Open Access

Clinical significance and effect of lncRNA BBOX1‑AS1 on the proliferation and migration of lung squamous cell carcinoma

  • Authors:
    • Yu Zhang
    • Xiao Wang
    • Xian-Kui Cheng
    • Yuan-Yuan Zong
    • Rong-Quan He
    • Gang Chen
    • Ye-Jun Qin
  • View Affiliations

  • Published online on: November 12, 2021     https://doi.org/10.3892/ol.2021.13135
  • Article Number: 17
  • Copyright: © Zhang 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

Long non‑coding RNAs (lncRNAs) have a role in the occurrence and development of lung squamous cell carcinoma (LUSC). lncRNA γ‑butyrobetaine hydroxylase 1 (BBOX1)‑antisense 1 (AS1) may contribute to disease development. However, there are no studies on the role of BBOX1‑AS1 in LUSC to date. In the present study, an in‑house gene microarray analysis was performed to detect the differentially expressed lncRNAs and mRNAs between three pairs of LUSC and normal lung tissues. Only one lncRNA, BBOX1‑AS1, was differentially expressed in the in‑house microarray and The Cancer Genome Atlas (TCGA), Gene Expression Omnibus (GEO) and ArrayExpress databases. Reverse transcription‑quantitative PCR (RT‑qPCR) was then performed and the original RNA‑sequencing data from the TCGA, GEO and ArrayExpress datasets were used to determine the expression and clinical value of BBOX1‑AS1 in LUSC. In addition, a Cell Counting Kit‑8 assay, cell cycle analysis and scratch assay were performed to explore whether BBOX1‑AS1 expression affected the proliferation and migration of LUSC cells in vitro. The results of the RT‑qPCR analysis and data obtained from the TCGA database, GEO datasets, in‑house gene microarray and standard mean deviation analysis all supported the upregulated expression level of BBOX1‑AS1 in LUSC. Furthermore, silencing of BBOX1‑AS1 inhibited the proliferation and migration of LUSC cells according to in vitro assays. In addition, the cells were arrested in S‑phase after knockdown of BBOX1‑AS1. In conclusion, the expression level of BBOX1‑AS1 was upregulated in LUSC tissues. BBOX1‑AS1 may exert an oncogenic effect on LUSC by regulating various biological functions. However, additional functional experiments should be performed to verify the exact mechanism.

Introduction

Lung cancer is one of the most common and lethal cancer types with a high rate of mortality worldwide (1,2). Lung cancer accounts for 14% of new cancer cases and has a 26% mortality rate in males; for females, it accounts for 13% of new cancer cases and has a 25% mortality rate (3). Non-small-cell lung cancer (NSCLC) is the primary subtype of lung cancer and is composed of two major histologic groups: Lung squamous cell carcinoma (LUSC) and lung adenocarcinoma. Treatment strategies, including surgical resection, radiotherapy and chemotherapy, have developed rapidly in recent years, but the overall survival of patients with lung cancer remains poor (4,5). Biomarkers that may be used to detect or diagnose LUSC, improve the survival rate and reduce recurrence are lacking (6). Therefore, it is necessary to develop novel therapeutic strategies for LUSC.

Long non-coding RNAs (lncRNAs) range from 200 nucleotides to 100 kb in length and are a class of non-protein-coding RNAs (7,8). lncRNAs are known to have various roles in disease development, regulation of metabolism, epigenetic gene control and transcriptional regulation (9,10). Over the past decade, lncRNAs have been determined to be widely involved in the proliferation, apoptosis, invasion and metastasis of malignancies (11,12). Increasing evidence in lung cancer has indicated that dysregulated lncRNAs may affect the development and occurrence of lung cancer by modifying the biological functions and self-renewal abilities of cancer cells (1214). Furthermore, it was reported that dysregulated lncRNAs have a role in the drug resistance of lung cancer (15,16).

lncRNA γ-butyrobetaine hydroxylase 1 (BBOX1)-antisense 1 (AS1) [National Center of Biotechnology Information (NCBI) Gene ID: 103695435] is located on chromosome 11p14.2-p14.1. As previously reported, BBOX1-AS1 has a role in the development of various diseases, including NSCLC, colorectal cancer, cervical cancer and gastric cancer (1720). BBOX1-AS1 was overexpressed in NSCLC and in colorectal, cervical and gastric cancers. BBOX1-AS1 may also facilitate the proliferation, invasion and migration of these cancer cells (1720). However, the functions of BBOX1-AS1 in LUSC have yet to be studied independently.

In the present study, the clinical significance and effect of the differential expression of BBOX1-AS1 in LUSC was investigated. In vitro assays and in silico analyses were performed to examine the biological functions and probable mechanisms of BBOX1-AS1 in LUSC. A flow diagram of the present study is presented in Fig. 1.

Materials and methods

Patient tissue samples

A total of three pairs of LUSC tissues and normal lung tissues obtained >5 cm from the edge of the LUSC lesion were collected between August 2017 and October 2017 from the Department of Pathology of the First Affiliated Hospital of the Guangxi Medical University (Nanning, China). The patients' age ranged from 51 to 58 years. All three patients were definitively diagnosed with LUSC and underwent surgical resection without any further treatment. The pathological diagnoses were independently performed by at least two blinded pathologists. All methods used adhered to the relevant guidelines and were approved by the Ethical Committee of the First Affiliated Hospital of Guangxi Medical University (Nanning, China). Consent forms for the use of tissues were signed by the doctors and patients involved in the study.

In-house microarray analysis

An in-house microarray analysis was performed to detect differentially expressed lncRNAs between LUSC and normal lung tissues. Microarray hybridization and sample analysis were performed by Kangchen Biotech. The detailed methods were performed as previously described by Zhang et al (21). The significantly differentially expressed lncRNAs between LUSC and normal lung tissues were selected using the criteria of |fold change| ≥2 and P≤0.05. Similarly, the differentially expressed mRNAs were identified using microarray analysis.

Validation of the expression and clinical role of BBOX1-AS1

Expression analysis in clinical samples was performed using the original RNA sequencing (RNA-seq) data from The Cancer Genome Atlas (TCGA, cancergenome.nih.gov) database to determine BBOX1-AS1 expression between LUSC and noncancerous lung tissues. The LUSC cohort comprised 502 LUSC cases and 49 corresponding noncancerous lung samples. The original expression data were normalized using the R package DESeq. Differences in the expression of BBOX1-AS1 between LUSC tissues and normal lung tissues were identified from the TCGA dataset (22,23). The relationships of BBOX-AS1 expression and clinicopathological parameters (age, sex, ethnicity, tumor stage, regional lymph nodes, distant metastasis and stage) were determined from the TCGA data. A receiver operating characteristic (ROC) curve was created to determine the clinical value of BBOX1-AS1 expression in distinguishing normal and LUAD tissue. Gene Expression Omnibus (GEO, http://www.ncbi.nlm.nih.gov/geo/) data and the ArrayExpress (ebi.ac.uk/arrayexpress/) database were used to verify BBOX1-AS1 expression in LUSC. Kaplan-Meier curve analysis was used to assess BBOX1-AS1 expression and the 5-year survival rates of patients with LUSC with an endpoint of death or the end of the study period.

The LUSC-related BBOX1-AS1 microarray and RNA-seq datasets were downloaded from GEO, ArrayExpress, the TCGA, Sequence Read Archive (SRA, http://trace.ncbi.nlm.nih.gov/Traces/sra/) and Oncomine (https://www.oncomine.org/). Publications related to BBOX1-AS1 in LUSC were also selected from 12 online databases: PubMed, EMBASE, Web of Science, Science Direct, Cochrane Central Register of Controlled Trials, Google Scholar, Ovid, Chinese CNKI, LILACS, Wiley Online Library, Wan Fang, Chong Qing VIP and China Biology Medicine disc. The retrieval date was set to September 15, 2020, with the following Boolean search terms: (lung OR pulmonary OR ‘NSCLC’ OR ‘LC’) AND (‘lncRNA’ OR ‘noncoding RNA’ OR ‘noncoding RNA’ OR ‘noncoding RNA’ OR gene). Two different investigators (YZ and XW) cross-checked the literature. A group discussion was used to resolve any disagreement. The number of false positives, true positives, false negatives and true negatives was extracted as previously reported (24,25). The standard mean deviation (SMD) and summary receiver operating characteristic (SROC) curve of BBOX1-AS1 in LUSC were presented in Stata 14.0 (Stata Corp.). A fixed-effects model was used to analyze the pooled SMD to determine the expression of BBOX1-AS1 in LUSC compared to that in the normal group. The funnel plot used to evaluate the publication bias. The asymmetry of the funnel plot could display the validity of conclusions.

Cell culture and transfection with small interfering (si)RNA

Three human LUSC cell lines (NCI-H1703, NCI-H226 and SK-MES-1) and human bronchial epithelial cells (BEAS-2B) were obtained from the American Type Culture Collection. These four cell lines were cultured in DMEM with 10% heat-inactivated fetal bovine serum (Invitrogen; Thermo Fisher Scientific, Inc.). All of the cells were incubated in a humidified atmosphere with 5% CO2 at 37°C.

RNA interference (RNAi) was performed to determine the underlying effect of BBOX1-AS1 on biological processes in LUSC. The lentiviral vector containing siRNA targeting BBOX1-AS1 was synthesized by GeneChem (sense, 5′-CTGCTTTGCTCTTCAGACTTATT-3′; antisense, 5′-TAAGTCTGAAGAGCAAAGCAG-3′). The siRNA vectors used to knock down the BBOX1-AS1 gene (KD) and the negative siRNA controls (NC; cat. no. CON077, GeneChem) were transfected into the NCI-H1703 and SK-MES-1 cell lines using Lipofectamine 2000® (Thermo Fisher Scientific, Inc.) when the cells reached 80% confluency. Puromycin (2 µg/ml; Corning, Inc.; cat. no. 21-031-CVR) was applied for filtering the stable cell lines at 48 h after transfection. A fluorescence microscope was used to observe cell growth after transfection.

Reverse transcription-quantitative PCR (RT-qPCR)

Total RNA from NCI-H1703, NCI-H266, SK-MES-1 and BEAS-2B cells was isolated using TRIzol reagent (Shanghai Pufei Biotech Co., Ltd.) and reverse-transcribed using a Promega M-MLV Kit (Shanghai Pufei Biotech Co., Ltd.). qPCR was performed using the LightCycler 480 Real-time PCR System (Roche). The specific primers were synthesized as follows: BBOX1-AS1 forward, 5′-GATGGGCACATTTGGAAGTT-3′ and reverse, 5′-CAGCGTTAGGTTTGGAGTTG-3′; and GAPDH (internal control) forward, 5′-TGACTTCAACAGCGACACCCA-3′ and reverse, 5′-CACCCTGTTGCTGTAGCCAAA-3′. All experiments were performed in triplicate. The levels of BBOX1-AS1 expression were standardized to those of GAPDH and calculated using the delta Cq method (26). Furthermore, the transfection efficiency of the lentiviral vector containing BBOX1-AS1 siRNA was detected via RT-qPCR.

Cell Counting Kit-8 (CCK-8) assay

A CCK-8 assay was performed to examine the proliferation of LUSC cells in two different groups (KD group and NC group). Cell viability was determined with a CCK-8 (Beyotime Institute of Biotechnology) following the manufacturer's instructions. The detailed methods were performed as previously described by Zhang et al (27). The viability of NCI-H1703 and SK-MES-1 cells at days one, two, three, four and five was assessed and the absorbance at 450 nm was read using a microplate reader (Infinite M200 Micro Plate Reader; Tecan Group). The fold change in the optical density at 450 nm was considered to represent changes in cell viability. Experiments were performed in triplicate.

Flow cytometric cell cycle analysis

Flow cytometry was used to measure the effect of BBOX1-AS1 on the cell cycle distribution. The detailed method of the flow cytometric cell cycle assay was previously described by Zhang et al (27). Cells in the different phases of the cell cycle (G1, S and G2) were identified using FACSCalibur (EMD Millipore) and data analysis was performed using ModFit 3.2.1 (Verity Software House, Inc.). Each test was performed in triplicate.

Scratch assay

A scratch assay was performed to observe the potential effect of BBOX1-AS1-siRNA on the migration of LUSC cells. The detailed method of the scratch assay was performed as previously described by Zhang et al (27,28). Several random fields of the monolayer were captured directly under a microscope at 0 and 24 h after scratching. All experiments were performed in triplicate.

Potential pathways and functions associated with BBOX1-AS1

Genes associated with BBOX1-AS1 in LUSC were subjected to Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analyses to determine fundamental biological functions and downstream pathways (2931). The genes associated with BBOX1-AS1 in the GEO datasets were determined using the Psych R programming package. The co-expressed genes in the TCGA database overlapping with the upregulated mRNAs in the in-house microarray for bioinformatics analysis were selected. The Database for Annotation, Visualization and Integrated Discovery (DAVID; http://david.abcc.ncifcrf.gov/) website was used for GO and KEGG analyses. The functional terms in the categories biological process (BP), cellular component (CC) and molecular function (MF) were obtained from the GO analysis and the GO functional network was drawn using Cytoscape (v3.5.1, http://cytoscape.org).

The interaction pairs of the co-expressed genes were determined using the Search Tool for the Retrieval of Interacting Genes (STRING; version 9.0; http://string-db.org) (32). STRING provides a global database for numerous organisms and the predicted and known interactions were scored. The interactional pairs in the protein-protein interaction (PPI) network were nominated based on a combined score of >0.4.

Fluorescence in situ hybridization (FISH)

The FISH assay was performed to determine the localization of BBOX1-AS1 in the nucleus or cytoplasm of LUSC tissues. Cy3-labeled BBOX1-AS1 probes were designed and synthesized by General Biol. Hybridization was performed overnight with BBOX1-AS1 probes according to the manufacturer's instructions. Images were collected using a Leica DM 4000B laser scanning confocal microscope (Leica Instrument, Inc.). The sequence of the BBOX1-AS1 probe for FISH was CAGGGTAACCGTAG CATGACCTAGAAATAGTCCTCTCA. All experiments were performed at least three times.

Statistical analysis

Statistical analysis was performed using SPSS v22.0 (IBM Corporation). Student's t-test was used to compare the differences between the two groups and the Kruskal-Wallis H-test was employed to evaluate differences among >2 groups. Spearman's rank correlation was utilized to evaluate the correlation between BBOX1-AS1 expression and clinicopathological features. A ROC curve was plotted to distinguish LUSC from normal lung tissues by gene expression. A two-sided P<0.05 was considered to indicate statistical significance.

Results

Differentially expressed lncRNAs via in-house microarray analysis

Based on the in-house microarray analysis, a total of 20,777 lncRNAs were identified. Only 533 upregulated lncRNAs and 1,031 downregulated lncRNAs were significantly differentially expressed between LUSC and normal lung tissues (|fold change| ≥2, P≤0.05; Fig. 2A). The upregulated lncRNAs were extracted from the TCGA, GEO and ArrayExpress databases. Only one upregulated lncRNA, BBOX1-AS1, overlapped among the TCGA, GEO, ArrayExpress and in-house microarray data (Fig. 2B).

Clinical significance of BBOX1-AS1 in LUSC

Increased relative expression of BBOX1-AS1 was observed in LUSC tissues (5.358±0.174) compared with that in noncancerous tissues based on TCGA (0.042±0.006, P<0.0001; Fig. 3A). High BBOX1-AS1 expression was related to the clinicopathological characteristic of lymphatic metastasis (P<0.05; Fig. 3B; Table I). No statistical significance was obtained for any of the other clinicopathological parameters in the TCGA database. Furthermore, the AUC of BBOX1-AS1 was 0.983 (95% CI: 0.973-0.993, P<0.0001; Fig. 3C), which indicated a high possibility that BBOX1-AS1 was able to discriminate LUSC from normal lung tissues. A trend that patients with low BBOX1-AS1 expression survived longer than patients with high BBOX1-AS1 expression was determined via Kaplan-Meier curve analysis (P=0.505; Fig. 3D).

Table I.

Correlation between BBOX1-AS1 expression and clinicopathological features based on TCGA database.

Table I.

Correlation between BBOX1-AS1 expression and clinicopathological features based on TCGA database.

BBOX1-AS1 expression

Clinicopathological featureNMean ± SDRP-value
Age, years 0.050.272
  <60915.752±3.939
  ≥604015.260±3.838
Sex 0.0540.230
  Male3735.243±3.951
  Female1285.723±3.736
Ethnicity 0.0770.004
  White3485.219±3.533
  Black315.777±4.309
  Asian99.209±4.086
T stage 0.0190.674
  T1+T24065.330±3.637
  T3+T4955.518±4.883
N stage 0.1350.140
  N0-N12805.096±3.376
  N2-N3376.721±6.450
M stage 0.0060.919
  M03215.134±3.567
  M165.286±3.639
Stage 0.0450.318
  I+II4085.310±3.641
  III+IV895.766±4.924

[i] T, primary tumor; N, regional lymph nodes; M, distant metastasis; SD, standard deviation; BBOX1-AS1, γ-butyrobetaine hydroxylase 1 antisense 1.

The RT-qPCR results suggested that BBOX1-AS1 was more highly expressed in LUSC cells (NCI-H1703, NCI-H266, SK-MES-1) than in BEAS-2B cells, particularly in NCI-H1703 and SK-MES-1 cells (Fig. 3E). NCI-H1703 and SK-MES-1 cells were selected for further research. The original data from six datasets (E-MTAB-5231, GSE4824-GPL97, GSE19188, GSE27489, GSE30219 and GSE103512) from GEO and the ArrayExpress database were used to verify the expression of BBOX1-AS1 in LUSC. Of these, four datasets (E-MTAB-5231, GSE19188, GSE30219 and GSE103512) confirmed high expression of BBOX1-AS1 (Fig. 3F-I).

The results of the SMD analysis (including 641 cases of LUSC and 159 controls) indicated that the combined SMD and 95% CI reached 1.91 (1.66, 2.16), suggesting significantly higher BBOX1-AS1 expression in the LUSC group than in the control group (P<0.001; Fig. 4A). Of note, publication bias was detected in the present study (P<0.05; Fig. S1A and B). The pooled sensitivity and specificity of BBOX1-AS1 were 0.85 (0.66-0.94) and 0.99 (0.91-1.00), respectively (Fig. S1C). The positive diagnostic likelihood ratio (DLR-positive) and negative diagnostic likelihood ratio (DLR-negative) scores were 83.24 (9.10-761.45) and 0.15 (0.06-0.38), respectively (Fig. S1D). LR<0.1 or >10.0 was indicative of high accuracy. The diagnostic score and odds ratio were 6.30 (3.82-8.77) and 542.07 (45.73-6425.35), respectively (Fig. S1E). The AUC of the SROC was 0.99 (0.98-1.00; Fig. 4B), which indicated a high potential utility of BBOX-AS1 for distinguishing between LUSC and noncancer cases. Publication bias occurred in the SROC analysis (P<0.05; Fig. S1B). A flow chart of the SMD and SROC analyses is provided in Fig. 5.

Effect of BBOX1-AS1 on the proliferation and migration of LUSC cells in vitro

The transfection efficiency of BBOX1-AS1 RNAi plasmid was observed using a light microscope and fluorescence microscope. The transfection efficiency in the BBOX1-AS1 RNAi group in LUSC cell lines was>80% and the knockdown efficiency of BBOX1-AS1 was >70%. Images of cells after transfection with BBOX1-AS1 RNAi and control lentivirus are provided in Fig. 6A and B. RT-qPCR indicated that BBOX1-AS1 expression was significantly lower in the BBOX1-AS1 RNAi groups than in the control groups in both NCI-H1703 and SK-MES-1 cells (P<0.05; Fig. 6C and D).

According to the CCK-8 assay, proliferation was inhibited in the KD group in two cell lines (NCI-H1703 and SK-MES-1) compared to the NC groups (Fig. 7). In NCI-H1703 and SK-MES-1 cells, no obvious difference was observed between the KD group and the NC group on the first day. The proliferation of SK-MES-1 cells in the KD group was markedly reduced on days 2–5 (P<0.05) and most significantly reduced on day five. There was no clear difference observed in NCI-H1703 cells on the second and third days, whereas a significant decrease was determined on day four (P<0.05). Based on these results, it may be concluded that silencing of BBOX1-AS1 inhibited the proliferation of LUSC cells.

In NCI-H1703 cells, the results of the cell cycle analysis revealed that BBOX1-AS1 RNAi caused cell cycle arrest in S phase (P<0.05; Fig. 8). Thus, BBOX1-AS1 RNAi may inhibit the proliferation of LUSC cells partly because silencing of BBOX1-AS1 arrested cell cycle progression.

Furthermore, the results of the Celigo scratch assay suggested that migration was inhibited after knockdown of BBOX1-AS1 in SK-MES-1 cells (P<0.01; Fig. 9A and B).

Potential pathways associated with BBOX1-AS1

The in-house microarray, TCGA and GEO datasets were used to explore the genes co-expressed with BBOX1-AS1. A total of 167 genes were selected for downstream analysis. The possible GO and KEGG pathways were examined based on these 167 genes. The GO and KEGG analyses suggested that these genes may participate in various important biological processes, including cell-cell junctions, protein complex binding, tissue morphogenesis, focal adhesion, ECM-receptor interaction and DNA replication (Fig. 10A; Tables II and III).

Table II.

Top enrichment GO terms in the categories BP, CC and MF by the co-expressed genes of γ-butyrobetaine hydroxylase 1 antisense 1.

Table II.

Top enrichment GO terms in the categories BP, CC and MF by the co-expressed genes of γ-butyrobetaine hydroxylase 1 antisense 1.

GO IDTermOntologyCountP-value
GO:0034621Cellular macromolecular complex subunit organizationBP100.004493
GO:0034622Cellular macromolecular complex assemblyBP90.007375
GO:0042476OdontogenesisBP40.012155
GO:0048730Epidermis morphogenesisBP30.020492
GO:0007155Cell adhesionBP130.021424
GO:0022610Biological adhesionBP130.021636
GO:0048729Tissue morphogenesisBP60.022049
GO:0007229Integrin-mediated signaling pathwayBP40.024231
GO:0048732Gland developmentBP50.032087
GO:0007398Ectoderm developmentBP60.032126
GO:0030057DesmosomeCC51.88E-05
GO:0070161Anchoring junctionCC85.32E-04
GO:0005911Cell-cell junctionCC70.004754
GO:0043296Apical junction complexCC50.008914
GO:0016327Apicolateral plasma membraneCC50.009878
GO:0044427Chromosomal partCC90.0141
GO:0031965Nuclear membraneCC40.022122
GO:0005694ChromosomeCC90.035488
GO:0019866Organelle inner membraneCC70.05378
GO:0030054Cell junctionCC90.062978
GO:0032403Protein complex bindingMF50.094009
GO:0005178Integrin bindingMF30.094724

[i] GO, Gene Ontology; BP, biological process; CC, cellular component; MF, molecular function.

Table III.

Enriched KEGG pathway terms by the co-expressed genes of γ-butyrobetaine hydroxylase 1 antisense 1.

Table III.

Enriched KEGG pathway terms by the co-expressed genes of γ-butyrobetaine hydroxylase 1 antisense 1.

KEGG IDKEGG termCountP-valueGene symbols
hsa04510Focal adhesion60.03381ITGA5, LAMA5, ITGA7, PIK3R5, COL1A1, FLNA
hsa04512ECM-receptor interaction40.039674ITGA5, LAMA5, ITGA7, COL1A1
hsa03030DNA replication30.041514RFC4, FEN1, MCM6
hsa05414Dilated cardiomyopathy40.049758ACTC1, ITGA5, ADCY5, ITGA7

[i] KEGG, Kyoto Encyclopedia of Genes and Genomes.

A gene network of the co-expressed genes of BBOX1-AS1 was also constructed (Fig. 10B), from which it was possible to detect relationships between BBOX1-AS1 and these co-expressed genes.

The PPI network was constructed using STRING online and 253 PPI pairs with a combined score >0.4 were selected (Fig. 10C). The interactions between BBOX1-AS1 and coexpressed genes were obtained through the nodes and strings in the network. Centromere protein A (CENPA) had the highest degree of connectivity (degree=15) according to the PPI network, which indicates that CENPA may be the key gene coexpressed with BBOX1-AS1 in LUSC. In addition, CENPA was downregulated in LUSC based on the TCGA and GEPIA datasets (Fig. S2A and B). Furthermore, the correlation between BBOX1-AS1 and CENPA was determined with the TCGA and GEPIA datasets, indicating that BBOX1-AS1 expression was positively correlated with CENPA in LUSC (R=0.1512; Fig. S2C and D).

Cellular localization of BBOX1-AS1

FISH analysis was performed to observe the cellular location of BBOX1-AS1 in LUSC cells. The majority of BBOX1-AS1 (indicated in red) was located in the cytoplasm (magnification, ×400), indicating that BBOX1-AS1 is able to act as a competing endogenous (ce)RNA in LUSC (Fig. 11).

Discussion

Previous studies have revealed the pivotal role of BBOX1-AS1 in NSCLC, as well as colorectal, cervical and gastric cancers (1720). Liu et al (17) indicated that BBOX1-AS1 was overexpressed in colorectal cancer cell lines and that BBOX1-AS1 knockdown enhanced cell proliferation, invasion and migration while decreasing apoptosis. In cervical cancer, Xu et al (19) determined that BBOX1-AS1 was upregulated. Increased expression of BBOX1-AS1 had stimulating effects on cervical cancer cell growth and migration. Yang et al (18) determined that BBOX1-AS1 was significantly upregulated in gastric cancer tissues and cells and that overexpression of BBOX1-AS1 promoted gastric cancer cell proliferation and inhibited gastric cancer cell apoptosis. Shi et al (20) revealed that KLF5-induced BBOX1-AS1 expression exerted a tumor-promoting role in NSCLC by sponging microRNA (miRNA/miR)-27a-5p. Several obvious differences were observed between the present study and that by Shi et al (20). First, the present study used an in-house microarray analysis to detect differentially expressed lncRNAs between LUSC and normal lung tissues. BBOX1-AS1 was the only overlapping upregulated lncRNA based on the datasets from the TCGA, GEO, ArrayExpress and the in-house microarray. Next, BBOX1-AS1 was indicated to act as a competing ‘sponge’ of miRNA, but this requires BBOX1-AS1 to be located in the cytoplasm. FISH analysis was performed in the present study to determine the location of BBOX1-AS1 in the nucleus or cytoplasm of LUSC tissues. Furthermore, SMD and SROC analyses were included in the present study. SMD analysis confirmed upregulated expression of BBOX1-AS1 in LUSC. SROC analysis indicated the high potential utility of BBOX-AS1 in distinguishing between LUSC and noncancer cases.

In the present study, RT-qPCR and in vitro verification were used to further determine the effect of BBOX1-AS1 in LUSC. LUSC cell proliferation and migration were reduced following BBOX1-AS1 RNAi. These discoveries confirmed the oncogenic role of BBOX1-AS1 in LUSC. NCI-H1703, NCI-H226 and SK-MES-1 cells are three well-known human LUSC cell lines. According to the RT-qPCR analysis, BBOX1-AS1 expression was higher in NCI-H1703 and SK-MES-1 cells than in NCI-H226 cells. Therefore, the NCI-H1703 and SK-MES-1 cell lines were selected for further transfection with BBOX1-AS1 RNAi, and it was indicated that the tumor cell clones were heterogeneous with respect to both morphology and function. Lung cancer is highly heterogeneous in cell origin, histology, clinical features, molecular biology and other aspects, resulting in a lack of effective and specific therapy strategies (33). Future therapies will undoubtedly focus on precision medicine and personalized treatments as the study of nosogenesis continues.

In the present study, it was discovered that high BBOX1-AS1 expression was associated with lymphatic metastasis, which is an important clinical finding in LUSC. Furthermore, the high AUC value indicated that BBOX1-AS1 expression is able to discriminate between LUSC and noncancerous tissues. First, lncRNA high-throughput chips and original RNA-seq data were collected from multiple databases to calculate the SMD and SROC curve of BBOX1-AS1 for LUSC. The combined values of sensitivity (0.85) and specificity (0.99) demonstrated the high accuracy of BBOX1-AS1 for the detection of LUSC. The present results verified that the SROC curve was located near the upper left corner. PLR and NLR were determined to predict the diagnostic accuracy of BBOX1-AS1. LR<0.1 or >10.0 was indicative of high accuracy. A PLR value of 83.24 suggested that patients with LUSC had an ~83.24-fold higher chance of expressing high levels of BBOX1-AS1. In addition, the NLR (0.15) suggested that if the BBOX1-AS1 result was negative, there was an ~15% chance that the patient had LUSC. SMD analysis confirmed higher expression of BBOX1-AS1 in LUSC, in accordance with the results of the RT-qPCR and in-house microarray analyses.

The GO and KEGG analyses indicated that the target genes may have various biological roles. Previous studies suggested that these enriched terms may have essential functions in various cancer types, including cervical, lung and liver cancer (34,35). These enriched functions may be associated with tumor occurrence, and the progression and development of lung cancer (34,36). CENPA had the highest degree of involvement based on the PPI network. CENPA was reported to be significantly involved in the prognosis and predicted outcome of targeted therapy of LUSC (37,38). It was hypothesized that CENPA is the key gene downstream of BBOX1-AS1 in LUSC. BBOX1-AS1 may affect the progression and development of LUSC by regulating various biological functions. However, the precise molecular mechanisms should be determined in future functional experiments.

lncRNAs may be located in the cytoplasm and function as ceRNAs in cancer (39,40). Kong et al (39) reported that lncRNA-CDC6 may act as a ceRNA to target CDC6 by sponging miR-215 to promote the progression of breast cancer. Tang and Yang (40) indicated that lncRNA SNHG14 may function as a ceRNA in hepatocellular carcinoma and the SNHG14/miR-656-3p/SIRT5 axis was able to aggravate the invasion and migration of hepatocellular carcinoma. The results of the FISH assay suggested that BBOX1-AS1 was located in the cytoplasm. Thus, BBOX1-AS1 may act as a ceRNA to affect the biological functions of LUSC. BBOX1-AS1/miR-361-3p/AEG-1 may have significant roles in the proliferation, invasion and metastasis of LUSC based on the literature and bioinformatic prediction software, but functional experiments should be performed for verification (17,19,41).

There were certain limitations to the SMD and SROC analyses. Publication bias was detected and heterogeneity (high I-square values) was unavoidable due to the nonsignificant results in the two GEO datasets. Furthermore, the use of blinding was inconsistent among the five different data sources (GEO, ArrayExpress, TCGA, RT-qPCR and in-house microarray), which contributed to the heterogeneity. To the best of our knowledge, the present study was the first to explore the relationship between BBOX1-AS1 expression and the biological activity of LUSC in vitro. It was determined that BBOX1-AS1 was able to promote the proliferation and migration of LUSC cells. Additional functional experiments should be performed to verify the potential molecular mechanism of BBOX1-AS1 in LUSC.

In conclusion, the present study indicated that BBOX1-AS1 may have an oncogenic effect in LUSC by regulating various biological functions. However, additional functional experiments should be designed to verify the exact mechanism.

Supplementary Material

Supporting Data

Acknowledgements

Not applicable.

Funding

The present study was supported by the Natural Science Foundation of Shandong, China (grant no. ZR202102240355).

Availability of data and materials

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

Authors' contributions

YZ and XW made a significant contribution to the study design, execution and acquisition of data. XKC, YYZ and RQH contributed to data analysis and interpretation. GC and YJQ analyzed the experimental data and wrote and revised the manuscript. YZ and GC confirm the authenticity of the raw data. All authors read and approved the final version of the manuscript.

Ethics approval and consent to participate

All methods used adhered to the relevant guidelines and were approved by the Ethical Committee of the First Affiliated Hospital of Guangxi Medical University (Nanning, China). Consent forms for the use of tissues were signed by the doctors and patients involved in the study.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Glossary

Abbreviations

Abbreviations:

lncRNA

long noncoding RNA

NSCLC

non-small cell lung cancer

LUSC

lung squamous cell carcinoma

RT-qPCR

reverse transcription-quantitative PCR

GEO

Gene Expression Omnibus

TCGA

The Cancer Genome Atlas

GO

Gene Ontology

KEGG

Kyoto Encyclopedia of Genes and Genomes

ROC

receiver operating characteristic

BP

biological process

CC

cellular component

MF

molecular function

DAVID

Database for Annotation, Visualization and Integrated Discovery

STRING

Search Tool for the Retrieval of Interacting Genes

NCBI

National Center of Biotechnology Information

SROC

summary receiver operating characteristic

SRA

Sequence Read Archive

siRNA

small interfering RNA

SMD

standard mean deviation

References

1 

Zhao Y, Wang XX, Wu W, Long H, Huang J, Wang Z, Li T, Tang S, Zhu B and Chen D: EZH2 regulates PD-L1 expression via HIF-1α in non-small cell lung cancer cells. Biochem Biophys Res Commun. 517:201–209. 2019. View Article : Google Scholar : PubMed/NCBI

2 

Laurans M, Botticella A, Moukasse Y, Lévy A and Le Péchoux C: Lung cancer and elective nodal irradiation: A solved issue? Cancer Radiother. 23:701–707. 2019.(In French). View Article : Google Scholar : PubMed/NCBI

3 

Buentzel J, Heinz J, Bleckmann A, Bauer C, Röver C, Bohnenberger H, Saha S, Hinterthaner M, Baraki H, Kutschka I, et al: Sarcopenia as Prognostic Factor in Lung Cancer Patients: A Systematic Review and Meta-analysis. Anticancer Res. 39:4603–4612. 2019. View Article : Google Scholar : PubMed/NCBI

4 

Huang N, Lin W, Shi X and Tao T: STK24 expression is modulated by DNA copy number/methylation in lung adenocarcinoma and predicts poor survival. Future Oncol. 14:2253–2263. 2018. View Article : Google Scholar : PubMed/NCBI

5 

Hoang LT, Domingo-Sabugo C, Starren ES, Willis-Owen SAG, Morris-Rosendahl DJ, Nicholson AG, Cookson WOCM and Moffatt MF: Metabolomic, transcriptomic and genetic integrative analysis reveals important roles of adenosine diphosphate in haemostasis and platelet activation in non-small-cell lung cancer. Mol Oncol. 13:2406–2421. 2019. View Article : Google Scholar : PubMed/NCBI

6 

Fu L, Wang H, Wei D, Wang B, Zhang C, Zhu T, Ma Z, Li Z, Wu Y and Yu G: The value of CEP55 gene as a diagnostic biomarker and independent prognostic factor in LUAD and LUSC. PLoS One. 15:e02332832020. View Article : Google Scholar : PubMed/NCBI

7 

Li K, Tian Y, Yuan Y, Fan X, Yang M, He Z and Yang D: Insights into the Functions of lncRNAs in Drosophila. Int J Mol Sci. 20:202019.

8 

Murillo-Maldonado JM and Riesgo-Escovar JR: The various and shared roles of lncRNAs during development. Dev Dyn. 248:1059–1069. 2019. View Article : Google Scholar : PubMed/NCBI

9 

Zhu J, Yu W, Wang Y, Xia K, Huang Y, Xu A, Chen Q, Liu B, Tao H, Li F, et al: lncRNAs: Function and mechanism in cartilage development, degeneration, and regeneration. Stem Cell Res Ther. 10:3442019. View Article : Google Scholar : PubMed/NCBI

10 

Wang S, Jin J, Xu Z and Zuo B: Functions and Regulatory Mechanisms of lncRNAs in Skeletal Myogenesis, Muscle Disease and Meat Production. Cells. 8:82019. View Article : Google Scholar

11 

Lim LJ, Wong SYS, Huang F, Lim S, Chong SS, Ooi LL, Kon OL and Lee CG: Roles and Regulation of Long Noncoding RNAs in Hepatocellular Carcinoma. Cancer Res. 79:5131–5139. 2019. View Article : Google Scholar : PubMed/NCBI

12 

Chi Y, Wang D, Wang J, Yu W and Yang J: Long Non-Coding RNA in the Pathogenesis of Cancers. Cells. 8:82019. View Article : Google Scholar

13 

Yang J, Qiu Q, Qian X, Yi J, Jiao Y, Yu M, Li X, Li J, Mi C, Zhang J, et al: Long noncoding RNA LCAT1 functions as a ceRNA to regulate RAC1 function by sponging miR-4715-5p in lung cancer. Mol Cancer. 18:1712019. View Article : Google Scholar : PubMed/NCBI

14 

Zhao Y, Feng C, Li Y, Ma Y and Cai R: lncRNA H19 promotes lung cancer proliferation and metastasis by inhibiting miR-200a function. Mol Cell Biochem. 460:1–8. 2019. View Article : Google Scholar : PubMed/NCBI

15 

Huang N, Guo W, Ren K, Li W, Jiang Y, Sun J, Dai W and Zhao W: lncRNA AFAP1-AS1 Supresses miR-139-5p and Promotes Cell Proliferation and Chemotherapy Resistance of Non-small Cell Lung Cancer by Competitively Upregulating RRM2. Front Oncol. 9:11032019. View Article : Google Scholar : PubMed/NCBI

16 

Jin D, Guo J, Wu Y, Du J, Yang L, Wang X, Di W, Hu B, An J, Kong L, et al: m6A mRNA methylation initiated by METTL3 directly promotes YAP translation and increases YAP activity by regulating the MALAT1-miR-1914-3p-YAP axis to induce NSCLC drug resistance and metastasis. J Hematol Oncol. 12:1352019. View Article : Google Scholar : PubMed/NCBI

17 

Liu J, Zhu J, Xiao Z, Wang X and Luo J: BBOX1-AS1 contributes to colorectal cancer progression by sponging hsa-miR-361-3p and targeting SH2B1. FEBS Open Bio. 2211–5463, 12802. 2020.

18 

Yang Y, Yu Q, Li B, Guan R, Huang C and Yang X: BBOX1-AS1 Accelerates Gastric Cancer Proliferation by Sponging miR-3940-3p to Upregulate BIRC5 Expression. Dig Dis Sci. 66:1054–1062. 2021. View Article : Google Scholar : PubMed/NCBI

19 

Xu J, Yang B, Wang L, Zhu Y, Zhu X, Xia Z, Zhao Z and Xu L: lncRNA BBOX1-AS1 upregulates HOXC6 expression through miR-361-3p and HuR to drive cervical cancer progression. Cell Prolif. 53:e128232020. View Article : Google Scholar : PubMed/NCBI

20 

Shi J, Yang C, An J, Hao D, Liu C, Liu J, Sun J and Jiang J: KLF5-induced BBOX1-AS1 contributes to cell malignant phenotypes in non-small cell lung cancer via sponging miR-27a-5p to up-regulate MELK and activate FAK signaling pathway. J Exp Clin Cancer Res. 40:1482021. View Article : Google Scholar : PubMed/NCBI

21 

Zhang Y, He RQ, Dang YW, Zhang XL, Wang X, Huang SN, Huang WT, Jiang MT, Gan XN, Xie Y, et al: Comprehensive analysis of the long noncoding RNA HOXA11-AS gene interaction regulatory network in NSCLC cells. Cancer Cell Int. 16:892016. View Article : Google Scholar : PubMed/NCBI

22 

Linehan WM and Ricketts CJ: The Cancer Genome Atlas of renal cell carcinoma: Findings and clinical implications. Nat Rev Urol. 16:539–552. 2019. View Article : Google Scholar : PubMed/NCBI

23 

Xia L, Zhang W and Gao L: Clinical and prognostic effects of CDKN2A, CDKN2B and CDH13 promoter methylation in ovarian cancer: A study using meta-analysis and TCGA data. Biomarkers. 24:700–711. 2019. View Article : Google Scholar : PubMed/NCBI

24 

Cai KT, Liu AG, Wang ZF, Jiang HW, Zeng JJ, He RQ, Ma J, Chen G and Zhong JC: Expression and potential molecular mechanisms of miR 204 5p in breast cancer, based on bioinformatics and a meta analysis of 2,306 cases. Mol Med Rep. 19:1168–1184. 2019.PubMed/NCBI

25 

Gan BL, Zhang LJ, Gao L, Ma FC, He RQ, Chen G, Ma J, Zhong JC and Hu XH: Downregulation of miR 224 5p in prostate cancer and its relevant molecular mechanism via TCGA, GEO database and in silico analyses. Oncol Rep. 40:3171–3188. 2018.PubMed/NCBI

26 

Rao X, Huang X, Zhou Z and Lin X: An improvement of the 2ˆ(−delta delta CT) method for quantitative real-time polymerase chain reaction data analysis. Biostat Bioinforma Biomath. 3:71–85. 2013.PubMed/NCBI

27 

Zhang Y, Chen WJ, Gan TQ, Zhang XL, Xie ZC, Ye ZH, Deng Y, Wang ZF, Cai KT, Li SK, et al: Clinical Significance and Effect of lncRNA HOXA11-AS in NSCLC: A Study Based on Bioinformatics, In Vitro and In Vivo Verification. Sci Rep. 7:55672017. View Article : Google Scholar : PubMed/NCBI

28 

Zhang Y, Li ZY, Hou XX, Wang X, Luo YH, Ying YP and Chen G: Clinical significance and effect of AEG-1 on the proliferation, invasion, and migration of NSCLC: A study based on immunohistochemistry, TCGA, bioinformatics, in vitro and in vivo verification. Oncotarget. 8:16531–16552. 2017. View Article : Google Scholar : PubMed/NCBI

29 

Cheng Q, Huang C, Cao H, Lin J, Gong X, Li J, Chen Y, Tian Z, Fang Z and Huang J: A Novel Prognostic Signature of Transcription Factors for the Prediction in Patients With GBM. Front Genet. 10:9062019. View Article : Google Scholar : PubMed/NCBI

30 

Ding J and Zhang Y: Analysis of key GO terms and KEGG pathways associated with carcinogenic chemicals. Comb Chem High Throughput Screen. Dec 18–2017.(Epub ahead of print). doi: 10.2174/1386207321666171218120133. PubMed/NCBI

31 

Zhou Z, Li Y, Hao H, Wang Y, Zhou Z, Wang Z and Chu X: Screening Hub Genes as Prognostic Biomarkers of Hepatocellular Carcinoma by Bioinformatics Analysis. Cell Transplant. 28 (Suppl 1):76S–86S. 2019. View Article : Google Scholar : PubMed/NCBI

32 

Liang Y, Zhang C, Ma MH and Dai DQ: Identification and prediction of novel non-coding and coding RNA-associated competing endogenous RNA networks in colorectal cancer. World J Gastroenterol. 24:5259–5270. 2018. View Article : Google Scholar : PubMed/NCBI

33 

de Sousa VML and Carvalho L: Heterogeneity in Lung Cancer. Pathobiology. 85:96–107. 2018. View Article : Google Scholar : PubMed/NCBI

34 

Aboubakar Nana F, Vanderputten M and Ocak S: Role of Focal Adhesion Kinase in Small-Cell Lung Cancer and Its Potential as a Therapeutic Target. Cancers (Basel). 11:112019. View Article : Google Scholar

35 

Mughal MJ, Mahadevappa R and Kwok HF: DNA replication licensing proteins: Saints and sinners in cancer. Semin Cancer Biol. 58:11–21. 2019. View Article : Google Scholar : PubMed/NCBI

36 

Yenerall P, Das AK, Wang S, Kollipara RK, Li LS, Villalobos P, Flaming J, Lin YF, Huffman K, Timmons BC, et al: RUVBL1/RUVBL2 ATPase Activity Drives PAQosome Maturation, DNA Replication and Radioresistance in Lung Cancer. Cell Chem Biol. 27:105–121.e14. 2020. View Article : Google Scholar : PubMed/NCBI

37 

Jiang L and Zhao L, Bi J, Guan Q, Qi A, Wei Q, He M, Wei M and Zhao L: Glycolysis gene expression profilings screen for prognostic risk signature of hepatocellular carcinoma. Aging (Albany NY). 11:10861–10882. 2019. View Article : Google Scholar : PubMed/NCBI

38 

Qi L, Gao C, Feng F, Zhang T, Yao Y, Wang X, Liu C, Li J, Li J and Sun C: MicroRNAs associated with lung squamous cell carcinoma: New prognostic biomarkers and therapeutic targets. J Cell Biochem. 120:18956–18966. 2019. View Article : Google Scholar : PubMed/NCBI

39 

Kong X, Duan Y, Sang Y, Li Y, Zhang H, Liang Y, Liu Y, Zhang N and Yang Q: lncRNA-CDC6 promotes breast cancer progression and function as ceRNA to target CDC6 by sponging microRNA-215. J Cell Physiol. 234:9105–9117. 2019. View Article : Google Scholar : PubMed/NCBI

40 

Tang SJ and Yang JB: lncRNA SNHG14 aggravates invasion and migration as ceRNA via regulating miR-656-3p/SIRT5 pathway in hepatocellular carcinoma. Mol Cell Biochem. 473:143–153. 2020. View Article : Google Scholar : PubMed/NCBI

41 

Yao H, Chen R, Yang Y and Jiang J: lncRNA BBOX1-AS1 Aggravates the Development of Ovarian Cancer by Sequestering miR-361-3p to Augment PODXL Expression. Reprod Sci. 28:736–744. 2021. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

January-2022
Volume 23 Issue 1

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
Zhang Y, Wang X, Cheng X, Zong Y, He R, Chen G and Qin Y: Clinical significance and effect of lncRNA BBOX1‑AS1 on the proliferation and migration of lung squamous cell carcinoma. Oncol Lett 23: 17, 2022
APA
Zhang, Y., Wang, X., Cheng, X., Zong, Y., He, R., Chen, G., & Qin, Y. (2022). Clinical significance and effect of lncRNA BBOX1‑AS1 on the proliferation and migration of lung squamous cell carcinoma. Oncology Letters, 23, 17. https://doi.org/10.3892/ol.2021.13135
MLA
Zhang, Y., Wang, X., Cheng, X., Zong, Y., He, R., Chen, G., Qin, Y."Clinical significance and effect of lncRNA BBOX1‑AS1 on the proliferation and migration of lung squamous cell carcinoma". Oncology Letters 23.1 (2022): 17.
Chicago
Zhang, Y., Wang, X., Cheng, X., Zong, Y., He, R., Chen, G., Qin, Y."Clinical significance and effect of lncRNA BBOX1‑AS1 on the proliferation and migration of lung squamous cell carcinoma". Oncology Letters 23, no. 1 (2022): 17. https://doi.org/10.3892/ol.2021.13135