Next Article in Journal / Special Issue
Endothelial to Mesenchymal Transition (EndoMT) in the Pathogenesis of Human Fibrotic Diseases
Previous Article in Journal / Special Issue
EMT Involved in Migration of Stem/Progenitor Cells for Pituitary Development and Regeneration
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Effect of Cigarette Smoking on Epithelial to Mesenchymal Transition (EMT) in Lung Cancer

Division of Hematology and Oncology, Department of Medicine, Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL 35233, USA
*
Author to whom correspondence should be addressed.
J. Clin. Med. 2016, 5(4), 44; https://doi.org/10.3390/jcm5040044
Submission received: 4 January 2016 / Revised: 4 April 2016 / Accepted: 6 April 2016 / Published: 11 April 2016
(This article belongs to the Special Issue Epithelial-Mesenchymal Transition)

Abstract

:
Epithelial to mesenchymal transition (EMT) is a process that allows an epithelial cell to acquire a mesenchymal phenotype through multiple biochemical changes resulting in an increased migratory capacity. During cancer progression, EMT is found to be associated with an invasive or metastatic phenotype. In this review, we focus on the discussion of recent studies about the regulation of EMT by cigarette smoking. Various groups of active compounds found in cigarette smoke such as polycyclic aromatic hydrocarbons (PAH), nicotine-derived nitrosamine ketone (NNK), and reactive oxygen specicies (ROS) can induce EMT through different signaling pathways. The links between EMT and biological responses to cigarette smoke, such as hypoxia, inflammation, and oxidative damages, are also discussed. The effect of cigarette smoke on EMT is not only limited to cancer types directly related to smoking, such as lung cancer, but has also been found in other types of cancer. Altogether, this review emphasizes the importance of understanding molecular mechanisms of the induction of EMT by cigarette smoking and will help in identifying novel small molecules for targeting EMT induced by smoking.

1. Introduction of EMT

Epithelial to mesenchymal transition (EMT) is a process through which epithelial cells undergo multiple biochemical changes to acquire mesenchymal phenotype and increase migratory capacity. Based on the physiological tissue context, EMT has been categorized to take place in three types of biological processes: (1) embryonic development and organ formation [1,2]; (2) wound healing and organ fibrosis [3]; and (3) cancer progression [4]. Type 3 EMT is found to be associated with an invasive or metastatic phenotype [5]. Defining morphological changes of EMT are the dissolution of the epithelial junctions, the disruption of the polarity complex, and the reorganization of the cytoskeletal architecture. At the molecular level, EMT is characterized by the downregulation of genes encoding for epithelial cell junction proteins (E-cadherin, claudins, and occludins) and the activation of genes, the protein products of which promote mesenchymal adhesion (vimentin, fibronectin and N-cadherin) [6]. Among the expression changes of epithelial and mesenchymal markers, the downregulation of E-cadherin is considered as a hallmark of EMT that leads to destabilization of adherens junctions [7].
Upregulation of EMT-related transcription factors and mesenchymal markers has been found in lung cancer and associated with increased rate of cancer recurrence and decreased survival in lung cancer patients (Table 1). Previous studies indicated that overexpression of EMT-related transcriptional factors, such as SNAIL1 [8,9,10], SLUG [11], TWIST [12,13,14], ZEB1 [15,16], FOXC2 [17], FOXQ1 [18], FOXC1 [19], and FOXM1 [20], are associated with invasiveness, metastasis and poor prognosis of lung cancer. Lung metastatic carcinomas show a significantly higher expression of these transcription factors than primary lung tumors, indicating their importance in the metastatic process. Loss or dysfunction of E-cadherin is associated with an invasive phenotype in lung cancer [21]. Furthermore, increased expression of mesenchymal proteins, such as N-Cadherin [22], Syndecan-1 [23], miR-21 [24], Vimentin [25], Periostin [26], and alpha-smooth muscle actin (α-SMA) [27], were also found in patients with poor prognosis.
The regulated expression and activity of several EMT-associated proteins have been shown to be involved in this process. There are three major groups of EMT-activating transcription factors: The SNAIL family of zinc-finger transcription factors SNAIL1/SNAIL2, the ZEB family of transcription factors ZEB1/ZEB2, and the TWIST family of bHLH transcription factors TWIST1/TWIST2. Due to distinct expression profiles, contributions of these transcription factors to EMT depend on the cell or tissue type and the signaling pathways that initiate EMT [6].
The SNAIL family of zinc-finger transcription factors, consisting of SNAIL1, SNAIL2, and SNAIL3 (also known as SNAIL, SLUG and SMUC respectively), activate EMT program during development, fibrosis and cancer [28]. The mechanism by which SNAIL proteins repress the expression of epithelial markers is represented by the activity of SNAIL1 on the promoter of E-cadherin. SNAIL1 suppresses expression of E-cadherin by binding to E-box DNA sequences and recruiting histone deacetylases (HDACs) [29], the Polycomb repressive complex 2 (PRC2) [30,31], Lys-specific demethylase 1 (LSD1) [32], G9a [33], and suppressor of variegation 3–9 homologue 1 (SUV39H1) [34]. The assembly leads to various histone modifications including methylation and acetylation at histone H3 Lys 4 (H3K4), H3K9 and H3K27, resulting in the suppression of E-cadherin promoter activity. In addition to repressing epithelial genes, SNAIL1 activates genes that contribute to the mesenchymal phenotype, such as fibronectin, N-cadherin, and collagen. SNAIL also induces EMT through upregulating other EMT transcription factors, such as SLUG [35], TWIST, ZEB2 [36], and ZEB1 [37]. Diverse signaling pathways cooperate in the initiation and progression of EMT by activating SNAIL1 expression. SNAIL1 expression has been known to be upregulated by Notch [38], TGF-β [38], and Wnt-β-catenin [39] signaling pathways.
The family of basic helix–loop–helix (bHLH) transcription factors plays essential roles in lineage commitment and differentiation. In this family, TWIST1 and TWIST2 have a key role in EMT regulation. TWIST1 downregulates epithelial gene expression and activates mesenchymal gene expression [40] independently of SNAIL and through association with other proteins. Importantly, TWIST1 recruits the methyltransferase SET8, which repress the E-cadherin and activate N-cadherin promoter via its H4K20 mono-methylation activity [41]. TWIST expression can be upregulated by multiple signaling pathways during tumorigenesis. EGF/EGFR signaling pathways upregulate TWIST expression in a manner dependent on STAT3 [42]. Under hypoxic conditions, the expression of TWIST can be induced by the transcription factor hypoxia-inducible factor 1α (HIF1α) [43]. In addition to expression regulation, MAPKs can regulate TWIST1 by phosphorylating TWIST1 at Ser68, stabilizing it from ubiquitin-mediated degradation and increasing its activity [44].
The ZEB family of transcription factors contains two members (ZEB1 and ZEB2), which bind regulatory gene sequences at E-boxes and function as transcriptional repressors and activators, thereby repressing some epithelial genes and activating mesenchymal genes. ZEBs mainly mediate transcriptional repression by recruiting C-terminal-binding protein (CTBP) co-repressor to E-boxes [45]. In addition to CTBP, ZEB1 can also recruit Switch/sucrose non-fermentable (SWI/SNF) chromatin remodeling protein BRG1 and repress E-cadherin expression [46]. ZEB1 can also interact with the transcriptional coactivator p300/CBP-associated factor (PCAF), which switches it from a transcriptional repressor to a transcriptional activator, promoting Smad signaling [47]. Additionally, ZEB1 can recruit the Lys-specific demethylase 1 (LSD1), possibly linking it to histone demethylation in EMT [48]. ZEB1 is directly regulated by SNAIL1. Moreover, TWIST1 cooperates with SNAIL1 in the induction of ZEB1 expression [49]. ZEB expression can be induced by TGF-β, WNT and MAPK signaling pathways.
In the past decade, an increasing number of studies have provided strong evidence for the critical role EMT plays in cancer progression and metastasis [50]. EMT allows tumor cells to adapt the constant changes of microenvironment in human tumors and successfully metastasize [6]. Theiry et al. proposed a metastasis model in which EMT is activated in primary epithelial tumor cells and allows tumor cells to spread through the body. Then, the tumors undergo mesenchymal to epithelial transition (MET) and form epithelial metastases through increasing epithelial markers including E-cadherin [51].

2. Effects of Cigarette Smoke on Cancer Development and Treatment Response

In 1950s, cigarette smoking was first found to be associated with lung cancer as people who smoked around 20 cigarettes a day had 26 times the lung cancer risk than non-smokers, and those who smoked three cigarettes a day had six times greater risk [52]. Since then, the association between cigarette smoke and lung cancer has been studied for more than six decades. Although cigarette smoke contains carcinogens, which convincingly cause lung tumor [53], the adverse effects of smoking are not limited to the lung cancer initiation in smokers. The effect of cigarette smoke on tumor growth and metastasis has been considered. Previous clinical studies reported the relationship between smoking history and cancer spread in lung cancer patients. Continued smoking was found to be associated with a significantly increased risk of mortality and development of a second primary tumor in early stage non-small cell lung cancer (NSCLC) [54]. Cancer patients who were previous or current smokers had an increased risk of metastatic disease at diagnosis [55]. Furthermore, the pro-metastatic effect of cigarette smoke is not found only at disease sites traditionally associated with cigarette smoking such as lung cancer but also at other type of cancer. Previous studies suggest a strong association between smoking behavior and pulmonary metastasis from breast [56], esophageal [57], oropharyngeal [58], and prostate [59] cancers.
Emerging data suggests that nicotine has negative effects on clinical outcome of cancer treatments. Smoking can reduce the efficacy of medical treatment and increase complications during and after surgery and adjunctive therapies [60,61]. Preclinical studies also suggest that nicotine may impair the therapeutic effects of chemotherapy and radiotherapy in vitro [62,63] and in a mouse model [64]. Furthermore, patients with limited-stage small-cell lung cancer who continue to smoke during chemotherapy and radiotherapy have poorer survival rates compared with those who do not [65].
However, the mechanism by which cigarette smoking confers drug resistance, reduced therapy efficacy and metastasis remains poorly understood. As EMT has been shown to be associated with drug resistance and metastasis, the link between cigarette smoke and EMT might hold the answer to the molecular network underlying adverse effects of cigarette smoke. It has been reported that there are higher levels of vimentin and other mesenchymal markers [66,67] and a decrease in the expression of E-cadherin in smokers compared with normal non-smokers. Several of the key pathways driving EMT are also aberrantly activated in cancer [68]. Previous in vitro studies also indicated that treatment with cigarette smoke extract (CSE) could promote EMT in lung cancer cells [69,70]. Importantly, EMT has been also found to be increased in patients with chronic obstructive pulmonary disease (COPD), which is mainly caused by smoking [67]. The coexistence of lung cancer and chronic obstructive pulmonary disease (COPD) is commonly detected in smokers, and the risk of developing lung cancer in smoking patients is significantly increased in the presence of COPD [71]. Although a detailed discussion of COPD is not within the scope of this review, it is important to emphasize that EMT can play an important role in the link between COPD and lung cancer diseases that are both induced by smoking. One important subsequent outcome with active EMT is the development of angiogenesis [68], a common feature of both lung cancer and COPD [72]. Recently, Fantozzi et al. showed that EMT could confer efficient tumorigenicity to breast cancer cells by elevated expression of the pro-angiogenic factor VEGF-A and by increased tumor angiogenesis [73]. Therefore, in this review, we will discuss current knowledge about the molecular mechanisms underlying EMT-inducing effects of cigarette smoke. This research area might provide potential therapeutic agents for targeting EMT in both lung cancer and COPD.

3. Molecular Mechanisms of Cigarette Smoke-Induced EMT

3.1. The EMT-Inducing Effect of Key Components of Cigarette Smoke

Cigarette smoke contains more than 4000 compounds overall and forty carcinogenic compounds classified into five major classes: Polycyclic aromatic hydrocarbons (PAH), nicotine-derived nitrosamines, aza-arenes, miscellaneous organic compounds, and inorganic compounds [74]. Among these compounds, nicotine, PAH, and nicotine-derived nitrosamine ketone (NNK) have been shown to induce EMT individually in cancer cells through different signaling pathways (Figure 1). The role of other classes of compounds in EMT remains unknown.

3.1.1. Nicotine

Although nicotine is not considered as a carcinogen, recent studies showed that nicotine can contribute to EMT and proliferation in lung cancer through the activation of nicotinic acetylcholine receptors (nAChRs) [75,76] which are expressed in a variety of non-neuronal cells including endothelial cells and several histological types of lung tissue [77]. The binding of nicotine to nAChRs causes the recruitment of β-arrestin and Src to the nicotinic receptors, which activate the MAPK pathway, resulting in induced cell proliferation [49]. Furthermore, nicotine was also found to induce invasion and migration of NSCLC cells through a7-nAChRs [50]. As a hallmark of pro-invasive effects, nicotine could induce changes in gene expression consistent with EMT, characterized by decreased expression of epithelial markers (E-cadherin, ZO-1) and concomitant increase in levels of mesenchymal proteins (vimentin, fibronectin) in both in vitro [50] and in vivo settings [78] of lung cancer. These results indicated that nicotine, through the nAChR signaling pathway, induces changes in gene expression pattern to facilitate EMT and tumor metastasis. In line with these studies, Pillai et al. emphasized the essential role of β-arrestin-1 in nicotine-induced EMT. β-arrestins function as scaffold proteins that recruit a broad spectrum of signaling molecules to membrane-bound receptors [79]. Stimulation of multiple NSCLC cell lines with nicotine led to enhanced recruitment of β-arrestin-1 and E2F1 on promoters and promote the expression of mesenchymal markers [80].
In addition to nAChRs, nicotine was also found to induce EMT through different oncogenic pathways. Nicotine can activate the Wnt3a signal pathway in lung cancer cells, leading to an upregulation of several mesenchymal markers such as vimentin, matrix metalloproteinases-9, and type I collagen as well as downregulation of E-cadherin [81]. This result is consistent with previous studies, which found that Wnt3a activates Wnt/β-catenin signaling and promotes EMT-like phenotypes in breast cancer [82]. Recently, periostin was identified as a novel target involved in nicotine-induced EMT. Periostin, also known as osteoblast-specific factor 2 (OSF-2), is an adhesion molecule that was initially identified in mouse osteoblastic cells as a secreted extracellular matrix protein. Overexpression of the periostin gene has been reported to cause EMT as well as cell migration, invasion and adhesion [83]. Periostin expression was found regulated by nicotine in NSCLC cells through nAChR-independent pathways [84].

3.1.2. Polycyclic Aromatic Hydrocarbons (PAH)

Role of major polycyclic aromatic hydrocarbons (PAH) in EMT induction was first noticed when Benzo[a]pyrene, a major PAH, was shown to induce expression of EMT-related genes including plasminogen activator inhibitor-1, fibronectin, TWIST, transforming growth factor-b2, basic fibroblast growth factor in A549 cells [85]. Furthermore, PAH can mediate EMT through the activation of aryl hydrocarbon receptor (AhR) [86]. AhR expression is upregulated in lung adenocarcinoma, suggesting that AhR and its expression might play an important role in the development of lung adenocarcinoma [87]. Prolonged AhR activation triggers a marked cytoskeleton remodeling associated with JNK activation and increased migration [88]. Upon activation by cigarette smoke, increased nuclear accumulation of AhR lead to transcriptional activation of SLUG, resulting in increased EMT.

3.1.3. Reactive Oxygen Species (ROS)

In addition to a majority of carcinogenic and mutagenic chemicals, tobacco smoke also contains stable and unstable free radicals and reactive oxygen species (ROS) in the particulate and the gas phase that can lead to oxidative damages [89]. Although the role of oxidative damage in cancer induced by cigarette smoke is unclear [90], it has been shown that various oxidants present in tobacco smoke such as peroxynitrite [91] and H2O2 [92] can activate Src, resulting in EMT initiation [93]. Src is the prototypic member of a family of non-receptor membrane associated tyrosine kinases, which are important regulators of a variety of cellular process including cytoskeletal organization, cell–cell contact, and cell–matrix adhesion. Activation of Src family kinases is involved in expression of mesenchymal proteins and other EMT events [94].

3.2. Cigarette Smoke Mediated Epigenetic Modifications and Oncogenic Pathways

In line with the studies on key compounds in cigarette smoke, a number of studies using cigarette smoke extract (CSE) have elucidated various signaling pathways and factors involved in the enhancing effect of cigarette smoke on EMT (Figure 2). Importantly, cigarette smoke can induce EMT in cancer cells through epigenetic modifications or through the activation of oncogenic pathways.

3.2.1. Epigenetic Regulatory Mechanisms

Our previous study showed that exposure of lung cancer cells to CSE induces EMT by downregulating E-cadherin at the transcriptional level. Our results reveal, for the first time, that smoking-mediated decrease in E-cadherin expression plays a key role in the induction in EMT in lung cancer [95]. Smoking-induced EMT is mediated through increased expression of SLUG. Furthermore, the recruitment of HDACs by SLUG is essential for E-cadherin suppression and EMT in cigarette smokers. Treatment with HDAC inhibitor MS-275 reverses CSE-induced EMT, migration, and invasion through the restoration of E-cadherin expression.
Chronic exposure of lung cancer cells to CSE leads to the activation of NF-κB, which is an important signaling pathway in inflammation. Enhancement in NF-κB activity increases the expression of TWIST-1 and downregulates miR-200c, a member of miR-200 family [69]. The miR-200 family (miR-200a, miR-200b, miR-200c, miR-141, and miR-429) has been considered as a novel repressor of EMT [96]. The miR-200 family members function as suppressors of EMT by targeting mRNAs encoding ZEB1 and ZEB2. Expression of the miR-200 family is lost in regions of metaplastic breast cancers lacking E-cadherin, whereas ZEB1 and ZEB2 are highly abundant in invasive mesenchymal cells. This study established a link between inflammatory response and CSE-induced EMT.
Recently, HOTAIR, a long noncoding RNA (lncRNA), was found to be upregulated by CSE and required for CSE-mediated EMT. CSE treatment induces secretion of IL-6, leading to the activation of STAT3, which binds to HOTAIR promoter regions and up-regulates HOTAIR in an autocrine manner [97]. Overexpression of HOTAIR was found in lung cancer tissue and linked to increased metastatic potential [98]. Enforced expression of HOTAIR in epithelial cancer cells induces genome-wide targeting of the Polycomb repressive complex 2, which alters histone H3 lysine 27 methylation, resulting in epigenetic silencing of metastasis suppressor genes [99].

3.2.2. Oncogenic Pathways

CSE-induced EMT was also found to be accompanied by increased expression of uPAR followed by the activation of AKT [100]. uPAR is a glycosylphosphatidylinositol (GPI)-anchored membrane protein that binds urokinase-type plasminogen activator (uPA) [101], which is involved in remodeling of the extracellular matrix, modulating cell adhesion and enhancing cell migration [102]. Increased uPAR expression has been implicated in the promotion of EMT in numerous cancers. uPAR gene silencing was sufficient to block CSE-induced EMT and the activation of AKT signaling [76].
Hypoxia is considered as a poor prognostic factor in non-small cell lung cancer [103]. Chronic exposure to carbon monoxide in cigarette smokers results in tissue hypoxia [104]. Previous studies showed that hypoxia induces an EMT phenotype in lung cancer and increases potential for migration, invasion, and metastasis [105,106]. It has been found that HIF-1 regulates the expression of TWIST by binding directly to the hypoxia-response element (HRE) in the TWIST proximal promoter [107]. CSE treatment induces hypoxic signaling in lung cancer cell lines as well as in mice lung tissue. The activation of HIF1α is important for the upregulation of mesenchymal marker expression and increased production of collagen in response to CSE [108].
CSE treatment can initiate EMT through increasing expression of Rac1 [109]. Rac1 is a member of an important small Rho GTPases family of proteins, which has a major role in cytoskeleton rearrangements and cancer cell metastasis [110]. The activation of Rac1 is critical for TWIST1-induced EMT [111]. Upregulation of Rac1 also leads to an increased TGF-β1 and activation of AKT signaling in pulmonary epithelial cells. Pharmacological inhibition or knockdown of Rac1 decreases CSE exposure induced TGF-β1 release and AKT activation, restraining CSE-induced changes in EMT-related markers [87].

4. Conclusion and Perspectives

The abnormal induction of EMT in cancer cells results in a variety of unfavorable biological outcomes including resistance to chemotherapy, enhanced stem cell properties, migration, invasion, and metastasis. In the past decade, it has been well accepted that EMT in normal and tumor epithelial cells has been controlled by complex transcription networks and various post-transcriptional modulators. In this review, we focus on the recent advances in understanding the role of cigarette smoke in EMT induction with emphasis on the following points.
(1)
In addition to carcinogenic effects, various compounds found in cigarette smoke such as PAH, nicotine, and ROS can induce EMT through different signaling pathways. The effects can be mediated through specific receptors such as nAChR and AhR for nicotine and PAH, respectively or through other molecular factors. Despite the diversity in signaling pathways and molecules involved in cigarette smoke-induced EMT, the effects are mainly associated with upregulation and enhanced activation of EMT-inducing transcription factors. As these regulators are also found to be associated with poor prognosis in lung cancer, it is essential to get further insight into the molecular network regulated by EMT-inducing cigarette smoke components.
(2)
The induction in EMT in cancer by cigarette smoke can be mediated by both changes in epigenetic regulation and activities of a variety of oncogenic signaling pathways. Importantly, several modifications are also linked to biological responses to cigarette smoke, such as inflammation, hypoxia and oxidative stress. Therefore, to effectively study the complex network underlying the effects of cigarette smoke on EMT, it is more applicable to use cigarette smoke extracts to mimic the environmental stresses caused by long-term cigarette smoking. Furthermore, by using CSE, scientists can study the crosstalk between different signaling pathways activated by concerted effects of individual cigarette components in picogram concentrations.
(3)
The effect of cigarette smoke on EMT has not only been found in lung cancer, the cancer type directly related to smoking, but also in other cancer types. This is consistent with previous observations that smoking also has adverse effects on health outcomes in patients with different types of cancer. However, it is not well established how cigarette smoking affects cancers other than lung cancer in vivo.
The potential of using inhibitors targeting cigarette smoke-induced EMT can be considered. Our previous study showed that using HDAC inhibitor MS-275 can reverse cigarette smoke-induced EMT, migration, and invasion through the restoration of E-cadherin expression [71]. Recently, Epigallocatechin-3-gallate (EGCG), the most abundant polyphenol found in green tea extract, has been reported to have negative effects on migration and epithelial to mesenchymal transition induced by nicotine in NSCLC cells. EGCG reverses the upregulation of HIF1α, vascular endothelial growth factor (VEGF), COX-2, p-AKT, p-ERK and vimentin protein levels, and the downregulation of p53 and beta-catenin protein levels mediated by nicotine in A549 cells. Additionally, a recent study also identified the extracellular-signal-regulated kinase 5 (ERK5) as a negative regulator for CSE-induced EMT [112]. ERK5 is a member of the mitogen-activated protein kinase (MAPK) family and has the Thr–Glu–Tyr (TEY) activation motif [113]. ERK5 is activated by growth factors and has an important role in the regulation of cell proliferation and cell differentiation. Several lines of evidence have suggested that activation of ERK5 suppresses EMT in cancer [114,115].
The persistent smoking after cancer diagnosis has been known to reduce the efficacy of medical treatment and increases complications. However, there are approximately 30 percent of cancer patients who smoked prior to their diagnosis continue to smoke [116]. Therefore, in parallel to placing more efforts for smoking cessation in cancer patients, it is essential to understand molecular mechanisms of the induction of EMT by cigarette smoke and identify novel targets for developing EMT-targeting strategies especially for smoking patients. Altogether, the observations discussed in this review describe a complicated network underlying cigarette smoke-induced EMT. The links between smoking and EMT, including the involvement of oncogenic pathways and epigenetic regulators, are important areas for future study.

Acknowledgments

We apologize to those colleagues whose work is not referenced because of space limitations and some of those are cited through reviews. This work was supported by R01 CA95195, Department of Veterans Affairs Merit Review Award, and a Faculty Development Award from UAB Comprehensive Cancer Center, P30 CA013148 (to P.K.D.).

Author Contributions

T.V. wrote the paper; L,J. helped revising the paper; P.K.D contributed to the writing and revising of the paper.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Lim, J.; Thiery, J.P. Epithelial to mesenchymal transitions: Insights from development. Development 2012, 139, 3471–3486. [Google Scholar] [PubMed]
  2. Shook, D.; Keller, R. Mechanisms, mechanics and function of epithelial to mesenchymal transitions in early development. Mech. Dev. 2003, 120, 1351–1383. [Google Scholar] [PubMed]
  3. Kalluri, R. EMT: When epithelial cells decide to become mesenchymal-like cells. J. Clin. Investig. 2009, 119, 1417–1419. [Google Scholar] [PubMed]
  4. Kang, Y.; Massague, J. Epithelial to mesenchymal transitions: Twist in development and metastasis. Cell 2004, 118, 277–279. [Google Scholar] [CrossRef] [PubMed]
  5. Singh, A.; Settleman, J. EMT, cancer stem cells and drug resistance: An emerging axis of evil in the war on cancer. Oncogene 2010, 29, 4741–4751. [Google Scholar] [PubMed]
  6. Lamouille, S.; Xu, J.; Derynck, R. Molecular mechanisms of epithelial to mesenchymal transition. Nat. Rev. Mol. Cell Biol. 2014, 15, 178–219. [Google Scholar] [CrossRef] [PubMed]
  7. Yilmaz, M.; Christofori, G. EMT, the cytoskeleton, and cancer cell invasion. Cancer Metastasis Rev. 2009, 28, 15–33. [Google Scholar] [CrossRef] [PubMed]
  8. Liu, C.W.; Li, C.H.; Peng, Y.J.; Cheng, Y.W.; Chen, H.W.; Liao, P.L.; Kang, J.J.; Yeng, M.H. SNAIL regulates Nanog status during the epithelial to mesenchymal transition via the Smad1/AKT/GSK3beta signaling pathway in non-small-cell lung cancer. Oncotarget 2014, 5, 3880–3894. [Google Scholar] [CrossRef] [PubMed]
  9. Merikallio, H.; Turpeenniemi-Hujanen, T.; Pääkkö, P.; Mäkitaro, R.; Riitta, K.; Salo, S.; Salo, T.; Harju, T.; Soini, Y. SNAIL promotes an invasive phenotype in lung carcinoma. Respir. Res. 2012, 13, 104–120. [Google Scholar] [CrossRef] [PubMed]
  10. Hung, J.J.; Yang, M.H.; Hsu, H.S.; Hsu, W.H.; Liu, J.S.; Wu, K.J. Prognostic significance of hypoxia-inducible factor-1alpha, TWIST1 and SNAIL expression in resectable non-small cell lung cancer. Thorax 2009, 64, 1082–1089. [Google Scholar] [CrossRef] [PubMed]
  11. Merikallio, H.; Pääkkö, P.; Mäkitaro, R.; Kaarteenaho, R.; Lehtonen, S.; Salo, S.; Salo, T.; Harju, T.; Soini, Y.H. SLUG is associated with poor survival in squamous cell carcinoma of the lung. Int. J. Clin. Exp. Pathol. 2014, 7, 5846. [Google Scholar] [PubMed]
  12. Wushou, A.; Hou, J.; Zhao, Y.J.; Shao, Z.M. Twist-1 up-regulation in carcinoma correlates to poor survival. Int. J. Mol. Sci. 2014, 15, 21621–21630. [Google Scholar] [CrossRef] [PubMed]
  13. Zeng, J.; Zhan, P.; Wu, G.; Yang, W.; Liang, W.; Lv, T.; Song, Y. Prognostic value of Twist in lung cancer: Systematic review and meta-analysis. Transl. Lung Cancer Res. 2015, 4, 236–241. [Google Scholar] [PubMed]
  14. Nakashima, H.; Hashimoto, N.; Aoyama, D.; Kohnoh, T.; Sakamoto, K.; Kusunose, M.; Imaizumi, K.; Takeyama, Y.; Sato, M.; Kawabe, T.; et al. Involvement of the transcription factor twist in phenotype alteration through epithelial to mesenchymal transition in lung cancer cells. Mol. Carcinog. 2012, 51, 400–410. [Google Scholar] [CrossRef] [PubMed]
  15. Merikallio, H.; Kaarteenaho, R.; Pääkkö, P.; Lehtonen, S.; Hirvikoski, P.; Mäkitaro, R.; Harju, T.; Soini, Y. Zeb1 and twist are more commonly expressed in metastatic than primary lung tumours and show inverse associations with claudins. J. Clin. Pathol. 2011, 64, 136–140. [Google Scholar] [CrossRef] [PubMed]
  16. Liu, L.; Chen, X.; Wang, Y.; Qu, Z.; Lu, Q.; Zhao, J.; Yan, X.; Zhang, H.; Zhou, Y. Notch3 is important for TGF-β-induced epithelial to mesenchymal transition in non-small cell lung cancer bone metastasis by regulating ZEB-1. Cancer Gene Ther. 2014, 21, 364–376. [Google Scholar] [CrossRef] [PubMed]
  17. Jiang, W.; Pang, X.G.; Wang, Q.; Shen, Y.X.; Chen, X.K.; Xi, J.J. Prognostic role of Twist, SLUG, and Foxc2 expression in stage I non-small-cell lung cancer after curative resection. Clin. Lung Cancer 2012, 13, 280–290. [Google Scholar] [CrossRef] [PubMed]
  18. Feng, J.; Zhang, X.; Zhu, H.; Wang, X.; Ni, S.; Huang, J. FoxQ1 overexpression influences poor prognosis in non-small cell lung cancer, associates with the phenomenon of EMT. PLoS ONE 2012, 7, 39937–39947. [Google Scholar] [CrossRef] [PubMed]
  19. Wei, L.X. High expression of FOXC1 is associated with poor clinical outcome in non-small cell lung cancer patients. Tumor Biol. 2013, 34, 941–950. [Google Scholar] [CrossRef] [PubMed]
  20. Xu, N. FoxM1 is associated with poor prognosis of non-small cell lung cancer patients through promoting tumor metastasis. PLoS ONE 2013, 8, 59412–59420. [Google Scholar] [CrossRef] [PubMed]
  21. Kase, S.; Sugio, K.; Yamazaki, K.; Okamoto, T.; Yano, T.; Sugimachi, K. Expression of E-cadherin and beta-catenin in human non-small cell lung cancer and the clinical significance. Clin. Cancer Res. 2000, 6, 4789–4798. [Google Scholar] [CrossRef]
  22. Zhang, X.; Liu, G.; Kang, Y.; Dong, Z.; Qian, Q.; Ma, X. N-cadherin expression is associated with acquisition of EMT phenotype and with enhanced invasion in erlotinib-resistant lung cancer cell lines. PLoS ONE 2013, 8, 57692–57700. [Google Scholar] [CrossRef] [PubMed]
  23. Anttonen, A.; Leppä, S.; Ruotsalainen, T.; Alfthan, H.; Mattson, K.; Joensuu, H. Pretreatment serum syndecan-1 levels and outcome in small cell lung cancer patients treated with platinum-based chemotherapy. Lung Cancer 2003, 41, 171–177. [Google Scholar] [CrossRef]
  24. Liu, X.G. High expression of serum miR-21 and tumor miR-200c associated with poor prognosis in patients with lung cancer. Med. Oncol. 2012, 29, 618–630. [Google Scholar] [CrossRef] [PubMed]
  25. Chen, D.; Zhang, Y.; Zhang, X.; Li, J.; Han, B.; Liu, S.; Wang, L.; Ling, Y.; Mao, S.; Wang, X. Overexpression of integrin-linked kinase correlates with malignant phenotype in non-small cell lung cancer and promotes lung cancer cell invasion and migration via regulating epithelial-mesenchymal transition (EMT)—Related genes. Acta Histochem. 2013, 115, 128–238. [Google Scholar] [CrossRef] [PubMed]
  26. Soltermann, A.; Tischler, V.; Arbogast, S.; Braun, J.; Probst-Hensch, N.; Weder, W.; Moch, H.; Kristiansen, G. Prognostic significance of epithelial-mesenchymal and mesenchymal-epithelial transition protein expression in non-small cell lung cancer. Clin. Cancer Res. 2008, 14, 7430. [Google Scholar] [CrossRef] [PubMed]
  27. Chen, Y.; Zou, L.; Zhang, Y.; Chen, Y.; Xing, P.; Yang, W.; Li, F.; Ji, X.; Liu, F.; Lu, X. Transforming growth factor-β1 and α-smooth muscle actin in stromal fibroblasts are associated with a poor prognosis in patients with clinical stage I–IIIA nonsmall cell lung cancer after curative resection. Tumor Biol. 2014, 35, 6707–6720. [Google Scholar] [CrossRef] [PubMed]
  28. Barrallo-Gimeno, A.; Nieto, M.A. The SNAIL genes as inducers of cell movement and survival: Implications in development and cancer. Development 2005, 132, 3151–3161. [Google Scholar] [CrossRef] [PubMed]
  29. Peinado, H.; Ballestar, E.; Esteller, M.; Cano, A. SNAIL mediates E-cadherin repression by the recruitment of the Sin3A/histone deacetylase 1 (HDAC1)/HDAC2 complex. Mol. Cell. Biol. 2004, 24, 306–319. [Google Scholar] [CrossRef] [PubMed]
  30. Herranz, N.; Pasini, D.; Díaz, V.M.; Francí, C.; Gutierrez, A.; Dave, N.; Escrivà, M.; Hernandez-Muñoz, I.; Di Croce, L.; Helin, K.; et al. Polycomb complex 2 is required for E-cadherin repression by the SNAIL1 transcription factor. Mol. Cell. Biol. 2008, 28, 4772–4781. [Google Scholar] [CrossRef] [PubMed]
  31. Tong, Z.T.; Cai, M.Y.; Wang, X.G.; Kong, L.L.; Mai, S.J.; Liu, Y.H.; Zhang, H.B.; Liao, Y.J.; Zheng, F.; Zhu, W.; et al. EZH2 supports nasopharyngeal carcinoma cell aggressiveness by forming a co-repressor complex with HDAC1/HDAC2 and SNAIL to inhibit E-cadherin. Oncogene 2012, 31, 583–594. [Google Scholar] [CrossRef] [PubMed]
  32. Lin, T.; Ponn, A.; Hu, X.; Law, B.K.; Lu, J. Requirement of the histone demethylase LSD1 in Snai1-mediated transcriptional repression during epithelial to mesenchymal transition. Oncogene 2010, 29, 4896–4904. [Google Scholar] [CrossRef] [PubMed]
  33. Dong, C.; Wu, Y.; Yao, J.; Wang, Y.; Yu, Y.; Rychahou, P.G.; Evers, B.M.; Zhou, B.P. G9a interacts with SNAIL and is critical for SNAIL-mediated E-cadherin repression in human breast cancer. J. Clin. Investig. 2012, 122, 1469–1486. [Google Scholar] [CrossRef] [PubMed]
  34. Dong, C.; Wu, Y.; Wang, Y.; Wang, C.; Kang, T.; Rychahou, P.G.; Chi, Y.I.; Evers, B.M.; Zhou, B.P. Interaction with Suv39H1 is critical for SNAIL-mediated E-cadherin repression in breast cancer. Oncogene 2013, 32, 1351–1362. [Google Scholar] [CrossRef] [PubMed]
  35. Thuault, S.; Tan, E.J.; Peinado, H.; Cano, A.; Heldin, C.H.; Moustakas, A. HMGA2 and Smads coregulate SNAIL1 expression during induction of epithelial toto-mesenchymal transition. J. Biol. Chem. 2008, 283, 33437–33446. [Google Scholar] [CrossRef] [PubMed]
  36. Smit, M.A.; Geiger, T.R.; Song, J.Y.; Gitelman, I.; Peeper, D.S. A Twist-SNAIL axis critical for TrkB-induced epithelial to mesenchymal transition-like transformation, anoikis resistance, and metastasis. Mol. Cell. Biol. 2009, 29, 3722–3737. [Google Scholar] [CrossRef] [PubMed]
  37. Guaita, S.; Puig, I.; Franci, C.; Garrido, M.; Dominguez, D.; Batlle, E.; Sancho, E.; Dedhar, S.; De Herreros, A.G.; Baulida, J. SNAIL induction of epithelial to mesenchymal transition in tumor cells is accompanied by MUC1 repression and ZEB1 expression. J. Biol. Chem. 2002, 277, 39209–39220. [Google Scholar] [CrossRef] [PubMed]
  38. Saad, S.; Stanners, S.R.; Yong, R.; Tang, O.; Pollock, C.A. Notch mediated epithelial to mesenchymal transformation is associated with increased expression of the SNAIL transcription factor. Int. J. Biochem. Cell Biol. 2010, 42, 1115–1122. [Google Scholar] [CrossRef] [PubMed]
  39. Yang, X.; Li, L.; Huang, Q.; Xu, W.; Cai, X.; Zhang, J.; Yan, W.; Song, D.; Liu, T.; Zhou, W.; et al. Wnt signaling through SNAIL1 and Zeb1 regulates bone metastasis in lung cancer. Am. J. Cancer Res. 2015, 5, 748–755. [Google Scholar] [PubMed]
  40. Yang, J.; Mani, S.A.; Donaher, J.L.; Ramaswamy, S.; Itzykson, R.A.; Come, C.; Savagner, P.; Gitelman, I.; Richardson, A.; Weinberg, R.A. Twist, a master regulator of morphogenesis, plays an essential role in tumor metastasis. Cell 2004, 117, 927–939. [Google Scholar] [CrossRef] [PubMed]
  41. Yang, F.; Sun, L.; Li, Q.; Han, X.; Lei, L.; Zhang, H.; Shang, Y. SET8 promotes epithelial to mesenchymal transition and confers TWIST dual transcriptional activities. EMBO J. 2012, 31, 110–123. [Google Scholar] [CrossRef] [PubMed]
  42. Lo, H.W.; Hsu, S.C.; Xia, W.; Cao, X.; Shih, J.Y.; Wei, Y.; Abbruzzese, J.L.; Hortobagyi, G.N.; Hung, M.C. Epidermal growth factor receptor cooperates with signal transducer and activator of transcription 3 to induce epithelial to mesenchymal transition in cancer cells via up-regulation of TWIST gene expression. Cancer Res. 2007, 67, 9066–9076. [Google Scholar] [CrossRef] [PubMed]
  43. Yang, M.H.; Wu, K.J. TWIST activation by hypoxia inducible factor-1 (HIF-1): Implications in metastasis and development. Cell Cycle 2008, 7, 2090–2096. [Google Scholar] [CrossRef] [PubMed]
  44. Hong, J.; Zhou, J.; Fu, J.; He, T.; Qin, J.; Wang, L.; Liao, L.; Xu, J. Phosphorylation of serine 68 of Twist1 by MAPKs stabilizes Twist1 protein and promotes breast cancer cell invasiveness. Cancer Res. 2011, 71, 3980–3990. [Google Scholar] [CrossRef] [PubMed]
  45. Postigo, A.A.; Dean, D.C. ZEB represses transcription through interaction with the corepressor CtBP. Proc. Natl. Acad. Sci. USA 1999, 96, 6683–6699. [Google Scholar] [CrossRef] [PubMed]
  46. Sánchez-Tilló, E.E. ZEB1 represses E-cadherin and induces an EMT by recruiting the SWI/SNF chromatin-remodeling protein BRG1. Oncogene 2010, 29, 3490–3510. [Google Scholar] [CrossRef] [PubMed]
  47. Postigo, A.A.; Depp, J.L.; Taylor, J.J.; Kroll, K.L. Regulation of Smad signaling through a differential recruitment of coactivators and corepressors by ZEB proteins. EMBO J. 2003, 22, 2453–2463. [Google Scholar] [CrossRef] [PubMed]
  48. Wang, J.; Scully, K.; Zhu, X.; Cai, L.; Zhang, J.; Prefontaine, G.G.; Krones, A.; Ohgi, K.A.; Zhu, P.; Garcia-Bassets, I.; et al. Opposing LSD1 complexes function in developmental gene activation and repression programmes. Nature 2007, 446, 882–890. [Google Scholar] [CrossRef] [PubMed]
  49. Dave, N.; Guaita-Esteruelas, S.; Gutarra, S.; Frias, À.; Beltran, M.; Peiró, S.; de Herreros, A.G. Functional cooperation between SNAIL1 and twist in the regulation of ZEB1 expression during epithelial to mesenchymal transition. J. Biol. Chem. 2011, 286, 12024–12034. [Google Scholar] [CrossRef] [PubMed]
  50. Tsai, J.H.; Yang, J. Epithelial to mesenchymal plasticity in carcinoma metastasis. Genes Dev. 2013, 27, 2192–2200. [Google Scholar] [CrossRef] [PubMed]
  51. Thiery, J.P. Epithelial–mesenchymal transitions in tumour progression. Nat. Rev. Cancer 2002, 2, 442–454. [Google Scholar] [CrossRef] [PubMed]
  52. Doll, R.; Hill, A.B. Smoking and carcinoma of the lung: Preliminary report. Br. Med. J. 1950, 2, 739–748. [Google Scholar] [CrossRef] [PubMed]
  53. Hecht, S.S.; Kassie, F.; Hatsukami, D.K. Chemoprevention of lung carcinogenesis in addicted smokers and ex-smokers. Nat. Rev. Cancer 2009, 9, 476–488. [Google Scholar] [CrossRef] [PubMed]
  54. Parsons, A.; Daley, A.; Begh, R.; Aveyard, P. Influence of smoking cessation after diagnosis of early stage lung cancer on prognosis: Systematic review of observational studies with meta-analysis. BMJ 2010, 340, 5569. [Google Scholar] [CrossRef] [PubMed]
  55. Kobrinsky, N.L.; Klug, M.G.; Hokanson, P.J.; Sjolander, D.E.; Burd, L. Impact of smoking on cancer stage at diagnosis. J. Clin. Oncol. 2003, 21, 907–913. [Google Scholar] [CrossRef] [PubMed]
  56. Murin, S.; Inciardi, J. Cigarette smoking and the risk of pulmonary metastasis from breast cancer. Chest 2001, 119, 1635–1640. [Google Scholar] [CrossRef] [PubMed]
  57. Abrams, J.A.; Lee, P.C.; Port, J.L.; Altorki, N.K.; Neugut, A.I. Cigarette smoking and risk of lung metastasis from esophageal cancer. Cancer Epidemiol. Biomark. Prev. 2008, 17, 2707–2713. [Google Scholar] [CrossRef] [PubMed]
  58. McBride, S.M.; Ali, N.N.; Margalit, D.N.; Chan, A.W. Active tobacco smoking and distant metastasis in patients with oropharyngeal cancer. Int. J. Radiat. Oncol. Biol. Phys. 2012, 84, 183–188. [Google Scholar] [CrossRef] [PubMed]
  59. Moreira, D.M.; Aronson, W.J.; Terris, M.K.; Kane, C.J.; Amling, C.L.; Cooperberg, M.R.; Boffetta, P.; Freedland, S.J. Cigarette smoking is associated with an increased risk of biochemical disease recurrence, metastasis, castration-resistant prostate cancer, and mortality after radical prostatectomy: Results from the SEARCH database. Cancer 2014, 120, 197–204. [Google Scholar] [CrossRef] [PubMed]
  60. Fujisawa, T.; Iizasa, T.; Saitoh, Y.; Sekine, Y.; Motohashi, S.; Yasukawa, T.; Shibuya, K.; Hiroshima, K.; Ohwada, H. Smoking before surgery predicts poor long-term survival in patients with stage I non-small-cell lung carcinomas. J. Clin. Oncol. 1999, 17, 2086–2091. [Google Scholar] [PubMed]
  61. Fox, J.L.; Rosenzweig, K.E.; Ostroff, J.S. The effect of smoking status on survival following radiation therapy for non-small cell lung cancer. Lung Cancer 2004, 44, 287–293. [Google Scholar] [CrossRef] [PubMed]
  62. Zhang, J.; Kamdar, O.; Le, W.; Rosen, G.D.; Upadhyay, D. Nicotine induces resistance to chemotherapy by modulating mitochondrial signaling in lung cancer. Am. J. Respir. Cell Mol. Biol. 2009, 40, 135–146. [Google Scholar] [CrossRef] [PubMed]
  63. Shen, T.; Le, W.; Yee, A.; Kamdar, O.; Hwang, P.H.; Upadhyay, D. Nicotine induces resistance to chemotherapy in nasal epithelial cancer. Am. J. Rhinol. Allergy 2010, 24, 73–77. [Google Scholar] [CrossRef] [PubMed]
  64. Warren, G.W.; Singh, A.K. Nicotine and lung cancer. J. Carcinog. 2013, 12, 135–146. [Google Scholar] [CrossRef] [PubMed]
  65. Videtic, G.M.; Stitt, L.W.; Dar, A.R.; Kocha, W.I.; Tomiak, A.T.; Truong, P.T.; Vincent, M.D.; Yu, E.W. Continued cigarette smoking by patients receiving concurrent chemoradiotherapy for limited-stage small-cell lung cancer is associated with decreased survival. J. Clin. Oncol. 2003, 21, 1544–1549. [Google Scholar] [CrossRef] [PubMed]
  66. Sohal, S.S.; Reid, D.; Soltani, A.; Ward, C.; Weston, S.; Muller, H.K.; Wood-Baker, R.; Walters, E.H. Reticular basement membrane fragmentation and potential epithelial mesenchymal transition is exaggerated in the airways of smokers with chronic obstructive pulmonary disease. Respirology 2010, 15, 930–938. [Google Scholar] [CrossRef] [PubMed]
  67. Milara, J.; Peiró, T.; Serrano, A.; Cortijo, J. Epithelial to mesenchymal transition is increased in patients with COPD and induced by cigarette smoke. Thorax 2013, 68, 410. [Google Scholar] [CrossRef] [PubMed]
  68. Sohal, S.S.; Mahmood, M.Q.; Walters, E.H. Clinical significance of epithelial mesenchymal transition (EMT) in chronic obstructive pulmonary disease (COPD): Potential target for prevention of airway fibrosis and lung cancer. Clin. Transl. Med. 2014, 3, 33. [Google Scholar] [CrossRef] [PubMed]
  69. Zhao, Y.; Xu, Y.; Li, Y.; Xu, W.; Luo, F.; Wang, B.; Pang, Y.; Xiang, Q.; Zhou, J.; Wang, X.; et al. NF-kappaB-mediated inflammation leading to EMT via miR-200c is involved in cell transformation induced by cigarette smoke extract. Toxicol. Sci. 2013, 135, 265–276. [Google Scholar] [CrossRef] [PubMed]
  70. Wang, J.; Chen, Y.; Lin, C.; Jia, J.; Tian, L.; Yang, K.; Zhao, L.; Lai, N.; Jiang, Q.; Sun, Y.; et al. Effects of chronic exposure to cigarette smoke on canonical transient receptor potential expression in rat pulmonary arterial smooth muscle. Am. J. Physiol. Cell Physiol. 2014, 306, 364–373. [Google Scholar] [CrossRef] [PubMed]
  71. Sohal, S.S.; Walters, E.H. Role of epithelial mesenchymal transition (EMT) in chronic obstructive pulmonary disease (COPD). Respir. Res. 2013, 14, 120. [Google Scholar] [CrossRef] [PubMed]
  72. Siafakas, N.M.; Antoniou, K.M.; Tzortzaki, E.G. Role of angiogenesis and vascular remodeling in chronic obstructive pulmonary disease. Int. J. Chron. Obstr. Pulm. Dis. 2007, 2, 453–462. [Google Scholar]
  73. Fantozzi, A.; Gruber, D.C.; Pisarsky, L.; Heck, C.; Kunita, A.; Yilmaz, M.; Meyer-Schaller, N.; Cornille, K.; Hopfer, U.; Bentires-Alj, M.; et al. VEGF-mediated angiogenesis links EMT-induced cancer stemness to tumor initiation. Cancer Res. 2014, 74, 1566–1575. [Google Scholar] [CrossRef] [PubMed]
  74. Fowles, J.; Dybing, E. Application of toxicological risk assessment principles to the chemical constituents of cigarette smoke. Tob. Control 2003, 12, 424–430. [Google Scholar] [CrossRef] [PubMed]
  75. Dasgupta, P.; Rizwani, W.; Pillai, S.; Kinkade, R.; Kovacs, M.; Rastogi, S.; Banerjee, S.; Carless, M.; Kim, E.; Coppola, D.; et al. Nicotine induces cell proliferation, invasion and epithelial to mesenchymal transition in a variety of human cancer cell lines. Int. J. Cancer 2009, 124, 36–45. [Google Scholar] [CrossRef] [PubMed]
  76. Dasgupta, P.; Rastogi, S.; Pillai, S.; Ordonez-Ercan, D.; Morris, M.; Haura, E.; Chellappan, S. Nicotine induces cell proliferation by beta-arrestin-mediated activation of Src and Rb-Raf-1 pathways. J. Clin. Investig. 2006, 116, 2208–2217. [Google Scholar] [CrossRef] [PubMed]
  77. Schuller, H.M.; Plummer, H.K., 3rd; Jull, B.A. Receptor-mediated effects of nicotine and its nitrosated derivative NNK on pulmonary neuroendocrine cells. Anat. Rec. A Discov. Mol. Cell. Evol. Biol. 2003, 270, 51–58. [Google Scholar] [CrossRef] [PubMed]
  78. Davis, R.; Rizwani, W.; Banerjee, S.; Kovacs, M.; Haura, E.; Coppola, D.; Chellappan, S. Nicotine promotes tumor growth and metastasis in mouse models of lung cancer. PLoS ONE 2009, 4, 7524. [Google Scholar] [CrossRef] [PubMed]
  79. Kommaddi, R.P.; Shenoy, S.K. Arrestins and protein ubiquitination. Prog. Mol. Biol. Transl. Sci. 2013, 118, 175–204. [Google Scholar] [PubMed]
  80. Pillai, S.; Trevino, J.; Rawal, B.; Singh, S.; Kovacs, M.; Li, X.; Schell, M.; Haura, E.; Bepler, G.; Chellappan, S. Beta-arrestin-1 mediates nicotine-induced metastasis through E2F1 target genes that modulate epithelial to mesenchymal transition. Cancer Res. 2015, 75, 1009–1020. [Google Scholar] [CrossRef] [PubMed]
  81. Zou, W.; Zou, Y.; Zhao, Z.; Li, B.; Ran, P. Nicotine-induced epithelial to mesenchymal transition via Wnt/beta-catenin signaling in human airway epithelial cells. Am. J. Physiol. Lung Cell. Mol. Physiol. 2013, 304, 199–209. [Google Scholar] [CrossRef] [PubMed]
  82. Wu, Z.Q.; Li, X.Y.; Hu, C.Y.; Ford, M.; Kleer, C.G.; Weiss, S.J. Canonical Wnt signaling regulates SLUG activity and links epithelial to mesenchymal transition with epigenetic Breast Cancer 1, Early Onset (BRCA1) repression. Proc. Natl. Acad. Sci. USA 2012, 109, 16654–16659. [Google Scholar] [CrossRef] [PubMed]
  83. Yan, W.; Shao, R. Transduction of a mesenchyme-specific gene periostin into 293T cells induces cell invasive activity through epithelial to mesenchymal transformation. J. Biol. Chem. 2006, 281, 19700–19708. [Google Scholar] [CrossRef] [PubMed]
  84. Wu, S.Q.; Lv, Y.E.; Lin, B.H.; Luo, L.M.; Lv, S.L.; Bi, A.H.; Jia, Y.S. Silencing of periostin inhibits nicotine-mediated tumor cell growth and epithelial to mesenchymal transition in lung cancer cells. Mol. Med. Rep. 2013, 7, 875–880. [Google Scholar] [CrossRef] [PubMed]
  85. Yoshino, I.; Kometani, T.; Shoji, F.; Osoegawa, A.; Ohba, T.; Kouso, H.; Takenaka, T.; Yohena, T.; Maehara, Y. Induction of epithelial to mesenchymal transition-related genes by benzo[a]pyrene in lung cancer cells. Cancer 2007, 110, 369–374. [Google Scholar] [CrossRef] [PubMed]
  86. Ikuta, T.; Kawajiri, K. Zinc finger transcription factor SLUG is a novel target gene of aryl hydrocarbon receptor. Exp. Cell Res. 2006, 312, 3585–3594. [Google Scholar] [CrossRef] [PubMed]
  87. Lin, P.; Chang, H.; Ho, W.L.; Wu, M.H.; Su, J.M. Association of aryl hydrocarbon receptor and cytochrome P4501B1 expressions in human non-small cell lung cancers. Lung Cancer 2003, 42, 255–261. [Google Scholar] [CrossRef]
  88. Diry, M.; Tomkiewicz, C.; Koehle, C.; Coumoul, X.; Bock, K.W.; Barouki, R.; Transy, C. Activation of the dioxin/aryl hydrocarbon receptor (AhR) modulates cell plasticity through a JNK-dependent mechanism. Oncogene 2006, 25, 5570–5574. [Google Scholar] [CrossRef] [PubMed]
  89. Valavanidis, A.; Vlachogianni, T.; Fiotakis, K. Tobacco smoke: Involvement of reactive oxygen species and stable free radicals in mechanisms of oxidative damage, carcinogenesis and synergistic effects with other respirable particles. Int. J. Environ. Res. Public Health 2009, 6, 445–462. [Google Scholar] [CrossRef] [PubMed]
  90. Pfeifer, G.P.; Denissenko, M.F.; Olivier, M.; Tretyakova, N.; Hecht, S.S.; Hainaut, P. Tobacco smoke carcinogens, DNA damage and p53 mutations in smoking-associated cancers. Oncogene 2002, 21, 7435–7451. [Google Scholar] [CrossRef] [PubMed]
  91. Mallozzi, C.; Di Stasi, A.M.; Minetti, M. Activation of src tyrosine kinases by peroxynitrite. FEBS Lett. 1999, 456, 201–206. [Google Scholar] [CrossRef]
  92. Mehdi, M.Z.; Pandey, N.R.; Pandey, S.K.; Srivastava, A.K. H2O2-induced phosphorylation of ERK1/2 and PKB requires tyrosine kinase activity of insulin receptor and c-Src. Antioxid. Redox Signal. 2005, 7, 1014–1020. [Google Scholar] [CrossRef] [PubMed]
  93. Zhang, H.; Liu, H.; Borok, Z.; Davies, K.J.; Ursini, F.; Forman, H.J. Cigarette smoke extract stimulates epithelial to mesenchymal transition through Src activation. Free Radic. Biol. Med. 2012, 52, 1437–1442. [Google Scholar] [CrossRef] [PubMed]
  94. Liu, X.; Feng, R. Inhibition of epithelial to mesenchymal transition in metastatic breast carcinoma cells by c-Src suppression. Acta Biochim. Biophys. Sin. 2010, 42, 496–501. [Google Scholar] [CrossRef] [PubMed]
  95. Nagathihalli, N.S.; Massion, P.P.; Gonzalez, A.L.; Lu, P.; Datta, P.K. Smoking induces epithelial to-mesenchymal transition in non-small cell lung cancer through HDAC-mediated downregulation of E-cadherin. Mol. Cancer Ther. 2012, 11, 2362–2372. [Google Scholar] [CrossRef] [PubMed]
  96. Park, S.M.; Gaur, A.B.; Lengyel, E.; Peter, M.E. The miR-200 family determines the epithelial phenotype of cancer cells by targeting the E-cadherin repressors ZEB1 and ZEB2. Genes Dev. 2008, 22, 894–907. [Google Scholar] [CrossRef] [PubMed]
  97. Liu, Y.; Luo, F.; Xu, Y.; Wang, B.; Zhao, Y.; Xu, W.; Shi, L.; Lu, X.; Liu, Q. Epithelial to mesenchymal transition and cancer stem cells, mediated by a long non-coding RNA, HOTAIR, are involved in cell malignant transformation induced by cigarette smoke extract. Toxicol. Appl. Pharmacol. 2015, 282, 9–19. [Google Scholar] [CrossRef] [PubMed]
  98. Zhao, W.; An, Y.; Liang, Y.; Xie, X.W. Role of HOTAIR long noncoding RNA in metastatic progression of lung cancer. Eur. Rev. Med. Pharmacol. Sci. 2014, 18, 1930–1936. [Google Scholar] [PubMed]
  99. Gupta, R.A.; Shah, N.; Wang, K.C.; Kim, J.; Horlings, H.M.; Wong, D.J.; Tsai, M.C.; Hung, T.; Argani, P.; Rinn, J.L.; et al. Long non-coding RNA HOTAIR reprograms chromatin state to promote cancer metastasis. Nature 2010, 464, 1071–1076. [Google Scholar] [CrossRef] [PubMed]
  100. Wang, Q. Activation of uPAR is required for cigarette smoke extract-induced epithelial to mesenchymal transition in lung epithelial cells. Oncol. Res. 2013, 21, 295. [Google Scholar] [CrossRef] [PubMed]
  101. Lund, L.R.; Ellis, V.; Rønne, E.; Pyke, C.; Danø, K. Transcriptional and post-transcriptional regulation of the receptor for urokinase-type plasminogen activator by cytokines and tumour promoters in the human lung carcinoma cell line A549. Biochem. J. 1995, 310, 345–352. [Google Scholar] [CrossRef] [PubMed]
  102. Duffy, M.J.; Duggan, C. The urokinase plasminogen activator system: A rich source of tumour markers for the individualised management of patients with cancer. Clin. Biochem. 2004, 37, 541–548. [Google Scholar] [CrossRef] [PubMed]
  103. Nieder, C.; Bremnes, R.M. Effects of smoking cessation on hypoxia and its potential impact on radiation treatment effects in lung cancer patients. Strahlenther. Onkol. 2008, 184, 605–609. [Google Scholar] [CrossRef] [PubMed]
  104. Sagone, A.L., Jr.; Lawrence, T.; Balcerzak, S.P. Effect of smoking on tissue oxygen supply. Blood 1973, 41, 845–850. [Google Scholar] [PubMed]
  105. Nurwidya, F.; Takahashi, F.; Kobayashi, I.; Murakami, A.; Kato, M.; Minakata, K.; Nara, T.; Hashimoto, M.; Yagishita, S.; Baskoro, H.; et al. Treatment with insulin-like growth factor 1 receptor inhibitor reverses hypoxia-induced epithelial to mesenchymal transition in non-small cell lung cancer. Biochem. Biophys. Res. Commun. 2014, 455, 332–338. [Google Scholar] [CrossRef] [PubMed]
  106. Shaikh, D.; Zhou, Q.; Chen, T.; Ibe, J.C.; Raj, J.U.; Zhou, G. cAMP-dependent protein kinase is essential for hypoxia-mediated epithelial to mesenchymal transition, migration, and invasion in lung cancer cells. Cell. Signal. 2012, 24, 2396–2400. [Google Scholar] [CrossRef] [PubMed]
  107. Yang, M.H. Direct regulation of TWIST by HIF-1alpha promotes metastasis. Nat. Cell Biol. 2008, 10, 295–300. [Google Scholar] [CrossRef] [PubMed]
  108. Eurlings, I.M.; Reynaert, N.L.; Van den Beucken, T.; Gosker, H.R.; de Theije, C.C.; Verhamme, F.M.; Bracke, K.R.; Wouters, E.F.; Dentener, M.A. Cigarette smoke extract induces a phenotypic shift in epithelial cells; involvement of HIF1alpha in mesenchymal transition. PLoS ONE 2014, 9, 245–250. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  109. Shen, H.J.; Sun, Y.H.; Zhang, S.J.; Jiang, J.X.; Dong, X.W.; Jia, Y.L.; Shen, J.; Guan, Y.; Zhang, L.H.; Li, F.F.; et al. Cigarette smoke-induced alveolar epithelial to mesenchymal transition is mediated by Rac1 activation. Biochim. Biophys. Acta 2014, 1840, 1838–1849. [Google Scholar] [CrossRef] [PubMed]
  110. Price, L.S.; Collard, J.G. Regulation of the cytoskeleton by Rho-family GTPases: Implications for tumour cell invasion. Semin. Cancer Biol. 2001, 11, 167–173. [Google Scholar] [CrossRef] [PubMed]
  111. Yang, W.H.; Lan, H.Y.; Huang, C.H.; Tai, S.K.; Tzeng, C.H.; Kao, S.Y.; Wu, K.J.; Hung, M.C.; Yang, M.H. RAC1 activation mediates Twist1-induced cancer cell migration. Nat. Cell Boil. 2012, 14, 366–370. [Google Scholar] [CrossRef] [PubMed]
  112. Liang, Z.; Xie, W.; Wu, R.; Geng, H.; Zhao, L.; Xie, C.; Li, X.; Huang, C.; Zhu, J.; Zhu, M.; et al. ERK5 negatively regulates tobacco smoke-induced pulmonary epithelial to mesenchymal transition. Oncotarget 2015, 6, 19605–19618. [Google Scholar] [CrossRef] [PubMed]
  113. Nishimoto, S.; Nishida, E. MAPK signalling: ERK5 versus ERK1/2. EMBO Rep. 2006, 7, 782–786. [Google Scholar] [CrossRef] [PubMed]
  114. Zuo, Y.; Wu, Y.; Wehrli, B.; Chakrabarti, S.; Chakraborty, C. Modulation of ERK5 is a novel mechanism by which Cdc42 regulates migration of breast cancer cells. J. Cell. Biochem. 2015, 116, 124–132. [Google Scholar] [CrossRef] [PubMed]
  115. Zhou, C.; Nitschke, A.M.; Xiong, W.; Zhang, Q.; Tang, Y.; Bloch, M.; Elliott, S.; Zhu, Y.; Bazzone, L.; Yu, D.; et al. Proteomic analysis of tumor necrosis factor-alpha resistant human breast cancer cells reveals a MEK5/Erk5-mediated epithelial to mesenchymal transition phenotype. Breast Cancer Res. 2008, 10, 105–120. [Google Scholar] [CrossRef] [PubMed]
  116. Schnoll, R.; Rothman, R.; Newman, H.; Lerman, C.; Miller, S.M.; Movsas, B.; Sherman, E.; Ridge, J.A.; Unger, M.; Langer, C.; et al. Characteristics of cancer patients entering a smoking cessation program and correlates of quit motivation: Implications for the development of tobacco control programs for cancer patients. Psycho-Oncol. 2004, 13, 346–358. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Overview of the molecular networks by which different compounds in cigarette smoke regulate EMT. Reactive oxygen species (ROS) found in cigarette smoke can activate SRC resulting in cytoskeletal modification. Nicotine can induce EMT through nAChRs-dependent and nAChRs-independent pathways. In the nAChRs-dependent pathway, the activation of nAChRs results in the recruitment of β-arrestin1 on to the promoters and promotes the expression of mesenchymal markers. Additionally, the activation of nAChRs also induces the expression of periostin, the overexpression of which has been reported to cause EMT. In the nAChRs-independent pathway, nicotine can induce EMT through upregulating miR-21 in a manner dependent on TGF-β signaling. Nicotine-derived nitrosamine ketone (NNK) has been found to upregulate the expression of the mesenchymal marker fibronectin through cyclooxygenase-2 (COX-2), which is mediated by the activation of a7-nAChR. Polycyclic aromatic hydrocarbons (PAH) can mediate EMT through the activation of aryl hydrocarbon receptor (aryl hydrocarbon receptor), which triggers a marked cytoskeleton remodeling associated with the activation of the JUN N-terminal kinase (JNK) pathway. Increased nuclear accumulation of AhR in response to PAH also leads to transcriptional activation of SLUG.
Figure 1. Overview of the molecular networks by which different compounds in cigarette smoke regulate EMT. Reactive oxygen species (ROS) found in cigarette smoke can activate SRC resulting in cytoskeletal modification. Nicotine can induce EMT through nAChRs-dependent and nAChRs-independent pathways. In the nAChRs-dependent pathway, the activation of nAChRs results in the recruitment of β-arrestin1 on to the promoters and promotes the expression of mesenchymal markers. Additionally, the activation of nAChRs also induces the expression of periostin, the overexpression of which has been reported to cause EMT. In the nAChRs-independent pathway, nicotine can induce EMT through upregulating miR-21 in a manner dependent on TGF-β signaling. Nicotine-derived nitrosamine ketone (NNK) has been found to upregulate the expression of the mesenchymal marker fibronectin through cyclooxygenase-2 (COX-2), which is mediated by the activation of a7-nAChR. Polycyclic aromatic hydrocarbons (PAH) can mediate EMT through the activation of aryl hydrocarbon receptor (aryl hydrocarbon receptor), which triggers a marked cytoskeleton remodeling associated with the activation of the JUN N-terminal kinase (JNK) pathway. Increased nuclear accumulation of AhR in response to PAH also leads to transcriptional activation of SLUG.
Jcm 05 00044 g001
Figure 2. Signaling pathways involved in the inducing effect of cigarette smoke on EMT. Cigarette Smoke Extract (CSE) treatment increases the expression of SLUG, which in turn recruits histone deacetylases (HDACs) to E-cadherin promoter and suppresses E-cadherin expression at the transcriptional level. CSE enhances NF-κB activity, which results in increased expression of TWIST-1 and downregulation of miR-200c. CSE treatment induces secretion of IL-6, leading to the activation of STAT3, which binds to HOTAIR promoter regions and up-regulates HOTAIR. CSE-induced EMT is also found to be accompanied by increased expression of uPAR followed by the activation of AKT. CSE treatment induces hypoxic signaling through the activation of HIF1α, which transcriptionally upregulates the expression of TWIST1. CSE treatment can initiate EMT by increasing the expression of Rac1. Upregulation of Rac1 also leads to an increase in the expression of TGF-β1 and activation of AKT signaling in pulmonary epithelial cells.
Figure 2. Signaling pathways involved in the inducing effect of cigarette smoke on EMT. Cigarette Smoke Extract (CSE) treatment increases the expression of SLUG, which in turn recruits histone deacetylases (HDACs) to E-cadherin promoter and suppresses E-cadherin expression at the transcriptional level. CSE enhances NF-κB activity, which results in increased expression of TWIST-1 and downregulation of miR-200c. CSE treatment induces secretion of IL-6, leading to the activation of STAT3, which binds to HOTAIR promoter regions and up-regulates HOTAIR. CSE-induced EMT is also found to be accompanied by increased expression of uPAR followed by the activation of AKT. CSE treatment induces hypoxic signaling through the activation of HIF1α, which transcriptionally upregulates the expression of TWIST1. CSE treatment can initiate EMT by increasing the expression of Rac1. Upregulation of Rac1 also leads to an increase in the expression of TGF-β1 and activation of AKT signaling in pulmonary epithelial cells.
Jcm 05 00044 g002
Table 1. Factors/Proteins involved in epithelial to mesenchymal transition (EMT) and their relevance to lung cancer.
Table 1. Factors/Proteins involved in epithelial to mesenchymal transition (EMT) and their relevance to lung cancer.
FactorsDescriptionRelevance to Lung CancerRefs
Transcription factors
SNAIL1Zinc-finger protein, E-box transcriptional repressorPositive expression associated with poor survival in squamous cell and adenocarcinomas[8,9,10]
SLUGZinc-finger protein, E-box transcriptional repressorUpregulation associated with poor survival in squamous cell carcinoma[11]
TWIST1bHLH factorOverexpression in primary NSCLCs associated with a shorter overall survival[12,13,14]
ZEB1Zinc-finger protein, E-box transcriptional repressorHigher expression found in metastatic lung tumors compared to primary tumors[15,16]
FOXC2Forkhead box transcription factorOverexpression associated with a worse overall survival and correlated with a shorter recurrence-free survival in patients with stage-I non-small cell lung cancer[17]
FOXQ1Forkhead box transcription factorUpregulation in NSCLC resulting in poor prognosis[18]
FOXC1Forkhead box transcription factorUpregulation correlated with poor tumor differentiation, tumor-node-metastasis stage, and lymph node metastasis in NSCLC patients[19]
FOXM1Forkhead box transcription factorOverexpression associated with poor prognosis of NSCLC patients and tumor metastasis[20]
Factors directly associated with EMT
E-cadherinAdhesion glycoproteinReduced E-cadherin expression significantly correlated with lymph node metastasis[21]
N-cadherinAdhesion glycoproteinOverexpression associated with a shorter overall survival[22]
Syndecan-1Transmembrane (type I) heparan sulfate proteoglycanHigh pretreatment serum syndecan-1 level associated with poor prognosis in SCLC treated with platinum-based chemotherapy[23]
miR-21Non-coding RNAUpregulation of serum miR-21 strongly associated with lymph node metastasis and advanced clinical stage of NSCLC[24]
α-SMAα-Smooth muscle actinOverexpression associated with a poor prognosis in patients with clinical stage I-IIIA NSCLC after curative resection[27]
VimentinMember of the intermediate filament familyUpregulation correlated with lymph node metastasis in squamous cell lung carcinoma[25]
PeriostinOsteoblast Specific FactorOverexpression associated with decreased progression-free survival[26]

Share and Cite

MDPI and ACS Style

Vu, T.; Jin, L.; Datta, P.K. Effect of Cigarette Smoking on Epithelial to Mesenchymal Transition (EMT) in Lung Cancer. J. Clin. Med. 2016, 5, 44. https://doi.org/10.3390/jcm5040044

AMA Style

Vu T, Jin L, Datta PK. Effect of Cigarette Smoking on Epithelial to Mesenchymal Transition (EMT) in Lung Cancer. Journal of Clinical Medicine. 2016; 5(4):44. https://doi.org/10.3390/jcm5040044

Chicago/Turabian Style

Vu, Trung, Lin Jin, and Pran K. Datta. 2016. "Effect of Cigarette Smoking on Epithelial to Mesenchymal Transition (EMT) in Lung Cancer" Journal of Clinical Medicine 5, no. 4: 44. https://doi.org/10.3390/jcm5040044

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