Abstract
The field of epitranscriptomics encompasses the study of post‐transcriptional RNA modifications and their regulatory enzymes. Among the numerous RNA modifications, N 6‐methyladenosine (m6A) has been identified as the most common internal modification of messenger RNA (mRNA). Although m6A modifications were first discovered in the 1970s, advances in technology have revived interest in this field, driving an abundance of research into the role of RNA modifications in various biological processes, including cancer. As analogs to epigenetic modifications, RNA modifications also play an important role in carcinogenesis by regulating gene expression post‐transcriptionally. A growing body of evidence suggests that carcinogens can modulate RNA modifications to alter the expression of oncogenes or tumor suppressors during cellular transformation. Additionally, the expression and activity of the enzymes that regulate RNA modifications can be dysregulated and contribute to carcinogenesis, making these enzymes promising targets of drug discovery. Here we summarize the roles of RNA modifications during carcinogenesis induced by exposure to various environmental carcinogens, with a main focus on the roles of the most widely studied m6A mRNA methylation.
Keywords: arsenic, carcinogenesis., chemical carcinogens, DNA damage response, environmental carcinogens, epitranscriptomics, m6A, metal, virus
1. INTRODUCTION
RNA species, including messenger RNA (mRNA), transfer RNA (tRNA), ribosomal RNA (rRNA), and noncoding RNAs (ncRNAs), can be patterned with over 100 different post‐transcriptional modifications, collectively referred to as the epitranscriptome. 1 , 2 Functionally, RNA modifications regulate gene expression by modulating RNA metabolism. 3 RNA modifications play a role in many biological processes, including development, the cellular stress response, aging, and diseases such as cancer. 4 , 5 , 6
In cancer, recent studies have demonstrated that RNA modifications and their associated regulatory enzymes are dysregulated, the effects of which are context‐dependent. 4 , 7 Although many cancers result from exposures to carcinogens, studies on the roles of RNA modifications during carcinogenesis from exogenous exposures are limited. It is important to consider how specific carcinogens may differentially regulate RNA modifications during cancer initiation, as new insights into these early mechanisms are critical for developing specialized therapies.
In this review, we summarize evidence for the role of RNA modifications in promoting carcinogenesis in response to specific metal, chemical, inhalation, dermal, and viral exposures. We focus primarily on carcinogens, which are classified as carcinogenic to humans (Group 1), as well as agents that are classified as probably (Group 2A) or possibly carcinogenic (Group 2B) to humans by the International Agency for Research on Cancer (IARC) and for which RNA modifications have been shown to have a potential role in promoting malignancy. 8
2. RNA MODIFICATIONS AND THEIR ENZYMES
The development of sensitive detection methods and high‐throughput RNA‐sequencing technologies has advanced the characterization and mapping of RNA modifications. 9 , 10 The most extensively studied RNA modification is N 6‐methyladenosine (m6A) on mRNA. Other modifications on mRNA described here include 1‐methyladenosine (m1A), 5‐methylcytosine (m5C), and N6, 2′‐O‐dimethyladenosine (m6Am). 11
Regulation of the epitranscriptome is mediated by three categories of regulatory enzymes: writers, erasers, and readers. Writers catalyze the addition of the modification to the RNA molecule; erasers catalyze the removal of the modifications. 12 Readers serve to recognize the modifications (Figure 1). 12 The coordination of these enzymes can drive molecular processes including transcription, RNA export, translation, and decay. 3 , 9 , 12
Figure 1.
RNA modifications and their regulatory enzymes. (A) Writers, erasers, and readers of N 6‐methyladenosine (m6A), 5‐methylcytosine (m5C), 1‐methyladenosine (m1A), and N6, 2′‐O‐dimethyladenosine (m6Am). (B) The m6A writer complex catalyzes the addition of m6A modifications to RNA. Erasers mediate their removal, whereas m6A readers detect the modifications.
2.1. m6A
m6A methylation is the most prevalent internal modification in eukaryotic mRNA. It is a reversible modification conserved across various organisms, including mammals, plants, viruses, and bacteria. 13 , 14 , 15 , 16 m6A is found on roughly 0.1–0.4% of total adenosine nucleotides in mammals. 17 , 18 Studies into the methylome landscape using m6A‐seq and next‐generation sequencing have revealed that m6A is found in over 7000 human genes and is enriched in long internal exons, near stop codons, and at the 3′‐untranslated region (UTR). 10 , 19
m6A is deposited by a multicomponent writer complex, comprising methyltransferase‐like family members 3 and 14 (METTL3 and METTL14), and Wilms‐tumor 1‐associated protein (WTAP). 20 This complex interacts with other proteins, including RBM15, KIAA1429, and ZC3H13. 20 , 21 , 22 , 23 m6A erasers, or demethylases, include fat mass and obesity associated (FTO) prand Alkb homolog 5 (ALKBH5). 24 , 25 m6A readers include YTH‐binding proteins 1, 2, and 3 (YTHDF1, YTHDF2, and YTHDF3), YTH domain‐containing 1 and 2 (YTHDC1 and YTHDC2), heterogeneous nuclear ribonucleoprotein A2/B1 and C (HNRNPA2B1 and HNRNPC), and IGF2BPs. 26 , 27
2.2. m5C
m5C is found on both tRNA and mRNA at the 5′‐ and 3′‐UTRs. 28 m5C writers include the human ortholog NOP2/Sun domain protein 2 (NSUN2) and other NSUN family members, NSUN1‐7, as well as DNA methyltransferase homolog (DNMT2). 29 , 30 m5C readers include the Aly/REF export factor (ALYREF) and Y‐box‐binding protein 1 (YBX1). 31 , 32
2.3. m1A
m1A modifications are less abundant than m6A, constituting about 0.02% of adenosine in human mRNA, and are preferentially enriched around the start codon. 33 , 34 m1A writers include tRNA Methyltransferases 6 and 61a (TRMT6 and TRMT61a). 35
2.4. m6Am
m6Am modification consists of a dimethylation of the adenosine nucleoside. 36 m6Am is found adjacent to the mRNA cap structure at the transcription start nucleotide and internally on U2 small nuclear RNA (snRNA); it is estimated that 50–80% of starting adenosine nucleotides contain an m6Am modification. 37 The writers of m6Am are phosphorylated CTD interacting factor 1 (PCIF1) and METTL4. 38 , 39
3. CARCINOGEN EXPOSURES
3.1. Metal exposures
Evidence suggests that exposures to toxic metals arsenic, cadmium, and nickel can cause carcinogenesis by modulating cellular pathways through RNA modifications. Further studies are needed to determine whether exposure to other toxic metals, such as cobalt, lead, and chromium, can regulate RNA modifications to drive cancer development.
3.1.1. Arsenic
Arsenic is a naturally occurring metalloid in Earth's crust and an IARC Group 1 carcinogen. 40 Exposure to inorganic arsenic through contaminated drinking water is associated with skin, lung, bladder, and liver cancer. 41 Several studies have found that exposure to arsenic resulted in decreased m6A levels (Figure 2). Cui et al. 42 showed that in human keratinocytes, chronic exposure to relevant low levels of arsenite (100 nM) for 28 weeks increased FTO levels through arsenic‐mediated inhibition of p62‐dependent selective autophagy, resulting in decreased global m6A levels. 42 This study also found that the tumor‐suppressive ubiquitin ligase Nedd4l was a gene target of FTO in this context. 42 Decreased m6A on the Nedd4l transcript decreased Nedd4l mRNA stability, leading to the activation of Wnt signaling and tumorigenesis. 42 Other studies have found decreased global m6A levels in arsenic‐treated adenocarcinomic human alveolar basal epithelial (A549) cells. 43 , 44 Gao et al. 43 found that treatment with 2 μM of arsenic decreased global m6A mRNA levels by upregulating FTO in A549 cells. In particular, this study found that FTO demethylated transcripts of Apobec3 (A3B), a DNA deaminase and key driver of arsenic‐induced mutagenicity. 43 This demethylation increased A3B expression by inhibiting YTHDF2‐mediated A3B mRNA decay, leading to DNA damage and hypermutation in arsenic‐induced carcinogenesis. 43 Cayir et al. 44 also found decreased global m6A levels in arsenic‐exposed A549 cells, although the mechanism was not investigated in this study.
Figure 2.
Proposed molecular mechanisms of arsenic‐induced carcinogenesis. (A) Upregulation of fat mass and obesity associated (FTO) results in demethylation and degradation of NEDDL4 transcripts, promoting Wnt signaling. (B) Upregulation of FTO demethylates AP3 transcripts to increase AP3 translation, causing DNA hypermutation. (C) Upregulation of the m6A methyltransferase complex results in degradation of PDRM1 and upregulated expression of MDM2 and YY1, leading to inhibited p53 signaling.
In contrast, Zhao et al. 45 found that keratinocytes treated with 1 μM of arsenite for 5 months showed increased m6A levels, increased expression of METTL3, METTL14, WTAP, KIAA1429, and YTHDF1, and decreased expression of FTO. In particular, elevated m6A methylation of the p53 activator PRDM2 and p53 inhibitors YY1 and MDM2 was observed. 45 YTHDF2‐mediated decay of PRDM2 transcripts, together with YTHDF1‐mediated upregulation of YY1 and MDM2 translation, resulted in the inhibition of the tumor suppressor p53 and keratinocyte transformation. 45 Similarly, in human bronchial epithelial (HBE) cells treated with 2.5 μM arsenite for 13 weeks, m6A levels and expression of METTL3, METTL14, and WTAP expression were increased, whereas FTO was decreased. 46 Furthermore, another study found that NSUN2 expression was decreased after arsenite treatment in mouse keratinocytes, resulting in loss of tRNA methylation. 47
3.1.2. Cadmium
Cadmium is a heavy metal and IARC Group 1 carcinogen, and is strongly associated with the development of pulmonary cancer. 48 Cadmium exposure is primarily occupational. 48 Yang et al. 49 found that cadmium‐induced transformation of SV‐HUC‐1 human uroepithelial cells increased overall m6A levels and increased m6A deposition on cancer‐promoting genes, including Cdcp11, which encodes for an oncogenic transmembrane glycoprotein. Mechanistically, METTL3 deposited m6A onto the 3′‐UTR of Cdcp1 mRNA, resulting in increased CDCP1 expression through a YTHDF1‐dependent mechanism in transformed cells. 49 Similarly, cadmium‐treated HBE (BEAS‐2B) cells displayed decreased m6A levels, along with increased expression of ALKBH5. 50 ALKBH5 was found to demethylate mRNA of the tumor suppressor Pten, decreasing Pten mRNA stability and reducing PTEN expression, which promoted cadmium‐induced cell transformation. 50 In addition, in a study of cadmium‐induced oxidative damage, pancreatic β‐cells exposed to cadmium sulfate displayed decreased levels of m6A, METTL3, and FTO, although the precise nature of this relationship was not elucidated. 51
3.1.3. Nickel
Nickel is an IARC Group 1 carcinogen that can cause cancers of the lung, nasal cavity, and paranasal sinuses. 52 Yang et al. 49 found that nickel‐induced transformation of SV‐HUC‐1 human uroepithelial cells resulted in increased m6A levels and increased m6A deposition on Cdcp1 mRNA, causing increased CDCP1 translation in a YTHDF1‐dependent manner.
3.1.4. Other metals
Chromium, cobalt, and lead are toxic heavy metals and IARC carcinogens. 52 , 53 , 54 These heavy metals are found in contaminated soil and water; occupational exposures also occur during industrial processes. 52 , 53 , 54 Few studies have assessed changes in RNA modifications after these exposures. In HEK293T cells, exposure to chromium for 24 h altered levels of inosine modification on mRNA and upregulated expression of the adenosine deaminase ADAR1 that catalyzes the A‐to‐I modification. 55 , 56 Additionally, in the cortexes of C57BL/6 mice exposed to cobalt for 30 days, decreased m6A levels were noted, coupled with increased expression of FTO and ALKBH5, and decreased expression of METTL3, METTL14, and WTAP. 57 Functionally, the effects of cobalt exposure in this context were associated with neurodegenerative damage. 57 Furthermore, in adolescent C57B16 mice exposed to lead for 20 days, the m6Am writer METTL4 was downregulated; however, changes in m6Am levels were not evaluated. 58
3.2. Chemical exposures
There is evidence that exposure to chemical carcinogens, such as Fumoisin B1 (FB1) and diethylnitrosamine, can modulate RNA modifications to promote carcinogenesis by affecting oncogenic cellular pathways. Current work on arecoline exposure suggests a role for RNA modifications in promoting existing cancers, but not in inducing cancer initiation. Lastly, recent studies into other chemical carcinogens, including aflatoxin B1 (AFB1), carbon nanotubes (CNTs), cyclophosphamides (CYPs), and endocrine‐disrupting chemical exposures, indicate that these carcinogens can modulate RNA modifications and influence other biological processes, but how these changes may promote cancer initiation is unknown (Table 1).
Table 1.
Summary of RNA modifications in response to various chemical exposures
Carcinogen | IARC group | Cells or organisms | Finding | Biological process | References |
---|---|---|---|---|---|
Fumoisin B1 | 2B | HepG2 cells | ↑m6A | Oxidative stress and hepatocarcinogenesis | 60 |
↑METTL3, ↑METTL14, ↑YTHDF1, ↑YTHDF2, ↑YTHDC2, ↑YTHDF3 | |||||
↓FTO, ↓ALKBH5 | |||||
Diethylnitrosamine | 2A | C57Bl/6N | ↑FTO | HCC carcinogenesis | 62 |
mice | |||||
Arecoline | 1 | SCC25 and CAL27‐A cells | ↑FTO, ↑METTL3 | OSCC proliferation | 65 |
Arecoline | 1 | OECM1, SAS, and CGHNC9 cells | ↓ YBX1 | OSCC | 66 |
Aflatoxin | 1 | BME cells | ↓m6A, | Cytotoxicity | 69 |
↓METTL3, ↓METTL14, | |||||
↑ALKBH5 | |||||
Aflatoxin | 1 | C57BL/6J mice | ↑m6A, ↑METTL3, | Oxidative stress | 70 |
↓FTO, ↓YTHDF2 | |||||
Aflatoxin | 1 | HepaRG cells | ↓TRMT6 | – | 71 |
Aflatoxin | 1 | Caco‐2 cells | ↑ALYREF | Colon cytotoxicity | 72 |
Carbon nanotubes | 2B | HBE cells (16HBE14o‐) | ↓m6A, | – | 75 |
↑ hm5c | |||||
Carbon nanotubes | 2B | C57BL/6 mice | ↑TRMT61A | – | 76 |
Cyclophosphamide | 1 | Neonatal rat cardiac myocytes | ↑m6A, | Cardiotoxicity | 79 |
↑METTL3 | |||||
Cyclophosphamide | 1 | hGCs and mouse ovary | ↑m6A, | Ovarian damage | 80 |
↑METTL3, ↑METTL14, ↑Zc3h13, ↑KIAA1429, | |||||
↓FTO, | |||||
↓YTHDF1, | |||||
↓YTHDF2, | |||||
↓YTHDC1, | |||||
↓YTHDC3 | |||||
PCB | 1 | C57BL/6J mice | ↑m6Am, | Nonalcoholic steatohepatitis | 83 |
↑PCIF1 | |||||
PCB | 1 | Human primary blood mononuclear cells | ↓WTAP, | – | 84 |
↓RBM15 | |||||
DHEP | 2B | Sprague–Dawley rats | ↑m6A, | Testicular injury | 86 |
↑YTHDC2, | |||||
↓FTO | |||||
Triclosan | – | Zebrafish larvae | ↓m6A, | Obesity and nonalcoholic fatty liver disease | 90 |
↑YTHDC1, | |||||
↑YTHDF1, | |||||
↑FTO | |||||
Bisphenol‐A | – | Zebrafish larvae | ↓m6A, | Obesity and nonalcoholic fatty liver disease | 90 |
↑YTHDC1 | |||||
↑YTHDF1 | |||||
Bisphenol‐A | – | A549 cells | ↓m6A | – | 44 |
Vinclozolin | – | A549 cells | ↓m6A | – | 44 |
Note: Refer to original reference for full list.
Abbreviations: ALKBH5, Alkb homolog 5; BME, bovine mammary epithelial; DHEP, Di(2‐ethylhexyl)phthalate; FTO, fat mass and obesity; HCC, hepatocellular carcinoma; hGCs, human ovarian granulosa cells; hm5C, 5‐hydroxymethylcytosine; METTL3, methyltransferase‐like family member 3; METTL14, methyltransferase‐like family member 14; m6A, N 6‐methyladenosine; PCIF1, phosphorylated CTD interacting factor 1; OSCC, oral squamous cell carcinoma; WTAP, Wilms‐tumor 1‐associated protein; YBX1, Y‐box‐binding protein 1; YTHDC1, YTH domain‐containing 1; YTHDC2, YTH domain‐containing 2; YTHDF1, YTH‐binding protein 1; YTHDF2, YTH‐binding protein 2; YTHDF3, YTH‐binding protein 3.
3.2.1. FB1
FB1 is a toxin produced by Fusarium molds found primarily in maize products and was considered possibly carcinogenic to humans (IARC Group 2B) after causing hepatocellular carcinoma (HCC), cholangiocarcinoma, and renal tubule carcinomas in mouse models. 59 Increased m6A levels were observed in HepG2 cells exposed to FB1 for 24 h, coupled with increased expression of writers METTL3 and METTL14, decreased expression of erasers FTO and ALKBH5, and increased expression of readers YTHDF1, YTHDF3, YTHDC2, and YTHDF2. The transcription factor NRF2 promotes an antioxidant response upon FB1 exposure and an increase of m6A‐modified Nrf2 transcripts upregulated NRF2 expression via a YTHDF1, YTHDF3, and/or YTHDC2 mechanism in response to FB1. 60 In contrast, expression of KEAP1, an NRF2 inhibitor, was decreased in response to FB1 as FB1‐induced increases in m6A on the Keap1 transcript resulted in decreased Keap1 expression via a YTHDF2 mechanism. 60 This suggests that the KEAP1‐NRF2 antioxidant response is activated in response to FB1‐induced reactive oxygen species and it is hypothesized that the prolonged activation of NRF2 signaling may be a mechanism by which FB1 promotes hepatocarcinogenesis. 60
3.2.2. Diethylnitrosamine
Diethylnitrosamine, or N‐Nitrodiethylamine (DEN), is a synthetic oil and hepatocarcinogen classified in Group 2A, and is found in gasoline and other industrial materials. 61 In a study of tumor initiation in HCC, male mice injected with 100 mg/kg DEN had elevated Fto mRNA and protein expression at 24 and 48 h postinjection. 62 CUL4A is an oncogene associated with HCC; in DEN‐exposed mice, Cul4a transcripts were demethylated by FTO, resulting in decreased Cul4a expression. 62 This suggests a protective role for FTO against DEN‐induced HCC development. 62
3.2.3. Arecoline
Arecoline is an alkaloid and IARC Group 1 carcinogen found in the areca nut, and chewing the areca nut is linked to a high risk of oral cancer due to arecoline exposure. 63 , 64 In oral squamous cell carcinoma (OSCC) cell lines treated with 1 μM arecoline for 90 days, FTO and METTL3 protein expression were increased. 65 The same study also found that FTO promoted cancer development and proliferation of arecoline‐transformed OSCC by maintaining cancer stemness and mediating cisplatin resistance. 65 In another study of arecoline‐induced OSCC, oral cancer cell lines treated with areca nut extract for 3 months showed decreased expression of the m5C reader YBX1. 66
3.2.4. AFB1
AFB1 is a mycotoxin which can pollute grains and animal products. 67 AFB1 is metabolized to AFM1 after consumption by cows and can contaminate dairy products such as milk. 67 AFB1 is classified as an IARC Group 1 carcinogen and it is associated with liver and breast cancers. 68 Wu et al. 69 found decreased levels of m6A in bovine mammary epithelial cells treated with AFB1, coupled with increased expression of the demethylase ALKBH5 and decreased expression of methyltransferases METTL3 and METTL14. However, how these changes regulate cellular response to AFB1 damage is unknown. 69 In another study, when C57BL/6J mice were fed a diet of 600 μg/kg AFB1 over 4 weeks, they showed higher levels of m6A modification in the liver as well as decreased expression of FTO and YTHDF2, and higher expression of METTL3. 70 The authors found that treatment with resveratrol, a naturally occurring antioxidant, could attenuate increased m6A levels in the liver and suggested that this may be a mechanism of promoting antioxidant gene expression to repair hepatic function. 70 Another study showed that AFB1 exposure decreases the m1A writer TRMT6 in HepaRG cells. 71 Furthermore, human colon Caco‐2 cells exposed to a chemical precursor of AFB1 called Versicolorin A exhibited increased expression of the m5C reader ALYREF. 72
3.2.5. CNTs
CNTs are composed of graphene sheets rolled into a cylindrical fiber and produce particulate matter similar to that of asbestos. 73 A type of CNT, a multiwalled CNT (MWCNT) called Mitsui‐7, was classified as possibly carcinogenic to humans (Group 2B) in 2014. 74 One study found that bronchial epithelial cells exposed to MWCNTs had decreased global m6A methylation and increased levels of 5‐hydroxymethylcytosine (hm5C) on RNA. 75 Additionally, exposure to single‐walled CNTs showed higher levels of hm5C on RNA. 75 Another study showed that mice exposed to low, medium, and high doses of MWCNT had upregulated levels of the m1A writer TRMT61A. 76
3.2.6. CYP
CYP is an antineoplastic and alkylating agent used to treat certain cancers and diseases. 77 CYP has been classified as an IARC Group 1 carcinogen and has been found to cause bladder cancer and acute myeloid leukemia. 78 A study of the effects of CYP on cardiotoxicity showed that rat neonatal cardiomyocytes treated with CYP for 2 days had increased m6A levels and increased METTL3 expression. 79 METTL3‐mediated m6A on junctophilin‐2 (Jph2) mRNA decreased Jph2 expression and promoted cardiac dysfunction under cardiotoxic stress. 79 In another study, treatment of normal human ovarian granulosa cells (hGCs) or mouse ovaries with CYP also showed increased levels of m6A in a time‐ and concentration‐dependent manner. 80 CYP‐treated hGCs and mouse ovaries also showed increased expression of METTL3, METTL14, ZC3H13, and KIAA1429, and decreased expression of FTO and the readers YTHDF1, YTHDF2, YTHDC1, and YTHDC3, in a time‐dependent manner. 80 The authors suggest that these effects may have a role in CYP‐induced ovarian damage. 80
3.2.7. Endocrine‐disrupting chemicals: Polychlorinated biphenyls (PCBs), Di(2‐ethylhexyl)phthalate (DHEP), triclosan, and bisphenol‐A (BPA)
PCBs are synthetic organic compounds that were widely used in dielectric fluids and sealants until 1979 and can contaminate soil, air, water, and food. 81 PCBs are IARC Group 1 carcinogens associated with melanoma, non‐Hodgkin's lymphoma, and breast cancer. 81 Aluru et al. 82 exposed zebrafish embryos to 10 nM PCB126 for 7 h and found 15 different RNA transcripts with m6A methylation. Interestingly, these 15 RNA transcripts were associated with AHR signaling, which is involved in responses to environmental stress. 82 In a study of toxicant‐associated steatohepatitis, C57BL/6J male mice exposed to PCB126 and a high‐fat diet (HFD) had altered levels of 10 types of RNA modifications in total RNA. 83 In particular, mice fed HFDs and exposed to a PCB mixture showed increased m6Am and PCIF1 expression. 83 Furthermore, a study of transcriptional profiling in response to mixed PCB exposure in primary blood mononuclear cells showed decreased expression of the m6A writers WTAP and RBM15. 84
DHEP is an IARC group 2B carcinogen and Zhao et al. 85 , 86 showed that exposure to DHEP in Sprague–Dawley rats resulted in increased m6A levels, increased YTHDC2 expression, and decreased FTO expression. In addition, DHEP exposure resulted in decreased NRF2 signaling and increased oxidative stress, which promoted testicular toxicity. 86 The carcinogenicity of triclosan, BPA, and vinclozolin is still under consideration. 87 , 88 , 89 In a study of nonalcoholic fatty liver disease, zebrafish larvae treated with BPA or triclosan showed decreased global m6A levels. 90 Both triclosan and BPA exposures showed increased expression of YTHDC1 and YTHDF1, whereas only triclosan exposure showed increased FTO expression. 90 Furthermore, another study found that exposure of A549 cells to BPA or vinclozolin similarly showed decreased m6A levels. 44
3.3. Inhalation exposures
There is evidence to suggest that cigarette smoke (CS) exposure can lead to the development of lung cancer, pancreatic ductal adenocarcinoma (PDAC), and esophageal squamous cell carcinoma (ESCC), through changes in RNA modifications. Studies have also found that particulate matter in outdoor air pollution can affect RNA modifications, but it is unclear whether this particulate matter can contribute to carcinogenesis.
3.3.1. CS
As of 2019, 1.14 billion individuals worldwide were estimated to be tobacco smokers. 91 Tobacco smoke is an IARC Group 1 carcinogen with associations with a variety of cancer types including lung, esophagus, and pancreatic cancer. 92 Tobacco is most commonly smoked as cigarettes and secondhand exposure to tobacco smoke or involuntary smoking is also considered a Group 1 carcinogen. 92 Jin et al. 93 showed that A549 cells exposed to CS condensate (CSC) had increased m6A levels (Table 2). 93 Gene‐specific m6A increases were found on the transcript of threonine/serine kinase Dapk2, a tumor suppressor in nonsmall cell lung cancer (NSCLC). 93 In particular, upregulated METTL3 expression increased m6A on the Dapk2 transcript, resulting in decreased Dapk2 mRNA stability and expression through a YTHDF2‐dependent mechanism. 93 Decreased DAPK2 resulted in downstream activation of oncogenic nuclear factor‐κB (NF‐κB) signaling, establishing a role for the METTL3‐DAPK2‐NF‐κB axis in promoting CSC‐induced NSCLC malignancy. 93
Table 2.
Role of RNA modifications during CS‐induced carcinogenesis
Exposure form | Dose | Cells or organisms | Cancer type | Finding | Proposed | References |
---|---|---|---|---|---|---|
pathway | ||||||
CS condensate | 100 μg/ml | A549 cells | NSCLC | ↑m6A, ↑METTL3 | METTL3‐DAPK2‐NF‐κB | 93 |
CS extract | 2% | HBE cells | Lung cancer | ↑m6A, ↑METTL3 | METTL3‐ YTHDF2‐ZBTB4 | 94 |
CS | 20% | BEAS‐2B cells | Lung cancer | ↓YTHDC2 | YTHDC2‐CYLD‐NF‐κB | 95 |
CS condensate | 100 μg/ml | HPDE6‐C7 cells | PDAC | ↑METTL3 | METTL3‐miR‐25‐3p‐PHLPP2‐AKT | 96 |
CS condensate | 100 μg/ml | Het1A cells | ESCC | ↑ALKBH5 | YTHDF1‐YY1BM‐eEF2K | 97 |
CS | – | Human subjects | – | ↓m6A, ↑METTL3 | – | 98 |
Abbreviations: ALKBH5, Alkb homolog 5; CS, cigarette smoke; ESCC, esophageal squamous cell carcinoma; HBE, human bronchial epithelial; m6A, N 6‐methyladenosine; METTL3, methyltransferase‐like family member 3; NSCLC, nonsmall cell lung cancer; PDAC, pancreatic ductal adenocarcinoma; YTHDC2, YTH domain‐containing 2.
Similarly, in HBE cells transformed with 2% CS extract, increased levels of m6A and upregulated expression of METTL3 were observed after 48 h of treatment. 94 In particular, METTL3‐mediated increases of m6A on the 3′‐UTR of the tumor suppressor Zbtb4 resulted in decreased ZBTB4 expression through YTHDF2‐mediated mRNA decay. 94 As a result, decreased ZBTB4 expression promoted CS‐induced epithelial‐mesenchymal transition and carcinogenesis. 94 In another study, HBE (BEAS‐2B) cells exposed to CS had downregulated expression of YTHDC2. 95 Mechanistically, YTHDC2 was found to bind m6A sites on the transcript of the tumor suppressor Cyld and promoted Cyld mRNA stability. 95 As CYLD normally inhibits the NF‐κB pathway, decreased YTHDC2 expression after CS exposure can promote cancer cell proliferation by modulating the CYLD/NF‐κB axis in CS‐induced lung cancer. 95
In a PDAC study, exposure to 100 μg/ml of CSC in human pancreatic duct epithelial (HPDE6C7) cells for 48 h increased the expression of METTL3 and the microRNA (miRNA) miR‐25‐3p. 96 METTL3 promoted the maturation of the miRNA miR‐25‐3p in an m6A‐dependent manner and the NF‐κB‐associated protein NKAP served as a reader of m6A on miR‐25‐p3. 96 Given that miR‐25‐3p has a role in inhibiting PHLPP2 and activating AKT signaling in cancer progression, the study suggests a role of the METTL3‐miR‐25‐3p‐PHLPP2‐AKT axis in promoting cigarette smoking‐induced PDAC tumorigenesis. 96
A study in cigarette smoking‐induced ESCC showed that the translation of a long noncoding RNA (LINC20078) into the micropeptide (YY1BM) was regulated by m6A modifications and mediated by YTHDF1. 97 In ESCC, YY1BM suppresses transcription of an oncogenic factor, EEF2K, to slow cancer progression. 97 CS exposure was found to upregulate ALKBH5, resulting in the demethylation of LINC20078 and reduced YY1BM levels, ultimately promoting ESCC carcinogenesis. 97
Finally, one study utilized data from the Beijing Truck Driver Air Pollution Study to correlate smoking exposure to m6A modification in human subjects. 98 Long‐term smoking was correlated with a 10.7% decrease in global m6A levels in blood leukocytes compared to nonsmokers, whereas acute smoking exposure did not correlate with changes in m6A. 98
3.3.2. Particulate matter
Particulate matter in outdoor air pollution is an IARC Group 1 carcinogen based on evidence that exposure can cause lung cancer. 99 This group includes fine inhalable particles with diameters smaller than 2.5 μm, referred to as PM2.5, and can comprise inorganic ions, metal oxides, and carbonaceous material. 99 HBE and A549 cells exposed to 62.5, 125, and 250 μg/ml of PM2.5 showed increases in m6A levels. 100 In particular, METTL3 promoted the mRNA stability of cell death regulator, OSGIN1, in an m6A‐dependent manner, resulting in increased apoptosis, cell cycle arrest, and autophagy in PM2.5‐exposed cells. 100 Similarly, another study showed that the lungs of C57BL.6J male mice exposed to PM2.5 had increased levels of m6A and increased expression of METTL3 and METTL14. 101 Interestingly, this effect could be reversed after exposure to purified air. 101
In contrast, a study in A549 cells exposed to >62 μg/ml of PM for 24 or 48 h showed decreased global levels of m6A. 44 The authors also reviewed a data set of human participants exposed to high levels of PM2.5 and found increased expression of the writers METTL3 and WTAP, the erasers FTO and ALKBH5, and the reader HNRNPC in leukocyte cells. 44 In a study of differentially expressed genes in HBE cells after exposure to 50 μg/ml PM2.5, the m6A reader HNRNPA2B1 was upregulated and the m5C reader ALYREF was downregulated. 102 Lastly, a study of pulmonary fibrosis noted increased mRNA and protein levels of the m5C writer NSUN2 in the lungs of C57BL/6 male mice exposed to an average of 60 μg/ml of PM2.5 for 12 weeks. 103
3.4. Dermal exposures
Evidence indicates that UV exposure can promote carcinogenesis through RNA modifications. In contrast, exposure to γ‐radiation can modulate RNA modification and regulatory enzymes, but more research is needed to determine how this could promote carcinogenesis.
3.4.1. UV radiation
Natural sources of UV exposure include solar radiation (UVA and UVB). 104 Solar UV radiation, as well as artificial sources of radiation such as tanning beds, can cause melanoma and other skin cancers. 104 Yang et al. 105 showed that in human keratinocytes exposed to UVB irradiation, METTL14 was downregulated and levels of m6A were decreased. Mechanistically, METTL14 levels were decreased through UVB‐induced NBR1‐dependent selective autophagy. 105 In response to UVB‐induced damage, METTL14 was found to regulate translation of the genome repair factor DDB2 through an m6A/YTHDF1‐dependent mechanism. 105 Furthermore, skin‐specific heterozygous deletion of METTL14 in mice accelerated UVB‐induced carcinogenesis, suggesting that METTL14 suppresses tumorigenesis by promoting global genome repair. 105
Other research has shown that m6A modification transiently accumulates at sites of DNA damage. METTL3, METTL14, and FTO were also found to localize to sites of DNA damage in response to UVA laser micro‐irradiation or UVC irradiation in U2OS cells, A375 melanoma cells, and HeLa cells. 106 Additionally, the DNA damage repair factor PARP1 was found to recruit METTL3 and METTL3‐mediated RNA methylation promoted DNA repair by recruiting DNA polymerase Polκ to damaged sites. 106 There is also evidence that METTL16 accumulates at UVA‐radiated lesions and methylates snRNAs during later stages of genome repair. 107 UVA radiation was also found to decrease global m1A modification levels. 107 In addition, it has been shown that UVB exposure can rapidly reduce RNA levels of the m5C writer Nsun2 in human epidermal and dermal cells. 47
3.4.2. γ‐Radiation
Exposure to γ‐radiation can occur through natural sources and medical use, and can lead to leukemia, breast cancer, and thyroid cancer, among others. 108 Bone marrow cells from C57BL/6J mice exposed to γ‐irradiation showed reduced m6A levels after 2 h, accompanied by increased expression of METTL14 and ALKBH5, and decreased expression of METTL3, WTAP, and FTO. 109 m6A modifications were suggested to play a role in the development of radiation toxicity, and inhibition of FTO and ALKBH5 reduced γ‐radiation hematopoietic injury. 109
3.5. Viral infections
Viruses can modulate RNA modifications in both viral and host RNAs to enhance replication, promote long‐term latency, and regulate life cycles. 15 Research suggests that Hepatitis B, Epstein–Barr virus (EBV), and Kaposi's sarcoma‐associated herpesvirus (KSHV) are all oncogenic viruses that can modulate the host's RNA modifications to promote carcinogenesis. 15
3.5.1. Hepatitis B virus (HBV)
HBV is an IARC Group 1 carcinogen. 110 Chronic infection with HBV can cause HCC and cholangiocarcinoma. 110 Kim et al. 111 found that in primary human hepatocytes, infection with HBV induced differential m6A changes in various host RNAs (Figure 3). In particular, HBV infection increased METTL3‐mediated m6A modification on the tumor suppressor Pten transcript, resulting in decreased Pten mRNA stability. 111 Decreased PTEN expression resulted in the activation of the PI3K/AKT pathway, contributing to HBV‐mediated HCC carcinogenesis. 111
Figure 3.
Pathways of carcinogenesis during hepatitis B infection. (A) Viral protein HBX upregulates m6A modification of circ‐ARL‐3 to increase its expression, leading to inhibition of tumor suppressor miR‐1305. (B) Upregulation of methyltransferase‐like family member 3 (METTL3) leads to degredation of Pten transcripts and activation of phosphatidylinositol 3‐kinase (PI3K)/AKT pathway. (C) Viral protein hepatitis B X‐interacting protein (HBXIP) upregulates METTL3 to promote increased translation of oncogene Myc. (D) Upregulation of Alkb homolog 5 (ALKBH5) leads to increased production of oncogenic HBX. 120
In addition, HBX, a Hepatitis‐B viral protein and a major etiological factor in HCC, 112 has been shown to promote m6A modification of a circRNA, circ‐ARL3, by upregulating METTL3 expression. 113 As a result, increased circ‐ARL3 could then bind an HCC tumor suppressor miRNA, miR‐1305, and promote HBV‐related HCC pathogenesis. 113 Additionally, Qu et al. 114 showed that HBV could upregulate ALKBH5 through an HBX‐WDR5‐H3K4me3 feedback loop, causing ALKBH5 to be highly expressed in HBV‐induced HCC. Additionally, ALKBH5 promoted Hbx mRNA stability by decreasing m6A modification, suggesting an oncogenic role for ALKBH5 in HBV‐induced HCC. 114
Furthermore, HBV infection is also associated with gastric cancer development, and a study showed that the hepatitis B X‐interacting protein could upregulate METTL3 during HBV infection. 115 Mechanistically, METTL3‐mediated m6A modification on the proto‐oncogene c‐Myc transcript resulted in increased MYC expression and gastric cancer progression. 115
3.5.2. EBV
EBV was the first oncogenic virus discovered, and is an IARC Group 1 carcinogen that causes Burkitt's lymphoma and Hodgkin's lymphoma, among others. 110 Lang et al. 116 showed that EBV‐infected cells had elevated levels of METTL14. METTL14 was also found to colocalize with the EBV latent antigen EBNA3C. 116 Increased METTL14 upregulated the expression of EBV latent antigens, including EBNA3C, in an m6A‐dependent manner, establishing a positive feedback loop that contributed to EBV‐induced oncogenesis. 116 Xia et al. 117 showed that YTHDF1 could repress EBV replication by degrading m6A‐modified viral transcripts through recruitment of ZAP, DDX17, and DCP2, making it a potential therapeutic target for EBV‐associated cancers. 117
3.5.3. KSHV
KSHV is a γ‐2 herpesvirus and an IARC Group 1 carcinogen that can cause Kaposi's sarcoma and primary effusion lymphoma. 110 Tan et al. 118 analyzed m6A and m6Am modifications together, and noted that in the host RNA of cells latently infected with KSHV, 5′‐UTR were hypomethylated and 3′‐UTR were hypermethylated compared with uninfected cells. These differentially methylated genes belonged to pathways involved in cellular transformation, including mTOR signaling, ephrin receptor signaling, and hypoxia, suggesting that KSHV may utilize RNA modifications to induce tumorigenesis. 118 In addition, m6A has been shown to be required for the onset of KSHV lytic replication, a major contributor to cancer development, and regulate pre‐mRNA splicing of the replication transcription factor. 119
4. CONCLUSIONS
Compelling evidence has demonstrated the crucial role of RNA modifications in maintaining the oncogenic traits of established cancers and cancer cells. Recent emerging evidence suggests that upon exposure to external carcinogens, RNA modifications are also implicated much earlier in the carcinogenic process. In this review, we summarize advances in the field, and consider how various exogenous exposures may use RNA modifications to promote oncogenesis.
For some carcinogens such as arsenic and CS, there are several described mechanisms by which RNA modifications promote carcinogenesis. More studies must be conducted to understand how the various mechanisms co‐exist and interact with one another to pinpoint where potential therapies may be most successful. However, for many carcinogens such as CNTs and γ‐radiation, a direct link to carcinogenic processes is yet to be established and the limited evidence presented here indicates that these would be areas worth pursuing.
Notably, the existing research on RNA modifications in carcinogenesis focuses heavily on m6A modifications and their writers, erasers, and readers. However, preliminary evidence for exposures to carcinogens such as UV exposure and PM suggests that m5C and m1A modifications may also play an important role in carcinogenesis. As research into RNA modifications continues to gain prominence, further studies must investigate how different types of chemical modifications in RNA affect carcinogenic processes. Finally, out of 121 Group 1 IARC carcinogens, only 16 were described in this review, indicating a large breadth of potential research into other carcinogenic exposures that are yet to be fully explored. Overall, the role of RNA modifications in carcinogenesis is a new emerging field for continuing to elucidate the molecular mechanisms of oncogenesis and for developing new cancer therapies.
AUTHOR CONTRIBUTIONS
Yu‐Ying He conceived the project. Michelle Verghese, Emma Wilkinson, and Yu‐Ying He wrote the paper.
ACKNOWLEDGMENTS
We apologize to those investigators whose work could not be directly referenced owing to space limitations. We thank Ann Motten for her critical reading of the manuscript. Work in the authors' laboratory was supported in part by NIH grants 5T32CA009594‐32 (EW), ES031534 (Y‐YH), ES024373 (Y‐YH), ES030576 (Y‐YH), the CACHET (NIH ES027792), the University of Chicago Comprehensive Cancer Center (NIH CA014599), the CTSA (NIH UL1 TR000430), and the University of Chicago Friends of Dermatology Endowment Fund. All figures were created with BioRender.com.
Verghese M, Wilkinson E, He Y‐Y. Role of RNA modifications in carcinogenesis and carcinogen damage response. Molecular Carcinogenesis. 2023;62:24‐37. 10.1002/mc.23418
DATA AVAILABILITY STATEMENT
Data from this study are available from the corresponding author upon request.
REFERENCES
- 1. Boccaletto P, Machnicka MA, Purta E, et al. MODOMICS: a database of RNA modification pathways. 2017 update. Nucleic Acids Res. 2018;46(D1):D303‐D307. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2. Motorin Y, Helm M. RNA nucleotide methylation. WIREs RNA. 2011;2(5):611‐631. [DOI] [PubMed] [Google Scholar]
- 3. Roundtree IA, Evans ME, Pan T, He C. Dynamic RNA modifications in gene expression regulation. Cell. 2017;169(7):1187‐1200. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4. Barbieri I, Kouzarides T. Role of RNA modifications in cancer. Nat Rev Cancer. 2020;20(6):303‐322. [DOI] [PubMed] [Google Scholar]
- 5. Zhang M, Zhai Y, Zhang S, Dai X, Li Z. Roles of N6‐Methyladenosine (m6A) in stem cell fate decisions and early embryonic development in mammals. Front Cell Dev Biol. 2020;8:782. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6. Min K‐W, Zealy RW, Davila S, et al. Profiling of m6A RNA modifications identified an age‐associated regulation of AGO2 mRNA stability. Aging Cell. 2018;17(3):e12753. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7. Nombela P, Miguel‐López B, Blanco S. The role of m6A, m5C and Ψ RNA modifications in cancer: novel therapeutic opportunities. Mol Cancer. 2021;20(1):18. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8. International Agency for Research in Cancer . IARC Monographs on the Identification of Carcinogenic Hazards to Humans. International Agency for Research in Cancer;1987.
- 9. Shi H, Wei J, He C. Where, when and how: context‐dependent functions of RNA methylation writers, readers, and erasers. Mol Cell. 2019;74(4):640‐650. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10. Dominissini D, Moshitch‐Moshkovitz S, Schwartz S, et al. Topology of the human and mouse m6A RNA methylomes revealed by m6A‐seq. Nature. 2012;485(7397):201‐206. [DOI] [PubMed] [Google Scholar]
- 11. Shima H, Igarashi K. N1‐methyladenosine (m1A) RNA modification: the key to ribosome control. J Biochem. 2020;167(6):535‐539. [DOI] [PubMed] [Google Scholar]
- 12. Boo SH, Kim YK. The emerging role of RNA modifications in the regulation of mRNA stability. Exp Mol Med. 2020;52(3):400‐408. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13. Yue H, Nie X, Yan Z, Weining S. N6‐methyladenosine regulatory machinery in plants: composition, function and evolution. Plant Biotech J. 2019;17(7):1194‐1208. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14. Canaani D, Kahana C, Lavi S, Groner Y. Identification and mapping of N6‐methyladenosine containing sequences in simian virus 40 RNA. Nucleic Acids Res. 1979;6(8):2879‐2899. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15. Wu F, Cheng W, Zhao F, Tang M, Diao Y, Xu R. Association of N6‐methyladenosine with viruses and related diseases. Virol J. 2019;16(1):133. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16. Deng X, Chen K, Luo G‐Z, et al. Widespread occurrence of N6‐methyladenosine in bacterial mRNA. Nucleic Acids Res. 2015;43(13):6557‐6567. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17. Kane SE, Beemon K. Precise localization of m6A in Rous sarcoma virus RNA reveals clustering of methylation sites: implications for RNA processing. Mol Cell Biol. 1985;5(9):2298‐2306. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18. Wei C‐M, Gershowitz A, Moss B. Methylated nucleotides block 5′ terminus of HeLa cell messenger RNA. Cell. 1975;4(4):379‐386. [DOI] [PubMed] [Google Scholar]
- 19. Meyer KD, Saletore Y, Zumbo P, Elemento O, Mason CE, Jaffrey SR. Comprehensive analysis of mRNA methylation reveals enrichment in 3′ UTRs and near stop codons. Cell. 2012;149(7):1635‐1646. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20. Liu J, Yue Y, Han D, et al. A METTL3‐METTL14 complex mediates mammalian nuclear RNA N6‐adenosine methylation. Nat Chem Biol. 2014;10(2):93‐95. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21. Wen J, Lv R, Ma H, et al. Zc3h13 regulates nuclear RNA m(6)A methylation and mouse embryonic stem cell self‐renewal. Mol Cell. 2018;69(6):1028‐1038.e1026. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22. Schwartz S, Mumbach MR, Jovanovic M, et al. Perturbation of m6A writers reveals two distinct classes of mRNA methylation at internal and 5' sites. Cell Rep. 2014;8(1):284‐296. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23. Patil DP, Chen CK, Pickering BF, et al. m(6)A RNA methylation promotes XIST‐mediated transcriptional repression. Nature. 2016;537(7620):369‐373. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24. Jia G, Fu Y, Zhao X, et al. N6‐methyladenosine in nuclear RNA is a major substrate of the obesity‐associated FTO. Nat Chem Biol. 2011;7(12):885‐887. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25. Zheng G, Dahl JA, Niu Y, et al. ALKBH5 is a mammalian RNA demethylase that impacts RNA metabolism and mouse fertility. Mol Cell. 2013;49(1):18‐29. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26. Liao S, Sun H, Xu C, YTH domain: a family of N6‐methyladenosine (m6A) readers. Genomics Insights. 2018;16(2):99‐107. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27. Esteve‐Puig R, Bueno‐Costa A, Esteller M. Writers, readers and erasers of RNA modifications in cancer. Cancer Lett. 2020;474:127‐137. [DOI] [PubMed] [Google Scholar]
- 28. Squires JE, Patel HR, Nousch M, et al. Widespread occurrence of 5‐methylcytosine in human coding and non‐coding RNA. Nucleic Acids Res. 2012;40(11):5023‐5033. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29. Motorin Y, Lyko F, Helm M. 5‐Methylcytosine in RNA: detection, enzymatic formation and biological functions. Nucleic Acids Res. 2010;38(5):1415‐1430. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30. Schaefer M, Pollex T, Hanna K, et al. RNA methylation by Dnmt2 protects transfer RNAs against stress‐induced cleavage. Genes Dev. 2010;24(15):1590‐1595. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31. Yang X, Yang Y, Sun B‐F, et al. 5‐Methylcytosine promotes mRNA export—NSUN2 as the methyltransferase and ALYREF as an m5C reader. Cell Res. 2017;27(5):606‐625. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32. Chen X, Li A, Sun B‐F, et al. 5‐methylcytosine promotes pathogenesis of bladder cancer through stabilizing mRNAs. Nature Cell Biol. 2019;21(8):978‐990. [DOI] [PubMed] [Google Scholar]
- 33. Li X, Xiong X, Wang K, et al. Transcriptome‐wide mapping reveals reversible and dynamic N1‐methyladenosine methylome. Nat Chem Biol. 2016;12(5):311‐316. [DOI] [PubMed] [Google Scholar]
- 34. Dominissini D, Nachtergaele S, Moshitch‐Moshkovitz S, et al. The dynamic N1‐methyladenosine methylome in eukaryotic messenger RNA. Nature. 2016;530(7591):441‐446. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35. Ozanick S, Krecic A, Andersland J, Anderson JT. The bipartite structure of the tRNA m1A58 methyltransferase from S. cerevisiae is conserved in humans. RNA. 2005;11(8):1281‐1290. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36. Keith JM, Ensinger MJ, Moss B. HeLa cell RNA (2'‐O‐methyladenosine‐N6‐)‐methyltransferase specific for the capped 5'‐end of messenger RNA. J Biol Chem. 1978;253(14):5033‐5039. [PubMed] [Google Scholar]
- 37. Sun H, Li K, Zhang X, et al. m6Am‐seq reveals the dynamic m6Am methylation in the human transcriptome. Nat Commun. 2021;12(1):4778. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38. Akichika S, Hirano S, Shichino Y, et al. Cap‐specific terminal N6‐methylation of RNA by an RNA polymerase II‐associated methyltransferase. Science. 2019;363. [DOI] [PubMed] [Google Scholar]
- 39. Chen H, Gu L, Orellana EA, et al. METTL4 is an snRNA m6Am methyltransferase that regulates RNA splicing. Cell Res. 2020;30(6):544‐547. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40. International Agency for Research in Cancer . Arsenic, metals, fibres and dusts International Agency for Research on Cancer. Vol 100C. IARC Monographs on the Evaluation of Carcinogenic Risks to Humans;2012;41‐85. [PMC free article] [PubMed]
- 41. Martinez VD, Vucic EA, Becker‐Santos DD, Gil L, Lam WL. Arsenic exposure and the induction of human cancers. J Toxicol. 2011;2011:431287. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42. Cui Y‐H, Yang S, Wei J, et al. Autophagy of the m6A mRNA demethylase FTO is impaired by low‐level arsenic exposure to promote tumorigenesis. Nat Commun. 2021;12(1):2183. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43. Gao M, Qi Z, Feng W, et al. m6A demethylation of cytidine deaminase APOBEC3B mRNA orchestrates arsenic‐induced mutagenesis. J Biol Chem. 2022;0:0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44. Cayir A, Barrow TM, Guo L, Byun H‐M. Exposure to environmental toxicants reduces global N6‐methyladenosine RNA methylation and alters expression of RNA methylation modulator genes. Environ Res. 2019;175:228‐234. [DOI] [PubMed] [Google Scholar]
- 45. Zhao T, Sun D, Zhao M, Lai Y, Liu Y, Zhang Z. N6‐methyladenosine mediates arsenite‐induced human keratinocyte transformation by suppressing p53 activation. Environ Pollut. 2020;259:113908. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46. Gu S, Sun D, Dai H, Zhang Z. N6‐methyladenosine mediates the cellular proliferation and apoptosis via microRNAs in arsenite‐transformed cells. Toxicol Lett. 2018;292:1‐11. [DOI] [PubMed] [Google Scholar]
- 47. Gkatza NA, Castro C, Harvey RF, et al. Cytosine‐5 RNA methylation links protein synthesis to cell metabolism. PLoS Biol. 2019;17(6):e3000297. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48. International Agency for Research in Cancer . Beryllium, cadmium, mercury, and exposures in the glass manufacturing industry. IARC Monographs on the Evaluation of Carcinogenic Risks to Humans. Vol 58. International Agency for Research in Cancer; 1993:119‐238. [PMC free article] [PubMed] [Google Scholar]
- 49. Yang F, Jin H, Que B, et al. Dynamic m6A mRNA methylation reveals the role of METTL3‐m6A‐CDCP1 signaling axis in chemical carcinogenesis. Oncogene. 2019;38(24):4755‐4772. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50. Li L, Zhou M, Chen B, et al. ALKBH5 promotes cadmium‐induced transformation of human bronchial epithelial cells by regulating PTEN expression in an m6A‐dependent manner. Ecotoxicol Environ Safety. 2021;224:112686. [DOI] [PubMed] [Google Scholar]
- 51. Qu T, Mou Y, Dai J, et al. Changes and relationship of N6‐methyladenosine modification and long non‐coding RNAs in oxidative damage induced by cadmium in pancreatic β‐cells. Toxicol Lett. 2021;343:56‐66. [DOI] [PubMed] [Google Scholar]
- 52. International Agency for Research in Cancer . Chromium, nickel and welding. IARC Monographs on the Evaluation of Carcinogenic Risks to Humans. Vol 49. International Agency for Research in Cancer; 1990:208‐213. [PMC free article] [PubMed] [Google Scholar]
- 53. International Agency for Research in Cancer . Cobalt in hard metals and cobalt sulfate, gallium arsenide, indium phosphide and vanadium pentoxide. IARC Monographs on the Evaluation of Carcinogenic Risks to Humans. Vol 86. International Agency for Research in Cancer; 2006:129‐132. [PMC free article] [PubMed] [Google Scholar]
- 54. International Agency for Research in Cancer . Inorganic and organic lead compounds. IARC Monographs on the Evaluation of Carcinogenic Risks to Humans. Vol 87. International Agency for Research in Cancer; 2006:370‐378. [PMC free article] [PubMed] [Google Scholar]
- 55. Solomon O, Di Segni A, Cesarkas K, et al. RNA editing by ADAR1 leads to context‐dependent transcriptome‐wide changes in RNA secondary structure. Nat Commun. 2017;8(1):1440. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56. Chen B, Xiong J, Ding J‐H, Yuan B‐F, Feng Y‐Q. Analysis of the effects of Cr(VI) exposure on mRNA modifications. Chem Res Toxicol. 2019;32(10):2078‐2085. [DOI] [PubMed] [Google Scholar]
- 57. Tang J, Zheng C, Zheng F, et al. Global N6‐methyladenosine profiling of cobalt‐exposed cortex and human neuroblastoma H4 cells presents epitranscriptomics alterations in neurodegenerative disease‐associated genes. Environ Pollut. 2020;266:115326. [DOI] [PubMed] [Google Scholar]
- 58. Dosunmu R, Alashwal H, Zawia NH. Genome‐wide expression and methylation profiling in the aged rodent brain due to early‐life Pb exposure and its relevance to aging. Mech Ageing Dev. 2012;133(6):435‐443. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59. International Agency for Research in Cancer . Some naturally occuring substances: food items and constituents, heterocyclic aromatic amines and mycotoxins. IARC Monographs on the Evaluation of Carcinogenic Risks to Humans. Vol 56. International Agency for Research in Cancer; 1993:445‐488.8411627 [Google Scholar]
- 60. Arumugam T, Ghazi T, Chuturgoon AA. Fumonisin B1 alters global m6A RNA methylation and epigenetically regulates Keap1‐Nrf2 signaling in human hepatoma (HepG2) cells. Arch Toxicol. 2021;95(4):1367‐1378. [DOI] [PubMed] [Google Scholar]
- 61. International Agency for Research in Cancer . Some N‐nitroso compounds. IARC Monographs on the Evaluation of the Carcinogenic Risk of Chemicals to Humans. Vol 17. International Agency for Research in Cancer; 1978:83‐124.355108 [Google Scholar]
- 62. Mittenbühler MJ, Saedler K, Nolte H, et al. Hepatic FTO is dispensable for the regulation of metabolism but counteracts HCC development in vivo. Mol Metab. 2020;42:101085. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63. International Agency for Research in Cancer . Acrolein, crotonaldehyde, and arecoline. IARC Monographs on the Evaluation of Carcinogenic Risks to Humans. Vol 128. International Agency for Research in Cancer; 2021:315‐317. [Google Scholar]
- 64. Li Y‐C, Cheng A‐J, Lee L‐Y, Huang Y‐C, Chang JT‐C. Multifaceted mechanisms of areca nuts in oral carcinogenesis: the molecular pathology from precancerous condition to malignant transformation. J Cancer. 2019;10(17):4054‐4062. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65. Li X, Xie X, Gu Y, et al. Fat mass and obesity‐associated protein regulates tumorigenesis of arecoline‐promoted human oral carcinoma. Cancer Med. 2021;10(18):6402‐6415. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66. Chiang C‐H, Wu C‐C, Lee L‐Y, et al. Proteomics analysis reveals involvement of Krt17 in areca nut‐induced oral carcinogenesis. J Proteome Res. 2016;15(9):2981‐2997. [DOI] [PubMed] [Google Scholar]
- 67. Streit E, Schatzmayr G, Tassis P, et al. Current situation of mycotoxin contamination and co‐occurrence in animal feed—focus on Europe. Toxins. 2012;4(10):788‐809. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68. International Agency for Research in Cancer . Some Naturally Occurring Substances. International Agency for Research on Cancer; 1993. [Google Scholar]
- 69. Wu K, Jia S, Zhang J, et al. Transcriptomics and flow cytometry reveals the cytotoxicity of aflatoxin B1 and aflatoxin M1 in bovine mammary epithelial cells. Ecotoxicol Environ Safety. 2021;209:111823. [DOI] [PubMed] [Google Scholar]
- 70. Wu J, Gan Z, Zhuo R, Zhang L, Wang T, Zhong X. Resveratrol attenuates aflatoxin B1‐induced ROS formation and increase of m6A RNA methylation. Animals. 2020;10(4):677. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71. Josse R, Dumont J, Fautrel A, Robin M‐A, Guillouzo A. Identification of early target genes of aflatoxin B1 in human hepatocytes, inter‐individual variability and comparison with other genotoxic compounds. Toxicol Appl Pharmacol. 2012;258(2):176‐187. [DOI] [PubMed] [Google Scholar]
- 72. Gauthier T, Duarte‐Hospital C, Vignard J, et al. Versicolorin A, a precursor in aflatoxins biosynthesis, is a food contaminant toxic for human intestinal cells. Environ Int. 2020;137:105568. [DOI] [PubMed] [Google Scholar]
- 73. Barbarino M, Giordano A. Assessment of the carcinogenicity of carbon nanotubes in the respiratory system. Cancers. 2021;13(6):1318. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74. Grosse Y, Loomis D, Guyton KZ, et al. Carcinogenicity of fluoro‐edenite, silicon carbide fibres and whiskers, and carbon nanotubes. Lancet Oncol. 2014;15(13):1427‐1428. [DOI] [PubMed] [Google Scholar]
- 75. Emerce E, Ghosh M, Öner D, et al. Carbon nanotube‐ and asbestos‐induced DNA and RNA methylation changes in bronchial epithelial cells. Chem Res Toxicol. 2019;32(5):850‐860. [DOI] [PubMed] [Google Scholar]
- 76. Søs Poulsen S, Jacobsen NR, Labib S, et al. Transcriptomic analysis reveals novel mechanistic insight into murine biological responses to multi‐walled carbon nanotubes in lungs and cultured lung epithelial cells. PLoS One. 2013;8(11):e80452. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77. Emadi A, Jones RJ, Brodsky RA. Cyclophosphamide and cancer: golden anniversary. Nat Rev Clin Oncol. 2009;6(11):638‐647. [DOI] [PubMed] [Google Scholar]
- 78. International Agency for Research in Cancer . Pharmaceuticals. IARC Monographs on the Evaluation of Carcinogenic Risks to Humans. Vol 100A. International Agency for Research on Cancer; 2012:82. [PMC free article] [PubMed] [Google Scholar]
- 79. Zhu M, Liu Y, Song Y, et al. The role of METTL3‐mediated N6‐methyladenosine (m6A) of JPH2 mRNA in cyclophosphamide‐induced cardiotoxicity. Front Cardiovasc Med. 2021;8:763469. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80. Huang B, Ding C, Zou Q, Wang W, Li H. Cyclophosphamide regulates N6‐methyladenosine and m6A RNA enzyme levels in human granulosa cells and in ovaries of a premature ovarian aging mouse model. Front Endocrinol. 2019;10:415. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81. International Agency for Research in Cancer . Polychlorinated biphenyls and polybrominated biphenyls. IARC Monographs on the Evaluation of Carcinogenic Risks to Humans. Vol 107. International Agency for Research in Cancer; 2016:423‐440. [PMC free article] [PubMed] [Google Scholar]
- 82. Aluru N, Karchner SI. PCB126 exposure revealed alterations in m6A RNA modifications in transcripts associated with AHR activation. Toxicol Sci. 2020;179(1):84‐94. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83. Klinge CM, Piell KM, Petri BJ, et al. Combined exposure to polychlorinated biphenyls and high‐fat diet modifies the global epitranscriptomic landscape in mouse liver. Environment Epigenet. 2021;7(1):dvab008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84. Ghosh S, Mitra PS, Loffredo CA, et al. Transcriptional profiling and biological pathway analysis of human equivalence PCB exposure in vitro: indicator of disease and disorder development in humans. Environ Res. 2015;138:202‐216. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85. International Agency for Research in Cancer . Some chemicals present in industrial and consumer products, food and drinking‐water. IARC Monographs on the Evaluation of Carcinogenic Risks to Humans. Vol 101. International Agency for Research in Cancer; 2013:260. [PMC free article] [PubMed] [Google Scholar]
- 86. Zhao T‐X, Wang J‐K, Shen L‐J, et al. Increased m6A RNA modification is related to the inhibition of the Nrf2‐mediated antioxidant response in di‐(2‐ethylhexyl) phthalate‐induced prepubertal testicular injury. Environ Pollut. 2020;259:113911. [DOI] [PubMed] [Google Scholar]
- 87. Seachrist DD, Bonk KW, Ho S‐M, Prins GS, Soto AM, Keri RA. A review of the carcinogenic potential of bisphenol A. Reproductive Toxicol. 2016;59:167‐182. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88. Dinwiddie MT, Terry PD, Chen J. Recent evidence regarding triclosan and cancer risk. Int J Environ Res Public Health. 2014;11(2):2209‐2217. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89. Hrelia P, Fimognari C, Maffei F, et al. The genetic and non‐genetic toxicity of the fungicide Vinclozolin. Mutagenesis. 1996;11(5):445‐453. [DOI] [PubMed] [Google Scholar]
- 90. Sun L, Ling Y, Jiang J, et al. Differential mechanisms regarding triclosan vs. bisphenol A and fluorene‐9‐bisphenol induced zebrafish lipid‐metabolism disorders by RNA‐Seq. Chemosphere. 2020;251:126318. [DOI] [PubMed] [Google Scholar]
- 91. Reitsma MB, Kendrick PJ, Ababneh E, et al. Spatial, temporal, and demographic patterns in prevalence of smoking tobacco use and attributable disease burden in 204 countries and territories, 1990–2019: a systematic analysis from the Global Burden of Disease Study 2019. Lancet. 2021;397(10292):2337‐2360. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92. International Agency for Research in Cancer . Tobacco smoke and involuntary smoking. IARC Monographs on the Evaluation of Carcinogenic Risks to Humans. Vol 83. International Agency for Research in Cancer; 2004:1179‐1413. [PMC free article] [PubMed] [Google Scholar]
- 93. Jin M, Li G, Liu W, et al. Cigarette smoking induces aberrant N6‐methyladenosine of DAPK2 to promote non‐small cell lung cancer progression by activating NF‐κB pathway. Cancer Lett. 2021;518:214‐229. [DOI] [PubMed] [Google Scholar]
- 94. Cheng C, Wu Y, Xiao T, et al. METTL3‐mediated m6A modification of ZBTB4 mRNA is involved in the smoking‐induced EMT in cancer of the lung. Mol Ther Nucleic Acids. 2020;23:487‐500. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
- 95. Wang J, Tan L, Jia B, et al. Downregulation of m6A reader YTHDC2 promotes the proliferation and migration of malignant lung cells via CYLD/NF‐κB pathway. Int J Biol Sci. 2021;17(10):2633‐2651. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96. Zhang J, Bai R, Li M, et al. Excessive miR‐25‐3p maturation via N6‐methyladenosine stimulated by cigarette smoke promotes pancreatic cancer progression. Nat Commun. 2019;10:1858. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97. Wu S, Zhang L, Deng J, et al. A novel micropeptide encoded by Y‐linked LINC00278 links cigarette smoking and AR signaling in male esophageal squamous cell carcinoma. Cancer Res. 2020;80(13):2790‐2803. [DOI] [PubMed] [Google Scholar]
- 98. Kupsco A, Gonzalez G, Baker BH, et al. Associations of smoking and air pollution with peripheral blood RNA N6‐methyladenosine in the Beijing truck driver air pollution study. Environ Int. 2020;144:106021. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99. International Agency for Research in Cancer . Outdoor air pollution. IARC Monographs on the Evaluation of Carcinogenic Risks to Humans. Vol 109. International Agency for Research in Cancer; 2016:36‐443. [PMC free article] [PubMed] [Google Scholar]
- 100. Yuan Q, Zhu H, Liu H, Wang M, Chu H, Zhang Z. METTL3 regulates PM2.5‐induced cell injury by targeting OSGIN1 in human airway epithelial cells. J Hazard Mater. 2021;415:125573. [DOI] [PubMed] [Google Scholar]
- 101. Li Z, Li N, Guo C, et al. The global DNA and RNA methylation and their reversal in lung under different concentration exposure of ambient air particulate matter in mice. Ecotoxicol Environ Safety. 2019;172:396‐402. [DOI] [PubMed] [Google Scholar]
- 102. Cai Y, Zheng K, Li RB, et al. Proteomics study on the differentially expressed proteins in c‐fos‐silenced cells exposed to PM2.5. Biomed Environ Sci. 2020;33(9):680‐689. [DOI] [PubMed] [Google Scholar]
- 103. Han X, Liu H, Zhang Z, et al. Epitranscriptomic 5‐methylcytosine profile in PM2.5‐induced mouse pulmonary fibrosis. Genomics Insights. 2020;18(1):41‐51. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104. International Agency for Research in Cancer . Solar and ultraviolet radiation. IARC Monographs on the Evaluation of Carcinogenic Risks to Humans. Vol 55. International Agency for Research in Cancer; 1992. [PMC free article] [PubMed] [Google Scholar]
- 105. Yang Z, Yang S, Cui Y‐H, et al. METTL14 facilitates global genome repair and suppresses skin tumorigenesis. PNAS. 2021;118(35). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106. Xiang Y, Laurent B, Hsu C‐H, et al. RNA m6A methylation regulates the ultraviolet‐induced DNA damage response. Nature. 2017;543(7646):573‐576. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107. Svobodová Kovaříková A, Stixová L, Kovařík A, et al. N6‐adenosine methylation in RNA and a reduced m3G/TMG level in non‐coding RNAs appear at microirradiation‐induced DNA lesions. Cells. 2020;9(2):360. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108. International Agency for Research in Cancer . Ionizing radiation, Part 1. X‐ and gamma (γ)‐radiation, and neutrons. IARC Monographs on the Evaluation of Carcinogenic Risks to Humans. Vol 75. International Agency for Research in Cancer; 2010:299‐301. [Google Scholar]
- 109. Zhang S, Dong J, Li Y, et al. Gamma‐irradiation fluctuates the mRNA N6‐methyladenosine (m6A) spectrum of bone marrow in hematopoietic injury. Environment Pollut. 2021;285:117509. [DOI] [PubMed] [Google Scholar]
- 110. International Agency for Research in Cancer . Biological agents. IARC Monographs on the Evaluation of Carcinogenic Risks to Humans. Vol 100B. International Agency for Research in Cancer; 2012:49‐196. [Google Scholar]
- 111. Kim G‐W, Imam H, Khan M, et al. HBV‐induced increased N6 methyladenosine modification of PTEN RNA affects innate immunity and contributes to HCC. Hepatology. 2021;73(2):533‐547. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112. Ng SA, Lee C. Hepatitis B virus X gene and hepatocarcinogenesis. J Gastroenterol. 2011;46(8):974‐990. [DOI] [PubMed] [Google Scholar]
- 113. Rao X, Lai L, Li X, Wang L, Li A, Yang Q. N6 ‐methyladenosine modification of circular RNA circ‐ARL3 facilitates Hepatitis B virus‐associated hepatocellular carcinoma via sponging miR‐1305. IUBMB Life. 2021;73(2):408‐417. [DOI] [PubMed] [Google Scholar]
- 114. Qu S, Jin L, Huang H, Lin J, Gao W, Zeng Z. A positive‐feedback loop between HBx and ALKBH5 promotes hepatocellular carcinogenesis. BMC Cancer. 2021;21:686. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115. Yang Z, Jiang X, Li D, Jiang X. HBXIP promotes gastric cancer via METTL3‐mediated MYC mRNA m6A modification. Aging. 2020;12(24):24967‐24982. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116. Lang F, Singh RK, Pei Y, Zhang S, Sun K, Robertson ES. EBV epitranscriptome reprogramming by METTL14 is critical for viral‐associated tumorigenesis. PLoS Pathog. 2019;15(6):e1007796. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 117. Xia T‐L, Li X, Wang X, et al. N(6)‐methyladenosine‐binding protein YTHDF1 suppresses EBV replication and promotes EBV RNA decay. EMBO Rep. 2021;22(4):e50128. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118. Tan B, Liu H, Zhang S, et al. Viral and cellular N6‐methyladenosine (m6A) and N6, 2′‐O‐dimethyladenosine (m6Am) epitranscriptomes in KSHV life cycle. Nat Microbiol. 2018;3(1):108‐120. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119. Ye F, Chen ER, Nilsen TW. Kaposi's sarcoma‐associated herpesvirus utilizes and manipulates RNA N6‐adenosine methylation to promote lytic replication. J Virol. 2017;91(16):e00466‐00417. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120. Oncolytic virus therapy (layout) by BioRender.com (2021). Retrieved from https://app.biorender.com/biorender-templates.
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.
Data Availability Statement
Data from this study are available from the corresponding author upon request.