Home A review of the potential adverse health impacts of atrazine in humans
Article
Licensed
Unlicensed Requires Authentication

A review of the potential adverse health impacts of atrazine in humans

  • Sina Arabi , Motahar Heidari-Beni , Parinaz Poursafa , Minaalsadat Roshanaei and Roya Kelishadi EMAIL logo
Published/Copyright: September 17, 2024

Abstract

Atrazine is a widely used chlorinated triazine herbicide in agricultural settings, which has raised concerns over its potential adverse effects on human health. The extensive application of atrazine has resulted in its pervasive presence in the environment, contaminating soil, groundwater, and surface water. While earlier research suggested that atrazine is unlikely to pose a health concern, recent evidence has indicated the necessity to reassess this point of view. This review aims to assess the recent evidence on atrazine’s adverse effects on human health, focusing on (i) Cancer, (ii) Metabolic Diseases, (iii) Reproductive System, (iv) Neural System, and (v) Epigenetic Effects. Strategies to mitigate atrazine contamination and limitations of previous studies are also discussed. We strongly believe that further investigation is necessary to determine the potential detrimental consequences of atrazine in humans, particularly in developing countries, where herbicides are widely used without stringent safety regulations. Therefore, the current review will be beneficial for guiding future research and regulatory measures concerning the use of atrazine.


Corresponding author: Roya Kelishadi, Professor, Department of Pediatrics, Child Growth and Development Research Center, Research Institute for Primordial Prevention of Non-Communicable Disease, Isfahan University of Medical Sciences, Isfahan, Iran, E-mail:

  1. Research ethics: Not applicable.

  2. Informed consent: Not applicable.

  3. Author contributions: Roya Kelishadi, Motahar Heidari-Beni, and Sina Arabi conceived of the presented idea. Data collection was performed by Sina Arabi and Parinaz Poursafa. Minaalsadat Roshanaei illustrated the visual abstracts (figures). The first draft of the manuscript was written by Sina Arabi in consultation with Roya Kelishadi and Motahar Heidari-Beni. All authors commented on previous versions of the manuscript and approved the final manuscript.

  4. Competing interests: The authors have no relevant financial or non-financial interests to disclose.

  5. Research funding: The authors declare that no funds, grants, or other support were received during the preparation of this manuscript.

  6. Data availability: Not applicable.

References

1. Ahn, C, Jeung, EB. Endocrine-disrupting chemicals and disease endpoints. Int J Mol Sci 2023;24:5342. https://doi.org/10.3390/ijms24065342.Search in Google Scholar PubMed PubMed Central

2. Sharma, A, Kumar, V, Shahzad, B, Tanveer, M, Sidhu, GPS, Handa, N, et al.. Worldwide pesticide usage and its impacts on ecosystem. SN Appl Sci 2019;1:1446. https://doi.org/10.1007/s42452-019-1485-1.Search in Google Scholar

3. Khatoon, A, Yasir Nawaz, M, Mehboob, G, Saleemi, MK, Gul, ST, Abbas, RZ, et al.. Unraveling the combined deleterious effects of ochratoxin A and atrazine upon broiler’s health: toxicopathological, serum biochemical and immunological perspectives. Toxicon 2023;236:107327. https://doi.org/10.1016/j.toxicon.2023.107327.Search in Google Scholar PubMed

4. Remigio, RV, Andreotti, G, Sandler, DP, Erickson, PA, Koutros, S, Albert, PS, et al.. An updated evaluation of atrazine-cancer incidence associations among pesticide applicators in the agricultural health study cohort. Environ Health Perspect 2024;132:27010. https://doi.org/10.1289/ehp13684.Search in Google Scholar PubMed PubMed Central

5. He, H, Liu, Y, You, S, Liu, J, Xiao, H, Tu, Z. A review on recent treatment technology for herbicide atrazine in contaminated environment. Int J Environ Res Publ Health 2019;16:5129. https://doi.org/10.3390/ijerph16245129.Search in Google Scholar PubMed PubMed Central

6. Roh, T, Aggarwal, A, Hasan, NT, Upadhyay, A, Trisha, NF. Pesticides and cancer. In: Bernicker, EH, editor. Environmental oncology: theory and impact. Cham: Springer International Publishing; 2023:177–211 pp.10.1007/978-3-031-33750-5_8Search in Google Scholar

7. Sass, JB, Colangelo, A. European Union bans atrazine, while the United States negotiates continued use. Int J Occup Environ Health 2006;12:260–7. https://doi.org/10.1179/oeh.2006.12.3.260.Search in Google Scholar PubMed

8. Vonberg, D, Hofmann, D, Vanderborght, J, Lelickens, A, Köppchen, S, Pütz, T, et al.. Atrazine soil core residue analysis from an agricultural field 21 years after its ban. J Environ Qual 2014;43:1450–9. https://doi.org/10.2134/jeq2013.12.0497.Search in Google Scholar PubMed

9. Liu, Y, Fan, X, Zhang, T, He, W, Song, F. Effects of the long-term application of atrazine on soil enzyme activity and bacterial community structure in farmlands in China. Environ Pollut 2020;262:114264. https://doi.org/10.1016/j.envpol.2020.114264.Search in Google Scholar PubMed

10. Freeman, LE, Rusiecki, JA, Hoppin, JA, Lubin, JH, Koutros, S, Andreotti, G, et al.. Atrazine and cancer incidence among pesticide applicators in the agricultural health study (1994–2007). Environ Health Perspect 2011;119:1253–9. https://doi.org/10.1289/ehp.1103561.Search in Google Scholar PubMed PubMed Central

11. Wetzel, LT, Luempert, LG3rd, Breckenridge, CB, Tisdel, MO, Stevens, JT, Thakur, AK, et al.. Chronic effects of atrazine on estrus and mammary tumor formation in female Sprague-Dawley and Fischer 344 rats. J Toxicol Environ Health 1994;43:169–82. https://doi.org/10.1080/15287399409531913.Search in Google Scholar PubMed

12. Hoar, ZS, Weisenburger, DD, Cantor, KP, Holmes, FF, Blair, A. Role of the herbicide atrazine in the development of non-Hodgkin’s lymphoma. Scand J Work Environ Health 1993;19:108–14. https://doi.org/10.5271/sjweh.1499.Search in Google Scholar PubMed

13. MacLennan, PA, Delzell, E, Sathiakumar, N, Myers, SL, Cheng, H, Grizzle, W, et al.. Cancer incidence among triazine herbicide manufacturing workers. J Occup Environ Med 2002;44:1048–58. https://doi.org/10.1097/00043764-200211000-00011.Search in Google Scholar PubMed

14. Jowa, L, Howd, R. Should atrazine and related chlorotriazines be considered carcinogenic for human health risk assessment? J Environ Sci Health C Environ Carcinog Ecotoxicol Rev 2011;29:91–144. https://doi.org/10.1080/10590501.2011.577681.Search in Google Scholar PubMed

15. Alavanja, MC, Samanic, C, Dosemeci, M, Lubin, J, Tarone, R, Lynch, CF, et al.. Use of agricultural pesticides and prostate cancer risk in the Agricultural Health Study cohort. Am J Epidemiol 2003;157:800–14. https://doi.org/10.1093/aje/kwg040.Search in Google Scholar PubMed

16. Lerro, CC, Beane Freeman, LE, DellaValle, CT, Andreotti, G, Hofmann, JN, Koutros, S, et al.. Pesticide exposure and incident thyroid cancer among male pesticide applicators in agricultural health study. Environ Int 2021;146:106187. https://doi.org/10.1016/j.envint.2020.106187.Search in Google Scholar PubMed PubMed Central

17. Reynolds, P, Hurley, SE, Goldberg, DE, Yerabati, S, Gunier, RB, Hertz, A, et al.. Residential proximity to agricultural pesticide use and incidence of breast cancer in the California Teachers Study cohort. Environ Res 2004;96:206–18. https://doi.org/10.1016/j.envres.2004.03.001.Search in Google Scholar PubMed

18. Wang, M, Chen, J, Zhao, S, Zheng, J, He, K, Liu, W, et al.. Atrazine promotes breast cancer development by suppressing immune function and upregulating MMP expression. Ecotoxicol Environ Saf 2023;253:114691. https://doi.org/10.1016/j.ecoenv.2023.114691.Search in Google Scholar PubMed

19. Sasikala, S, Minu Jenifer, M, Velavan, K, Sakthivel, M, Sivasamy, R, Fenwick Antony, ER. Predicting the relationship between pesticide genotoxicity and breast cancer risk in South Indian women in in vitro and in vivo experiments. Sci Rep 2023;13:9712. https://doi.org/10.1038/s41598-023-35552-3.Search in Google Scholar PubMed PubMed Central

20. Sathiakumar, N, MacLennan, PA, Mandel, J, Delzell, E. A review of epidemiologic studies of triazine herbicides and cancer. Crit Rev Toxicol 2011;41:1–34. https://doi.org/10.3109/10408444.2011.554793.Search in Google Scholar PubMed

21. Donna, A, Crosignani, P, Robutti, F, Betta, PG, Bocca, R, Mariani, N, et al.. Triazine herbicides and ovarian epithelial neoplasms. Scand J Work Environ Health 1989;15:47–53. https://doi.org/10.5271/sjweh.1882.Search in Google Scholar PubMed

22. Inoue-Choi, M, Weyer, PJ, Jones, RR, Booth, BJ, Cantor, KP, Robien, K, et al.. Atrazine in public water supplies and risk of ovarian cancer among postmenopausal women in the Iowa Women’s Health Study. Occup Environ Med 2016;73:582–7. https://doi.org/10.1136/oemed-2016-103575.Search in Google Scholar PubMed PubMed Central

23. Chen, J, Liu, J, Wu, S, Liu, W, Xia, Y, Zhao, J, et al.. Atrazine promoted epithelial ovarian cancer cells proliferation and metastasis by inducing low dose reactive oxygen species (ROS). Iran J Biotechnol 2021;19:e2623. https://doi.org/10.30498/IJB.2021.2623.Search in Google Scholar PubMed PubMed Central

24. Hu, K, Tian, Y, Du, Y, Huang, L, Chen, J, Li, N, et al.. Atrazine promotes RM1 prostate cancer cell proliferation by activating STAT3 signaling. Int J Oncol 2016;48:2166–74. https://doi.org/10.3892/ijo.2016.3433.Search in Google Scholar PubMed

25. Ward, ZJ, Bleich, SN, Cradock, AL, Barrett, JL, Giles, CM, Flax, C, et al.. Projected U.S. state-level prevalence of adult obesity and severe obesity. N Engl J Med 2019;381:2440–50. https://doi.org/10.1056/nejmsa1909301.Search in Google Scholar

26. Diamanti-Kandarakis, E, Bourguignon, JP, Giudice, LC, Hauser, R, Prins, GS, Soto, AM, et al.. Endocrine-disrupting chemicals: an Endocrine Society scientific statement. Endocr Rev 2009;30:293–342. https://doi.org/10.1210/er.2009-0002.Search in Google Scholar PubMed PubMed Central

27. Lobstein, T, Brownell, KD. Endocrine-disrupting chemicals and obesity risk: a review of recommendations for obesity prevention policies. Obes Rev 2021;22:e13332. https://doi.org/10.1111/obr.13332.Search in Google Scholar PubMed

28. Song, Y, Chou, EL, Baecker, A, You, NC, Song, Y, Sun, Q, et al.. Endocrine-disrupting chemicals, risk of type 2 diabetes, and diabetes-related metabolic traits: a systematic review and meta-analysis. J Diabetes 2016;8:516–32. https://doi.org/10.1111/1753-0407.12325.Search in Google Scholar PubMed

29. Heindel, JJ, Blumberg, B, Cave, M, Machtinger, R, Mantovani, A, Mendez, MA, et al.. Metabolism disrupting chemicals and metabolic disorders. Reprod Toxicol 2017;68:3–33. https://doi.org/10.1016/j.reprotox.2016.10.001.Search in Google Scholar PubMed PubMed Central

30. Lim, S, Ahn, SY, Song, IC, Chung, MH, Jang, HC, Park, KS, et al.. Chronic exposure to the herbicide, atrazine, causes mitochondrial dysfunction and insulin resistance. PLoS One 2009;4:e5186. https://doi.org/10.1371/journal.pone.0005186.Search in Google Scholar PubMed PubMed Central

31. LaVerda, NL, Goldsmith, DF, Alavanja, MC, Hunting, KL. Pesticide exposures and body mass index (BMI) of pesticide applicators from the agricultural health study. J Toxicol Environ Health A 2015;78:1255–76. https://doi.org/10.1080/15287394.2015.1074844.Search in Google Scholar

32. Wei, D, Wang, L, Xu, Q, Wang, J, Shi, J, Ma, C, et al.. Exposure to herbicides mixtures in relation to type 2 diabetes mellitus among Chinese rural population: results from different statistical models. Ecotoxicol Environ Saf 2023;261:115109. https://doi.org/10.1016/j.ecoenv.2023.115109.Search in Google Scholar

33. Kongtip, P, Nankongnab, N, Kallayanatham, N, Pundee, R, Choochouy, N, Yimsabai, J, et al.. Thyroid hormones in conventional and organic farmers in Thailand. Int J Environ Res Publ Health 2019;16:2704. https://doi.org/10.3390/ijerph16152704.Search in Google Scholar

34. de Albuquerque, FP, de Oliveira, JL, Moschini-Carlos, V, Fraceto, LF. An overview of the potential impacts of atrazine in aquatic environments: perspectives for tailored solutions based on nanotechnology. Sci Total Environ 2020;700:134868. https://doi.org/10.1016/j.scitotenv.2019.134868.Search in Google Scholar

35. Hayes, TB, Anderson, LL, Beasley, VR, de Solla, SR, Iguchi, T, Ingraham, H, et al.. Demasculinization and feminization of male gonads by atrazine: consistent effects across vertebrate classes. J Steroid Biochem Mol Biol 2011;127:64–73. https://doi.org/10.1016/j.jsbmb.2011.03.015.Search in Google Scholar

36. Stoker, TE, Laws, SC, Guidici, DL, Cooper, RL. The effect of atrazine on puberty in male Wistar rats: an evaluation in the protocol for the assessment of pubertal development and thyroid function. Toxicol Sci 2000;58:50–9. https://doi.org/10.1093/toxsci/58.1.50.Search in Google Scholar

37. Kniewald, J, Jakominić, M, Tomljenović, A, Simić, B, Romać, P, Vranesić, D, et al.. Disorders of male rat reproductive tract under the influence of atrazine. J Appl Toxicol 2000;20:61–8. https://doi.org/10.1002/(sici)1099-1263(200001/02)20:1<61::aid-jat628>3.3.co;2-v.10.1002/(SICI)1099-1263(200001/02)20:1<61::AID-JAT628>3.3.CO;2-VSearch in Google Scholar

38. Swan, SH. Semen quality in fertile US men in relation to geographical area and pesticide exposure. Int J Androl 2006;29:62–8. https://doi.org/10.1111/j.1365-2605.2005.00620.x. ; discussion 105–8.Search in Google Scholar

39. Abarikwu, SO, Ezim, OE, Ikeji, CN, Farombi, EO. Atrazine: cytotoxicity, oxidative stress, apoptosis, testicular effects and chemopreventive interventions. Front Toxicol 2023;5:1246708. https://doi.org/10.3389/ftox.2023.1246708.Search in Google Scholar

40. Abarikwu, SO, Farombi, EO, Kashyap, MP, Pant, AB. Atrazine induces transcriptional changes in marker genes associated with steroidogenesis in primary cultures of rat Leydig cells. Toxicol Vitro 2011;25:1588–95. https://doi.org/10.1016/j.tiv.2011.06.002.Search in Google Scholar

41. Laws, SC, Ferrell, JM, Stoker, TE, Schmid, J, Cooper, RL. The effects of atrazine on female Wistar rats: an evaluation of the protocol for assessing pubertal development and thyroid function. Toxicol Sci 2000;58:366–76. https://doi.org/10.1093/toxsci/58.2.366.Search in Google Scholar PubMed

42. Rayner, JL, Enoch, RR, Fenton, SE. Adverse effects of prenatal exposure to atrazine during a critical period of mammary gland growth. Toxicol Sci 2005;87:255–66. https://doi.org/10.1093/toxsci/kfi213.Search in Google Scholar PubMed

43. Rayner, JL, Wood, C, Fenton, SE. Exposure parameters necessary for delayed puberty and mammary gland development in Long-Evans rats exposed in utero to atrazine. Toxicol Appl Pharmacol 2004;195:23–34. https://doi.org/10.1016/j.taap.2003.11.005.Search in Google Scholar PubMed

44. Wirbisky, SE, Freeman, JL. Atrazine exposure and reproductive dysfunction through the hypothalamus–pituitary–gonadal (HPG) axis. Toxics 2015;3:414–50. https://doi.org/10.3390/toxics3040414.Search in Google Scholar PubMed PubMed Central

45. Foradori, CD, Zimmerman, AD, Hinds, LR, Zuloaga, KL, Breckenridge, CB, Handa, RJ. Atrazine inhibits pulsatile gonadotropin-releasing hormone (GnRH) release without altering GnRH messenger RNA or protein levels in the female rat. Biol Reprod 2013;88:9. https://doi.org/10.1095/biolreprod.112.102277.Search in Google Scholar PubMed

46. Foradori, CD, Hinds, LR, Hanneman, WH, Handa, RJ. Effects of atrazine and its withdrawal on gonadotropin-releasing hormone neuroendocrine function in the adult female Wistar rat. Biol Reprod 2009;81:1099–105. https://doi.org/10.1095/biolreprod.109.077453.Search in Google Scholar PubMed

47. Stradtman, SC, Freeman, JL. Mechanisms of neurotoxicity associated with exposure to the herbicide atrazine. Toxics 2021;9:207. https://doi.org/10.3390/toxics9090207.Search in Google Scholar PubMed PubMed Central

48. James, KA, Hall, DA. Groundwater pesticide levels and the association with Parkinson disease. Int J Toxicol 2015;34:266–73. https://doi.org/10.1177/1091581815583561.Search in Google Scholar PubMed

49. Wei, H, Zhang, X, Yang, X, Yu, Q, Deng, S, Guan, Q, et al.. Prenatal exposure to pesticides and domain-specific neurodevelopment at age 12 and 18 months in Nanjing, China. Environ Int 2023;173:107814. https://doi.org/10.1016/j.envint.2023.107814.Search in Google Scholar PubMed

50. Genovese, T, Siracusa, R, Fusco, R, D’Amico, R, Impellizzeri, D, Peritore, AF, et al.. Atrazine inhalation causes neuroinflammation, apoptosis and accelerating brain aging. Int J Mol Sci 2021;22:7938. https://doi.org/10.3390/ijms22157938.Search in Google Scholar PubMed PubMed Central

51. Klein, MO, Battagello, DS, Cardoso, AR, Hauser, DN, Bittencourt, JC, Correa, RG. Dopamine: functions, signaling, and association with neurological diseases. Cell Mol Neurobiol 2019;39:31–59. https://doi.org/10.1007/s10571-018-0632-3.Search in Google Scholar PubMed PubMed Central

52. Das, PC, McElroy, WK, Cooper, RL. Differential modulation of catecholamines by chlorotriazine herbicides in pheochromocytoma (PC12) cells in vitro. Toxicol Sci 2000;56:324–31. https://doi.org/10.1093/toxsci/56.2.324.Search in Google Scholar PubMed

53. Walters, JL, Lansdell, TA, Lookingland, KJ, Baker, LE. The effects of gestational and chronic atrazine exposure on motor behaviors and striatal dopamine in male Sprague-Dawley rats. Toxicol Appl Pharmacol 2015;289:185–92. https://doi.org/10.1016/j.taap.2015.09.026.Search in Google Scholar PubMed PubMed Central

54. Song, XY, Li, JN, Wu, YP, Zhang, B, Li, BX. Atrazine causes autophagy- and apoptosis-related neurodegenerative effects in dopaminergic neurons in the rat nigrostriatal dopaminergic system. Int J Mol Sci 2015;16:13490–506. https://doi.org/10.3390/ijms160613490.Search in Google Scholar PubMed PubMed Central

55. Bardullas, U, Giordano, M, Rodríguez, VM. Atrazine is primarily responsible for the toxicity of long-term exposure to a combination of atrazine and inorganic arsenic in the nigrostriatal system of the Albino rat. Neurotoxicol Teratol 2013;40:59–66. https://doi.org/10.1016/j.ntt.2013.10.003.Search in Google Scholar PubMed

56. Li, G, Wang, K, Zuo, K, Shi, G, Cai, Q, Huang, M. TDP-43 is a potential marker of dopaminergic neuronal damage caused by atrazine exposure. Ecotoxicol Environ Saf 2023;255:114780. https://doi.org/10.1016/j.ecoenv.2023.114780.Search in Google Scholar PubMed

57. Jo, M, Lee, S, Jeon, Y-M, Kim, S, Kwon, Y, Kim, H-J. The role of TDP-43 propagation in neurodegenerative diseases: integrating insights from clinical and experimental studies. Exp Mol Med 2020;52:1652–62. https://doi.org/10.1038/s12276-020-00513-7.Search in Google Scholar PubMed PubMed Central

58. Zhu, SY, Jiang, JZ, Lin, J, Liu, L, Guo, JY, Li, JL. Lycopene ameliorates atrazine-induced spatial learning and memory impairments by inhibiting ferroptosis in the hippocampus of mice. Food Chem Toxicol 2023;174:113655. https://doi.org/10.1016/j.fct.2023.113655.Search in Google Scholar PubMed

59. Li, J, Bi, H. Integrating network pharmacology and in vitro model to investigate hippocampal neurotoxicity induced by atrazine. Toxicol Mech Methods 2022;32:259–67. https://doi.org/10.1080/15376516.2021.1995917.Search in Google Scholar PubMed

60. Ma, K, Wu, HY, Wang, SY, Li, BX. The Keap1/Nrf2-ARE signaling pathway is involved in atrazine induced dopaminergic neurons degeneration via microglia activation. Ecotoxicol Environ Saf 2021;226:112862. https://doi.org/10.1016/j.ecoenv.2021.112862.Search in Google Scholar PubMed

61. Ma, K, Wu, HY, Zhang, B, He, X, Li, BX. Neurotoxicity effects of atrazine-induced SH-SY5Y human dopaminergic neuroblastoma cells via microglial activation. Mol Biosyst 2015;11:2915–24. https://doi.org/10.1039/c5mb00432b.Search in Google Scholar PubMed

62. Shi, G, Zhang, C, Li, G, Wang, K, Cai, Q, Huang, M. Atrazine induces phagocytotic dysfunction of microglia depends on nucleocytoplasmic translocation of acetylated HMGB1. Ecotoxicol Environ Saf 2023;252:114583. https://doi.org/10.1016/j.ecoenv.2023.114583.Search in Google Scholar PubMed

63. Chen, R, Kang, R, Tang, D. The mechanism of HMGB1 secretion and release. Exp Mol Med 2022;54:91–102. https://doi.org/10.1038/s12276-022-00736-w.Search in Google Scholar PubMed PubMed Central

64. Shan, W, Hu, W, Wen, Y, Ding, X, Ma, X, Yan, W, et al.. Evaluation of atrazine neurodevelopment toxicity in vitro-application of hESC-based neural differentiation model. Reprod Toxicol 2021;103:149–58. https://doi.org/10.1016/j.reprotox.2021.06.009.Search in Google Scholar PubMed

65. Alavian-Ghavanini, A, Rüegg, J. Understanding epigenetic effects of endocrine disrupting chemicals: from mechanisms to novel test methods. Basic Clin Pharmacol Toxicol 2018;122:38–45. https://doi.org/10.1111/bcpt.12878.Search in Google Scholar PubMed

66. Hao, C, Gely-Pernot, A, Kervarrec, C, Boudjema, M, Becker, E, Khil, P, et al.. Exposure to the widely used herbicide atrazine results in deregulation of global tissue-specific RNA transcription in the third generation and is associated with a global decrease of histone trimethylation in mice. Nucleic Acids Res 2016;44:9784–802. https://doi.org/10.1093/nar/gkw840.Search in Google Scholar PubMed PubMed Central

67. Sánchez, OF, Lin, L, Bryan, CJ, Xie, J, Freeman, JL, Yuan, C. Profiling epigenetic changes in human cell line induced by atrazine exposure. Environ Pollut 2020;258:113712. https://doi.org/10.1016/j.envpol.2019.113712.Search in Google Scholar PubMed

68. Xie, J, Lin, L, Sánchez, OF, Bryan, C, Freeman, JL, Yuan, C. Pre-differentiation exposure to low-dose of atrazine results in persistent phenotypic changes in human neuronal cell lines. Environ Pollut 2021;271:116379. https://doi.org/10.1016/j.envpol.2020.116379.Search in Google Scholar PubMed

69. Cai, Y, Zhang, Y, Loh, YP, Tng, JQ, Lim, MC, Cao, Z, et al.. H3K27me3-rich genomic regions can function as silencers to repress gene expression via chromatin interactions. Nat Commun 2021;12:719. https://doi.org/10.1038/s41467-021-20940-y.Search in Google Scholar PubMed PubMed Central

70. Sánchez, OF, Mendonca, A, Min, A, Liu, J, Yuan, C. Monitoring histone methylation (H3K9me3) changes in live cells. ACS Omega 2019;4:13250–9. https://doi.org/10.1021/acsomega.9b01413.Search in Google Scholar PubMed PubMed Central

71. Zhou, M, Yan, JQ, Chen, QX, Yang, YZ, Li, YL, Ren, YX, et al.. Association of H3K9me3 with breast cancer prognosis by estrogen receptor status. Clin Epigenetics 2022;14:135. https://doi.org/10.1186/s13148-022-01363-y.Search in Google Scholar PubMed PubMed Central

72. Thorson, JLM, Beck, D, Ben Maamar, M, Nilsson, EE, McBirney, M, Skinner, MK. Epigenome-wide association study for atrazine induced transgenerational DNA methylation and histone retention sperm epigenetic biomarkers for disease. PLoS One 2020;15:e0239380. https://doi.org/10.1371/journal.pone.0239380.Search in Google Scholar PubMed PubMed Central

73. Beck, D, Nilsson, EE, Ben Maamar, M, Skinner, MK. Environmental induced transgenerational inheritance impacts systems epigenetics in disease etiology. Sci Rep 2022;12:5452. https://doi.org/10.1038/s41598-022-09336-0.Search in Google Scholar PubMed PubMed Central

74. Hao, N, Xin, H, Shi, X, Xin, J, Zhang, H, Guo, S, et al.. Paternal reprogramming-escape histone H3K4me3 marks located within promoters of RNA splicing genes. Bioinformatics 2021;37:1039–44. https://doi.org/10.1093/bioinformatics/btaa920.Search in Google Scholar PubMed PubMed Central

75. Hoang, TT, Qi, C, Paul, KC, Lee, M, White, JD, Richards, M, et al.. Epigenome-wide DNA methylation and pesticide use in the agricultural lung health study. Environ Health Perspect 2021;129:97008. https://doi.org/10.1289/ehp8928.Search in Google Scholar

76. Almberg, KS, Turyk, ME, Jones, RM, Rankin, K, Freels, S, Stayner, LT. Atrazine contamination of drinking water and adverse birth outcomes in community water systems with elevated atrazine in Ohio, 2006–2008. Int J Environ Res Publ Health 2018;15:1889. https://doi.org/10.3390/ijerph15091889.Search in Google Scholar PubMed PubMed Central

77. Mohd, GR, Nik Yusoff, NR, Abdul Halim, NS, Wahab, IRA, Ab Latif, N, Hasmoni, SH, et al.. Health effects of herbicides and its current removal strategies. Bioengineered 2023;14:2259526. https://doi.org/10.1080/21655979.2023.2259526.Search in Google Scholar PubMed PubMed Central

78. Rostami, S, Jafari, S, Moeini, Z, Jaskulak, M, Keshtgar, L, Badeenezhad, A, et al.. Current methods and technologies for degradation of atrazine in contaminated soil and water: a review. Environ Technol Innovat 2021;24:102019. https://doi.org/10.1016/j.eti.2021.102019.Search in Google Scholar

79. Abarikwu, SO, Costa, GMJ, de Lima, EMLN, Lacerda, S, de França, LR. Atrazine impairs testicular function in BalB/c mice by affecting Leydig cells. Toxicology 2021;455:152761. https://doi.org/10.1016/j.tox.2021.152761.Search in Google Scholar PubMed

80. Pogrmic-Majkic, K, Samardzija, D, Stojkov-Mimic, N, Vukosavljevic, J, Trninic-Pjevic, A, Kopitovic, V, et al.. Atrazine suppresses FSH-induced steroidogenesis and LH-dependent expression of ovulatory genes through PDE-cAMP signaling pathway in human cumulus granulosa cells. Mol Cell Endocrinol 2018;461:79–88. https://doi.org/10.1016/j.mce.2017.08.015.Search in Google Scholar PubMed

Received: 2024-06-15
Accepted: 2024-08-22
Published Online: 2024-09-17
Published in Print: 2025-06-26

© 2024 Walter de Gruyter GmbH, Berlin/Boston

Articles in the same Issue

  1. Frontmatter
  2. Reviews
  3. Analytical methods, source, concentration, and human risks of microplastics: a review
  4. Solid fuel use and low birth weight: a systematic review and meta-analysis
  5. The human health effects of unconventional oil and gas (UOG) chemical exposures: a scoping review of the toxicological literature
  6. WHO to build neglect of RF-EMF exposure hazards on flawed EHC reviews? Case study demonstrates how “no hazards” conclusion is drawn from data showing hazards
  7. The role of environmental pollution in the development of pulmonary exacerbations in cystic fibrosis: a narrative review
  8. Semi-IPN polysaccharide-based hydrogels for effective removal of heavy metal ions and dyes from wastewater: a comprehensive investigation of performance and adsorption mechanism
  9. A structured review of the associations between breast cancer and exposures to selected organic solvents
  10. A review of the potential adverse health impacts of atrazine in humans
  11. Comprehensive approach to clinical decision-making strategy, illustrated by the Gulf War
  12. A systematic review and quality assessment of estimated daily intake of microplastics through food
  13. Adapting to heat-health vulnerability in temperate climates: current adaptation and mitigation responses and future predictions in Aotearoa New Zealand
  14. Evaluation of the impact of environmental pollutants on the sex ratio: a systematic review
  15. A critical review on the toxicological and epidemiological evidence integration for assessing human health risks to environmental chemical exposures
  16. The association between screen exposure and autism spectrum disorder in children: meta-analysis
  17. The association between maternal perfluoroalkylated substances exposure and neonatal birth weight: a system review and meta-analysis
  18. School built environment and children’s health: a scientometric analysis
  19. Letter to the Editors
  20. Underground power lines as a confounding factor in observational studies concerning magnetic fields and childhood leukemia
  21. A critical appraisal of the WHO 2024 systematic review of the effects of RF-EMF exposure on tinnitus, migraine/headache, and non-specific symptoms
Downloaded on 28.10.2025 from https://www.degruyterbrill.com/document/doi/10.1515/reveh-2024-0094/html
Scroll to top button