Circadian clock gene Per2 plays an important role in cell proliferation, apoptosis and cell cycle progression in human oral squamous cell carcinoma

  • Authors:
    • Qingqing Wang
    • Yiran Ao
    • Kai Yang
    • Hong Tang
    • Dan Chen
  • View Affiliations

  • Published online on: April 1, 2016     https://doi.org/10.3892/or.2016.4724
  • Pages: 3387-3394
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Previous studies have shown that the aberrant expression of period circadian clock 2 (Per2) is closely related to the occurrence and development of cancers, but the specific mechanism remains unclear. In the present study, we used shRNA to downregulate Per2 in oral squamous cell carcinoma (OSCC) Tca8113 cells, and then detected the alterations in cell cycle, cell proliferation and apoptosis by flow cytometric analysis and mRNA expression alterations in all the important genes in the cyclin/cyclin-dependent protein kinase (CDK)/cyclin-dependent kinase inhibitor (CKI) cell cycle network by RT-qPCR. We found that in the Tca8113 cells, after Per2 downregulation, the mRNA expression levels of cyclin A2, B1 and D1, CDK4, CDK6 and E2F1 were significantly increased (P<0.05), the mRNA expression levels of p53, p16 and p21 were significantly decreased (P<0.05), cell proliferation was significantly higher (P<0.05), apoptosis was significantly lower (P<0.05) and the number of cells in the G1/G0 phase was significantly decreased (P<0.05). The present study proves that in OSCC, clock gene Per2 plays an important role in cell cycle progression and the balance of cell proliferation and apoptosis by regulation of the cyclin/CDK/CKI cell cycle network. Further research on Per2 may provide a new effective molecular target for cancer treatments.

Introduction

In mammals, many life activities vary in an approximate 24 h periodic fluctuation, which is called the circadian rhythm (1,2). The circadian rhythm, which is one of the basically intrinsic characteristics of life activities, plays an important role in maintaining complicated life activities in a highly coordinated and orderly manner (3,4). The clock genes, whose rhythmic expression is responsible for circadian rhythms, exist in almost all cells in the body (5,6). To date, at least 14 core clock genes have been described, including Per1, period circadian clock 2 (Per2), Per3, Cry1, Cry2, Tim, Ck1ε, Clock, Bmal1, Rors, Rev-Erbs, Npas2, Dec1 and Dec2 (7,8). In mammals, circadian rhythms play an important role in physiological activities, including cell proliferation, metabolism and hormone secretion (9,10). Aberrant circadian rhythms lead to cardiovascular diseases, gastrointestinal diseases, nervous and mental diseases and cancers (1113).

Per2 is an important clock gene that works as a pacemaker of circadian rhythms in mammals (14). The absence of Per2 can lead to loss of circadian rhythms (1517). In recent years, studies have shown that the aberrant expression of Per2 is responsible for not only circadian rhythm alterations but also the occurrence and development of cancers (6,18). Per2 is reduced in various types of solid cancers, including breast and skin cancer, hepatocellular carcinoma, colorectal cancer, renal carcinoma, gastric cancer and head and neck squamous cell carcinoma (11,1924). Since many downstream cell cycle genes are regulated by Per2, the aberrant expression of Per2 affects cell cycle progression and leads to carcinogenesis (6,22,25).

Imbalance of cell proliferation and apoptosis caused by cell cycle disorder is the main reason for carcinogenesis (26,27). The cell cycle is under the control of the cyclin/cyclin-dependent protein kinase (CDK)/cyclin-dependent kinase inhibitor (CKI) cell cycle network composed of cyclins, CDKs and CKIs (28,29). To date, there have been only dispersed research studies on the Per2 regulation of downstream cell cycle genes, and these are cyclin A, B1, D1 and E, p53, c-myc, Rb, Mdm-2 and Gadd45α, which are mainly focused on the Per2 regulation of cyclins (6,18,25,30). However, the role Per2 plays in CDKs and CKIs, which are the two other important aspects of the cyclin/CDK/CKI network, remains unclear. We speculated that Per2 may regulate numerous molecules in all the three aspects of the cyclin/CDK/CKI cell cycle network. In the present study, we downregulated Per2 in oral squamous cell carcinoma (OSCC) cell line Tca8113, and then detected the alterations in the cell cycle, cell proliferation and apoptosis and all the important genes in the cyclin/CDK/CKI network to further illustrate the relationship of Per2 with the occurrence and development of cancers.

Materials and methods

Cell culture

Normal oral mucosa was collected from the Department of Maxillofacial Plastic Surgery at the Affiliated Hospital of Stomatology, Chongqing Medical University following approval by the local ethics committee. Patient samples were obtained after informed consent following the tenets of the Declaration of Helsinki, and written consent was obtained from all patients prior to surgery. The mucosa was kept in 1.0 U/ml Dispase II (Roche, Indianapolis, IN, USA) at 4°C overnight. Complete epithelial layer was separated from the mucosa under a microscope (Leica, Wetzlar, Germany). The epithelial layer was digested in 0.25% TrypLE Express (Gibco, Grand Island, NY, USA) at 37°C, and then inoculated in a 6-well plate paved by rat tail collagen using OKM (ScienCell, San Diego, CA, USA) at 37°C in a humidified atmosphere of 95% air and 5% CO2. The oral mucosal epithelial cells were continuously passaged/4 days. The second generation of cells was used for RNA and protein extraction, and the third generation of cells was used for making cell climbing slices. Tca8113 cells (Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, China) were cultured in RPMI-1640 medium with 10% fetal bovine serum (both from HyClone, Logan, UT, USA) at 37°C in an atmosphere of 95% humidity and 5% CO2.

Purity identification of the oral mucosal epithelial cells

Cell climbing slices were fixed by 4% paraformaldehyde, and then incubated for 10 min with 3% H2O2 at 37°C. After permeabilization in 0.1% Triton X-100 for 15 min, the slices were blocked using goat serum for 30 min at 37°C. The slices were incubated firstly with the rabbit monoclonal anti-keratin antibody (1:100; ZA-0540) overnight at 4°C, and secondly with the rabbit SP kit (SP-9001) (both from ZSGB-BIo, Beijing, China) for 1 h at 37°C according to the manufacturer's recommended protocol. Finally, slices were examined under a microscope (Olympus, Tokyo, Japan). In negative controls all reagents were used except the primary antibody.

Downregulation of Per2 in Tca8113 cells by shRNA plasmids

The plasmids pGPU6-Per2-shRNA-I-III and pGPU6-control-shRNA were obtained from Chengdu Biotechnology Co., Ltd. (Table I). The day before transfection, the Tca8113 cells were incubated into a 6-well plate to ensure that by the time of transfection the cells reached ~30% confluency. Approximately 4 μg of the plasmids were transfected using Lipofectamine 2000 (Invitrogen, Carlsbad, CA, USA) according to the manufacturer's protocol. The effect of Per2 downregulation was examined 36–72 h later. There were five groups in our experiment: Per2-shRNA-I, Per2-shRNA-II, Per2-shRNA-III, control-shRNA and Tca8113 group. Per2-shRNA-I, Per2-shRNA-II, Per2-shRNA-III and the control-shRNA group were transfected with pGPU6-Per2-shRNA-I, pGPU6-Per2-shRNA-II, pGPU6-Per2-shRNA-III and pGPU6-control-shRNA, respectively; the Tca8113 group did not accept any reagents.

Table I

The RNA oligos of shRNA and negative control plasmids.

Table I

The RNA oligos of shRNA and negative control plasmids.

PlasmidRNA oligos (5′-3′)
pGPU6-Per2-shRNA-I 5′-CACCGAAGTACGCCCTCAGGAGCTTCAAGAGAGCTCCTGAGGGCGTACTTCTTTTTTG-3′
5′-GATCCAAAAAAGAAGTACGCCCTCAGGAGCTCTCTTGAAGCTCCTGAGGGCGTACTTC-3′
pGPU6-Per2-shRNA-II 5′-CACCGTGAAGAATGCCGATATGTTTCAAGAGAACATATCGGCA TTCTTCACTTTTTTG-3′
5′-GATCCAAAAAAGTGAAGAATGCCGATATGTTCTCTTGAAACATATCGGCATTCTTCAC-3′
pGPU6-Per2-shRNA-III 5′-CACCGAAGTACGCCCTCAGGAGCTTCAAGAGAGCTCCTGAGGGCGTACTTCTTTTTTG-3′
5′-GATCCAAAAAAGAAGTACGCCCTCAGGAGCTCTCTTGAAGCTCCTGAGGGCGTACTTC-3′
pGPU6-control-shRNA 5′-CCGGGCACTACGAGAGCTAACTCAGCTCGAGCTGAGTTAGCTCTCGTAGTGCTTTTTG-3′
5′-AATTCAAAAAGCACTACGAGAGCTAACTCAGCTCGAGCTGAGTTAGCTCTCGTAGTGC-3′
Western blotting

The cells were lysed using RIPA + PMSF (Beyotime, Jiangsu, China) and centrifuged for 2 min, at 4°C and 167.7 × g. The concentration of cell protein was detected using the enhanced BCA protein assay kit (Beyotime). Proteins (50 μg) were seperated by 8% SDS-PAGE using Mini-PROTEAN 3 system (Bio-Rad, Hercules, CA, USA) and transferred to polyvinylidene difluoride (PVDF) membranes (Pierce, Rockford, IL, USA) using Trans-Blot SD Semi-Dry Transfer Cell (Bio-Rad). The membranes were incubated with mouse monoclonal anti-Per2 antibody (1:500; 19-J6:sc-101105; Santa Cruz Biotechnology, Inc., Santa Cruz, CA, USA) and mouse monoclonal anti-β-actin antibody (1:1,000; 60008-1-lg), respectively, overnight at 4°C, followed by goat monoclonal anti-mouse IgG (1:1,000) (SA00001-1) (both from ProteinTech, Chicago, IL, USA) for 1 h at 37°C. Blots were detected using enhanced chemiluminescence reagent (Pierce) under a fluorescent chemiluminescence imaging system (ChemiDoc XRS+; Bio-Rad). The software Quality one (Bio-Rad) was used to analyze the blots. To ensure accuracy the experiment was performed in triplicate.

Flow cytometric analysis

For analysis of the cell cycle, after a 48-h transient transfection, the cells were harvested and fixed using 70% ethanol overnight at 4°C. The cells were stained with propidium iodide solution (Cell Cycle Detection kit; KGA, China) for 30 min at 4°C in the dark, and subsequently detected by a flow cytometer (FACSVantage; BD Biosciences, San Jose CA, USA). The following formula was used to calculate the proliferation index (PI) of the cells: PI = (S + G2/M)/(G0/G1 + S + G2/M) × 100% (G0, G1, S, G2 and M represent the corresponding cell cycle phases). To ensure accuracy, the experiment was performed in triplicate. For analysis of apoptosis, after a 48-h transient transfection, the cells were harvested and stained with the Annexin V-FITC cell apoptosis analysis kit (with propidium iodide) (Sungene, Tianjin, China). Apoptotic cells were quantified by a flow cytometer (FACSVantage). The following formula was used to calculate apoptotic index (AI) of the cells: AI = (number of apoptotic cells/number of total detected cells) × 100%. To ensure accuracy, the experiment was performed in triplicate.

Quantitative real-time PCR (RT-qPCR)

After a 36-h transient transfection, total RNA was isolated from the cells using RNAiso Plus (9109; Takara, Japan). The optical density and concentration of RNA were detected by an ultraviolet spectrophotometer (NanoDrop, USA). RNA was reverse-transcribed with 20 μl of the system with a Prime Script RT reagent kit (RR047A; Takara) on a T100 thermal cycler (Bio-Rad, Singapore) according to the manufacturer's instructions. Oligo 17.0 software was used to design the specific primers of p53, p16, p21, cyclin A2, B1, D1 and E, CDK1, CDK2, CDK4, CDK6, E2F1, c-myc, cdc25, Wee1, Rb1 and GAPDH (endogenous reference) (Table II). According to the manufacturer's instructions, cDNA was used as the template for amplification using SYBR Premix Ex Taq™ II (RR820A; Takara) on a CFX96 Real-Time PCR Detection system (Bio-Rad). The threshold cycle (Ct) value was acquired. The relative expression levels of p53, p16, p21, cyclin A2, B1, D1 and E, CDK1, CDK2, CDK4, CDK6, E2F1, c-myc, cdc25, Wee1 and Rb1 mRNA in cells were calculated using the 2−ΔΔCt method. To ensure accuracy, the experiment was performed in triplicate.

Table II

Primers used for real-time PCR amplification of gene expression

Table II

Primers used for real-time PCR amplification of gene expression

GeneForward primer sequenceReverse primer sequence
Per2 5′-CGTGTTCCACAGTTTCACCT-3′ 5′-GGTAGCGGATTTCATTCTCG-3′
cyclin A2 5′-ATGTCACCGTTCCTCCTTG-3′ 5′-AGGGCATCTTCACGCTCTA-3′
cyclin B1 5′-TGGTTGATACTGCCTCTCCA-3′ 5′-TGACTGCTTGCTCTTCCTCA-3′
cyclin D1 5′-GTGTATCGAGAGGCCAAAGG-3′ 5′-CAACCAGAAATGCACAGACC-3′
cyclin E 5′-CTGGATGTTGACTGCCTTGA-3′ 5′-ATGTCGCACCACTGATACCC-3′
c-myc 5′-ATCCTGTCCGTCCAAGCA-3′ 5′-CGCACAAGAGTTCCGTAG-3′
p53 5′-GTCCAACAACACCAGCTCCT-3′ 5′-CTCTCGGAACATCTCGAAGC-3′
CDK1 5′-GTCCGCAACAGGGAAGAAC-3′ 5′-CGAAAGCCAAGATAAGCAACT-3′
CDK2 5′-CAGGATGTGACCAAGCCAGT-3′ 5′-TGAGTCCAAATAGCCCAAGG-3′
CDK4 5′-CTGGACACTGAGAGGGCAAT-3′ 5′-TGGGAAGGAGAAGGAGAAGC-3′
CDK6 5′-TCTTCATTCACACCGAGTAGTGC-3′ 5′-TGAGGTTAGAGCCATCTGGAAA-3′
Rb1 5′-CACAAGCAACCTCAGCCTTC-3′ 5′-GCGTTCACAAAGTGTATTTAGCC-3′
E2F1 5′-CCAACTCCCTCTACCCTTGA-3′ 5′-GTCTCCCTCCCTCACTTTCC-3′
Wee1 5′-TGTGGTGGTGTGCTGCTTAT-3′ 5′-TTCAAAGGGAGGGTATGTCTG-3′
cdc25 5′-TACTCGGCCATGTCACCCTT-3′ 5′-GGGTCGTATCGCCCTCATC-3′
p16 5′-ACCAGAGGCAGTAACCATGC-3′ 5′-TGATCTAAGTTTCCCGAGGTTT-3′
p21 5′-TTAGCAGCGGAACAAGGAGT-3′ 5′-CGTTAGTGCCAGGAAAGACA-3′
GAPDH 5′-ACAACTTTGGTATCGTGGAAGG-3′ 5′-GCCATCACGCCACAGTTTC-3′
Statistical analysis

The SPSS 17.0 statistical software package was used to analyze and calculate the mean ± SD of the data. The difference in Per2 expression between epithelial and Tca8113 cells was analyzed using group t-test. In addition, one-way ANOVA was used to analyze the differences between the various groups transfected with the different plasmids. A value of P<0.05 was considered statistically significant.

Results

Culture and purity identification of the oral mucosal epithelial cells

Under microscope, the oral mucosal epithelial cells had a paving stone appearance (Fig. 1A), and keratin was expressed in 100% of the cells (Fig. 1B), which identified the purity of the oral mucosal epithelial cells.

Expression of Per2 mRNA and protein in the oral mucosal epithelial and Tca8113 cells

In the oral mucosal epithelial and Tca8113 cells, the expression of Per2 mRNA was 2.41±0.21 and 1.00±0.12, respectively; and the expression of Per2 protein was 2.87±0.26 and 1.11±0.13, respectively (Fig. 2). The expression levels of Per2 mRNA and protein in the Tca8113 cells were significantly lower than those of the oral mucosal epithelial cells (P<0.05), indicating that Per2 expression is reduced in OSCC.

Alterations of Per2 mRNA and protein expression after transfections in the Tca8113 cells

In the Tca8113, control-shRNA, Per2-shRNA-I, Per2-shRNA-II and Per2-shRNA-III groups, the expression levels of Per2 mRNA were 3.20±0.52, 3.01±0.11, 1.67±0.30, 1.45±0.34 and 1.00±0.13, respectively; and the expression levels of Per2 protein were 3.21±0.42, 3.18±0.52, 1.52±0.11, 1.22±0.15 and 0.87±0.21, respectively (Fig. 3). Per2 mRNA and protein expression showed no significant difference between the control-shRNA and Tca8113 groups (P>0.05). However, Per2 mRNA and protein expression levels were significantly lower in the Per2-shRNA-III group than those noted in the control-shRNA and Tca8113 groups (P<0.05). Therefore, Per2 was effectively reduced in the Per2-shRNA-III group, and the Per2-shRNA-III group was adopted for subsequent experiment.

Effects of Per2 downregulation on cell cycle distribution, PI and AI in the Tca8113 cells

The cells from the different groups were collected and their cell cycle distribution (Fig. 4A; Table III) and apoptosis (Fig. 4B; Table III) were analyzed, and then PI and AI were calculated (Table III). Compared with those of the Tca8113 and control-shRNA groups, the Per2-shRNA-III group had a significantly decreased number of cells in the G1/G0 phase (P<0.05), significantly increased PI (P<0.05), and significantly decreased AI (P<0.05). The number of cells in the G1/G0 phase, and PI and AI showed no significant difference between the Tca8113 and control-shRNA group (P>0.05).

Table III

Effects of shRNA on cell cycle distribution, PI and AI in Tca8113 cells (mean ± SD).

Table III

Effects of shRNA on cell cycle distribution, PI and AI in Tca8113 cells (mean ± SD).

IndexTca8113Control-shRNAPer2-shRNA-IIIPP1P2P3
G1/G0 (%)51.00±2.1251.83±2.2746.04±1.760.0280.0260.0140.642
S (%)29.23±1.1027.41±4.1930.93±1.940.3550.4760.1670.447
G2/M (%)19.77±2.7520.76±3.1723.03±0.710.3190.1550.3010.638
PI (%)49.00±2.1248.17±2.2753.96±1.770.0280.0260.0140.642
AI (%)14.66±1.9314.13±0.9410.26±1.020.0150.0080.0140.651

[i] The cells from different groups were collected and their cell cycle distribution, PI and AI were analyzed. Compared with those of the Tca8113 and control-shRNA groups, in the Per2-shRNA-III group the number of cells at the G1/G0 phase was significantly decreased (P<0.05), PI was significantly increased (P<0.05) and AI was significantly decreased (P<0.05). There was no significant difference in the number of cells at the G1/G0 phase, PI and AI between the Tca8113 and control-shRNA group (P>0.05). P represents one-way ANOVA among the three groups. P1, P2 and P3 represent the pairwise comparison with the Per2-shRNA-III and Tca8113 group, Per2-shRNA-III and control-shRNA group and control-shRNA and Tca8113 group, respectively. A value of P<0.05 represents a significant difference. PI, proliferation index; AI, apoptosis index; G0/G1, cell number at the G0 and G1 phase; G2/M, cell number at the G2 and M phase.

Effects of Per2 downregulation on mRNA expression of cell cycle-related genes in the Tca8113 cells

Results of the RT-qPCR are shown in Table IV. Compared with those of the Tca8113 and control-shRNA groups, the Per2-shRNA-III group had a significantly decreased mRNA expression of p53, p16 and p21 (P<0.05), significantly increased mRNA expression of cyclin A2, B1 and D1, CDK4, CDK6, E2F1 (P<0.05), and a similar mRNA expression of c-myc, cyclin E, CDK1, CDK2, cdc25, Wee1 and Rb1 (P>0.05). Between the Tca8113 and control-shRNA group, there was no significant difference in mRNA expression of p53, p16, p21 cyclin A2, B1 and D1, CDK4, CDK6 and E2F1 (P>0.05).

Table IV

Effects of shRNA on the mRNA expression of cell cycle-related genes in the Tca8113 cells (mean ± SD).

Table IV

Effects of shRNA on the mRNA expression of cell cycle-related genes in the Tca8113 cells (mean ± SD).

GeneTca8113Control-shRNAPer2-shRNA-IIIPP1P2P3
Cyclin A20.99±0.020.96±0.073.36±0.530.0000.0000.0000.911
Cyclin B10.97±0.351.02±0.222.77±0.830.010.0060.0070.926
c-myc1.10±0.171.21±0.371.39±0.330.5360.2860.5120.653
Cyclin D10.86±0.131.01±0.473.11±0.550.0010.0010.0010.672
Cyclin E1.45±0.391.31±0.531.27±0.470.8840.6510.9240.719
p532.24±0.462.38±0.470.57±0.020.0020.0020.0010.667
CDK11.14±0.241.04±0.111.07±0.130.7710.6480.8150.496
CDK21.10±0.131.23±0.401.08±0.100.7310.9060.4770.549
CDK41.06±0.280.87±0.221.79±0.330.0160.0190.0070.447
CDK61.00±0.001.00±0.252.74±0.740.0050.0030.0030.997
p161.60±0.131.70±0.080.92±0.140.0000.0000.0000.354
p213.88±0.214.05±0.601.10±0.260.0000.0000.0000.633
cdc251.46±0.401.32±0.551.62±0.540.7750.7090.4930.747
Wee11.38±0.861.11±0.201.32±0.660.8670.9170.7000.627
Rb11.16±0.391.31±0.271.25±0.250.830.7350.8020.560
E2F10.92±0.561.13±0.543.46±0.870.0060.0040.0050.710

[i] mRNA expression of p53, p16, p21, cyclin A2, B1 and D1, CDK4, CDK6, E2F1, c-myc, cyclin E, CDK1, CDK2, cdc25, Wee1 and Rb1 in the Tca8113, control-shRNA and Per2-shRNA-III groups was calculated using the 2−ΔΔCt method (mean ± SD). P represents one-way ANOVA in the three groups. P1, P2 and P3 represent the pairwise comparison with the Per2-shRNA-III and Tca8113 group, Per2-shRNA-III and control-shRNA group and control-shRNA and Tca8113 group, respectively. A value of P<0.05 represents a significant difference. PI, proliferation index; AI, apoptosis index; G0/G1, cell number at the G0 and G1 phase; G2/M, cell number at the G2 and M phase.

Discussion

Previous studies have shown that Per2 expression is reduced in various types of solid cancers, including breast and skin cancer, hepatocellular carcinoma, colorectal cancer, renal carcinomas, gastric cancers and head and neck squamous cell carcinomas (11,1924). The alteration in Per2 expression has a close relationship with the occurrence and development of cancers (6,18). The present study found that Per2 expression in OSCC Tca8113 cells was significantly lower than that in the oral mucosal epithelial cells; in Tca8113 cells, downregulation of Per2 significantly increased PI, decreased AI and altered the cell cycle distribution by significantly decreasing the number of cells in the G1/G0 phase, which suggested that the clock gene Per2 has a tumor-suppressor role in OSCC.

Cell cycle disorder is the main reason for carcinogenesis (26,27). Normal cell cycle strictly and chronologically progresses along the G1, S and G2 M phase under the precise control of the cyclin/CDK/CKI cell cycle molecular network (29). CDKs are the core of the cell cycle, to which cyclins and CKIs are the positive and negative regulators, respectively (27,29). Cyclin A2, B1, D1 and E play an important role in cyclins; and CDK1, CDK2, CDK4 and CDK6 play an important role in CDKs (27,29). CKIs contain the Ink4 family and Cip/Kip family, in which p16 and p21 play an important role, respectively (27,29). In the different cell cycle phases, cyclins, CDKs and CKIs vary. Cyclin/CDK complex formed by combinations of cyclins and CDKs can promote orderly cell cycle progression by activating CDKs. While CKIs inhibit CDKs by combining with the corresponding CDKs or the cyclin/CDK complex, which may inhibit the transformation of the cell cycle phase (27,29). To date, studies have confirmed that Per2 can regulate cyclins and p53 which is a regulator of the cell cycle checkpoint, and the mutation of Per2 expression is responsible for the aberrant expression of cyclin A, B1, D1 and E, and p53 (6,18,25,31). Both changes in cell cycle progression and imbalance of cell proliferation and apoptosis induce cancers. Previous studies have mainly focused on the role of Per2 in cyclins (6,18,25,31), but there is little research concerning the role of Per2 in the other two important aspects of the cyclin/CDK/CKI network.

At the G1 phase, the p16/p21-cyclin D1-CDK4/6-Rb1-E2F1 pathway which is an important transduction pathway of molecular information is related to the occurrence and development of tumors (32). E2F1 plays an important role in promoting the transition of cells from the G1 to the S phase (33). At the G0 and the early G1 phase, the transcriptional activity of E2F1 is inhibited by combining with unphosphorylated Rb1 at specific binding sites. At the late G1 phase, the cyclin D1/CDK4 and cyclin D1/CDK6 complex, formed by combination of cyclin D1 and CDK4/6, phosphorylate Rb1 from which E2F1 is consequently released to start DNA biosynthesis and promote cells into the S phase (34). p16 and p21, as CDKIs, can inhibit the activities of CDK4/6 by competing with cyclin D1 for CDK4/6 binding (35). The present study found that downregulation of Per2 in Tca8113 cells significantly reduced the expression of p16 and p21, significantly increased the expression of cyclin D1, CDK4, CDK6 and E2F1 and significantly reduced the number of cells at the G1 phase. These results illustrate that in Tca8113 cells, Per2 downregulation decreases the expression of p16 and p21, and consequently increases the binding of cyclin D1 and CDK4/6, which can phosphorylate Rb1 to release more E2F1 from the Rb1/E2F1 complex, resulting in promotion of the cell transformation during the G1/S phase. Fu et al (6) and Yang et al (36) both reported that the downregulation of Per2 increased the expression of cyclin D1. In the present study, there was no significant difference in the expression of Rb1 mRNA, and a difference in phosphorylated Rb1 and unphosphorylated Rb1 was not detected.

p53 is an important regulator of the G1/S cell cycle checkpoint in the cyclin/CDK/CKI network (37). In the G1/S checkpoint, p53 is activated by damaged DNA to stagnate the progression of the cell cycle, leading to either repair of the damaged DNA or apoptosis (14,27). Meanwhile p53 in the cytoplasm can directly react with the BCL-2 family to promote cell permeabilization of mitochondria and apoptosis (18). The present study found that Per2 downregulation in the Tca8113 cells significantly reduced the expression of p53, the number of cells at G1 and AI. This suggests that in Tca8113 cells, Per2 downregulation reduces the expression of p53, leading to a reduction in the repair of damaged DNA in the G1/S phase checkpoint and a decreased ability to induce apoptosis, resulting in the damaged DNA being translated into the S phase. This can destroy the integrity and stability of the cell genome, which promotes cell malignant transformation. Meanwhile, p53 can reduce the expression of cyclin B1 (25). During the G2/M phase, the absence of cyclin B1 could block cells at the G2 phase, resulting in the inability to enter into the M phase (25). Thus, in the present study, Per2 downregulation significantly reduced the expression of p53, and subsequently significantly increased the expression of cyclin B1, which accelerated mitosis and significantly increased the PI. Gotoh et al reported that Per2 is at the key site of the transcription mediated by p53, and Per2 downregulation reduces p53 expression (31). Sun et al reported that in leukemic K562 cells Per2 downregulation decreased p53 expression, and Per2 overexpression increased p53 expression (25). Hua et al reported that Per2 overexpression increased p53 expression in Lewis lung cancer cells (LLCs), decreased cell proliferation and accelerated apoptosis in LLCs and breast cancer cells (EMT6) (18). The present study was in accordance with the above reports. The present study also proved that Per2 down-regulation significantly increased and decreased the expression of E2F1 and p21, respectively, which are the dual-directional regulators of apoptosis (38,39), resulting in a worsening of the imbalance of cell proliferation and apoptosis.

The present study found that circadian clock gene Per2 was reduced in OSCC. In the Tca8113 cells, Per2 downregulation significantly increased the mRNA expression of cyclin A2, B1 and D1, CDK4, CDK6 and E2F1, while significantly decreased the mRNA expression of p53, p16 and p21. Cell proliferation was significantly higher, apoptosis was significantly lower, and progression of the cell cycle was altered. The present study represents the first demonstration that in OSCC, the clock gene Per2 plays an important role in the G1/S checkpoint and the three aspects of the cyclin/CDK/CKI network at the transcriptional level. On this basis, further research of Per2 at the protein level and the modification level after protein translation may further define the interaction of the circadian rhythm and the cell cycle, and their relationship with carcinogenesis. This may provide effective new molecular targets for the treatment of cancers.

Acknowledgments

We thank Wen-Ping Luo for her technical assistance. The present study was supported by the Project Supported by the Program for Innovation Team Building at Institutions of Higher Education in Chongqing in 2013, and the Project Supported by Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education.

References

1 

Eismann EA, Lush E and Sephton SE: Circadian effects in cancer-relevant Psychoneuroendocrine and immune pathways. Psychoneuroendocrinology. 35:963–976. 2010. View Article : Google Scholar : PubMed/NCBI

2 

Zieker D, Jenne I, Koenigsrainer I, Zdichavsky M, Nieselt K, Buck K, Zieker J, Beckert S, Glatzle J, Spanagel R, et al: Circadian expression of clock- and tumor suppressor genes in human oral mucosa. Cell Physiol Biochem. 26:155–166. 2010. View Article : Google Scholar : PubMed/NCBI

3 

Hara Y, Onishi Y, Oishi K, Miyazaki K, Fukamizu A and Ishida N: Molecular characterization of Mybbp1a as a co-repressor on the Period2 promoter. Nucleic Acids Res. 37:1115–1126. 2009. View Article : Google Scholar : PubMed/NCBI

4 

Reppert SM and Weaver DR: Coordination of circadian timing in mammals. Nature. 418:935–941. 2002. View Article : Google Scholar : PubMed/NCBI

5 

Schibler U, Ripperger J and Brown SA: Peripheral circadian oscillators in mammals: Time and food. J Biol Rhythms. 18:250–260. 2003. View Article : Google Scholar : PubMed/NCBI

6 

Fu L, Pelicano H, Liu J, Huang P and Lee C: The circadian gene Period2 plays an important role in tumor suppression and DNA damage response in vivo. Cell. 111:41–50. 2002. View Article : Google Scholar : PubMed/NCBI

7 

Rana S and Mahmood S: Circadian rhythm and its role in malignancy. J Circadian Rhythms. 8:3–17. 2010. View Article : Google Scholar : PubMed/NCBI

8 

Savvidis C and Koutsilieris M: Circadian rhythm disruption in cancer biology. Mol Med. 18:1249–1260. 2012. View Article : Google Scholar : PubMed/NCBI

9 

Mullenders J, Fabius AW, Madiredjo M, Bernards R and Beijersbergen RL: A large scale shRNA barcode screen identifies the circadian clock component ARNTL as putative regulator of the p53 tumor suppressor pathway. PLoS one. 4:e47982009. View Article : Google Scholar : PubMed/NCBI

10 

Kang TH, Reardon JT, Kemp M and Sancar A: Circadian oscillation of nucleotide excision repair in mammalian brain. Proc Natl Acad Sci USA. 106:2864–2867. 2009. View Article : Google Scholar : PubMed/NCBI

11 

Chen ST, Choo KB, Hou MF, Yeh KT, Kuo SJ and Chang JG: Deregulated expression of the PER1, PER2 and PER3 genes in breast cancers. Carcinogenesis. 26:1241–1246. 2005. View Article : Google Scholar : PubMed/NCBI

12 

Viswanathan AN, Hankinson SE and Schernhammer ES: Night shift work and the risk of endometrial cancer. Cancer Res. 67:10618–10622. 2007. View Article : Google Scholar : PubMed/NCBI

13 

Sack RL, Auckley D, Auger RR, Carskadon MA, Wright KP Jr, Vitiello MV and Zhdanova IV; American Academy of Sleep Medicine: Circadian rhythm sleep disorders: Part I, basic principles, shift work and jet lag disorders. An American Academy of Sleep Medicine Review. Sleep. 30:1460–1483. 2007.PubMed/NCBI

14 

Rengarajan T, Nandakumar N, Rajendran P, Haribabu L, Nishigaki I and Balasubramanian MP: D-pinitol promotes apoptosis in MCF-7 cells via induction of p53 and Bax and inhibition of Bcl-2 and NF-κB. Asian Pac J Cancer Prev. 15:1757–1762. 2014. View Article : Google Scholar

15 

Zhu L, Yu J, Zhang W, Xie B and Zhu Y: Research progress on the central mechanism underlying regulation of visceral biological rhythm by per2 (Review). Mol Med Rep. 10:2241–2248. 2014.PubMed/NCBI

16 

Qu X, Metz RP, Porter WW, Neuendorff N, Earnest BJ and Earnest DJ: The clock genes period 1 and period 2 mediate diurnal rhythms in dioxin-induced Cyp1A1 expression in the mouse mammary gland and liver. Toxicol Lett. 196:28–32. 2010. View Article : Google Scholar : PubMed/NCBI

17 

Yang S, Liu A, Weidenhammer A, Cooksey RC, McClain D, Kim MK, Aguilera G, Abel ED and Chung JH: The role of mPer2 clock gene in glucocorticoid and feeding rhythms. Endocrinology. 150:2153–2160. 2009. View Article : Google Scholar : PubMed/NCBI

18 

Hua H, Wang Y, Wan C, Liu Y, Zhu B, Yang C, Wang X, Wang Z, Cornelissen-Guillaume G and Halberg F: Circadian gene mPer2 overexpression induces cancer cell apoptosis. Cancer Sci. 97:589–596. 2006. View Article : Google Scholar : PubMed/NCBI

19 

Okabe T, Kumagai M, Nakajima Y, Shirotake S, Kodaira K, Oyama M, Ueno M and Ikeda M: The impact of HIF1α on the Per2 circadian rhythm in renal cancer cell lines. PLoS One. 9:e1096932014. View Article : Google Scholar

20 

Hsu CM, Lin SF, Lu CT, Lin PM and Yang MY: Altered expression of circadian clock genes in head and neck squamous cell carcinoma. Tumour Biol. 33:149–155. 2012. View Article : Google Scholar

21 

Lengyel Z, Lovig C, Kommedal S, Keszthelyi R, Szekeres G, Battyáni Z, Csernus V and Nagy AD: Altered expression patterns of clock gene mRNAs and clock proteins in human skin tumors. Tumour Biol. 34:811–819. 2013. View Article : Google Scholar

22 

Lin YM, Chang JH, Yeh KT, Yang MY, Liu TC, Lin SF, Su WW and Chang JG: Disturbance of circadian gene expression in hepatocellular carcinoma. Mol Carcinog. 47:925–933. 2008. View Article : Google Scholar : PubMed/NCBI

23 

Soták M, Polidarová L, Ergang P, Sumová A and Pácha J: An association between clock genes and clock-controlled cell cycle genes in murine colorectal tumors. Int J Cancer. 132:1032–1041. 2013. View Article : Google Scholar

24 

Hu ML, Yeh KT, Lin PM, Hsu CM, Hsiao HH, Liu YC, Lin HY, Lin SF and Yang MY: Deregulated expression of circadian clock genes in gastric cancer. BMC Gastroenterol. 14:67–76. 2014. View Article : Google Scholar : PubMed/NCBI

25 

Sun CM, Huang SF, Zeng JM, Liu DB, Xiao Q, Tian WJ, Zhu XD, Huang ZG and Feng WL: Per2 inhibits k562 leukemia cell growth in vitro and in vivo through cell cycle arrest and apoptosis induction. Pathol Oncol Res. 16:403–411. 2010. View Article : Google Scholar

26 

Murphy PJ and Campbell SS: Physiology of the circadian system in animals and humans. J Clin Neurophysiol. 13:2–16. 1996. View Article : Google Scholar : PubMed/NCBI

27 

Soták M, Sumová A and Pácha J: Cross-talk between the circadian clock and the cell cycle in cancer. Ann Med. 46:221–232. 2014. View Article : Google Scholar : PubMed/NCBI

28 

Borgs L, Beukelaers P, Vandenbosch R, Belachew S, Nguyen L and Malgrange B: Cell 'circadian' cycle: New role for mammalian core clock genes. Cell Cycle. 8:832–837. 2009. View Article : Google Scholar : PubMed/NCBI

29 

Lim S and Kaldis P: Cdks, cyclins and CKIs: Roles beyond cell cycle regulation. Development. 140:3079–3093. 2013. View Article : Google Scholar : PubMed/NCBI

30 

Štorcelová M, Vicián M, Reis R, Zeman M and Herichová I: Expression of cell cycle regulatory factors hus1, gadd45a, rb1, cdkn2a and mre11a correlates with expression of clock gene per2 in human colorectal carcinoma tissue. Mol Biol Rep. 40:6351–6361. 2013. View Article : Google Scholar : PubMed/NCBI

31 

Gotoh T, Vila-Caballer M, Santos CS, Liu J, Yang J and Finkielstein CV: The circadian factor Period 2 modulates p53 stability and transcriptional activity in unstressed cells. Mol Biol Cell. 25:3081–3093. 2014. View Article : Google Scholar : PubMed/NCBI

32 

Brennan P, Hainaut P and Boffetta P: Genetics of lung-cancer susceptibility. Lancet Oncol. 12:399–408. 2011. View Article : Google Scholar

33 

Bell LA and Ryan KM: Life and death decisions by E2F-1. Cell Death Differ. 11:137–142. 2004. View Article : Google Scholar

34 

Rivadeneira DB, Mayhew CN, Thangavel C, Sotillo E, Reed CA, Graña X and Knudsen ES: Proliferative suppression by CDK4/6 inhibition: Complex function of the retinoblastoma pathway in liver tissue and hepatoma cells. Gastroenterology. 138:1920–1930. 2010. View Article : Google Scholar : PubMed/NCBI

35 

Herbst RS, Heymach JV and Lippman SM: Lung cancer. N Engl J Med. 359:1367–1380. 2008. View Article : Google Scholar : PubMed/NCBI

36 

Yang X, Wood PA, Oh EY, Du-Quiton J, Ansell CM and Hrushesky WJ: Down regulation of circadian clock gene Period 2 accelerates breast cancer growth by altering its daily growth rhythm. Breast Cancer Res Treat. 117:423–431. 2009. View Article : Google Scholar

37 

Levine AJ: p53, the cellular gatekeeper for growth and division. Cell. 88:323–331. 1997. View Article : Google Scholar : PubMed/NCBI

38 

Hallstrom TC, Mori S and Nevins JR: An E2F1-dependent gene expression program that determines the balance between proliferation and cell death. Cancer Cell. 13:11–22. 2008. View Article : Google Scholar : PubMed/NCBI

39 

Lin PY, Fosmire SP, Park SH, Park JY, Baksh S, Modiano JF and Weiss RH: Attenuation of PTEN increases p21 stability and cytosolic localization in kidney cancer cells: A potential mechanism of apoptosis resistance. Mol Cancer. 6:16–31. 2007. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

June-2016
Volume 35 Issue 6

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

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Wang Q, Ao Y, Yang K, Tang H and Chen D: Circadian clock gene Per2 plays an important role in cell proliferation, apoptosis and cell cycle progression in human oral squamous cell carcinoma. Oncol Rep 35: 3387-3394, 2016
APA
Wang, Q., Ao, Y., Yang, K., Tang, H., & Chen, D. (2016). Circadian clock gene Per2 plays an important role in cell proliferation, apoptosis and cell cycle progression in human oral squamous cell carcinoma. Oncology Reports, 35, 3387-3394. https://doi.org/10.3892/or.2016.4724
MLA
Wang, Q., Ao, Y., Yang, K., Tang, H., Chen, D."Circadian clock gene Per2 plays an important role in cell proliferation, apoptosis and cell cycle progression in human oral squamous cell carcinoma". Oncology Reports 35.6 (2016): 3387-3394.
Chicago
Wang, Q., Ao, Y., Yang, K., Tang, H., Chen, D."Circadian clock gene Per2 plays an important role in cell proliferation, apoptosis and cell cycle progression in human oral squamous cell carcinoma". Oncology Reports 35, no. 6 (2016): 3387-3394. https://doi.org/10.3892/or.2016.4724