Ethyl pyruvate suppresses the growth, invasion and migration and induces the apoptosis of non‑small cell lung cancer cells via the HMGB1/RAGE axis and the NF‑κB/STAT3 pathway

  • Authors:
    • Qiongqiong Liu
    • Yansong Huo
    • Hong Zheng
    • Jing Zhao
    • Li Jia
    • Peiguo Wang
  • View Affiliations

  • Published online on: May 30, 2019     https://doi.org/10.3892/or.2019.7176
  • Pages: 817-825
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

As an inhibitor of high mobility group protein B1 (HMGB1), ethyl pyruvate (EP) has been associated with various inflammatory diseases. Recent studies have investigated the relationship between EP and cancer. The present study aimed to determine the antitumor efficacy of EP in non‑small cell lung cancer (NSCLC) cells and elucidate the underlying mechanism. A549, H520 and PC‑9 cells were treated with EP in suitable concentrations. RT‑qPCR and western blot analysis were performed to evaluate HMGB1 and RAGE expression levels. MTT and colony formation assays assessed the effect of EP on cell growth. A Transwell assay was used to evaluate invasion and migration potential and flow cytometry was performed to analyze cell apoptosis. Bcl‑2 family proteins were identified by western blot analysis. The results demonstrated that an increased EP concentration effectively reduced HMGB1 and RAGE expression, thus inhibiting the HMGB1/RAGE axis. EP decreased level of PCNA and MMP‑9 and increased P53 levels. Bcl‑2 and Mcl‑1 were also decreased, whereas Bax expression was increased. Furthermore, a high concentration of EP (30 mmol/l) significantly inhibited NF‑κB and STAT3 activation. In summary, EP inhibited NSCLC cell growth, invasion and migration and induced apoptosis by suppressing the HMGB1/RAGE axis and the NF‑κB/STAT3 pathway, thus suggesting that EP may be a valuable therapeutic agent for NSCLC.

Introduction

Lung cancer is a leading cause of cancer-related mortality, with non-small cell lung cancer (NSCLC) accounting for ~80% of all lung cancer cases. Lung cancer incidence and mortality have significantly increased worldwide (1). The 5-year overall survival (OS) rate is only 15% among patients treated with traditional chemotherapeutic drugs due to the development of side effects and drug resistance. Approximately 1.7 million patients worldwide succumb to lung cancer yearly (2). It is therefore important to identify new therapies that may prolong the survival of lung cancer patients.

Cytokines, including inflammatory factors in the microenvironment, affect tumor cell proliferation and survival (3,4). Various inflammatory factors, including TNF-α, IL-6 and IL-8, promote tumor growth through Toll-like receptor (TLR)-mediated signaling pathways, thus promoting ERK, NF-κB and STAT3 activation (57). Damage-associated molecular patterns (DAMPs) are released by stressed, injured or dying cells, and initiate non-infectious inflammatory responses (8,9). High mobility group protein B1 (HMGB1), one of the DAMPs, is released from damaged, inflamed, and cancerous cells, in turn promoting tumor cell survival through its receptor RAGE. HMGB1, which organizes DNA and regulates transcription, has various biological functions in and outside the cell, and promotes inflammation and tumorigenesis. The HMGB1/RAGE axis may cause pro-inflammatory gene activation (10). Due to enhanced levels of HMGB1 in certain chronic diseases, the RAGE receptor is believed to exert a promoting effect in inflammatory diseases and various tumors. NF-κB is a heterodimeric protein belonging to the Rel family, and plays a major role in stress-induced immune and inflammatory responses (11). It was previously demonstrated that the NF-κB transcription factor group has an important association with tumor progression by affecting programmed cell death, proliferation control and tumorigenesis (12). Recent studies suggest that STAT proteins (particularly STAT3) play a crucial role in the carcinogenic inflammatory microenvironment at the early stages of malignant transformation, as well as during cancer progression (13,14). Several human malignancies, including lymphoma, leukemia and multiple myeloma, are associated with STAT3 (15). Therefore, inhibiton of STAT3 signaling may be a promising approach to cancer treatment.

Ethyl pyruvate (EP), a lipophilic ester, is derived from pyruvic acid and is a non-toxic food additive (16,17). EP has pharmacological benefits, namely alleviation of redox-caused cellular and tissue damage (18), anti-inflammatory properties (19,20) and promotion of apoptosis (21). EP inhibits HMGB1 release (22). Although EP may suppress various tumors to different degrees, its effect on lung cancer remains unclear. The aim of the present study was to investigate whether EP exerts antitumor effects on lung cancer cells and elucidate the underlying mechanism.

Materials and methods

Materials

The NSCLC cell lines A549, H520 and PC-9 were obtained from Tianjin Medical University Cancer Institute. The primers for HMGB1, RAGE, MMP-9, PCNA, P53 and ACTIN were all obtained from Invitrogen; Thermo Fisher Scientific, Inc. HMGB1 (cat. no. ab227168), RAGE (cat. no. ab3611), MMP-9 (cat. no. ab38898) antibodies were all purchased from Abcam. ACTIN (cat. no. sc-58673), GAPDH (cat. no. sc-365062), P53 (cat. no. sc-126), pCNA (cat. no. sc-9857), Bax (cat. no. sc-4239), Bcl-2 (cat. no. sc-7382), Mcl-1 (cat. no. sc-12756) and STAT3 (cat. no. sc-8019) antibodies were purchased from Santa Cruz Biotechnology, Inc. The antibodies against NF-κB (cat. no. 8242), p-NF-κB (cat. no. 3033 and p-STAT3 (cat. no. 9131) were all purchased from Cell Signaling Technology, Inc.

Drugs and reagents

MTT was purchased from Roche Molecular Biochemicals. EP was purchased from Sigma-Aldrich; Merck KGaA and RPMI-1640 from Thermo Fisher Scientific, Inc. Fetal bovine serum (FBS) was obtained from Gibco; Thermo Fisher Scientific, Inc. and TRIzol reagent was purchased from Invitrogen; Thermo Fisher Scientific, Inc. The PrimeScript RT Reagent kit (Perfect Real-Time) and SYBR Premix Ex Taq (Tli RNaseH Plus) were obtained from Takara Biotechnology Co., Ltd. The cell apoptosis kit with propidium iodide and Annexin V-FITC was from BD Pharmingen.

Cell culture

The cell lines A549, H520 and PC-9 were all cultured in RPMI-1640 medium. All three cultures were supplemented with 10% FBS, penicillin (100 U/ml) and streptomycin (100 mg/ml). The cell lines were all maintained at 37°C in a humidified atmosphere containing 5% CO2.

MTT assay

The MTT assay was used to analyze cell growth. Following treatment with EP, the cells were incubated in a 96-well-plate at a density of 3×104 cells/well. MTT (10 µl, 5 mg/ml) was added to the wells at 0, 1, 2, 3 and 4 days of culture. After incubation for 2 h, the crystals were dissolved by adding DMSO (150 µl/well) and mixed well with a multichannel pipette. The optical density (OD) of the soluble formazan in each well was measured at 490 nm with a microplate reader (Thermo Fisher Scientific, Inc.). All experiments were performed in triplicate.

RT-qPCR

mRNA expression levels were determined by RT-qPCR. Total RNA was extracted with TRIzol according to the manufacturer's specifications. Reverse transcription was carried out using a Prime Script RT Reagent kit (Perfect Real-Time) and cDNA amplification was conducted using SYBR Premix Ex Taq (Tli RNaseH Plus) according to the manufacturer's instructions. Target genes were amplified using oligonucleotide primers. The ACTIN gene was used as endogenous control. The PCR primer sequences are listed in Table I. Data were analyzed using the comparative Cq method (2−ΔΔCq) (23). Each experiment was conducted in triplicate.

Table I.

Primer sequences for PCR.

Table I.

Primer sequences for PCR.

GenesPrimer sequences (5′→3′)
HMGB1Forward ATATGGCAAAAGCGGACAAG
Reverse AGGCCAGGATGTTCTCCTTT
RAGEForward GTCATGGAACTGCCCAAACT
Reverse TCCTTCTGCGGATCTGTCTT
PCNAForward GCCGAGATCTCAGCCATATT
Reverse ATGTACTTAGAGGTACAAAT
MMP-9Forward CGCAGACATCGTCATCCAGT
Reverse GGATTGGCCTTGGAAGATG
P53Forward AACGGTACTCCGCCACC
Reverse CGTGTCACCGTCGTGGA
ACTINForward CTGGAACGGTGAAGGTGACA
Reverse AAGGGACTTCCTGTAACAATGCA
Western blot analysis

The cells of each group were collected and treated with EP (0, 5, 10, 20 and 30 mmol/l). Subsequently, proteins were extracted with CellLytic M cell Lysis Reagent with protease inhibitor and phosphatase inhibitor cocktails (all from Sigma-Aldrich; Merck KGaA). The proteins were quantified using a BCA protein assay kit. Proteins (30 µg) were separated by 10% SDS-PAGE. Polyvinylidene difluoride (PVDF) membrane were blocked with a blocking reagent [Tris-buffered saline-0.1% Tween-20 (TBST) containing 5% polyvinyl pyrrolidone (PVP), 5% FBS] at room temperature for 1 h and then incubated with primary antibody (dilution 1:1,000) overnight at 4°C. After washing the membranes three times, PVDF membranes were incubated with HRP-conjugated secondary antibodies (Cell Signaling Technology; dilution 1:2,000) for 1 h at normal temperature. Immunoreactive bands were visualized by an Imaging System (Tanon Science and Technology) after adding chemiluminescent HRP substrate (Millipore Corp.). Each experiment was conducted in triplicate.

Colony formation assay

The A549, H520 and PC-9 cells were treated with EP and seeded in 6-well plates with 800 cells/clone. The culture medium was replaced every 3 days. Colonies were counted when visible with the naked eye. The cells were then fixed with methanol and stained with 5% crystal violet solution. Each experiment was conducted in triplicate.

Cell apoptosis analysis

The percentage of apoptotic cells was calculated by means of an Annexin-V fluorescein isothiocyanate (FITC) and propidium iodide (PI) apoptosis detection kit (BD Biosciences) according to the manufacturer's instructions. The percentage of apoptotic cells (Annexin+/PI+) was analyzed via flow cytometry (BD Biosciences). Each experiment was conducted in triplicate.

Transwell assay

Transwell chambers (8 µm) were used for cell migration and invasion assays (Corning, Inc.). For the migration assay, 1×105 cells were harvested and seeded on the upper chamber. For the invasion assay, 3×105 cells were harvested and placed in the upper chamber coated with Matrigel (300 µg/ml, 100 µl). RPMI-1640 (600 µl) supplemented with 20% FBS was added to the lower chamber. The migrating or invading cells were fixed with 100% methanol for 30 min and stained with 5% crystal violet solution for 8 min post 24 h. Cells in 5 random fields (×4) were calculated using Nikon MA100 optical microscope (Nikon Corp., Tokyo. Japan). Each assay was performed in triplicate.

Statistical analysis

All statistical analyses were carried out using SPSS 17.0 software (SPSS, Inc., Chicago, IL, USA). Results are presented as mean ± standard deviation. The capability of cell migration and invasion between 20 mmol/l EP and 0 mmol/l EP in Fig. 3 (bar graph) were calculated using independent sample t-tests. Furthermore, the statistical analysis method used for the data of mRNA expression level and cell apoptosis index in Figs. 1, 2 and 4 (bar graphs) was the Kruskal-Wallis followed by Dunnett's post hoc test when the 0 mmol/l EP group was compared with the 5, 10, 20 and 30 mmol/l EP groups. P<0.05 was considered to indicate a statistically significant difference.

Results

Administration of EP suppresses the expression of the HMGB1/RAGE axis and the NF-κB/STAT3 pathway in lung cancer cells

RAGE, also known as AGER, is a transmembrane receptor that binds to a variety of ligands, including HMGB1 (24,25). A549, H520 and PC-9 cells were treated with EP (0, 5, 10, 20 and 30 mmol/l) in order to investigate its effects on HMGB1 and RAGE protein as well as mRNA expression. The protein and mRNA expression levels of HMGB1 were significantly decreased with increasing EP dose (Fig. 1A and B). Similarly, the mRNA and protein expression levels of RAGE were also decreased with increasing EP dose (Fig. 1C and D).

NF-κB regulates DNA transcription, cytokine production and cell survival (26). In the present study, A549, H520 and PC-9 cells were treated with 30 mmol/l EP for 2, 4, 6 and 8 h in order to examine NF-κB and p-NF-κB levels. The protein expression of p-NF-κB was gradually decreased over time (Fig. 1E). STAT3 is a member of the STAT protein family (27). A549, H520, PC-9 cells were treated with 30 mmol/l EP for 2, 4, 6 and 8 h in order to examine STAT3 and p-STAT3 levels. The protein expression level of p-STAT3 was also decreased over the specified time period (Fig. 1F).

Administration of EP suppresses the growth of lung cancer cells

A hallmark of cancer is enhanced cell proliferation (28). A549 H520 and PC-9 cell growth was assessed via MTT and colony formation assays. It was observed that A549, H520 and PC-9 cell growth was markedly suppressed by EP in a dose/time-dependent manner (Fig. 2A). The colony formation rate of A549, H520 and PC-9 cells was significantly decreased in the EP-treated groups (Fig. 2B and C).

Proliferating cell nuclear antigen (PCNA) is a DNA clamp that is essential for replication (29) and tumor progression. Therefore, anticancer treatment effectiveness may be determined by PCNA (30). PCNA was investigated by western blot analysis and RT-qPCR. The data demonstrated that the expression of PCNA was decreased in the EP-treated groups (Fig. 2D and E).

Administration of EP inhibits the migration and invasion of lung cancer cells

A Transwell assay demonstrated that the migration and invasion potential of A549, H520 and PC-9 cells in the EP-treated groups was reduced compared with that of the control group (Fig. 3A-D).

Matrix metallopeptidase 9 (MMP-9) participates in the degradation of the extracellular matrix (31). MMP-9 may be associated with the development of various human malignancies by affecting invasion, metastasis, growth and angiogenesis (32,33). Western blot analysis revealed downregulation of MMP-9 (Fig. 3E). The results were consistent with those of RT-qPCR (Fig. 3F).

Administration of EP promotes apoptosis of lung cancer cells

Flow cytometry was used to determine whether EP affects the apoptotic process of lung cancer cells. The results demonstrated that the apoptotic index of A549, H520 and PC-9 cells in the EP-treated groups was significantly higher compared with that of the control group (Fig. 4A and B).

P53, a tumor suppressor, directly participates in the endogenous apoptotic pathway by interacting with members of the Bcl-2 family (34,35). Western blot analysis and qPCR were used to determine P53 changes. The data demonstrated that the protein and mRNA levels of P53 increased with increasing EP concentration (Fig. 4C and D).

The Bcl-2 family plays an important role in the regulation of apoptosis, a type of programmed cell death. Bcl-2 family members may promote or inhibit apoptosis (36). Western blot analysis revealed that EP treatment increased the pro-apoptotic protein Bax and decreased the anti-apoptotic proteins Bcl-2 and Mcl-1 (Fig. 4E).

Discussion

In the present study, it was established that ethyl pyruvate (EP), a lipophilic ester, suppressed lung cancer cell growth, invasion and migration in vitro, and promoted apoptosis. Our results showed that the EP-mediated suppression of lung cancer cell growth was mediated via inhibition of the HMGB1/RAGE axis and the NF-κB/STAT3 pathway.

The results revealed that inflammatory factors play an important role in tumor initiation and progression. As an anti-inflammatory drug, EP has been shown to decrease organ dysfunction in several inflammation-induced disease models (19,20). Several trials have attempted to improve the overall surivial (OS) rate of cancer patients via EP administration. It was previously demonstrated that tumor progression may be suppressed by EP. The OS of animals in several tumor models, including hepatic, gastric, gallbladder cancer and mesothelioma, may be increased via EP (3742). As an HMGB1 inhibitor, the antitumor effect of EP is mediated by inhibiting the HMGB1/RAGE axis (37,40,42). Previous studies have investigated the expression levels of HMGB1 and its receptor RAGE in lung cancer (43), and reported that growth, invasion and migration of lung cancer cells are dependent on these signaling molecules (44). In the present study, a significant reduction in HMGB1 and RAGE protein levels was observed in EP-treated lung cancer cells. Furthermore, RT-qPCR demonstrated that HMGB1-induced RAGE mRNA expression was specifically suppressed by EP treatment. Therefore, the antitumor effects exerted by EP on lung cancer cell lines may be explained by disruption of the HMGB1/RAGE loop. Using varying concentrations of EP, decreased non-small cell lung cancer (NSCLC) cell growth, invasion and migration, as well as increased apoptosis, were observed. To the best of our knowledge, this is the first study to investigate EP as a potential therapeutic agent for NSCLC. NF-κB has been found to be active in tumor cells; therefore, NF-κB suppression may prevent tumor cell proliferation, thus enhancing the effectiveness of antitumor agents (45,46). STAT3 regulates gene expression and plays a critical role in a number of cellular processes, such as cell growth and apoptosis (47). In the present study, EP suppressed the RELA protein expression in a time-dependent manner in three lung cancer cell lines via phosphorylation of Ser536. In regards to the STAT3 pathway, EP suppressed STAT3 in a time-dependent manner via phosphorylation of its Tyr705 residue.

EP may be proven to be useful as an antitumor agent. Unlike other chemical compounds, EP is associated with no toxicity and has a good safety profile (16). EP is widely used as a food additive and is found in caramel, brandy, rum, chocolate and other food spices. Its lack of toxicity has been verified in several previous studies conducted on animal models. EP treatment in humans was also proven to be risk-free (48). Preclinical research has demonstrated that tumor cell sensitivity to other antitumor agents is significantly increased by inhibiting the release of HMGB1 via EP, and chemotherapy-related cytotoxicity is decreased by EP (49). This evidence strongly supports the use of EP as an adjuvant lung cancer therapy.

Moreover, human malignant mesothelioma (MM) arises from the malignant transformation of mesothelial cells in the pleura, peritoneum and pericardial cavity. In a recent study by Pellegrini et al, it was observed that HMGB1 targeting by EP inhibited the development of MM (39). EP was shown to suppress the viability, motility and migration of MM cell lines (REN, HP3 and PPM-MILL) by inhibiting the HMGB1/RAGE and NF-κB pathways. Furthermore, Pellegrini et al found that EP inhibited orthotopic tumor growth in MM xenografts. Similarly, in our present study, we found that EP suppressed the growth, invasion and migration and induced apoptosis of NSCLC cells via the HMGB1/RAGE axis and the NF-κB/STAT3 pathway. However, our findings were not tested in vivo. Taken together, the findings of Pellegrini et al (39) and those of the present study indicate that EP may be useful as an adjuvant treatment for thoracic malignancies.

Acknowledgements

Not applicable.

Funding

The present study was supported by the National Science Foundation (NNSF) of China (grant no. 81570194 to LJ).

Availability of data and materials

All the datasets generated and analyzed in the present study are included in this published manuscript.

Authors' contributions

QL, PW, LJ and YH conceived and designed the study. QL, HZ and JZ performed the experiments. QL, PW and YH wrote the manuscript. QL, YH, HZ, JZ and LJ reviewed and edited the manuscript. All authors read and approved the manuscript and agree to be accountable for all aspects of the research in ensuring that the accuracy or integrity of any part of the work are appropriately investigated and resolved.

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.

References

1 

Zou S, Pan X, Hua C, Wu M, He B and Chen Z: Myeloperoxidase-463 G/A polymorphism is associated with lung cancer risk: A meta-analysis with 7420 cases and 9132 controls. J Cancer Res Ther. 14 (Suppl):S282–S287. 2018. View Article : Google Scholar : PubMed/NCBI

2 

Ziebarth NR: Lung cancer risk perception biases. Prev Med. 110:16–23. 2018. View Article : Google Scholar : PubMed/NCBI

3 

Liu FT, Jia L, Wang P, Farren T, Li H, Hao X and Agrawal SG: CD126 and targeted therapy with tocilizumab in chronic lymphocytic leukemia. Clin Cancer Res. 22:2462–2469. 2016. View Article : Google Scholar : PubMed/NCBI

4 

Park BS, Jo HW, Park C, Huh Y, Jung J and Jeong NY: A novel effect of ethyl pyruvate in Schwann cell de-differentiation and proliferation during Wallerian degeneration. Anim Cells Syst. 19:262–268. 2015. View Article : Google Scholar

5 

Li X, Jiang S and Tapping RI: Toll-like receptor signaling in cell proliferation and survival. Cytokine. 49:1–9. 2010. View Article : Google Scholar : PubMed/NCBI

6 

Gruffaz M, Vasan K, Tan B, Ramos da Silva S and Gao SJ: TLR4-mediated inflammation promotes KSHV-induced cellular transformation and tumorigenesis by activating the STAT3 pathway. Cancer Res. 77:7094–7108. 2017. View Article : Google Scholar : PubMed/NCBI

7 

Su X, Wang H, Zhao J, Pan H and Mao L: Beneficial effects of ethyl pyruvate through inhibiting high-mobility group box 1 expression and TLR4/NF-κB pathway after traumatic brain injury in the rat. Mediators Inflamm. 2011:8071422012.

8 

Shao Y, Nanayakkara G, Cheng J, Cueto R, Yang WY, Park JY, Wang H and Yang X: Lysophospholipids and their receptors serve as conditional DAMPs and DAMP receptors in tissue oxidative and inflammatory injury. Antioxid Redox Signal. Apr 26–2017.(Epub ahead of print). doi: 10.1089/ars.2017.7069.

9 

Lin TJ, Lin HT, Chang WT, Mitapalli SP, Hsiao PW, Yin SY and Yang NS: Shikonin-enhanced cell immunogenicity of tumor vaccine is mediated by the differential effects of DAMP components. Mol Cancer. 14:1742015. View Article : Google Scholar : PubMed/NCBI

10 

Bierhaus A, Schiekofer S, Schwaninger M, Andrassy M, Humpert PM, Chen J, Hong M, Luther T, Henle T, Klöting I, et al: Diabetes-associated sustained activation of the transcription factor nuclear factor-kappaB. Diabetes. 50:2792–2808. 2001. View Article : Google Scholar : PubMed/NCBI

11 

Baldwin AS Jr: The NF-kappa B and I kappa B proteins: New discoveries and insights. Annu Rev Immunol. 14:649–683. 1996. View Article : Google Scholar : PubMed/NCBI

12 

Müller M, Morotti A and Ponzetto C: Activation of NF-kappaB is essential for hepatocyte growth factor-mediated proliferation and tubulogenesis. Mol Cell Biol. 22:1060–1072. 2002. View Article : Google Scholar : PubMed/NCBI

13 

Mantovani A, Allavena P, Sica A and Balkwill F: Cancer-related inflammation. Nature. 454:436–444. 2008. View Article : Google Scholar : PubMed/NCBI

14 

Grivennikov S, Karin E, Terzic J, Mucida D, Yu GY, Vallabhapurapu S, Scheller J, Rose-John S, Cheroutre H, Eckmann L and Karin M: IL-6 and Stat3 are required for survival of intestinal epithelial cells and development of colitis-associated cancer. Cancer Cell. 15:103–113. 2009. View Article : Google Scholar : PubMed/NCBI

15 

Shen Y, Devgan G, Darnell JE Jr and Bromberg JF: Constitutively activated Stat3 protects fibroblasts from serum withdrawal and UV-induced apoptosis and antagonizes the proapoptotic effects of activated Stat1. Proc Natl Acad Sci USA. 98:1543–1548. 2001. View Article : Google Scholar : PubMed/NCBI

16 

Pathak M, Mishra R, Agarwala PK, Ojha H, Singh B, Singh A and Kukreti S: Binding of ethyl pyruvate to bovine serum albumin: Calorimetric, spectroscopic and molecular docking studies. Thermochim Acta. 633:140–148. 2016. View Article : Google Scholar

17 

Cook VL, Holcombe SJ, Gandy JC, Corl CM and Sordillo LM: Ethyl pyruvate decreases proinflammatory gene expression in lipopolysaccharide-stimulated equine monocytes. Vet Immunol Immunopathol. 141:92–99. 2011. View Article : Google Scholar : PubMed/NCBI

18 

Wagner N, Dieteren S, Franz N, Köhler K, Mörs K, Nicin L, Schmidt J, Perl M, Marzi I and Relja B: Ethyl pyruvate ameliorates hepatic injury following blunt chest trauma and hemorrhagic shock by reducing local inflammation, NF-kappaB activation and HMGB1 release. PLoS One. 13:e0192171doi: 10.1371/journal.pone.0192171. eCollection. 2018. View Article : Google Scholar : PubMed/NCBI

19 

Wang FC, Xie Y and Zhou NJ: Ethyl pyruvate reduced h pylori- induced inflammation through inhibition of HMGB1/TLR4 pathways. J Gastroenterol Hepatol. 29:27. 2014.

20 

Shen HX, Hu XM, Liu C, Wang SP, Zhang WT, Gao H, Steder RA, Gao YQ and Chen J: Ethyl pyruvate protects against hypoxic-ischemic brain injury via anti-cell death and anti-inflammatory mechanisms. Neurobiol Dis. 37:711–722. 2010. View Article : Google Scholar : PubMed/NCBI

21 

Shen M, Lu J, Dai W, Wang F, Xu L, Chen K, He L, Cheng P, Zhang Y, Wang C, et al: Ethyl pyruvate ameliorates hepatic ischemia-reperfusion injury by inhibiting intrinsic pathway of apoptosis and autophagy. Mediators Inflamm. 2013:4615362013. View Article : Google Scholar : PubMed/NCBI

22 

Chakhtoura M, Chain R, Varghese L and Gallucci S: Ethyl pyruvate, an inhibitor of high-mobility group box 1 (HMGB1) release, modulates dendritic cell activation and survival (TRAN3P.897). J Immunol 192 (1 Suppl). S202–S236. 2014.

23 

Livak KJ and Schmittgen TD: Analysis of relative gene expression data using real-time quantitative PCR and the 2(-Delta Delta C(T)) method. Methods. 25:402–408. 2011. View Article : Google Scholar

24 

Ibrahim ZA, Armour CL, Phipps S and Sukkar MB: RAGE and TLRs: Relatives, friends or neighbours? Mol Immunol. 56:739–744. 2013. View Article : Google Scholar : PubMed/NCBI

25 

Han SH, Kim YH and Mook-Jung I: RAGE: The beneficial and deleterious effects by diverse mechanisms of actions. Mol Cells. 31:91–97. 2011. View Article : Google Scholar : PubMed/NCBI

26 

Perkins ND: Integrating cell-signalling pathways with NF-kappaB and IKK function. Nat Rev Mol Cell Biol. 8:49–62. 2007. View Article : Google Scholar : PubMed/NCBI

27 

Yu H, Pardoll D and Jove R: STATs in cancer inflammation and immunity: A leading role for STAT3. Nat Rev Cancer. 9:798–809. 2009. View Article : Google Scholar : PubMed/NCBI

28 

Hanahan D and Weinberg RA: Hallmarks of cancer: The next generation. Cell. 144:646–674. 2011. View Article : Google Scholar : PubMed/NCBI

29 

Actis M, Inoue A, Evison B, Perry S, Punchihewa C and Fujii N: Small molecule inhibitors of PCNA/PIP-box interaction suppress translesion DNA synthesis. Bioorg Med Chem. 21:1972–1977. 2013. View Article : Google Scholar : PubMed/NCBI

30 

Abike F, Tapisiz OL, Zergeroglu S, Dunder I, Temizkan O and Payasli A: PCNA and Ki-67 in endometrial hyperplasias and evaluation of the potential of malignancy. Eur J Gynaecol Oncol. 32:77–80. 2011.PubMed/NCBI

31 

Himelstein BP, Canete-Soler R, Bernhard EJ, Dilks DW and Muschel RJ: Metalloproteinases in tumor progression: The contribution of MMP-9. Invasion Metastasis. 14:246–258. 1994.PubMed/NCBI

32 

Morini M, Mottolese M, Ferrari N, Ghiorzo F, Buglioni S, Mortarini R, Noonan DM, Natali PG and Albini A: The alpha 3 beta 1 integrin is associated with mammary carcinoma cell metastasis, invasion, and gelatinase B (MMP-9) activity. Int J Cancer. 87:336–342. 2000. View Article : Google Scholar : PubMed/NCBI

33 

Farina AR and Mackay AR: Gelatinase B/MMP-9 in tumour pathogenesis and progression. Cancers (Basel). 6:240–296. 2014. View Article : Google Scholar : PubMed/NCBI

34 

Zhang L, Ma L, Yan T, Han X, Xu J, Xu J and Xu X: Activated mitochondrial apoptosis in hESCs after dissociation involving the PKA/p-p53/Bax signaling pathway. Exp Cell Res. 369:226–233. 2018. View Article : Google Scholar : PubMed/NCBI

35 

Yao TH, Pataer P, Regmi KP, Gu XW, Li QY, Du JT, Ge SM and Tu JB: Propranolol induces hemangioma endothelial cell apoptosis via a p53-BAX mediated pathway. Mol Med Rep. 18:684–694. 2018.PubMed/NCBI

36 

Youle RJ and Strasser A: The BCL-2 protein family: Opposing activities that mediate cell death. Nat Rev Mol Cell Biol. 9:47–59. 2008. View Article : Google Scholar : PubMed/NCBI

37 

Zhang J, Zhu JS, Zhou Z, Chen WX and Chen NW: Inhibitory effects of ethyl pyruvate administration on human gastric cancer growth via regulation of the HMGB1-RAGE and Akt pathways in vitro and in vivo. Oncol Rep. 27:1511–1519. 2012.PubMed/NCBI

38 

Birkenmeier G, Hemdan NYA, Kurz S, Bigl M, Pieroh P, Debebe T, Buchold M, Thieme R, Wichmann G and Dehghani F: Ethyl pyruvate combats human leukemia cells but spares normal blood cells. PLoS One. 11:e01615712016. View Article : Google Scholar : PubMed/NCBI

39 

Pellegrini L, Xue J, Larson D, Pastorino S, Jube S, Forest KH, Saad-Jube ZS, Napolitano A, Pagano I, Negi VS, et al: HMGB1 targeting by ethyl pyruvate suppresses malignant phenotype of human mesothelioma. Oncotarget. 8:22649–22661. 2017. View Article : Google Scholar : PubMed/NCBI

40 

Cheng P, Dai W, Wang F, Lu J, Shen M, Chen K, Li JJ, Zhang Y, Wang C, Yang J, et al: Ethyl pyruvate inhibits proliferation and induces apoptosis of hepatocellular carcinoma via regulation of the HMGB1-RAGE and AKT pathways. Biochem Biophys Res Commun. 443:1162–1168. 2014. View Article : Google Scholar : PubMed/NCBI

41 

Baunacke M, Horn LC, Trettner S, Engel KM, Hemdan NY, Wiechmann V, Stolzenburg JU, Bigl M and Birkenmeier G: Exploring glyoxalase 1 expression in prostate cancer tissues: Targeting the enzyme by ethyl pyruvate defangs some malignancy-associated properties. Prostate. 74:48–60. 2014. View Article : Google Scholar : PubMed/NCBI

42 

Li ML, Wang XF, Tan ZJ, Dong P, Gu J, Lu JH, Wu XS, Zhang L, Ding QC, Wu WG, et al: Ethyl pyruvate administration suppresses growth and invasion of gallbladder cancer cells via downregulation of HMGB1-RAGE axis. Int J Immunopathol Pharmacol. 25:955–965. 2012. View Article : Google Scholar : PubMed/NCBI

43 

Queisser MA, Kouri FM, Königshoff M, Wygrecka M, Schubert U, Eickelberg O and Preissner KT: Loss of RAGE in pulmonary fibrosis: Molecular relations to functional changes in pulmonary cell types. Am J Respir Cell Mol Biol. 39:337–345. 2008. View Article : Google Scholar : PubMed/NCBI

44 

Xu X, Zhu H, Wang T, Sun Y, Ni P, Liu Y, Tian S, Amoah Barnie P, Shen H, Xu W, et al: Exogenous high-mobility group box 1 inhibits apoptosis and promotes the proliferation of lewis cells via RAGE/TLR4-dependent signal pathways. Scand J Immunol. 79:386–394. 2014. View Article : Google Scholar : PubMed/NCBI

45 

Gilmore TD: Introduction to NF-kappaB: Players, pathways, perspectives. Oncogene. 25:6680–6684. 2006. View Article : Google Scholar : PubMed/NCBI

46 

Escárcega RO, Fuentes-Alexandro S, Garcia-Carrasco M, Gatica A and Zamora A: The transcription factor nuclear factor-kappa B and cancer. Clin Oncol (R Coll Radiol). 19:154–561. 2007. View Article : Google Scholar : PubMed/NCBI

47 

Yuan ZL, Guan YJ, Wang L, Wei W, Kane AB and Chin YE: Central role of the threonine residue within the p+1 loop of receptor tyrosine kinase in STAT3 constitutive phosphorylation in metastatic cancer cells. Mol Cell Biol. 24:9390–9400. 2004. View Article : Google Scholar : PubMed/NCBI

48 

Bennett-Guerrero E, Swaminathan M, Grigore AM, Roach GW, Aberle LG, Johnston JM and Fink MP: A phase II multicenter double-blind placebo-controlled study of ethyl pyruvate in high-risk patients undergoing cardiac surgery with cardiopulmonary bypass. J Cardiothorac Vasc Anesth. 23:324–329. 2009. View Article : Google Scholar : PubMed/NCBI

49 

Tang D, Kang R, Cheh CW, Livesey KM, Liang X, Schapiro NE, Benschop R, Sparvero LJ, Amoscato AA, Tracey KJ, et al: HMGB1 release and redox regulates autophagy and apoptosis in cancer cells. Oncogene. 29:5299–5310. 2010. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

August-2019
Volume 42 Issue 2

Print ISSN: 1021-335X
Online ISSN:1791-2431

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Liu Q, Huo Y, Zheng H, Zhao J, Jia L and Wang P: Ethyl pyruvate suppresses the growth, invasion and migration and induces the apoptosis of non‑small cell lung cancer cells via the HMGB1/RAGE axis and the NF‑κB/STAT3 pathway. Oncol Rep 42: 817-825, 2019
APA
Liu, Q., Huo, Y., Zheng, H., Zhao, J., Jia, L., & Wang, P. (2019). Ethyl pyruvate suppresses the growth, invasion and migration and induces the apoptosis of non‑small cell lung cancer cells via the HMGB1/RAGE axis and the NF‑κB/STAT3 pathway. Oncology Reports, 42, 817-825. https://doi.org/10.3892/or.2019.7176
MLA
Liu, Q., Huo, Y., Zheng, H., Zhao, J., Jia, L., Wang, P."Ethyl pyruvate suppresses the growth, invasion and migration and induces the apoptosis of non‑small cell lung cancer cells via the HMGB1/RAGE axis and the NF‑κB/STAT3 pathway". Oncology Reports 42.2 (2019): 817-825.
Chicago
Liu, Q., Huo, Y., Zheng, H., Zhao, J., Jia, L., Wang, P."Ethyl pyruvate suppresses the growth, invasion and migration and induces the apoptosis of non‑small cell lung cancer cells via the HMGB1/RAGE axis and the NF‑κB/STAT3 pathway". Oncology Reports 42, no. 2 (2019): 817-825. https://doi.org/10.3892/or.2019.7176