Next Article in Journal
EGFR- and Integrin αVβ3-Targeting Peptides as Potential Radiometal-Labeled Radiopharmaceuticals for Cancer Theranostics
Previous Article in Journal
Synthesis, Optimization and Molecular Self-Assembly Behavior of Alginate-g-Oleylamine Derivatives Based on Ugi Reaction for Hydrophobic Drug Delivery
Previous Article in Special Issue
Mutations in Genes Encoding Subunits of the RNA Exosome as a Potential Novel Cause of Thrombotic Microangiopathy
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Exploring the Utility of Long Non-Coding RNAs for Assessing the Health Consequences of Vaping

Department of Population & Public Health Sciences, USC Keck School of Medicine, University of Southern California, M/C 9603, Los Angeles, CA 90033, USA
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2024, 25(15), 8554; https://doi.org/10.3390/ijms25158554
Submission received: 4 July 2024 / Revised: 31 July 2024 / Accepted: 2 August 2024 / Published: 5 August 2024

Abstract

:
Electronic cigarette (e-cig) use, otherwise known as “vaping”, is widespread among adolescent never-smokers and adult smokers seeking a less-harmful alternative to combustible tobacco products. To date, however, the long-term health consequences of vaping are largely unknown. Many toxicants and carcinogens present in e-cig vapor and tobacco smoke exert their biological effects through epigenetic changes that can cause dysregulation of disease-related genes. Long non-coding RNAs (lncRNAs) have emerged as prime regulators of gene expression in health and disease states. A large body of research has shown that lncRNAs regulate genes involved in the pathogenesis of smoking-associated diseases; however, the utility of lncRNAs for assessing the disease-causing potential of vaping remains to be fully determined. A limited but growing number of studies has shown that lncRNAs mediate dysregulation of disease-related genes in cells and tissues of vapers as well as cells treated in vitro with e-cig aerosol extract. This review article provides an overview of the evolution of e-cig technology, trends in use, and controversies on the safety, efficacy, and health risks or potential benefits of vaping relative to smoking. While highlighting the importance of lncRNAs in cell biology and disease, it summarizes the current and ongoing research on the modulatory effects of lncRNAs on gene regulation and disease pathogenesis in e-cig users and in vitro experimental settings. The gaps in knowledge are identified, priorities for future research are highlighted, and the importance of empirical data for tobacco products regulation and public health is underscored.

1. Introduction

Electronic cigarettes (e-cigs) are electronic nicotine delivery systems (ENDS) that simulate tobacco smoking [1,2]. E-cigs are battery-powered devices that heat a solution (e-liquid) containing varying concentrations of humectants, nicotine, and flavors to produce a vapor, which users inhale through a mouthpiece [1,3]. E-cig use is also known as “vaping”, and e-cig users are commonly referred to as “vapers” [4,5]. E-cigs have been promoted originally as safe and subsequently as a less-harmful alternative to tobacco cigarettes [6,7]. This perception has promulgated by the fact that e-cigs “heat” e-liquid to render inhalable vapor, unlike traditional cigarettes that burn tobacco leaves to generate smoke [1,7]. Importantly, chemical analyses of e-cig vapor and e-liquid have shown the presence of many of the same toxicants and carcinogens as those found in cigarette smoke, although generally at substantially lower levels [1,4,8]. Currently, e-cig use is widespread among adolescent never-smokers and adult smokers seeking a reduced-harm substitute for combustible cigarettes [2,6,7,9]. To date, however, the long-term health effects of vaping are largely unknown [7,8,10]. Investigating the biological consequences of e-cig use can clarify the health risks or potential benefits of vaping in adult former-smokers as well as in youth never-smokers who have taken up this controvertible habit [5,8].
Many toxic and carcinogenic compounds present in e-cig vapor and cigarette smoke exert their biological effects through epigenetic changes that can cause dysregulation of disease-related genes [11,12,13,14,15,16,17,18,19,20,21,22]. The toxic and carcinogenic constituents of e-cig vapor and cigarette smoke, such as carbonyl compounds, volatile organic compounds (VOC), free radicals, and heavy metals, can induce a wide range of DNA lesions [1,10], many of which may interfere with the epigenetic machinery, e.g., by inhibiting the binding of epigenetic enzymes to their targets [15,23]. There may also exist a competitive demand for the metabolism of the chemicals present in e-cig vapor and cigarette smoke [15] and the cofactors/cosubstrates required for the epigenetic modifying enzymes [23]. Moreover, oxidative DNA damage induced by e-cig vapor and cigarette smoke can affect transcriptional regulatory elements and other epigenetic modifications [15,24].
Long non-coding RNAs (lncRNAs) have emerged as prime regulators of gene expression in health and disease states [25,26,27,28]. There is a mounting recognition of lncRNA-mediated dysregulation of genes in human diseases, including tobacco-related diseases [29,30,31,32,33,34,35,36]. Also, growing evidence shows the modulatory effects of lncRNAs on gene expression in response to specific cues or external stimuli such as tobacco product use [37,38]. While extensive research has shown that lncRNAs regulate genes involved in the pathogenesis of smoking-associated diseases [39,40,41,42], the utility of lncRNAs for assessing the disease-causing potential of vaping remains to be fully explored. Determining how lncRNAs regulate disease-related genes in e-cig users can lead to the identification of novel biomarkers of exposure and early effects for vaping. These biomarkers will have significant utility for assessing the health risks or potential benefits of vaping relative to smoking. This information is urgently needed by the regulatory agencies, including the United States Food and Drug Administration (FDA), which has the authority to regulate the manufacturing, marketing, and distribution of tobacco products to protect public health [6,9,43].
There is limited but growing research on lncRNA-mediated gene regulation in biospecimens from e-cig users and in cells treated in vitro with e-cig aerosol extract [13,44,45]. The present review provides an overview of the evolution of e-cigs as a highly consequential ENDS product, describes trends in e-cig use, and discusses the competing views on the public health impact of vaping. While highlighting the importance of lncRNAs in cell biology and disease, it summarizes the existing research on the modulatory effects of lncRNAs on disease-related molecular pathways and gene networks in e-cig users and in vitro experimental settings. The gaps in knowledge are identified, priorities for future research are highlighted, and the importance of empirical data for tobacco products regulation and public health is underscored.

2. Search Strategy and Selection Criteria

PubMed search was conducted to identify references using the following terms: “vaping”, “smoking”, “tobacco”, “cigarette”, “electronic cigarette”, “e-cigs”, “electronic nicotine delivery systems”, “ENDS”, “youth vaping”, “long non-coding RNA”, “lncRNA”, and “non-coding RNA”. The search terms were used both individually and in combination with each other. All English-written references, published on or before 30 June 2024, were considered. Where appropriate, publicly available databases and scientific reports from regulatory agencies and/or academia as well as news publications were considered; in all cases, cited sources were identified with a link to the published materials. To limit the number of citations, updated reviews were used rather than individual research articles, unless otherwise indicated.

3. E-Cig Technology

E-cigs have been promoted as a safe or less harmful alternative to tobacco cigarettes and as an aid to smoking cessation [2,5,8]. E-cigs are handheld, battery-powered devices that heat a liquid and convert it into a vapor, which users inhale into their lungs [1,3,46]. The liquid, also known as “e-liquid/e-juice”, is a mixture of propylene glycol (PG), glycerin/vegetable glycerin (VG), flavorings, and varying concentrations of nicotine, although nicotine-free e-liquid is also available [1,10,46]. Since the introduction of e-cigs to the U.S. market in 2007, these devices have evolved rapidly and substantially, from the first-generation “cig-a-like”, which was designed to look like a traditional tobacco cigarette, to the second-generation vape pens, third-generation box mods, and the currently popular fourth-generation pod-based devices [1,10]. The fast and significant changes in the design and features of e-cig devices have been accompanied by massive diversification of e-liquids [47,48,49]. With hundreds of chemicals being added to e-liquids to produce appealing flavors to every man, woman, and child, there are numerous combinations of chemicals in the ~20,000 e-liquids currently available in the market [49,50,51]. The combination of these chemicals, when vaporized altogether, can give rise to many more chemicals with uncertain toxicity profiles [52,53,54,55,56,57,58,59].

4. Chemical Composition of E-Cig Liquid and Vapor

Chemical analyses of e-cig liquid and vapor have revealed the presence of some of the same toxicants and carcinogens as those found in cigarette smoke, although mostly at substantially lower levels—these chemicals include carbonyl compounds, VOC, free radicals, and heavy metals, among others [1,3,10,60,61]. E-cig vapor also contains chemicals that are not present in cigarette smoke [51,62]. The latter compounds likely arise from mixing of the e-liquid ingredients and/or vaporization of humectants (PG/VG), flavorings, or chemicals leached from the device components [53,63]. At least seven categories of harmful and potentially harmful substances have been identified in e-cig liquid and vapor, including carbonyls, VOC, nicotine, nanoparticles, trace metal elements, bacterial endotoxins, and β-glucans [10]. The reduced levels of toxicants and carcinogens in e-cig vapor are consistent with the fact that e-cigs, unlike conventional cigarettes, do not “burn” tobacco to produce aerosol for inhalation [3,4]. This has led, in part, to the perception that e-cig use is safe or less-harmful than tobacco smoking [2]. While the lower levels of toxicants and carcinogens in e-cig vapor may imply reduced health risks, they cannot, however, equate to no risk [64]. In fact, exposure to many of the same constituents of e-cig vapor, at varying concentrations, has been associated with a variety of diseases, including cardiovascular-, immune-related (inflammatory), and respiratory diseases and cancer, among others [1,3,10,15,23,65].

5. Safety, Efficacy, and Health Risks or Benefits of E-Cig Use

There are competing views on e-cigs safety, efficacy, and health risks/benefits [6,66,67,68,69]. On the one hand, advocates for e-cigs claim that vaping, especially when combined with counseling and behavioral therapy, helps smokers quit; meta-analyses of dozens of randomized clinical trials mostly support this claim [7,9]. An important caveat is that e-cigs as a medical intervention but not a consumer product may help adult smokers quit [2,9,70,71,72,73]. Opponents of e-cigs argue that numerous population-based studies [74,75,76] confirm that e-cigs as a consumer product are not effective for smoking cessation [7,9,77]. As it stands, e-cigs are not approved as a medical intervention anywhere in the world; instead, e-cigs are consumer products that can be bought if one is of a certain age—e-cigs can be used however one wishes, as much as one likes, and as frequently as one wants [76,78]. Proponents of e-cigs also claim that the use of e-cigs has led to a decline in youth smoking [2,79]. Opponents, however, counter that e-cigs are addicting a new generation of teens and youth who would have never experimented with smoking in the first place [68,78]. It is also argued that vaping may lead to smoking and “renormalization” of this unhealthy habit [72,80,81]. While advocates of e-cigs claim that e-cigs cause less harm than tobacco cigarettes, opponents disagree [6,66,68,78,82].

6. Disentangling the Biological Effects of E-Cig Use in Adult Vapers

Adult e-cig users are likely to have a prior history of smoking (i.e., ex-smokers/current vapers) or co-use e-cigs and combustible cigarettes (i.e., dual users) [2,6,9,64]. The existing literature on the “potential” health risks of vaping is often criticized by the fact that the study participants in many reports include adult e-cig users with “past” or “current” smoking habits (ex-smokers/current vapers or dual users, respectively) [4,67]. This has complicated the interpretation of the results, as it is less than certain whether the observed effects in e-cig users are due to (i) persistent effects of past smoking (in former smokers) or current smoking (in dual users), (ii) current vaping only, or (iii) a combination of the two [2,4,6].

7. LncRNAs: Mechanisms and Functions

Although the vast majority of the human genome is transcribed into RNA, only ~2% of this RNA is protein-coding [83,84,85,86]. The most abundant and diverse class of non-coding RNAs is lncRNAs [85,87]. LncRNAs (≥200 nucleotides in length) are increasingly recognized as versatile regulators of gene expression at the transcriptional, post-transcriptional, translational, or post-translational levels [28,88,89,90]. LncRNA are often classified into different sub-types, including intergenic (long intergenic noncoding RNAs (lincRNAs)), intronic, and sense and antisense lncRNAs, with each sub-type having different genomic position in relation to genes and exons [91,92]. The majority of lncRNAs are overlapping sense and antisense transcripts, which mostly include protein-coding genes [83], thus resulting in a complex hierarchy of overlapping isoforms [93]. Genomic sequences within these transcriptional foci are frequently shared within a number of coding and non-coding transcripts in the sense and antisense directions relative to annotated genes [94]. LncRNAs are also derived from “pseudogenes” that are abundantly present in the human genome. Some of the ~15,000 pseudogenes identified in the human genome [95] have been shown to be functional [96,97]. In mammals, lncRNAs are dynamically expressed at various stages of development [98] and during differentiation of stem, muscle, mammary gland, immune, and neural cells, among others [99,100]. There is a transition in lncRNA expression during development, with broadly expressed and conserved lncRNAs evolving into an increasing number of lineage-specific and organ-specific lncRNAs [101,102]. The regulatory function of lncRNAs is primarily attributed to their roles as (1) “signals” in response to different stimuli or combinatorial actions of transcription factors (TFs); (2) “guides” to recruit histone-modifying enzymes or chromatin modifiers to the positions of target genes either in cis or in trans action; (3) “decoys” or “sponges” to titrate TFs or other intermediary regulatory entities such as RNA/DNA molecules (e.g., microRNAs (miRNA)) and sequester them away from their respective target site; and (4) “scaffolds” to recruit protein partners together by forming functional ribonucleoprotein complexes [103,104,105,106,107,108]. Various sub-types of lncRNAs exert their regulatory effects through participation in competing endogenous RNAs (ceRNAs) regulation, transcription regulation, and epigenetic regulation [106,107,108]. There is a growing appreciation of lncRNA-mediated dysregulation of genes in human diseases, such as cardiovascular, respiratory, and immune diseases and cancer, among others [29,30,31,32,33,34,35,36]. Emerging data show the modulatory effects of lncRNAs on gene expression in response to specific cues or external stimuli such as tobacco product use [37,38].

LncRNAs Classification, Modes of Action, and Regulation in Health and Disease

Although consensus on how to classify lncRNAs is yet to be reached [102], one widely employed method of classification is based on the genomic position of lncRNAs relative to other genes, e.g., protein-coding genes [91]. Accordingly, lncRNAs are classified into three categories. (I) Intronic lncRNAs are transcribed from the intron of a sense or antisense gene; (II) intergenic lncRNA genes do not overlap with other genes; and (III) antisense or divergent lncRNA genes either overlap or are in close proximity to a sense gene and are localized on the opposite DNA strand [91,92,102]. Another method of classification relies on the mode of action and regulation of lncRNAs; these include but are not limited to target gene regulation through either cis or trans action [91]; molecular role, e.g., enhancer RNAs [109], competitive endogenous RNAs [110], and architectural RNAs [111]; transcriptional regulation, e.g., damage-induced lncRNAs [112] and stress-induced promoter-associated antisense lncRNAs [113]; or physiological relevance, e.g., angio-LncRs (MALAT1, MANTIS, PUNISHER, MEG3, MIAT, SENCR, and GATA6-AS) [114]. The process of transcription per se has also been suggested to give an additional dimension to the original function of lncRNAs [28]. For example, some lncRNAs may not be exclusively non-coding, and while retaining their original function, they may also give rise to small functional peptides otherwise known as micropeptides [115]. Alternatively, some lncRNA loci may be part of a 3D nuclear construct permissive to the chromatin environment and gene regulation at the neighboring loci [116]. LncRNAs can be found anywhere in the cell, although the majority of lncRNAs are localized to the nucleus, which may stem from inefficient splicing events [27]. Unlike mRNAs, most lncRNAs have low expression levels, are evolutionarily less well conserved, and exhibit high cell-type specificity or tissue specificity [28]. Promoter regions of lncRNAs contain fewer TF binding motifs [115]. LncRNAs can form secondary and tertiary structures and contain functional RNA elements and nuclear localization sequences, which are key components of gene regulation [27,117]. Together, these features enable lncRNAs to serve a wide range of regulatory functions in physiologic and pathologic conditions [115]. For example, in mammals, lncRNAs have been demonstrated to be instrumental in processes such as the p53-mediated response to DNA damage [118], V(D)J recombination and class-switch recombination in immune cells [119], cytokine expression [120], endotoxic shock [121], inflammation and neuropathic pain [122,123,124], cholesterol biosynthesis and homeostasis [125,126], growth hormone and prolactin production [127], glucose metabolism [128,129], cellular signal transduction and transport pathways [130,131,132], synapse function [133,134], and learning [135]. A growing body of research shows the importance of lncRNAs in the regulation of genes involved in disease pathogenesis [102]. Altered expression of lncRNAs has been demonstrated in a wide variety of diseases ranging from cancer to cardiovascular, respiratory, and immune diseases, among others [29,30,31,32,33,34,35,36].

8. LncRNAs in Vaping Research

Aberrant expression of lncRNAs has been demonstrated in biospecimens from e-cig users [13,44] and cells treated in vitro with e-cig aerosol extract [45]. Le et al. [45] performed microarray expression analysis on human induced pluripotent stem cell-derived endothelial cells (iPSC-ECs) treated in vitro with menthol-flavored e-cig aerosol extract. The iPSC-ECs were generated from four healthy donors. There were 183 upregulated and 297 downregulated lncRNAs together with 132 upregulated and 413 downregulated mRNAs in the treated iPSC-ECs. Co-expression network analysis of the top five upregulated and downregulated lncRNAs and the associated 412 differentially expressed mRNAs revealed that the downregulated lncRNAs were associated with genes participating in fatty acid metabolism, cell cycle, cell division, and cell adhesion, while the upregulated lncRNAs were associated with genes involved in iron ion binding, protein binding, and proton-transporting ATPase activity. Small interfering RNA (siRNA) knock down of lncRNA LUCAT1, which was significantly upregulated in the treated cells, led to attenuation of the enhanced cell permeability and reactive oxygen species (ROS) production while partially restoring cell migration ability [45].
Kaur et al. [44] compared the expression profiles of lncRNA in plasma-derived exosomes obtained from e-cig users, cigarette smokers, waterpipe smokers, dual smokers (both cigarettes and waterpipe), and non-users. Six to eight subjects were studied per group. The GeneChipTM WT Pico kit was used for expression profiling. Differentially expressed lncRNAs were detectable in various groups as compared to non-users as follows: e-cig users (13 lncRNAs: 8 upregulated and 5 downregulated), cigarette smokers (7 lncRNAs: 2 upregulated and 5 downregulated), waterpipe smokers (18 lncRNAs: 9 upregulated and 9 downregulated), and dual smokers (9 lncRNAs: 4 upregulated and 5 downregulated). The differentially expressed lncRNAs in e-cig users vs. non-users were unique and did not overlap with those identified in other groups when compared to non-users. There were few overlapping lncRNAs that were commonly dysregulated across other comparison groups. Functional annotation of the differentially expressed lncRNAs by FunRich gene enrichment analysis showed significant enrichment for genes involved in steroid metabolism and steroid binding in e-cig users vs. non-users. The differentially expressed lncRNAs in other comparison groups were significantly enriched for genes involved in important biological processes, including cell differentiation and proliferation [44].
We performed RNA-seq analysis on total RNA isolated from oral epithelial cells of healthy adult “exclusive” vapers, “exclusive” cigarette smokers, and non-users (N = 42, 24, and 27, respectively) [13]. Our choice of oral epithelial cells for gene expression analysis was based on the following: (i) oral epithelium is the first site of “direct” exposure to known toxicants and carcinogens present in e-cig vapor and cigarette smoke [136,137,138,139,140]; (ii) oral epithelial cells are a major target for tumor development and other anomalies associated with tobacco use [141,142]; (iii) over 90% of all human cancers are of epithelial origin [143,144]; (iv) oral epithelial cells and lung epithelial cells show striking similarities in response to respiratory toxicants and carcinogens, as evidenced by the comparable patterns of genotoxic [145,146,147,148,149,150,151,152,153,154,155,156,157,158], epigenetic [159,160,161,162], and transcriptomic effects [163,164,165,166,167,168,169] in smokers’ oral cells and lung cells, respectively; and (v) oral epithelial cells are readily available for sampling by non-invasive techniques [136,166,170].
As shown in Figure 1A, there were large numbers of aberrantly expressed gene transcripts in both vapers and smokers as compared to non-users; however, smokers had nearly 50% more dysregulated genes than vapers (1726 vs. 1152). The aberrantly expressed genes in vapers and smokers can be divided into three categories: (I) vape-specific (853 transcripts); (II) smoke-specific (1427 transcripts); and (III) common to vape and smoke (299 transcripts) (Figure 1B). Gene ontology analysis revealed that cancer was the top disease associated with the dysregulated genes in both vapers (62%) and smokers (79%). The cancer-related dysregulated genes consisted of 361 genes specific to vapers, 1040 genes specific to smokers, and 182 genes common to vapers and smokers (total: 1583) (Figure 1C). The dysregulated genes in both vapers and smokers were also associated with other diseases and conditions, including inflammation. Of significance, some of the dysregulated genes in vapers and smokers are known to be frequently targeted in the early stages of diseases such as oral epithelial dysplasia, which can progress to malignancy [43].
Importantly, 47% of the aberrantly expressed genes in vapers were non-coding (vs. 21% in smokers). Of these, 23% were lncRNAs in vapers (vs. 8% in smokers) (Figure 2A). More specifically, there were 261 and 142 differentially expressed lncRNA genes in vapers and smokers, respectively (p < 0.0001) (Figure 2B). The differentially expressed lncRNAs in vapers consisted of 241 upregulated and 20 downregulated lncRNAs, whereas in smokers, the respective numbers were 128 and 14 (Figure 2B). Figure 2C shows sub-types of the differentially expressed lncRNAs detected in vapers and smokers. There was no statistically significant difference in number of differentially expressed lncRNA of any sub-types between vapers and smokers.
Molecular pathway and functional network analysis of the dysregulated genes identified the “Wnt/Ca+ pathway” as the most affected pathway in vapers, while the “integrin signaling pathway” was most impacted in smokers. The Wnt/Ca2+ signaling pathway, which is activated by the tumor suppressor WNT5A in the presence of a “frizzled” class receptor, is downregulated in several types of human cancer [171]. Of significance, the WNT5A gene and the frizzled receptor FDZ7 gene were both downregulated in vapers, likely inhibiting the downstream effectors of the cascade. The integrin signaling pathway is shown to control cell proliferation, survival, and migration. When dysregulated, the integrin signaling pathway can promote tumor invasion and metastasis [172]. The “Rho family GTPases signaling pathway” was the most common dysregulated pathway in vapers and smokers, although the number of affected targets was three times higher in smokers than vapers (27 vs. 9). The GTPase family of small GTP-binding proteins comprises a group of signaling molecules that are activated by growth factors, hormones, integrins, cytokines, and adhesion molecules [173]. They regulate reorganization of the actin cytoskeleton, transcriptional regulation, vesicle trafficking, morphogenesis, neutrophil activation, phagocytosis, mitogenesis, apoptosis, and tumorigenesis. Rho GTPases are also implicated in the DNA-damage response following genotoxin treatment [173]. More in-depth examination of the dysregulated genes and integrative analysis of the lncRNA and mRNA data are currently underway in our laboratory.

9. Concluding Remarks and Future Directions

A growing body of research shows the importance of lncRNAs in regulation of genes involved in the development of human diseases [29,30,31,32,33,34,35,36]. LncRNAs are emerging as promising biomarkers for assessing exposure to disease-causing agents [40,42,174] as well as elucidating the underlying mechanisms of disease development [33,37,38,175]. A limited but burgeoning number of studies has shown the potential of lncRNAs for assessing the biological consequences of vaping [13,44,45]. The existing data show that lncRNAs mediate dysregulation of key disease-related genes in cells and tissues of vapers [13,44] and in cell treated in vitro with e-cig aerosol extract [45]. The dysregulated lncRNAs and associated molecular pathways and gene networks in e-cig users have been shown to be partly similar to and partly distinct from those found in smokers [13,44]. The aberrantly expressed lncRNAs common to vapers and smokers can be attributed to shared exposure of both groups to chemicals present in e-cig vapers and tobacco smoke [13,55,176]. On the other hand, the unique dysregulated lncRNAs specific to vapers or smokers are likely due to distinct exposure of each group to chemicals present only in e-cig vapor or tobacco smoke [13,46,176]. Dual users of e-cigs and combustible tobacco products exhibited dysregulation of some of the same lncRNAs that are aberrantly expressed in exclusive vapers and smokers [45]. Nonetheless, dual users have also shown unique dysregulated lncRNAs, which may be ascribed to the interactive effects of combined use of e-cigs and combustible tobacco products [45,67].
Thus far, the (relatively) small size of the studied populations has precluded examination of the contribution of product characteristics, e.g., e-cig device type or features and e-liquid content (i.e., flavor type, nicotine concentration, and humectants), to the observed effects in vapers [13,44]. Follow-up studies with large sample size and improved statistical power should determine the role of product characteristics in the lncRNA-mediated dysregulation of disease-related genes observed in vapers. Future investigations should consider the dynamic changes as well as uniformities in epigenetic marks across different cell types [11,12,159,177,178] when comparing the expression profiles of lncRNA in various biospecimens from e-cig users. In addition, the effect of local vs. systemic exposure to chemicals present in e-cig vapor should be taken into account when analyzing different cells and tissues from e-cig users, e.g., oral or nasal epithelia vs. peripheral blood.
Because many transcriptomic changes occur in the early stages of disease—often prior to clinical manifestation of the disease [179,180,181]—it is all but certain that one should detect dysregulated disease-related coding and non-coding genes, e.g., lncRNAs, in apparently healthy vapers, smokers, and dual users, as shown by us [13,23,176] and others [44]. The target lncRNAs/mRNAs in healthy vapers and/or conventional tobacco product users are likely to be dysregulated to a lower extent than those in patients diagnosed with diseases. Substantial and long-term exposure of chronic vapers and smokers to toxicants and carcinogens present in e-cig vapor and tobacco smoke should lead to transcriptomic changes including differentially expressed lncRNAs similar to those found in the patient population, although patients are likely to have more pronounced changes. The dysregulated lncRNAs in vapers and/or smokers have been associated with diseases like cancer, respiratory diseases, cardiovascular disease, and/or immune diseases [13,44,45]. This is consistent with fact that these diseases are most commonly caused by or linked to tobacco product use [1,3,5,8,10,15,23,65,176,182,183,184].
Since an individual lncRNA can have multiple mRNA targets [83,84,85,86], most dysregulated lncRNAs in vapers and/or smokers may be associated with multiple diseases. Of note, a biological pathway is rarely, if ever, affected in one disease only. To minimize noise and facilitate data interpretation, future investigations should use statistical and bioinformatic approaches to prioritize selection of lncRNA–disease pairs in vapers and/or smokers with the highest association specificity and sensitivity. Lastly, while association studies of molecular changes and disease are widely used for biomarker discovery in humans [179,185,186,187,188,189], follow-up functional studies involving RNA interference (RNAi) and antisense oligonucleotides [190,191,192,193] should be conducted to verify whether the dysregulated lncRNAs found in vapers and/or smokers can be causally linked to disease development.

Author Contributions

H.B. performed experiments and collected data; S.T. performed experiments and collected data, analyzed data, and interpreted the results; A.B. conceived and designed the study, performed experiments and collected data, analyzed data, interpreted the results, and wrote the manuscript. All authors have read and agreed to the published version of the manuscript.

Funding

The work of the authors is supported by grants from the National Institute of Dental and Craniofacial Research, National Cancer Institute, and National Institute on Drug Abuse of the National Institutes of Health (R01DE031863, R21CA268197, and R21DA058342) and the University of California Tobacco-Related Disease Research Program (T31IR1839, T32IR5144, and T33IR6680). The sponsors of the study had no role in study design, data collection, data analysis, data interpretation, writing of the report, or in the decision to submit for publication. The bioinformatics software and computing resources used in the analysis were funded by the USC Office of Research and the Norris Medical Library.

Data Availability Statement

All data are contained within the article.

Conflicts of Interest

All the authors declare no conflicts of interest.

Abbreviations

ceRNAs, competing endogenous RNAs; e-cig, electronic cigarette; ENDS, electronic nicotine delivery systems; FDA, United States Food and Drug Administration; iPSC-ECs, induced pluripotent stem cell-derived endothelial cells; lincRNAs, long intergenic noncoding RNAs; lncRNAs, long non-coding RNAs; miRNA, microRNAs; PG, propylene glycol; ROS, reactive oxygen species; siRNA, small interfering RNA; TF, transcription factor; VG, vegetable glycerin; VOC, volatile organic compounds

References

  1. Gordon, T.; Karey, E.; Rebuli, M.E.; Escobar, Y.H.; Jaspers, I.; Chen, L.C. E-Cigarette Toxicology. Annu. Rev. Pharmacol. Toxicol. 2022, 62, 301–322. [Google Scholar] [CrossRef] [PubMed]
  2. Hartmann-Boyce, J.; McRobbie, H.; Lindson, N.; Bullen, C.; Begh, R.; Theodoulou, A.; Notley, C.; Rigotti, N.A.; Turner, T.; Butler, A.R.; et al. Electronic cigarettes for smoking cessation. Cochrane Database Syst. Rev. 2021, 4, CD010216. [Google Scholar] [PubMed]
  3. Gotts, J.E.; Jordt, S.E.; McConnell, R.; Tarran, R. What are the respiratory effects of e-cigarettes? BMJ 2019, 366, l5275. [Google Scholar] [CrossRef] [PubMed]
  4. Besaratinia, A.; Tommasi, S. Vaping epidemic: Challenges and opportunities. Cancer Causes Control CCC 2020, 31, 663–667. [Google Scholar] [CrossRef] [PubMed]
  5. Bozier, J.; Chivers, E.K.; Chapman, D.G.; Larcombe, A.N.; Bastian, N.A.; Masso-Silva, J.A.; Byun, M.K.; McDonald, C.F.; Crotty Alexander, L.E.; Ween, M.P. The Evolving Landscape of e-Cigarettes: A Systematic Review of Recent Evidence. Chest 2020, 157, 1362–1390. [Google Scholar] [CrossRef] [PubMed]
  6. Balfour, D.J.K.; Benowitz, N.L.; Colby, S.M.; Hatsukami, D.K.; Lando, H.A.; Leischow, S.J.; Lerman, C.; Mermelstein, R.J.; Niaura, R.; Perkins, K.A.; et al. Balancing Consideration of the Risks and Benefits of E-Cigarettes. Am. J. Public Health 2021, 111, 1661–1672. [Google Scholar] [CrossRef] [PubMed]
  7. Glantz, S.A.; Nguyen, N.; Oliveira da Silva, A.L. Population-Based Disease Odds for E-Cigarettes and Dual Use versus Cigarettes. NEJM Evid. 2024, 3, EVIDoa2300229. [Google Scholar] [CrossRef] [PubMed]
  8. Wills, T.A.; Soneji, S.S.; Choi, K.; Jaspers, I.; Tam, E.K. E-cigarette use and respiratory disorders: An integrative review of converging evidence from epidemiological and laboratory studies. Eur. Respir. J. 2021, 57, 1901815. [Google Scholar] [CrossRef] [PubMed]
  9. Wang, R.J.; Bhadriraju, S.; Glantz, S.A. E-Cigarette Use and Adult Cigarette Smoking Cessation: A Meta-Analysis. Am. J. Public Health 2021, 111, 230–246. [Google Scholar] [CrossRef]
  10. Park, J.A.; Crotty Alexander, L.E.; Christiani, D.C. Vaping and Lung Inflammation and Injury. Annu. Rev. Physiol. 2022, 84, 611–629. [Google Scholar] [CrossRef]
  11. Baylin, S.B.; Jones, P.A. Epigenetic Determinants of Cancer. Cold Spring Harb. Perspect. Biol. 2016, 8, a019505. [Google Scholar] [CrossRef] [PubMed]
  12. Feinberg, A.P.; Koldobskiy, M.A.; Gondor, A. Epigenetic modulators, modifiers and mediators in cancer aetiology and progression. Nat. Rev. Genet. 2016, 17, 284–299. [Google Scholar] [CrossRef] [PubMed]
  13. Tommasi, S.; Caliri, A.W.; Caceres, A.; Moreno, D.E.; Li, M.; Chen, Y.; Siegmund, K.D.; Besaratinia, A. Deregulation of Biologically Significant Genes and Associated Molecular Pathways in the Oral Epithelium of Electronic Cigarette Users. Int. J. Mol. Sci. 2019, 20, 738. [Google Scholar] [CrossRef] [PubMed]
  14. Andersen, A.; Reimer, R.; Dawes, K.; Becker, A.; Hutchens, N.; Miller, S.; Dogan, M.; Hundley, B.; Mills, J.A.; Long, J.D.; et al. DNA methylation differentiates smoking from vaping and non-combustible tobacco use. Epigenetics 2021, 1–13. [Google Scholar] [CrossRef]
  15. Caliri, A.W.; Tommasi, S.; Besaratinia, A. Relationships among smoking, oxidative stress, inflammation, macromolecular damage, and cancer. Mutat. Res. 2021, 787, 108365. [Google Scholar] [CrossRef]
  16. Yan, R.; Chen, X.L.; Xu, Y.M.; Lau, A.T.Y. Epimutational effects of electronic cigarettes. Environ. Sci. Pollut. Res. Int. 2021, 28, 17044–17067. [Google Scholar] [CrossRef]
  17. Besaratinia, A.; Caceres, A.; Tommasi, S. DNA Hydroxymethylation in Smoking-Associated Cancers. Int. J. Mol. Sci. 2022, 23, 2657. [Google Scholar] [CrossRef] [PubMed]
  18. Goodman, S.; Chappell, G.; Guyton, K.Z.; Pogribny, I.P.; Rusyn, I. Epigenetic alterations induced by genotoxic occupational and environmental human chemical carcinogens: An update of a systematic literature review. Mutat. Res. Rev. Mutat. Res. 2022, 789, 108408. [Google Scholar] [CrossRef] [PubMed]
  19. Tommasi, S.; Kim, S.I.; Zhong, X.; Wu, X.; Pfeifer, G.P.; Besaratinia, A. Investigating the epigenetic effects of a prototype smoke-derived carcinogen in human cells. PLoS ONE 2010, 5, e10594. [Google Scholar] [CrossRef]
  20. Mori, K.M.; McElroy, J.P.; Weng, D.Y.; Chung, S.; Fadda, P.; Reisinger, S.A.; Ying, K.L.; Brasky, T.M.; Wewers, M.D.; Freudenheim, J.L.; et al. Lung mitochondrial DNA copy number, inflammatory biomarkers, gene transcription and gene methylation in vapers and smokers. EBioMedicine 2022, 85, 104301. [Google Scholar] [CrossRef]
  21. Song, M.A.; Mori, K.M.; McElroy, J.P.; Freudenheim, J.L.; Weng, D.Y.; Reisinger, S.A.; Brasky, T.M.; Wewers, M.D.; Shields, P.G. Accelerated epigenetic age, inflammation, and gene expression in lung: Comparisons of smokers and vapers with non-smokers. Clin. Epigenetics 2023, 15, 160. [Google Scholar] [CrossRef]
  22. Herzog, C.; Jones, A.; Evans, I.; Raut, J.R.; Zikan, M.; Cibula, D.; Wong, A.; Brenner, H.; Richmond, R.C.; Widschwendter, M. Cigarette Smoking and E-cigarette Use Induce Shared DNA Methylation Changes Linked to Carcinogenesis. Cancer Res. 2024, 84, 1898–1914. [Google Scholar] [CrossRef] [PubMed]
  23. Caliri, A.W.; 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, 816–829. [Google Scholar] [CrossRef] [PubMed]
  24. Hahm, J.Y.; Park, J.; Jang, E.S.; Chi, S.W. 8-Oxoguanine: From oxidative damage to epigenetic and epitranscriptional modification. Exp. Mol. Med. 2022, 54, 1626–1642. [Google Scholar] [CrossRef] [PubMed]
  25. Quinn, J.J.; Chang, H.Y. Unique features of long non-coding RNA biogenesis and function. Nat. Rev. Genet. 2016, 17, 47–62. [Google Scholar] [CrossRef] [PubMed]
  26. Yao, R.W.; Wang, Y.; Chen, L.L. Cellular functions of long noncoding RNAs. Nat. Cell Biol. 2019, 21, 542–551. [Google Scholar] [CrossRef]
  27. Rinn, J.L.; Chang, H.Y. Long Noncoding RNAs: Molecular Modalities to Organismal Functions. Annu. Rev. Biochem. 2020, 89, 283–308. [Google Scholar] [CrossRef] [PubMed]
  28. Statello, L.; Guo, C.J.; Chen, L.L.; Huarte, M. Gene regulation by long non-coding RNAs and its biological functions. Nat. Rev. Mol. Cell Biol. 2021, 22, 96–118. [Google Scholar] [CrossRef] [PubMed]
  29. Ishii, N.; Ozaki, K.; Sato, H.; Mizuno, H.; Susumu, S.; Takahashi, A.; Miyamoto, Y.; Ikegawa, S.; Kamatani, N.; Hori, M.; et al. Identification of a novel non-coding RNA, MIAT, that confers risk of myocardial infarction. J. Hum. Genet. 2006, 51, 1087–1099. [Google Scholar] [CrossRef]
  30. Fitzgerald, K.A.; Caffrey, D.R. Long noncoding RNAs in innate and adaptive immunity. Curr. Opin. Immunol. 2014, 26, 140–146. [Google Scholar] [CrossRef]
  31. Elling, R.; Chan, J.; Fitzgerald, K.A. Emerging role of long noncoding RNAs as regulators of innate immune cell development and inflammatory gene expression. Eur. J. Immunol. 2016, 46, 504–512. [Google Scholar] [CrossRef]
  32. Ferreira, H.J.; Esteller, M. Non-coding RNAs, epigenetics, and cancer: Tying it all together. Cancer Metastasis Rev. 2018, 37, 55–73. [Google Scholar] [CrossRef] [PubMed]
  33. Devadoss, D.; Long, C.; Langley, R.J.; Manevski, M.; Nair, M.; Campos, M.A.; Borchert, G.; Rahman, I.; Chand, H.S. Long Noncoding Transcriptome in Chronic Obstructive Pulmonary Disease. Am. J. Respir. Cell Mol. Biol. 2019, 61, 678–688. [Google Scholar] [CrossRef] [PubMed]
  34. Tsagakis, I.; Douka, K.; Birds, I.; Aspden, J.L. Long non-coding RNAs in development and disease: Conservation to mechanisms. J. Pathol. 2020, 250, 480–495. [Google Scholar] [CrossRef] [PubMed]
  35. Moukette, B.; Barupala, N.P.; Aonuma, T.; Sepulveda, M.; Kawaguchi, S.; Kim, I.M. Interactions between noncoding RNAs as epigenetic regulatory mechanisms in cardiovascular diseases. Methods Cell Biol. 2021, 166, 309–348. [Google Scholar]
  36. Wang, P.S.; Wang, Z.; Yang, C. Dysregulations of long non-coding RNAs-The emerging “lnc” in environmental carcinogenesis. Semin. Cancer Biol. 2021, 76, 163–172. [Google Scholar] [CrossRef]
  37. Karlsson, O.; Baccarelli, A.A. Environmental Health and Long Non-coding RNAs. Curr. Environ. Health Rep. 2016, 3, 178–187. [Google Scholar] [CrossRef]
  38. Goodall, G.J.; Wickramasinghe, V.O. RNA in cancer. Nat. Rev. Cancer 2021, 21, 22–36. [Google Scholar] [CrossRef] [PubMed]
  39. Thai, P.; Statt, S.; Chen, C.H.; Liang, E.; Campbell, C.; Wu, R. Characterization of a novel long noncoding RNA, SCAL1, induced by cigarette smoke and elevated in lung cancer cell lines. Am. J. Respir. Cell Mol. Biol. 2013, 49, 204–211. [Google Scholar] [CrossRef]
  40. Gu, W.; Yuan, Y.; Wang, L.; Yang, H.; Li, S.; Tang, Z.; Li, Q. Long non-coding RNA TUG1 promotes airway remodelling by suppressing the miR-145-5p/DUSP6 axis in cigarette smoke-induced COPD. J. Cell Mol. Med. 2019, 23, 7200–7209. [Google Scholar] [CrossRef]
  41. Xie, J.; Wu, Y.; Tao, Q.; Liu, H.; Wang, J.; Zhang, C.; Zhou, Y.; Wei, C.; Chang, Y.; Jin, Y.; et al. The role of lncRNA in the pathogenesis of chronic obstructive pulmonary disease. Heliyon 2023, 9, e22460. [Google Scholar] [CrossRef] [PubMed]
  42. Zheng, R.; Gao, F.; Mao, Z.; Xiao, Y.; Yuan, L.; Huang, Z.; Lv, Q.; Qin, C.; Du, M.; Zhang, Z.; et al. LncRNA BCCE4 Genetically Enhances the PD-L1/PD-1 Interaction in Smoking-Related Bladder Cancer by Modulating miR-328-3p-USP18 Signaling. Adv. Sci. (Weinh) 2023, 10, e2303473. [Google Scholar] [CrossRef] [PubMed]
  43. Besaratinia, A.; Tommasi, S. Vaping: A growing global health concern. EClinicalMedicine 2019, 17, 100208. [Google Scholar] [CrossRef] [PubMed]
  44. Kaur, G.; Singh, K.; Maremanda, K.P.; Li, D.; Chand, H.S.; 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, e0243065. [Google Scholar] [CrossRef] [PubMed]
  45. Le, H.H.T.; Liu, C.W.; Denaro, P., 3rd; Jousma, J.; Shao, N.Y.; Rahman, I.; Lee, W.H. Genome-wide differential expression profiling of lncRNAs and mRNAs in human induced pluripotent stem cell-derived endothelial cells exposed to e-cigarette extract. Stem Cell Res. Ther. 2021, 12, 593. [Google Scholar] [CrossRef] [PubMed]
  46. Besaratinia, A. COVID-19: A pandemic converged with global tobacco epidemic and widespread vaping-state of the evidence. Carcinogenesis 2021, 42, 1009–1022. [Google Scholar] [CrossRef]
  47. Soule, E.; Bansal-Travers, M.; Grana, R.; McIntosh, S.; Price, S.; Unger, J.B.; Walton, K. Electronic cigarette use intensity measurement challenges and regulatory implications. Tob. Control 2021, 32, 124–129. [Google Scholar] [CrossRef] [PubMed]
  48. Snoderly, H.T.; Nurkiewicz, T.R.; Bowdridge, E.C.; Bennewitz, M.F. E-Cigarette Use: Device Market, Study Design, and Emerging Evidence of Biological Consequences. Int. J. Mol. Sci. 2021, 22, 12452. [Google Scholar] [CrossRef] [PubMed]
  49. Havermans, A.; Krüsemann, E.J.Z.; Pennings, J.; de Graaf, K.; Boesveldt, S.; Talhout, R. Nearly 20 000 e-liquids and 250 unique flavour descriptions: An overview of the Dutch market based on information from manufacturers. Tob. Control 2021, 30, 57–62. [Google Scholar] [CrossRef]
  50. Schneller, L.M.; Bansal-Travers, M.; Goniewicz, M.L.; McIntosh, S.; Ossip, D.; O’Connor, R.J. Use of flavored electronic cigarette refill liquids among adults and youth in the US-Results from Wave 2 of the Population Assessment of Tobacco and Health Study (2014–2015). PLoS ONE 2018, 13, e0202744. [Google Scholar] [CrossRef]
  51. Wei, B.; O’Connor, R.J.; Goniewicz, M.L.; Hyland, A. Emerging Chemicals of Health Concern in Electronic Nicotine Delivery Systems. Chem. Res. Toxicol. 2020, 33, 2637–2646. [Google Scholar] [CrossRef] [PubMed]
  52. Bitzer, Z.T.; Goel, R.; Reilly, S.M.; Elias, R.J.; Silakov, A.; Foulds, J.; Muscat, J.; Richie, J.P., Jr. Effect of flavoring chemicals on free radical formation in electronic cigarette aerosols. Free Radic. Biol. Med. 2018, 120, 72–79. [Google Scholar] [CrossRef] [PubMed]
  53. Erythropel, H.C.; Jabba, S.V.; DeWinter, T.M.; Mendizabal, M.; Anastas, P.T.; Jordt, S.E.; Zimmerman, J.B. Formation of flavorant-propylene Glycol Adducts With Novel Toxicological Properties in Chemically Unstable E-Cigarette Liquids. Nicotine Tob. Res. Off. J. Soc. Res. Nicotine Tob. 2019, 21, 1248–1258. [Google Scholar] [CrossRef] [PubMed]
  54. Son, Y.; Mishin, V.; Laskin, J.D.; Mainelis, G.; Wackowski, O.A.; Delnevo, C.; Schwander, S.; Khlystov, A.; Samburova, V.; Meng, Q. Hydroxyl Radicals in E-Cigarette Vapor and E-Vapor Oxidative Potentials under Different Vaping Patterns. Chem. Res. Toxicol. 2019, 32, 1087–1095. [Google Scholar] [CrossRef] [PubMed]
  55. Besaratinia, A.; Pfeifer, G.P. Enhancement of the mutagenicity of benzo(a)pyrene diol epoxide by a nonmutagenic dose of ultraviolet A radiation. Cancer Res. 2003, 63, 8708–8716. [Google Scholar] [PubMed]
  56. Son, Y.; Bhattarai, C.; Samburova, V.; Khlystov, A. Carbonyls and Carbon Monoxide Emissions from Electronic Cigarettes Affected by Device Type and Use Patterns. Int. J. Environ. Res. Public Health 2020, 17, 2767. [Google Scholar] [CrossRef] [PubMed]
  57. Tommasi, S.; Bates, S.E.; Behar, R.Z.; Talbot, P.; Besaratinia, A. Limited mutagenicity of electronic cigarettes in mouse or human cells in vitro. Lung Cancer 2017, 112, 41–46. [Google Scholar] [CrossRef] [PubMed]
  58. Correia-Alvarez, E.; Keating, J.E.; Glish, G.; Tarran, R.; Sassano, M.F. Reactive Oxygen Species, Mitochondrial Membrane Potential, and Cellular Membrane Potential Are Predictors of E-Liquid Induced Cellular Toxicity. Nicotine Tob. Res. 2020, 22 (Suppl. S1), S4–S13. [Google Scholar] [CrossRef]
  59. Barhdadi, S.; Mertens, B.; Van Bossuyt, M.; Van De Maele, J.; Anthonissen, R.; Canfyn, M.; Courselle, P.; Rogiers, V.; Deconinck, E.; Vanhaecke, T. Identification of flavouring substances of genotoxic concern present in e-cigarette refills. Food Chem. Toxicol. An. Int. J. Publ. Br. Ind. Biol. Res. Assoc. 2021, 147, 111864. [Google Scholar] [CrossRef]
  60. McDonough, S.R.; Rahman, I.; Sundar, I.K. Recent updates on biomarkers of exposure and systemic toxicity in e-cigarette users and EVALI. Am. J. Physiol. Lung Cell. Mol. Physiol. 2021, 320, L661–l679. [Google Scholar] [CrossRef]
  61. Besaratinia, A.; Caliri, A.W.; Tommasi, S. The interplay of DNA damage and repair, gene expression, and mutagenesis in mammalian cells during oxidative stress. Carcinogenesis 2024. [Google Scholar] [CrossRef] [PubMed]
  62. Tehrani, M.W.; Newmeyer, M.N.; Rule, A.M.; Prasse, C. Characterizing the Chemical Landscape in Commercial E-Cigarette Liquids and Aerosols by Liquid Chromatography-High-Resolution Mass Spectrometry. Chem. Res. Toxicol. 2021, 34, 2216–2226. [Google Scholar] [CrossRef] [PubMed]
  63. Khlystov, A.; Samburova, V. Flavoring Compounds Dominate Toxic Aldehyde Production during E-Cigarette Vaping. Environ. Sci. Technol. 2016, 50, 13080–13085. [Google Scholar] [CrossRef] [PubMed]
  64. Besaratinia, A.; Tommasi, S. The consequential impact of JUUL on youth vaping and the landscape of tobacco products: The state of play in the COVID-19 era. Prev. Med. Rep. 2021, 22, 101374. [Google Scholar] [CrossRef] [PubMed]
  65. Shields, P.G.; Berman, M.; Brasky, T.M.; Freudenheim, J.L.; Mathe, E.; McElroy, J.P.; Song, M.A.; Wewers, M.D. A Review of Pulmonary Toxicity of Electronic Cigarettes in the Context of Smoking: A Focus on Inflammation. Cancer Epidemiol. Biomark. Prev. 2017, 26, 1175–1191. [Google Scholar] [CrossRef] [PubMed]
  66. Dockrell, M.; Newton, J.N. Tobacco Control Leaders Call for a Balanced Assessment of the Risks and Benefits of Nicotine Vaping. Am. J. Public Health 2021, 111, 1570–1571. [Google Scholar] [CrossRef] [PubMed]
  67. Besaratinia, A. From Tobacco Cigarettes to Electronic Cigarettes: The Two Sides of a Nicotine Coin. Front. Oral. Health 2021, 2, 790634. [Google Scholar] [CrossRef]
  68. Samet, J.M.; Barrington-Trimis, J. E-Cigarettes and Harm Reduction: An Artificial Controversy Instead of Evidence and a Well-Framed Decision Context. Am. J. Public Health 2021, 111, 1572–1574. [Google Scholar] [CrossRef]
  69. Mendelsohn, C.P.; Hall, W.; Borland, R.; Wodak, A.; Beaglehole, R.; Benowitz, N.L.; Britton, J.; Bullen, C.; Etter, J.F.; McNeill, A.; et al. A critique of the Australian National Health and Medical Research Council CEO statement on electronic cigarettes. Addiction 2023, 118, 1184–1192. [Google Scholar] [CrossRef]
  70. Chan, G.C.K.; Stjepanović, D.; Lim, C.; Sun, T.; Shanmuga Anandan, A.; Connor, J.P.; Gartner, C.; Hall, W.D.; Leung, J. A systematic review of randomized controlled trials and network meta-analysis of e-cigarettes for smoking cessation. Addict. Behav. 2021, 119, 106912. [Google Scholar] [CrossRef]
  71. Grabovac, I.; Oberndorfer, M.; Fischer, J.; Wiesinger, W.; Haider, S.; Dorner, T.E. Effectiveness of Electronic Cigarettes in Smoking Cessation: A Systematic Review and Meta-analysis. Nicotine Tob. Res. 2021, 23, 625–634. [Google Scholar] [CrossRef] [PubMed]
  72. Hanewinkel, R.; Niederberger, K.; Pedersen, A.; Unger, J.B.; Galimov, A. E-cigarettes and nicotine abstinence: A meta-analysis of randomised controlled trials. Eur. Respir. Rev. 2022, 31, 210215. [Google Scholar] [CrossRef]
  73. Levett, J.Y.; Filion, K.B.; Reynier, P.; Prell, C.; Eisenberg, M.J. Efficacy and Safety of E-Cigarette Use for Smoking Cessation: A Systematic Review and Meta-Analysis of Randomized Controlled Trials. Am. J. Med. 2023, 136, 804–813.e4. [Google Scholar] [CrossRef]
  74. Osibogun, O.; Bursac, Z.; Maziak, W. Longitudinal transition outcomes among adult dual users of e-cigarettes and cigarettes with the intention to quit in the United States: PATH Study (2013–2018). Prev. Med. Rep. 2022, 26, 101750. [Google Scholar] [CrossRef]
  75. Chen, R.; Pierce, J.P.; Leas, E.C.; Benmarhnia, T.; Strong, D.R.; White, M.M.; Stone, M.; Trinidad, D.R.; McMenamin, S.B.; Messer, K. Effectiveness of e-cigarettes as aids for smoking cessation: Evidence from the PATH Study cohort, 2017–2019. Tob. Control 2023, 32, e145–e152. [Google Scholar] [CrossRef] [PubMed]
  76. Banks, E.; Yazidjoglou, A.; Brown, S.; Nguyen, M.; Martin, M.; Beckwith, K.; Daluwatta, A.; Campbell, S.; Joshy, G. Electronic cigarettes and health outcomes: Umbrella and systematic review of the global evidence. Med. J. Aust. 2023, 218, 267–275. [Google Scholar] [CrossRef] [PubMed]
  77. Hedman, L.; Galanti, M.R.; Ryk, L.; Gilljam, H.; Adermark, L. Electronic cigarette use and smoking cessation in cohort studies and randomized trials: A systematic review and meta-analysis. Tob. Prev. Cessat. 2021, 7, 62. [Google Scholar] [CrossRef]
  78. Glantz, S.A. E-Cigarettes as Consumer Products. Am. J. Public Health 2022, 112, e4–e5. [Google Scholar] [CrossRef]
  79. McNeill, A.; Brose, L.S.; Calder, R.; Bauld, L.; Robson, D. Evidence Review of e-Cigarettes and Heated Tobacco Products 2018; Public Health England: London, UK, 2018. [Google Scholar]
  80. Khouja, J.N.; Suddell, S.F.; Peters, S.E.; Taylor, A.E.; Munafò, M.R. Is e-cigarette use in non-smoking young adults associated with later smoking? A systematic review and meta-analysis. Tob. Control 2020, 30, 8–15. [Google Scholar] [CrossRef]
  81. Gades, M.S.; Alcheva, A.; Riegelman, A.L.; Hatsukami, D.K. The Role of Nicotine and Flavor in the Abuse Potential and Appeal of Electronic Cigarettes for Adult Current and Former Cigarette and Electronic Cigarette Users: A Systematic Review. Nicotine Tob. Res. 2022, 24, 1332–1343. [Google Scholar] [CrossRef]
  82. Tommasi, S.; Zheng, A.; Yoon, J.I.; Besaratinia, A. Epigenetic targeting of the Nanog pathway and signaling networks during chemical carcinogenesis. Carcinogenesis 2014, 35, 1726–1736. [Google Scholar] [CrossRef] [PubMed]
  83. Kapranov, P.; Cheng, J.; Dike, S.; Nix, D.A.; Duttagupta, R.; Willingham, A.T.; Stadler, P.F.; Hertel, J.; Hackermüller, J.; Hofacker, I.L.; et al. RNA maps reveal new RNA classes and a possible function for pervasive transcription. Science 2007, 316, 1484–1488. [Google Scholar] [CrossRef] [PubMed]
  84. Djebali, S.; Davis, C.A.; Merkel, A.; Dobin, A.; Lassmann, T.; Mortazavi, A.; Tanzer, A.; Lagarde, J.; Lin, W.; Schlesinger, F.; et al. Landscape of transcription in human cells. Nature 2012, 489, 101–108. [Google Scholar] [CrossRef] [PubMed]
  85. Chen, L.L. Linking Long Noncoding RNA Processing and Function to RNA Biology. Trends Biochem. Sci. 2016, 41, 733–734. [Google Scholar] [CrossRef] [PubMed]
  86. Lee, H.; Zhang, Z.; Krause, H.M. Long Noncoding RNAs and Repetitive Elements: Junk or Intimate Evolutionary Partners? Trends Genet. TIG 2019, 35, 892–902. [Google Scholar] [CrossRef] [PubMed]
  87. Volders, P.J.; Anckaert, J.; Verheggen, K.; Nuytens, J.; Martens, L.; Mestdagh, P.; Vandesompele, J. LNCipedia 5: Towards a reference set of human long non-coding RNAs. Nucleic Acids Res. 2019, 47, D135–D139. [Google Scholar] [CrossRef] [PubMed]
  88. Yoon, J.H.; Abdelmohsen, K.; Gorospe, M. Posttranscriptional gene regulation by long noncoding RNA. J. Mol. Biol. 2013, 425, 3723–3730. [Google Scholar] [CrossRef] [PubMed]
  89. Marchese, F.P.; Raimondi, I.; Huarte, M. The multidimensional mechanisms of long noncoding RNA function. Genome Biol. 2017, 18, 206. [Google Scholar] [CrossRef] [PubMed]
  90. Bunch, H. Gene regulation of mammalian long non-coding RNA. Mol. Genet. Genom. 2018, 293, 1–15. [Google Scholar] [CrossRef]
  91. Ma, L.; Bajic, V.B.; Zhang, Z. On the classification of long non-coding RNAs. RNA Biol. 2013, 10, 925–933. [Google Scholar] [CrossRef]
  92. Fernandes, J.C.R.; Acuña, S.M.; Aoki, J.I.; Floeter-Winter, L.M.; Muxel, S.M. Long Non-Coding RNAs in the Regulation of Gene Expression: Physiology and Disease. Noncoding RNA 2019, 5, 17. [Google Scholar] [CrossRef] [PubMed]
  93. Kapranov, P.; Willingham, A.T.; Gingeras, T.R. Genome-wide transcription and the implications for genomic organization. Nat. Rev. Genet. 2007, 8, 413–423. [Google Scholar] [CrossRef] [PubMed]
  94. Birney, E.; Stamatoyannopoulos, J.A.; Dutta, A.; Guigó, R.; Gingeras, T.R.; Margulies, E.H.; Weng, Z.; Snyder, M.; Dermitzakis, E.T.; Thurman, R.E.; et al. Identification and analysis of functional elements in 1% of the human genome by the ENCODE pilot project. Nature 2007, 447, 799–816. [Google Scholar] [PubMed]
  95. Kapusta, A.; Feschotte, C. Volatile evolution of long noncoding RNA repertoires: Mechanisms and biological implications. Trends Genet. TIG 2014, 30, 439–452. [Google Scholar] [CrossRef] [PubMed]
  96. Struhl, K. Transcriptional noise and the fidelity of initiation by RNA polymerase II. Nat. Struct. Mol. Biol. 2007, 14, 103–105. [Google Scholar] [CrossRef] [PubMed]
  97. Chen, J.; Shishkin, A.A.; Zhu, X.; Kadri, S.; Maza, I.; Guttman, M.; Hanna, J.H.; Regev, A.; Garber, M. Evolutionary analysis across mammals reveals distinct classes of long non-coding RNAs. Genome Biol. 2016, 17, 19. [Google Scholar] [CrossRef] [PubMed]
  98. Umlauf, D.; Goto, Y.; Cao, R.; Cerqueira, F.; Wagschal, A.; Zhang, Y.; Feil, R. Imprinting along the Kcnq1 domain on mouse chromosome 7 involves repressive histone methylation and recruitment of Polycomb group complexes. Nat. Genet. 2004, 36, 1296–1300. [Google Scholar] [CrossRef]
  99. Espinoza, C.A.; Goodrich, J.A.; Kugel, J.F. Characterization of the structure, function, and mechanism of B2 RNA, an ncRNA repressor of RNA polymerase II transcription. Rna 2007, 13, 583–596. [Google Scholar] [CrossRef]
  100. Katayama, S.; Tomaru, Y.; Kasukawa, T.; Waki, K.; Nakanishi, M.; Nakamura, M.; Nishida, H.; Yap, C.C.; Suzuki, M.; Kawai, J.; et al. Antisense transcription in the mammalian transcriptome. Science 2005, 309, 1564–1566. [Google Scholar] [CrossRef]
  101. Sleutels, F.; Zwart, R.; Barlow, D.P. The non-coding Air RNA is required for silencing autosomal imprinted genes. Nature 2002, 415, 810–813. [Google Scholar] [CrossRef]
  102. Mattick, J.S.; Amaral, P.P.; Carninci, P.; Carpenter, S.; Chang, H.Y.; Chen, L.L.; Chen, R.; Dean, C.; Dinger, M.E.; Fitzgerald, K.A.; et al. Long non-coding RNAs: Definitions, functions, challenges and recommendations. Nat. Rev. Mol. Cell Biol. 2023, 24, 430–447. [Google Scholar] [CrossRef] [PubMed]
  103. Wang, K.C.; Chang, H.Y. Molecular mechanisms of long noncoding RNAs. Mol. Cell 2011, 43, 904–914. [Google Scholar] [CrossRef] [PubMed]
  104. Guttman, M.; Rinn, J.L. Modular regulatory principles of large non-coding RNAs. Nature 2012, 482, 339–346. [Google Scholar] [CrossRef]
  105. Ulitsky, I.; Bartel, D.P. lincRNAs: Genomics, evolution, and mechanisms. Cell 2013, 154, 26–46. [Google Scholar] [CrossRef] [PubMed]
  106. Zampetaki, A.; Albrecht, A.; Steinhofel, K. Long Non-coding RNA Structure and Function: Is There a Link? Front. Physiol. 2018, 9, 1201. [Google Scholar] [CrossRef] [PubMed]
  107. Mattick, J.S. The State of Long Non-Coding RNA Biology. Noncoding RNA 2018, 4, 17. [Google Scholar] [CrossRef] [PubMed]
  108. Wu, M.; Yang, L.Z.; Chen, L.L. Long noncoding RNA and protein abundance in lncRNPs. Rna 2021, 27, 1427–1440. [Google Scholar] [CrossRef] [PubMed]
  109. Kim, T.K.; Hemberg, M.; Gray, J.M. Enhancer RNAs: A class of long noncoding RNAs synthesized at enhancers. Cold Spring Harb. Perspect. Biol. 2015, 7, a018622. [Google Scholar] [CrossRef] [PubMed]
  110. Tay, Y.; Rinn, J.; Pandolfi, P.P. The multilayered complexity of ceRNA crosstalk and competition. Nature 2014, 505, 344–352. [Google Scholar] [CrossRef]
  111. Chujo, T.; Yamazaki, T.; Hirose, T. Architectural RNAs (arcRNAs): A class of long noncoding RNAs that function as the scaffold of nuclear bodies. Biochim. Biophys. Acta 2016, 1859, 139–146. [Google Scholar] [CrossRef]
  112. Michelini, F.; Pitchiaya, S.; Vitelli, V.; Sharma, S.; Gioia, U.; Pessina, F.; Cabrini, M.; Wang, Y.; Capozzo, I.; Iannelli, F.; et al. Damage-induced lncRNAs control the DNA damage response through interaction with DDRNAs at individual double-strand breaks. Nat. Cell Biol. 2017, 19, 1400–1411. [Google Scholar] [CrossRef] [PubMed]
  113. Giannakakis, A.; Zhang, J.; Jenjaroenpun, P.; Nama, S.; Zainolabidin, N.; Aau, M.Y.; Yarmishyn, A.A.; Vaz, C.; Ivshina, A.V.; Grinchuk, O.V.; et al. Contrasting expression patterns of coding and noncoding parts of the human genome upon oxidative stress. Sci. Rep. 2015, 5, 9737. [Google Scholar] [CrossRef] [PubMed]
  114. Yu, B.; Wang, S. Angio-LncRs: LncRNAs that regulate angiogenesis and vascular disease. Theranostics 2018, 8, 3654–3675. [Google Scholar] [CrossRef] [PubMed]
  115. Oo, J.A.; Brandes, R.P.; Leisegang, M.S. Long non-coding RNAs: Novel regulators of cellular physiology and function. Pflug. Arch. 2022, 474, 191–204. [Google Scholar] [CrossRef] [PubMed]
  116. Ali, T.; Grote, P. Beyond the RNA-dependent function of LncRNA genes. Elife 2020, 9, e60583. [Google Scholar] [CrossRef] [PubMed]
  117. Wan, Y.; Kertesz, M.; Spitale, R.C.; Segal, E.; Chang, H.Y. Understanding the transcriptome through RNA structure. Nat. Rev. Genet. 2011, 12, 641–655. [Google Scholar] [CrossRef] [PubMed]
  118. Huarte, M.; Guttman, M.; Feldser, D.; Garber, M.; Koziol, M.J.; Kenzelmann-Broz, D.; Khalil, A.M.; Zuk, O.; Amit, I.; Rabani, M.; et al. A large intergenic noncoding RNA induced by p53 mediates global gene repression in the p53 response. Cell 2010, 142, 409–419. [Google Scholar] [CrossRef] [PubMed]
  119. Rothschild, G.; Zhang, W.; Lim, J.; Giri, P.K.; Laffleur, B.; Chen, Y.; Fang, M.; Chen, Y.; Nair, L.; Liu, Z.P.; et al. Noncoding RNA transcription alters chromosomal topology to promote isotype-specific class switch recombination. Sci. Immunol. 2020, 5, eaay5864. [Google Scholar] [CrossRef]
  120. Fanucchi, S.; Fok, E.T.; Dalla, E.; Shibayama, Y.; Börner, K.; Chang, E.Y.; Stoychev, S.; Imakaev, M.; Grimm, D.; Wang, K.C.; et al. Immune genes are primed for robust transcription by proximal long noncoding RNAs located in nuclear compartments. Nat. Genet. 2019, 51, 138–150. [Google Scholar] [CrossRef]
  121. Vollmers, A.C.; Covarrubias, S.; Kuang, D.; Shulkin, A.; Iwuagwu, J.; Katzman, S.; Song, R.; Viswanathan, K.; Vollmers, C.; Wakeland, E.; et al. A conserved long noncoding RNA, GAPLINC, modulates the immune response during endotoxic shock. Proc. Natl. Acad. Sci. USA 2021, 118, e2016648118. [Google Scholar] [CrossRef]
  122. Atianand, M.K.; Hu, W.; Satpathy, A.T.; Shen, Y.; Ricci, E.P.; Alvarez-Dominguez, J.R.; Bhatta, A.; Schattgen, S.A.; McGowan, J.D.; Blin, J.; et al. A Long Noncoding RNA lincRNA-EPS Acts as a Transcriptional Brake to Restrain Inflammation. Cell 2016, 165, 1672–1685. [Google Scholar] [CrossRef] [PubMed]
  123. Hu, G.; Gong, A.Y.; Wang, Y.; Ma, S.; Chen, X.; Chen, J.; Su, C.J.; Shibata, A.; Strauss-Soukup, J.K.; Drescher, K.M.; et al. LincRNA-Cox2 Promotes Late Inflammatory Gene Transcription in Macrophages through Modulating SWI/SNF-Mediated Chromatin Remodeling. J. Immunol. 2016, 196, 2799–2808. [Google Scholar] [CrossRef] [PubMed]
  124. Zhao, X.; Tang, Z.; Zhang, H.; Atianjoh, F.E.; Zhao, J.Y.; Liang, L.; Wang, W.; Guan, X.; Kao, S.C.; Tiwari, V.; et al. A long noncoding RNA contributes to neuropathic pain by silencing Kcna2 in primary afferent neurons. Nat. Neurosci. 2013, 16, 1024–1031. [Google Scholar] [CrossRef] [PubMed]
  125. Ruan, X.; Li, P.; Ma, Y.; Jiang, C.F.; Chen, Y.; Shi, Y.; Gupta, N.; Seifuddin, F.; Pirooznia, M.; Ohnishi, Y.; et al. Identification of human long noncoding RNAs associated with nonalcoholic fatty liver disease and metabolic homeostasis. J. Clin. Investig. 2021, 131, e136336. [Google Scholar] [CrossRef] [PubMed]
  126. Hennessy, E.J.; van Solingen, C.; Scacalossi, K.R.; Ouimet, M.; Afonso, M.S.; Prins, J.; Koelwyn, G.J.; Sharma, M.; Ramkhelawon, B.; Carpenter, S.; et al. The long noncoding RNA CHROME regulates cholesterol homeostasis in primate. Nat. Metab. 2019, 1, 98–110. [Google Scholar] [CrossRef] [PubMed]
  127. Du, Q.; Hoover, A.R.; Dozmorov, I.; Raj, P.; Khan, S.; Molina, E.; Chang, T.C.; de la Morena, M.T.; Cleaver, O.B.; Mendell, J.T.; et al. MIR205HG Is a Long Noncoding RNA that Regulates Growth Hormone and Prolactin Production in the Anterior Pituitary. Dev. Cell 2019, 49, 618–631.e5. [Google Scholar] [CrossRef] [PubMed]
  128. Zhang, P.; Cao, L.; Fan, P.; Mei, Y.; Wu, M. LncRNA-MIF, a c-Myc-activated long non-coding RNA, suppresses glycolysis by promoting Fbxw7-mediated c-Myc degradation. EMBO Rep. 2016, 17, 1204–1220. [Google Scholar] [CrossRef] [PubMed]
  129. Zheng, X.; Han, H.; Liu, G.P.; Ma, Y.X.; Pan, R.L.; Sang, L.J.; Li, R.H.; Yang, L.J.; Marks, J.R.; Wang, W.; et al. LncRNA wires up Hippo and Hedgehog signaling to reprogramme glucose metabolism. EMBO J. 2017, 36, 3325–3335. [Google Scholar] [CrossRef]
  130. McClintock, M.A.; Dix, C.I.; Johnson, C.M.; McLaughlin, S.H.; Maizels, R.J.; Hoang, H.T.; Bullock, S.L. RNA-directed activation of cytoplasmic dynein-1 in reconstituted transport RNPs. Elife 2018, 7, e36312. [Google Scholar] [CrossRef]
  131. Lin, A.; Hu, Q.; Li, C.; Xing, Z.; Ma, G.; Wang, C.; Li, J.; Ye, Y.; Yao, J.; Liang, K.; et al. The LINK-A lncRNA interacts with PtdIns(3,4,5)P(3) to hyperactivate AKT and confer resistance to AKT inhibitors. Nat. Cell Biol. 2017, 19, 238–251. [Google Scholar] [CrossRef]
  132. Ma, Y.; Zhang, J.; Wen, L.; Lin, A. Membrane-lipid associated lncRNA: A new regulator in cancer signaling. Cancer Lett. 2018, 419, 27–29. [Google Scholar] [CrossRef] [PubMed]
  133. Wang, F.; Wang, Q.; Liu, B.; Mei, L.; Ma, S.; Wang, S.; Wang, R.; Zhang, Y.; Niu, C.; Xiong, Z.; et al. The long noncoding RNA Synage regulates synapse stability and neuronal function in the cerebellum. Cell Death Differ. 2021, 28, 2634–2650. [Google Scholar] [CrossRef] [PubMed]
  134. Samaddar, S.; Banerjee, S. Far from the nuclear crowd: Cytoplasmic lncRNA and their implications in synaptic plasticity and memory. Neurobiol. Learn. Mem. 2021, 185, 107522. [Google Scholar] [CrossRef]
  135. Wei, W.; Zhao, Q.; Wang, Z.; Liau, W.S.; Basic, D.; Ren, H.; Marshall, P.R.; Zajaczkowski, E.L.; Leighton, L.J.; Madugalle, S.U.; et al. ADRAM is an experience-dependent long noncoding RNA that drives fear extinction through a direct interaction with the chaperone protein 14-3-3. Cell Rep. 2022, 38, 110546. [Google Scholar] [CrossRef]
  136. Holland, N.; Bolognesi, C.; Kirsch-Volders, M.; Bonassi, S.; Zeiger, E.; Knasmueller, S.; Fenech, M. The micronucleus assay in human buccal cells as a tool for biomonitoring DNA damage: The HUMN project perspective on current status and knowledge gaps. Mutat. Res. 2008, 659, 93–108. [Google Scholar] [CrossRef] [PubMed]
  137. Fenech, M.; Holland, N.; Zeiger, E.; Chang, W.P.; Burgaz, S.; Thomas, P.; Bolognesi, C.; Knasmueller, S.; Kirsch-Volders, M.; Bonassi, S. The HUMN and HUMNxL international collaboration projects on human micronucleus assays in lymphocytes and buccal cells--past, present and future. Mutagenesis 2011, 26, 239–245. [Google Scholar] [CrossRef]
  138. Besaratinia, A.; Bates, S.E.; Pfeifer, G.P. Mutational signature of the proximate bladder carcinogen N-hydroxy-4-acetylaminobiphenyl: Inconsistency with the p53 mutational spectrum in bladder cancer. Cancer Res. 2002, 62, 4331–4338. [Google Scholar]
  139. Torres-Bugarin, O.; Zavala-Cerna, M.G.; Nava, A.; Flores-Garcia, A.; Ramos-Ibarra, M.L. Potential uses, limitations, and basic procedures of micronuclei and nuclear abnormalities in buccal cells. Dis. Markers 2014, 2014, 956835. [Google Scholar] [CrossRef]
  140. Cheng, G.; Guo, J.; Carmella, S.G.; Lindgren, B.; Ikuemonisan, J.; Jensen, J.; Hatsukami, D.K.; Balbo, S.; Hecht, S.S. Increased Acrolein-DNA Adducts in Buccal Brushings of e-Cigarette Users. Carcinogenesis 2022, 43, 437–444. [Google Scholar] [CrossRef]
  141. Silverman, S. Oral Cancer, 5th ed.; BC Decker, Inc.: Hamilton, ON, Canada, 2003. [Google Scholar]
  142. Proia, N.K.; Paszkiewicz, G.M.; Nasca, M.A.; Franke, G.E.; Pauly, J.L. Smoking and smokeless tobacco-associated human buccal cell mutations and their association with oral cancer—A review. Cancer Epidemiol. Biomark. Prev. 2006, 15, 1061–1077. [Google Scholar] [CrossRef]
  143. Cairns, J. Mutation selection and the natural history of cancer. Nature 1975, 255, 197–200. [Google Scholar] [CrossRef] [PubMed]
  144. Rosin, M.P. The use of the micronucleus test on exfoliated cells to identify anti-clastogenic action in humans: A biological marker for the efficacy of chemopreventive agents. Mutat. Res. 1992, 267, 265–276. [Google Scholar] [CrossRef] [PubMed]
  145. Foiles, P.G.; Miglietta, L.M.; Quart, A.M.; Quart, E.; Kabat, G.C.; Hecht, S.S. Evaluation of 32P-postlabeling analysis of DNA from exfoliated oral mucosa cells as a means of monitoring exposure of the oral cavity to genotoxic agents. Carcinogenesis 1989, 10, 1429–1434. [Google Scholar] [CrossRef] [PubMed]
  146. Zhang, Y.; Chen, S.Y.; Hsu, T.; Santella, R.M. Immunohistochemical detection of malondialdehyde-DNA adducts in human oral mucosa cells. Carcinogenesis 2002, 23, 207–211. [Google Scholar] [CrossRef]
  147. Besaratinia, A.; Maas, L.M.; Brouwer, E.M.; Kleinjans, J.C.; Van Schooten, F.J. Comparison between smoking-related DNA adduct analysis in induced sputum and peripheral blood lymphocytes. Carcinogenesis 2000, 21, 1335–1340. [Google Scholar]
  148. Schwartz, J.L.; Muscat, J.E.; Baker, V.; Larios, E.; Stephenson, G.D.; Guo, W.; Xie, T.; Gu, X.; Chung, F.L. Oral cytology assessment by flow cytometry of DNA adducts, aneuploidy, proliferation and apoptosis shows differences between smokers and non-smokers. Oral. Oncol. 2003, 39, 842–854. [Google Scholar] [CrossRef]
  149. Schwartz, J.L.; Baker, V.; Larios, E.; Chung, F.L. Molecular and cellular effects of green tea on oral cells of smokers: A pilot study. Mol. Nutr. Food Res. 2005, 49, 43–51. [Google Scholar] [CrossRef]
  150. Saatci, C.; Ozkul, Y.; Tahiri, S.; Caglayan, A.O.; Turhan, A.B.; Dundar, M. The effect of maras powder on DNA methylation and micronucleus formation in human buccal tissue. J. Toxicol. Environ. Health Part A 2008, 71, 396–404. [Google Scholar] [CrossRef]
  151. Slebos, R.J.; Li, M.; Vadivelu, S.; Burkey, B.B.; Netterville, J.L.; Sinard, R.; Gilbert, J.; Murphy, B.; Chung, C.H.; Shyr, Y.; et al. Microsatellite mutations in buccal cells are associated with aging and head and neck carcinoma. Br. J. Cancer 2008, 98, 619–626. [Google Scholar] [CrossRef]
  152. Besaratinia, A.; Kleinjans, J.C.; Van Schooten, F.J. Biomonitoring of tobacco smoke carcinogenicity by dosimetry of DNA adducts and genotyping and phenotyping of biotransformational enzymes: A review on polycyclic aromatic hydrocarbons. Biomarkers 2002, 7, 209–229. [Google Scholar]
  153. Shani, T.; Onn, A.; Kabha, A.; Ben-Dov, I.; Adam, I.; Amariglio, N.; Yahalom, R.; Rechavi, G.; Trakhtenbrot, L.; Hirshberg, A. Chromosomal numerical aberrations in apparently normal oral mucosa of heavy smokers affected by lung cancer. Oral. Oncol. 2010, 46, 96–99. [Google Scholar] [CrossRef]
  154. Tommasi, S.; Blumenfeld, H.; Besaratinia, A. Vaping dose, device type, and e-liquid flavor are determinants of DNA damage in electronic cigarette users. Nicotine Tob. Res. 2023, 25, 1145–1154. [Google Scholar] [CrossRef] [PubMed]
  155. Bonassi, S.; Coskun, E.; Ceppi, M.; Lando, C.; Bolognesi, C.; Burgaz, S.; Holland, N.; Kirsh-Volders, M.; Knasmueller, S.; Zeiger, E.; et al. The HUman MicroNucleus project on eXfoLiated buccal cells (HUMN(XL)): The role of life-style, host factors, occupational exposures, health status, and assay protocol. Mutat. Res. 2011, 728, 88–97. [Google Scholar] [CrossRef] [PubMed]
  156. Greenspan, E.J.; Lee, H.; Dyba, M.; Pan, J.; Mekambi, K.; Johnson, T.; Blancato, J.; Mueller, S.; Berry, D.L.; Chung, F.L. High-throughput, quantitative analysis of acrolein-derived DNA adducts in human oral cells by immunohistochemistry. J. Histochem. Cytochem. 2012, 60, 844–853. [Google Scholar] [CrossRef]
  157. Pan, J.; Awoyemi, B.; Xuan, Z.; Vohra, P.; Wang, H.T.; Dyba, M.; Greenspan, E.; Fu, Y.; Creswell, K.; Zhang, L.; et al. Detection of acrolein-derived cyclic DNA adducts in human cells by monoclonal antibodies. Chem. Res. Toxicol. 2012, 25, 2788–2795. [Google Scholar] [CrossRef] [PubMed]
  158. Stepanov, I.; Muzic, J.; Le, C.T.; Sebero, E.; Villalta, P.; Ma, B.; Jensen, J.; Hatsukami, D.; Hecht, S.S. Analysis of 4-hydroxy-1-(3-pyridyl)-1-butanone (HPB)-releasing DNA adducts in human exfoliated oral mucosa cells by liquid chromatography-electrospray ionization-tandem mass spectrometry. Chem. Res. Toxicol. 2013, 26, 37–45. [Google Scholar] [CrossRef] [PubMed]
  159. Teschendorff, A.E.; Yang, Z.; Wong, A.; Pipinikas, C.P.; Jiao, Y.; Jones, A.; Anjum, S.; Hardy, R.; Salvesen, H.B.; Thirlwell, C.; et al. Correlation of Smoking-Associated DNA Methylation Changes in Buccal Cells With DNA Methylation Changes in Epithelial Cancer. JAMA Oncol. 2015, 1, 476–485. [Google Scholar] [CrossRef] [PubMed]
  160. De Oliveira, S.R.; Da Silva, I.C.; Mariz, B.A.; Pereira, A.M.; De Oliveira, N.F. DNA methylation analysis of cancer-related genes in oral epithelial cells of healthy smokers. Arch. Oral Biol. 2015, 60, 825–833. [Google Scholar] [CrossRef] [PubMed]
  161. Bhutani, M.; Pathak, A.K.; Fan, Y.H.; Liu, D.D.; Lee, J.J.; Tang, H.; Kurie, J.M.; Morice, R.C.; Kim, E.S.; Hong, W.K.; et al. Oral epithelium as a surrogate tissue for assessing smoking-induced molecular alterations in the lungs. Cancer Prev. Res. 2008, 1, 39–44. [Google Scholar] [CrossRef]
  162. Wangsri, S.; Subbalekha, K.; Kitkumthorn, N.; Mutirangura, A. Patterns and possible roles of LINE-1 methylation changes in smoke-exposed epithelia. PLoS ONE 2012, 7, e45292. [Google Scholar] [CrossRef]
  163. Shah, V.; Sridhar, S.; Beane, J.; Brody, J.S.; Spira, A. SIEGE: Smoking Induced Epithelial Gene Expression Database. Nucleic Acids Res. 2005, 33, D573–D579. [Google Scholar] [CrossRef] [PubMed]
  164. Spivack, S.D.; Hurteau, G.J.; Jain, R.; Kumar, S.V.; Aldous, K.M.; Gierthy, J.F.; Kaminsky, L.S. Gene-environment interaction signatures by quantitative mRNA profiling in exfoliated buccal mucosal cells. Cancer Res. 2004, 64, 6805–6813. [Google Scholar] [CrossRef]
  165. Smith, R.V.; Schlecht, N.F.; Childs, G.; Prystowsky, M.B.; Belbin, T.J. Pilot study of mucosal genetic differences in early smokers and nonsmokers. Laryngoscope 2006, 116, 1375–1379. [Google Scholar] [CrossRef]
  166. Boyle, J.O.; Gumus, Z.H.; Kacker, A.; Choksi, V.L.; Bocker, J.M.; Zhou, X.K.; Yantiss, R.K.; Hughes, D.B.; Du, B.; Judson, B.L.; et al. Effects of cigarette smoke on the human oral mucosal transcriptome. Cancer Prev. Res. 2010, 3, 266–278. [Google Scholar] [CrossRef] [PubMed]
  167. Sridhar, S.; Schembri, F.; Zeskind, J.; Shah, V.; Gustafson, A.M.; Steiling, K.; Liu, G.; Dumas, Y.M.; Zhang, X.; Brody, J.S.; et al. Smoking-induced gene expression changes in the bronchial airway are reflected in nasal and buccal epithelium. BMC Genom. 2008, 9, 259. [Google Scholar] [CrossRef] [PubMed]
  168. Kupfer, D.M.; White, V.L.; Jenkins, M.C.; Burian, D. Examining smoking-induced differential gene expression changes in buccal mucosa. BMC Med. Genom. 2010, 3, 24. [Google Scholar] [CrossRef]
  169. Schlage, W.K.; Iskandar, A.R.; Kostadinova, R.; Xiang, Y.; Sewer, A.; Majeed, S.; Kuehn, D.; Frentzel, S.; Talikka, M.; Geertz, M.; et al. In vitro systems toxicology approach to investigate the effects of repeated cigarette smoke exposure on human buccal and gingival organotypic epithelial tissue cultures. Toxicol. Mech. Methods 2014, 24, 470–487. [Google Scholar] [CrossRef]
  170. Garcia-Closas, M.; Egan, K.M.; Abruzzo, J.; Newcomb, P.A.; Titus-Ernstoff, L.; Franklin, T.; Bender, P.K.; Beck, J.C.; Le Marchand, L.; Lum, A.; et al. Collection of genomic DNA from adults in epidemiological studies by buccal cytobrush and mouthwash. Cancer Epidemiol. Biomark. Prev. 2001, 10, 687–696. [Google Scholar]
  171. De, A. Wnt/Ca2+ signaling pathway: A brief overview. Acta Biochim. Biophys. Sin. 2011, 43, 745–756. [Google Scholar] [CrossRef]
  172. Guo, W.; Giancotti, F.G. Integrin signalling during tumour progression. Nat. Rev. Mol. Cell Biol. 2004, 5, 816–826. [Google Scholar] [CrossRef]
  173. Fritz, G.; Henninger, C. Rho GTPases: Novel Players in the Regulation of the DNA Damage Response? Biomolecules 2015, 5, 2417–2434. [Google Scholar] [CrossRef] [PubMed]
  174. Moawad, A.M.; Hassan, F.M.; Sabry Abdelfattah, D.; Basyoni, H.A.M. Long non-coding RNA ENST00000414355 as a biomarker of cadmium exposure regulates DNA damage and apoptosis. Toxicol. Ind. Health 2021, 37, 745–751. [Google Scholar] [CrossRef] [PubMed]
  175. Liang, J.; Liu, X.H.; Chen, X.M.; Song, X.L.; Li, W.; Huang, Y. Emerging Roles of Non-Coding RNAs in Childhood Asthma. Front. Pharmacol. 2022, 13, 856104. [Google Scholar] [CrossRef] [PubMed]
  176. Tommasi, S.; Pabustan, N.; Li, M.; Chen, Y.; Siegmund, K.D.; Besaratinia, A. A novel role for vaping in mitochondrial gene dysregulation and inflammation fundamental to disease development. Sci. Rep. 2021, 11, 22773. [Google Scholar] [CrossRef]
  177. Besaratinia, A.; Tommasi, S. Epigenetics of human melanoma: Promises and challenges. J. Mol. Cell Biol. 2014, 6, 356–367. [Google Scholar] [CrossRef] [PubMed]
  178. Zhong, J.; Agha, G.; Baccarelli, A.A. The Role of DNA Methylation in Cardiovascular Risk and Disease: Methodological Aspects, Study Design, and Data Analysis for Epidemiological Studies. Circ. Res. 2016, 118, 119–131. [Google Scholar] [CrossRef] [PubMed]
  179. Gesthalter, Y.B.; Vick, J.; Steiling, K.; Spira, A. Translating the transcriptome into tools for the early detection and prevention of lung cancer. Thorax 2015, 70, 476–481. [Google Scholar] [CrossRef] [PubMed]
  180. Billatos, E.; Vick, J.L.; Lenburg, M.E.; Spira, A.E. The Airway Transcriptome as a Biomarker for Early Lung Cancer Detection. Clin. Cancer Res. 2018, 24, 2984–2992. [Google Scholar] [CrossRef]
  181. Li, Y.; Wang, C.; Li, T.; Ma, L.; Fan, F.; Jin, Y.; Shen, J. The whole transcriptome and proteome changes in the early stage of myocardial infarction. Cell Death Discov. 2019, 5, 73. [Google Scholar] [CrossRef]
  182. Besaratinia, A.; Tommasi, S. An opportune and unique research to evaluate the public health impact of electronic cigarettes. Cancer Causes Control CCC 2017, 28, 1167–1171. [Google Scholar] [CrossRef]
  183. Benowitz, N.L.; Fraiman, J.B. Cardiovascular effects of electronic cigarettes. Nat. Rev. Cardiol. 2017, 14, 447–456. [Google Scholar] [CrossRef] [PubMed]
  184. Besaratinia, A.; Pfeifer, G.P. Second-hand smoke and human lung cancer. Lancet Oncol. 2008, 9, 657–666. [Google Scholar] [CrossRef] [PubMed]
  185. Moore, L.E.; Pfeiffer, R.M.; Poscablo, C.; Real, F.X.; Kogevinas, M.; Silverman, D.; Garcia-Closas, R.; Chanock, S.; Tardon, A.; Serra, C.; et al. Genomic DNA hypomethylation as a biomarker for bladder cancer susceptibility in the Spanish Bladder Cancer Study: A case-control study. Lancet Oncol. 2008, 9, 359–366. [Google Scholar] [CrossRef] [PubMed]
  186. Zou, A.E.; Ku, J.; Honda, T.K.; Yu, V.; Kuo, S.Z.; Zheng, H.; Xuan, Y.; Saad, M.A.; Hinton, A.; Brumund, K.T.; et al. Transcriptome sequencing uncovers novel long noncoding and small nucleolar RNAs dysregulated in head and neck squamous cell carcinoma. Rna 2015, 21, 1122–1134. [Google Scholar] [CrossRef] [PubMed]
  187. Münzel, T.; Hahad, O.; Kuntic, M.; Keaney, J.F.; Deanfield, J.E.; Daiber, A. Effects of tobacco cigarettes, e-cigarettes, and waterpipe smoking on endothelial function and clinical outcomes. Eur. Heart J. 2020, 41, 4057–4070. [Google Scholar] [CrossRef] [PubMed]
  188. Ali, S.A.; Peffers, M.J.; Ormseth, M.J.; Jurisica, I.; Kapoor, M. The non-coding RNA interactome in joint health and disease. Nat. Rev. Rheumatol. 2021, 17, 692–705. [Google Scholar] [CrossRef]
  189. Toden, S.; Goel, A. Non-coding RNAs as liquid biopsy biomarkers in cancer. Br. J. Cancer 2022, 126, 351–360. [Google Scholar] [CrossRef] [PubMed]
  190. Fok, E.T.; Scholefield, J.; Fanucchi, S.; Mhlanga, M.M. The emerging molecular biology toolbox for the study of long noncoding RNA biology. Epigenomics 2017, 9, 1317–1327. [Google Scholar] [CrossRef]
  191. Salehi, S.; Taheri, M.N.; Azarpira, N.; Zare, A.; Behzad-Behbahani, A. State of the art technologies to explore long non-coding RNAs in cancer. J. Cell Mol. Med. 2017, 21, 3120–3140. [Google Scholar] [CrossRef]
  192. Ramanathan, M.; Porter, D.F.; Khavari, P.A. Methods to study RNA-protein interactions. Nat. Methods 2019, 16, 225–234. [Google Scholar] [CrossRef]
  193. Chowdhary, A.; Satagopam, V.; Schneider, R. Long Non-coding RNAs: Mechanisms, Experimental, and Computational Approaches in Identification, Characterization, and Their Biomarker Potential in Cancer. Front. Genet. 2021, 12, 649619. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Aberrantly expressed genes in the oral epithelial cells of vapers and smokers as compared to non-users. (A) Numbers of upregulated and downregulated gene transcripts in vapers and smokers are indicated. Venn diagrams of all dysregulated genes (B) and cancer-related dysregulated genes (C) in vapers and smokers are shown. DEGs = differentially expressed genes. Data are from our previous publication (Ref. [13]). Detailed descriptions of data pre-processing, alignment, quantification, and differential expression analysis are provided in Ref. [13].
Figure 1. Aberrantly expressed genes in the oral epithelial cells of vapers and smokers as compared to non-users. (A) Numbers of upregulated and downregulated gene transcripts in vapers and smokers are indicated. Venn diagrams of all dysregulated genes (B) and cancer-related dysregulated genes (C) in vapers and smokers are shown. DEGs = differentially expressed genes. Data are from our previous publication (Ref. [13]). Detailed descriptions of data pre-processing, alignment, quantification, and differential expression analysis are provided in Ref. [13].
Ijms 25 08554 g001
Figure 2. Classification of aberrantly expressed transcripts in the oral epithelial cells of vapers and smokers as compared to non-users. (A) Percentages of protein coding genes (PC), lncRNAs, and others (including (un)processed pseudogene, transcribed (un)processed pseudogene, to be experimentally confirmed (TEC), Ig V gene, rRNA, scaRNA, snoRNA, scaRNA/snoRNA, snRNA, and unitary pseudogene) are shown. (B) Numbers of upregulated and downregulated lncRNAs in vapers and smokers are indicated. (C) Sub-types of dysregulated lncRNAs in vapers and smokers. Total number of each sub-type of dysregulated lncRNAs is indicated. Data are from our previous publication (Ref. [13]). Detailed descriptions of data pre-processing, alignment, quantification, and differential expression analysis are provided in Ref. [13].
Figure 2. Classification of aberrantly expressed transcripts in the oral epithelial cells of vapers and smokers as compared to non-users. (A) Percentages of protein coding genes (PC), lncRNAs, and others (including (un)processed pseudogene, transcribed (un)processed pseudogene, to be experimentally confirmed (TEC), Ig V gene, rRNA, scaRNA, snoRNA, scaRNA/snoRNA, snRNA, and unitary pseudogene) are shown. (B) Numbers of upregulated and downregulated lncRNAs in vapers and smokers are indicated. (C) Sub-types of dysregulated lncRNAs in vapers and smokers. Total number of each sub-type of dysregulated lncRNAs is indicated. Data are from our previous publication (Ref. [13]). Detailed descriptions of data pre-processing, alignment, quantification, and differential expression analysis are provided in Ref. [13].
Ijms 25 08554 g002
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Besaratinia, A.; Blumenfeld, H.; Tommasi, S. Exploring the Utility of Long Non-Coding RNAs for Assessing the Health Consequences of Vaping. Int. J. Mol. Sci. 2024, 25, 8554. https://doi.org/10.3390/ijms25158554

AMA Style

Besaratinia A, Blumenfeld H, Tommasi S. Exploring the Utility of Long Non-Coding RNAs for Assessing the Health Consequences of Vaping. International Journal of Molecular Sciences. 2024; 25(15):8554. https://doi.org/10.3390/ijms25158554

Chicago/Turabian Style

Besaratinia, Ahmad, Hannah Blumenfeld, and Stella Tommasi. 2024. "Exploring the Utility of Long Non-Coding RNAs for Assessing the Health Consequences of Vaping" International Journal of Molecular Sciences 25, no. 15: 8554. https://doi.org/10.3390/ijms25158554

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop