaibani ES, El-Morshedi NA. 2014. Histopathological and ultrastructural effect of zinc oxide nanoparticles on male Wistar rats submandibular glands. IOSR J Pharm Biol Sci 9:5-9.

2.

Ben-Slama I, Mrad I, Rihane N, Mir LE, Sakly M, Amara S. 2015. Sub-acute oral toxicity of zinc oxide nanoparticles in male rats. J Nanomed Nanotechnol 6:100284.

3.

Chupani L, Zusková E, Niksirat H, Panáček A, Lünsmann V, Haange SB, von Bergen M, Jehmlich N. 2017. Effects of chronic dietary exposure of zinc oxide nanoparticles on the serum protein profile of juvenile common carp (Cyprinus carpio L.). Sci Total Environ 579:1504-1511.

4.

Dhawan A, Sharma V. 2010. Toxicity assessment of nanomaterials: Methods and challenges. Anal Bioanal Chem 398:589-605.

5.

Elshama SS, Salem RR, Osman HEH, El-Kenawy AEM. 2017. Toxic effect of sub-chronic use of zinc oxide nanoparticles on the lymphatic system of adult albino rats. Curr Top Toxicol 13.

6.

Espanani HR, Faghfoori Z, Izadpanah M, Babadi VY. 2015. Toxic effect of nano-zinc oxide. Bratisl Lek Listy 116: 616-620.

7.

Fenech M. 2011. Micronuclei and their association with sperm abnormalities, infertility, pregnancy loss, pre-eclampsia and intra-uterine growth restriction in humans. Mutagenesis 26:63-67.

8.

Heddle JA, Stuart E, Salamone MF. 1984. The bone marrow micronucleus test. In Handbook of mutagenicity test procedures. 2nd ed. Kilbey BJ, Legator M, Nichols W, Ramel C (eds). Elsevier, Amsterdam, Netherland. pp 441-457.

9.

Hong JS, Park MK, Kim MS, Lim JH, Park GJ, Meang EH, Shin JH, Kim MK, Jeong J, Park JA, Kim JC, Shin HC. 2014. Prenatal development toxicity study of zinc oxide nanoparticles in rats. Int J Nanomed 9:159-171.

10.

Iavicoli I, Leso V, Beezhold DH, Shvedova AA. 2017. Nanotechnology in agriculture: Opportunities, toxicological implications, and occupational risks. Toxicol Appl Pharmacol 329:96-111.

11.

Kirsch-Volders M, Vanhauwaert A, Eichenlaub-Ritter U, Decordier I. 2003. Indirect mechanisms of genotoxicity. Toxicol Lett 140–141:63-74.

12.

Kisin ER, Murray AR, Keane MJ, Shi XC, Schwegler-Berry D, Gorelik O, Arepalli S, Castranova V, Wallace WE, Kagan VE, Shvedova AA. 2007. Single-walled carbon nanotubes: Geno- and cytotoxic effects in lung fibroblast V79 cells. J Toxicol Environ Health A 70:2071-2079.

13.

Kumar A, Dhawan A. 2013. Genotoxic and carcinogenic potential of engineered nanoparticles: An update. Arch Toxicol 87:1883-1900.

14.

Liu J, Kang Y, Zhang W, Song B, Wei L, Chen L, Shao L. 2017. From the cover: Ion-shedding zinc oxide nanoparticles induce microglial BV2 cell proliferation via the ERK and Akt signaling pathways. Toxicol Sci 156:167-178.

15.

Liu HL, Yang HL, Lin BC, Zhang W, Tian L, Zhang HS, Xi ZG. 2015. Toxic effect comparison of three typical sterilization nanoparticles on oxidative stress and immune inflammation response in rats. Toxicol Res 4:486-493.

16.

McKelvey-Martin VJ, Green MHL, Schmezer P, Pool-Zobel BL, De Méo MP, Collins A. 1993. The single cell gel electrophoresis assay (comet assay): A European review. Mutat Res 288:47-63.

17.

Minetto D, Volpi Ghirardini A, Libralato G. 2016. Saltwater ecotoxicology of Ag, Au, CuO, TiO2, ZnO and C60 engineered nanoparticles: An overview. Environ Int 92-93:189-201.

18.

Olive PL, Banáth JP, Durand RE. 1990. Heterogeneity in radiation-induced DNA damage and repair in tumor and normal cells measured using the “comet” assay. Radiat Res 122:86-94.

19.

Sano H, Hosokawa K, Kidoya H, Takakura N. 2006. Negative regulation of VEGF-induced vascular leakage by blockade of angiotensin II type 1 receptor. Arterioscler Thromb Vasc Biol 26:2673-2680.

20.

Schmid W. 1975. The micronucleus test. Mutat Res Environ Mutagen Relat Subj 31:9-15.

21.

Sharma V, Singh P, Pandey AK, Dhawan A. 2012. Induction of oxidative stress, DNA damage and apoptosis in mouse liver after sub-acute oral exposure to zinc oxide nanoparticles. Mutat Res Genet Toxicol Environ Mutagen 745:84-91.

22.

Shirvani H, Noori A, Mashayekh AM. 2014. The effect of ZnO nanoparticles on the growth and puberty of newborn male Wistar rats. Int J Basic Sci Appl Res 3:180-185.

23.

Singh NP, McCoy MT, Tice RR, Schneider EL. 1988. A simple technique for quantitation of low levels of DNA damage in individual cells. Exp Cell Res 175:184-191.

24.

Somayeh B, Mohammad F. 2014. Vitamin C can reduce toxic effects of nano zinc oxide. Int Res J Biol Sci 3:65-70.

25.

Srivastav AK, Kumar M, Ansari NG, Jain AK, Shankar J, Arjaria N, Jagdale P, Singh D. 2016. A comprehensive toxicity study of zinc oxide nanoparticles versus their bulk in Wistar rats: Toxicity study of zinc oxide nanoparticles. Hum Exp Toxicol 35:1286-1304.

26.

Suzuki Y, Nagae Y, Li J, Sakaba H, Mozawa K, Takahashi A, Shimizu H. 1989. The micronucleus test and erythropoiesis. Effects of erythropoietin and a mutagen on the ratio of polychromatic to normochromatic erythrocytes (P/N ratio). Mutagenesis 4:420-424.

27.

Terradas M, Martín M, Tusell L, Genescà A. 2010. Genetic activities in micronuclei: Is the DNA entrapped in micronuclei lost for the cell? Mutat Res Rev Mutat Res 705:60-67.

28.

Tice RR, Agurell E, Anderson D, Burlinson B, Hartmann A, Kobayashi H, Miyamae Y, Rojas E, Ryu JC, Sasaki YF. 2000. Single cell gel/comet assay: Guidelines for in vitro and in vivo genetic toxicology testing. Environ Mol Mutagen 35:206-221.

29.

Tripathi R, Pancholi SS, Tripathi P. 2012. Genotoxicity of ibuprofen in mouse bone marrow cells in vivo. Drug Chem Toxicol 35:389-392.

30.

Villarini M, Moretti M, Pasquini R, Scassellati-Sforzolini G, Fatigoni C, Marcarelli M, Monarca S, Rodrı´guez AV. 1998. In vitro genotoxic effects of the insecticide deltamethrin in human peripheral blood leucocytes: DNA damage (‘comet’ assay) in relation to the induction of sister-chromatid exchanges and micronuclei. Toxicology 130:129-139.

31.

Wang B, Feng W, Wang M, Wang T, Gu Y, Zhu M, Ouyang H, Shi J, Zhang F, Zhao Y, Chai Z, Wang H, Wang J. 2008. Acute toxicological impact of nano- and submicro-scaled zinc oxide powder on healthy adult mice. J Nanopart Res 10:263-276.

32.

Wang C, Lu J, Zhou L, Li J, Xu J, Li W, Zhang L, Zhong X, Wang T. 2016. Effects of long-term exposure to zinc oxide nanoparticles on development, zinc metabolism and biodistribution of minerals (Zn, Fe, Cu, Mn) in mice. PLOS ONE 11:0164434.

33.

Yang Y, Qin Z, Zeng W, Yang T, Cao Y, Mei C, Kuang Y. 2016. Toxicity assessment of nanoparticles in various systems and organs. Nanotechnol Rev 6:279-289.

34.

Yeh TK, Chen JK, Lin CH, Yang MH, Yang CS, Chou FI, Peir JJ, Wang MY, Chang WH, Tsai MH, Tsai HT, Lin P. 2012. Kinetics and tissue distribution of neutron-activated zinc oxide nanoparticles and zinc nitrate in mice: Effects of size and particulate nature. Nanotechnology 23:085102.

35.

Zayed NA, Luaibi NM. 2018. Effect of ZnO NPs on body and organ weights in male rat. Int J Innov Sci Eng Technol 5:12-25.