Open Access

lncRNA PLAC2 activated by H3K27 acetylation promotes cell proliferation and invasion via the activation of Wnt/β‑catenin pathway in oral squamous cell carcinoma

  • Authors:
    • Fubo Chen
    • Shengcai Qi
    • Xu Zhang
    • Jinjin Wu
    • Xi Yang
    • Raorao Wang
  • View Affiliations

  • Published online on: February 1, 2019     https://doi.org/10.3892/ijo.2019.4707
  • Pages: 1183-1194
  • Copyright: © Chen 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

As a new group of important effector molecules involved in multiple cancer types, including breast cancer, lung cancer and oral squamous cell carcinoma, long noncoding RNAs (lncRNAs) have attracted considerable attention recently. However, the underlying cause that induces the dysregulated lncRNAs in cancer remains poorly understood. In the present study, the regulatory model of the lncRNA placenta‑specific protein 2 (PLAC2) upregulation in oral squamous cell carcinoma (OSCC) was investigated and its biological functions in OSCC malignant progression was identified. A reverse transcription‑quantitative polymerase chain reaction assay identified that PLAC2 is upregulated in OSCC cell lines and primary tissue samples. Furthermore, bioinformatic analysis followed by chromatin immunoprecipitation verified an enriched histone H3 on lysine 27 (H3K27) acetylation (H3K27ac) at the promoter region of the PLAC2 gene. Knockdown of cAMP‑response element binding protein‑binding protein (CBP) significantly reduced the enrichment level of H3K27ac, and thereby induced a decreased expression of PLAC2. Functionally, overexpression of PLAC2 promotes OSCC cell proliferation, migration and invasion, whereas knockdown of PLAC2 exerted an opposite effect. Furthermore, the Wnt/β‑catenin signaling pathway was activated by PLAC2 and mediated the PLAC2‑induced malignant progress of OSCC. In conclusion, the present results indicated that lncRNA PLAC2 is transcriptionally activated by H3K27ac modification at the promoter region in OSCC, and promotes cell growth and metastasis via activating Wnt/β‑catenin signaling pathway. Therefore, PLAC2 may serve as a promising biomarker for OSCC prognosis and therapy.

Introduction

Oral squamous cell carcinoma (OSCC) is one of the most aggressive neoplasms among head and neck malignant tumors (1). Smoking and alcohol consumption are the primary environmental factors affecting the development of OSCC. Annually, ~540,000 new cases are diagnosed as OSCC, and mortality rates have not reduced significantly in recent years (2). Although advances have been made in its treatment, the prognosis of OSCC remains poor, with a 5-year survival ratio of nearly 50% in USA in 2009 (3). Improved understanding of the genetic and molecular disorders of the disease is the key to early diagnosis, appropriate treatment and improved prognosis of patients with OSCC.

Long non-coding RNAs (lncRNAs) are a class of poorly conserved endogenous RNAs >200 nucleotides in length that do not encode proteins, but regulate gene expression (4). They are involved in gene regulation by titration of transcription factors, splicing alteration, sponging of microRNAs and recruitment of chromatin modifying enzymes (5-7). Additionally, dysregulated lncRNAs can influence carcinogenesis and chemoresistance in patients with cancer (8). Increasing evidence has demonstrated that a number of lncRNAs, including HOX transcript antisense RNA, H19 and PVT1, are dysregulated and serve a role in the processes of cell proliferation, differentiation, apoptosis and cancer metastasis (9). However, it remains unknown why lncRNAs are upregulated and which signaling pathways are regulated by these lncRNAs.

The discovery of dysregulated lncRNAs represents a new layer of complexity in the molecular architecture of human disease (10). However, numerous gaps in our current understanding of lncRNA function remain. Placenta-specific protein 2 (PLAC2; ENSG00000223573), also known as TINCR, is a 3.7-kb lncRNA located in Chromosome 19p13.3 (11), and no reports have examined the role of lncRNA PLAC2 in OSCC. PLAC2 is upregulated in a number of other cancer types, including gastric, bladder and breast cancer (12). However, whether PLAC2 is dysregulated and regulates the carcinogenesis of OSCC remains unknown.

Currently, the underlying mechanism of lncRNA dysregulation in cancer remains not defined. Histone acetylation, such as histone H3 on lysine 27 (H3K27) acetylation (H3K27ac), is one of the most common types of histone post-translational modifications (13). H3K27ac was first discovered in yeast, and is present in animals and plants (14). H3K27ac is frequently associated with active enhancer regulatory elements (15), and genes associated with these enhancers are expressed at increased levels, compared with those lacking H3K27ac. Previously, Ding et al (16) demonstrated that upregulation of the lncRNA gastric carcinoma proliferation enhancing transcript 1 (GHET1) is due to the H3K27ac modification at the promoter region of the GHET1 gene. However, whether other activated lncRNAs, including PLAC2, are also induced by H3K27ac remains unknown.

In the present study, the aim was to identify the expression level of PLAC2 in OSCC and reveal the underlying mechanism that caused the dysregulation of PLAC2. It was determined that PLAC2 is upregulated and activated by H3K27ac in OSCC. Furthermore, enhanced PLAC2 promotes OSCC progression by regulating the Wnt/β-catenin pathway.

Materials and methods

Clinical samples and ethics statement

The present study included OSCC tissues samples from 48 patients (21 females and 27 males; age range, 35-67 years; median age, 47 years) with OSCC who underwent partial or total surgical resection at the Department of Oral and Maxillofacial Head and Neck Oncology, Ninth People's Hospital, Shanghai JiaoTong University School of Medicine (Shanghai, China) between June 2010 and July 2012. Primary cancer tissues and adjacent non-tumor tissues (>2 cm distal from the cancer area) were collected by surgical resection (no biopsy samples) and were used to investigate the clinical diagnostic and prognostic role of PLAC2. No patients were received radiotherapy or chemotherapy prior to surgical resection. Tissue samples were immediately snap-frozen in liquid nitrogen upon resection and then stored at -80°C until use. The present study was approved by Research Scientific Ethics Committee of Ninth People's Hospital, Shanghai JiaoTong University School of Medicine. All participants signed informed consent prior to using the tissues for scientific research.

Cell culture and regents

A total of 2 human OSCC cell lines SCC-9 and CAL-27 were purchased from Chinese Academy of Sciences (Shanghai, China). The human normal oral epithelial keratinocytes (HOK) were purchased from ScienCell Research Laboratories, Inc. (cat. no. 2610; San Diego, CA, USA). CAL-27 and SCC-9 cells were cultured in Dulbecco's modified Eagle's medium: nutrient Mixture F-12 (DMEM/F12; Gibco; Thermo Fisher Scientific, Inc., Waltham, MA, USA) with 10% fetal bovine serum (FBS; Sigma-Aldrich; Merck KGaA, Darmstadt, Germany). HOK was cultured in DMEM supplemented with 10% FBS and penicillin-streptomycin (100 U/ml and 100 µg/ml, respectively). The culture condition was 37°C in an atmosphere containing 5% CO2. C646 (Selleck Chemicals, Houston, TX, USA) was used at the concentration of 10 µmol/l for 24 h as required.

Vector construction and cell transfection

The synthetic silencing oligonucleotides used for interfering PLAC2 (si-PLAC2) and cAMP-response element binding protein-binding protein (CBP; si-CBP) were synthesized by Guangzhou RiboBio Co., Ltd. (Guangzhou, China). Negative control si-NC was purchased from Invitrogen (cat. no. 12935-110; Thermo Fisher Scientific, Inc.). Additionally, the overexpression plasmid containing oligonucleotides of PLAC2 (p-PLAC2) and negative control vector (p-vector) was generated by Guangzhou RiboBio Co., Ltd. and were loaded into lentivirus vectors (Lv-PLAC2 and Lv-NC, respectively). Cells were transfected with DNA plasmids using TransFast transfection reagent (Promega Corporation, Madison, WI, USA). A total of 5×105 cells was seeded into each well of a 6-well plate and transfected with respective oligoribonucleotides upon reaching 70-80% confluence. For transfection, ~4 µg RNA for each well was used and the final transfection concentration was 100 nM. The transfection effects were assessed by RT-qPCR after transfection for 24 h. The cells were then subjected to further functional assays. The oligonucleotides sequences used for silencing are: si-PLAC2: 5′-UAUUCCUUCAGCCAGUACCCAGGUC-3′; and si-CBP: 5′-CGGCACAGCCTCTCAGTCA-3′.

Reverse transcription-quantitative polymerase chain reaction (RT-qPCR)

Total RNA was extracted from OSCC cells using a RNeasy plus mini kit (Qiagen, Inc., Valencia, CA, USA), according to the manufacturer's protocol. The 20 µl RT reactions were performed using a PrimeScript® RT reagent kit (Takara Biotechnology Co., Ltd., Dalian, China), and incubated for 30 min at 37°C and 5 sec at 85°C. For RT-qPCR, 2 µl diluted RT product was mixed with 23 µl reaction buffer from a SYBR® Green PCR kit (Takara Biotechnology Co., Ltd.) to a final volume of 25 µl. All reactions were conducted using an Eppendorf Mastercycler EP Gradient S (Eppendorf, Hamburg, Germany) under the following conditions: 95°C for 30 sec, followed by 45 cycles of 95°C for 5 sec and 60°C for 30 sec. The expression levels of detected RNAs were normalized to GAPDH using the comparative 2-ΔΔCq method (17). The primer sequences for RT-qPCR are as follows: PLAC2 (sense) 5′-TGT GGCCCAAACTCAGGGATACAT-3′, and (antisense) 5′-AGATGACAGTGGCTGGAGTTGTCA-3′; CBP (sense) 5′-GTGCTGGCTGAGACCCTAAC-3′, and (antisense) 5′-GGCTGTCCAAATGGACTTGT-3′; and GAPDH (sense) 5′-GCACCGTCAAGGCTGAGAAC-3′, and (antisense) 5′-ATGGTGGTGAAGACGCCAGT-3′.

Bioinformatics analysis

The putative binding site of H3K27ac at the promoter region of PLAC2 gene was predicted by UCSC Genome Browser (http://genome.ucsc.edu/). The full sequence of PLAC2 gene can be accessed at NCBI Reference Sequence: NC_000019.10 (https://www.ncbi.nlm.nih.gov/nuccore/NC_000019.10?report=fasta&from=5558167&to=5568034&strand=true).

Cell proliferation assay

The altered cell proliferation following transfection with si-PLAC2 was assayed using an MTT kit (cat. no. 2348-71-2; Dojindo Molecular Technologies, Inc., Rockville, MD, USA). Cells were seeded onto 96-well plates at a density of 3×103 cells/well and cultured in 200 µl DMEM/F12 cell culture medium followed by treatment with si-PLAC2 for 12, 24, 36 or 72 h at the concentration of 100 µM under room temperature. A total of 10 µl MTT (5 mg/ml; pH 7.4; prepared with PBS) was added to culture the cells for 2 h at 37°C in an atmosphere containing 5% CO2. After the medium was removed, the precipitate was diluted in 100 µl dimethyl sulfoxide. An enzyme-linked immunosorbent plate reader was utilized to determine the absorbance of each well.

Cell migration and invasion assay

A wound-healing assay was used here to determine the migration ability of OSCC cells. In brief, cells were seeded at a density of 5×105 cell/well onto 6-well plates. After 12 h with respective vectors, the layer of cells was scratched to form wounds using a sterile 20 µl pipette tip. The non-adherent cells were washed away with culture medium (DMEM/F12) under room temperature, and then the cells were further cultured for 48 h under room temperature followed by imaging with an inverted light confocal microscope at a ×20 magnification (Leica Microsystems GmbH, Wetzlar, Germany) to identify the gap area. Cell invasive ability was evaluated using a Matrigel invasion assay with Boyden chambers (BD Biosciences; Becton, Dickinson and Company, Franklin Lakes, NJ, USA) that had 8-μm pore size membranes with Matrigel. 1×105 cells in the serum-free Opti-MEM media (Gibco; Thermo Fisher Scientific) were placed into the upper chamber of an insert. DMEM/F12 culture medium containing 10% FBS was added to the lower chamber. After 12 h of incubation, the cells that had invaded through the membrane were stained with methanol (15 min) and 0.1% crystal violet (5 min) under room temperature and imaged using an inverted light confocal microscope (Leica Microsystems GmbH).

Immunofluorescence

Cells on slides were permeabilized with 0.3% Triton X-100 (Beyotime Institute of Biotechnology, Shanghai, China) for 15 min after being fixed with 4% paraformaldehyde for 15 min under room temperature. The cells were blocked using 5% goat serum (Beijing Solarbio Science & Technology Co., Ltd., Beijing, China) for 1 h at room temperature followed by incubation with anti-Ki67 antibody (cat. no. ab15580) or anti-CBP antibody (cat. no. ab2832) or anti-β-catenin (cat. no. ab32572) (all from Abcam, Cambridge, MA, USA; all 1:100) overnight at 4°C. The slides were then incubated with anti-rabbit Alexa Fluor 488 (cat. no. 111-545-003; 1:200; Jackson ImmunoResearch Laboratories, Inc., West Grove, PA, USA) for 1 h at room temperature. DAPI was used for nuclear counterstaining at room temperature for 1 h. The samples were observed under a fluorescence microscope (×40; IX73; Olympus Corporation, Tokyo, Japan).

Signal transduction reporter array

The Cignal 45-pathway Reporter ArrayTM (Qiagen, Inc.; cat. no. CCA-901L) was used to determine the potential activity alterations of the 45 untranslated regions in response to overexpression of PLAC2, according to the manufacturer's protocols. Cells (5×105) were suspended in Opti-MEM supplemented with 10% of FBS and 0.1 mM non-essential amino acids (Sigma-Aldrich; Merck KGaA), and then 50 µl of the cell suspension, equivalent to 1×104 cells, was added into each plate well and mixed with DNA resident (Qiagen, Inc.; cat. no. CCA-901L). Cells were transfected with PLAC2-overexpression plasmid (Guangzhou RiboBio Co., Ltd.) for 24 h and were subsequently transfected with a mixture of a transcription factor-responsive firefly luciferase reporter and a constitutively expressing Renilla construct (Qiagen, Inc.) using the TransFast transfection reagent. The cells were incubated for 48 h at 5% CO2 and 37°C. The relative activity of each pathway was decided by luciferase/Renilla, normalized by untreated controls and measured with a Luciferase Reporter Assay System (Promega Corporation).

Cytosolic/nuclear fraction and RNA florescent in situ hybridization (RNA FISH)

The cellular fraction was isolated to locate the sublocation of PLAC2. Briefly, 1×107 cells were harvested, resuspended in 1 ml ice-cold RNase-free PBS, 1 ml buffer C1 (1.28 M Sucrose, 40 mM Tris-HCl, pH 7.5, 20 mM MgCl2 and 4% Triton X-100) and 3 ml RNase-free water, and incubated for 15 min on ice. The cells were then centrifuged for 15 min at 3,000 × g at 4°C, and the supernatant containing the cytoplasmic constituents and the nuclear pellet were retained for RNA extraction.

For RNA FISH, OSCC cells were seeded and fixed with 4% paraformaldehyde for 15 min at 4°C, treated with 0.5% Triton in PBS for 10 min at room temperature, followed by pre-hybridization. They were then hybridized with the PLAC2 probe (5 µM) overnight at 4°C. The PLAC2 probes were synthesized by Sangon (Shanghai Biotech Co., Ltd., Shanghai, China). The cells were visualized under a light confocal microscope at ×40 magnification (Leica Microsystems GmbH).

Chromatin immunoprecipitation (ChIP)

An EZ-Magna ChIP kit (EMD Millipore, Billerica, MA, USA) was used for the ChIP experiment, according to the manufacturer's protocol. OSCC cells were treated with formaldehyde and incubated for 10 min at room temperature to generate DNA-protein cross-links. Cell lysates were then sonicated to generate chromatin fragments of 200-300 bp and immunoprecipitated for 1 h at room temperature with antibodies, including anti-H3K27ac (Abcam; 1:100; cat. no. ab4729), anti-CBP (Abcam; 1:100; cat. no. ab2832) or normal mouse IgG polyclonal antibody (EMD Millipore; 1:100; cat. no. 12-371). Precipitated chromatin DNA was recovered and analyzed by RT-qPCR as aforementioned.

Western blots and antibodies

Radioimmunoprecipitation assay buffer (Sigma Aldrich; Merck KGaA) was used for the cell lysis to obtain total protein-containing lysates. Protein concentration was measured by using the bicinchoninic acid method (Sigma Aldrich; Merck KGaA). Subsequently, quantified protein (25 µg) was transferred onto polyvinylidene fluoride (PVDF) membranes following the separation process by performing a routine 10% gel electrophoresis. The membrane was then blocked with TBS with 10% Tween-20 solution containing 5% non-fat dry milk for 1 h at room temperature followed by treatment with respective primary antibodies (1:1,000) at 4°C overnight. Subsequently, they were incubated with a goat anti-rabbit polyclonal horseradish peroxidase-conjugated secondary antibody (1:5,000; Abcam; cat. no. ab7090) for 1 h at room temperature. The primary antibodies are as follows: anti-transcription factor 4 (TCF-4) antibody (Abcam; cat. no. ab76151), anti-matrix metallopeptidase-7 (MMP-7) antibody (Abcam; cat. no. ab205525), anti-CyclinD1 antibody (Abcam; cat. no. ab16663), anti-MMP-9 antibody (Abcam; cat. no. ab76003), anti-β-catenin antibody (Abcam; cat. no. ab8932) and anti-GAPDH antibody (Invitrogen; Thermo Fisher Scientific, Inc.; cat. no. PA1-987). Protein bands were detected using enhanced chemiluminescence reagent (GE Healthcare, Chicago, IL, USA) and exposed on an X-ray film. Gray analysis by image analysis was performed using the software Gel-Pro Analyzer version 4 (United States Biochemical, Salem, MA, USA) following scanning. The semi-quantitative analysis was performed according to the relative expression of objective protein and internal control protein GAPDH, namely the ratio of objective protein and GAPDH.

In vivo tumorigenesis assay

A total of 12 male BALB/c nude mice (19-22 g; 6 weeks old) were obtained from Animal Center of the Chinese Academy of Science (Shanghai, China). They were randomly divided into two groups of 6 mice and housed 3 per cage. All mice were housed and maintained under specific pathogen-free conditions at 18-22°C, with 20% humidity, a 12/12 h light/dark cycle and feeding ad libitum. The experimental protocol was approved by the Committee on the Animal Ethics of Shanghai Tenth People's Hospital (Shanghai, China). PLAC2 stably expressing cell lines were established by infecting SCC-9 cells with lentivirus as aforementioned and selected by puromycin (Sigma-Aldrich; Merck KGaA). OSCC cells were subcutaneously injected into right side of the armpit regions of mice. The mice were housed for 5 weeks, then were sacrificed and the formed tumors were stripped, and tumor mass was measured. Xenograft volumes were evaluated by caliper measurements of two perpendicular diameters and calculated as formula: Volume = a x b2/2, where ‘a’ represents the length and ‘b’ represents the width). The length of the largest subcutaneous tumor observed was 13.1 mm, and maximum tumor burden was 6.14% (tumor mass/body weight, 1.35/22.0 g). No multiple tumors were exhibited in any mouse.

Another 12 male BALB/c nude mice (19-22 g; 6 weeks old) were also obtained from Animal Center of the Chinese Academy of Science (Shanghai, China). They were randomly divided into two groups of 6 mice and housed 3 per cage with the same conditions aforementioned. Experimental lung metastases were induced by injections of a single-cell suspension (2×106 cells in 100 µl) into the mouse lateral tail vein. Cells were stably infected with Lv-PLAC2 or Lv-NC as aforementioned, and all cell injections were administered in a total volume of 500 µl PBS containing 0.1% bovine serum albumin (Gibco; Thermo Fisher Scientific, Inc.) over 60 sec (18). At 5 weeks after intravenous injection into the nude mice, mice were sacrificed, then the lungs were isolated and tumor nodule numbers and areas were analyzed in a blinded fashion. For luciferase imaging, mice were injected with luciferin (25 mg/ml in 0.1 ml PBS). At 15 min after injection, animals were imaged using an IVIS-100 system at the UAB Small Animal Imaging Core Facility (Birmingham, AL, USA).

All surgeries (subcutaneous or lateral tail vein cell injection, tumor volume measurements and luciferase imaging) were performed under sodium pentobarbital anesthesia (75 mg/kg), and all efforts were made to minimize suffering. During anesthesia (15-20 min) and while recovering, mice were kept warm under a red heat lamp. To prevent the cornea from drying out, sterile eye ointment (Viscotears®; Novartis International AG, Basel, Switzerland) was carefully applied to the eyes during all anesthesia interventions. The endpoint criteria for removing the mice before finishing the study were approved by the Committee on the Ethics of Shanghai Tenth People's Hospital. If one of the endpoint criteria was observed the mouse was sacrificed. For our experiments the following endpoint criteria was used: i) weight loss >15% from the initial weight; ii) ruffled fur, hunched back, closed eyes for more than one day; iii) immobilization/disability because of tumor size or tumor volume >2×103 mm3; iv) breathing difficulties; v) no contact reactions; vi) no palm gripping reflex; and vii) signs of infection.

All mice were sacrificed using CO2 euthanization methods. Briefly, the mice were placed in a transparent polycarbonate shoebox euthanasia chamber (44.0×23.5×21.0 cm). The euthanasia chamber was covered with an acrylic lid, with ports for gas inlet and outlet. Chamber air was replaced with CO2 at a rate of 30% of the chamber volume per minute. Compressed CO2 gas was provided from a cylinder (Weiler Welding, Moraine, OH) and controlled by a CO2-specific regulator (Weiler Welding). A total of 3 mice were euthanized each time. The CO2 inflation time was 6-8 min until the mice were confirmed as dead. The death of the mice was verified comprehensively by the stop of breathing and heartbeat, and the disappearance of pupil and nerve reflex.

Immunohistochemistry staining

Anti-β-catenin antibody (rabbit, polyclonal antibody, 1:100; cat. no. ab32572; Abcam) was used for the immunohistochemistry detection of protein expression in the tumor tissues from mice (4 µm) at 4°C overnight. Endogenous peroxidase was inhibited by incubation with freshly prepared 3% hydrogen peroxide with 0.1% sodium azide for 1 h at room temperature. Non-specific staining was blocked with 0.5% casein and 5% normal serum (Sigma-Aldrich; Merck KGaA) for 1 h at room temperature. The tissue samples were incubated with horseradish peroxidase goat anti-rabbit antibody (Absin, Shanghai, China; cat. no. abs957) for 1 h at room temperature. Staining was developed with diaminobenzidine substrate and the sections were counterstained with hematoxylin for 15 sec at room temperature. Images were visualized using a Nikon ECLIPSE Ti (Nikon Corporation, Tokyo, Japan) microscope system at a ×10 or ×40 magnification and processed using Nikon software version 3.0.

Statistical analysis

The Kolmogorov-Smirnov test was applied for data analysis with the distribution of each group samples. Data were presented as the median (interquartile range). Mann-Whitney U test was used for the comparison of datasets containing two groups. The Kruskal-Wallis test (post-hoc Mann-Whitney U test with Bonferroni's) was used for analyzing statistical difference among multiple groups. Receiver Operation Characteristic (ROC) analysis was used to evaluate the diagnostic performance of PLAC2. The correlations between PLAC2 and CBP expression, and PLAC2 and β-catenin expression were analyzed using Spearman's correlation test. Kaplan-Meier analysis followed by log-rank test was performed to determine the prognostic performance of PLAC2. P<0.05 was considered to indicate a statistically significant difference. Statistical analysis was performed using GraphPad Prism 5 (GraphPad Software, Inc., La Jolla, CA, USA).

Results

PLAC2 is transcriptionally activated by H3K27ac in OSCC

The expression level of PLAC2 in OSCC was measured by RT-qPCR. Fig. 1A demonstrates that PLAC2 expression is significantly increased in OSCC cells, CAL-27 and SCC-9, compared with HOK. Additionally, PLAC2 was also significantly increased in 48 OSCC tissues, compared with the paired precancerous tissues (Fig. 1B). Recent research demonstrated that aberrant expression of lncRNAs is attributed to transcriptionally activation, including acetylation (19). To understand the cause of PLAC2 upregulation in OSCC, the probable mechanisms were investigated by genome bioinformatics analysis. Fig. 1C demonstrates that the promoter region (~1,000 bp) of PLAC2 was predicted with high density of H3K27ac enrichment. To validate the histone acetylation at the promoter region of PLAC2, ChIP assay was performed using an anti-H3K27ac antibody. As depicted in Fig. 1D, H3K27ac was enriched at the promoter region of PLAC2 in both OSCC cells and normal epithelial HOK cells. Furthermore, the enrichment level of H3K27ac was significantly increased in both OSCC cell lines, compared with HOK cells. ChIP assay was further conducted using 5 paired tissues, which were identified with significantly increased PLAC2 level in tumor tissues, compared with non-tumor tissues. As expected, the H3K27ac enrichment level was significantly upregulated in tumor tissues, compared with the paired non-tumor tissues (Fig. 1E). Furthermore, when OSCC cells were treated with histone acetyltransferase (HAT) inhibitor C646, the expression level of PLAC2 was significantly downregulated (Fig. 1F).

CBP facilitate the H3K27ac activation of PLAC2

Subsequently, whether other key enzymes participate in the process of H3K27ac was investigated. As CBP is essential for chromatin acetylation (20), therefore it was hypothesized that CBP may be important for the enhanced acetylation. To test this hypothesis, immunofluorescence was conducted to detect the CBP expression in OSCC cells and normal HOK cells. As expected, the expression level of CBP was increased in both OSCC cell lines, compared with HOK cells (Fig. 2A). Notably, CBP protein was enriched by the PLAC2 gene and the enrichment level was increased in OSCC cells, compared with normal HOK cells (Fig. 2B). Following knockdown of the CBP gene with specific siRNA (Fig. 2C), a ChIP assay was conducted to assess the influence of CBP on H3K27ac enrichment. As expected, the enriched H3K27ac with PLAC2 gene was significantly decreased upon knockdown of CBP (Fig. 2D). Furthermore, knockdown of CBP also caused a significantly decreased expression of PLAC2 in OSCC cells (Fig. 2E). Collectively, the results indicate that upregulated PLAC2 in OSCC is attributed to the CBP-mediated H3K27ac at the promoter region of the PLAC2 gene.

Knockdown of PLAC2 inhibits proliferation and invasion of OSCC cells

As CAL-27 cells exhibited a relatively high expression of PLAC2 while SCC-9 cells exhibited a low expression level, the PLAC2-overexpression vector was built in SCC-9 cells and the PLAC2-knockdown vector in CAL-27 cells (Fig. 3A). Subsequently, an MTT assay was conducted to assess the effects of PLAC2 on cell proliferation over 6 days. As expected, knockdown of PLAC2 significantly suppressed CAL-27 cell proliferation while overexpression of PLAC2 promoted proliferation of SCC-9 cells (Fig. 3B). Additionally, an immunofluorescence assay was performed by an antibody against Ki-67 protein, which is a well-known proliferation marker (21). Fig. 3C demonstrates that knockdown of PLAC2 in CAL-27 cells suppressed the expression level of Ki-67, whereas overexpression of PLAC2 in SCC-9 cells demonstrated the opposite effect. Furthermore, a wound-healing assay indicated that knockdown of PLAC2 significantly suppressed the cell migration ability, while overexpression of PLAC2 increased cell migration (Fig. 3D). Similarly, a Matrigel assay demonstrated a decreased invasion ability caused by PLAC2 knockdown and an increased invasion ability induced by PLAC2 overexpression (Fig. 3E). These results indicate that PLAC2 may serve an oncogenic role in OSCC.

PLAC2 promotes OSCC malignant progression via the activation of the Wnt/β-catenin pathway

The subcellular localization of an lncRNA is associated with its biological mechanism. To verify how PLAC2 contributes to OSCC progression, a cellular fractionation assay and RNA-FISH were performed. It was demonstrated that PLAC2 was distributed primarily in the cytoplasm of SCC-9 cells (Fig. 4A and B). Subsequently, a Signal Reporter Array was conducted to simultaneously investigate the potential signaling pathways in SCC-9 cells affected by overexpression of PLAC2. Notably, the Wnt reporter pathway was reported as one of the most activated pathways upon overexpression of PLAC2 (Fig. 4C). It is previously reported that CBP could interact with Wnt/β-catenin and may serve as a co-activator of Wnt/β-catenin-mediated transcription (22). Furthermore, activation of the Wnt/β-catenin pathway could enhance the proliferation and invasion of cancer cells (23). Therefore, it was hypothesized that PLAC2 may promote malignant progression of OSCC via activating the Wnt/β-catenin signaling pathway. To test this hypothesis, western blot analysis was conducted. The results demonstrated that overexpression of PLAC2 in SCC-9 cells increased the expression of proteins in the Wnt/β-catenin pathway, including β-catenin, TCF-4, MMP-7, MMP-9 and CyclinD1, while knockdown of PLAC2 in CAL-27 cells demonstrated the opposite effect (Fig. 4D). Additionally, the results of the immunofluorescence staining demonstrated that β-catenin trans-located from the cytoplasm to the nucleus in SCC-9 cells upon overexpression of PLAC2 (Fig. 4E). Collectively, the results indicated that PLAC2 may promote OSCC malignant development through activating the Wnt/β-catenin pathway.

PLAC2 promotes tumor growth and metastasis in vivo

Subsequently, an in vivo xenograft model was constructed in male BALB/C nude mice by planting SCC-9 cells, which were stably infected with Lv-PLAC2 or Lv-NC. The mice bearing xenografts were fed for 5 weeks and then tumor xenografts were stripped from nude mice (Fig. 5A). By measuring the volume and mass of the obtained xenograft, it was verified that enhanced PLAC2 expression notably promoted the tumor growth, compared with cells infected with Lv-NC (Fig. 5B). Furthermore, a RT-qPCR assay revealed that the transcript expression of PLAC2, CBP and β-catenin in implanted OSCC tissues were significantly increased in the Lv-PLAC2 group, compared with the Lv-NC group (Fig. 5C). Immunohistochemistry analysis further revealed a significant upregulation of β-catenin in the tumor tissues from the Lv-PLAC2 group, compared with the Lv-NC group (Fig. 5D). Furthermore, a lung-metastasis model was established by injections of a single-cell suspension into the mouse lateral tail vein. As depicted in Fig. 5E and F, the luciferase flux count of lung metastases and visible number of metastases on lung surface were significantly increased in the Lv-PLAC2 group, compared with the Lv-NC group. To conclude, the results confirmed the functional role of PLAC2 in an in vivo xenograft.

Upregulated expression of PLAC2 is associated with poor prognosis of OSCC patients

Based on the aforementioned observations, the role of PLAC2 expression in the diagnosis and prognosis of OSCC was investigated. By performing Spearman's correlation test, positive correlations were identified between the transcript expression of PLAC2 and CBP, or PLAC2 and β-catenin in 48 OSCC tissues (Fig. 6A). Subsequently, the diagnostic role of PLAC2 was investigated by establishing ROC curves using the 48 paired OSCC and adjacent non-tumor tissues (Fig. 6B). The area under the curve of the ROC curve was 0.871, with a diagnostic sensitivity and specificity reaching 75.0 and 85.4% (95% confidence interval, 0.787-0.930), respectively. Analysis of association between PLAC2 expression and clinical pathological factors revealed that PLAC2 expression is positively associated with tumor size and Tumor-Node-Metastasis stage, but not with other factors, including sex, age and histological grade (Table I). By using the median value of PLAC2 as a cut-off, patients with OSCC were stratified into a high and a low PLAC2 expressing group. Kaplan-Meier analysis of the survival data demonstrated that patients with high PLAC2 expression were associated with reduced overall survival and progressive-free survival rates, compared with patients with low expression of PLAC2 (Fig. 6C and D). This indicates a potential prognostic significance of PLAC2 for patients with OSCC.

Table I

Clinical characteristics of 48 patients and the expression of PLAC2 in primary OSCC tissues.

Table I

Clinical characteristics of 48 patients and the expression of PLAC2 in primary OSCC tissues.

Characteristics (46)CasePLAC2 median (range)P-value
Age (years)0.830
 <55270.15 (0.10-0.32)
 ≥55210.16 (0.11-0.28)
Sex0.185
 Male310.17 (0.10-0.32)
 Female170.15 (0.10-0.30)
Tumor size0.036
 <5 cm320.13 (0.10-0.29)
 ≥5 cm160.18 (0.12-0.32)
Histological grade0.087
 Well100.14 (0.11-0.31)
 Moderate270.16 (0.10-0.30)
 Poor110.17 (0.14-0.32)
TNM stage0.005
 I-Ⅱ220.12 (0.10-0.26)
 III-IV260.19 (0.14-0.32)

[i] PLAC2, placenta-specific protein 2; OSCC, oral squamous cell carcinoma; TNM, Tumor-Node-Metastasis.

Collectively, the present study demonstrates that the H3K27ac-induced upregulation of PLAC2 promotes OSCC cell growth and metastasis via the activation of Wnt/β-catenin signaling pathway (Fig. 6E). Therefore, PLAC2 may be a potential prognostic indicator of OSCC.

Discussion

Efforts have been devoted to improve the understanding of molecular characteristics and functional roles of lncRNAs in cancer progression; however, their expression levels and specific roles remain largely unknown in cancer, including OSCC (24). Therefore, identification of the expression level role and functional of lncRNAs, and developing lncRNA-based targeted therapy, may be important for improving the clinical outcome of OSCC. In the present study, the upregulation of lncRNA PLAC2 was verified and the regulatory mechanism that induced the upregulation PLAC2 was revealed. Additionally, it the oncogenic role of PLAC2 in OSCC via the activation of Wnt/β-catenin pathway was also demonstrated.

The prognosis of patients with advanced OSCC remains dismal, and an improved understanding of the underlying mechanisms is critical for identifying effective targets with therapeutic potential to improve the survival of patients with OSCC. Currently, a number of lncRNAs have been studied in OSCC, including HOX transcript antisense RNA (25), metastasis associated lung adenocarcinoma transcript 1 (26) and protein disulfide isomerase family A member 3 (27); however, further investigation is required. The underlying mechanism that regulates the dysregulation of lncRNAs is largely unknown, which makes it difficult to develop effective targeted therapies. In the present study, it was identified that PLAC2 was upregulated in OSCC tissue specimens and cell lines, and further uncovered the epigenetic regulation of PLAC2.

The epigenetic factors, including chromatin modifications, have not only been implicated in the spatio-temporal regulation of gene expression, but also influences how these transcripts are processed (28). H3K27ac was first discovered in yeast (29), and previous advancements in DNA sequencing technology have enabled the analysis of histone acetylation distribution patterns through the whole genome (30). The dysregulation of histone H3K27ac is associated with cancer initiation and malignant progression (13). Notably, lncRNA GHET1 is upregulated and activated in hepatocellular carcinoma due to the H3K27ac modification at the promoter region (16). Therefore, the promoter region of PLAC2 was analyzed by genome bioinformatics analysis and a high-level enrichment of H3K27ac was identified. Histone proteins have long flexible N-terminal tails that are subject to a number of covalent modifications, including acetylation (31). Additionally, numerous Lys residues of histones are involved in interacting with DNA, and this acetylation neutralizes the positive charge of Lys, resulting in the weakening of the DNA-histone interaction and subsequent activation of transcription (32). This indicates that PLAC2 may be transcriptionally activated by the modification of histone acetylation. To test this hypothesis, a ChIP assay was performed and enriched H3K27ac at PLAC2 promoter region was identified. Furthermore, the enrichment level was notably increased in both OSCC cell lines, compared with normal HOK cell lines, which is further confirmed by an increased enrichment of H3K27ac in tumor tissues, compared with non-tumor tissues.

Enzymes that catalyze the reversible post-translational modification of histones on lysine residues by acetylation have received attention as potential drug targets in cancer, aging and embryonic development (33). The histone acetylation process is controlled by HATs and histone deacetylases (34). It is considered that acetylated histones are associated with active transcription, whereas the hypoacetylated histones are involved in gene repression (35). CBP is a HAT that primarily acetylates H3 histones and has a strong association with tumor initiation and progression (36,37). Previously, it was reported that blocking HAT activity of CBP offers a valid target for rationale-based designing of chemical modulators, which could regulate p53 transcriptional functions (38). By using steady-state HAT assays, Bose et al (20) demonstrated that a RNA binding region in the HAT domain of CBP, a regulatory motif unique to CBP, allows RNA to stimulate CBP's HAT activity. Therefore, whether CBP is involved in the H3K27ac modification of PLAC2 promoter was investigated. As expected, knockdown of CBP induced a decreased enrichment of H3K27ac and thereafter a decreased expression of PLAC2.

Following verification of PLAC2 upregulation, its function in OSCC malignant progression was investigated. As expected, increased expression of PLAC2 notably promoted cell proliferation, migration and invasion, whereas decreased PLAC2 demonstrated the opposite effect. The function of PLAC2 has been investigated in other cancer types and was identified as oncogenes in gastric cancer (39), breast cancer (40) and hepatocellular carcinoma (41). Notably, PLAC2 were verified as an oncogene in colorectal and lung cancer by Zheng et al (42) and Zhu et al (43), respectively. However, a contradictory conclusion was developed by Zhang et al (44) and Liu et al (45), which indicates a tumor-suppressive role of PLAC2 in colorectal cancer and lung cancer, respectively. This suggests that the functional role of PLAC2 requires further studies. The present study demonstrated that PLAC2 served an oncogenic function in OSCC. Notably, PLAC2 promotes progression of OSCC by the activation of Wnt/β-catenin signaling pathway as indicates with a Signal Transduction Reporter array followed by western blot analysis. Additionally, miRNA-relevant experiments were not performed; however, it was considered that miRNA could participate in the cancer initiation and progression. Therefore, this hypothesis will be investigated in a follow-up study.

In conclusion, it was demonstrated that the CBP-mediated H3K27ac modification at the promoter region of PLAC2 gene induced an upregulation of PLAC2. Furthermore, enhanced PLAC2 expression promotes OSCC malignant progression via activation of the downstream Wnt/β-catenin signaling pathway. Therefore, PLAC2 may be a potential biological biomarker for OSCC progression.

Funding

No funding was received.

Availability of data and materials

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

Authors' contributions

FC and SQ acquired the data and created a draft of the manuscript. XY, XZ and JW collected clinical samples and performed the in vitro and in vivo assays. FC, XZ and RW analyzed and interpreted the data, and performed statistical analysis. FC, XZ and RW reviewed the manuscript, figures, and tables. All authors read and approved the final manuscript.

Ethics approval and consent to participate

The experimental protocol of animal study was approved by the Committee on the Animal Ethics of Shanghai Tenth People's Hospital (Shanghai, China). The clinical study protocol was approved by the Clinical Research Ethics Committee of Ninth People's Hospital, Shanghai JiaoTong University School of Medicine. Written informed consent was obtained from each participant prior to tissue collection.

Patient consent for publication

All patients enrolled in this study have approved the publication of this study.

Competing interests

The authors declare that they have no competing interests.

Acknowledgments

Not applicable.

References

1 

Binmadi NO and Basile JR: Perineural invasion in oral squamous cell carcinoma: A discussion of significance and review of the literature. Oral Oncol. 47:1005–1010. 2011. View Article : Google Scholar : PubMed/NCBI

2 

Yan L, Chen F, Liu F, Qiu Y, Wang J, Wu J, Bao X, Hu Z, Peng X, Lin X, et al: Differences in modifiable factors of oral squamous cell carcinoma in the upper and lower of oral fissure. Oncotarget. 8:75094–75101. 2017. View Article : Google Scholar : PubMed/NCBI

3 

Glazer CA, Chang SS, Ha PK and Califano JA: Applying the molecular biology and epigenetics of head and neck cancer in everyday clinical practice. Oral Oncol. 45:440–446. 2009. View Article : Google Scholar

4 

Loewen G, Jayawickramarajah J, Zhuo Y and Shan B: Functions of lncRNA HOTAIR in lung cancer. J Hematol Oncol. 7:902014. View Article : Google Scholar : PubMed/NCBI

5 

Nishizawa Y, Konno M, Asai A, Koseki J, Kawamoto K, Miyoshi N, Takahashi H, Nishida N, Haraguchi N, Sakai D, et al: Hypoxia stimulates the cytoplasmic localization of oncogenic long noncoding RNA LINC00152 in colorectal cancer. Int J Oncol. 52:453–460. 2018.PubMed/NCBI

6 

Zhang W, Cai X, Yu J, Lu X, Qian Q and Qian W: Exosome-mediated transfer of lncRNA RP11 838N2.4 promotes erlotinib resistance in non-small cell lung cancer. Int J Oncol. 53:527–538. 2018.PubMed/NCBI

7 

Zhang X, Feng W, Zhang J, Ge L, Zhang Y, Jiang X, Peng W, Wang D, Gong A and Xu M: Long non coding RNA PVT1 promotes epithelial mesenchymal transition via the TGF β/Smad pathway in pancreatic cancer cells. Oncol Rep. 40:1093–1102. 2018.PubMed/NCBI

8 

Han P, Li JW, Zhang BM, Lv JC, Li YM, Gu XY, Yu ZW, Jia YH, Bai XF, Li L, et al: The lncRNA CRNDE promotes colorectal cancer cell proliferation and chemoresistance via miR-181a-5p-mediated regulation of Wnt/β-catenin signaling. Mol Cancer. 16:92017. View Article : Google Scholar

9 

Bassett AR, Akhtar A, Barlow DP, Bird AP, Brockdorff N, Duboule D, Ephrussi A, Ferguson-Smith AC, Gingeras TR, Haerty W, et al: Considerations when investigating lncRNA function in vivo. eLife. 3:e030582014. View Article : Google Scholar : PubMed/NCBI

10 

Wapinski O and Chang HY: Long noncoding RNAs and human disease. Trends Cell Biol. 21:354–361. 2011. View Article : Google Scholar : PubMed/NCBI

11 

Hu YW, Kang CM, Zhao JJ, Nie Y, Zheng L, Li HX, Li X, Wang Q and Qiu YR: LncRNA PLAC2 down-regulates RPL36 expression and blocks cell cycle progression in glioma through a mechanism involving STAT1. J Cell Mol Med. 22:497–510. 2018. View Article : Google Scholar

12 

Kretz M: TINCR, staufen1, and cellular differentiation. RNA Biol. 10:1597–1601. 2013. View Article : Google Scholar : PubMed/NCBI

13 

Wang S, Zang C, Xiao T, Fan J, Mei S, Qin Q, Wu Q, Li X, Xu K, He HH, et al: Modeling cis-regulation with a compendium of genome-wide histone H3K27ac profiles. Genome Res. 26:1417–1429. 2016. View Article : Google Scholar : PubMed/NCBI

14 

Zubritskiy A and Medvedeva YA: DNA sequence features in the establishing of H3K27ac. F1000 Res. 7:1652018. View Article : Google Scholar

15 

Creyghton MP, Cheng AW, Welstead GG, Kooistra T, Carey BW, Steine EJ, Hanna J, Lodato MA, Frampton GM, Sharp PA, et al: Histone H3K27ac separates active from poised enhancers and predicts developmental state. Proc Natl Acad Sci USA. 107:21931–21936. 2010. View Article : Google Scholar : PubMed/NCBI

16 

Ding G, Li W, Liu J, Zeng Y, Mao C, Kang Y and Shang J: LncRNA GHET1 activated by H3K27 acetylation promotes cell tumorigenesis through regulating ATF1 in hepatocellular carcinoma. Biomed Pharmacother. 94:326–331. 2017. View Article : Google Scholar : PubMed/NCBI

17 

Tuomi JM, Voorbraak F, Jones DL and Ruijter JM: Bias in the Cq value observed with hydrolysis probe based quantitative PCR can be corrected with the estimated PCR efficiency value. Methods. 50:313–322. 2010. View Article : Google Scholar : PubMed/NCBI

18 

Minn AJ, Gupta GP, Siegel PM, Bos PD, Shu W, Giri DD, Viale A, Olshen AB, Gerald WL and Massagué J: Genes that mediate breast cancer metastasis to lung. Nature. 436:518–524. 2005. View Article : Google Scholar : PubMed/NCBI

19 

Jain AK, Xi Y, McCarthy R, Allton K, Akdemir KC, Patel LR, Aronow B, Lin C, Li W, Yang L, et al: LncPRESS1 Is a p53-Regulated LncRNA that Safeguards Pluripotency by Disrupting SIRT6-Mediated De-acetylation of Histone H3K56. Mol Cell. 64:967–981. 2016. View Article : Google Scholar : PubMed/NCBI

20 

Bose DA, Donahue G, Reinberg D, Shiekhattar R, Bonasio R and Berger SL: RNA Binding to CBP Stimulates Histone Acetylation and Transcription. Cell. 168:135–149.e22. 2017. View Article : Google Scholar : PubMed/NCBI

21 

Miller I, Min M, Yang C, Tian C, Gookin S, Carter D and Spencer SL: Ki67 is a Graded Rather than a Binary Marker of Proliferation versus Quiescence. Cell Reports. 24:1105–1112.e5. 2018. View Article : Google Scholar : PubMed/NCBI

22 

Arensman MD, Telesca D, Lay AR, Kershaw KM, Wu N, Donahue TR and Dawson DW: The CREB-binding protein inhibitor ICG-001 suppresses pancreatic cancer growth. Mol Cancer Ther. 13:2303–2314. 2014. View Article : Google Scholar : PubMed/NCBI

23 

Han F, Xu Q, Zhao J, Xiong P and Liu J: ERO1L promotes pancreatic cancer cell progression through activating the Wnt/catenin pathway. J Cell Biochem. 119:8996–9005. 2018. View Article : Google Scholar : PubMed/NCBI

24 

Tian T, Wang M, Lin S, Guo Y, Dai Z, Liu K, Yang P, Dai C, Zhu Y, Zheng Y, et al: The Impact of lncRNA Dysregulation on Clinicopathology and Survival of Breast Cancer: A Systematic Review and Meta-analysis. Mol Ther Nucleic Acids. 12:359–369. 2018. View Article : Google Scholar : PubMed/NCBI

25 

Chang SM and Hu WW: Long non-coding RNA MALAT1 promotes oral squamous cell carcinoma development via microRNA-125b/STAT3 axis. J Cell Physiol. 233:3384–3396. 2018. View Article : Google Scholar

26 

Zhang Wu Y, Zhang L, Wang L, Li Y, Ren H, Wei X, Yu F, Liu W, Wang TX, et al: Long non-coding RNA HOTAIR promotes tumor cell invasion and metastasis by recruiting EZH2 and repressing E-cadherin in oral squamous cell carcinoma. Int J Oncol. 46:2586–2594. 2015. View Article : Google Scholar : PubMed/NCBI

27 

Sun CC, Zhang L, Li G, Li SJ, Chen ZL, Fu YF, Gong FY, Bai T, Zhang DY, Wu QM, et al: 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

28 

Piunti A and Pasini D: Epigenetic factors in cancer development: Polycomb group proteins. Future Oncol. 7:57–75. 2011. View Article : Google Scholar

29 

Suka N, Suka Y, Carmen AA, Wu J and Grunstein M: Highly specific antibodies determine histone acetylation site usage in yeast heterochromatin and euchromatin. Mol Cell. 8:473–479. 2001. View Article : Google Scholar : PubMed/NCBI

30 

Tie F, Banerjee R, Conrad PA, Scacheri PC and Harte PJ: Histone demethylase UTX and chromatin remodeler BRM bind directly to CBP and modulate acetylation of histone H3 lysine 27. Mol Cell Biol. 32:2323–2334. 2012. View Article : Google Scholar : PubMed/NCBI

31 

Tak YG, Hung Y, Yao L, Grimmer MR, Do A, Bhakta MS, O'Geen H, Segal DJ and Farnham PJ: Effects on the transcriptome upon deletion of a distal element cannot be predicted by the size of the H3K27Ac peak in human cells. Nucleic Acids Res. 44:4123–4133. 2016. View Article : Google Scholar : PubMed/NCBI

32 

Kouzarides T: Chromatin modifications and their function. Cell. 128:693–705. 2007. View Article : Google Scholar : PubMed/NCBI

33 

Benton CB, Fiskus W and Bhalla KN: Targeting Histone Acetylation: Readers and Writers in Leukemia and Cancer. Cancer J. 23:286–291. 2017. View Article : Google Scholar : PubMed/NCBI

34 

Pradeepa MM, Grimes GR, Kumar Y, Olley G, Taylor GC, Schneider R and Bickmore WA: Histone H3 globular domain acetylation identifies a new class of enhancers. Nat Genet. 48:681–686. 2016. View Article : Google Scholar : PubMed/NCBI

35 

Gräff J and Tsai LH: Histone acetylation: Molecular mnemonics on the chromatin. Nat Rev Neurosci. 14:97–111. 2013. View Article : Google Scholar : PubMed/NCBI

36 

Hamzaoui H, Rizk-Rabin M, Gordon J, Offutt C, Bertherat J and Bouizar Z: PTHrP P3 p romoter activity in breast cancer cell lines: Role of Ets1 and CBP (CREB binding protein). Mol Cell Endocrinol. 268:75–84. 2007. View Article : Google Scholar : PubMed/NCBI

37 

Tang Z, Yu W, Zhang C, Zhao S, Yu Z, Xiao X, Tang R, Xuan Y, Yang W, Hao J, et al: CREB-binding protein regulates lung cancer growth by targeting MAPK and CPSF4 signaling pathway. Mol Oncol. 10:317–329. 2016. View Article : Google Scholar

38 

Hsu CH, Chang MD, Tai KY, Yang YT, Wang PS, Chen CJ, Wang YH, Lee SC, Wu CW and Juan LJ: HCMV IE2-mediated inhibition of HAT activity downregulates p53 function. EMBO J. 23:2269–2280. 2004. View Article : Google Scholar : PubMed/NCBI

39 

Xu TP, Wang YF, Xiong WL, Ma P, Wang WY, Chen WM, Huang MD, Xia R, Wang R, Zhang EB, et al: E2F1 induces TINCR transcriptional activity and accelerates gastric cancer progression via activation of TINCR/STAU1/CDKN2B signaling axis. Cell Death Dis. 8:e28372017. View Article : Google Scholar : PubMed/NCBI

40 

Liu Y, Du Y, Hu X, Zhao L and Xia W: Up-regulation of ceRNA TINCR by SP1 contributes to tumorigenesis in breast cancer. BMC Cancer. 18:3672018. View Article : Google Scholar : PubMed/NCBI

41 

Tian F, Xu J, Xue F, Guan E and Xu X: TINCR expression is associated with unfavorable prognosis in patients with hepato-cellular carcinoma. Biosci Rep. 37:372017. View Article : Google Scholar

42 

Zheng Y, Yang C, Tong S, Ding Y, Deng W, Song D and Xiao K: Genetic variation of long non-coding RNA TINCR contribute to the susceptibility and progression of colorectal cancer. Oncotarget. 8:33536–33543. 2017.PubMed/NCBI

43 

Zhu ZJ and He JK: TINCR facilitates non-small cell lung cancer progression through BRAF-activated MAPK pathway. Biochem Biophys Res Commun. 497:971–977. 2018. View Article : Google Scholar : PubMed/NCBI

44 

Zhang ZY, Lu YX, Zhang ZY, Chang YY, Zheng L, Yuan L, Zhang F, Hu YH, Zhang WJ and Li XN: Loss of TINCR expression promotes proliferation, metastasis through activating EpCAM cleavage in colorectal cancer. Oncotarget. 7:22639–22649. 2016.PubMed/NCBI

45 

Liu X, Ma J, Xu F and Li L: TINCR suppresses proliferation and invasion through regulating miR-544a/FBXW7 axis in lung cancer. Biomed Pharmacother. 99:9–17. 2018. View Article : Google Scholar : PubMed/NCBI

46 

Pollaers K, Hinton-Bayre A, Friedland PL and Farah CS: AJCC 8th Edition oral cavity squamous cell carcinoma staging - Is it an improvement on the AJCC 7th Edition? Oral Oncol. 82:23–28. 2018. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

April-2019
Volume 54 Issue 4

Print ISSN: 1019-6439
Online ISSN:1791-2423

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Chen F, Qi S, Zhang X, Wu J, Yang X and Wang R: lncRNA PLAC2 activated by H3K27 acetylation promotes cell proliferation and invasion via the activation of Wnt/β‑catenin pathway in oral squamous cell carcinoma. Int J Oncol 54: 1183-1194, 2019
APA
Chen, F., Qi, S., Zhang, X., Wu, J., Yang, X., & Wang, R. (2019). lncRNA PLAC2 activated by H3K27 acetylation promotes cell proliferation and invasion via the activation of Wnt/β‑catenin pathway in oral squamous cell carcinoma. International Journal of Oncology, 54, 1183-1194. https://doi.org/10.3892/ijo.2019.4707
MLA
Chen, F., Qi, S., Zhang, X., Wu, J., Yang, X., Wang, R."lncRNA PLAC2 activated by H3K27 acetylation promotes cell proliferation and invasion via the activation of Wnt/β‑catenin pathway in oral squamous cell carcinoma". International Journal of Oncology 54.4 (2019): 1183-1194.
Chicago
Chen, F., Qi, S., Zhang, X., Wu, J., Yang, X., Wang, R."lncRNA PLAC2 activated by H3K27 acetylation promotes cell proliferation and invasion via the activation of Wnt/β‑catenin pathway in oral squamous cell carcinoma". International Journal of Oncology 54, no. 4 (2019): 1183-1194. https://doi.org/10.3892/ijo.2019.4707