lncRNA PDIA3P regulates cell proliferation and invasion in non‑small cell lung cancer

  • Authors:
    • Xiaogang Yang
    • Baojun Yang
  • View Affiliations

  • Published online on: August 13, 2019     https://doi.org/10.3892/etm.2019.7882
  • Pages: 3184-3190
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Increasing evidence has shown that long non‑coding RNAs (lncRNAs) serve a critical role in tumor progression. The lncRNA protein disulfide isomerase family A member 3 pseudogene 1 (PDIA3P) is associated with the progression of oral squamous cell carcinoma and multiple myeloma. However, the roles of lncRNA PDIA3P in non‑small cell lung cancer (NSCLC) remain unknown. In the present study, it was demonstrated that lncRNA PDIAP3 expression was significantly increased in patients with NSCLC, particularly in patients with an advanced Tumor‑Node‑Metastasis stages and lymph‑node metastasis. The Gene Expression Profiling Interactive Analysis database revealed that patients with NSCLC with high lncRNA PDIA3P expression had a poorer disease‑free survival rate. Furthermore, functional assays suggested that PDIA3P inhibition significantly suppressed the proliferation and invasion of NSCLC cells in vitro and reduced tumor growth in vivo. In addition, lncRNA PDIA3P enhanced the activity of the Wnt/β‑catenin pathway in progression of NSCLC. The present study showed that lncRNA PDIA3P promoted NSCLC progression by regulating Wnt/β‑catenin signaling, highlighting a potential therapeutic target for treating patients with NSCLC treatment.

Introduction

Lung cancer is the leading cause of cancer-associated death worldwide, of which non-small-cell lung cancer (NSCLC) accounts for >80% of lung cancer cases (1,2). Although there have been significant advances in treatment methods, including surgery combined with radiotherapy and/or chemotherapy, as a result of a high rate of local recurrence and metastasis, the 5-year survival rate remains poor (3,4). Therefore, a better understanding of the underlying molecular mechanisms involved in NSCLC progression is required.

Long non-coding RNAs (lncRNAs) are a type of non-coding RNA >200 nucleotides in length, with limited or no coding potential (5,6). There are an increasing number of studies which have demonstrated that lncRNAs are involved in multiple cellular processes, including in cancer (7,8). For example, Zhang et al (9) demonstrated that that upregulation of lncRNA metastasis associated lung adenocarcinoma transcript 1 was associated with tumor progression and poor prognosis in clear cell renal cell carcinoma. Gao et al (10) demonstrated that ZNFX1 antisense RNA 1 exhibited an oncogenic role in glioma progression by regulating epithelial-mesenchymal transition (EMT) and the Notch signaling pathway. Chen et al (11) showed that lncRNA colon cancer associated transcript 1 promotes the progression of multiple myeloma by acting as a molecular sponge of microRNA (miR)-181a-5p, thus modulating the expression of homeobox A1.

Protein disulfide isomerase family A member 3 pseudogene 1 (PDIA3P1) is a 2,099-nucleotide lncRNA that is mapped to human chromosome 1q21.1. Sun et al (12) reported that lncRNA PDIA3P was upregulated and interacted with miR-185-5p to promote the proliferation of oral squamous cell carcinoma cells by targeting cyclin D2. Yang et al (13) showed that lncRNA PDIA3P interacted with c-myc to regulate cell proliferation by activating the pentose phosphate pathway in multiple myeloma. The aim of the present study was to reveal the functions of lncRNA PDIA3P in the progression of NSCLC.

Materials and methods

Tumor specimens

A total of 73 pairs of NSCLC tissues and the adjacent normal tissues were obtained from patients who received surgery at The Affiliated Hospital of Hebei University of Engineering (Handan, China) between January 2013 and December 2016. All of the specimens were immediately frozen in liquid nitrogen and stored at −80°C until they were used for RNA extraction. None of the patients received previous local or systemic treatment prior to the operation. Clinical information was obtained from the medical records of the patients. All the patients in the present study provided written informed consent. The present study was approved by The Ethics Committee of Hebei University of Engineering (approval no. HBU-2018-01127).

Cell culture and transfection

The human NSCLC cell lines A549, H1299, H1703, H520 and SK-MES-1, and the normal human bronchial epithelium cell line BEAS-2B were obtained from The Cell Bank of Type Culture Collection of the Chinese Academy of Sciences. All cells were cultured in RPMI-1640 medium (Gibco; Thermo Fisher Scientific, Inc.) supplemented with 10% fetal bovine serum (HyClone; GE Healthcare Life Sciences) at 37°C with 5% CO2. The concentrations of siRNA and plasmids were 50 µM and 2 µg/ml, respectively. Small interfering (si)-PDIA3P and the negative control (NC) were purchased from Shanghai GenePharma Co., Ltd. The pcDNA3.1-PDIA3P plasmid was prepared in our previous study using RNA interference sequences (12) and an empty pcDNA3.1 vector (Shanghai GenePharma Co., Ltd.) was used as a negative control. The Wnt pathway inhibitor IWR-1-endo was obtained from Cayman Chemical Company. Aliquots of 2 mM in DMSO were stored at −20°C and working concentrations (5 µM) were prepared prior to use. Lipofectamine® 2000 (Invitrogen; Thermo Fisher Scientific, Inc.) was used for transfection, according to the manufacturer's protocol (14). The sequences of si-PDIA3P were as follows: si-RNA-1, 5′-AACCACTGGGGAGGACTAGG-3′; si-RNA-2, 5′-TGGTAGCAGAGAATTTGAT-3′; and si-NC, 5′-AATTCTCCGAACGTGTCACGT-3′. Further experiments were completed 24 h following transfection.

Reverse transcription-quantitative (RT-q)PCR

Total RNAs were extracted from NSCLC tissues and cell lines using TRIzol® (Invitrogen; Thermo Fisher Scientific, Inc.) according to the manufacturer's protocol. Total RNA was reverse transcribed into cDNA using the PrimeScript RT reagent kit (Takara Bio, Inc.) according to the manufacturer's protocol. qPCR analysis was performed using a SYBR Green qPCR Master Mix kit (Promega Corporation), according to the manufacturer's instructions. The thermocycling conditions were as follows: Initial denaturation at 95°C for 5 min followed by 40 cycles at 95°C for 10 sec and 60°C for 1 min. Relative expression was normalized to GAPDH and calculated using the 2−ΔΔCq method (15). The sequences of the PCR primers were as follows: PDIA3P forward, 5′-AACCACTGGGGAGGACTAGG-3′ and reverse, 5′-CAGTGCAGCTAAGAAATGGCT-3′; and GAPDH forward, 5′-ATGGGGAAGGTGAAGGTCG-3′ and reverse, 5′-GGGTCATTGATGGCAACAATATC-3′.

Cell proliferation assay

A Cell Counting Kit-8 (Dojindo Molecular Technologies, Inc.) assay was performed to determine the proliferative rate of lung cancer cells. Briefly, cells were seeded into 96-well plates and cultured for the indicated times (24, 48 and 72 h). Subsequently, 10 µl CCK-8 solution was added and incubated for another 2 h at 37°C. The absorbance at 450 nm was determined using a microplate reader (Bio-Rad Laboratories, Inc.).

Colony formation assay

A total of 2 ml complete medium containing 2×103 transfected cells were added to 6-well plates and incubated for 2 weeks. Subsequently, the supernatant was discarded and the cells were washed with PBS, fixed using 500 µl methanol at room temperature for 20 min and stained with 0.1% crystal violet (Nanjing KeyGen Biotech Co., Ltd.) for 20 min at room temperature. The number of colonies >10 cells were counted using an optical light microscope at ×50 magnification (Olympus Corporation).

Cell invasion assay

The invasive capability of the cells was examined using a Transwell chamber assay with an 8-µM pore (EMD Millipore). Briefly, 1×104 cells were seeded in the upper chamber coated with Matrigel (Sigma-Aldrich; Merck KGaA) and incubated for 48 h. Cells which had not invaded were removed using a swab and the cells which had invaded through to the lower surface were fixed with methanol for 35 min at room temperature, stained with crystal violet for 50 min at room temperature, washed with PBS and counted under a light microscope (magnification, ×50; Nikon Corporation).

Animal experiments

The transfected A549 cells were subcutaneously injected into 4 week-old female BALB/c nude mice (Beijing Experimental Animal Research Center, Beijing, China). Each group included 4 mice, and they were housed under specific pathogen free conditions at 20–26°C, 40–70% humidity and a 12/12 h light/dark cycle. The mice had free access to food and water. At the end of the 7-week observation period, the mice were sacrificed by cervical dislocation, and the tumor tissues were removed for subsequent experiments (16). All experimental procedures were approved by the committee on animal experimentation of Hebei University of Engineering.

Western blotting

Proteins were lysed using RIPA buffer (Beyotime Institute of Biotechnology) and the concentrations were measured using a bicinchoninic acid protein assay (Beyotime Institute of Biotechnology). Proteins (20 µg/per lane) were separated using a 10% SDS-PAGE gel and transferred to a PVDF membrane (EMD Millipore). After incubation with antibodies against β-catenin (1:1,000; cat. no. ab32572), c-myc (1:1,000; cat. no. ab32072), glycogen synthase kinase (GSK)-3β (1:1,000; cat. no. ab32391) and GAPDH (1:5,000; cat. no. ab181602) (all from Abcam) overnight at 4°C, the membranes were incubated with a goat anti-rabbit horse radish peroxidase-conjugated secondary antibody (1:5,000; cat. no. ab97051; Abcam) at room temperature for 1 h. The signals were visualized using an enhanced chemiluminescence reagent (EMD Millipore). Protein bands were visualized using ImageJ 1.48 software (National Institutes of Health).

Clinical databases

The online database Gene Expression Profiling Interactive Analysis (GEPIA; http://gepia.cancerpku.cn/index.html.) was used to analyze the RNA sequencing expression data relevant to the present study based on The Cancer Genome Atlas and the Genotype-Tissue Expression databases (17). GEPIA performs survival analyses based on gene expression levels and uses a log-rank test for hypothesis evaluation.

Immunohistochemistry

The expression of Ki-67 in nude mice injected with A549 cells was measured using immunohistochemistry. The immunohistochemistry assay was conducted according to previous report (18). The sections were treated with rabbit polyclonal anti-Ki-67 antibody (1:100; cat. no. ab833; Abcam) at 4°C overnight. After successfully completing the previous steps, the sections were incubated with a secondary goat anti-rabbit antibody (1:1,000; cat. no. ab6721; Abcam) for 20 min at 37°C. Image acquisition was performed by light microscope (magnification, ×100; Nikon Corporation) and Image-Pro Plus version 6.0 (Media Cybernetics, Inc.) was used to analyze the integrated optical density values of the brown area.

Statistical analysis

All data were analyzed using SPSS 18.0 software (SPSS, Inc.) and are expressed as the mean ± SD from at least three independent experiments. The differences between the two groups were evaluated using a Student's t-test or one-way ANOVA followed by a Bonferroni post hoc test. P<0.05 was considered to indicate a statistically significant difference.

Results

lncRNA PDIA3P is upregulated in NSCLC

In the present study, PDIA3P expression in NSCLC tissues was examined. RT-qPCR analysis showed that PDIA3P expression was significantly higher in NSCLC tissues (Fig. 1A; P<0.05) compared with the control. High PDIA3P expression levels were inversely associated with TNM stages III and IV, and the presence of lymph node metastasis (Fig. 1B and C; Table I; P<0.05). Furthermore, data from the GEPIA database showed that PDIA3P expression was increased in NSCLC (lung adenocarcinoma and lung squamous cell carcinoma) tissues compared with normal tissues (Fig. 1D; P<0.05). In addition, high PDIA3P expression was associated with advanced tumor stage and poor disease-free survival of patients with NSCLC (Fig. 1E and F; P<0.05).

Table I.

Associations between PDIA3P expression and clinical features of patients with NSCLC.

Table I.

Associations between PDIA3P expression and clinical features of patients with NSCLC.

PDIA3P expression

CharacteristicsHigh, n=37Low, n=36P-value
Age, years 0.415
  >602117
  ≤601619
Sex 0.736
  Male2220
  Female1516
TNM stage 0.003a
  I/II1022
  III/IV2714
Tumor size, cm 0.295
  >32318
  ≤31418
Lymph node metastasis 0.009a
  Negative1929
  Positive187

a P<0.01. TNM, Tumor-Node-Metastasis.

lncRNA PDIA3P-silencing suppresses the proliferation and invasion of NSCLC cells

To determine the biological functions of PDIA3P in NSCLC progression, the expression levels of PDIA3P were first determined in NSCLC cell lines (A549, H1299, H1703, H520 and SK-MES-1) and the normal human bronchial epithelium cell line BEAS-2B, and A549 and H520 cell lines were selected for further study as they exhibited high PDIA3P expression. RT-qPCR analysis showed that PDIA3P expression was significantly increased in all the NSCLC cell lines compared with BEAS-2B cells (Fig. 2A; P<0.05). Therefore, PDIA3P was silenced in both A549 and H520 cells by si-PDIA3P (Fig. 2B; P<0.05). A CCK-8 assay showed that PDIA3P knockdown decreased the proliferative rate of A549 and H520 cells compared with the negative control (Fig. 2C; P<0.05), and the cell colony formation was significantly decreased following PDIA3P silencing in A549 and H520 cells compared with the negative control (Fig. 2D; P<0.05). Furthermore, the migratory and invasive ability of A549 and H520 cells was reduced following silencing of PDIA3P compared with the control (Fig. 2E and F; P<0.05).

lncRNA PDIA3P silencing reduces tumor growth in vivo

The effects of PDIA3P on NSCLC growth in vivo were determined. The results showed that PDIA3P inhibition significantly reduced the tumor volume compared with the negative control (Fig. 3A; P<0.05). At 7 weeks after the injection, the mice were sacrificed, and tumor weight was determined. The results showed that PDIA3P silencing reduced the tumor weight (Fig. 3B; P<0.05). Furthermore, immunohistochemical staining showed that the Ki67 expression was decreased in PDIA3P-silenced xenograft tumor tissues (Fig. 3C; P<0.05).

lncRNA PDIA3P activates the Wnt/β-catenin signaling pathway

To determine the underlying mechanism by which PDIA3P affects NSCLC progression, RT-qPCR and western blotting were performed to examine the effects of PDIA3P on the Wnt/β-catenin pathway, which is frequently aberrantly activated in human cancer (19). The results showed that PDIA3P inhibition decreased both the mRNA and protein expression levels of β-catenin and c-myc, and increased the expression levels of GSK-3β in A549 cells (Fig. 4A and B; P<0.05). A549 cell line was used as it exhibited the highest expression. PDIA3P overexpression upregulated β-catenin and c-myc expression, and decreased GSK-3β expression in A549 cells, both at the mRNA and protein level (Fig. 4C-E; P<0.05). Furthermore, the Wnt pathway inhibitor IWR-1-endo partly reversed the effects of PDIA3P expression on proliferation (Fig. 4F; P<0.05) and invasion (Fig. 4G; P<0.05) in A549 cells. Therefore, PDIA3P may promote the progression of NSCLC, at least partly by regulating the Wnt/β-catenin pathway.

Discussion

There has been an increase in the number of studies demonstrating the potential roles of various lncRNAs as key regulators of cancer progression over the past decade (2022), and genetic and epigenetic alterations of lncRNAs may serve key roles in tumorigenesis (23). Furthermore, abnormal expression of lncRNAs may serve important roles in cancer progression, including in NSCLC. For example, Xie et al (24) showed that the lncRNA gastric cancer associated transcript 2 is downregulated and associated with poor prognosis in patients with NSCLC. Cui et al (25) found that upregulation of the lncRNA small nucleolar RNA host gene 1 contributes to NSCLC progression via inhibition of miR-101-3p and activation of the Wnt/β-catenin signaling pathway. Gao et al (26) found that lncRNA FLVCR1 divergent transcript contributes to proliferation and invasion by sponging miR-573 to upregulate E2F transcription factor 3 expression in lung cancer.

In the present study, the function and underlying mechanisms of PDIA3P expression in NSCLC progression were determined. The results showed that PDIA3P expression was significantly increased in NSCLC tissues compared with adjacent non-tumor tissues. High PDIA3P expression was associated with advanced TNM stage cancer, lymph-node metastasis of cancer and poor disease-free survival of patients with NSCLC. PDIA3P functions in lung cancer were examined both in vitro and in vivo. The results showed that PDIA3P knockdown significantly inhibited the growth of NSCLC cells both in vitro and in vivo. Similarly, wound healing assays and Transwell assays showed that PDIA3P inhibition decreased the migration and invasion of lung cancer cells in vitro. In a further study the effects of PDIA3P on proliferation markers, such as proliferating cell nuclear antigen, and migration markers, such as matrix metalloproteinases, will be determined. Taken together, the results of the present study showed that PDIA3P may act as an oncogenic lncRNA in the progression of NSCLC.

The Wnt/β-catenin pathway is frequently activated in a wide range of different types of cancer, and is known to promote tumor invasion and metastasis through upregulation of factors regulating EMT (27,28). Recent studies demonstrated that certain lncRNAs affect the Wnt/β-catenin signaling pathway and thus regulate cancer progression. For example, Ma et al (29) showed that the lncRNA CCAL regulates the progression of colorectal cancer by activating the Wnt/β-catenin pathway through suppression of activator protein 2α. Zhao et al (30) demonstrated that upregulation of the lncRNA HNF1A antisense RNA 1 promotes cell proliferation and metastasis in osteosarcoma through activation of the Wnt/β-catenin pathway. However, the association between PDIA3P expression and the Wnt/β-catenin pathway in NSCLC remains unclear. In the present study, it was demonstrated that PDIA3P inhibition significantly reduced β-catenin and c-myc expression, and increased GSK-3β expression in NSCLC cells, while ectopic PDIA3P expression resulted in the opposite effects. In addition, in vitro functional assays showed that IWR-1 endo (Wnt pathway inhibitor) (31) attenuated the effects of PDIA3P on the proliferation and invasive ability of NSCLC cells, suggesting that PDIA3P promotes NSCLC progression at least partly through the Wnt/β-catenin pathway.

There are certain limitations to the present study. The effects of IWR-1 alone on b-catenin, or on cell viability and invasion ability, were not assessed.

In conclusion, PDIA3P is significantly increased in NSCLC, and promotes the proliferation and invasion of NSCLC by regulating the Wnt/β-catenin pathway. Future experiments should examine the effects of PDIA3P on EMT. The findings of the present study indicated that PDIA3P may serve as a potential therapeutic target for the treatment of patients with NSCLC.

Acknowledgements

Not applicable.

Funding

No funding was received.

Availability of data and materials

The datasets used and/or analyzed during the present study are available from the corresponding author on reasonable request.

Authors' contributions

BY designed the current study. XY and BY performed the experiments and analyzed the data. BY drafted the manuscript. XY and BY wrote the manuscript. All authors read and approved the final manuscript.

Ethics approval and consent to participate

All the patients in the present study provided written informed consent. The present study was approved by The Ethics Committee of Hebei University of Engineering (approval no. HBU-2018-01127). All experimental procedures involving animals were approved by the committee on animal experimentation of Hebei University of Engineering.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Torre LA, Bray F, Siegel RL, Ferlay J, Lortet-Tieulent J and Jemal A: Global cancer statistics, 2012. CA Cancer J Clin. 65:87–108. 2015. View Article : Google Scholar : PubMed/NCBI

2 

Chen W, Zheng R, Baade PD, Zhang S, Zeng H, Bray F, Jemal A, Yu XQ and He J: Cancer statistics in China, 2015. CA Cancer J Clin. 66:115–132. 2016. View Article : Google Scholar : PubMed/NCBI

3 

Dela Cruz CS, Tanoue LT and Matthay RA: Lung cancer: Epidemiology, etiology, and prevention. Clin Chest Med. 32:605–644. 2011. View Article : Google Scholar : PubMed/NCBI

4 

Didkowska J, Wojciechowska U, Mańczuk M and Łobaszewski J: Lung cancer epidemiology: Contemporary and future challenges worldwide. Ann Transl Med. 4:1502016. View Article : Google Scholar : PubMed/NCBI

5 

Mattick JS and Makunin IV: Non-coding RNA. Hum Mol Genet. 15:R17–R29. 2006. View Article : Google Scholar : PubMed/NCBI

6 

Fatica A and Bozzoni I: Long non-coding RNAs: New players in cell differentiation and development. Nat Rev Genet. 15:7–21. 2014. View Article : Google Scholar : PubMed/NCBI

7 

Spizzo R, Almeida MI, Colombatti A and Calin GA: Long non-coding RNAs and cancer: A new frontier of translational research. Oncogene. 31:4577–4587. 2012. View Article : Google Scholar : PubMed/NCBI

8 

Qi P and Du X: The long non-coding RNAs, a new cancer diagnostic and therapeutic gold mine. Mod Pathol. 26:155–165. 2013. View Article : Google Scholar : PubMed/NCBI

9 

Zhang HM, Yang FQ, Chen SJ, Che J and Zheng JH: Upregulation of long non-coding RNA MALAT1 correlates with tumor progression and poor prognosis in clear cell renal cell carcinoma. Tumor Biol. 36:2947–2955. 2015. View Article : Google Scholar

10 

Gao K, Ji Z, She K, Yang Q and Shao L: Long non-coding RNA ZFAS1 is an unfavourable prognostic factor and promotes glioma cell progression by activation of the Notch signaling pathway. Biomed Pharmacother. 87:555–560. 2017. View Article : Google Scholar : PubMed/NCBI

11 

Chen L, Hu N, Wang C, Zhao H and Gu Y: Long non-coding RNA CCAT1 promotes multiple myeloma progression by acting as a molecular sponge of miR-181a-5p to modulate HOXA1 expression. Cell Cycle. 17:319–329. 2018. View Article : Google Scholar : PubMed/NCBI

12 

Sun CC, Zhang L, Li G, Li SJ, Chen ZL, Fu YF, Gong FY, Bai T, Zhang DY, Wu QM and Li DJ: The lncRNA PDIA3P interacts with miR-185-5p to modulate oral squamous cell carcinoma progression by targeting cyclin D2. Mol Ther-Nucleic Acids. 9:100–110. 2017. View Article : Google Scholar : PubMed/NCBI

13 

Yang X, Ye H, He M, Zhou X, Sun N, Guo W, Lin X, Huang H, Lin Y, Yao R and Wang H: LncRNA PDIA3P interacts with c-Myc to regulate cell proliferation via induction of pentose phosphate pathway in multiple myeloma. Biochem Biophys Res Commun. 498:207–213. 2018. View Article : Google Scholar : PubMed/NCBI

14 

Yang FQ, Zhang HM, Chen SJ, Yan Y and Zheng JH: MiR-506 is down-regulated in clear cell renal cell carcinoma and inhibits cell growth and metastasis via targeting FLOT1. PLoS One. 10:e01202582015. View Article : Google Scholar : PubMed/NCBI

15 

Ou L, Wang D, Zhang H, Yu Q and Hua F: Decreased expression of MiR-138-5p by LncRNA H19 in cervical cancer promotes tumor proliferation. Oncol Res. 26:401–410. 2018. View Article : Google Scholar : PubMed/NCBI

16 

Gu Y, Xiao X and Yang S: LncRNA MALAT1 acts as an oncogene in multiple myeloma through sponging miR-509-5p to modulate FOXP1 expression. Oncotarget. 8:101984–101993. 2017. View Article : Google Scholar : PubMed/NCBI

17 

Tang Z, Li C, Kang B, Gao G, Li C and Zhang Z: GEPIA: A web server for cancer and normal gene expression profiling and interactive analyses. Nucleic Acids Res. 45:W98–W102. 2017. View Article : Google Scholar : PubMed/NCBI

18 

Tu J, Yu Y, Liu W and Chen S: Significance of human epidermal growth factor receptor 2 expression in colorectal cancer. Exp Ther Med. 9:17–24. 2015. View Article : Google Scholar : PubMed/NCBI

19 

Pridgeon MG, Grohar PJ, Steensma MR and Williams BO: Wnt signaling in ewing sarcoma, osteosarcoma, and malignant peripheral nerve sheath tumors. Curr Osteoporosis Rep. 15:239–246. 2017. View Article : Google Scholar

20 

Yang G, Lu X and Yuan L: LncRNA: A link between RNA and cancer. Biochim Biophys Acta. 1839:1097–1109. 2014. View Article : Google Scholar : PubMed/NCBI

21 

Schmitt AM and Chang HY: Long noncoding RNAs in cancer pathways. Cancer Cell. 29:452–463. 2016. View Article : Google Scholar : PubMed/NCBI

22 

Peng WX, Koirala P and Mo Y: LncRNA-mediated regulation of cell signaling in cancer. Oncogene. 36:5661–5667. 2017. View Article : Google Scholar : PubMed/NCBI

23 

Tang Q, Ni Z, Cheng Z, Xu J, Yu H and Yin P: Three circulating long non-coding RNAs act as biomarkers for predicting NSCLC. Cell Physiol Biochem. 37:1002–1009. 2015. View Article : Google Scholar : PubMed/NCBI

24 

Xie X, Liu HT, Mei J, Ding FB, Xiao HB, Hu FQ, Hu R and Wang MS: LncRNA HMlincRNA717 is down-regulated in non-small cell lung cancer and associated with poor prognosis. Int J Clin Exp Pathol. 7:8881–8886. 2014.PubMed/NCBI

25 

Cui Y, Zhang F, Zhu C, Geng L, Tian T and Liu H: Upregulated lncRNA SNHG1 contributes to progression of non-small cell lung cancer through inhibition of miR-101-3p and activation of Wnt/β-catenin signaling pathway. Oncotarget. 8:17785–17794. 2017.PubMed/NCBI

26 

Gao X, Zhao S, Yang X, Zang S and Yuan X: Long non-coding RNA FLVCR1-AS1 contributes to the proliferation and invasion of lung cancer by sponging miR-573 to upregulate the expression of E2F transcription factor 3. Biochem Biophys Res Commun. 505:931–938. 2018. View Article : Google Scholar : PubMed/NCBI

27 

MacDonald BT, Tamai K and He X: Wnt/Beta-catenin signaling: Components, mechanisms, and diseases. Dev Cell. 17:9–26. 2009. View Article : Google Scholar : PubMed/NCBI

28 

Clevers H and Nusse R: Wnt/β-catenin signaling and disease. Cell. 149:1192–1205. 2012. View Article : Google Scholar : PubMed/NCBI

29 

Ma Y, Yang Y, Wang F, Moyer MP, Wei Q, Zhang P, Yang Z, Liu W, Zhang H, Chen N, et al: Long non-coding RNA CCAL regulates colorectal cancer progression by activating Wnt/β-catenin signalling pathway via suppression of activator protein 2α. Gut. 65:1494–1504. 2016. View Article : Google Scholar : PubMed/NCBI

30 

Zhao H, Hou W, Tao J, Zhao Y, Wan G, Ma C and Xu H: Upregulation of lncRNA HNF1A-AS1 promotes cell proliferation and metastasis in osteosarcoma through activation of the Wnt/β-catenin signaling pathway. Am J Transl Res. 8:3503–3512. 2016.PubMed/NCBI

31 

Huang SM, Mishina YM, Liu S, Cheung A, Stegmeier F, Michaud GA, Charlat O, Wiellette E, Zhang Y, Wiessner S, et al: Tankyrase inhibition stabilizes axin and antagonizes Wnt signalling. Nature. 461:614–620. 2009. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

October-2019
Volume 18 Issue 4

Print ISSN: 1792-0981
Online ISSN:1792-1015

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Yang X and Yang X: lncRNA PDIA3P regulates cell proliferation and invasion in non‑small cell lung cancer. Exp Ther Med 18: 3184-3190, 2019
APA
Yang, X., & Yang, X. (2019). lncRNA PDIA3P regulates cell proliferation and invasion in non‑small cell lung cancer. Experimental and Therapeutic Medicine, 18, 3184-3190. https://doi.org/10.3892/etm.2019.7882
MLA
Yang, X., Yang, B."lncRNA PDIA3P regulates cell proliferation and invasion in non‑small cell lung cancer". Experimental and Therapeutic Medicine 18.4 (2019): 3184-3190.
Chicago
Yang, X., Yang, B."lncRNA PDIA3P regulates cell proliferation and invasion in non‑small cell lung cancer". Experimental and Therapeutic Medicine 18, no. 4 (2019): 3184-3190. https://doi.org/10.3892/etm.2019.7882