Skip to main content
Function logoLink to Function
. 2021 Apr 23;2(3):zqab022. doi: 10.1093/function/zqab022

Perspectives on Epigenetics Alterations Associated with Smoking and Vaping

Zidian Xie 1, Irfan Rahman 2, Maciej L Goniewicz 3, Dongmei Li 1,
PMCID: PMC8788872  PMID: 35330676

Abstract

Epigenetic alterations, including DNA methylation, microRNA, and long noncoding RNA, play important roles in the pathogenesis of numerous respiratory health conditions and diseases. Exposure to tobacco smoking has been found to be associated with epigenetic changes in the respiratory tract. Marketed as a less harmful alternative to combustible cigarettes, electronic cigarette (e-cigarette) has rapidly gained popularity in recent years, especially among youth and young adults. Accumulative evidence from both animal and human studies has shown that e-cigarette use (vaping) is also linked to similar respiratory health conditions as observed with cigarette smoking, including wheezing, asthma, and COPD. This review aims to provide an overview of current studies on associations of smoking and vaping with epigenetic alterations in respiratory cells and provide future research directions in epigenetic studies related to vaping.

Keywords: epigenetics, e-cigarette, DNA methylation, miRNA, lncRNA

Introduction

Electronic cigarette (e-cigarette) is a battery-operated device that heats a liquid and allows users to inhale an aerosol, which usually contains nicotine, ultrafine particles, flavorings such as cytotoxic cinnamaldehyde and diacetyl (a chemical linked to a serious lung disease), volatile organic compounds such as respiratory irritants acrolein and acrylamide, lung cancer-causing chemicals such as formaldehyde and acetaldehyde, and heavy metals such as nickel, tin, and lead.1–4 Those respiratory toxicants and irritants present in aerosols generated from e-cigarettes are delivered to lungs with every puff taken by e-cigarette user. E-cigarettes have rapidly gained popularity in the United States in recent several years, especially among youth and young adults.1,5 E-cigarettes use (vaping) in youth has also been shown to be associated with subsequent cigarette smoking.6,7 Using large national survey data, our epidemiology studies have found the association of vaping with self-reported wheezing and chronic obstructive pulmonary disease (COPD) in US adults,8–10 and the association of vaping with self-reported difficulty concentrating, remembering, or making decisions in both US youth and adults.11,12 Our recent findings that e-cigarette aerosols cause oxidative stress, DNA damage, and inflammatory responses in human lung epithelial cells and mouse lungs indicated respiratory disease risk associated with e-cigarette use, which is consistent with previous studies.13–18 While flavoring chemicals are commonly used in e-cigarettes, there is limited information on the adverse health effects of those flavorings in e-cigarettes.19 We have shown that e-cigarette flavoring chemicals such as acetoin, ortho-vanillin, and maltol can stimulate the release of IL-8 in human bronchial epithelial cells (Beas2B) and human lung fibroblasts (HFL-1) to trigger inflammatory responses.14,20 A recent review of toxicological effects of tobacco and menthol-flavored e-cigarettes summarized the cytotoxicity and genotoxicity of tobacco and menthol-flavored e-cigarette inhalation exposure from both mice and human cell line studies, including increased oxidative stress, apoptosis, inflammation, DNA damage, and epithelial barrier dysfunction.21

Epigenetic alterations, including DNA methylation changes, dysregulation of microRNAs (miRNAs), and long noncoding RNAs (lncRNAs), have been found to play some important roles in the initiation and development of human diseases, as well as the interactions between genetic and environmental factors.22–33 DNA methylation is an epigenetic mechanism involving transferring a methyl group to the C5 position of the cytosine to form 5-methylcytosine, which could regulate gene expression.34 DNA methylation has been involved in autoimmune diseases, metabolic disorders, neurological disorders, and the aging process.35 miRNAs are a class of small noncoding RNAs with 21–25 nucleotides that play important regulatory roles in a wide range of cellular and biological processes such as immune regulation and inflammatory response.36–39 miRNAs have been reported to play crucial roles in pathophysiology of chronic inflammatory lung diseases and lung cancers.40–43 lncRNAs are a type of noncoding RNAs with more than 200 nucleotides in length.44–46 lncRNAs are a relative abundant component of transcriptome, which have been identified to have important cellular functions including regulation of gene transcription, cell differentiation, cancer cell invasion, and metastasis, and chromatin remodeling.47–50 Increasing evidence showed that lncRNAs regulate many physiological and pathological responses, including immune cell differentiation and activation, metabolism and glucose homeostasis, cardiovascular development, brain temporal and spatial expression patterns, and responses to environmental exposures. Deregulation of lncRNA is responsible for numerous diseases in mammals, and lncRNA has shown their significance as biomarkers in cancer prognosis and diagnosis.49–53

Epigenetic Alterations Associated with Smoking

Cigarette smoking is a well-known risk factor for cancer, cardiovascular disease, and COPD.54 Smoking exposure was considered a strong environmental modifier and has been found to be associated with epigenetic changes across tissue types in several studies.55–58 Smoking has been shown to modulate DNA methyltransferase 1 (DNMT1) and histone modification enzymes that are involved in pathogenesis of lung cancer and COPD.59 A previous metaanalysis of genome-wide DNA methylation studies found the association of smoking with DNA methylation changes (2623 CpG sites) linked with pulmonary functions, cancers, inflammatory diseases, and heart disease.60 Another study showed that smoking-associated DNA methylation biomarkers had a strong association with cognitive function, brain structure, physical health, and psychosocial health.58 Investigating the DNA methylation alterations associated with smoking not only helps us understand the mechanisms in pathogenesis of those diseases associated with smoking exposure, but also identifies biomarkers used for cigarette consumption prediction. For example, DNA methylation status at locus cg05575921 examined either in human whole blood or saliva samples could be used as a biomarker to differentiate smokers from nonsmokers as well as a predictor for daily cigarette consumption.61,62

In addition to DNA methylation alterations, smoking exposure can lead to noncoding RNAs change such as miRNAs and lncRNAs. Using whole blood samples from Framingham Heart Study participants, a six-miRNA signature of smoking was found to be associated with smoking-induced inflammation and reduced pulmonary functions.63 A genome-wide lncRNA expression in human lung tissue study showed that hundreds of lncRNAs were differentially expressed between five smokers with COPD, five smokers without COPD, and three nonsmokers without COPD, which suggest that smoking can change the expression of many lncRNAs.64 Gene enrichment analysis of identified significant lncRNAs showed changes in key pathogenic processes of COPD due to smoking.65 In vitro human bronchial epithelial (HBE) cell studies indicated the involvement of lncRNAs in the epithelial–mesenchymal transition and malignant transformation of the HBE cells induced by cigarette smoke extract.66

Epigenetic Alterations Associated with Vaping

Very few studies have investigated the epigenetic changes associated with vaping, and the potential association of DNA methylations, miRNAs, and lncRNAs with the health effects of vaping (Table 1). Previous mouse studies showed that maternal e-cigarette exposure could lead to global DNA methylation changes and cognitive problems such as deficits in short-term memory, reduced anxiety, and hyperactivity in the offspring.67,68 Using 45 human peripheral blood samples from exclusive vapers, smokers, and controls (nonusers), recent study showed significantly reduced methylation levels in LINE-1 repeat elements and global DNA hydroxymethylation in both vapers and smokers compared with controls.69 Meanwhile, no significant difference in those DNA methylation levels was observed between exclusive vapers and smokers.69

Table 1.

Current Literature on Epigenetic Alterations Associated with Vaping

Epigenetic alterations Study sample Tissue Groups Sample size Study results Reference
DNA methylation Mice Brain
  • Ambient air

  • E-cigarette aerosols with nicotine

  • E-cigarette aerosols without nicotine

8 in each group
  • Global DNA methylation was significantly increased in Group 3 compared to Group 1.

  • No significant change between Group 1 and Group 2.

Nguyen et al.67
DNA methylation Mice Lung
  • Ambient air

  • E-cigarette aerosols with nicotine

  • E-cigarette aerosols without nicotine

3 in each group Global DNA methylation was significantly increased in Groups 2 and 3 compared to Group 1. Chen et al.68
DNA methylation Human Peripheral blood
  • Vapers

  • Nonsmokers and nonvapers

15 in each group Demethylation in the LINE-1 repeat elements and decreased global methylation was significant between groups. Caliri et al.69
Exosomal miRNAs Human Plasma
  • Vapers

  • Nonsmokers

7 vapers and 8 nonsmokers 13 upregulated and 4 downregulated miRNAs were significant between groups. Singh et al.70
Exosomal lncRNAs Human Plasma
  • Vapers

  • Nonsmokers

6 in each group 13 upregulated and 5 downregulated lncRNAs were significant between groups Kaur et al.71

Exosomes have been reported recently to mediate cell-to-cell communication and affect many physiological processes.72–75 Exosomes are small nano-sized vesicles released by different cell types such as immune and structure cells.76 Exosomes contain enriched amount of surface proteins, regulatory proteins, mRNAs, miRNAs, and lncRNAs.77 Our recent study using plasma exosomes from seven cigarette smokers, seven vapers, and eight nonsmokers identified 24 significant miRNAs between smokers and nonsmokers, and 17 significant miRNAs between vapers and nonsmokers.70 Examination of the 24 miRNAs and 17 miRNAs showed 9 overlapped miRNAs, which indicated both similarities and differences in the miRNA perturbations between smoking and vaping.70 Identified miRNAs have been found to be involved in multiple biological pathways and processes in respiratory tract, such as regulation of nucleotide and nucleic acid metabolism and transcription factor activities using functional enrichment analysis. One of the identified significant miRNA hsa-let-7a-5p was found to be able to differentiate nonsmokers from tobacco users.70 Another study by our group using human plasma exosomes from six smokers, six vapers, and six nonsmokers identified seven significant lncRNAs between smokers and nonsmokers, 13 significant lncRNAs between vapers and nonsmokers.71 Examination of the 7 lncRNAs and 13 lncRNAs did not show overlapped lncRNAs, which indicated the differences in the lncRNA perturbations between smoking and vaping.71 Functional analysis of identified lncRNAs showed the involvement of those significant lncRNAs in the biological process such as steroid metabolism, hemopoiesis, and regulation of cell proliferation.71

Conclusions and Future Perspectives

Emerging evidences from mice and human studies suggest potential association of vaping with epigenetic alterations. Given the similarities (eg, nicotine) and differences (eg, combustion byproducts) in chemical compositions between tobacco smoke and e-cigarette aerosols, our recent studies found both common and different epigenetic changes when comparing smokers and vapers with controls.70,71 How these epigenetic alterations could assist us to understand relative risks of vaping compared to smoking is still unclear. Meanwhile, whether the differences in the epigenetic changes between vapers and cigarette smokers could result in toxicities unique to vaping such as the EVALI (e-cigarette, or vaping, product use-associated lung injury) cases in vapers awaits further investigation.78 However, the role of those identified epigenetic biomarkers in the etiology of vaping-associated diseases remains unanswered. All of current studies are cross-sectional. Thus, within-subject epigenetic alterations during the e-cigarette initiation and cessation process warrant further investigation. Meanwhile, how the epigenetic level changes when e-cigarette users switch flavors such as switching from fruit flavor (mainly including maltol and furaneol flavoring chemicals) to menthol (L-menthol flavoring chemicals) or tobacco (mainly 2,3,5-trimethylpyrazine flavoring chemicals) flavor in response to the US Food and Drug Administration (FDA) flavor enforcement policy are unknown, which need to be investigated in the future.

With the development of new methods and technologies in epigenetic studies, the epigenetic changes could be examined in an increasingly higher resolution to allow new discoveries in epigenetic changes associated with smoking and vaping. For example, the Perturb-ATAC approach that combines multiplexed CRISPR technique with chromatin accessibility analysis within a single cell to determine the role of transregulatory factors, the new method of selecting DNA methylation-based biomarkers through different biophysical properties to distinguish cancer cells from noncancerous cells, and a multiplexed mass cytometry assay to investigate the global levels of 40 different histone modifications at single-cell resolution.79–81 Meanwhile, studies on cell-type-specific and tissue-specific epigenetic changes will allow us to have a better and deeper understanding of the role of epigenetic changes plays in the etiology of disease development. These new technologies on epigenetic studies as well as tissue and cell-specific epigenetic studies could facilitate us to understand how smoking and vaping affect the epigenetic changes in different cell or tissue types, and potential health risks associated with them. Scientific endeavors in terms of understanding the epigenetic/epigenomic changes based on multiomics and spatial transcriptomics will help determine the toxicities of vaping at cellular/subcellular levels.

Funding

Research reported in this publication was supported by the National Cancer Institute of the National Institutes of Health (NIH) and the Food and Drug Administration (FDA) Center for Tobacco Products under Award Number U54CA228110. The content is solely the responsibility of the authors and does not necessarily represent the official views of the NIH or the Food and Drug Administration (FDA).

Conflict of Interest Statement

M.L.G. reports research grant from Pfizer and personal fees from Johnson & Johnson, outside this work. Other authors have no potential conflict of interest to declare.

Authors’ Contributions

Z.X., I.R., M.L.G., and D.L. conceived and designed the study; Z.X. and D.L. wrote the original manuscript; Z.X., I.R., M.L.G., and D.L. contributed to editing the manuscript.

References

  • 1.Callahan-Lyon P. Electronic cigarettes: human health effects. Tob Control 2014;231(Suppl 2):ii36–ii40. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Goniewicz ML, Gawron M, Smith DM, Peng M, Jacob P 3rd, Benowitz NL.. Exposure to nicotine and selected toxicants in cigarette smokers who switched to electronic cigarettes: a longitudinal within-subjects observational study. Nicotine Tob Res 2017;19(2):160–167. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Lerner CA, Sundar IK, Yao H, et al. Vapors produced by electronic cigarettes and e-juices with flavorings induce toxicity, oxidative stress, and inflammatory response in lung epithelial cells and in mouse lung. PLoS One 2015;10(2):e0116732. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Soussy S, El-Hellani A, Baalbaki R, Salman R, Shihadeh A, Saliba NA.. Detection of 5-hydroxymethylfurfural and furfural in the aerosol of electronic cigarettes. Tob Control 2016;25(Suppl 2):ii88–ii93. [DOI] [PubMed] [Google Scholar]
  • 5.Cullen KA, Ambrose BK, Gentzke AS, Apelberg BJ, Jamal A, King BA.. Notes from the field: use of electronic cigarettes and any tobacco product among middle and high school students - United States, 2011-2018. MMWR Morb Mortal Wkly Rep 2018;67(45):1276–1277. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Barrington-Trimis JL, Berhane K, Unger JB, et al. The e-cigarette social environment, e-cigarette use, and susceptibility to cigarette smoking. J Adolesc Health 2016;59(1):75–80. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Barrington-Trimis JL, Urman R, Berhane K, et al. E-cigarettes and future cigarette use. Pediatrics 2016;138(1):e20160379. doi: 10.1542/peds.2016-0379. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Li D, Sundar IK, McIntosh S, et al. Association of smoking and electronic cigarette use with wheezing and related respiratory symptoms in adults: cross-sectional results from the Population Assessment of Tobacco and Health (PATH) study, wave 2. Tob Control 2020;29(2):140–147. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Li D, Xie Z.. Cross-sectional association of lifetime electronic cigarette use with wheezing and related respiratory symptoms in U.S. adults. Nicotine Tob Res 2020;22(Suppl 1):S85–S92. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Xie Z, Ossip DJ, Rahman I, Li D.. Use of electronic cigarettes and self-reported chronic obstructive pulmonary disease diagnosis in adults. Nicotine Tob Res 2020;22(7):1155–1161. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Xie C, Xie Z, Li D.. Association of electronic cigarette use with self-reported difficulty concentrating, remembering, or making decisions in US youth. Tob Induc Dis 2020;18:106. doi: 10.18332/tid/130925. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Xie Z, Ossip DJ, Rahman I, O'Connor RJ, Li D.. Electronic cigarette use and subjective cognitive complaints in adults. PLoS One 2020;15(11):e0241599. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Dimitrov S, Hulteng E, Hong S.. Inflammation and exercise: inhibition of monocytic intracellular TNF production by acute exercise via β2-adrenergic activation. Brain Behav Immun 2017;61:60–8. doi: 10.1016/j.bbi.2016.12.017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Gerloff J, Sundar IK, Freter R, et al. Inflammatory response and barrier dysfunction by different e-cigarette flavoring chemicals identified by gas chromatography-mass spectrometry in e-liquids and e-vapors on human lung epithelial cells and fibroblasts. Appl In Vitro Toxicol 2017;3(1):28–40. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Muthumalage T, Prinz M, Ansah KO, Gerloff J, Sundar IK, Rahman I.. Inflammatory and Oxidative responses induced by exposure to commonly used e-cigarette flavoring chemicals and flavored e-liquids without nicotine. Front Physiol 2017;8:1130. doi: 10.3389/fphys.2017.01130. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Solleti SK, Bhattacharya S, Ahmad A, et al. MicroRNA expression profiling defines the impact of electronic cigarettes on human airway epithelial cells. Sci Rep 2017;7(1):1081. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Kaur G, Muthumalage T, Rahman I.. Mechanisms of toxicity and biomarkers of flavoring and flavor enhancing chemicals in emerging tobacco and non-tobacco products. Toxicol Lett 2018;288:143–155. doi: 10.1016/j.toxlet.2018.02.025. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Sundar IK, Rashid K, Gerloff J, Rangel-Moreno J, Li D, Rahman I.. Genetic ablation of histone deacetylase 2 leads to lung cellular senescence and lymphoid follicle formation in COPD/emphysema. FASEB J 2018;32(9):4955–4971. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Leigh NJ, Lawton RI, Hershberger PA, Goniewicz ML.. Flavourings significantly affect inhalation toxicity of aerosol generated from electronic nicotine delivery systems (ENDS). Tob Control 2016;25(Suppl 2):ii81–ii87. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Vreeke S, Peyton DH, Strongin RM.. Triacetin enhances levels of acrolein, formaldehyde hemiacetals, and acetaldehyde in electronic cigarette aerosols. ACS Omega 2018;3(7):7165–7170. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Kaur G, Gaurav A, Lamb T, Perkins M, Muthumalage T, Rahman I.. Current perspectives on characteristics, compositions, and toxicological effects of e-cigarettes containing tobacco and menthol/mint flavors. Front Physiol 2020;11:613948. doi: 10.3389/fphys.2020.613948. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Forouzanfar F, Shojapour M, Asgharzade S, Amini E.. Causes and consequences of microRNA dysregulation following cerebral ischemia-reperfusion injury. CNS Neurol Disord Drug Targets 2019;18(3):212–221. [DOI] [PubMed] [Google Scholar]
  • 23.Tan L, Yu JT, Tan L.. Causes and consequences of microRNA dysregulation in neurodegenerative diseases. Mol Neurobiol 2015;51(3):1249–1262. [DOI] [PubMed] [Google Scholar]
  • 24.Nieto-Diaz M, Esteban FJ, Reigada D, et al. MicroRNA dysregulation in spinal cord injury: causes, consequences and therapeutics. Front Cell Neurosci 2014;8:53. doi: 10.3389/fncel.2014.00053. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Iorio MV, Croce CM.. Causes and consequences of microRNA dysregulation. Cancer J 2012;18(3):215–222. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Croce CM. Causes and consequences of microRNA dysregulation in cancer. Nat Rev Genet 2009;10(10):704–714. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Maass PG, Luft FC, Bahring S.. Long non-coding RNA in health and disease. J Mol Med (Berl) 2014;92(4):337–346. [DOI] [PubMed] [Google Scholar]
  • 28.DiStefano JK. The emerging role of long noncoding RNAs in human disease. Methods Mol Biol 2018;1706:91–110. doi: 10.1007/978-1-4939-7471-9_6. [DOI] [PubMed] [Google Scholar]
  • 29.Esteller M. Non-coding RNAs in human disease. Nat Rev Genet 2011;12(12):861–874. [DOI] [PubMed] [Google Scholar]
  • 30.Mongelli A, Martelli F, Farsetti A, Gaetano C.. The dark that matters: long non-coding RNAs as master regulators of cellular metabolism in non-communicable diseases. Front Physiol 2019;10:369. doi: 10.3389/fphys.2019.00369. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Mittelstrass K, Waldenberger M.. DNA methylation in human lipid metabolism and related diseases. Curr Opin Lipidol 2018;29(2):116–124. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Gao D, Zhu B, Sun H, Wang X.. Mitochondrial DNA methylation and related disease. Adv Exp Med Biol 2017;1038:117–132. doi: 10.1007/978-981-10-6674-0_9. [DOI] [PubMed] [Google Scholar]
  • 33.Fragou D, Pakkidi E, Aschner M, Samanidou V, Kovatsi L.. Smoking and DNA methylation: correlation of methylation with smoking behavior and association with diseases and fetus development following prenatal exposure. Food Chem Toxicol 2019;129:312–327. doi: 10.1016/j.fct.2019.04.059. [DOI] [PubMed] [Google Scholar]
  • 34.Moore LD, Le T, Fan G.. DNA methylation and its basic function. Neuropsychopharmacology 2013;38(1):23–38. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Jin Z, Liu Y.. DNA methylation in human diseases. Genes Dis 2018;5(1):1–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Shivdasani RA. MicroRNAs: regulators of gene expression and cell differentiation. Blood 2006;108(12):3646–3653. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Ambros V, Chen X.. The regulation of genes and genomes by small RNAs. Development 2007;134(9):1635–1641. [DOI] [PubMed] [Google Scholar]
  • 38.Wahid F, Shehzad A, Khan T, Kim YY.. MicroRNAs: synthesis, mechanism, function, and recent clinical trials. Biochim Biophys Acta 2010;1803(11):1231–1243. [DOI] [PubMed] [Google Scholar]
  • 39.Yang P, Cai Z, Wu K, Hu Y, Liu L, Liao M.. Identification of key microRNAs and genes associated with abdominal aortic aneurysm based on the gene expression profile. Exp Physiol 2019;105(1):160–173. [DOI] [PubMed] [Google Scholar]
  • 40.Zhang W, Liu J, Wang G.. The role of microRNAs in human breast cancer progression. Tumour Biol 2014;35(7):6235–6244. [DOI] [PubMed] [Google Scholar]
  • 41.Peng Y, Croce CM.. The role of MicroRNAs in human cancer. Signal Transduct Target Ther 2016;1:15004. doi: 10.1038/sigtrans.2015.4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Zheng Q, Chen C, Guan H, Kang W, Yu C.. Prognostic role of microRNAs in human gastrointestinal cancer: a systematic review and meta-analysis. Oncotarget 2017;8(28):46611–46623. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Lamichhane SR, Thachil T, De Ieso P, Gee H, Moss SA, Milic N.. Prognostic role of microRNAs in human non-small-cell lung cancer: a systematic review and meta-analysis. Dis Markers 2018;2018:8309015. doi: 10.1155/2018/8309015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Kung JT, Colognori D, Lee JT.. Long noncoding RNAs: past, present, and future. Genetics 2013;193(3):651–669. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Zampetaki A, Albrecht A, Steinhofel K.. Long non-coding RNA structure and function: is there a link? Front Physiol 2018;9:1201. doi: 10.3389/fphys.2018.01201. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Paralkar VR, Weiss MJ.. Long noncoding RNAs in biology and hematopoiesis. Blood 2013;121(24):4842–4846. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Dhanoa JK, Sethi RS, Verma R, Arora JS, Mukhopadhyay CS.. Long non-coding RNA: its evolutionary relics and biological implications in mammals: a review. J Anim Sci Technol 2018;60:25. doi: 10.1186/s40781-018-0183-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Wei CW, Luo T, Zou SS, Wu AS.. The role of long noncoding RNAs in central nervous system and neurodegenerative diseases. Front Behav Neurosci 2018;12:175. doi: 10.3389/fnbeh.2018.00175. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Chi J, Liu T, Shi C, et al. Long non-coding RNA LUCAT1 promotes proliferation and invasion in gastric cancer by regulating miR-134-5p/YWHAZ axis. Biomed Pharmacother 2019;118:109201. doi: 10.1016/j.biopha.2019.109201. [DOI] [PubMed] [Google Scholar]
  • 50.Rankin CR, Shao L, Elliott J, et al. The IBD associated long non-coding RNA IFNG-AS1 regulates the balance between inflammatory and anti-inflammatory cytokine production after T-cell stimulation. Am J Physiol Gastrointest Liver Physiol 2019;318(1):G34-G40. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Vakkilainen S, Skoog T, Einarsdottir E, et al. The human long non-coding RNA gene RMRP has pleiotropic effects and regulates cell-cycle progression at G2. Sci Rep 2019;9(1):13758. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Wang H, Liao S, Li H, Chen Y, Yu J.. Long non-coding RNA TUG1 sponges Mir-145a-5p to regulate microglial polarization after oxygen-glucose deprivation. Front Mol Neurosci 2019;12:215. doi: 10.3389/fnmol.2019.00215. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Zhang L, Meng X, Zhu XW, et al. Long non-coding RNAs in oral squamous cell carcinoma: biologic function, mechanisms and clinical implications. Mol Cancer 2019;18(1):102. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Lee KW, Pausova Z.. Cigarette smoking and DNA methylation. Front Genet 2013;4:132. doi: 10.3389/fgene.2013.00132. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Ho SM, Johnson A, Tarapore P, Janakiram V, Zhang X, Leung YK.. Environmental epigenetics and its implication on disease risk and health outcomes. ILAR J 2012;53(3–4):289–305. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Nielsen CH, Larsen A, Nielsen AL.. DNA methylation alterations in response to prenatal exposure of maternal cigarette smoking: a persistent epigenetic impact on health from maternal lifestyle? Arch Toxicol 2016;90(2):231–245. [DOI] [PubMed] [Google Scholar]
  • 57.Zong D, Liu X, Li J, Ouyang R, Chen P.. The role of cigarette smoke-induced epigenetic alterations in inflammation. Epigenetics Chromatin 2019;12(1):65. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Corley J, Cox SR, Harris SE, et al. Epigenetic signatures of smoking associate with cognitive function, brain structure, and mental and physical health outcomes in the Lothian Birth Cohort 1936. Transl Psychiatry 2019;9(1):248. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Sundar IK, Mullapudi N, Yao H, Spivack SD, Rahman I.. Lung cancer and its association with chronic obstructive pulmonary disease: update on nexus of epigenetics. Curr Opin Pulm Med 2011;17(4):279–285. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Joehanes R, Just AC, Marioni RE, et al. Epigenetic signatures of cigarette smoking. Circ Cardiovasc Genet 2016;9(5):436–447. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Philibert R, Dogan M, Beach SRH, Mills JA, Long JD.. AHRR methylation predicts smoking status and smoking intensity in both saliva and blood DNA. Am J Med Genet B Neuropsychiatr Genet 2020;183(1):51–60. [DOI] [PubMed] [Google Scholar]
  • 62.Dawes K, Andersen A, Vercande K, et al. Saliva DNA methylation detects nascent smoking in adolescents. J Child Adolesc Psychopharmacol 2019;29(7):535–544. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Willinger CM, Rong J, Tanriverdi K, et al. MicroRNA signature of cigarette smoking and evidence for a putative causal role of microRNAs in smoking-related inflammation and target organ damage. Circ Cardiovasc Genet 2017;10(5):e001678. doi: 10.1161/CIRCGENETICS.116.001678. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Karlsson O, Baccarelli AA.. Environmental health and long non-coding RNAs. Curr Environ Health Rep 2016;3(3):178–187. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Bi H, Zhou J, Wu D, et al. Microarray analysis of long non-coding RNAs in COPD lung tissue. Inflamm Res 2015;64(2):119–126. [DOI] [PubMed] [Google Scholar]
  • 66.Lu L, Luo F, Liu Y, et al. Posttranscriptional silencing of the lncRNA MALAT1 by miR-217 inhibits the epithelial-mesenchymal transition via enhancer of zeste homolog 2 in the malignant transformation of HBE cells induced by cigarette smoke extract. Toxicol Appl Pharmacol 2015;289(2):276–285. [DOI] [PubMed] [Google Scholar]
  • 67.Nguyen T, Li GE, Chen H, Cranfield CG, McGrath KC, Gorrie CA.. Maternal e-cigarette exposure results in cognitive and epigenetic alterations in offspring in a mouse model. Chem Res Toxicol 2018;31(7):601–611. [DOI] [PubMed] [Google Scholar]
  • 68.Chen H, Li G, Chan YL, et al. Maternal e-cigarette exposure in mice alters DNA methylation and lung cytokine expression in offspring. Am J Respir Cell Mol Biol 2018;58(3):366–377. [DOI] [PubMed] [Google Scholar]
  • 69.Caliri AW, Caceres A, Tommasi S, Besaratinia A.. Hypomethylation of LINE-1 repeat elements and global loss of DNA hydroxymethylation in vapers and smokers. Epigenetics 2020;15(8):816–829. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Nolte-'t Hoen E, Cremer T, Gallo RC, Margolis LB.. Extracellular vesicles and viruses: are they close relatives? Proc Natl Acad Sci USA 2016;113(33):9155–9161. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Balducci E, Leroyer AS, Lacroix R, et al. Extracellular vesicles from T cells overexpress miR-146b-5p in HIV-1 infection and repress endothelial activation. Sci Rep 2019;9(1):10299. doi: 10.1038/s41598-019-44743-w. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Han L, Lam EWF, Sun Y.. Extracellular vesicles in the tumor microenvironment: old stories, but new tales. Mol Cancer 2019;18(1):59. doi: 10.1186/s12943-019-0980-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Li N, Zhao L, Wei YK, Ea VL, Nian H, Wei R.. Recent advances of exosomes in immune-mediated eye diseases. Stem Cell Res Ther 2019;10(1):278. doi: 10.1186/s13287-019-1372-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Paolicelli RC, Bergamini G, Rajendran L.. Cell-to-cell communication by extracellular vesicles: focus on microglia. Neuroscience 2019;405:148–157. doi: 10.1016/j.neuroscience.2018.04.003. [DOI] [PubMed] [Google Scholar]
  • 75.Seo Y, Kim HS, Hong IS.. Stem cell-derived extracellular vesicles as immunomodulatory therapeutics. Stem Cells Int 2019;2019:5126156. doi: 10.1155/2019/5126156. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Singh KP, Maremanda KP, Li D, Rahman I.. Exosomal microRNAs are novel circulating biomarkers in cigarette, waterpipe smokers, E-cigarette users and dual smokers. BMC Med Genomics 2020;13(1):128. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Kaur G, Singh K, Maremanda KP, Li D, Chand HS, Rahman I.. Differential plasma exosomal long non-coding RNAs expression profiles and their emerging role in E-cigarette users, cigarette, waterpipe, and dual smokers. PLoS One 2020;15(12):e0243065. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.McDonough SR, Rahman I, Sundar IK.. Recent updates on biomarkers of exposure and systemic toxicity in e-cigarette users and EVALI. Am J Physiol Lung Cell Mol Physiol 2021;320(5):L661-L679. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Rubin AJ, Parker KR, Satpathy AT, et al. Coupled single-cell CRISPR screening and epigenomic profiling reveals causal gene regulatory networks. Cell 2019;176(1–2):361–376 e317. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Sina AA, Carrascosa LG, Liang Z, et al. Epigenetically reprogrammed methylation landscape drives the DNA self-assembly and serves as a universal cancer biomarker. Nat Commun 2018;9(1):4915. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Cheung P, Vallania F, Warsinske HC, et al. Single-cell chromatin modification profiling reveals increased epigenetic variations with aging. Cell 2018;173(6):1385–1397 e1314. [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from Function are provided here courtesy of American Physiological Society

RESOURCES