Skip to main content
Erschienen in: European Journal of Medical Research 1/2023

Open Access 01.12.2023 | Review

The potential roles of cigarette smoke-induced extracellular vesicles in oral leukoplakia

verfasst von: Qiao Peng, Ning Duan, Xiang Wang, Wenmei Wang

Erschienen in: European Journal of Medical Research | Ausgabe 1/2023

Abstract

Background

The onset of oral leukoplakia (OLK), the most common oral lesion with a high risk of malignant transformation, is closely associated with the exposure of cigarette smoke. Cigarette smoke is a complicated mixture of more than 4500 different chemicals including various oxidants and free radical, which contributes to the onset of immune and inflammatory response or even carcinogenesis. Recent studies have proved that the exposure of cigarette smoke leads to the onset and aggravation of many diseases via significantly changed the production and components of extracellular vesicles. The extracellular vesicles are membrane-enclosed nanosized particles secreted by diverse cells and involved in cell–cell communication because of their ability to deliver a number of bioactive molecules including proteins, lipids, DNAs and RNAs. Getting insight into the mechanisms of extracellular vesicles in regulating OLK upon cigarette smoke stimulation contributes to unravel the pathophysiology of OLK in-depth. However, evidence done on the role of extracellular vesicles in cigarette smoke-induced OLK is still in its infancy.

Materials and methods

Relevant literatures on cigarette smoke, oral leukoplakia and extracellular vesicles were searched in PubMed database.

Conclusions

In this review, we summarize the recent findings about the function of extracellular vesicles in the pathogenesis of cigarette smoke-induced diseases, and to infer their potential utilizations as diagnostic biomarkers, prognostic evaluation, and therapeutic targets of OLK in the future.
Hinweise

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Introduction

A robust body of evidence have accumulated documenting that cigarette smoke (CS) is an essential risk factor for multiple human diseases worldwide including cancers, cardiovascular disease, chronic lung diseases, asthma and inflammatory skin disorders [14]. The prevalent use of CS and the series of diseases induced by CS result in serious economic and health burden to the world, especially for the undeveloped countries [5]. A cross-over study performed by Kerr et al. found that the heart rate of healthy individuals increased only after 4 h use of tobacco smoking/electronic cigarettes [6]. Moreover, the peak expiratory flow remarkably down-regulated in electronic cigarettes group [6]. These data suggested the acute exposure to CS influenced vascular and respiratory function. It is reported that CS is a complicated mixture of chemicals that contains about 1015–17 oxidants/free radicals and up to 4500–4700 different compounds, such as nicotine, tobacco-specific nitrosamines, reactive aldehydes and quinones, per puff [7, 8]. Most of these components manifest great cellular toxicity pathogenesis by modulating the cellular survival, apoptosis, ferroptosis, autophagy, proliferation and angiogenesis [911]. They also act as mutagen and carcinogen factors promoting tumorigenesis [12, 13].
The tissues of oral cavity are always inevitably to suffer damage, because it is the first part stimulated by CS. In fact, CS is one of the most established major risk factors for many oral diseases, including periodontal disease, dental caries, aphthous ulcers, primary Sjögren’s syndrome and peri-implantitis [1416]. According to a meta-analysis performed by Leite et al., smoking increased the risk of periodontitis by 85% and this risk could be decreased by about 14% if smoking factor is eliminated in this population, which well-demonstrated the detrimental effect of smoking on periodontitis [17]. CS also represents the most important etiological factor that promotes oral potentially malignant disorders (OPMD) and oral squamous cell carcinoma (OSCC) [12, 18, 19]. Among the OPMDs, oral leukoplakia (OLK) is the most common potentially premalignant lesion that showing a primary white change of oral mucosa [20, 21]. The prevalence for OLK is between 1.5% and 2.6%, and the risk malignant transformation is up to 5–36% [22, 23]. Strong evidence have been found that the incidence of OLK was closely associated with the addiction of tobacco [24, 25]. In addition, the correlation between CS and OLK is underlined by a dose-dependent manner [26]. The complicated pathogenic impacts of CS are considered as the attribution that leads to OLK and even its malignant transformation, In a prior study by Ye et al., they found that the immortalized human oral mucosa epithelial cell line leuk1 could significantly accumulated reactive oxygen species (ROS) upon cigarette smoke extract (CSE) exposure compared to the unexposed co-cultured cells, indicating the destructive effect of oxidative stress on the structure and function of oral epithelium [27].
Recent evidence identify that the oxidative stress contributes to the release of extracellular vesicles (EVs) [2830]. EVs are a heterogeneous group of cell-derived membranous structures that are categories as exosomes and ectosomes according to their different mechanism of formation and approximate size [31] (Fig. 1). Exosomes are EVs in the size range of 40–160 nm in diameter with an endosome origin, termed a multivesicular body biogenesis [31, 32]. Different from exosomes, the ectosomes, including microvesicles and microparticles, are ubiquitous vesicles directly released from the plasma membrane through outward budding with a size range of 50 nm–1 μm in diameter [31, 32] (Fig. 1). EVs are derived from almost all types of cells and can be found in diverse biological fluids, including plasma, serum, lymph, saliva, urine, bile, cerebrospinal fluid and human milk [33, 34] (Fig. 2). EVs have emerged as important modulators in intercellular information transmission by delivering their biological cargo, such as proteins, lipids, RNAs and DNAs [35, 36] (Fig. 2).
Currently, emerging evidence have attempted to explain how CS affects the generation and release of EVs, and what is the specific molecular mechanism or pathways they interfered that cause the disease occurrence, especially for lung, cardiovascular and cancer-related diseases. However, little is known about how CS-induced EVs regulate the pathophysiological processes of OLK, even though the oral mucosa is the first-line site affected by CS. Thus, attempts to understand the roles of CS-induced EVs in the microenvironment of OLK are of great importance, which not only helps to clarify the immunopathogenesis of OLK in-depth, but also provides novel strategies for the diagnosis and treatment of OLK, even inhibition of its malignant transformation.

Advances of EVs as biological biomarkers

Although the traditional tissue biopsies remains the gold standard for disease diagnosis, the procedure is invasive and commonly constrained by its repeatability and brings economic and mental burden to patients [37, 38]. These limitations of traditional tissue biopsy promote the application of biofluids/liquid biopsy technique in clinical practice. Interestingly, EVs display great superiority in liquid biopsy with the following reasons (Fig. 2): first, the prevalence of EVs that exist in almost all body fluids makes it possible to implement less invasive operation and to apply in different types of diseases [33, 34]. Second, it is time saving, sample repeatable and cost effective due to the rapid advancement in EVs-related purification and identification techniques [39, 40]. Third, EVs are secreted continuously by living cells, providing exciting opportunities for real time monitoring of lesions, disease diagnosis, prediction and surveillance [41]. Fourth, the EVs contain differentially expressed cargos and specific surface markers from their parental cells, which could accurately reflect the pathological factors of diseases on both molecular and cellular levels [42, 43]. This might largely contribute to the progression of targeted therapy in future. Finally, the membranous shell of EVs show high stability to guarantee the long-term existence of their contents [44], which would not be affected by the different processing time of samples, ensuring the stable and reliable detection results.

CS-induced EVs as biomarkers for the diagnosis and treatment of diseases

The exposure of CS could increase the production of EVs from diverse types of cells, including endothelial cells, epithelial cells and fibroblasts, which might rely on the redox modifications of protein thiols [45, 46]. Serban et al. showed that CS-induced endothelial microparticles (EMPs) contained predominantly exosomes that were largely enriched in let-7d, miR-191, miR-126 and miR-125a [47], which may be important in predicting potential function of exosomes as paracrine effectors in patients with chronic obstructive pulmonary disease (COPD). In addition, these EMPs were ceramide-rich and required the ceramide-synthesis enzyme acid sphingomyelinase (aSMase) for microparticle release [47]. The aSMase is an enzyme that is classically associated with stress-induced apoptosis and shown higher activity in plasma of patients with COPD or of CS-exposed mice. These reveal that CS-induced EMPs could be a promising indicator of the functional state of the endothelium in smokers. Stassen et al. identified that CSE promoted the secretion of CD63+CD81+ extracellular vesicles (exosomes) by bronchial epithelial cells [48]. Moreover, these exosomes were enriched in tissue factor (TF) and exerted a TF-dependent procoagulant activity compared with exosomes by unexposed cells [49], suggesting that TF+ exosomes might contribute to the elevated cardiovascular risk in smokers and consider as biomarkers of exposure to CSE.

The EVs-borne cargo are changed by CS exposure

The change of proteins in EVs exposure to CS

The protein composition of CS-induced EVs could be influenced by many factors including gender, age, hormones and smoking status. In the case of gender, Varming et al., by analyzing the proteins of plasma exosomes, have found a lower level of amphiregulin (AREG), MUC1, MUC18 (CD146), CD13 and tumor susceptibility gene 101 (TSG101) in female smokers compared to the group of non-smoking females, and this was not seen for male smokers [50]. However, the male smokers showed a higher expression level of CD171, programmed death-ligand 1 (PD-L1) and TSG101 when comparing to the smoking females and non-smoking males [50]. With respect to age, Mobarrez et al. identified higher expression levels of CD41, CD235, TF, and phosphatidylserine (PS) in CS-induced circulating microparticles from younger and older groups compared to those of middle age [51].
In our preliminary study, we have collected the plasma exosomes from patients with OLK and healthy individuals. By proteomic analysis, we identified 10 differentially expressed proteins between the two groups. To be specific, there were 4 upregulated proteins (P01742, Q9BXG8, P06310, P19652) and 6 downregulated proteins (A0A0B4J1X8, Q9NZP8, P61604, Q01459, Q6UXB8, Q9Y5Y7) in patients with CS-induced OLK compared to healthy individuals (data unpublished), suggesting that the alteration of circulating exosomal proteins might play key roles in promoting OLK development and be considered as biomarkers for early diagnosis of OLK.

The change of cytokines in EVs exposure to CS

CS could change the expression level of cytokine/chemokines in EVs. In patients with CS-induced COPD, the pro-inflammatory Wnt5a and cytokines including interleukin (IL)-8, IL-1β, IL-6 and tumor necrosis factor (TNF)-α in serum EVs were significantly elevated [52]. The functionality of Wnt5a is multifaceted. Apart from as the pro-inflammatory Wnt ligand, it can prohibit the transcription of anti-inflammatory peroxisome proliferator-activated receptor γ and modulate the polarization of M1/M2 macrophages. IL-6, as the NF-κB-dependent cytokines, is a prototypical cytokine and functions like a double-edged sword for the host. The appropriate expression of IL-6 plays a positive role in eliminating infectious agents and restoring damaged tissues, whereas excessive expression of IL-6 exerts a positive role in the progression of different inflammatory diseases even tumors [53]. For patients with OLK, an elevation of salivary IL-6 levels was found compared to healthy controls, with a positive relationship with the severity of dysplasia [54, 55]. Moreover, use of tobacco was seen to significantly promote the elevation of IL-6. These evidence pose the possibility that aberrant expression of EVs-derived IL-6 play a pathological role in the dysplasia of patients with OLK.
Kumar et al. found that the packaging of CD9 in rat plasma-derived EVs was significantly increased after self-administered nicotine, one of the major addictive ingredient in CS [56]. The increased expression of CD9 in plasma EVs might indicate an effect of nicotine on EVs formation, because CD9 is reported an important organizer of proteins and lipid complexes at the EV membrane [57]. In addition, an increased expression of cytochrome P450 (CYP2A6) in plasma EVs was also found [56]. CYP2A6 is responsible for the metabolism of nicotine. These results suggest that the elevated CYP2A6 in plasma EVs upon nicotine self-administered could cause the increased production of toxic metabolites and induction of oxidative stress, and ultimately result in tissue damage and disease progression. The enhanced release of EVs by reactive carbonyl species (RCS) and ROS stimulation could be blocked by antioxidant glutathione S-transferase (GSH) [48]. Interestingly, Ye et al. found a significantly accumulation of ROS in CSE-exposed leuk1 cells compared with non-exposed cells [27]. Simultaneously, the Nrf2 was activated in both CS-treated animal model and leuk1 cells, which alleviated smoking-induced oxidative stress [27]. In addition, the use of sulphoraphane (SFN), a natural antioxidant agent present in cruciferous vegetables that activates Nrf2, on leuk1 cells increased the expression of GSH [27]. Thus, these data indicate that SFN might be a promising therapeutic strategy to interfere the release of CS-induced EVs.

The change of microRNAs (miRNAs) in EVs exposure to CS

The functionality of miRNAs in EVs, especially in exosomes, are of particular interest at present, because the proportion of miRNAs in exosomes accounts for 50%, which is much higher than in their parental cells [58, 59]. Evidence proved that plasma exosomes of different types of smokers including cigarette smokers, waterpipe smokers, electronic cigarettes users and dual smokers have common differential expression of miRNAs, especially hsa-let-7a-5p [60]. Sundar et al. also identified that the hsa-miR-144-3p, hsa-miR-486-5p, hsa-miR-92a-3p and hsa-miR-93-5p were significantly reduced in CS-induced exosomes from plasma compared to non-smokers, whereas the hsa-miR-29a-3p was significantly elevated [61]. Xu et al. found that the level of serum exosomal miR-21 from smokers were higher than those for non-smokers [62]. In addition, the levels of serum exosomal miR-21 inversely correlated with forced expiratory volume in 1 s/forced vital capacity ratio (FEV1/FVC%), an indicator representing the percentage of the lung size that can be exhaled in 1 s [62]. Taken together, these findings might pave the way for considering circulating exosomal miRNAs as new biomarkers or theranostic factors in smoking-induced diseases.
It is worth noting that the differentially expressed miRNAs in OLK are identified to associate with its malignant transformation [6366]. In the latest scoping review, the authors have included fifteen articles for analysis and identified 21 differentially expressed miRNAs in OLK [67]. To be specific, 6 miRNAs (miR-21, miR-181b, miR-184, miR-345, miR-146a and miR-208-3p) that were categories as oncogenic, 5 miRNAs (miR-31*, miR-3065-5p, miR-129-2-3p, miR-204-5p and miR-375) that were classified as tumor suppressors and 10 miRNAs (miR-138, miR-639, miR-27a-3p, miR-15b, miR-485-5p, miR-200c, miR-19a, miR-424, miR-205 and miR-21) that were related to epithelial–mesenchymal transition, invasion and migration [67]. Among these miRNAs, the authors concluded that miR-21, miR-345, miR-181b, miR-31* might be potential markers of malignant transformation of OLK.

The change of long noncoding RNAs (lncRNAs) in EVs exposure to CS

The lncRNAs are operationally classified as transcripts > 200 nucleotides that do not have any evident protein-coding potential [68, 69]. The distribution of lncRNAs is diverse, which is present in both the nucleus and cytoplasm [70]. LncRNAs are demonstrated to play important roles in multiple biological processed because of their capability to affect gene expression by interacting with DNA, RNAs and proteins [71]. Recently, lncRNAs are found to be selectively packaged into exosomes and function as a messenger in intercellular communication to regulate the recipient cells [72, 73]. Notably, CS exposure could change the lncRNA profile in exosomes. Compared to the non-smokers, Kaur et al. identified 2 upregulated and 5 downregulated lncRNAs in the exosomes from blood plasma of cigarette smokers [74]. In regard to E-cigarette smokers, 13 differentially expressed lncRNAs were found [74]. Among these, the expression level of Bcl2 interacting protein 3-like protein (BNIP3L) revealed approximately fourfold increase [74]. BNIP3L/Nix is a mitochondrial and pro-apoptotic protein integrated in the mitochondrial outer membrane [75]. The most well-accepted mechanism underlying BNIP3L-induced mitophagy is through the binding to Atg8 family including LC3 and BECN1 [76]. Furthermore, BNIP3L-mediated mitophagy might function as a tumor promoter with the evidence that Bnip3l depletion significantly delays the progression of pancreatic cancer and improves the survival in a murine model of pancreatic ductal adenocarcinoma [77].
Liu et al. have found that the expression of LC3-II and Beclin-1/BECN1 were upregulated in leuk-1 cells upon CS stimulation and this upregulation was in a concentration- and time-dependent manner [78], suggesting that CS exposure could trigger the autophagy of human oral epithelial cells. Similarly, Lima et al. also confirmed the significant elevation in the percentage of LC3-II positive cells from the normal oral mucosa to the leukoplakia samples, especially in the upper layers of the epithelium [79]. Interestingly, the aberrant expression of lncRNAs was also found in OLK. By performing microarray, Liu et al. for the first time identified a novel key lncRNA n386251 that was highly expressed in OLK. They named it as lncRNA oral leukoplakia progressed associated 1 (LOLA1) because of their ability to affect the migration, invasion, and epithelial–mesenchymal transition of cells, which was associated with the malignant transformation of OLK [80]. In addition to LOLA1, the lncRNA IFITM4P was also significantly upregulated in OLK samples compared to oral normal mucosa and was the highest in OSCC samples [81]. Moreover, IFITM4P could lead to increased cell growth and colony formation in leuk-1 cells [81], indicating the contribution of IFITM4P as an oncogene in oral carcinogenesis.

CS-induced EVs as mediators for intra/inter-cellular message transmission

The changes of circulating EVs induced by CS are in essence due to the changes of EVs secreted by various cells under the stimulation of CS, and finally released into the peripheral blood or local tissues. The altered composition in EVs upon CS exposure ultimately leads to their dysregulated functions after being engulfed by recipient cells. Researches on the production of EVs, the specific changes of contents, and the mechanism of modulating cells upon CS stimulation could contribute to our deeper understanding in the field of EVs, which will benefits for future use of EVs as diagnostic biomarkers and therapeutic targets of smoke-induced-related diseases.

The change of epithelial cells-derived EVs exposure to CS

The oral mucosa is the first line that encounters with CS, which makes the epithelial cells in oral cavity are inevitably as the first type of cells affected by CS. However, the researches on oral epithelial cells-derived EVs exposure to CS at present are scare and mainly focused on airway epithelial cells (AECs). The proteomic analysis revealed that about 201 proteins were differentially expressed in BEAS-2B AECs-derived EVs exposed to 1% cigarette smoke condensate (CSE) for 24 h compared with the EVs unexposed to CSE. Among these, 24 proteins, including TF, of the pathway haemostasis were remarkably elevated, indicating the procoagulant properties of CS-induced EVs [49]. In addition, the RNA content of EVs released from human small airway epithelial cells (SAE) were also changed upon exposure to CSE. Corsello et al. proved that a total of 289 miRNAs and 62 piwi-interacting RNA (piRNAs) were changed. To be specific, the expression levels of hsa-miR-3913-5p, hsa-miR-574-5p, hsa-miR-500a-5p and piR-50603 in SAE-derived EVs exposed to CSE were significantly upregulated, whereas the expression levels of hsa-miR-618 and pi-R52900 were significantly downregulated [82]. These data promote to identify biomarkers for diagnosis and novel targets for therapeutic strategies of CS-related diseases.
Notably, the altered composition in EVs implies the alteration of functionality of EVs on the recipient cells. Serban et al. showed that the engulfment of EMPs by peripheral blood monocytes-derived macrophages was correlated with significant inhibition of efferocytosis, indicating that EMPs might play an important role in the pathogenesis of diseases linked to inflammation in response to CS [47]. It was reported that the upregulation of miR-21-3p or miR-27b-3p in CSE-treated Beas-2B-derived exosomes could not only induce macrophages RAW264.7 to express CD86, CD80, CD163 and CD206, but also promote them to produce TNF-α, IL-6, inducible nitric oxide synthase (iNOS), IL-10, Arginase (Arg)-1 and transforming growth factor-beta (TGF-β), suggesting their ability to polarize the macrophages into M1 phenotype and M2 phenotype simultaneously [83]. Interestingly, Zhu et al. found that exposure of CSE could promote the polarization of M2 macrophages (F4/80+CD206+ cells) with significantly increased expression of Arg-1 and IL-10 by investigating the oral mucosa epithelial tissue in OLK mouse model [84]. Taken together, in conjunction with the aforementioned increased expression of exosomal miR-21 in patients with OLK, we propose that exposure of CSE increases the secretion of miR-21+ exosomes, which promotes the polarization and expression of Arg-1 and IL-10 of M2 macrophages.
Another study reported by Fujita et al. have confirmed that the expression of miR-210 in human bronchial epithelial cells (HBECs)-derived exosomes induced by CS was significantly elevated, which contributed to myofibroblast differentiation by suppressing its target gene ATG7, a key composition of autophagy [85]. These results introduced the involvement of autophagy processes mediated by CS-induced exosomes that might open a novel path for us to better understand the pathological process of diseases [85]. Evidence showed that, under the CS stimulation, the autophagy of oral epithelial cells also changed. To be specific, the classical autophagy-related proteins LC3-II and Beclin-1 were upregulated in a concentration- and time-dependent manner [78]. As we mentioned above, the LC3-II was also overexpressed in the leukoplakia samples, especially in the upper layers of the epithelium, compared to the normal tissues [79]. Based on the data, we infer that the alteration of CSE-induced EVs might upregulate the autophagy of oral mucosal epithelial cells by upregulating LC3-II and Beclin-1, and ultimately contributes to the progression of OLK.
It has been found that the CS exposure of bronchial epithelial cells enhanced the encapsulation of matured miR-93 in EVs. These EVs-born miR-93 that were taken up by macrophages suppressed dual-specificity phosphatase 2 (DUSP2) and then activated the JNK pathway, which upregulated the expression of Matrix metalloproteinases (MMP)-9 and MMP-12 and leaded to elastin degradation [86]. The MMP-9 is one of the most complex MMPs and belongs to gelatinase family to degrade the extracellular matrix (ECM) components during tissue remodeling, which plays an essential role in tumor invasion and metastasis [87, 88]. The expression of MMP-9 has proven to be a reliable diagnostic and prognostic marker in oral cancer [8991]. Prior studies also have documented the significant elevation of MMP-9 in patients with OLK [9294]. In addition, the expression of MMP-9 increased higher in lesions with more severe dysplasia compared to those with lower degree of dysplasia, indicating their positive correlation with the disease progression.

The change of macrophages-derived EVs exposure to CS

CS was proven to exert different effects on the release of EVs from different types of macrophages. Specifically, exposure to CS contributes to the production of EVs from proinflammatory macrophages, but inhibits the secretion of EVs from anti-inflammatory macrophages. For example, the expression level of MMP-14 in EVs from proinflammatory macrophages are 3 times high after exposed to CS than non-exposed controls [95]. This increase could greatly affect the tissue remodeling and fibrogenesis due to the gelatinolytic and collagenolytic activity of MMP-14. Among the MMPs family, MMP-14 shows the broadest substrate specificity of all membrane-bound MMPs, especially to the composition of pericellular ECMs, which affects the cell–cell or cell–ECM communication [96, 97]. The effects are versatile including the promotion of matrix degradation and remodeling, angiogenesis, inflammation, tumor invasion and metastasis [96, 97]. Thus, the enrichment of MMP-14+ MVs might be a mediator that destroy tissue integrity and organs of individuals exposed to CS. It is reported that the suppressor of cytokine signaling (SOCS) 1 and SOCS3 in EVs taken up by epithelial cells downregulate the inflammatory cytokine signaling under homeostatic conditions [98]. However, after CS exposure, both SOCS1 and SOCS3 in EVs from anti-inflammatory macrophages are decreased, indicating the aggravation of inflammation [98].

The change of T cells-derived EVs exposure to CS

Apart from macrophages, it was reported that exposure of CS can also change the component of T cells-derived EVs and arouse the dysregulation of EVs functions. Donate et al. found that the Aryl hydrocarbon receptor (AhR) on Th17 cells were activated upon CS exposure, and then resulted in the increased expression of EVs-loaded miR-132 derived from Th17 cells. These CS-induced EVs had no obvious effect on the number and cytokine production (TNF, IL-6, IL-23) of dendritic cells, as well as the polarization of M0 macrophages, but they acted as a proinflammatory factor that induced osteoclastogenesis by inhibiting cyclooxygenase-2 (COX-2), revealing a mechanism of CS signaling and offering a potential target for therapeutic interference of inflammatory disease [99].

The change of monocytes-derived EVs exposure to CS

Interestingly, the human mononuclear cells-derived EVs stimulated by CSE also manifested a proinflammatory effect. Cordazzo et al. found a rapid generation of MPs by mononuclear cells upon exposure to CSE for 15 min on calcium-dependent mechanism. The MPs from CSE treated mononuclear cells showed a 2.6-fold, significant increase of TF activity indicating their elevated procoagulant potential [100]. Moreover, these MPs increased the expression of proinflammatory mediators including IL-18, intercellular adhesion molecule (ICAM-1), and monocyte chemoattractant protein (MCP)-1 of lung epithelial cells [100]. These data well-supplemented Li’ observations who identified that the enhanced production of MPs was induced by apoptosis of human monocytes/macrophages exposed to CSE [101].

Conclusions

The health of oral mucosal epithelial is prone to be damaged, because it is the first affected site by CS. Exposure to CS is identified as a major risk factor for OLK and its malignant transformation to OSCC. However, evidence done on the role of EVs in CS-induced OLK is still in its infancy. Based on the prior literature, upon CS stimulation, the biogenesis of EVs is significantly elevated, the composition of EVs and the signaling transduction via EVs are obviously changed, which is closely associated with the occurrence and progression of disease. Exposure to cigarette smoke contributes to the production of EVs from different types of cells including epithelial cells, macrophages, T cells, and monocytes. The bioactive cargo (proteins, cytokines, RNAs, and DNAs) of these EVs induced by CS stimulation are significantly changed, which acts as mediators for intra/inter-cellular communication. When these EVs are engulfed, they can change the biological behavior of the recipient/target cells and finally lead to the onset of OLK pathogenesis or even carcinogenesis (Fig. 3). Further researches should be performed to identify these assumptions. Elucidating the functions of CS-induced EVs in the pathophysiological processes of OLK is of great significance for the development of diagnostic biomarkers, prognostic evaluation, and therapeutic targets of OLK.

Declarations

Not applicable.

Competing interests

The authors declare that there is no competing interests.
Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://​creativecommons.​org/​licenses/​by/​4.​0/​. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Literatur
1.
Zurück zum Zitat Lugg ST, Scott A, Parekh D, Naidu B, Thickett DR. Cigarette smoke exposure and alveolar macrophages: mechanisms for lung disease. Thorax. 2022;77:94–101.PubMedCrossRef Lugg ST, Scott A, Parekh D, Naidu B, Thickett DR. Cigarette smoke exposure and alveolar macrophages: mechanisms for lung disease. Thorax. 2022;77:94–101.PubMedCrossRef
2.
Zurück zum Zitat Prieux R, Eeman M, Rothen-Rutishauser B, Valacchi G. Mimicking cigarette smoke exposure to assess cutaneous toxicity. Toxicol In Vitro. 2020;62: 104664.PubMedCrossRef Prieux R, Eeman M, Rothen-Rutishauser B, Valacchi G. Mimicking cigarette smoke exposure to assess cutaneous toxicity. Toxicol In Vitro. 2020;62: 104664.PubMedCrossRef
3.
Zurück zum Zitat Rezk-Hanna M, Benowitz NL. Cardiovascular effects of hookah smoking: potential implications for cardiovascular risk. Nicotine Tob Res. 2019;21:1151–61.PubMedCrossRef Rezk-Hanna M, Benowitz NL. Cardiovascular effects of hookah smoking: potential implications for cardiovascular risk. Nicotine Tob Res. 2019;21:1151–61.PubMedCrossRef
4.
Zurück zum Zitat Mravec B, Tibensky M, Horvathova L, Babal P. E-cigarettes and cancer risk. Cancer Prev Res. 2020;13:137–44.CrossRef Mravec B, Tibensky M, Horvathova L, Babal P. E-cigarettes and cancer risk. Cancer Prev Res. 2020;13:137–44.CrossRef
5.
Zurück zum Zitat Rohde P, Kahler CW, Lewinsohn PM, Brown RA. Psychiatric disorders, familial factors, and cigarette smoking: II. Associations with progression to daily smoking. Nicotine Tob Res. 2004;6:119–32.PubMedCrossRef Rohde P, Kahler CW, Lewinsohn PM, Brown RA. Psychiatric disorders, familial factors, and cigarette smoking: II. Associations with progression to daily smoking. Nicotine Tob Res. 2004;6:119–32.PubMedCrossRef
6.
Zurück zum Zitat Kerr DMI, Brooksbank KJM, Taylor RG, Pinel K, Rios FJ, Touyz RM, et al. Acute effects of electronic and tobacco cigarettes on vascular and respiratory function in healthy volunteers: a cross-over study. J Hypertens. 2019;37:154–66.PubMedCrossRef Kerr DMI, Brooksbank KJM, Taylor RG, Pinel K, Rios FJ, Touyz RM, et al. Acute effects of electronic and tobacco cigarettes on vascular and respiratory function in healthy volunteers: a cross-over study. J Hypertens. 2019;37:154–66.PubMedCrossRef
7.
Zurück zum Zitat Yang SR, Wright J, Bauter M, Seweryniak K, Kode A, Rahman I. Sirtuin regulates cigarette smoke-induced proinflammatory mediator release via RelA/p65 NF-kappaB in macrophages in vitro and in rat lungs in vivo: implications for chronic inflammation and aging. Am J Physiol Lung Cell Mol Physiol. 2007;292:L567–76.PubMedCrossRef Yang SR, Wright J, Bauter M, Seweryniak K, Kode A, Rahman I. Sirtuin regulates cigarette smoke-induced proinflammatory mediator release via RelA/p65 NF-kappaB in macrophages in vitro and in rat lungs in vivo: implications for chronic inflammation and aging. Am J Physiol Lung Cell Mol Physiol. 2007;292:L567–76.PubMedCrossRef
8.
Zurück zum Zitat Mossina A, Lukas C, Merl-Pham J, Uhl FE, Mutze K, Schamberger A, et al. Cigarette smoke alters the secretome of lung epithelial cells. Proteomics. 2017;17:1600243.CrossRef Mossina A, Lukas C, Merl-Pham J, Uhl FE, Mutze K, Schamberger A, et al. Cigarette smoke alters the secretome of lung epithelial cells. Proteomics. 2017;17:1600243.CrossRef
10.
Zurück zum Zitat Sepand MR, Maghsoudi AS, Shadboorestan A, Mirnia K, Aghsami M, Raoufi M. Cigarette smoke-induced toxicity consequences of intracellular iron dysregulation and ferroptosis. Life Sci. 2021;281: 119799.PubMedCrossRef Sepand MR, Maghsoudi AS, Shadboorestan A, Mirnia K, Aghsami M, Raoufi M. Cigarette smoke-induced toxicity consequences of intracellular iron dysregulation and ferroptosis. Life Sci. 2021;281: 119799.PubMedCrossRef
11.
Zurück zum Zitat Kono Y, Colley T, To M, Papaioannou AI, Mercado N, Baker JR, et al. Cigarette smoke-induced impairment of autophagy in macrophages increases galectin-8 and inflammation. Sci Rep. 2021;11:335.PubMedPubMedCentralCrossRef Kono Y, Colley T, To M, Papaioannou AI, Mercado N, Baker JR, et al. Cigarette smoke-induced impairment of autophagy in macrophages increases galectin-8 and inflammation. Sci Rep. 2021;11:335.PubMedPubMedCentralCrossRef
12.
Zurück zum Zitat Asthana S, Labani S, Kailash U, Sinha DN, Mehrotra R. Association of smokeless tobacco use and oral cancer: a systematic global review and meta-analysis. Nicotine Tob Res. 2019;21:1162–71.PubMedCrossRef Asthana S, Labani S, Kailash U, Sinha DN, Mehrotra R. Association of smokeless tobacco use and oral cancer: a systematic global review and meta-analysis. Nicotine Tob Res. 2019;21:1162–71.PubMedCrossRef
13.
Zurück zum Zitat Aredo JV, Luo SJ, Gardner RM, Sanyal N, Choi E, Hickey TP, et al. Tobacco smoking and risk of second primary lung cancer. J Thorac Oncol. 2021;16:968–79.PubMedPubMedCentralCrossRef Aredo JV, Luo SJ, Gardner RM, Sanyal N, Choi E, Hickey TP, et al. Tobacco smoking and risk of second primary lung cancer. J Thorac Oncol. 2021;16:968–79.PubMedPubMedCentralCrossRef
14.
16.
Zurück zum Zitat Olsson P, Bodewes ILA, Nilsson AM, Turesson C, Jacobsson LTH, Theander E, et al. Associations of cigarette smoking with disease phenotype and type I interferon expression in primary Sjogren’s syndrome. Rheumatol Int. 2019;39:1575–84.PubMedCrossRef Olsson P, Bodewes ILA, Nilsson AM, Turesson C, Jacobsson LTH, Theander E, et al. Associations of cigarette smoking with disease phenotype and type I interferon expression in primary Sjogren’s syndrome. Rheumatol Int. 2019;39:1575–84.PubMedCrossRef
17.
Zurück zum Zitat Leite FRM, Nascimento GG, Scheutz F, Lopez R. Effect of smoking on periodontitis: a systematic review and meta-regression. Am J Prev Med. 2018;54:831–41.PubMedCrossRef Leite FRM, Nascimento GG, Scheutz F, Lopez R. Effect of smoking on periodontitis: a systematic review and meta-regression. Am J Prev Med. 2018;54:831–41.PubMedCrossRef
18.
Zurück zum Zitat Kerr AR, Lodi G. Management of oral potentially malignant disorders. Oral Dis. 2021;27:2008–25.PubMedCrossRef Kerr AR, Lodi G. Management of oral potentially malignant disorders. Oral Dis. 2021;27:2008–25.PubMedCrossRef
19.
Zurück zum Zitat Warnakulasuriya S. Oral potentially malignant disorders: a comprehensive review on clinical aspects and management. Oral Oncol. 2020;102: 104550.PubMedCrossRef Warnakulasuriya S. Oral potentially malignant disorders: a comprehensive review on clinical aspects and management. Oral Oncol. 2020;102: 104550.PubMedCrossRef
20.
Zurück zum Zitat Carrard VC, van der Waal I. A clinical diagnosis of oral leukoplakia; a guide for dentists. Med Oral Patol Oral Cir Bucal. 2018;23:e59–64.PubMed Carrard VC, van der Waal I. A clinical diagnosis of oral leukoplakia; a guide for dentists. Med Oral Patol Oral Cir Bucal. 2018;23:e59–64.PubMed
21.
Zurück zum Zitat Lodi G, Franchini R, Warnakulasuriya S, Varoni EM, Sardella A, Kerr AR, et al. Interventions for treating oral leukoplakia to prevent oral cancer. Cochrane Database Syst Rev. 2016;7:Cd001829.PubMed Lodi G, Franchini R, Warnakulasuriya S, Varoni EM, Sardella A, Kerr AR, et al. Interventions for treating oral leukoplakia to prevent oral cancer. Cochrane Database Syst Rev. 2016;7:Cd001829.PubMed
22.
Zurück zum Zitat Villa A, Sonis S. Oral leukoplakia remains a challenging condition. Oral Dis. 2018;24:179–83.PubMedCrossRef Villa A, Sonis S. Oral leukoplakia remains a challenging condition. Oral Dis. 2018;24:179–83.PubMedCrossRef
23.
Zurück zum Zitat Aguirre-Urizar JM, Lafuente-Ibáñez de Mendoza I, Warnakulasuriya S. Malignant transformation of oral leukoplakia: systematic review and meta-analysis of the last 5 years. Oral Dis. 2021;27:1881–95.PubMedCrossRef Aguirre-Urizar JM, Lafuente-Ibáñez de Mendoza I, Warnakulasuriya S. Malignant transformation of oral leukoplakia: systematic review and meta-analysis of the last 5 years. Oral Dis. 2021;27:1881–95.PubMedCrossRef
24.
Zurück zum Zitat Kumar S, Muniyandi M. Tobacco use and oral leukoplakia: cross-sectional study among the Gond tribe in Madhya Pradesh. Asian Pac J Cancer Prev. 2015;16:1515–8.PubMedCrossRef Kumar S, Muniyandi M. Tobacco use and oral leukoplakia: cross-sectional study among the Gond tribe in Madhya Pradesh. Asian Pac J Cancer Prev. 2015;16:1515–8.PubMedCrossRef
25.
Zurück zum Zitat Bánóczy J, Gintner Z, Dombi C. Tobacco use and oral leukoplakia. J Dent Educ. 2001;65:322–7.PubMedCrossRef Bánóczy J, Gintner Z, Dombi C. Tobacco use and oral leukoplakia. J Dent Educ. 2001;65:322–7.PubMedCrossRef
26.
Zurück zum Zitat Dietrich T, Reichart PA, Scheifele C. Clinical risk factors of oral leukoplakia in a representative sample of the US population. Oral Oncol. 2004;40:158–63.PubMedCrossRef Dietrich T, Reichart PA, Scheifele C. Clinical risk factors of oral leukoplakia in a representative sample of the US population. Oral Oncol. 2004;40:158–63.PubMedCrossRef
27.
Zurück zum Zitat Ye P, Chen W, Huang F, Liu Q, Zhu YN, Wang X, et al. Smoking increases oral mucosa susceptibility to Candida albicans infection via the Nrf2 pathway: in vitro and animal studies. J Cell Mol Med. 2021;25:7948–60.PubMedPubMedCentralCrossRef Ye P, Chen W, Huang F, Liu Q, Zhu YN, Wang X, et al. Smoking increases oral mucosa susceptibility to Candida albicans infection via the Nrf2 pathway: in vitro and animal studies. J Cell Mol Med. 2021;25:7948–60.PubMedPubMedCentralCrossRef
28.
Zurück zum Zitat Sekar D. Extracellular vesicles are involved in oxidative stress and mitochondrial homeostasis in pulmonary arterial hypertension. Hypertens Res. 2021;44:1028–9.PubMedCrossRef Sekar D. Extracellular vesicles are involved in oxidative stress and mitochondrial homeostasis in pulmonary arterial hypertension. Hypertens Res. 2021;44:1028–9.PubMedCrossRef
29.
Zurück zum Zitat Chiaradia E, Tancini B, Emiliani C, Delo F, Pellegrino RM, Tognoloni A, et al. Extracellular vesicles under oxidative stress conditions: biological properties and physiological roles. Cells. 2021;10:1763.PubMedPubMedCentralCrossRef Chiaradia E, Tancini B, Emiliani C, Delo F, Pellegrino RM, Tognoloni A, et al. Extracellular vesicles under oxidative stress conditions: biological properties and physiological roles. Cells. 2021;10:1763.PubMedPubMedCentralCrossRef
30.
Zurück zum Zitat Ho J, Chaiswing L, St Clair DK. Extracellular vesicles and cancer therapy: insights into the role of oxidative stress. Antioxidants. 2022;11:1194.PubMedPubMedCentralCrossRef Ho J, Chaiswing L, St Clair DK. Extracellular vesicles and cancer therapy: insights into the role of oxidative stress. Antioxidants. 2022;11:1194.PubMedPubMedCentralCrossRef
32.
Zurück zum Zitat Cocucci E, Meldolesi J. Ectosomes and exosomes: shedding the confusion between extracellular vesicles. Trends Cell Biol. 2015;25:364–72.PubMedCrossRef Cocucci E, Meldolesi J. Ectosomes and exosomes: shedding the confusion between extracellular vesicles. Trends Cell Biol. 2015;25:364–72.PubMedCrossRef
34.
Zurück zum Zitat Zhu L, Sun HT, Wang S, Huang SL, Zheng Y, Wang CQ, et al. Isolation and characterization of exosomes for cancer research. J Hematol Oncol. 2020;13:152.PubMedPubMedCentralCrossRef Zhu L, Sun HT, Wang S, Huang SL, Zheng Y, Wang CQ, et al. Isolation and characterization of exosomes for cancer research. J Hematol Oncol. 2020;13:152.PubMedPubMedCentralCrossRef
35.
Zurück zum Zitat Pitt JM, Kroemer G, Zitvogel L. Extracellular vesicles: masters of intercellular communication and potential clinical interventions. J Clin Invest. 2016;126:1139–43.PubMedPubMedCentralCrossRef Pitt JM, Kroemer G, Zitvogel L. Extracellular vesicles: masters of intercellular communication and potential clinical interventions. J Clin Invest. 2016;126:1139–43.PubMedPubMedCentralCrossRef
36.
Zurück zum Zitat O’Brien K, Breyne K, Ughetto S, Laurent LC, Breakefield XO. RNA delivery by extracellular vesicles in mammalian cells and its applications. Nat Rev Mol Cell Biol. 2020;21:585–606.PubMedPubMedCentralCrossRef O’Brien K, Breyne K, Ughetto S, Laurent LC, Breakefield XO. RNA delivery by extracellular vesicles in mammalian cells and its applications. Nat Rev Mol Cell Biol. 2020;21:585–606.PubMedPubMedCentralCrossRef
37.
Zurück zum Zitat Macias M, Alegre E, Diaz-Lagares A, Patino A, Perez-Gracia JL, Sanmamed M, et al. Liquid biopsy: from basic research to clinical practice. Adv Clin Chem. 2018;83:73–119.PubMedCrossRef Macias M, Alegre E, Diaz-Lagares A, Patino A, Perez-Gracia JL, Sanmamed M, et al. Liquid biopsy: from basic research to clinical practice. Adv Clin Chem. 2018;83:73–119.PubMedCrossRef
38.
Zurück zum Zitat Yu W, Hurley J, Roberts D, Chakrabortty SK, Enderle D, Noerholm M, et al. Exosome-based liquid biopsies in cancer: opportunities and challenges. Ann Oncol. 2021;32:466–77.PubMedCrossRef Yu W, Hurley J, Roberts D, Chakrabortty SK, Enderle D, Noerholm M, et al. Exosome-based liquid biopsies in cancer: opportunities and challenges. Ann Oncol. 2021;32:466–77.PubMedCrossRef
40.
41.
Zurück zum Zitat Skog J, Wurdinger T, van Rijn S, Meijer DH, Gainche L, Sena-Esteves M, et al. Glioblastoma microvesicles transport RNA and proteins that promote tumour growth and provide diagnostic biomarkers. Nat Cell Biol. 2008;10:1470–6.PubMedPubMedCentralCrossRef Skog J, Wurdinger T, van Rijn S, Meijer DH, Gainche L, Sena-Esteves M, et al. Glioblastoma microvesicles transport RNA and proteins that promote tumour growth and provide diagnostic biomarkers. Nat Cell Biol. 2008;10:1470–6.PubMedPubMedCentralCrossRef
42.
Zurück zum Zitat Zhou B, Xu K, Zheng X, Chen T, Wang J, Song Y, et al. Application of exosomes as liquid biopsy in clinical diagnosis. Signal Transduct Target Ther. 2020;5:144.PubMedPubMedCentralCrossRef Zhou B, Xu K, Zheng X, Chen T, Wang J, Song Y, et al. Application of exosomes as liquid biopsy in clinical diagnosis. Signal Transduct Target Ther. 2020;5:144.PubMedPubMedCentralCrossRef
43.
Zurück zum Zitat Ma YS, Yang XL, Xin R, Liu JB, Fu D. Power and promise of exosomes as clinical biomarkers and therapeutic vectors for liquid biopsy and cancer control. Biochim et Biophys Acta Rev Cancer. 2021;1875: 188497.CrossRef Ma YS, Yang XL, Xin R, Liu JB, Fu D. Power and promise of exosomes as clinical biomarkers and therapeutic vectors for liquid biopsy and cancer control. Biochim et Biophys Acta Rev Cancer. 2021;1875: 188497.CrossRef
44.
Zurück zum Zitat Tkach M, Thery C. Communication by extracellular vesicles: where we are and where we need to go. Cell. 2016;164:1226–32.PubMedCrossRef Tkach M, Thery C. Communication by extracellular vesicles: where we are and where we need to go. Cell. 2016;164:1226–32.PubMedCrossRef
45.
Zurück zum Zitat Benedikter BJ, Weseler AR, Wouters EFM, Savelkoul PHM, Rohde GGU, Stassen FRM. Redox-dependent thiol modifications: implications for the release of extracellular vesicles. Cell Mol Life Sci. 2018;75:2321–37.PubMedPubMedCentralCrossRef Benedikter BJ, Weseler AR, Wouters EFM, Savelkoul PHM, Rohde GGU, Stassen FRM. Redox-dependent thiol modifications: implications for the release of extracellular vesicles. Cell Mol Life Sci. 2018;75:2321–37.PubMedPubMedCentralCrossRef
46.
Zurück zum Zitat Benedikter BJ, Koenen RR. Vaping, vapor, vesicles! Electronic cigarettes provoke vascular extracellular vesicle release in healthy volunteers. Atherosclerosis. 2020;301:79–81.PubMedCrossRef Benedikter BJ, Koenen RR. Vaping, vapor, vesicles! Electronic cigarettes provoke vascular extracellular vesicle release in healthy volunteers. Atherosclerosis. 2020;301:79–81.PubMedCrossRef
47.
Zurück zum Zitat Serban KA, Rezania S, Petrusca DN, Poirier C, Cao D, Justice MJ, et al. Structural and functional characterization of endothelial microparticles released by cigarette smoke. Sci Rep. 2016;6:31596.PubMedPubMedCentralCrossRef Serban KA, Rezania S, Petrusca DN, Poirier C, Cao D, Justice MJ, et al. Structural and functional characterization of endothelial microparticles released by cigarette smoke. Sci Rep. 2016;6:31596.PubMedPubMedCentralCrossRef
48.
Zurück zum Zitat Benedikter BJ, Volgers C, van Eijck PH, Wouters EFM, Savelkoul PHM, Reynaert NL, et al. Cigarette smoke extract induced exosome release is mediated by depletion of exofacial thiols and can be inhibited by thiol-antioxidants. Free Radic Biol Med. 2017;108:334–44.PubMedCrossRef Benedikter BJ, Volgers C, van Eijck PH, Wouters EFM, Savelkoul PHM, Reynaert NL, et al. Cigarette smoke extract induced exosome release is mediated by depletion of exofacial thiols and can be inhibited by thiol-antioxidants. Free Radic Biol Med. 2017;108:334–44.PubMedCrossRef
49.
Zurück zum Zitat Benedikter BJ, Bouwman FG, Heinzmann ACA, Vajen T, Mariman EC, Wouters EFM, et al. Proteomic analysis reveals procoagulant properties of cigarette smoke-induced extracellular vesicles. J Extracell Vesicles. 2019;8:1585163.PubMedPubMedCentralCrossRef Benedikter BJ, Bouwman FG, Heinzmann ACA, Vajen T, Mariman EC, Wouters EFM, et al. Proteomic analysis reveals procoagulant properties of cigarette smoke-induced extracellular vesicles. J Extracell Vesicles. 2019;8:1585163.PubMedPubMedCentralCrossRef
50.
Zurück zum Zitat Baek R, Varming K, Jorgensen MM. Does smoking, age or gender affect the protein phenotype of extracellular vesicles in plasma? Transfus Apher Sci. 2016;55:44–52.PubMedCrossRef Baek R, Varming K, Jorgensen MM. Does smoking, age or gender affect the protein phenotype of extracellular vesicles in plasma? Transfus Apher Sci. 2016;55:44–52.PubMedCrossRef
51.
Zurück zum Zitat Mobarrez F, Antoniewicz L, Bosson JA, Kuhl J, Pisetsky DS, Lundback M. The effects of smoking on levels of endothelial progenitor cells and microparticles in the blood of healthy volunteers. PLoS ONE. 2014;9: e90314.PubMedPubMedCentralCrossRef Mobarrez F, Antoniewicz L, Bosson JA, Kuhl J, Pisetsky DS, Lundback M. The effects of smoking on levels of endothelial progenitor cells and microparticles in the blood of healthy volunteers. PLoS ONE. 2014;9: e90314.PubMedPubMedCentralCrossRef
52.
Zurück zum Zitat Feller D, Kun J, Ruzsics I, Rapp J, Sarosi V, Kvell K, et al. Cigarette smoke-induced pulmonary inflammation becomes systemic by circulating extracellular vesicles containing wnt5a and inflammatory cytokines. Front Immunol. 2018;9:1724.PubMedPubMedCentralCrossRef Feller D, Kun J, Ruzsics I, Rapp J, Sarosi V, Kvell K, et al. Cigarette smoke-induced pulmonary inflammation becomes systemic by circulating extracellular vesicles containing wnt5a and inflammatory cytokines. Front Immunol. 2018;9:1724.PubMedPubMedCentralCrossRef
53.
Zurück zum Zitat Tanaka T, Narazaki M, Masuda K, Kishimoto T. Regulation of IL-6 in immunity and diseases. Adv Exp Med Biol. 2016;941:79–88.PubMedCrossRef Tanaka T, Narazaki M, Masuda K, Kishimoto T. Regulation of IL-6 in immunity and diseases. Adv Exp Med Biol. 2016;941:79–88.PubMedCrossRef
54.
Zurück zum Zitat Sharma M, Bairy I, Pai K, Satyamoorthy K, Prasad S, Berkovitz B, et al. Salivary IL-6 levels in oral leukoplakia with dysplasia and its clinical relevance to tobacco habits and periodontitis. Clin Oral Investig. 2011;15:705–14.PubMedCrossRef Sharma M, Bairy I, Pai K, Satyamoorthy K, Prasad S, Berkovitz B, et al. Salivary IL-6 levels in oral leukoplakia with dysplasia and its clinical relevance to tobacco habits and periodontitis. Clin Oral Investig. 2011;15:705–14.PubMedCrossRef
55.
Zurück zum Zitat Rhodus NL, Ho V, Miller CS, Myers S, Ondrey F. NF-kappaB dependent cytokine levels in saliva of patients with oral preneoplastic lesions and oral squamous cell carcinoma. Cancer Detect Prev. 2005;29:42–5.PubMedCrossRef Rhodus NL, Ho V, Miller CS, Myers S, Ondrey F. NF-kappaB dependent cytokine levels in saliva of patients with oral preneoplastic lesions and oral squamous cell carcinoma. Cancer Detect Prev. 2005;29:42–5.PubMedCrossRef
56.
Zurück zum Zitat Kumar A, Sinha N, Haque S, Kodidela S, Wang T, Martinez AG, et al. Nicotine self-administration with menthol and audiovisual cue facilitates differential packaging of CYP2A6 and cytokines/chemokines in rat plasma extracellular vesicles. Sci Rep. 2021;11:17393.PubMedPubMedCentralCrossRef Kumar A, Sinha N, Haque S, Kodidela S, Wang T, Martinez AG, et al. Nicotine self-administration with menthol and audiovisual cue facilitates differential packaging of CYP2A6 and cytokines/chemokines in rat plasma extracellular vesicles. Sci Rep. 2021;11:17393.PubMedPubMedCentralCrossRef
57.
Zurück zum Zitat Charrin S, le Naour F, Silvie O, Milhiet PE, Boucheix C, Rubinstein E. Lateral organization of membrane proteins: tetraspanins spin their web. Biochem J. 2009;420:133–54.PubMedCrossRef Charrin S, le Naour F, Silvie O, Milhiet PE, Boucheix C, Rubinstein E. Lateral organization of membrane proteins: tetraspanins spin their web. Biochem J. 2009;420:133–54.PubMedCrossRef
58.
60.
Zurück zum Zitat 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 Genom. 2020;13:128.CrossRef 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 Genom. 2020;13:128.CrossRef
61.
Zurück zum Zitat Sundar IK, Li D, Rahman I. Small RNA-sequence analysis of plasma-derived extracellular vesicle miRNAs in smokers and patients with chronic obstructive pulmonary disease as circulating biomarkers. J Extracell Vesicles. 2019;8:1684816.PubMedPubMedCentralCrossRef Sundar IK, Li D, Rahman I. Small RNA-sequence analysis of plasma-derived extracellular vesicle miRNAs in smokers and patients with chronic obstructive pulmonary disease as circulating biomarkers. J Extracell Vesicles. 2019;8:1684816.PubMedPubMedCentralCrossRef
62.
Zurück zum Zitat Xu H, Ling M, Xue J, Dai X, Sun Q, Chen C, et al. Exosomal microRNA-21 derived from bronchial epithelial cells is involved in aberrant epithelium-fibroblast cross-talk in COPD induced by cigarette smoking. Theranostics. 2018;8:5419–33.PubMedPubMedCentralCrossRef Xu H, Ling M, Xue J, Dai X, Sun Q, Chen C, et al. Exosomal microRNA-21 derived from bronchial epithelial cells is involved in aberrant epithelium-fibroblast cross-talk in COPD induced by cigarette smoking. Theranostics. 2018;8:5419–33.PubMedPubMedCentralCrossRef
63.
Zurück zum Zitat Kaunein N, Ramani RS, Koo K, Moore C, Celentano A, McCullough M, et al. A systematic review of microRNA signatures associated with the progression of leukoplakia with and without epithelial dysplasia. Biomolecules. 2021;11:1879.PubMedPubMedCentralCrossRef Kaunein N, Ramani RS, Koo K, Moore C, Celentano A, McCullough M, et al. A systematic review of microRNA signatures associated with the progression of leukoplakia with and without epithelial dysplasia. Biomolecules. 2021;11:1879.PubMedPubMedCentralCrossRef
64.
Zurück zum Zitat Chen H, Liu X, Jin Z, Gou C, Liang M, Cui L, et al. A three miRNAs signature for predicting the transformation of oral leukoplakia to oral squamous cell carcinoma. Am J Cancer Res. 2018;8:1403–13.PubMedPubMedCentral Chen H, Liu X, Jin Z, Gou C, Liang M, Cui L, et al. A three miRNAs signature for predicting the transformation of oral leukoplakia to oral squamous cell carcinoma. Am J Cancer Res. 2018;8:1403–13.PubMedPubMedCentral
65.
Zurück zum Zitat Roy R, Singh R, Chattopadhyay E, Ray A, Sarkar N, Aich R, et al. MicroRNA and target gene expression based clustering of oral cancer, precancer and normal tissues. Gene. 2016;593:58–63.PubMedCrossRef Roy R, Singh R, Chattopadhyay E, Ray A, Sarkar N, Aich R, et al. MicroRNA and target gene expression based clustering of oral cancer, precancer and normal tissues. Gene. 2016;593:58–63.PubMedCrossRef
66.
Zurück zum Zitat Brito JA, Gomes CC, Guimaraes AL, Campos K, Gomez RS. Relationship between microRNA expression levels and histopathological features of dysplasia in oral leukoplakia. J Oral Pathol Med. 2014;43:211–6.PubMedCrossRef Brito JA, Gomes CC, Guimaraes AL, Campos K, Gomez RS. Relationship between microRNA expression levels and histopathological features of dysplasia in oral leukoplakia. J Oral Pathol Med. 2014;43:211–6.PubMedCrossRef
67.
Zurück zum Zitat Niklander S, Guerra D, Contreras F, Gonzalez-Arriagada W, Marin C. MicroRNAs and their role in the malignant transformation of oral leukoplakia: a scoping review. Med Oral Patol Oral Cir Bucal. 2022;27:e77–84.PubMedCrossRef Niklander S, Guerra D, Contreras F, Gonzalez-Arriagada W, Marin C. MicroRNAs and their role in the malignant transformation of oral leukoplakia: a scoping review. Med Oral Patol Oral Cir Bucal. 2022;27:e77–84.PubMedCrossRef
68.
Zurück zum Zitat Grammatikakis I, Lal A. Significance of lncRNA abundance to function. Mamm Genome. 2022;33:271–80.PubMedCrossRef Grammatikakis I, Lal A. Significance of lncRNA abundance to function. Mamm Genome. 2022;33:271–80.PubMedCrossRef
69.
Zurück zum Zitat Ferrè F, Colantoni A, Helmer-Citterich M. Revealing protein-lncRNA interaction. Brief Bioinform. 2016;17:106–16.PubMedCrossRef Ferrè F, Colantoni A, Helmer-Citterich M. Revealing protein-lncRNA interaction. Brief Bioinform. 2016;17:106–16.PubMedCrossRef
70.
Zurück zum Zitat Rinn JL, Chang HY. Genome regulation by long noncoding RNAs. Annu Rev Biochem. 2012;81:145–66.PubMedCrossRef Rinn JL, Chang HY. Genome regulation by long noncoding RNAs. Annu Rev Biochem. 2012;81:145–66.PubMedCrossRef
72.
Zurück zum Zitat Sun Z, Yang S, Zhou Q, Wang G, Song J, Li Z, et al. Emerging role of exosome-derived long non-coding RNAs in tumor microenvironment. Mol Cancer. 2018;17:82.PubMedPubMedCentralCrossRef Sun Z, Yang S, Zhou Q, Wang G, Song J, Li Z, et al. Emerging role of exosome-derived long non-coding RNAs in tumor microenvironment. Mol Cancer. 2018;17:82.PubMedPubMedCentralCrossRef
73.
Zurück zum Zitat Dai J, Su Y, Zhong S, Cong L, Liu B, Yang J, et al. Exosomes: key players in cancer and potential therapeutic strategy. Signal Transduct Target Ther. 2020;5:145.PubMedPubMedCentralCrossRef Dai J, Su Y, Zhong S, Cong L, Liu B, Yang J, et al. Exosomes: key players in cancer and potential therapeutic strategy. Signal Transduct Target Ther. 2020;5:145.PubMedPubMedCentralCrossRef
74.
Zurück zum Zitat 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: e0243065.PubMedPubMedCentralCrossRef 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: e0243065.PubMedPubMedCentralCrossRef
75.
Zurück zum Zitat Humpton TJ, Alagesan B, DeNicola GM, Lu D, Yordanov GN, Leonhardt CS, et al. Oncogenic KRAS induces NIX-mediated mitophagy to promote pancreatic cancer. Cancer Discov. 2019;9:1268–87.PubMedPubMedCentralCrossRef Humpton TJ, Alagesan B, DeNicola GM, Lu D, Yordanov GN, Leonhardt CS, et al. Oncogenic KRAS induces NIX-mediated mitophagy to promote pancreatic cancer. Cancer Discov. 2019;9:1268–87.PubMedPubMedCentralCrossRef
76.
Zurück zum Zitat Li Y, Zheng W, Lu Y, Zheng Y, Pan L, Wu X, et al. BNIP3L/NIX-mediated mitophagy: molecular mechanisms and implications for human disease. Cell Death Dis. 2021;13:14.PubMedPubMedCentralCrossRef Li Y, Zheng W, Lu Y, Zheng Y, Pan L, Wu X, et al. BNIP3L/NIX-mediated mitophagy: molecular mechanisms and implications for human disease. Cell Death Dis. 2021;13:14.PubMedPubMedCentralCrossRef
77.
Zurück zum Zitat Zhang T, Liu Q, Gao W, Sehgal SA, Wu H. The multifaceted regulation of mitophagy by endogenous metabolites. Autophagy. 2022;18:1216–39.PubMedCrossRef Zhang T, Liu Q, Gao W, Sehgal SA, Wu H. The multifaceted regulation of mitophagy by endogenous metabolites. Autophagy. 2022;18:1216–39.PubMedCrossRef
78.
Zurück zum Zitat Liu Q, Zhao M, Chen W, Xu K, Huang F, Qu J, et al. Mainstream cigarette smoke induces autophagy and promotes apoptosis in oral mucosal epithelial cells. Arch Oral Biol. 2020;111: 104646.PubMedCrossRef Liu Q, Zhao M, Chen W, Xu K, Huang F, Qu J, et al. Mainstream cigarette smoke induces autophagy and promotes apoptosis in oral mucosal epithelial cells. Arch Oral Biol. 2020;111: 104646.PubMedCrossRef
79.
Zurück zum Zitat de Lima TB, Paz AHR, Rados PV, Leonardi R, Bufo P, Pedicillo MC, et al. Autophagy analysis in oral carcinogenesis. Pathol Res Pract. 2017;213:1072–7.PubMedCrossRef de Lima TB, Paz AHR, Rados PV, Leonardi R, Bufo P, Pedicillo MC, et al. Autophagy analysis in oral carcinogenesis. Pathol Res Pract. 2017;213:1072–7.PubMedCrossRef
80.
Zurück zum Zitat Liu W, Yao Y, Shi L, Tang G, Wu L. A novel lncRNA LOLA1 may predict malignant progression and promote migration, invasion, and EMT of oral leukoplakia via the AKT/GSK-3beta pathway. J Cell Biochem. 2021;122:1302–12.PubMedCrossRef Liu W, Yao Y, Shi L, Tang G, Wu L. A novel lncRNA LOLA1 may predict malignant progression and promote migration, invasion, and EMT of oral leukoplakia via the AKT/GSK-3beta pathway. J Cell Biochem. 2021;122:1302–12.PubMedCrossRef
81.
Zurück zum Zitat Shi L, Yang Y, Li M, Li C, Zhou Z, Tang G, et al. LncRNA IFITM4P promotes immune escape by up-regulating PD-L1 via dual mechanism in oral carcinogenesis. Mol Ther. 2022;30:1564–77.PubMedPubMedCentralCrossRef Shi L, Yang Y, Li M, Li C, Zhou Z, Tang G, et al. LncRNA IFITM4P promotes immune escape by up-regulating PD-L1 via dual mechanism in oral carcinogenesis. Mol Ther. 2022;30:1564–77.PubMedPubMedCentralCrossRef
82.
Zurück zum Zitat Corsello T, Kudlicki AS, Garofalo RP, Casola A. Cigarette smoke condensate exposure changes RNA content of extracellular vesicles released from small airway epithelial cells. Cells. 2019;8:1652.PubMedPubMedCentralCrossRef Corsello T, Kudlicki AS, Garofalo RP, Casola A. Cigarette smoke condensate exposure changes RNA content of extracellular vesicles released from small airway epithelial cells. Cells. 2019;8:1652.PubMedPubMedCentralCrossRef
83.
Zurück zum Zitat Chen Z, Wu H, Shi R, Fan W, Zhang J, Su W, et al. miRNAomics analysis reveals the promoting effects of cigarette smoke extract-treated Beas-2B-derived exosomes on macrophage polarization. Biochem Biophys Res Commun. 2021;572:157–63.PubMedCrossRef Chen Z, Wu H, Shi R, Fan W, Zhang J, Su W, et al. miRNAomics analysis reveals the promoting effects of cigarette smoke extract-treated Beas-2B-derived exosomes on macrophage polarization. Biochem Biophys Res Commun. 2021;572:157–63.PubMedCrossRef
84.
Zurück zum Zitat Zhu Y, Zhang S, Sun J, Wang T, Liu Q, Wu G, et al. Cigarette smoke promotes oral leukoplakia via regulating glutamine metabolism and M2 polarization of macrophage. Int J Oral Sci. 2021;13:25.PubMedPubMedCentralCrossRef Zhu Y, Zhang S, Sun J, Wang T, Liu Q, Wu G, et al. Cigarette smoke promotes oral leukoplakia via regulating glutamine metabolism and M2 polarization of macrophage. Int J Oral Sci. 2021;13:25.PubMedPubMedCentralCrossRef
85.
Zurück zum Zitat Fujita Y, Araya J, Ito S, Kobayashi K, Kosaka N, Yoshioka Y, et al. Suppression of autophagy by extracellular vesicles promotes myofibroblast differentiation in COPD pathogenesis. J Extracell Vesicles. 2015;4:28388.PubMedCrossRef Fujita Y, Araya J, Ito S, Kobayashi K, Kosaka N, Yoshioka Y, et al. Suppression of autophagy by extracellular vesicles promotes myofibroblast differentiation in COPD pathogenesis. J Extracell Vesicles. 2015;4:28388.PubMedCrossRef
86.
Zurück zum Zitat Xia H, Wu Y, Zhao J, Li W, Lu L, Ma H, et al. The aberrant cross-talk of epithelium-macrophages via METTL3-regulated extracellular vesicle miR-93 in smoking-induced emphysema. Cell Biol Toxicol. 2022;38:167–83.PubMedCrossRef Xia H, Wu Y, Zhao J, Li W, Lu L, Ma H, et al. The aberrant cross-talk of epithelium-macrophages via METTL3-regulated extracellular vesicle miR-93 in smoking-induced emphysema. Cell Biol Toxicol. 2022;38:167–83.PubMedCrossRef
87.
Zurück zum Zitat Mondal S, Adhikari N, Banerjee S, Amin SA, Jha T. Matrix metalloproteinase-9 (MMP-9) and its inhibitors in cancer: a minireview. Eur J Med Chem. 2020;194: 112260.PubMedCrossRef Mondal S, Adhikari N, Banerjee S, Amin SA, Jha T. Matrix metalloproteinase-9 (MMP-9) and its inhibitors in cancer: a minireview. Eur J Med Chem. 2020;194: 112260.PubMedCrossRef
89.
Zurück zum Zitat Shin YJ, Vu H, Lee JH, Kim HD. Diagnostic and prognostic ability of salivary MMP-9 for oral squamous cell carcinoma: a pre-/post-surgery case and matched control study. PLoS ONE. 2021;16: e0248167.PubMedPubMedCentralCrossRef Shin YJ, Vu H, Lee JH, Kim HD. Diagnostic and prognostic ability of salivary MMP-9 for oral squamous cell carcinoma: a pre-/post-surgery case and matched control study. PLoS ONE. 2021;16: e0248167.PubMedPubMedCentralCrossRef
90.
Zurück zum Zitat Nanda DP, Dutta K, Ganguly KK, Hajra S, Mandal SS, Biswas J, et al. MMP-9 as a potential biomarker for carcinoma of oral cavity: a study in eastern India. Neoplasma. 2014;61:747–57.PubMedCrossRef Nanda DP, Dutta K, Ganguly KK, Hajra S, Mandal SS, Biswas J, et al. MMP-9 as a potential biomarker for carcinoma of oral cavity: a study in eastern India. Neoplasma. 2014;61:747–57.PubMedCrossRef
91.
Zurück zum Zitat Enășescu DA, Moisescu MG, Imre M, Greabu M, Ripszky Totan A, Stanescu-Spinu I, et al. Lutein treatment effects on the redox status and metalloproteinase-9 (MMP-9) in oral cancer squamous cells-are there therapeutical hopes? Materials. 2021;14:2968.PubMedPubMedCentralCrossRef Enășescu DA, Moisescu MG, Imre M, Greabu M, Ripszky Totan A, Stanescu-Spinu I, et al. Lutein treatment effects on the redox status and metalloproteinase-9 (MMP-9) in oral cancer squamous cells-are there therapeutical hopes? Materials. 2021;14:2968.PubMedPubMedCentralCrossRef
92.
Zurück zum Zitat Chang PY, Kuo YB, Wu TL, Liao CT, Sun YC, Yen TC, et al. Association and prognostic value of serum inflammation markers in patients with leukoplakia and oral cavity cancer. Clin Chem Lab Med. 2013;51:1291–300.PubMedCrossRef Chang PY, Kuo YB, Wu TL, Liao CT, Sun YC, Yen TC, et al. Association and prognostic value of serum inflammation markers in patients with leukoplakia and oral cavity cancer. Clin Chem Lab Med. 2013;51:1291–300.PubMedCrossRef
93.
Zurück zum Zitat de Carvalho Fraga CA, Farias LC, de Oliveira MV, Domingos PL, Pereira CS, Silva TF, et al. Increased VEGFR2 and MMP9 protein levels are associated with epithelial dysplasia grading. Pathol Res Pract. 2014;210:959–64.PubMedCrossRef de Carvalho Fraga CA, Farias LC, de Oliveira MV, Domingos PL, Pereira CS, Silva TF, et al. Increased VEGFR2 and MMP9 protein levels are associated with epithelial dysplasia grading. Pathol Res Pract. 2014;210:959–64.PubMedCrossRef
94.
Zurück zum Zitat Smith J, Rattay T, McConkey C, Helliwell T, Mehanna H. Biomarkers in dysplasia of the oral cavity: a systematic review. Oral Oncol. 2009;45:647–53.PubMedCrossRef Smith J, Rattay T, McConkey C, Helliwell T, Mehanna H. Biomarkers in dysplasia of the oral cavity: a systematic review. Oral Oncol. 2009;45:647–53.PubMedCrossRef
95.
Zurück zum Zitat Li CJ, Liu Y, Chen Y, Yu D, Williams KJ, Liu ML. Novel proteolytic microvesicles released from human macrophages after exposure to tobacco smoke. Am J Pathol. 2013;182:1552–62.PubMedPubMedCentralCrossRef Li CJ, Liu Y, Chen Y, Yu D, Williams KJ, Liu ML. Novel proteolytic microvesicles released from human macrophages after exposure to tobacco smoke. Am J Pathol. 2013;182:1552–62.PubMedPubMedCentralCrossRef
96.
Zurück zum Zitat Gonzalez-Molina J, Gramolelli S, Liao Z, Carlson JW, Ojala PM, Lehti K. MMP14 in sarcoma: a regulator of tumor microenvironment communication in connective tissues. Cells. 2019;8:991.PubMedPubMedCentralCrossRef Gonzalez-Molina J, Gramolelli S, Liao Z, Carlson JW, Ojala PM, Lehti K. MMP14 in sarcoma: a regulator of tumor microenvironment communication in connective tissues. Cells. 2019;8:991.PubMedPubMedCentralCrossRef
97.
98.
Zurück zum Zitat Bourdonnay E, Zaslona Z, Penke LR, Speth JM, Schneider DJ, Przybranowski S, et al. Transcellular delivery of vesicular SOCS proteins from macrophages to epithelial cells blunts inflammatory signaling. J Exp Med. 2015;212:729–42.PubMedPubMedCentralCrossRef Bourdonnay E, Zaslona Z, Penke LR, Speth JM, Schneider DJ, Przybranowski S, et al. Transcellular delivery of vesicular SOCS proteins from macrophages to epithelial cells blunts inflammatory signaling. J Exp Med. 2015;212:729–42.PubMedPubMedCentralCrossRef
99.
Zurück zum Zitat Donate PB, Alves de Lima K, Peres RS, Almeida F, Fukada SY, Silva TA, et al. Cigarette smoke induces miR-132 in Th17 cells that enhance osteoclastogenesis in inflammatory arthritis. Proc Natl Acad Sci USA. 2021;118: e2017120118.PubMedCrossRef Donate PB, Alves de Lima K, Peres RS, Almeida F, Fukada SY, Silva TA, et al. Cigarette smoke induces miR-132 in Th17 cells that enhance osteoclastogenesis in inflammatory arthritis. Proc Natl Acad Sci USA. 2021;118: e2017120118.PubMedCrossRef
100.
Zurück zum Zitat Cordazzo C, Petrini S, Neri T, Lombardi S, Carmazzi Y, Pedrinelli R, et al. Rapid shedding of proinflammatory microparticles by human mononuclear cells exposed to cigarette smoke is dependent on Ca2+ mobilization. Inflamm Res. 2014;63:539–47.PubMedCrossRef Cordazzo C, Petrini S, Neri T, Lombardi S, Carmazzi Y, Pedrinelli R, et al. Rapid shedding of proinflammatory microparticles by human mononuclear cells exposed to cigarette smoke is dependent on Ca2+ mobilization. Inflamm Res. 2014;63:539–47.PubMedCrossRef
101.
Zurück zum Zitat Li M, Yu D, Williams KJ, Liu ML. Tobacco smoke induces the generation of procoagulant microvesicles from human monocytes/macrophages. Arterioscler Thromb Vasc Biol. 2010;30:1818–24.PubMedPubMedCentralCrossRef Li M, Yu D, Williams KJ, Liu ML. Tobacco smoke induces the generation of procoagulant microvesicles from human monocytes/macrophages. Arterioscler Thromb Vasc Biol. 2010;30:1818–24.PubMedPubMedCentralCrossRef
Metadaten
Titel
The potential roles of cigarette smoke-induced extracellular vesicles in oral leukoplakia
verfasst von
Qiao Peng
Ning Duan
Xiang Wang
Wenmei Wang
Publikationsdatum
01.12.2023
Verlag
BioMed Central
Erschienen in
European Journal of Medical Research / Ausgabe 1/2023
Elektronische ISSN: 2047-783X
DOI
https://doi.org/10.1186/s40001-023-01217-0

Weitere Artikel der Ausgabe 1/2023

European Journal of Medical Research 1/2023 Zur Ausgabe