Open Access

Regulatory function of peroxiredoxin I on 4‑(methylnitrosamino)‑1‑(3‑pyridyl)‑1‑butanone‑induced lung cancer development (Review)

  • Authors:
    • Hu-Nan Sun
    • Chen-Xi Ren
    • Yi-Xi Gong
    • Dan-Ping Xie
    • Taeho Kwon
  • View Affiliations

  • Published online on: April 12, 2021     https://doi.org/10.3892/ol.2021.12726
  • Article Number: 465
  • Copyright: © Sun et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Smoking is a major cause of lung cancer, and 4‑(methylnitrosamino)‑1‑(3‑pyridyl)‑1‑butanone (NNK) is one of the most important carcinogens in cigarette smoke. NNK modulates the expression of peroxiredoxin (Prdx) I in lung cancer. Prdx1 is upregulated in lung squamous cell carcinoma and lung adenocarcinoma, and considered a potential biomarker for lung cancer. The current article reviewed the role and regulatory mechanisms of Prdx1 in NNK‑induced lung cancer cells. Prdx1 protects erythrocytes and DNA from NNK‑induced oxidative damage, prevents malignant transformation of cells and promotes cytotoxicity of natural killer cells, hence suppressing tumor formation. In addition, Prdx1 has the ability to prevent NNK‑induced lung tumor metabolic activity and generation of large amount of reactive oxygen species (ROS) and ROS‑induced apoptosis, thus promoting tumor cell survival. In contrast to this, Prdx1, together with NNK, can promote the epithelial‑mesenchymal transition and migration of lung tumor cells. The signaling pathways associated with NNK and Prdx1 in lung cancer cells have been discussed in present review; however, numerous potential pathways are yet to be studied. To develop novel methods for treating NNK‑induced lung cancer, and improve the survival rate of patients with lung cancer, further research is needed to understand the complete mechanism associated with NNK.

Introduction

Environmental risk factors, such as cigarette smoking and asbestos, lead to increased risk of lung cancer. Before the 20th century, incidences of lung cancer were very rare, up to 1898, only 140 cases of lung cancer were reported in the world medical literature (1). Since the start of tobacco usage, the morbidity and mortality rates of lung cancer have been gradually rising (2,3), and according to WHO statistics, lung cancer has the highest incidence and mortality rate among the 36 most common cancers in the world in 2018 (3). Studies have shown that smoking is directly related to lung cancer (4). According to the statistical data from the World Health Organization, lung cancer was the leading cause of cancer-associated death in 2018; currently there are 300 million tobacco users worldwide, and there are 8 million deaths every year (3).

Multiple studies have shown that 4-(methylnitrosamino)-1-(3-pyridyl)-1-butanone (NNK) is the most potent carcinogen in tobacco that causes lung cancer (5,6). NNK can induce DNA strand breaks and DNA adduct formation, while its metabolism results in the generation of hydroxyl and other reactive oxygen radica9 ls, which in turn causes lung cancer (7). A study by Yeh et al (8) demonstrated that the incubation of A549 lung cancer cells with NNK results in increased levels of reactive oxygen species (ROS) formation (8). Furthermore, it has been shown that NNK induces oxidative stress by increasing ROS level in cells and promotes lung cancer progression, which may be associated with the changes in the expression of the genes related to ROS metabolism (9).

The antioxidant defense system in mammalian cells prevents excessive ROS accumulation (10) and maintains the intracellular redox balance. The peroxiredoxin (Prdx) family of proteins function in the cellular oxidative defense system, which eliminates ROS (11) and affects various cellular activities, such as cell proliferation, differentiation (12), apoptosis (13) and gene expression (14). Prdx1 inhibits NNK-induced DNA damage and prevents the development of lung tumors (1517). NNK-induced changes in the expression of peroxide redox proteins in lung cancer cells indicates that Prdx1 may be involved in the detoxification of ROS during NNK-induced oxidative stress (17). Hence, Prdx1 protects the cells, DNA and proteins from NNK-induced damage, and thus development of lung cancer. The interplay between NNK and Prdx1 has recently gained attention (12,1618), and an improved understanding of the role of Prdx1 in the development of NNK-induced lung cancer may shed new light towards the development of therapeutic strategies against lung cancer.

NNK

NNK, an aromatic compound, is the most potent carcinogen in tobacco smoke (19). In multiple organs, the nicotine in tobacco is rapidly metabolized by cytochrome (CY) P450. In the liver, it is hydroxylated by CYP2A26 at the 2′position to form an amino ketone intermediate, which is subsequently nitrosated to produce NNK (20).

In vivo experiments show that NNK is metabolized by three methods: Carbonyl reduction, pyridine oxidation and α-hydroxylation (21). In the carbonyl reduction process, NNK is carbonylated by 11β-hydroxysteroid dehydrogenase to form 4-(methylnitrosamino)-1-(3-pyridyl)-1-butanol (NNAL), which is then metabolized by glucuronidation to produce NNAL-glucuronic acid (22). During oxidation of pyridine nitrogen, CYP450 2B1 and CYP3A4 metabolize NNK to NNK-N-oxide (23). The α-hydroxylation process includes two modes: A-hydroxylation of the methyl carbon adjacent to the N-nitroso nitrogen and α-hydroxylation of the methylene carbon adjacent to the N-nitroso nitrogen. NNK is hydroxylated at the methyl group adjacent to N-nitroso to form α-hydroxymethyl-NNK, which then decomposes to form formaldehyde and 4-(3-pyridyl)-4-oxobutane-1-diazohydroxide. Finally, the latter reacts with water to form ketone alcohol. The methylene carbon of NNK can also be hydroxylated to generate an unstable α-methylene hydroxyl-NNK, which quickly decomposes to methane diazohydroxide and keto aldehyde, and finally keto aldehyde oxidizes to form keto acid (19) (Fig. 1).

Using Syrian golden hamster tissue sections, it has been shown that lung tissue has a lower NNK total metabolic rate compared with that of kidney and liver tissues (24). The oxidative metabolism of NNK to DNA-reactive intermediates by α-hydroxylation accounts for 13–31%, pyridine nitrogen oxidation accounts for 5–22%, while carbonyl group reduction of NNK to NNAL accounts for 47–81% of the total metabolism of NNK in the lung. The total metabolism of NNAL in all the tissues is ~10 times lower compared with that of NNK (24). The difference in the metabolic rate of various NNK metabolites is one of the reasons that the lung is more susceptible to the NNK carcinogen (24).

Prdx1

Prdxs, a class of antioxidant protective proteins, play an important role in the elimination of ROS and cancer development (25). Prdx1 is a member of the Prdxs family of proteins, which is primarily localized in the cytosol, as well as found in the nucleus, plasma, membrane and centrosome (11). Prdx1 is considered to be an important antioxidant protein (15), and exerts an antioxidant effect by forming a homodimer. The Cys52 sulfhydryl group on one peptide chain and the Cys172 sulfhydryl group on the other peptide chain are dehydrogenated to form an intermolecular disulfide bond, Cys52S-SCys172, which reduces peroxides by providing hydrogen ions, thereby detoxifying them (26). Prdx1 is highly sensitive to hydrogen peroxide (H2O2), among various peroxides (11).

In most cancer cells, such as breast, esophageal and lung cancer, Prdx1 can removes excess intracellular ROS, maintains ROS balance and protects the cells from oxidative stress-induced DNA damage (27). Furthermore, by eliminating ROS, Prdx1 prevents oxidative stress-induced mutations in the P53 and K-Ras genes, thereby inhibiting tumor formation, suppressing lung cancer cell proliferation, invasion and migration, and increasing radiation sensitivity of the cancer cells (15,28). Cys52 is the active center of Prdx1 (29), which either reacts with H2O2 (30) or combines with heme; hence, Prdx1 is also called as heme-binding protein 23 (31). Prdx1 acts as a scavenger for the cytoplasmic heme and has an important inhibitory effect on heme toxicity (32). Furthermore, Prdx1 enhances the immune activity of natural killer (NK) cells against tumor cells, and is also known as natural killer enhancement factor (11). It has been shown that Prdx1 can effectively prevent lung cancer progression by enhancing the tumor killing effects of NK cells (33,34). Prdx1 is overexpressed in non-small cell lung cancer (NSCLC) cells, which has been demonstrated to promote transforming growth factor-β1 (TGF-β1)-induced epithelial mesenchymal transition (EMT) and A549 cell migration (35). In addition, the interaction between Prdx1 and nuclear erythroid 2-related factor 2 (Nrf2) can significantly affect the proliferation of lung cancer cells (28,36). In a rat acute lung injury model, overexpression of Prdx1 increased the expression of proinflammatory cytokines interleukin-6 (IL-6), IL-8 and tumor necrosis factor-α (37). Inflammatory factors play an important role in the development of lung cancer. In addition, Prdx I affects the proliferation, migration and invasion of lung cancer cells by regulating various cytokines, in turn modulating different cell signaling pathways (38,39).

On one hand, Prdx1 protects macromolecules, such as proteins and DNA, from oxidative damage and suppresses malignant transformation of normal cells, thus preventing tumor development. On the other hand, Prdx1 inhibits ROS-induced apoptosis of cancer cells and promotes tumor cell survival (27). Hence, understanding its mechanism of action may provide novel insights into the development of better therapeutic strategies for lung cancer.

Prdx1 and NNK affect the growth and development of lung cancer by acting on P53 and K-Ras genes

NNK mainly undergoes metabolic activation through α-methyl and α-methylene hydroxylation, thereby producing DNA adducts and promoting cancer development. α-methyl hydroxylated metabolites of NNK can pyridyloxobutylate DNA and produce DNA pyridyloxobutyl adducts, whereas α-methylene hydroxylation generates α-methylenehydroxy-NNK, methane diazohydroxide and methyldiazonium ions. These react with DNA and yield 7-methyl guanine, O6−methyl guanine and O4-methyl thymine adducts (7). NNK induces oxidative stress by increasing the level of intracellular ROS, which in turn leads to the mutation of K-Ras and P53 oncogenes. In addition, the NNK metabolites have been shown to result in the mutation of K-Ras and P53 oncogenes in the lung. Thus, these deleterious effects of NNK on DNA may promote the development of lung cancer.

Prdx1 is considered a potential marker for NSCLC, and the interaction between Prdx1 and ROS plays an important role in the development of tumors (40). ROS plays a role in cell growth, differentiation, immune response and apoptosis (41,42). Increase in intracellular levels of ROS activates the expression of P53 (15), which in turn induces the expression of apoptotic factors, such as Bak and Bax, under oxidative stress, promotes the activation of caspases and finally activates the mitochondrial apoptotic signaling pathway (27). P53 in its active form suppresses the proliferation of abnormal cells, thereby exerting a tumor suppressor effect (43). In addition, P53 plays an important role in detecting DNA damage. P53 status after reducing the expression of Prdx1 is the major determinant of tumor growth and response of lung cancer cells to treatment (15). K-Ras mutations are known to cause uncontrolled division of human lung adenocarcinoma cells (4446). Furthermore, K-Ras mutations and ROS-induced oxidative stress are the major causes of NSCLC development. Prdx1 inhibits the activation of ROS/ERK/cyclin D1 pathway, thus results in Nrf2-dependent inhibition of K-Ras-driven lung tumorigenesis (28).

NNK may promote lung cancer development by increasing the intracellular ROS level and inducing mutations in important oncogenes. Prdx1 effectively eliminates excess ROS and prevents gene mutations caused by oxidative damage of DNA. Prdx1 prevents the occurrence of mutations in the P53 gene, enabling it to detect and repair damaged DNA. Furthermore, activation of the Nrf2 pathway results in upregulation of Prdx1. Prdx1 inhibits the ROS/ERK/cyclin D1 signaling pathway and suppresses the development of lung tumors (Fig. 2) (42,43,47,48).

Prdx1 and NNK affect the growth and development of lung cancer by reacting with heme and hemoglobin

It has been demonstrated that NNK-induced DNA damage is significantly reduced by antioxidants (BHT), catalase and superoxide dismutase (SOD) in A549 cells. The order of the effectiveness has been indicated to be BHT > catalase > SOD (8). Thus, it is speculated that NNK mainly induces generation of H2O2 (8,49,50). The α-methyl hydroxylation of NNK results in the formation of unstable α-hydroxymethyl NNK. The decomposition of a-hydroxymethyl NNK results in the formation of electrophilic 4-(3-pyridyl)-4-oxybutyl diazoxide, which can react with hemoglobin to form hemoglobin adduct (7,51). NNK α-methyl hydroxylation results in globulin methylation and pyridyloxybutylation (15), and the hemoglobin adduct formed by pyridyloxybutylation releases 4-hydroxy-1-(3-pyridyl)-1-butanone by alkaline hydrolysis (51). Studies have shown that keto alcohol-releasing adducts were formed by treatment of hemoglobin with NNK (5153). Phenylethyl isothiocyanate treatment can significantly inhibit NNK-mediated lung tumorigenesis by reducing the release of ketone alcohol products (54).

Prdx1 can bind to heme (53), which is abundant in red blood cells. Furthermore, heme is widely distributed in organelles, such as the nucleus, endoplasmic reticulum and plasma membrane (55,56), and it is involved in a processes in mammalian cells, including respiration, metabolism, transcription, DNA binding and protein degradation (55,57). Heme is synthesized in mitochondria and loosely bound to Prdx1 (31), which is proposed to facilitate the transport of heme to other organelles (55,56,58). Heme is insoluble in aqueous solutions and is toxic to the cells (59), and the toxicity is further manifested by the generation of ROS (57). However, binding of heme to Prdx1 reduces heme toxicity and promotes H2O2-mediated heme degradation (60). Thus, Prdx1 protects free heme from peroxidation but loses its peroxidase activity when bound to heme (31). NNK induces the generation H2O2 and Prdx1 is more sensitive to H2O2. H2O2 causes erythrocyte lysis, leading to the release of large amounts of heme and hemoglobin (61). Heme interacts with oxygen to produce ROS (62), which further destroys red blood cells. The binding of heme and Prdx1 reduces heme cytotoxicity; however, Prdx1 loses its ROS scavenging ability. NNK metabolites produce adducts with hemoglobin, thereby promoting the development of lung tumors, and Prdx1 plays a role in ROS scavenging, which in turn protects red blood cells from oxidative damage and inhibits lung tumorigenesis (Fig. 3).

Prdx1 and NNK affect the growth and development of lung cancer by acting on alveolar macrophages (AMs) and NK cells

NNK metabolites inhibit AM-mediated production of interleukin-12 (IL-12), nitric oxide and TNF; however, they also induce the production of IL-10 (63), which may promote the growth and development of lung tumors (64). Additionally, NNK has an inhibitory effect on TNF-dependent cytotoxicity of AMs (49). Furthermore, metabolites produced by NNK α-methyl hydroxylation may be involved in the regulation of AM function. For example, keto acid inhibits IL-12 production by AMs, while keto alcohol may inhibit the AM production of TNF and IL-12 (65). NNK may induce the expression of cyclooxygenase 2 (COX-2) and upregulate prostaglandin E2 (PGE2); PGE2 in turn upregulates IL-10 (63,66). Nicotinic acetylcholine receptors (nAChRs) are present in immune cells (67), and NNK has high affinity towards nAChRs. Thus, interaction of NNK and nAChRs may activate the production of IL-10 (68). Moreover, IL-10 suppresses the production of IL-12 (69), and inhibition of IL-12 leads to decreased expression of interferon-γ (70,71). Furthermore, IL-12 is mainly produced by phagocytes (monocytes/macrophages and neutrophils) and dendritic cells (72) and enhances the cytotoxicity of AM and NK cells (73). NK cells are a group of lymphocytes that can kill tumor cells (74). Therefore, it can be speculated that NNK attenuates the toxic effects of NK cells on tumor cells and further promotes the development of lung cancer.

All six Prdxs are expressed by human lung cells; however, AMs mainly express Prdx1 and III (75). Prdx1 may affect the production of pro-inflammatory cytokines in macrophages (76) and has the ability to enhance NK cell toxicity in vitro. Furthermore, it has been reported that free thiol groups are required to maintain NK cell toxicity against tumor cells (42). The alkylation of free sulfhydryl groups in Prdx1 upon reduction decreases its ability to enhance NK cell toxicity, further indicating the requirement of free thiol groups for enhanced cytotoxicity of NK cells (16). Therefore, Prdx1 not only protects the cells from oxidative damage, but also selectively promotes the killing effect of AM and NK cells in certain tumors.

NNK may activate AMs to produce H2O2. NNK inhibits the production of IL-12 and TNF by AMs, thereby reducing the cytotoxicity of AMs and NKs against the tumor cells. Prdx1 is expressed mainly in AMs and may play a role in immune regulation by affecting inflammatory factors. Prdx1 may inhibit lung tumors by enhancing the killing effect of NK cells. However, the exact role of NNK and Prdx1 in AM-mediated killing of tumor cells is still unclear (Fig. 4) (64,70,7476).

Prdx1 and NNK affect EMT

Long-term exposure of lung alveolar cells to NNK results in their proliferation and eventually malignant transformation (77). After the cells are exposed to NNK, the expression of intracellular β-catenin and F-actin decrease, whereas that of fibronectin, vimentin and matrix metalloproteinase-2 increase. This in turn promotes EMT (77). EMT is a process by which epithelial cells are transformed into the mesenchymal phenotype, which increases their invasion and migration capabilities. Morphologically, the epithelial cells are loosely connected and the cytoskeleton structure is reorganized. The EMT process is accompanied by various changes in protein expression, including a decrease in E-cadherin and an increase in fibronectin expression levels (77). Low E-cadherin expression decreases cell adhesion, and downregulation of E-cadherin is also considered as a sign of EMT (78). NNK and ROS induce EMT through different signaling pathways (79). In cancer cells, NNK may promote the production of ROS, such as H2O2, which in turn activates c-Src (80,81), leading to cytoskeletal modifications (79) and initiation of EMT (82). Most tissues usually do not express COX-2 or express it at a low level (83). The induced expression of COX-2 inhibits apoptosis and increases the migration potential of cancer cells (84). The combination of NNK and a7-nAChR can induce the expression of COX-2, which upregulates fibronectin and promotes EMT (79).

Furthermore, Prdx1 is a type of peroxidase reductase, which can play the role of scavenging ROS, thus inhibiting the EMT process. It has been observed that Prdx 1 can promote EMT in breast (85), pancreatic (86) and colon cancer (87). Furthermore, high expression levels of Prdx1 downregulates E-cadherin, whereas, at lower levels it upregulates E-cadherin in A549 lung adenocarcinoma cells (88). TGF-β1 is a pleiotropic cytokine that is involved in apoptosis, differentiation and proliferation of cells and is the primary inducer of EMT (83,8991). It has been shown that the overexpression of Prdx1 in lung cancer cells significantly enhances TGF-β1-mediated EMT and cell migration (92).

In A549 lung cancer cells, NNK treatment results in a significant increase in Prdx1 expression. NNK not only results in ROS production, but also upregulates the expression of fibronectin via COX-2 and promotes EMT. Prdx1 can play a role in scavenging ROS, and Prdx1 can also inhibit EMT process by scavenging ROS caused by NNK. Additionally, high levels of Prdx1 results in upregulation of E-cadherin, which promotes EMT (Fig. 5) (7782,84,85).

Signaling pathways associated with Prdx1 and NNK in lung cancer cells

NADPH oxidase of Nox family is expressed in both normal and cancer cells, and is related to ROS production and tumorigenicity in various cancer cells. For example, Nox1 is highly expressed in human colon cancer and prostate cancer, and lung cancer A549 cells also express Nox1, 2 and 4 (16,93). NNK induces the expression of NOX protein and results in the production of large amount of ROS, which causes damage to protein and DNA through oxidative stress. In A549 lung cancer cells, NNK-mediated generation of ROS through NOX activates phosphatidylinositol-3-kinase (PI3K)/protein kinase B (AKT) and Wnt signaling pathways, which leads to the development of drug resistance in lung cancer cells (16) and is associated with lower survival rate of patients with stage 1 lung tumors in Tumor-Node-Metastasis staging system (94). NNK increases the expression of thromboxane A2 (TxA2) and Tx receptor in lung cancer cells by elevating COX and Tx synthase expression (95). NNK has been shown to promote adhesion and invasion of CL1.0 cells through α7-nAChR/ERK/Contactin 1 signaling (96). Furthermore, NNK prevents PH domain leucine-rich repeat-containing protein phosphatase 2-mediated AKT dephosphorylation, activates AKT to inhibit E-cadherin expression and promotes lung cancer cell migration (97). The combination of NNK and α7-nAChRs activates c-Src and protein kinase C, and promotes the dissociation of phosphorylated Bad from Bcl-xl, which in turn inhibits apoptosis (98).

In tumor cells, Prdx1 has been reported to eliminate large quantities of ROS produced by tumor cell metabolism and thus, suppresses tumor cell death. Additionally, Prdx1 inhibits the oxidative stress-induced PI3K/AKT signaling pathway by eliminating ROS. It is known that the ROS-induced activation of PI3K/AKT is due to the oxidative inactivation of phosphatase and tensin homolog (PTEN) protein (99). Furthermore, Prdx1 protects PTEN lipid phosphatase activity from oxidative inactivation, thereby preventing AKT from driving tumor cell proliferation and inducing apoptosis (26). C-Abl plays a vital role in oxidative stress-induced cell death (100). Prdx1 can be used as a physiological inhibitor of C-Abl (18) to inhibit apoptosis induced by the C-Abl/P38/MAPK signaling pathway.

NNK induces Nox protein to produce ROS, and activates the PI3K/Akt signaling pathway; however, NNK inhibits the same pathway by removing ROS or preventing oxidative inactivation of phosphatase and PTEN. In addition, NNK activates α7-nAChRs and downstream signaling pathways, and hence promotes apoptosis and migration of lung cancer cells. Furthermore, both NNK and Prdx1 can regulate apoptosis-related proteins and thus, control the apoptosis of lung cancer cells (99,101103) (Fig. 6).

Conclusions

In conclusion, on the one hand, Prdx1 has the ability to protect erythrocytes and DNA from NNK-induced oxidative damage, prevent malignant transformation of cells and promote cytotoxicity of NK cells, suppressing tumor formation. In addition, Prdx1 prevents NNK-induced generation of large amount of ROS and hence, ROS-induced apoptosis, and promotes tumor cell survival. On the other hand, together with NNK, Prdx1 promotes EMT and migration of lung tumor cells. The signaling pathways of NNK and Prdx1 in lung cancer cells are intricate, and the associated mechanisms are yet to be explored.

Acknowledgments

Not applicable.

Funding

This research was supported by the Basic Science Research Program through the National Research Foundation of Korea funded by the Ministry of Education (grant no. 2020R1I1A2052417), The Korean Research Institute of Bioscience and Biotechnology Research Information System (grant no. RBM0112112) and The Project of Sanzong (grant no. ZRCPY202030) by Heilongjiang Bayi Agricultural University.

Availability of data and materials

Data sharing is not applicable to this article, as no datasets were generated or analyzed during the current study.

Authors' contributions

HNS, CXR, YXG and TK conceived and designed the review. HNS, CXR, YXG, DPX and TK wrote the manuscript and prepared the figures. HNS and TK reviewed and edited the manuscript. TK acquired the funding. All authors read and approved the final manuscript.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Glossary

Abbreviations

Abbreviations:

NNK

4-(methylnitrosamino)-1-(3-pyridyl)-1-butanone

Prdx1

peroxiredoxin I

ROS

reactive oxygen species

Prdx

peroxiredoxins

NK cells

natural killer cells

NSCLC

non-small cell lung cancer

TGF-β1

transforming growth factor-β1

EMT

epithelial mesenchymal transition

Nrf2

nucleosome 2-related factor 2

SOD

superoxide dismutase

H2O2

hydrogen peroxide

AM

alveolar macrophages

IL-12

interleukin-12

TNF

tumor necrosis factor

IL-10

interleukin 10

COX-2

cyclooxygenase 2

PGE2

prostaglandin E2

nAChRs

nicotinic acetylcholine receptors

PI3K

phosphatidylinositol-3-kinase

AKT

protein kinase B

TxA2

thromboxane A2

PTEN

phosphatase and tensin homolog

References

1 

Proctor RN: Tobacco and the global lung cancer epidemic. Nat Rev Cancer. 1:82–86. 2001. View Article : Google Scholar : PubMed/NCBI

2 

Thun M, Peto R, Boreham J and Lopez AD: Stages of the cigarette epidemic on entering its second century. Tob Control. 21:96–101. 2012. View Article : Google Scholar : PubMed/NCBI

3 

Bray F, Ferlay J, Soerjomataram I, Siegel RL, Torre LA and Jemal A: Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 68:394–424. 2018. View Article : Google Scholar : PubMed/NCBI

4 

Witschi H: Carcinogenic activity of cigarette smoke gas phase and its modulation by beta-carotene and N-acetylcysteine. Toxicol Sci. 84:81–87. 2005. View Article : Google Scholar : PubMed/NCBI

5 

Jin Z, Gao F, Flagg T and Deng X: Tobacco-specific nitrosamine 4-(methylnitrosamino)-1-(3-pyridyl)-1-butanone promotes functional cooperation of Bcl2 and c-Myc through phosphorylation in regulating cell survival and proliferation. J Biol Chem. 279:40209–40219. 2004. View Article : Google Scholar : PubMed/NCBI

6 

Maser E: Significance of reductases in the detoxification of the tobacco-specific carcinogen NNK. Trends Pharmacol Sci. 25:235–237. 2004. View Article : Google Scholar : PubMed/NCBI

7 

Yalcin E and de la Monte S: Tobacco nitrosamines as culprits in disease: Mechanisms reviewed. J Physiol Biochem. 72:107–120. 2016. View Article : Google Scholar : PubMed/NCBI

8 

Yeh SL, Wang WY, Huang CS and Hu ML: Flavonoids suppresses the enhancing effect of beta-carotene on DNA damage induced by 4-(methylnitrosamino)-1-(3-pyridyl)-1-butanone (NNK) in A549 cells. Chem Biol Interact. 160:175–182. 2006. View Article : Google Scholar : PubMed/NCBI

9 

Lehtonen ST, Svensk AM, Soini Y, Pääkkö P, Hirvikoski P, Kang SW, Säily M and Kinnula VL: Peroxiredoxins, a novel protein family in lung cancer. Int J Cancer. 111:514–521. 2004. View Article : Google Scholar : PubMed/NCBI

10 

Gorrini C, Harris IS and Mak TW: Modulation of oxidative stress as an anticancer strategy. Nat Rev Drug Discov. 12:931–947. 2013. View Article : Google Scholar : PubMed/NCBI

11 

Rhee SG and Kil IS: Multiple functions and regulation of mammalian peroxiredoxins. Annu Rev Biochem. 86:749–775. 2017. View Article : Google Scholar : PubMed/NCBI

12 

Bajor M, Zych AO, Graczyk-Jarzynka A, Muchowicz A, Firczuk M, Trzeciak L, Gaj P, Domagala A, Siernicka M, Zagozdzon A, et al: Targeting peroxiredoxin 1 impairs growth of breast cancer cells and potently sensitises these cells to prooxidant agents. Br J Cancer. 119:873–884. 2018. View Article : Google Scholar : PubMed/NCBI

13 

Han YH, Zhang YQ, Jin MH, Jin YH, Qiu MY, Li WL, He C, Yu LY, Hyun JW, Lee J, et al: Peroxiredoxin I deficiency increases keratinocyte apoptosis in a skin tumor model via the ROS-p38 MAPK pathway. Biochem Biophys Res Commun. 529:635–641. 2020. View Article : Google Scholar : PubMed/NCBI

14 

Hampton MB, Vick KA, Skoko JJ and Neumann CA: Peroxiredoxin involvement in the initiation and progression of human cancer. Antioxid Redox Signal. 28:591–608. 2018. View Article : Google Scholar : PubMed/NCBI

15 

Chen MF, Chen WC, Wu CT, Lin PY, Shau H, Liao SK, Yang CT and Lee KD: p53 status is a major determinant of effects of decreasing peroxiredoxin I expression on tumor growth and response of lung cancer cells to treatment. Int J Radiat Oncol Biol Phys. 66:1461–1472. 2006. View Article : Google Scholar : PubMed/NCBI

16 

Hirata N, Yamada S, Sekino Y and Kanda Y: Tobacco nitrosamine NNK increases ALDH-positive cells via ROS-Wnt signaling pathway in A549 human lung cancer cells. J Toxicol Sci. 42:193–204. 2017. View Article : Google Scholar : PubMed/NCBI

17 

Shi GQ, Zhou WS, Li M, Ren F and Han YW: Characterization and expression analysis of peroxiredoxin genes in NNK-induced V79 cells. Biomed Environ Sci. 30:224–228. 2017.PubMed/NCBI

18 

Immenschuh S and Baumgart-Vogt E: Peroxiredoxins, oxidative stress, and cell proliferation. Antioxid Redox Signal. 7:768–777. 2005. View Article : Google Scholar : PubMed/NCBI

19 

Akopyan G and Bonavida B: Understanding tobacco smoke carcinogen NNK and lung tumorigenesis. Int J Oncol. 29:745–752. 2006.PubMed/NCBI

20 

Hecht SS, Hochalter JB, Villalta PW and Murphy SE: 2′-Hydroxylation of nicotine by cytochrome P450 2A6 and human liver microsomes: Formation of a lung carcinogen precursor. Proc Natl Acad Sci USA. 97:12493–12497. 2000. View Article : Google Scholar : PubMed/NCBI

21 

Peterson LA: Context matters: Contribution of specific DNA adducts to the genotoxic properties of the tobacco-specific nitrosamine NNK. Chem Res Toxicol. 30:420–433. 2017. View Article : Google Scholar : PubMed/NCBI

22 

Ashmore JH, Luo S, Watson CJW and Lazarus P: Carbonyl reduction of NNK by recombinant human lung enzymes: Identification of HSD17β12 as the reductase important in (R)-NNAL formation in human lung. Carcinogenesis. 39:1079–1088. 2018. View Article : Google Scholar : PubMed/NCBI

23 

Leslie EM, Ghibellini G, Nezasa K and Brouwer KL: Biotransformation and transport of the tobacco-specific carcinogen 4-(methylnitrosamino)-1-(3-pyridyl)-1-butanone (NNK) in bile duct-cannulated wild-type and Mrp2/Abcc2-deficient (TR) Wistar rats. Carcinogenesis. 28:2650–2656. 2007. View Article : Google Scholar : PubMed/NCBI

24 

Richter E, Friesenegger S, Engl J and Tricker AR: Use of precision-cut tissue slices in organ culture to study metabolism of 4-(methylnitrosamino)-1-(3-pyridyl)-1-butanone (NNK) and 4-(methylnitrosamino)-1-(3-pyridyl)-1-butanol (NNAL) by hamster lung, liver and kidney. Toxicology. 144:83–91. 2000. View Article : Google Scholar : PubMed/NCBI

25 

Rhee SG, Kang SW, Chang TS, Jeong W and Kim K: Peroxiredoxin, a novel family of peroxidases. IUBMB Life. 52:35–41. 2001. View Article : Google Scholar : PubMed/NCBI

26 

Neumann CA, Cao J and Manevich Y: Peroxiredoxin 1 and its role in cell signaling. Cell Cycle. 8:4072–4078. 2009. View Article : Google Scholar : PubMed/NCBI

27 

Ding C, Fan X and Wu G: Peroxiredoxin 1-an antioxidant enzyme in cancer. J Cell Mol Med. 21:193–202. 2017. View Article : Google Scholar : PubMed/NCBI

28 

Park YH, Kim SU, Lee BK, Kim HS, Song IS, Shin HJ, Han YH, Chang KT, Kim JM, Lee DS, et al: Prx I suppresses K-ras-driven lung tumorigenesis by opposing redox-sensitive ERK/cyclin D1 pathway. Antioxid Redox Signal. 19:482–496. 2013. View Article : Google Scholar : PubMed/NCBI

29 

Chang TS, Jeong W, Choi SY, Yu S, Kang SW and Rhee SG: Regulation of peroxiredoxin I activity by Cdc2-mediated phosphorylation. J Biol Chem. 277:25370–25376. 2002. View Article : Google Scholar : PubMed/NCBI

30 

Poole LB: The basics of thiols and cysteines in redox biology and chemistry. Free Radic Biol Med. 80:148–157. 2015. View Article : Google Scholar : PubMed/NCBI

31 

Watanabe Y, Ishimori K and Uchida T: Dual role of the active-center cysteine in human peroxiredoxin 1: Peroxidase activity and heme binding. Biochem Biophys Res Commun. 483:930–935. 2017. View Article : Google Scholar : PubMed/NCBI

32 

Gozzelino R, Jeney V and Soares MP: Mechanisms of cell protection by heme oxygenase-1. Annu Rev Pharmacol Toxicol. 50:323–354. 2010. View Article : Google Scholar : PubMed/NCBI

33 

Guo Y, Patil NK, Luan L, Bohannon JK and Sherwood ER: The biology of natural killer cells during sepsis. Immunology. 153:190–202. 2018. View Article : Google Scholar : PubMed/NCBI

34 

Aktaş ON, Öztürk AB, Erman B, Erus S, Tanju S and Dilege Ş: Role of natural killer cells in lung cancer. J Cancer Res Clin Oncol. 144:997–1003. 2018. View Article : Google Scholar

35 

Li S, Wang R, Zhang M, Wang L and Cheng S: Proteomic analysis of non-small cell lung cancer tissue interstitial fluids. World J Surg Oncol. 11:1732013. View Article : Google Scholar : PubMed/NCBI

36 

Chen MF, Keng PC, Shau H, Wu CT, Hu YC, Liao SK and Chen WC: Inhibition of lung tumor growth and augmentation of radiosensitivity by decreasing peroxiredoxin I expression. Int J Radiat Oncol Biol Phys. 64:581–591. 2006. View Article : Google Scholar : PubMed/NCBI

37 

Liu D, Mao P, Huang Y, Liu Y, Liu X, Pang X and Li Y: Proteomic analysis of lung tissue in a rat acute lung injury model: Identification of PRDX1 as a promoter of inflammation. Mediators Inflamm. 2014:4693582014. View Article : Google Scholar : PubMed/NCBI

38 

Brenner DR, Fanidi A, Grankvist K, Muller DC, Brennan P, Manjer J, Byrnes G, Hodge A, Severi G, Giles GG, et al: Inflammatory cytokines and lung cancer risk in 3 prospective studies. Am J Epidemiol. 185:86–95. 2017. View Article : Google Scholar : PubMed/NCBI

39 

DeCotiis C, Hu Y, Greenberg AK, Huie M, Tsay JC, Pass H, Goldberg JD and Rom WN: Inflammatory cytokines and non-small cell lung cancer in a CT-scan screening cohort: Background review of the literature. Cancer Biomark. 16:219–233. 2016. View Article : Google Scholar : PubMed/NCBI

40 

Chang JW, Lee SH, Jeong JY, Chae HZ, Kim YC, Park ZY and Yoo YJ: Peroxiredoxin-I is an autoimmunogenic tumor antigen in non-small cell lung cancer. FEBS Lett. 579:2873–2877. 2005. View Article : Google Scholar : PubMed/NCBI

41 

Finkel T and Holbrook NJ: Oxidants, oxidative stress and the biology of ageing. Nature. 408:239–247. 2000. View Article : Google Scholar : PubMed/NCBI

42 

Moloney JN and Cotter TG: ROS signalling in the biology of cancer. Semin Cell Dev Biol. 80:50–64. 2018. View Article : Google Scholar : PubMed/NCBI

43 

Macip S, Igarashi M, Berggren P, Yu J, Lee SW and Aaronson SA: Influence of induced reactive oxygen species in p53-mediated cell fate decisions. Mol Cell Biol. 23:8576–8585. 2003. View Article : Google Scholar : PubMed/NCBI

44 

Moll HP, Pranz K, Musteanu M, Grabner B, Hruschka N, Mohrherr J, Aigner P, Stiedl P, Brcic L, Laszlo V, et al: Afatinib restrains K-RAS-driven lung tumorigenesis. Sci Transl Med. 10:eaao23012018. View Article : Google Scholar : PubMed/NCBI

45 

Aran V and Omerovic J: Current approaches in NSCLC targeting K-RAS and EGFR. Int J Mol Sci. 20:57012019. View Article : Google Scholar : PubMed/NCBI

46 

Scheffler M, Ihle MA, Hein R, Merkelbach-Bruse S, Scheel AH, Siemanowski J, Brägelmann J, Kron A, Abedpour N, Ueckeroth F, et al: K-ras mutation subtypes in NSCLC and associated Co-occuring mutations in other oncogenic pathways. J Thorac Oncol. 14:606–616. 2019. View Article : Google Scholar : PubMed/NCBI

47 

Peeper DS, Upton TM, Ladha MH, Neuman E, Zalvide J, Bernards R, DeCaprio JA and Ewen ME: Ras signalling linked to the cell-cycle machinery by the retinoblastoma protein. Nature. 386:177–181. 1997. View Article : Google Scholar : PubMed/NCBI

48 

Slebos RJ, Kibbelaar RE, Dalesio O, Kooistra A, Stam J, Meijer CJ, Wagenaar SS, Vanderschueren RG, van Zandwijk N, Mooi WJ, et al: K-ras oncogene activation as a prognostic marker in adenocarcinoma of the lung. N Engl J Med. 323:561–565. 1990. View Article : Google Scholar : PubMed/NCBI

49 

Proulx LI, Paré G and Bissonnette EY: Alveolar macrophage cytotoxic activity is inhibited by 4-(methylnitrosamino)-1-(3-pyridyl)-1-butanone (NNK), a carcinogenic component of cigarette smoke. Cancer Immunol Immunother. 56:831–838. 2007. View Article : Google Scholar : PubMed/NCBI

50 

Rioux N and Castonguay A: The induction of cyclooxygenase-1 by a tobacco carcinogen in U937 human macrophages is correlated to the activation of NF-kappaB. Carcinogenesis. 21:1745–1751. 2000. View Article : Google Scholar : PubMed/NCBI

51 

Wang Y, Narayanapillai SC, Hu Q, Fujioka N and Xing C: Detection and quantification of 4-hydroxy-1-(3-pyridyl)-1-butanone (HPB) from smoker albumin and its potential as a surrogate biomarker of tobacco-specific nitrosamines exposure and bioactivation. Toxicol Lett. 311:11–16. 2019. View Article : Google Scholar : PubMed/NCBI

52 

Peterson LA, Carmella SG and Hecht SS: Investigations of metabolic precursors to hemoglobin and DNA adducts of 4-(methylnitrosamino)-1-(3-pyridyl)-1-butanone. Carcinogenesis. 11:1329–1333. 1990. View Article : Google Scholar : PubMed/NCBI

53 

Hecht SS, Stepanov I and Carmella SG: Exposure and metabolic activation biomarkers of carcinogenic tobacco-specific nitrosamines. Acc Chem Res. 49:106–114. 2016. View Article : Google Scholar : PubMed/NCBI

54 

Hecht SS, Trushin N, Rigotty J, Carmella SG, Borukhova A, Akerkar S, Desai D, Amin S and Rivenson A: Inhibitory effects of 6-phenylhexyl isothiocyanate on 4-(methylnitrosamino)-1-(3-pyridyl)-1-butanone metabolic activation and lung tumorigenesis in rats. Carcinogenesis. 17:2061–2067. 1996. View Article : Google Scholar : PubMed/NCBI

55 

Tsiftsoglou AS, Tsamadou AI and Papadopoulou LC: Heme as key regulator of major mammalian cellular functions: Molecular, cellular, and pharmacological aspects. Pharmacol Ther. 111:327–345. 2006. View Article : Google Scholar : PubMed/NCBI

56 

Severance S and Hamza I: Trafficking of heme and porphyrins in metazoa. Chem Rev. 109:4596–4616. 2009. View Article : Google Scholar : PubMed/NCBI

57 

Shimizu T, Lengalova A, Martínek V and Martínková M: Heme: Emergent roles of heme in signal transduction, functional regulation and as catalytic centres. Chem Soc Rev. 48:5624–5657. 2019. View Article : Google Scholar : PubMed/NCBI

58 

Khan AA and Quigley JG: Control of intracellular heme levels: Heme transporters and heme oxygenases. Biochim Biophys Acta. 1813:668–682. 2011. View Article : Google Scholar : PubMed/NCBI

59 

Roumenina LT, Rayes J, Lacroix-Desmazes S and Dimitrov JD: Heme: Modulator of plasma systems in hemolytic diseases. Trends Mol Med. 22:200–213. 2016. View Article : Google Scholar : PubMed/NCBI

60 

Vlasova II: Peroxidase activity of human hemoproteins: Keeping the fire under control. Molecules. 23:25612018. View Article : Google Scholar : PubMed/NCBI

61 

Peralta IN, Cogoi L, Filip R and Anesini C: Prevention of hydrogen peroxide-induced red blood cells lysis by Ilex paraguariensis aqueous extract: Participation of phenolic and xanthine compounds. Phytother Res. 27:192–198. 2013. View Article : Google Scholar : PubMed/NCBI

62 

Hahl P, Hunt R, Bjes ES, Skaff A, Keightley A and Smith A: Identification of oxidative modifications of hemopexin and their predicted physiological relevance. J Biol Chem. 292:13658–13671. 2017. View Article : Google Scholar : PubMed/NCBI

63 

Therriault MJ, Proulx LI, Castonguay A and Bissonnette EY: Immunomodulatory effects of the tobacco-specific carcinogen, NNK, on alveolar macrophages. Clin Exp Immunol. 132:232–238. 2003. View Article : Google Scholar : PubMed/NCBI

64 

Liu Y and Cao X: The origin and function of tumor-associated macrophages. Cell Mol Immunol. 12:1–4. 2015. View Article : Google Scholar : PubMed/NCBI

65 

Proulx LI, Castonguay A and Bissonnette EY: Cytokine production by alveolar macrophages is down regulated by the alpha-methylhydroxylation pathway of 4-(methylnitrosamino)-1-(3-pyridyl)-1-butanone (NNK). Carcinogenesis. 25:997–1003. 2004. View Article : Google Scholar : PubMed/NCBI

66 

Proulx LI, Gaudreault M, Turmel V, Augusto LA, Castonguay A and Bissonnette EY: 4-(Methylnitrosamino)-1-(3-pyridyl)-1-butanone, a component of tobacco smoke, modulates mediator release from human bronchial and alveolar epithelial cells. Clin Exp Immunol. 140:46–53. 2005. View Article : Google Scholar : PubMed/NCBI

67 

Mashimo M: Dual roles of α7 nicotinic acetylcholine receptors expressed in immune cells in T cell differentiation-α7 nAChRs exert different actions between antigen-presenting cells and CD4(+) T cells. Yakugaku Zasshi. 140:1421–1425. 2020.(In Japanese). View Article : Google Scholar : PubMed/NCBI

68 

Schuller HM, Jull BA, Sheppard BJ and Plummer HK: Interaction of tobacco-specific toxicants with the neuronal alpha(7) nicotinic acetylcholine receptor and its associated mitogenic signal transduction pathway: Potential role in lung carcinogenesis and pediatric lung disorders. Eur J Pharmacol. 393:265–277. 2000. View Article : Google Scholar : PubMed/NCBI

69 

Rahim SS, Khan N, Boddupalli CS, Hasnain SE and Mukhopadhyay S: Interleukin-10 (IL-10) mediated suppression of IL-12 production in RAW 264.7 cells also involves c-rel transcription factor. Immunology. 114:313–321. 2005. View Article : Google Scholar : PubMed/NCBI

70 

Zirnheld AL, Villard M, Harrison AM, Kosiewicz MM and Alard P: β-Catenin stabilization in NOD dendritic cells increases IL-12 production and subsequent induction of IFN-γ-producing T cells. J Leukoc Biol. 106:1349–1358. 2019. View Article : Google Scholar : PubMed/NCBI

71 

Chakraborty K, Zhou Z, Wakamatsu N and Guerrero-Plata A: Interleukin-12p40 modulates human metapneumovirus-induced pulmonary disease in an acute mouse model of infection. PLoS One. 7:e371732012. View Article : Google Scholar : PubMed/NCBI

72 

Guo Y, Cao W and Zhu Y: Immunoregulatory functions of the IL-12 family of cytokines in antiviral systems. Viruses. 11:7722019. View Article : Google Scholar : PubMed/NCBI

73 

Trinchieri G: Interleukin-12 and the regulation of innate resistance and adaptive immunity. Nat Rev Immunol. 3:133–146. 2003. View Article : Google Scholar : PubMed/NCBI

74 

Caligiuri MA: Human natural killer cells. Blood. 112:461–469. 2008. View Article : Google Scholar : PubMed/NCBI

75 

Kinnula VL, Lehtonen S, Kaarteenaho-Wiik R, Lakari E, Pääkkö P, Kang SW, Rhee SG and Soini Y: Cell specific expression of peroxiredoxins in human lung and pulmonary sarcoidosis. Thorax. 57:157–164. 2002. View Article : Google Scholar : PubMed/NCBI

76 

Tae Lim Y, Sup Song D, Joon Won T, Lee YJ, Yoo JS, Eun Hyung K, Won Yoon J, Park SY and Woo Hwang K: Peroxiredoxin-1, a possible target in modulating inflammatory cytokine production in macrophage like cell line RAW264.7. Microbiol Immunol. 56:411–419. 2012. View Article : Google Scholar : PubMed/NCBI

77 

Mennecier G, Torres LN, Cogliati B, Sanches DS, Mori CM, Latorre AO, Chaible LM, Mackowiak II, Nagamine MK, Da Silva TC, et al: Chronic exposure of lung alveolar epithelial type II cells to tobacco-specific carcinogen NNK results in malignant transformation: A new in vitro lung carcinogenesis model. Mol Carcinog. 53:392–402. 2014. View Article : Google Scholar : PubMed/NCBI

78 

Yilmaz M and Christofori G: EMT, the cytoskeleton, and cancer cell invasion. Cancer Metastasis Rev. 28:15–33. 2009. View Article : Google Scholar : PubMed/NCBI

79 

Vu T, Jin L and Datta PK: Effect of cigarette smoking on epithelial to mesenchymal transition (EMT) in lung cancer. J Clin Med. 5:442016. View Article : Google Scholar : PubMed/NCBI

80 

Mehdi MZ, Pandey NR, Pandey SK and Srivastava AK: H2O2-induced phosphorylation of ERK1/2 and PKB requires tyrosine kinase activity of insulin receptor and c-Src. Antioxid Redox Signal. 7:1014–1020. 2005. View Article : Google Scholar : PubMed/NCBI

81 

Shen J, Xu L, Owonikoko TK, Sun SY, Khuri FR, Curran WJ and Deng X: NNK promotes migration and invasion of lung cancer cells through activation of c-Src/PKCι/FAK loop. Cancer Lett. 318:106–113. 2012. View Article : Google Scholar : PubMed/NCBI

82 

Zhang H, Liu H, Borok Z, Davies KJ, Ursini F and Forman HJ: Cigarette smoke extract stimulates epithelial-mesenchymal transition through Src activation. Free Radic Biol Med. 52:1437–1442. 2012. View Article : Google Scholar : PubMed/NCBI

83 

Shintani Y, Okimura A, Sato K, Nakagiri T, Kadota Y, Inoue M, Sawabata N, Minami M, Ikeda N, Kawahara K, et al: Epithelial to mesenchymal transition is a determinant of sensitivity to chemoradiotherapy in non-small cell lung cancer. Ann Thorac Surg. 92:1794–1804. 2011. View Article : Google Scholar : PubMed/NCBI

84 

Tseng YC, Tsai YH, Tseng MJ, Hsu KW, Yang MC, Huang KH, Li AF, Chi CW, Hsieh RH, Ku HH and Yeh TS: Notch2-induced COX-2 expression enhancing gastric cancer progression. Mol Carcinog. 51:939–951. 2012. View Article : Google Scholar : PubMed/NCBI

85 

Ocaña OH, Córcoles R, Fabra A, Moreno-Bueno G, Acloque H, Vega S, Barrallo-Gimeno A, Cano A and Nieto MA: Metastatic colonization requires the repression of the epithelial-mesenchymal transition inducer Prrx1. Cancer Cell. 22:709–724. 2012. View Article : Google Scholar

86 

Reichert M, Takano S, von Burstin J, Kim SB, Lee JS, Ihida-Stansbury K, Hahn C, Heeg S, Schneider G, Rhim AD, et al: The Prrx1 homeodomain transcription factor plays a central role in pancreatic regeneration and carcinogenesis. Genes Dev. 27:288–300. 2013. View Article : Google Scholar : PubMed/NCBI

87 

Lee JM, Dedhar S, Kalluri R and Thompson EW: The epithelial-mesenchymal transition: New insights in signaling, development, and disease. J Cell Biol. 172:973–981. 2006. View Article : Google Scholar : PubMed/NCBI

88 

Ha B, Kim EK, Kim JH, Lee HN, Lee KO, Lee SY and Jang HH: Human peroxiredoxin 1 modulates TGF-β1-induced epithelial-mesenchymal transition through its peroxidase activity. Biochem Biophys Res Commun. 421:33–37. 2012. View Article : Google Scholar : PubMed/NCBI

89 

Xu J, Lamouille S and Derynck R: TGF-beta-induced epithelial to mesenchymal transition. Cell Res. 19:156–172. 2009. View Article : Google Scholar : PubMed/NCBI

90 

Yang Y, Pan X, Lei W, Wang J and Song J: Transforming growth factor-beta1 induces epithelial-to-mesenchymal transition and apoptosis via a cell cycle-dependent mechanism. Oncogene. 25:7235–7244. 2006. View Article : Google Scholar : PubMed/NCBI

91 

Derynck R and Akhurst RJ: Differentiation plasticity regulated by TGF-beta family proteins in development and disease. Nat Cell Biol. 9:1000–1004. 2007. View Article : Google Scholar : PubMed/NCBI

92 

Gotzmann J, Huber H, Thallinger C, Wolschek M, Jansen B, Schulte-Hermann R, Beug H and Mikulits W: Hepatocytes convert to a fibroblastoid phenotype through the cooperation of TGF-beta1 and Ha-Ras: Steps towards invasiveness. J Cell Sci. 115:1189–1202. 2002.PubMed/NCBI

93 

Ushio-Fukai M and Nakamura Y: Reactive oxygen species and angiogenesis: NADPH oxidase as target for cancer therapy. Cancer Lett. 266:37–52. 2008. View Article : Google Scholar : PubMed/NCBI

94 

Jiang F, Qiu Q, Khanna A, Todd NW, Deepak J, Xing L, Wang H, Liu Z, Su Y, Stass SA and Katz RL: Aldehyde dehydrogenase 1 is a tumor stem cell-associated marker in lung cancer. Mol Cancer Res. 7:330–338. 2009. View Article : Google Scholar : PubMed/NCBI

95 

Huang RY, Li MY, Hsin MK, Underwood MJ, Ma LT, Mok TS, Warner TD and Chen GG: 4-Methylnitrosamino- 1-3-pyridyl-1-butanone (NNK) promotes lung cancer cell survival by stimulating thromboxane A2 and its receptor. Oncogene. 30:106–116. 2011. View Article : Google Scholar : PubMed/NCBI

96 

Hung YH and Hung WC: 4-(Methylnitrosamino)-1- (3-pyridyl)-1-butanone (NNK) enhances invasiveness of lung cancer cells by up-regulating contactin-1 via the alpha7 nicotinic acetylcholine receptor/ERK signaling pathway. Chem Biol Interact. 179:154–159. 2009. View Article : Google Scholar : PubMed/NCBI

97 

Yan J, Wong N, Hung C, Chen WX and Tang D: Contactin-1 reduces E-cadherin expression via activating AKT in lung cancer. PLoS One. 8:e654632013. View Article : Google Scholar : PubMed/NCBI

98 

Jin Z, Xin M and Deng X: Survival function of protein kinase C{iota} as a novel nitrosamine 4-(methylnitrosamino)-1-(3-pyridyl)-1-butanone-activated bad kinase. J Biol Chem. 280:16045–16052. 2005. View Article : Google Scholar : PubMed/NCBI

99 

Wang KC, Liu YC, El-Shazly M, Shih SP, Du YC, Hsu YM, Lin HY, Chen YC, Wu YC, Yang SC and Lu MC: The antioxidant from ethanolic extract of Rosa cymosa fruits activates phosphatase and tensin homolog in vitro and in vivo: A new insight on its antileukemic effect. Int J Mol Sci. 20:19352019. View Article : Google Scholar : PubMed/NCBI

100 

Neumann CA and Fang Q: Are peroxiredoxins tumor suppressors? Curr Opin Pharmacol. 7:375–380. 2007. View Article : Google Scholar : PubMed/NCBI

101 

Nauseef WM: Biological roles for the NOX family NADPH oxidases. J Biol Chem. 283:16961–16965. 2008. View Article : Google Scholar : PubMed/NCBI

102 

Ge GZ, Xu TR and Chen C: Tobacco carcinogen NNK-induced lung cancer animal models and associated carcinogenic mechanisms. Acta Biochim Biophys Sin (Shanghai). 47:477–487. 2015. View Article : Google Scholar : PubMed/NCBI

103 

Hong WG, Kim JY, Cho JH, Hwang SG, Song JY, Lee E, Chang TS, Um HD and Park JK: AMRI-59 functions as a radiosensitizer via peroxiredoxin I-targeted ROS accumulation and apoptotic cell death induction. Oncotarget. 8:114050–114064. 2017. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

June-2021
Volume 21 Issue 6

Print ISSN: 1792-1074
Online ISSN:1792-1082

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Sun H, Ren C, Gong Y, Xie D and Kwon T: Regulatory function of peroxiredoxin I on 4‑(methylnitrosamino)‑1‑(3‑pyridyl)‑1‑butanone‑induced lung cancer development (Review). Oncol Lett 21: 465, 2021
APA
Sun, H., Ren, C., Gong, Y., Xie, D., & Kwon, T. (2021). Regulatory function of peroxiredoxin I on 4‑(methylnitrosamino)‑1‑(3‑pyridyl)‑1‑butanone‑induced lung cancer development (Review). Oncology Letters, 21, 465. https://doi.org/10.3892/ol.2021.12726
MLA
Sun, H., Ren, C., Gong, Y., Xie, D., Kwon, T."Regulatory function of peroxiredoxin I on 4‑(methylnitrosamino)‑1‑(3‑pyridyl)‑1‑butanone‑induced lung cancer development (Review)". Oncology Letters 21.6 (2021): 465.
Chicago
Sun, H., Ren, C., Gong, Y., Xie, D., Kwon, T."Regulatory function of peroxiredoxin I on 4‑(methylnitrosamino)‑1‑(3‑pyridyl)‑1‑butanone‑induced lung cancer development (Review)". Oncology Letters 21, no. 6 (2021): 465. https://doi.org/10.3892/ol.2021.12726