Epigenetic alterations induced by genotoxic occupational and environmental human chemical carcinogens: An update of a systematic literature review

Mutation Research/Reviews in Mutation Research - Tập 789 - Trang 108408 - 2022
Samantha Goodman1, Grace A. Chappell2, Kathryn Z. Guyton3, Igor P. Pogribny4, Ivan Rusyn1
1Department of Veterinary Integrative Biosciences, Texas A & M University, College Station, TX, USA
2ToxStrategies, Inc., Asheville, NC, USA
3International Agency for Research on Cancer, Lyon, France
4National Center for Toxicological Research, US Food and Drug Administration, Jefferson, AR, USA

Tóm tắt

Từ khóa


Tài liệu tham khảo

Samet, 2020, The IARC monographs: updated procedures for modern and transparent evidence synthesis in cancer hazard identification, J. Natl. Cancer Inst., 112, 30, 10.1093/jnci/djz169

Smith, 2016, Key characteristics of carcinogens as a basis for organizing data on mechanisms of carcinogenesis, Environ. Health Perspect., 124, 713, 10.1289/ehp.1509912

Chiu, 2018, Use of high-throughput in vitro toxicity screening data in cancer hazard evaluations by IARC Monograph Working Groups, ALTEX, 35, 51, 10.14573/altex.1703231

Smith, 2020, The key characteristics of carcinogens: relationship to the hallmarks of Cancer, relevant biomarkers, and assays to measure them, Cancer Epidemiol. Biomarkers Prev., 29, 1887, 10.1158/1055-9965.EPI-19-1346

La Merrill, 2020, Consensus on the key characteristics of endocrine-disrupting chemicals as a basis for hazard identification, Nat. Rev. Endocrinol., 16, 45, 10.1038/s41574-019-0273-8

Arzuaga, 2019, Proposed key characteristics of male reproductive toxicants as an approach for organizing and evaluating mechanistic evidence in human health hazard assessments, Environ. Health Perspect., 127, 65001, 10.1289/EHP5045

Luderer, 2019, Proposed key characteristics of female reproductive toxicants as an approach for organizing and evaluating mechanistic data in hazard assessment, Environ. Health Perspect., 127, 75001, 10.1289/EHP4971

Rusyn, 2021, Key characteristics of human hepatotoxicants as a basis for identification and characterization of the causes of liver toxicity, Hepatology, 74, 3486, 10.1002/hep.31999

Lind, 2021, Key characteristics of cardiovascular toxicants, Environ. Health Perspect., 129, 95001, 10.1289/EHP9321

Jones, 2007, The epigenomics of cancer, Cell, 128, 683, 10.1016/j.cell.2007.01.029

Chappell, 2016, Epigenetic alterations induced by genotoxic occupational and environmental human chemical carcinogens: a systematic literature review, Mutat. Res. Rev. Mutat. Res., 768, 27, 10.1016/j.mrrev.2016.03.004

Jones, 2002, The fundamental role of epigenetic events in cancer, Nat.Rev.Genet., 3, 415, 10.1038/nrg816

Feinberg, 2006, The epigenetic progenitor origin of human cancer, Nat. Rev. Genet., 7, 21, 10.1038/nrg1748

Martin, 2018, Environmental influences on the epigenome: exposure- associated DNA methylation in human populations, Annu. Rev. Public Health, 39, 309, 10.1146/annurev-publhealth-040617-014629

Inbar-Feigenberg, 2013, Basic concepts of epigenetics, Fertil. Steril., 99, 607, 10.1016/j.fertnstert.2013.01.117

Pogribny, 2013, Environmental toxicants, epigenetics, and cancer, Adv. Exp. Med. Biol., 754, 215, 10.1007/978-1-4419-9967-2_11

Herceg, 2013, Towards incorporating epigenetic mechanisms into carcinogen identification and evaluation, Carcinogenesis, 34, 1955, 10.1093/carcin/bgt212

Shapiro, 2018, Software tools to facilitate systematic review used for cancer hazard identification, Environ. Health Perspect., 126, 10.1289/EHP4224

Chen, 2006, Establishment and maintenance of DNA methylation patterns in mammals, Curr. Top. Microbiol. Immunol., 301, 179

Wu, 2017, TET-mediated active DNA demethylation: mechanism, function and beyond, Nat. Rev. Genet., 18, 517, 10.1038/nrg.2017.33

Ito, 2015, Epigenetic modifications in DNA could mimic oxidative DNA damage: a double-edged sword, DNA Repair (Amst), 32, 52, 10.1016/j.dnarep.2015.04.013

Bird, 1986, CpG-rich islands and the function of DNA methylation, Nature, 321, 209, 10.1038/321209a0

Mohandas, 1981, Reactivation of an inactive human X chromosome: evidence for X inactivation by DNA methylation, Science, 211, 393, 10.1126/science.6164095

Gartler, 1983, Mammalian X-chromosome inactivation, Annu. Rev. Genet., 17, 155, 10.1146/annurev.ge.17.120183.001103

Reik, 1987, Genomic imprinting determines methylation of parental alleles in transgenic mice, Nature, 328, 248, 10.1038/328248a0

Pan, 2018, DNA methylation profiles in cancer diagnosis and therapeutics, Clin. Exp. Med., 18, 1, 10.1007/s10238-017-0467-0

Man, 2015, Suppression of SOX7 by DNA methylation and its tumor suppressor function in acute myeloid leukemia, Blood, 125, 3928, 10.1182/blood-2014-06-580993

Min, 2016, Epigenetic silencing of the putative tumor suppressor gene GLDC (Glycine dehydrogenase) in gastric carcinoma, Anticancer Res., 36, 179

Gaudet, 2003, Induction of tumors in mice by genomic hypomethylation, Science, 300, 489, 10.1126/science.1083558

Baylin, 1998, Alterations in DNA methylation: a fundamental aspect of neoplasia, Adv. Cancer Res., 72, 141, 10.1016/S0065-230X(08)60702-2

Koch, 2018, Analysis of DNA methylation in cancer: location revisited, Nat. Rev. Clin. Oncol., 15, 459, 10.1038/s41571-018-0004-4

Gonzalgo, 1998, The role of DNA methylation in expression of the p19/p16 locus in human bladder cancer cell lines, Cancer Res., 58, 1245

Homma, 2006, Spreading of methylation within RUNX3 CpG island in gastric cancer, Cancer Sci., 97, 51, 10.1111/j.1349-7006.2005.00133.x

Deng, 1999, Methylation of CpG in a small region of the hMLH1 promoter invariably correlates with the absence of gene expression, Cancer Res., 59, 2029

Licchesi, 2010, Transcriptional regulation of Wnt inhibitory factor-1 by Miz-1/c-Myc, Oncogene, 29, 5923, 10.1038/onc.2010.322

Yoshikawa, 2001, SOCS-1, a negative regulator of the JAK/STAT pathway, is silenced by methylation in human hepatocellular carcinoma and shows growth-suppression activity, Nat. Genet., 28, 29, 10.1038/ng0501-29

Michaud, 2020, DNA methylation-derived immune cell profiles, CpG markers of inflammation, and pancreatic cancer risk, Cancer Epidemiol. Biomarkers Prev., 29, 1577, 10.1158/1055-9965.EPI-20-0378

Gouil, 2019, Latest techniques to study DNA methylation, Essays Biochem., 63, 639, 10.1042/EBC20190027

Kurdyukov, 2016, DNA methylation analysis: choosing the right method, Biology (Basel), 5, 3

Olkhov-Mitsel, 2012, Strategies for discovery and validation of methylated and hydroxymethylated DNA biomarkers, Cancer Med., 1, 237, 10.1002/cam4.22

Yong, 2016, Profiling genome-wide DNA methylation, Epigenetics Chromatin, 9, 26, 10.1186/s13072-016-0075-3

Audia, 2016, Histone modifications and cancer, Cold Spring Harb. Perspect. Biol., 8, 10.1101/cshperspect.a019521

Saha, 2020, Histone modifications and other facets of epigenetic regulation in trypanosomatids: leaving their mark, MBio, 11, e01079, 10.1128/mBio.01079-20

Fujisawa, 2017, Functions of bromodomain-containing proteins and their roles in homeostasis and cancer, Nat. Rev. Mol. Cell Biol., 18, 246, 10.1038/nrm.2016.143

Josling, 2012, The role of bromodomain proteins in regulating gene expression, Genes (Basel), 3, 320, 10.3390/genes3020320

Eissenberg, 2010, Histone H3 lysine 4 (H3K4) methylation in development and differentiation, Dev. Biol. (Basel), 339, 240, 10.1016/j.ydbio.2009.08.017

Kouzarides, 2007, Chromatin modifications and their function, Cell, 128, 693, 10.1016/j.cell.2007.02.005

Herz, 2013, SET for life: biochemical activities and biological functions of SET domain-containing proteins, Trends Biochem. Sci., 38, 621, 10.1016/j.tibs.2013.09.004

Seligson, 2005, Global histone modification patterns predict risk of prostate cancer recurrence, Nature, 435, 1262, 10.1038/nature03672

Bannister, 2011, Regulation of chromatin by histone modifications, Cell Res., 21, 381, 10.1038/cr.2011.22

Venkatesh, 2015, Histone exchange, chromatin structure and the regulation of transcription, Nat. Rev. Mol. Cell Biol., 16, 178, 10.1038/nrm3941

Waters, 2015, Histone modification and chromatin remodeling during NER, DNA Repair (Amst), 36, 105, 10.1016/j.dnarep.2015.09.013

O’Hagan, 2014, Chromatin modifications during repair of environmental exposure-induced DNA damage: a potential mechanism for stable epigenetic alterations, Environ. Mol. Mutagen., 55, 278, 10.1002/em.21830

Pierozan, 2020, Perfluorooctane sulfonate (PFOS) and perfluorooctanoic acid (PFOA) induce epigenetic alterations and promote human breast cell carcinogenesis in vitro, Arch. Toxicol., 94, 3893, 10.1007/s00204-020-02848-6

Costa, 2008, Non-coding RNAs, epigenetics and complexity, Gene, 410, 9, 10.1016/j.gene.2007.12.008

Amaral, 2008, The eukaryotic genome as an RNA machine, Science, 319, 1787, 10.1126/science.1155472

Ghildiyal, 2009, Small silencing RNAs: an expanding universe, Nat. Rev. Genet., 10, 94, 10.1038/nrg2504

Ruvkun, 2001, Molecular biology. Glimpses of a tiny RNA world, Science, 294, 797, 10.1126/science.1066315

Mishra, 2016, Exploring miRNA based approaches in cancer diagnostics and therapeutics, Crit. Rev. Oncol. Hematol., 98, 12, 10.1016/j.critrevonc.2015.10.003

Lee, 2009, MicroRNAs in cancer, Annu. Rev. Pathol., 4, 199, 10.1146/annurev.pathol.4.110807.092222

Garzon, 2006, MicroRNA expression and function in cancer, Trends Mol. Med., 12, 580, 10.1016/j.molmed.2006.10.006

Jonas, 2015, Towards a molecular understanding of microRNA-mediated gene silencing, Nat. Rev. Genet., 16, 421, 10.1038/nrg3965

Paraskevopoulou, 2016, Analyzing MiRNA-LncRNA interactions, Methods Mol. Biol., 1402, 271, 10.1007/978-1-4939-3378-5_21

Huarte, 2015, The emerging role of lncRNAs in cancer, Nat. Med., 21, 1253, 10.1038/nm.3981

Wang, 2011, Molecular mechanisms of long noncoding RNAs, Mol. Cell, 43, 904, 10.1016/j.molcel.2011.08.018

Zhang, 2018, The application of lncRNAs in cancer treatment and diagnosis, Recent Pat. Anticancer Drug Discov., 13, 292, 10.2174/1574892813666180226121819

Rathinasamy, 2018, Role of lncRNAs in the cancer development and progression and their regulation by various phytochemicals, Biomed. Pharmacother., 102, 242, 10.1016/j.biopha.2018.03.077

Zhao, 2016, Long noncoding RNAs in DNA methylation: new players stepping into the old game, Cell Biosci., 6, 45, 10.1186/s13578-016-0109-3

Jin, 2016, Cigarette smoking, BPDE-DNA adducts, and aberrant promoter methylations of tumor suppressor genes (TSGs) in NSCLC from Chinese population, Cancer Invest., 34, 173, 10.3109/07357907.2016.1156689

Jin, 2017, Tobacco-specific carcinogens induce hypermethylation, DNA adducts, and DNA damage in bladder Cancer, Cancer Prev. Res. (Phila), 10, 588, 10.1158/1940-6207.CAPR-17-0198

Jiang, 2017, Air pollution and DNA methylation alterations in lung cancer: a systematic and comparative study, Oncotarget, 8, 1369, 10.18632/oncotarget.13622

He, 2017, TRIM36 hypermethylation is involved in polycyclic aromatic hydrocarbons-induced cell transformation, Environ Pollut, 225, 93, 10.1016/j.envpol.2017.03.001

Xia, 2016, Repression of biotin-related proteins by Benzo[a]Pyrene-Induced epigenetic modifications in human bronchial epithelial cells, Int. J. Toxicol., 35, 336, 10.1177/1091581816637071

Bhargava, 2020, Mapping the mitochondrial regulation of epigenetic modifications in association with carcinogenic and noncarcinogenic polycyclic aromatic hydrocarbon exposure, Int. J. Toxicol., 39, 465, 10.1177/1091581820932875

Bojang, 2018, Epigenetic reactivation of LINE-1 retrotransposon disrupts NuRD corepressor functions and induces oncogenic transformation in human bronchial epithelial cells, Mol. Oncol., 12, 1342, 10.1002/1878-0261.12329

Zeng, 2021, Poly(ADP-ribose) glycohydrolase silencing-mediated H2B expression inhibits benzo(a)pyrene-induced carcinogenesis, Environ. Toxicol., 36, 291, 10.1002/tox.23034

Zeng, 2018, Poly (ADP-ribose) glycohydrolase silencing-mediated maintenance of H2A and downregulation of H2AK9me protect human bronchial epithelial cells from benzo(a)pyrene-induced carcinogenesis, Toxicol. Lett., 295, 270, 10.1016/j.toxlet.2018.07.003

Li, 2020, MicroRNA expression profiling and the role of ALCAM modulating tumor growth and metastasis in benzo[a]pyrene-transformed 16HBE cells, Toxicology, 442, 10.1016/j.tox.2020.152539

Malik, 2018, Mechanistic evidence that benzo[a]pyrene promotes an inflammatory microenvironment that drives the metastatic potential of human mammary cells, Arch. Toxicol., 92, 3223, 10.1007/s00204-018-2291-z

Malik, 2019, Interleukin-6 selectively induces drug metabolism to potentiate the genotoxicity of dietary carcinogens in mammary cells, Arch. Toxicol., 93, 3005, 10.1007/s00204-019-02558-8

Chanyshev, 2019, Effect of benzo[a]pyrene on the expression of miR-126, miR-190a and their target genes EGFL7, TP53INP1 and PHLPP1 in primary endometrial cells, J. Biochem. Mol. Toxicol., 33, 10.1002/jbt.22314

Wu, 2020, Benzo(a)pyrene regulated A549 cell migration, invasion and epithelial-mesenchymal transition by up-regulating long non-coding RNA linc00673, Toxicol. Lett., 320, 37, 10.1016/j.toxlet.2019.11.024

Fu, 2018, LncRNA H19 interacts with S-adenosylhomocysteine hydrolase to regulate LINE-1 Methylation in human lung-derived cells exposed to Benzo[a]pyrene, Chemosphere, 207, 84, 10.1016/j.chemosphere.2018.05.048

Liu, 2016, STAT3 methylation in white blood cells as a novel sensitive biomarker for the toxic effect of low-dose benzene exposure, Toxicol. Res. (Camb), 5, 800, 10.1039/C5TX00445D

Wang, 2020, [Study of methylation of mitochondrial MT-COI of benzene poisoning], Zhonghua Lao Dong Wei Sheng Zhi Ye Bing Za Zhi, 38, 664

Jamebozorgi, 2018, Aberrant DNA methylation of two tumor suppressor genes, p14(ARF) and p15(INK4b), after chronic occupational exposure to Low Level of benzene, Int. J. Occup. Environ. Med., 9, 145, 10.15171/ijoem.2018.1317

Silvestre, 2020, Hypermethylation in gene promoters are induced by chronic exposure to benzene, toluene, ethylbenzene and xylenes, Pak. J. Biol. Sci., 23, 518, 10.3923/pjbs.2020.518.525

Ren, 2020, Promoter hypermethylation in CSF3R induces peripheral neutrophil reduction in benzene-exposure poisoning, Environ. Mol. Mutagen., 61, 786, 10.1002/em.22382

Zheng, 2017, Association between promoter methylation of gene ERCC3 and benzene hematotoxicity, Int. J. Environ. Res. Public Health, 14, 921, 10.3390/ijerph14080921

Ren, 2019, The prevalence and persistence of aberrant promoter DNA methylation in benzene-exposed Chinese workers, PLoS One, 14, 10.1371/journal.pone.0220500

Zhang, 2017, Do mutations in DNMT3A/3B affect global DNA hypomethylation among benzene-exposed workers in Southeast China?: effects of mutations in DNMT3A/3B on global DNA hypomethylation, Environ. Mol. Mutagen., 58, 678, 10.1002/em.22136

Zhang, 2020, Up-regulation of DNMT3b contributes to HOTAIRM1 silencing via DNA hypermethylation in cells transformed by long-term exposure to hydroquinone and workers exposed to benzene, Toxicol. Lett., 322, 12, 10.1016/j.toxlet.2019.12.028

Yu, 2019, Benzene metabolite 1,2,4-benzenetriol changes DNA methylation and histone acetylation of erythroid-specific genes in K562 cells, Arch. Toxicol., 93, 137, 10.1007/s00204-018-2333-6

Yu, 2017, Changes in DNA methylation of erythroid-specific genes in K562 cells exposed to catechol in long term, Toxicol. In Vitro, 43, 21, 10.1016/j.tiv.2017.05.019

Tang, 2016, Long-term exposure of K562 cells to benzene metabolites inhibited erythroid differentiation and elevated methylation in erythroid specific genes, Toxicol. Res. (Camb), 5, 1284, 10.1039/C6TX00143B

Chen, 2016, Histone deacetylase inhibitors trichostatin a and MCP30 relieve benzene-induced hematotoxicity via restoring topoisomerase IIalpha, PLoS One, 11

Li, 2018, Enhanced H3K4me3 modifications are involved in the transactivation of DNA damage responsive genes in workers exposed to low-level benzene, Environ Pollut, 234, 127, 10.1016/j.envpol.2017.11.042

Song, 2015, Blood miRNAs as sensitive and specific biological indicators of environmental and occupational exposure to volatile organic compound (VOC), Int. J. Hyg. Environ. Health, 218, 590, 10.1016/j.ijheh.2015.06.002

Hu, 2016, Overexpression of miR-221 in peripheral blood lymphocytes in petrol station attendants: a population based cross-sectional study in southern China, Chemosphere, 149, 8, 10.1016/j.chemosphere.2016.01.083

Liu, 2016, Analysis of plasma microRNA expression profiles in a Chinese population occupationally exposed to benzene and in a population with chronic benzene poisoning, J. Thorac. Dis., 8, 403, 10.21037/jtd.2016.02.56

Jiang, 2020, Exosomal miR-221 derived from hydroquinone-transformed malignant human bronchial epithelial cells is involved in cell viability of recipient cells, J. Appl. Toxicol., 40, 224, 10.1002/jat.3898

Liang, 2018, Down-regulation of miRNA-451a and miRNA-486-5p involved in benzene-induced inhibition on erythroid cell differentiation in vitro and in vivo, Arch. Toxicol., 92, 259, 10.1007/s00204-017-2033-7

Ma, 2020, MiR-486-5p-directed MAGI1/Rap1/RASSF5 signaling pathway contributes to hydroquinone-induced inhibition of erythroid differentiation in K562 cells, Toxicol. In Vitro, 66, 10.1016/j.tiv.2020.104830

Gui, 2019, Poly(ADP-ribose) polymerase-1 promotes expression of miR-155 by the up-regulation of methyl-CpG binding domain protein 2 in TK6 cells exposed to hydroquinone, Toxicol. In Vitro, 55, 51, 10.1016/j.tiv.2018.11.007

Luo, 2018, miR-7-5p overexpression suppresses cell proliferation and promotes apoptosis through inhibiting the ability of DNA damage repair of PARP-1 and BRCA1 in TK6 cells exposed to hydroquinone, Chem. Biol. Interact., 283, 84, 10.1016/j.cbi.2018.01.019

Wang, 2021, Changes in miR-222 expression, DNA repair capacity, and MDM2-p53 axis in association with low-dose benzene genotoxicity and hematotoxicity, Sci. Total Environ., 765, 142740, 10.1016/j.scitotenv.2020.142740

Chen, 2019, lncRNAVNN3 mediated benzene-induced hematotoxicity through promoting autophagy and apoptosis, Ecotoxicol. Environ. Saf., 185, 10.1016/j.ecoenv.2019.109672

Sun, 2017, Benzene and its metabolite decreases cell proliferation via LncRNA-OBFC2A-mediated anti-proliferation effect involving NOTCH1 and KLF15, Oncotarget, 8, 40857, 10.18632/oncotarget.16588

Guo, 2020, LncRNA-OBFC2A targeted to Smad3 regulated Cyclin D1 influences cell cycle arrest induced by 1,4-benzoquinone, Toxicol. Lett., 332, 74, 10.1016/j.toxlet.2020.07.004

Yuan, 2020, Regulatory loop between lncRNA FAS-AS1 and DNMT3b controls FAS expression in hydroquinone-treated TK6 cells and benzene-exposed workers, Environ Pollut, 261, 10.1016/j.envpol.2020.114147

Rieswijk, 2016, Aflatoxin B1 induces persistent epigenomic effects in primary human hepatocytes associated with hepatocellular carcinoma, Toxicology, 350-352, 31, 10.1016/j.tox.2016.05.002

Tryndyak, 2018, Effect of aflatoxin B1, benzo[a]pyrene, and methapyrilene on transcriptomic and epigenetic alterations in human liver HepaRG cells, Food Chem. Toxicol., 121, 214, 10.1016/j.fct.2018.08.034

Tryndyak, 2019, Gene expression and cytosine DNA methylation alterations in induced pluripotent stem-cell-derived human hepatocytes treated with low doses of chemical carcinogens, Arch. Toxicol., 93, 3335, 10.1007/s00204-019-02569-5

Zhang, 2020, Single-cell sequencing reveals novel mechanisms of Aflatoxin B1-induced hepatotoxicity in S phase-arrested L02 cells, Cell Biol. Toxicol., 36, 603, 10.1007/s10565-020-09547-z

Soni, 2021, Epigenetic alterations induced by aflatoxin B1: an in vitro and in vivo approach with emphasis on enhancer of zeste homologue-2/p21 axis, Sci. Total Environ., 762, 10.1016/j.scitotenv.2020.143175

Soni, 2018, Aflatoxin B1 induced multiple epigenetic modulators in human epithelial cell lines, Toxicon, 151, 119, 10.1016/j.toxicon.2018.07.011

Wu, 2017, Diagnostic and prognostic potential of serum microRNA-4651 for patients with hepatocellular carcinoma related to aflatoxin B1, Oncotarget, 8, 81235, 10.18632/oncotarget.16027

Livingstone, 2019, Serum miR-182 is a predictive biomarker for dichotomization of risk of hepatocellular carcinoma in rats, Mol. Carcinog., 58, 2017, 10.1002/mc.23093

Shirani, 2021, miR-155 influences cell-mediated immunity in Balb/c mice treated with aflatoxin M1, Drug Chem. Toxicol., 44, 39, 10.1080/01480545.2018.1556682

Zhang, 2019, Analysis of miRNA-mRNA regulatory network revealed key genes induced by aflatoxin B1 exposure in primary human hepatocytes, Mol. Genet. Genomic Med., 7, e971, 10.1002/mgg3.971

Liu, 2020, AFB1 induced transcriptional regulation related to apoptosis and lipid metabolism in liver of chicken, Toxins (Basel), 12, 290, 10.3390/toxins12050290

Liu, 2019, CYP1A1 methylation mediates the effect of smoking and occupational polycyclic aromatic hydrocarbons co-exposure on oxidative DNA damage among Chinese coke-oven workers, Environ. Health, 18, 69, 10.1186/s12940-019-0508-0

Yang, 2018, Urinary 1-hydroxypyrene and smoking are determinants of LINE-1 and AhRR promoter methylation in coke oven workers, Mutat. Res. Genet. Toxicol. Environ. Mutagen., 826, 33, 10.1016/j.mrgentox.2018.01.001

He, 2019, FLT1 hypermethylation is involved in polycyclic aromatic hydrocarbons-induced cell transformation, Environ Pollut, 252, 607, 10.1016/j.envpol.2019.05.137

Xing, 2020, Association between H3K36me3 modification and methylation of LINE-1 and MGMT in peripheral blood lymphocytes of PAH-exposed workers, Toxicol. Res. (Camb), 9, 661, 10.1093/toxres/tfaa074

Zhang, 2016, Specific histone modifications were associated with the PAH-induced DNA damage response in coke oven workers, Toxicol. Res. (Camb), 5, 1193, 10.1039/C6TX00112B

Zhang, 2021, Global H3K79 di-methylation mediates DNA damage response to PAH exposure in Chinese coke oven workers, Environ Pollut, 268, 10.1016/j.envpol.2020.115956

Huang, 2016, Polycyclic aromatic hydrocarbons-associated MicroRNAs and heart rate variability in coke oven workers, J. Occup. Environ. Med., 58, e24, 10.1097/JOM.0000000000000564

Gao, 2016, Specific long non-coding RNAs response to occupational PAHs exposure in coke oven workers, Toxicol. Rep., 3, 160, 10.1016/j.toxrep.2015.12.011

Simons, 2018, Sulfur mustard-induced epigenetic modifications over time - a pilot study, Toxicol. Lett., 293, 45, 10.1016/j.toxlet.2017.11.010

Khafaei, 2015, Evaluation of miR-9 and miR-143 expression in urine specimens of sulfur mustard exposed patients, Interdiscip. Toxicol., 8, 169, 10.1515/intox-2015-0026

Gharbi, 2018, MicroRNA expression in serum samples of sulfur mustard veterans as a diagnostic gateway to improve care, PLoS One, 13, 10.1371/journal.pone.0194530

Salimi, 2019, Expression of miR-15b-5p, miR-21-5p, and SMAD7 in lung tissue of sulfur mustard-exposed individuals with long-term pulmonary complications, Iran. J. Allergy Asthma Immunol., 18, 332

Rothmiller, 2018, Alteration of miRNA expression in a sulfur mustard resistant cell line, Toxicol. Lett., 293, 38, 10.1016/j.toxlet.2017.08.014

Rana, 2020, MicroRNA-mediated inflammation and coagulation effects in rats exposed to an inhaled analog of sulfur mustard, Ann. N. Y. Acad. Sci., 1479, 148, 10.1111/nyas.14416

Barbosa, 2019, Increase of global DNA methylation patterns in beauty salon workers exposed to low levels of formaldehyde, Environ. Sci. Pollut. Res. Int., 26, 1304, 10.1007/s11356-018-3674-7

Chen, 2017, Regulation of chromatin assembly and cell transformation by formaldehyde exposure in human cells, Environ. Health Perspect., 125, 10.1289/EHP1275

Lewis, 2019, Sex-specific differences in genotoxic and epigenetic effects of 1,3-butadiene among mouse tissues, Arch. Toxicol., 93, 791, 10.1007/s00204-018-2374-x

Lewis, 2019, Population-based analysis of DNA damage and epigenetic effects of 1,3-butadiene in the mouse, Chem. Res. Toxicol., 32, 887, 10.1021/acs.chemrestox.9b00035

Israel, 2018, Tissue- and strain-specific effects of a genotoxic carcinogen 1,3-butadiene on chromatin and transcription, Mamm. Genome, 29, 153, 10.1007/s00335-018-9739-6

Chappell, 2017, Variation in DNA-Damage responses to an inhalational carcinogen (1,3-Butadiene) in relation to strain-specific differences in chromatin accessibility and gene transcription profiles in C57BL/6J and CAST/EiJ mice, Environ. Health Perspect., 125, 10.1289/EHP1937

Qiu, 2019, Association between methylation of DNA damage response-related genes and DNA damage in hepatocytes of rats following subchronic exposure to vinyl chloride, Chemosphere, 227, 323, 10.1016/j.chemosphere.2019.04.058

Feng, 2017, Analysis of microRNA expression and micronuclei frequency in workers exposed to vinyl chloride monomer in China, Epigenomics, 9, 1093, 10.2217/epi-2017-0028

Sisto, 2019, Circulating microRNAs as potential biomarkers of occupational exposure to low dose organic solvents, Toxicol. Rep., 6, 126, 10.1016/j.toxrep.2019.01.001

Duan, 2020, Epigenetic-based biomarkers in the malignant transformation of BEAS-2B cells induced by coal tar pitch extract, Medicina (Kaunas), 57, 24, 10.3390/medicina57010024

Li, 2020, Identification and analysis of key lncRNAs in malignant-transformed BEAS-2B cells induced with coal tar pitch by microarray analysis, Environ. Toxicol. Pharmacol., 79, 10.1016/j.etap.2020.103376

Li, 2019, LncRNA-ENST00000501520 promotes the proliferation of malignant-transformed BEAS-2B cells induced with coal tar pitch mediated by target genes, Environ. Toxicol., 34, 869, 10.1002/tox.22759

Lin, 2020, 4-Aminobiphenyl inhibits the DNA homologous recombination repair in human liver cells: the role of miR-630 in downregulating RAD18 and MCM8, Toxicology, 440, 10.1016/j.tox.2020.152441

IARC Working Group on the Evaluation of Carcinogenic Risks to Humans, 2016, Outdoor air pollution, IARC Monogr. Eval. Carcinog. Risks Hum., 109, 9

Vos, 2021, Mitochondrial DNA methylation in placental tissue: a proof of concept study by means of prenatal environmental stressors, Epigenetics, 16, 121, 10.1080/15592294.2020.1790923

Wang, 2020, Genome-wide DNA methylation analysis reveals significant impact of long-term ambient air pollution exposure on biological functions related to mitochondria and immune response, Environ Pollut, 264, 10.1016/j.envpol.2020.114707

Eze, 2020, Genome-wide DNA methylation in peripheral blood and long-term exposure to source-specific transportation noise and air pollution: the SAPALDIA study, Environ. Health Perspect., 128, 67003, 10.1289/EHP6174

Wang, 2020, Long-term ambient fine particulate matter and DNA methylation in inflammation pathways: results from the Sister Study, Epigenetics, 15, 524, 10.1080/15592294.2019.1699894

Wang, 2020, DNA methylation of insulin-like growth factor 2 and H19 cluster in cord blood and prenatal air pollution exposure to fine particulate matter, Environ. Health, 19, 129, 10.1186/s12940-020-00677-9

Merid, 2021, Integration of gene expression and DNA methylation identifies epigenetically controlled modules related to PM2.5 exposure, Environ. Int., 146, 10.1016/j.envint.2020.106248

Feng, 2020, GPR61 methylation in cord blood: a potential target of prenatal exposure to air pollutants, Int. J. Environ. Health Res., 1

Gruzieva, 2019, Prenatal particulate air pollution and DNA methylation in newborns: an epigenome-wide meta-analysis, Environ. Health Perspect., 127, 57012, 10.1289/EHP4522

Callahan, 2018, Lifetime exposure to ambient air pollution and methylation of tumor suppressor genes in breast tumors, Environ. Res., 161, 418, 10.1016/j.envres.2017.11.040

Cantone, 2020, Particulate air pollution, clock gene methylation, and stroke: effects on stroke severity and disability, Int. J. Mol. Sci., 21, 10.3390/ijms21093090

Sun, 2020, Inflammatory cytokines and DNA methylation in healthy young adults exposure to fine particulate matter: a randomized, double-blind crossover trial of air filtration, J. Hazard. Mater., 398, 10.1016/j.jhazmat.2020.122817

Calderon-Garciduenas, 2020, Reduced repressive epigenetic marks, increased DNA damage and Alzheimer’s disease hallmarks in the brain of humans and mice exposed to particulate urban air pollution, Environ. Res., 183, 10.1016/j.envres.2020.109226

Vrijens, 2020, Association of prenatal exposure to ambient air pollution with circulating histone levels in maternal cord blood, JAMA Netw Open, 3, 10.1001/jamanetworkopen.2020.5156

Mancini, 2020, microRNA expression profiles and personal monitoring of exposure to particulate matter, Environ Pollut, 263, 10.1016/j.envpol.2020.114392

Tsamou, 2020, Prenatal particulate air pollution exposure and expression of the miR-17/92 cluster in cord blood: findings from the ENVIRONAGE birth cohort, Environ. Int., 142, 10.1016/j.envint.2020.105860

Li, 2020, Particulate matter air pollution and the expression of microRNAs and pro-inflammatory genes: association and mediation among children in Jinan, China, J. Hazard. Mater., 389, 10.1016/j.jhazmat.2019.121843

Chen, 2020, Effects of ambient ozone exposure on circulating extracellular vehicle microRNA levels in coronary artery disease patients, J Toxicol Environ Health A, 83, 351, 10.1080/15287394.2020.1762814

IARC Working Group on the Evaluation of Carcinogenic Risks to Humans, 2014, Diesel and gasoline engine exhausts and some nitroarenes, IARC Monogr. Eval. Carcinog. Risks Hum., 105, 9

Zhang, 2016, Associations between DNA methylation in DNA damage response-related genes and cytokinesis-block micronucleus cytome index in diesel engine exhaust-exposed workers, Arch. Toxicol., 90, 1997, 10.1007/s00204-015-1598-2

Jiang, 2014, Short-term diesel exhaust inhalation in a controlled human crossover study is associated with changes in DNA methylation of circulating mononuclear cells in asthmatics, Part. Fibre Toxicol., 11, 71, 10.1186/s12989-014-0071-3

Lei, 2018, Differentially DNA methylation changes induced in vitro by traffic-derived nanoparticulate matter, Toxicology, 395, 54, 10.1016/j.tox.2017.11.005

Zhang, 2018, Diesel exhaust and house dust mite allergen lead to common changes in the airway methylome and hydroxymethylome, Environ. Epigenet., 4, dvy020, 10.1093/eep/dvy020

Clifford, 2017, Inhalation of diesel exhaust and allergen alters human bronchial epithelium DNA methylation, J. Allergy Clin. Immunol., 139, 112, 10.1016/j.jaci.2016.03.046

Li, 2021, Predominant DNMT and TET mediate effects of allergen on the human bronchial epithelium in a controlled air pollution exposure study, J. Allergy Clin. Immunol., 147, 1671, 10.1016/j.jaci.2020.08.044

Tachibana, 2015, Prenatal diesel exhaust exposure disrupts the DNA methylation profile in the brain of mouse offspring, J. Toxicol. Sci., 40, 1, 10.2131/jts.40.1

Goodson, 2017, In utero exposure to diesel exhaust particulates is associated with an altered cardiac transcriptional response to transverse aortic constriction and altered DNA methylation, FASEB J., 31, 4935, 10.1096/fj.201700032R

Goodson, 2019, In utero exposure to diesel exhaust is associated with alterations in neonatal cardiomyocyte transcription, DNA methylation and metabolic perturbation, Part. Fibre Toxicol., 16, 17, 10.1186/s12989-019-0301-9

Belinsky, 2002, Aberrant CpG island methylation of the p16(INK4a) and estrogen receptor genes in rat lung tumors induced by particulate carcinogens, Carcinogenesis, 23, 335, 10.1093/carcin/23.2.335

Cao, 2007, COX-2 expression induced by diesel particles involves chromatin modification and degradation of HDAC1, Am. J. Respir. Cell Mol. Biol., 37, 232, 10.1165/rcmb.2006-0449OC

Jardim, 2009, Disruption of microRNA expression in human airway cells by diesel exhaust particles is linked to tumorigenesis-associated pathways, Environ. Health Perspect., 117, 1745, 10.1289/ehp.0900756

Yadav, 2017, MIR155 regulation of Ubiquilin1 and Ubiquilin2: implications in cellular protection and tumorigenesis, Neoplasia, 19, 321, 10.1016/j.neo.2017.02.001

Bleck, 2013, MicroRNA-375 regulation of thymic stromal lymphopoietin by diesel exhaust particles and ambient particulate matter in human bronchial epithelial cells, J. Immunol., 190, 3757, 10.4049/jimmunol.1201165

Zhou, 2015, Effects of diesel exhaust particles on microRNA-21 in human bronchial epithelial cells and potential carcinogenic mechanisms, Mol. Med. Rep., 12, 2329, 10.3892/mmr.2015.3655

Zhang, 2020, MicroRNA-382-5p is involved in pulmonary inflammation induced by fine particulate matter exposure, Environ Pollut, 262, 10.1016/j.envpol.2020.114278

Rider, 2016, Controlled diesel exhaust and allergen coexposure modulates microRNA and gene expression in humans: effects on inflammatory lung markers, J. Allergy Clin. Immunol., 138, 1690, 10.1016/j.jaci.2016.02.038

Yamamoto, 2013, MicroRNA expression in response to controlled exposure to diesel exhaust: attenuation by the antioxidant N-acetylcysteine in a randomized crossover study, Environ. Health Perspect., 121, 670, 10.1289/ehp.1205963

Rynning, 2019, Bulky DNA adducts, microRNA profiles, and lipid biomarkers in Norwegian tunnel finishing workers occupationally exposed to diesel exhaust, Occup. Environ. Med., 76, 10, 10.1136/oemed-2018-105445

2018, IARC working group on the evaluation of carcinogenic risks to humans

Agrahari, 2019, Overexpression of cerebral cytochrome P450s in prenatally exposed offspring modify the toxicity of lindane in rechallenged offspring, Toxicol. Appl. Pharmacol., 371, 20, 10.1016/j.taap.2019.03.022

Huang, 2019, Maternal pentachlorophenol exposure induces developmental toxicity mediated by autophagy on pregnancy mice, Ecotoxicol. Environ. Saf., 169, 829, 10.1016/j.ecoenv.2018.11.073

IARC working Group on the Evaluation of Carcinogenic Risks to Humans, 2016, Polychlorinated biphenyls and polybrominated biphenyls, IARC Monogr. Eval. Carcinog. Risks Hum., 107, 9

Rusiecki, 2008, Global DNA hypomethylation is associated with high serum-persistent organic pollutants in Greenlandic Inuit, Environ. Health Perspect., 116, 1547, 10.1289/ehp.11338

Itoh, 2014, Association between serum organochlorines and global methylation level of leukocyte DNA among Japanese women: a cross-sectional study, Sci. Total Environ., 490, 603, 10.1016/j.scitotenv.2014.05.035

Lind, 2013, Global DNA hypermethylation is associated with high serum levels of persistent organic pollutants in an elderly population, Environ. Int., 59, 456, 10.1016/j.envint.2013.07.008

van den Dungen, 2017, Association between DNA methylation profiles in leukocytes and serum levels of persistent organic pollutants in Dutch men, Environ. Epigenet., 3, dvx001, 10.1093/eep/dvx001

Lee, 2017, Association between serum persistent organic pollutants and DNA methylation in Korean adults, Environ. Res., 158, 333, 10.1016/j.envres.2017.06.017

Leung, 2018, Identification of sex-specific DNA methylation changes driven by specific chemicals in cord blood in a Faroese birth cohort, Epigenetics, 13, 290, 10.1080/15592294.2018.1445901

Consales, 2016, Exposure to persistent organic pollutants and sperm DNA methylation changes in Arctic and European populations, Environ. Mol. Mutagen., 57, 200, 10.1002/em.21994

Ouidir, 2020, Concentrations of persistent organic pollutants in maternal plasma and epigenome-wide placental DNA methylation, Clin. Epigenetics, 12, 103, 10.1186/s13148-020-00894-6

Kobayashi, 2017, Gender-specific association of exposure to non-dioxin-like polychlorinated biphenyls during pregnancy with methylation levels of H19 and long interspersed nuclear element-1 in cord blood in the Hokkaido study, Toxicology, 390, 135, 10.1016/j.tox.2017.08.010

Kim, 2018, Prenatal exposure to persistent organic pollutants and methylation of LINE-1 and imprinted genes in placenta: a CHECK cohort study, Environ. Int., 119, 398, 10.1016/j.envint.2018.06.039

Su, 2019, Perinatal polychlorinated biphenyls and polychlorinated dibenzofurans exposure are associated with DNA methylation changes lasting to early adulthood: findings from Yucheng second generation, Environ. Res., 170, 481, 10.1016/j.envres.2019.01.001

Cui, 2016, Epigenetic toxicity of trichloroethylene: a single-molecule perspective, Toxicol. Res. (Camb), 5, 641, 10.1039/C5TX00454C

Georgiadis, 2019, DNA methylation profiling implicates exposure to PCBs in the pathogenesis of B-cell chronic lymphocytic leukemia, Environ. Int., 126, 24, 10.1016/j.envint.2019.01.068

Wang, 2019, Study on the exposure of polychlorinated biphenyl contamination and DNA methylation in male employees in an e-waste dismantling area in Tianjin, Zhonghua Yu Fang Yi Xue Za Zhi, 53, 376

Lee, 2018, Association of low-dose exposure to persistent organic pollutants with E-cadherin promoter methylation in healthy Koreans, Biomarkers, 23, 293, 10.1080/1354750X.2017.1417482

Eguchi, 2019, An altered DNA methylation status in the human umbilical cord is correlated with maternal exposure to polychlorinated biphenyls, Int. J. Environ. Res. Public Health, 16, 2786, 10.3390/ijerph16152786

Park, 2015, Persistent organic pollutants and promoter hypermethylation of the O(6)-methylguanine-DNA methyltransferase gene, Biomarkers, 20, 136, 10.3109/1354750X.2014.1002806

Formisano, 2011, The repressor element 1-silencing transcription factor is a novel molecular target for the neurotoxic effect of the polychlorinated biphenyl mixture aroclor 1254 in neuroblastoma SH-SY5Y cells, J. Pharmacol. Exp. Ther., 338, 997, 10.1124/jpet.111.181289

Casati, 2012, Polychlorinated biphenyls affect histone modification pattern in early development of rats: a role for androgen receptor-dependent modulation?, Epigenomics, 4, 101, 10.2217/epi.11.110

Cai, 2016, Polychlorinated biphenyls impair endometrial receptivity in vitro via regulating mir-30d expression and epithelial mesenchymal transition, Toxicology, 365, 25, 10.1016/j.tox.2016.07.017

Li, 2015, Exploring the associations between microRNA expression profiles and environmental pollutants in human placenta from the National Children’s Study (NCS), Epigenetics, 10, 793, 10.1080/15592294.2015.1066960

Krauskopf, 2017, MicroRNA profile for health risk assessment: environmental exposure to persistent organic pollutants strongly affects the human blood microRNA machinery, Sci. Rep., 7, 9262, 10.1038/s41598-017-10167-7

Wahlang, 2016, Polychlorinated biphenyl exposure alters the expression profile of microRNAs associated with vascular diseases, Toxicol. In Vitro, 35, 180, 10.1016/j.tiv.2016.06.001

Lesiak, 2014, The environmental neurotoxicant PCB 95 promotes synaptogenesis via ryanodine receptor-dependent miR132 upregulation, J. Neurosci., 34, 717, 10.1523/JNEUROSCI.2884-13.2014

Topper, 2015, Sexually dimorphic effects of gestational endocrine-disrupting chemicals on microRNA expression in the developing rat hypothalamus, Mol. Cell. Endocrinol., 414, 42, 10.1016/j.mce.2015.07.013

Shan, 2020, 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) and polychlorinated biphenyl coexposure alters the expression profile of MicroRNAs in the liver associated with atherosclerosis, Biomed Res. Int., 2020, 10.1155/2020/2652756

Zhu, 2012, Differential expression profile of MicroRNAs during differentiation of cardiomyocytes exposed to polychlorinated biphenyls, Int. J. Mol. Sci., 13, 15955, 10.3390/ijms131215955

Kim, 2010, Association of low-dose exposure to persistent organic pollutants with global DNA hypomethylation in healthy Koreans, Environ. Health Perspect., 118, 370, 10.1289/ehp.0901131

Pittman, 2020, Dioxin-like compound exposures and DNA methylation in the anniston community health survey phase II, Sci. Total Environ., 742, 10.1016/j.scitotenv.2020.140424

Vorrink, 2014, Epigenetic determinants of CYP1A1 induction by the aryl hydrocarbon receptor agonist 3,3’,4,4’,5-pentachlorobiphenyl (PCB 126), Int. J. Mol. Sci., 15, 13916, 10.3390/ijms150813916

Aluru, 2018, Role of DNA methylation in altered gene expression patterns in adult zebrafish (Danio rerio) exposed to 3, 3’, 4, 4’, 5-pentachlorobiphenyl (PCB 126), Environ. Epigenet., 4, dvy005, 10.1093/eep/dvy005

He, 2019, Effects of 2,3’,4,4’5-pentachlorobiphenyl exposure during pregnancy on epigenetic imprinting and maturation of offspring’s oocytes in mice, Arch. Toxicol., 93, 2575, 10.1007/s00204-019-02529-z

Qu, 2018, Effect of 2,3’,4,4’,5-Pentachlorobiphenyl exposure on endometrial receptivity and the methylation of HOXA10, Reprod. Sci., 25, 256, 10.1177/1933719117711258

Liu, 2015, Exposure to coplanar PCBs induces endothelial cell inflammation through epigenetic regulation of NF-kappaB subunit p65, Toxicol. Appl. Pharmacol., 289, 457, 10.1016/j.taap.2015.10.015

Liu, 2016, EGCG prevents PCB-126-induced endothelial cell inflammation via epigenetic modifications of NF-kappaB target genes in human endothelial cells, J. Nutr. Biochem., 28, 164, 10.1016/j.jnutbio.2015.10.003

Ovesen, 2011, Aryl hydrocarbon receptor ligands of widely different toxic equivalency factors induce similar histone marks in target gene chromatin, Toxicol. Sci., 121, 123, 10.1093/toxsci/kfr032

Guida, 2013, Association between exposure to dioxin-like polychlorinated biphenyls and miR-191 expression in human peripheral blood mononuclear cells, Mutat. Res., 753, 36, 10.1016/j.mrgentox.2012.12.018

Chen, 2020, Transcriptome analysis in normal human liver cells exposed to 2, 3, 3’, 4, 4’, 5 - Hexachlorobiphenyl (PCB 156), Chemosphere, 239, 10.1016/j.chemosphere.2019.124747

IARC Working Group on the Evaluation of Carcinogenic Risks to Humans, 2014, Trichloroethylene, tetrachloroethylene, and some other chlorinated agents, IARC Monogr. Eval. Carcinog. Risks Hum., 106, 1

Byrum, 2019, Continuous developmental and early life trichloroethylene exposure promoted DNA methylation alterations in polycomb protein binding sites in Effector/Memory CD4(+) t cells, Front. Immunol., 10, 2016, 10.3389/fimmu.2019.02016

Gilbert, 2017, Trichloroethylene-induced alterations in DNA methylation were enriched in polycomb protein binding sites in effector/memory CD4(+) T cells, Environ. Epigenet., 3, dvx013, 10.1093/eep/dvx013

Gilbert, 2016, Chronic exposure to water pollutant trichloroethylene increased epigenetic drift in CD4(+) T cells, Epigenomics, 8, 633, 10.2217/epi-2015-0018

Gilbert, 2016, Chronic exposure to trichloroethylene increases DNA methylation of the Ifng promoter in CD4(+) T cells, Toxicol. Lett., 260, 1, 10.1016/j.toxlet.2016.08.017

Jiang, 2014, Trichloroethylene-induced gene expression and DNA methylation changes in B6C3F1 mouse liver, PLoS One, 9, 10.1371/journal.pone.0116179

Tao, 1999, Effect of trichloroethylene on DNA methylation and expression of early-intermediate protooncogenes in the liver of B6C3F1 mice, J. Biochem. Mol. Toxicol., 13, 231, 10.1002/(SICI)1099-0461(1999)13:5<231::AID-JBT2>3.0.CO;2-F

Tao, 1998, Effect of dichloroacetic acid and trichloroacetic acid on DNA methylation in liver and tumors of female B6C3F1 mice, Toxicol. Sci., 43, 139, 10.1093/toxsci/43.2.139

Tao, 2004, Hypomethylation of DNA and the insulin-like growth factor-II gene in dichloroacetic and trichloroacetic acid-promoted mouse liver tumors, Toxicology, 196, 127, 10.1016/j.tox.2003.11.011

Tao, 2000, Hypomethylation and overexpression of c-jun and c-myc protooncogenes and increased DNA methyltransferase activity in dichloroacetic and trichloroacetic acid-promoted mouse liver tumors, Cancer Lett., 158, 185, 10.1016/S0304-3835(00)00518-8

Pereira, 2004, Prevention by methionine of dichloroacetic acid-induced liver cancer and DNA hypomethylation in mice, Toxicol. Sci., 77, 243, 10.1093/toxsci/kfh031

Tabish, 2012, Epigenetic factors in cancer risk: effect of chemical carcinogens on global DNA methylation pattern in human TK6 cells, PLoS One, 7, 10.1371/journal.pone.0034674

Zhang, 2014, Analysis of trichloroethylene-induced global DNA hypomethylation in hepatic L-02 cells by liquid chromatography-electrospray ionization tandem mass spectrometry, Biochem. Biophys. Res. Commun., 446, 590, 10.1016/j.bbrc.2014.03.015

Lai, 2019, DNA methyltransferase expression and DNA hypomethylation status in human hepatocytes following trichloroacetic acid exposure, Biochem. Biophys. Res. Commun., 511, 266, 10.1016/j.bbrc.2019.02.017

Ruan, 2018, Trichloroethylene-induced abnormal methylation on promoter region of SET in hepatic L-02 cells, Zhonghua Lao Dong Wei Sheng Zhi Ye Bing Za Zhi, 36, 165

Lu, 2018, SET promotes H2Ak9 acetylation by suppressing HDAC1 in trichloroethylene-induced hepatic cytotoxicity, Environ. Toxicol. Pharmacol., 59, 125, 10.1016/j.etap.2018.03.011

Deng, 2017, An investigation of trichloroethylene-induced effects on histone methylation in L-02 hepatic cells, Zhonghua Yu Fang Yi Xue Za Zhi, 51, 347

Xie, 2016, Effect of SET deficiency on the trichloroethylene-induced alteration of cell proliferation and cell apoptosis and DNA methylation in human hepatic L-02 cells, Zhonghua Lao Dong Wei Sheng Zhi Ye Bing Za Zhi, 34, 161

Lai, 2021, Specific epigenetic microenvironment and the regulation of tumor-related gene expression by trichloroethylene in human hepatocytes, Ecotoxicol. Environ. Saf., 208, 10.1016/j.ecoenv.2020.111453

Stermer, 2019, Trichloroethylene exposure alters dimethylated histone three lysine four in protein kinase A signaling pathway chromatin of rat spermdagger, Biol. Reprod., 101, 875, 10.1093/biolre/ioz155

Aoki, 2020, MicroRNA biomarkers for chemical hazard screening identified by RNA deep sequencing analysis in mouse embryonic stem cells, Toxicol. Appl. Pharmacol., 392, 10.1016/j.taap.2020.114929

Harting, 2016, The effect of dichloroacetate in canine prostate adenocarcinomas and transitional cell carcinomas in vitro, Int. J. Oncol., 49, 2341, 10.3892/ijo.2016.3720

Huang, 2020, Downregulation of miR-133a contributes to the cardiac developmental toxicity of trichloroethylene in zebrafish, Chemosphere, 251, 10.1016/j.chemosphere.2020.126610

Lee, 2019, Association between occupational exposure to trichloroethylene and serum levels of microRNAs: a cross-sectional molecular epidemiology study in China, Int. Arch. Occup. Environ. Health, 92, 1077, 10.1007/s00420-019-01448-x

Ren, 2019, Trichloroethylene-induced downregulation of miR-199b-5p contributes to SET-mediated apoptosis in hepatocytes, Cell Biol. Toxicol., 35, 565, 10.1007/s10565-019-09479-3

Jiang, 2017, The role of miR-182-5p in hepatocarcinogenesis of trichloroethylene in mice, Toxicol. Sci., 156, 208

Jiang, 2019, Role of miR-182-5p overexpression in trichloroethylene-induced abnormal cell cycle functions in human HepG2 cells, J Toxicol Environ Health A, 82, 920, 10.1080/15287394.2019.1666550

Harting, 2017, Dichloroacetate affects proliferation but not apoptosis in canine mammary cell lines, PLoS One, 12, 10.1371/journal.pone.0178744

2018, IARC working group on the evaluation of carcinogenic risks to humans

Li, 2015, Oxidative stress, telomere shortening, and DNA methylation in relation to low-to-moderate occupational exposure to welding fumes, Environ. Mol. Mutagen., 56, 684, 10.1002/em.21958

Hossain, 2015, Exposure to welding fumes is associated with hypomethylation of the F2RL3 gene: a cardiovascular disease marker, Occup. Environ. Med., 72, 845, 10.1136/oemed-2015-102884

Xu, 2017, Occupational exposure to particles and mitochondrial DNA - relevance for blood pressure, Environ. Health, 16, 22, 10.1186/s12940-017-0234-4

Lu, 2016, Short-term exposure to engineered nanomaterials affects cellular epigenome, Nanotoxicology, 10, 140

Shoeb, 2020, A possible relationship between telomere length and markers of neurodegeneration in rat brain after welding fume inhalation exposure, Environ. Res., 180, 10.1016/j.envres.2019.108900

Shoeb, 2017, Oxidative stress, DNA methylation, and telomere length changes in peripheral blood mononuclear cells after pulmonary exposure to metal-rich welding nanoparticles, NanoImpact, 5, 61, 10.1016/j.impact.2017.01.001

Amrani, 2020, Exposure to metal fumes and circulating miRNAs in Algerian welders, Int. Arch. Occup. Environ. Health, 93, 553, 10.1007/s00420-019-01509-1

Zhao, 2019, Epigenetic modifications of histones in cancer, Genome Biol., 20, 245, 10.1186/s13059-019-1870-5

Guyton, 2018, Application of the key characteristics of carcinogens in cancer hazard identification, Carcinogenesis, 39, 614, 10.1093/carcin/bgy031