Open Access

KIF22 promotes bladder cancer progression by activating the expression of CDCA3

Retraction in: /10.3892/ijmm.2023.5295

  • Authors:
    • Kai Li
    • Song Li
    • Shuai Tang
    • Minghao Zhang
    • Zhen Ma
    • Qi Wang
    • Fangmin Chen
  • View Affiliations

  • Published online on: October 7, 2021     https://doi.org/10.3892/ijmm.2021.5044
  • Article Number: 211
  • Copyright: © Li 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

Bladder cancer is a common malignant tumor of the urinary system and is associated with a high morbidity and mortality, due to the difficulty in the accurate diagnosis of patients with early‑stage bladder cancer and the lack of effective treatments for patients with advanced bladder cancer. Thus, novel therapeutic targets are urgently required for this disease. Kinesin family member 22 (KIF22) is a kinesin‑like DNA binding protein belonging to kinesin family, and is involved in the regulation of mitosis. KIF22 has also been reported to promote the progression of several types of cancer, such as breast cancer and melanoma. The present study demonstrates the high expression of KIF22 in human bladder cancer tissues. KIF22 was found to be associated with clinical features, including clinical stage (P=0.003) and recurrence (P=0.016), and to be associated with the prognosis of patients with bladder cancer. Furthermore, it was found that KIF22 silencing inhibited the proliferation of bladder cancer cells in vitro and tumor progression in mice. Additionally, it was noted that KIF22 transcriptionally activated cell division cycle‑associated protein 3 expression, which was also confirmed in tumors in mice. Taken together, the present study investigated the molecular mechanisms underlying the promotion of bladder cancer by KIF22 and provide a novel therapeutic target for the treatment of bladder cancer.
Introduction

Introduction

Bladder cancer is one of the most common malignant tumors of the urinary system and is associated with a high morbidity and mortality. It is also the fourth most common solid tumor among males and the seventh most common among females worldwide (1-3). Early-stage bladder cancer is difficult to diagnose due to the lack of obvious symptoms (4). The most commonly used diagnostic methods, such as urine cytology, are limited due to their high cost and high invasiveness (5,6). In addition, the treatment of advanced bladder cancer remains a challenge, and existing treatment methods, such as surgical treatment, radiotherapy and chemotherapy do not achieve satisfactory therapeutic effects (7). Notably, targeted treatment has some effect; however, existing treatment targets, such as VEGF/VEGFR and EGFR, still have limited effect (8-10). To combat this disease, novel therapeutic targets are urgently required.

Kinesin family members (KIFs) are a group of molecular motor proteins involved in the transport of cargo along the microtubule in an adenosine triphosphate (ATP)-dependent manner (11). KIFs mediate a variety of cellular functions, such as mitosis, ciliary assembly and signaling transduction (12-14). KIF22 is a kinesin-like DNA binding protein (15,16). KIF22 is essential for cell division, and is involved in spindle formation and the regulation of mitosis (17). KIF22 can also be phosphorylated by CDK1 to enhance its ability to bind to chromosomes (18). KIF22 has been reported to regulate the progression of several tumor types, such as breast cancer and melanoma (17,19). KIF22 can also promote cancer cell proliferation by coordinating CAR and EGFR dynamics (20). KIF22 has also bene found to be highly expressed in tumor cells and to promote tumor development by stimulating the transcription of cell division cycle-associated protein (CDC)25C, and it can also mediate cancer progression via the regulation of cell cycle-related proteins (18). However, the effects of KIF22 on bladder cancer remain unknown. Thus, whether KIF22 affects bladder cancer progression through transcriptional regulation is worthy of investigation.

CDCA3, a component of Skip1-cullin-F-box, has been reported to mediate the process of cell mitosis (21). A number of studies have demonstrated that CDCA3 plays an important role in cancer development. The role of CDCA3 in the regulation of the cell cycle has been well revealed. CDCA3 has been shown to promote oral cancer progression by stimulating G1 phase arrest and to affect non-small cell lung cancer by the regulation of cell cycle (22,23). In addition, CDCA3 has been found to be involved in the regulation of hepatocellular carcinoma and prostate cancer (24,25). In view of the effects of CDCA3 on multiple types of tumor, whether CDCA3 is involved in the progression of bladder cancer warrants further investigation.

In the present study, it was found KIF22 was associated with the clinicopathological features and the prognosis of patients with bladder cancer. Further analyses confirmed that KIF22 promoted the proliferation of bladder cancer both in vitro and in mice in vivo. It was also found KIF22 promoted the transcription of CDCA3. The present study demonstrated that KIF22 may serve as a potential therapeutic target for bladder cancer.

Materials and methods

Biological information

Biological information was obtained to investigate the mRNA levels of KIF22 in tumor and normal tissues and investigate the association between KIF2C and patient prognosis. Data on survival rates were obtained from The Cancer Genome Atlas (TCGA) database. Gene Expression Profiling Interactive Analysis (http://gepia.cancer-pku.cn/detail.php?gene=KIF22/) was used to collate and analyze TCGA (https://www.cancer.gov/about-nci/organization/ccg/research/structural-genomics/tcga) data with a threshold of P<0.05 and LogFC>1 or <−1 for differential genes; the median was used as the basis for dividing patients into two groups: i) The high expression, or ii) low expression groups for Kaplan-Meier survival analysis. The log rank test was used to determine any statistically significant differences in patient survival.

Antibodies, primers and plasmids

The following antibodies were used in the present study: Anti-KIF22 [1:200 dilution for immunohistochemistry (IHC), 1:2,000 dilution for western blot analysis and 1:50 dilution for chromatin immunoprecipitation (ChIP) assay; MA5-15912; Invitrogen; Thermo Fisher Scientfic, Inc.], anti-CDCA3 (1:200 dilution; ab166902; Abcam), anti-β-actin (1:2,000 dilution; 60008-1-Ig; ProteinTech Group, Inc.), anti-Ki67 (1:1,000 dilution; 27309-1-AP; ProteinTech Group, Inc.), anti-proliferating cell nuclear antigen (PCNA) (1:500 dilution; SAB2108448; Sigma-Aldrich; Merck KGaA), anti-cyclin D1 (1:1,000 dilution; ab16663, Abcam) and anti-cyclin A2 (1:1,000 dilution; ab181591; Abcam).

The primer sequences used for reverse transcription-quantitative PCR (RT-qPCR) were as follows: KIF22 forward, 5′-GAT CTC AGG AGC TGG TCG C-3′ and reverse, 5′-GTT CCA TCC ACA AAT GGC CG-3′; CDCA3 forward, 5′-TGG TAT TGC ACG GAC ACC TA-3′ and reverse, 5′-TGT TTC ACC AGT GGG CTT G-3′; and GAPDH forward, 5′-CGA CCA CTT TGT CAA GCT CA-3′ and reverse, 5′-GGT TGA GCA CAG GGT ACT TTA TT-3′.

The shRNA clone of KIF22 was purchased from Addgene, Inc. The pcDNA3.1-KIF22, pcDNA-CDCA3 and pGL-CDCA3 plasmids were constructed in the laboratory of Department of Urology of Tianjin Third Central Hospital Affiliated to Nankai University. The pcDNA3.1-vector served as the control of pcDNA3.1-KIF22 and pcDNA3.1-CDCA3, and the pGL-vector served as the control of pGL-CDCA3. The scrambled control plasmid (5′-ATG GTA CTG ACC TCC AGA G-3′) was used as the negative control (NC). The method used to harvest viral supernatant was through ultracentrifugation (600 × g, 5 min, 4°C). A total of 1×105 T24 or 5637 cells were seeded into 6-well plates and 0.5 µg plasmids were used. The cells were transfected using 10 µl Lipofectamine® 3000 (Invitrogen; Thermo Fisher Scientific, Inc.) in each well. Following incubation for 20 min at 20°C, the transfection was completed. The efficiency was measured using both reverse transcription-quantitative PCR (RT-qPCR) and western blot analysis after 48 h. Two shRNAs were initially used to avoid off-target effects, and the one with a higher silencing efficiency was selected for use in subsequent experiments in vitro and in vivo. The high silencing efficiency shRNA sequence of KIF22 was 5′-AAG CAA GAT TGG AGC TAC TCG TC-3′. The shRNA sequence of the negative control was 5′-CTT GGA GAA TGA GGC AGG GCA GA-3′.

Human tissue samples

A total of 131 human bladder cancer tissues were obtained from Tianjin Third Central Hospital (Tianjin, China) from patients who underwent transurethral resection of the bladder tumor. All procedures in this experiment were approved and conducted in accordance with the standards upheld by the Ethics Committee of Tianjin Third Central Hospital Affiliated to Nankai University. All patients signed informed consent.

IHC

The samples were fixed with 10% formalin for 24 h at 98°C, embedded with resin (Epoxy resin; Sigma-Aldrich; Merck KGaA), and divided into 5-µm-thick sections. The sections were dewaxed with xylene at 65°C, then rehydrated in a gradient ethanol series. The samples were immersed in citrate buffer (pH 6.0) at 98°C for 30 min and placed in a microwave for incubation for 10 min for antigen retrieval at 20°C. Hydrogen peroxide was then added to block endogenous peroxidase activity and the samples were incubated at 20°C for 10 min, followed by blocking with 2% BSA (Sigma-Aldrich; Merck KGaA) for 20 min at room temperature. Subsequently, the samples were incubated with the primary antibody of KIF22 and CDCA3 at room temperature for 2 h. Finally, the samples were washed with PBS four times and incubated with the secondary antibody (anti-Rabbit HRP; 1:200 dilution; ab205718; Abcam). Diaminobenzidine was used as a chromogen substrate. Images were captured using an Olympus inverted fluorescence microscope (IX71; Carl Zeiss AG).

The KIF22 protein is located in both the cytoplasm and nucleus of bladder cancer tissues (26). The expression level of KIF22 was classified into four groups based on the staining intensity (0, negative; 1, low; 2, medium; and 3, high). Additionally, the proportion of stained cells was as follows: 0, 0% stained cells; 1,1-25% stained cells; 2, 26-50% stained cells; and 3, 51-100% stained cells. A staining intensity score x the score of the percentage of stained cells <2 was considered as weak staining, 2-3 as moderate staining and >4 as strong staining.

A staining index (score, 0-12) was determined by multiplying the score for the positive area and the staining intensity. The expression of CDCA3 was scored as follows: 0, negative; 1, weak; 2, moderate; 3, strong). The property of positive cells was defined as follows: 0, <5% positively stained cells; 1, 5-25; 2, 26-50; 3, 51-75; and 4, >75% positively stained cells). A score of 0 was considered negative, scores of 1-6 were considered low expression and scores of 7-12 were considered as a high CDCA3 expression.

The sections of each patient were observed within five visual fields, and an experienced pathologist examined the sections.

Cell culture and transfection

The T24 and 5637 human bladder cancer cell lines were purchased from ATCC. Both cells were maintained in RPMI-1640 culture medium, supplemented with 10% of fetal bovine serum and incubated at 37°C in a 5% CO2 incubator.

A total of 0.5 µg control or KIF22 shRNA plasmids were transfected into the bladder cancer cells using Lipofectamine® 3000 (Invitrogen; Thermo Fisher Scientific, Inc.). The cells in the shControl (shNC) group were transfected with a negative control plasmid, and those in the shKIF22 group were transfected with a KIF22 shRNA plasmid. After 48 h, the subsequent assays were performed. The stable KIF22 knockdown cell line was screened using lentivirus infection and used in the in vivo assays.

RT-qPCR

Total RNA was extracted from the T24 and 5637 cells using TRIzol® reagent (Invitrogen; Thermo Fisher Scientific, Inc.). Subsequently, the total RNA was reverse transcribed using M-MLV reverse transcriptase at 42°C for 60 min (Promega Corporation). qPCR was conducted using the SYBR PrimeScript RT-PCR kit II (cat. no. DRR083; Takara Biotechnology Co., Ltd.) and the relative expression levels of KIF22 was normalized to the mRNA expression levels of β-actin. The following thermocycling conditions were used: Initial denaturation at 95°C for 3 min; followed by 30 cycles of denaturation at 95°C for 30 sec, annealing at 58°C for 30 sec and extension at 72°C for 30 sec. The 2−ΔΔCq method was used to quantify the results (27). The relative expression level of KIF22 was normalized to GAPDH. The primer sequences used for RT-qPCR are described above.

Western blot analysis

Bladder cancer cells or tissue samples were lysed with lysis buffer (60 mM Tris-HCl, pH 6.8, 2% SDS, 20% glycerol, 0.25% bromophenol blue, 1.25% 2-mercaptoethanol and protease inhibitor cocktail, Beyotime Institute of Biotechnology). Total protein was separated by 10% SDS-PAGE and sequentially transferred onto PVDF membranes (IPSN07852; EMD Millipore). The PVDF membranes were then blocked with 5% dry milk at room temperature for 2 h in TBST buffer and subsequently incubated with the primary antibodies, including KIF22, Ki67, PCNA, cyclin D1, cyclin A2, or β-actin antibody for 2 h at room temperature. After washing with TBST 3 times, the membranes were incubated with secondary antibody (rabbit; 1:5,000 dilution; cat. no. ab205718; Abcam) for 45 min at room temperature. Each blot was subsequently visualized with the use of an ECL kit (RPN 2109; Cytiva). The blot intensity was analyzed using ImageJ 9.0 software (National Institutes of Health).

Colony formation assay

The T24 and 5637 cells were re-suspended and plated into 6-well plates at a density of 2,000 cells/well and grown for 2 weeks. The colonies were then fixed with methanol at −20°C for 5 min and stained with 0.1% crystal violet for 20 min. Colonies were then photographed using an Olympus inverted fluorescence microscope and images captured (IX71; Carl Zeiss AG) and the differences in colony numbers between the control and KIF22-silenced bladder cancer cells were calculated. A colony was counted when it included >100 cells. The number of colonies per visual field area visible under an Olympus inverted fluorescence microscope (IX71; Carl Zeiss AG) was counted.

MTT and CCK-8 assays

Both bladder cancer cells were plated in 96-well plates at a density of 5×104 cells/well and cultured for 48 h (MTT) or 5 days (CCK-8) at 37°C. For the MTT assays, the cells were then treated with MTT (Beyotime Institute of Biotechnology) for 3 h and washed with PBS. Cells were then extracted using 150 µl DMSO and the absorbance value at a wavelength of 570 nm was measured and analyzed (28). For CCK-8 assays, the cells were then treated with CCK-8 (Beyotime, China) for 2 h and the absorbance value at a wave-length of 490 nm was measured using a Multiscan Spectrum (K3; Thermo Fisher Scientific, Inc.).

Cell cycle assay

Following transfection for 48 h, the cells were collected and washed with PBS twice. The cells were then fixed with precooled in 70% ethanol at −20°C for 1 h. Subsequently, the fixed cells were washed with PBS twice and subjected to RNase I (BD Biosciences) treatment at 37°C for 30 min. Finally, the cells were stained with propidium iodide (PI, 200 µg/ml; BD Biosciences) at 4°C for a further 30 min and analyzed using a BD FACSCalibur™ flow cytometer (BD Biosciences).

Tumor growth in vivo assay

The experiments involving animals were approved by the Institutional Animal Care and Use Committee (IACUC) of the Tianjin Third Central Hospital Affiliated to Nankai University (approval no. SYXK 2019-0318). Nude BALB/c mice were purchased from Beijing Vital River Laboratory Animal Technology Co., Ltd. A total of 16 male nude BalB/c mice (8 in each group, 8-weeks-old; 18-22 g) were purchased from Beijing Vital River Laboratory Animal Technology Co., Ltd., and fed with food and water ad libitum and at specific pathogen-free conditions (20°C; 60% humidity and alternating 12-h light/dark cycles).

For the tumor growth assay, T24 cells stably transfected with control or KIF22 shRNA lentivirus were subcutaneously injected into the right flanks of female nude mice. Almost 2 weeks later, tumors (150 mm3) were established, and the tumor volume was measured each week and calculated [length x (width)2/2]. The mice were euthanized by an intraperitoneal injection of 120 mg/kg sodium pentobarbital before the tumors were removed at the 49-day time point. The hearts of the mice were then monitored, and death was confirmed by cardiac arrest. Tumor growth curves were plotted according to the tumor volume in the different groups.

ChIP and luciferase assays

ChIP assay was performed using a ChIP assay kit (ab500; Abcam). T24 cells (~108) were cross-linked with 1% formaldehyde (Sigma-Aldrich; Merck KGaA), resuspended and lysed by RIPA buffer (Beyotime Institute of Biotechnology), then sonicated to shear the DNA into a range of 500-1,000 bp. The DNA and protein complex were then immunoprecipitated with anti-KIF22 antibody for 2 h at room temperature, and the complex was enriched using protein A agarose (Beyotime Institute of Biotechnology). Magnetic beads (Beyotime Institute of Biotechnology) were isolated and washed. Isolated DNA was further purified using a QIAquick PCR Purification kit (cat. no. 28104, Qiagen, Inc.) and amplified using AmpliTaq Gold® 360 Master Mix (Life Technologies; Thermo Fisher Scientific, Inc.).

Luciferase assay was performed using the luciferase assay system as per the manufacturer's instructions (E1500, Promega Corporation) to detect the activities of the promoter of the CDCA3 gene from which the 3′ untranslated regions (UTRs) were obtained. Briefly, T24 cells were cultured and transfected with pGL-CDCA3, pGL-Basic and pcDNA3.1 plasmids (0.5 µg) overnight using Lipofectamine® 3000 (10 µl; Invitrogen; Thermo Fisher Scientific, Inc.). Following transfection for 48 h, the cells were washed and the luciferase activities were measured following the addition of prepared solutions. The relative luciferase activities were calculated by normalizing the Firefly luciferase activity to Renilla luciferase activity.

Statistical analysis

GraphPad 5.0 software (GraphPad Software, Inc.), was used to perform the statistical analysis in the present study. Data are represented as the mean ± SEM. The statistical significance of the differences between two groups was analyzed using a unpaired Student's t-test. The statistical significance of the difference among more than two groups was analyzed using one-way ANOVA and a Turkey's post hoc test. A value of P<0.05 was considered to indicate a statistically significant difference. Kaplan-Meier survival analysis with the log-rank test was performed to assess patient prognosis, and the Chi-squared test (χ2 test) was performed to assess the association between protein expression levels and the clinical features of patients. Pearson's correlation coefficient (Pearson's R) was used to analyze the correlation between the expression of KIF22 and CDCA3 in bladder cancer tissues.

Results

KIF22 is highly expressed in tumor tissues of patients with bladder cancer

To investigate the role of KIF22 in the development of bladder cancer, the present study used tumor and adjacent tissue samples from 131 patients with surgically treated bladder cancer to detect its expression. IHC assays were conducted to detect the expression of KIF22. It was found KIF22 was localized and distributed in the cytoplasm and nucleus of the bladder cancer tissues (Fig. 1A). However, the expression of KIF22 was evidently low in the normal adjacent tissues (Fig. 1B), compared with that in the tumor tissues. These findings demonstrated the high expression of KIF22 in human bladder cancer tissues, suggesting a potential link between KIF22 and bladder cancer.

KIF22 is associated with the clinicopathological features and a poor prognosis of patients with bladder cancer

Subsequently, according to the staining intensity of KIF22 expression in the bladder tumor tissues, the samples were classified into two groups as follows: The KIF22-low (n=44) and KIF22-high (n=87; Fig. 1A) expression groups. The differences in the clinicopathological characteristics of the patients between KIF22-high and KIF22-low groups were then analyzed. The data demonstrated that KIF22 expression in bladder cancer was significantly associated with tumor stage (P=0.003) and recurrence (P=0.016), whereas no obvious association was revealed between KIF22 and other clinical features, such as patient age (P=0.422), gender (P=0.856), tumor grade (P=0.198) and lymph node metastasis (P=0.354; Table I).

Table I

Association between KIF22 expression and clinicopathological characteristics of the 131 patients with bladder cancer in the present study.

Table I

Association between KIF22 expression and clinicopathological characteristics of the 131 patients with bladder cancer in the present study.

CharacteristicAll patients (n=131)KIF22 expression
χ2 test valueP-value
Low n=44High n=87
Age (years)0.6430.422
 <65662046
 ≥65652441
Sex0.0330.856
 Male702446
 Female612041
Tumor stage9.1480.003a
 T2482424
 T3/T4832063
Tumor grade1.6590.198
 Low411724
 High902763
Lymph node metastasis0.8600.354
 Yes241014
 No1073473
Recurrence5.7930.016a
 Yes671651
 No642836

a P<0.05 (statistically significant differences). KIF22, kinesin family member 22.

Through bioinformatics analysis, it was found that KIF22 was highly expressed in bladder cancer clinical samples (Fig. 1B), consistent with the authors' expectations. In addition, by performing Kaplan-Meier analysis, it was revealed that patients with a low expression of KIF22 had a higher overall survival and disease-free survival rate, compared with those with a high KIF22 expression (Fig. 1C). Collectively, these results confirmed that KIF22 was associated with the clinical characteristics and the prognosis of patients with bladder cancer.

KIF22 knockdown inhibits the proliferation of bladder cancer cells in vitro

The present study then investigated whether KIF22 affects bladder cancer progression via the regulation of cell proliferation. KIF22 expression was silenced using KIF22 shRNA plasmids in the T24 and 5637 human bladder cancer cell lines. The silencing efficiency of KIF22 shRNA was determined using RT-qPCR and western blot analysis. The results revealed that KIF22 expression in the KIF22-shRNA-transfected T24 and 5637 bladder cancer cells was significantly decreased (Fig. 2).

On this basis, the role of KIF22 in the proliferation of bladder cancer was explored using colony formation and MTT assays. The results revealed significantly decreased colony numbers in the KIF22-silenced T24 and 5637 cells (Fig. 3A and B). Furthermore, MTT assays revealed KIF22 silencing led to a significant decrease in the proliferation of the two bladder cancer cell lines (Fig. 3C). In addition, using CCK-8 assays, it was found that the OD value decreased from day 3 following KIF22 silencing in the T24 and 5637 cells (Fig. 3D).

Subsequently, the expression of the cell proliferation-related markers, PCNA and Ki67, was examined. A markedly decreased expression of PCNA and Ki67 was observed in the group in which KIF22 was silenced, indicating a decline in the cell proliferative ability induced by KIF22 silencing (Fig. 3E-G).

KIF22 knockdown leads to bladder cancer cell cycle arrest

The disruption of the cell cycle leads to an abnormal proliferation and further triggers tumorigenesis. Thus, the present study detected the differences in the cell cycle between the cells in which KIF22 was silenced and the controls. It was noted that KIF22 silencing significantly increased the percentage of cells in the S phase and decreased that of cells in the G2/M phase (Fig. 4A). The expression of cyclin D1 and cyclin A2 was then detected and a significant decrease in expression was observed in the KIF22-silenced T24 and 5637 cells (Fig. 4B). Therefore, the knockdown of KIF22 led to a significant arrest of the cell cycle, thus suppressing the proliferation of bladder cancer cells.

KIF22 promotes the growth of bladder cancer in mice

The aforementioned results revealed a critical role of KIF22 in the regulation of the proliferation of bladder cancer cells. To further determine whether KIF22 silencing suppresses bladder cancer progression, an in vivo assay was performed. T24 cells transfected with control or KIF22 shRNA lentivirus were injected into nude mice, and after 2 weeks, the tumor volume was measured each week. Representative tumor photographs from each group were obtained and are presented in Fig. 5A. Tumor growth curves were drawn and the tumor volume in the mice injected with KIF22-silenced cells was significantly smaller than that of the controls (Fig. 5B). KIF22 expression was then detected in the tumor tissues, and the results of both western blot analysis and IHC confirmed that the expression of KIF22 in the tumor tissues from mice injected with KIF22-silenced cells was markedly decreased compared with the controls (Fig. 5C and D). Thus, these results confirmed that KIF22 promoted bladder tumorigenesis in vivo.

KIF22 promotes the growth of bladder cancer by transcriptionally activating CDCA3

The present study then investigated the mechanisms underlying the promotion of the proliferation of bladder cancer cells by KIF22. Previous research has confirmed that CDCA3 is involved in cancer progression (24).

In the present study, to confirm the transcriptional regulation of CDCA3 by KIF22 in bladder cancer cells, RT-qPCR was performed to examine the alteration in CDCA3 expression in KIF22-silenced T24 cells. As was expected, the overexpression of KIF22 led to the increased expression of CDCA3, and KIF22 silencing suppressed the expression of CDCA3 in the T24 and 5637 cells (Fig. 6A). The overexpression efficiency of KIF22 and CDCA3 in the T24 cells was further confirmed using western blot analysis (Fig. S1). Additionally, using western blot analysis, it was found that KIF22 overexpression promoted the expression of CDCA3, whereas its silencing led to a decrease in CDCA3 expression (Fig. 6B).

Figure 6

KIF22 transcriptionally activates the expression of CDCA3. (A) CDCA3 mRNA expression in the T24 (left panel) or 5637 (right panel) cells transfected with the pcDNA3.1-vector, pcDNA3.1-KIF22, control or KIF22 shRNA plasmids examined using reverse transcription-quantitative PCR. (B) CDCA3 expression in T24 cells transfected with pcDNA3.1-vector, pcDNA3.1-KIF22, control or KIF22 shRNA plasmids examined using western blot analysis. (C) CCK-8 assays exhibited the difference in the proliferation of the T24 (left panel) and 5637 (right panel) cells transfected with the indicated shRNAs and/or plasmids. (D) The effects of KIF22 on proliferation in T24 cells transfected the indicated shRNAs or plasmids were detected using colony formation assays, and the colony numbers were quantified. (E) Luciferase activity of pGL3-Basic, pGL3-CDCA3 in T24 cells co-transfected with pcDNA3.1-KIF22 or pcDNA3.1-vector plasmids examined using by dual-luciferase reporter assays. (F) PCR amplification of the anti-KIF22 antibody-enriched CDCA3 promoter fragment in T24 cells using ChIP assay. (G) Western blot analysis revealed the difference in Ki67 expression in the T24 (left panel) and 5637 (right panel) cells transfected with the indicated shRNAs and/or plasmids. The in vitro independent experiments were repeated three times. The results are presented as the mean ± SEM, *P<0.05. **P<0.01 and ***P<0.001; ns, not significant; KIF22, kinesin family member 22; cell division cycle-associated protein 3.

Furthermore, rescue assays were performed to verify whether KIF22 regulates the proliferation of bladder cancer through CDCA3. The pcDNA3.1-CDCA3 plasmid was used to reverse the defects in the proliferation of T24 cells induced by KIF22 silencing. According to the results of MTT assays, the pcDNA3.1-KIF22 plasmid was transfected into the T24 and 5637 cells, and this evidently increased cell proliferation compared with the controls (Fig. 6C). However, cell proliferation was markedly suppressed by KIF22 knockdown. Of note, it was found that CDCA3 overexpression significantly attenuated the defects in proliferation induced by KIF22 silencing in both the T24 and 5637 cells, suggesting that KIF22 promotes the proliferation of bladder cancer cells via CDCA3 (Fig. 6C). Similarly, using colony formation assays, it was found that KIF22 overexpression induced cell proliferation, and its silencing markedly suppressed cell proliferation. Additionally, it was found that CDCA3 overexpression markedly attenuated the decrease in cell proliferation induced by KIF22 silencing in the T24 and 5637 cells (Fig. 6D).

In addition, luciferase reporter assay was used to determine whether KIF22 regulates the transcription of CDCA3. The pGL-CDCA3 plasmid, which contained the promoter region of CDCA3, was co-transfected with pcDNA3.1-vector or pcDNA3.1-KIF22 plasmid (Fig. 6E). Using ChIP assay in the T24 cells, it was found that the promoter fragment of CDCA3 was specifically co-immunoprecipitated by KIF22 antibody, but not by IgG, indicating the binding of KIF22 with the promoter of CDCA3 (Fig. 6F). There results revealed that KIF22 promotes the transcriptional activation of CDCA3 promoter in T24 cells.

Furthermore, western blot analysis was performed to confirm the findings. The expression of Ki67, a marker of proliferating cells, was detected in the T24 cells in the different treatment groups. It was found that KIF22 overexpression induced the expression of Ki67, and its silencing decreased Ki67 expression. Notably, Ki67 expression was increased following the overexpression of CDCA3 in KIF22-silenced cells (Fig. 6G). Collectively, these results revealed that KIF22 transcriptionally activated the expression of CDCA3.

Co-expression of KIF22 and CDCA3 in bladder cancer tissues

The aforementioned data revealed that KIF22 could bind to the promoter site of CDCA3 and promoted its transcription, and further promoted bladder tumor development through CDCA3. The present study then aimed to confirm these findings in bladder cancer tissues. IHC was performed using tumor tissues from patients with bladder cancer. The expression level of both KIF22 and CDCA3 was detected, and the association between the expression of KIF22 and CDCA3 in bladder cancer tissues was further analyzed (Fig. 7A). Of note, it was found that the expression of CDCA3 was mainly located in the nucleus and was lower or higher in the KIF22 low or high expression groups, suggesting an evident positive association between the expression of KIF22 and CDCA3 (P<0.001, respectively) (Fig. 7A and Table II). Similarly, through Pearson's correlation analysis, it was noted that the expression of KIF22 correlated with the expression of CDCA3 in human bladder cancer tissues (Fig. 7B). Based on these data, we further confirmed that KIF22 promoted bladder cancer through regulating the transcription of CDCA3 in bladder cancer tissues.

Table II

Association and correlation between of KIF22 and CDCA3 expression in the 131 patients with bladder cancer in the present study.

Table II

Association and correlation between of KIF22 and CDCA3 expression in the 131 patients with bladder cancer in the present study.

All patients (n=131)KIF22 expression
χ2 test valueP-valuePearson's R
LowHigh
CDCA3 expression19.350<0.0010.384
 Low3228
 High1259

[i] KIF22, kinesin family member 22; cell division cycle-associated protein 3.

Therefore, it was considered that KIF22 was closely associated with the prognosis and pathological features (clinical stage and recurrence) of patients with bladder cancer. KIF22 can regulate bladder cancer cell cycle and proliferation by activating the transcription of CDCA3 and thus promoting the development of bladder cancer (Fig. 8).

Discussion

In view of the inconspicuous early symptoms of bladder cancer and the lack of effective treatment for patients with advanced bladder cancer, the mortality rate of affected patients has increased in recent years (29,30). Due to the high metastasis of bladder cancer, the existing treatment methods, such as surgical resection, radiotherapy and chemotherapy do not provide sufficient therapeutic effects (31). Targeted therapy for bladder cancer is still not highly effective (30). Given the heterogeneity of bladder cancer, novel therapeutic targets are still urgently needed (32). In the present study, it was demonstrated that KIF22 may serve as a novel molecular target for the treatment of bladder cancer.

KIFs have been reported to affect the progression of several tumor types. KIFC1 and KIF2A are highly expressed in breast cancer and have been shown to promote the proliferation and migration of breast cancer cells (33,34). KIF3B and KIF14 have been shown to be associated with the prognosis of patients with hepatocellular carcinoma (35,36). KIF1B can promote the migration of glioma cells (37). As an important member of KIFs, the present study found that KIF22 was involved in the development and progression of bladder cancer. Mechanistic analyses are required however, to better understand their specific roles in tumor growth and progression.

In the present study, it was found that KIF22 expression was markedly increased in bladder cancer tissues compared with corresponding non-tumor normal tissue. Consistent with the present findings, KIF22 has been shown to exhibit a high expression in and to promote a number of types of cancers, such as cervical, ovarian, lung and breast cancer (18-20). Additionally, it was further indicated that KIF22 promoted bladder cancer growth by stimulating cell proliferation. Similarly, several KIFs, including KIF14 and KIF18A, have been reported to be involved in mitosis, and the silencing of these KIFs inhibits cell proliferation and cancer progression (38,39). These KIFs, together with KIF22 in the present study, may be potential anti-proliferation targets for the treatment of multiple cancers. The knockdown of KIF22 led to the inhibition of bladder cancer cell proliferation, suggesting that KIF22 mediates the bladder cancer cell cycle. Notably, KIF22 silencing resulted in cell cycle arrest. The deficiency in KIF22 is known to induce abnormal mitosis, and can thus lead to a decrease in proliferation by promoting cell cycle arrest (38,39).

KIF22 has the ability of DNA binding and the activation of the transcription of downstream genes, and previous research has confirmed the transcriptional regulation function of KIF22 (40). KIF22 has been previously identified to bind to the CDC25C promoter region and negatively regulate the expression of CDC25C expression at both the mRNA and protein level (18). KIF22 silencing further improves CDK1 activity by promoting CDC25C expression, thus affecting breast cancer (18).

Of note, the present study found that KIF22 could bind to the promoter region of CDCA3 to promote its expression. Furthermore, the inhibition of cell proliferation induced by KIF22 silencing was attenuated by CDCA3 overexpression, indicating that the regulatory effects of KIF22 on cell proliferation were partly due to the function of CDCA3. Previous studies have demonstrated that the expression of CDCA3 is a potential biomarker and therapeutic target in several types of cancer, such as lung and prostate cancer (22,24).

Since CDCA3 is a factor that regulates the cell cycle, the present study focused on the effects of its potential upstream protein, KIF22, on the cell cycle and proliferation. The results suggested that KIF22 affects the proliferation of bladder cancer cells by regulating CDCA3 transcription. In fact, KIF22 may also affect the apoptosis, migration and epithelial-mesenchymal transition of bladder cancer cells; however, these were not examined in the present study, and thus the molecular mechanism of these cellular processes need to be investigated in future studies.

In the present study, it was found that KIF22 transcriptionally regulated CDCA3, and subsequent in vitro and in vivo experiments and mechanistic analyses confirmed the regulatory effects of KIF22 on CDCA3. However, it remains unknown as to whether other mechanisms may be involved in the regulatory effects of KIF22 on bladder cancer cell proliferation; thus, further studies are required on this matter. Consistent with these studies, the present study demonstrated that KIF22 transcriptionally regulated CDCA3 and therefore promoted cell proliferation.

Supplementary Data

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

KL, SL and FC contributed to the conception and design of the study. ST and MZ performed the data analysis and wrote the manuscript. ZM and QW participated in the design of the study and in the statistical analysis of the data. KL and SL confirm the authenticity of all the raw data. All authors have read and approved the final manuscript.

Ethics approval and consent to participate

All procedures performed in the present study were approved by the Ethics Committee of the Tianjin Third Central Hospital Affiliated to Nankai University. Written informed consent was obtained from all patients or their families. The animal experiments were approved by the Institutional Animal Care and Use Committee (IACUC) of Tianjin Third Central Hospital Affiliated to Nankai University (Approval no. SYXK 2019-0318).

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Acknowledgments

Not applicable.

References

1 

Franekova M, Halasova E, Bukovska E, Luptak J and Dobrota D: Gene polymorphisms in bladder cancer. Urol Oncol. 26:1–8. 2008. View Article : Google Scholar : PubMed/NCBI

2 

Kochańska-Dziurowicz AA, Mielniczuk MJ, Bijak A and Palugniok R: Estimation of usefulness of monitoring tissue polypeptide antigen-TPA-M concentrations in the effectiveness surgical treatment of urinary bladder cancer. Nucl Med Rev Cent East Eur. 5:109–111. 2002.

3 

McLellan RA, French CG and Bell DG: Trends in the incidence of bladder cancer in Nova Scotia: A twenty-year perspective. Can J Urol. 10:1880–1884. 2003.PubMed/NCBI

4 

Dalbagni G: The management of superficial bladder cancer. Nat Clin Pract Urol. 4:254–260. 2007. View Article : Google Scholar : PubMed/NCBI

5 

Kiyoshima K, Akitake M, Shiota M, Takeuchi A, Takahashi R, Inokuchi J, Tatsugami K, Yokomizo A and Eto M: Prognostic significance of preoperative urine cytology in low-grade non-muscleinvasive bladder cancer. Anticancer Res. 36:799–802. 2016.PubMed/NCBI

6 

Lavery HJ, Zaharieva B, McFaddin A, Heerema N and Pohar KS: A prospective comparison of UroVysion FISH and urine cytology in bladder cancer detection. BMC Cancer. 17:2472017. View Article : Google Scholar : PubMed/NCBI

7 

Ikushima H, Iwamoto S, Osaki K, Furutani S, Yamashita K, Kawanaka T, Kubo A, Takegawa Y, Kudoh T, Kanayama H and Nishitani H: Effective bladder preservation strategy with low-dose radiation therapy and concurrent intraarterial chemotherapy for muscle-invasive bladder cancer. Radiat Med. 26:156–163. 2008. View Article : Google Scholar : PubMed/NCBI

8 

Black PC, Agarwal PK and Dinney CP: Targeted therapies in bladder cancer-an update. Urol Oncol. 25:433–438. 2007. View Article : Google Scholar : PubMed/NCBI

9 

Li Y, Yang X, Su LJ and Flaig TW: VEGFR and EGFR inhibition increases epithelial cellular characteristics and chemotherapy sensitivity in mesenchymal bladder cancer cells. Oncol Rep. 24:1019–1028. 2010.PubMed/NCBI

10 

Huang Z, Zhang M, Chen G, Wang W, Zhang P, Yue Y, Guan Z, Wang X and Fan J: Bladder cancer cells interact with vascular endothelial cells triggering EGFR signals to promote tumor progression. Int J Oncol. 54:1555–1566. 2019.PubMed/NCBI

11 

Miki T, Nishina M and Goshima G: RNAi screening identifies the armadillo repeat-containing kinesins responsible for microtubule-dependent nuclear positioning in Physcomitrella patens. Plant Cell Physiol. 56:737–749. 2015. View Article : Google Scholar : PubMed/NCBI

12 

Gould R, Freund C, Palmer F, Knapp PE, Huang J, Morrison H and Feinstein DL: Messenger RNAs for kinesins and dynein are located in neural processes. Biol Bull. 197:259–260. 1999. View Article : Google Scholar : PubMed/NCBI

13 

Hu Z, Liang Y, Meng D, Wang L and Pan J: Microtubule depolymerizing kinesins in the regulation of assembly, disassembly, and length of cilia and flagella. Int Rev Cell Mol Biol. 317:241–265. 2015. View Article : Google Scholar

14 

Vicente JJ and Wordeman L: Mitosis, microtubule dynamics and the evolution of kinesins. Exp Cell Res. 334:61–69. 2015. View Article : Google Scholar : PubMed/NCBI

15 

Min BJ, Kim N, Chung T, Kim OH, Nishimura G, Chung CY, Song HR, Kim HW, Lee HR, Kim J, et al: Whole-exome sequencing identifies mutations of KIF22 in spondyloepimetaphyseal dysplasia with joint laxity, leptodactylic type. Am J Hum Genet. 89:760–766. 2011. View Article : Google Scholar : PubMed/NCBI

16 

Park SM, Littleton JT, Park HR and Lee JH: Drosophila homolog of human KIF22 at the autism-linked 16p112 loci influences synaptic connectivity at larval neuromuscular junctions. Exp Neurobiol. 25:33–39. 2016. View Article : Google Scholar : PubMed/NCBI

17 

Bruzzoni-Giovanelli H, Fernandez P, Veiga L, Podgorniak MP, Powell DJ, Candeias MM, Mourah S, Calvo F and Marín M: Distinct expression patterns of the E3 ligase SIAH-1 and its partner Kid/KIF22 in normal tissues and in the breast tumoral processes. J Exp Clin Cancer Res. 29:102010. View Article : Google Scholar : PubMed/NCBI

18 

Yu Y, Wang XY, Sun L, Wang YL, Wan YF, Li XQ and Feng YM: Inhibition of KIF22 suppresses cancer cell proliferation by delaying mitotic exit through upregulating CDC25C expression. Carcinogenesis. 35:1416–1425. 2014. View Article : Google Scholar : PubMed/NCBI

19 

Manning CS, Hooper S and Sahai EA: Intravital imaging of SRF and Notch signalling identifies a key role for EZH2 in invasive melanoma cells. Oncogene. 34:4320–4332. 2015. View Article : Google Scholar :

20 

Pike R, Ortiz-Zapater E, Lumicisi B, Santis G and Parsons M: KIF22 coordinates CAR and EGFR dynamics to promote cancer cell proliferation. Sci Signal. 11:eaaq10602018. View Article : Google Scholar : PubMed/NCBI

21 

Ayad NG, Rankin S, Murakami M, Jebanathirajah J, Gygi S and Kirschner MW: Tome-1, a trigger of mitotic entry, is degraded during G1 via the APC. Cell. 113:101–113. 2003. View Article : Google Scholar : PubMed/NCBI

22 

Adams MN, Burgess JT, He Y, Gately K, Snell C, Zhang SD, Hooper JD, Richard DJ and O'Byrne KJ: Expression of CDCA3 is a prognostic biomarker and potential therapeutic target in non-small cell lung cancer. J Thorac Oncol. 12:1071–1084. 2017. View Article : Google Scholar : PubMed/NCBI

23 

Uchida F, Uzawa K, Kasamatsu A, Takatori H, Sakamoto Y, Ogawara K, Shiiba M, Tanzawa H and Bukawa H: Overexpression of cell cycle regulator CDCA3 promotes oral cancer progression by enhancing cell proliferation with prevention of G1 phase arrest. BMC Cancer. 12:3212012. View Article : Google Scholar : PubMed/NCBI

24 

Chen J, Zhu S, Jiang N, Shang Z, Quan C and Niu Y: HoxB3 promotes prostate cancer cell progression by transactivating CDCA3. Cancer Lett. 330:217–224. 2013. View Article : Google Scholar

25 

Hu Q, Fu J, Luo B, Huang M, Guo W, Lin Y, Xie X and Xiao S: OY-TES-1 may regulate the malignant behavior of liver cancer via NANOG, CD9, CCND2 and CDCA3: A bioinformatic analysis combine with RNAi and oligonucleotide microarray. Oncol Rep. 33:1965–1975. 2015. View Article : Google Scholar : PubMed/NCBI

26 

Li B, Zhu FC, Yu SX, Liu SJ and Li BY: Suppression of KIF22 inhibits cell proliferation and xenograft tumor growth in colon cancer. Cancer Biother Radiopharm. 35:50–57. 2020. View Article : Google Scholar

27 

Livak KJ and Schmittgen TD: Analysis of relative gene expression data using real-time quantitative PCR and the 2(-Delta Delta C(T)) method. Methods. 25:402–408. 2001. View Article : Google Scholar

28 

Zhao Y, Zhao Z, Cui Y, Chen X, Chen C, Xie C, Qin B and Yang Y: Redox-responsive glycosylated combretastatin A-4 derivative as novel tubulin polymerization inhibitor for glioma and drug delivery. Drug Develop Res. Sep 29–2021.Epub ahead of print. View Article : Google Scholar

29 

Obermann EC, Meyer S, Hellge D, Zaak D, Filbeck T, Stoehr R, Hofstaedter F, Hartmann A and Knuechel R: Fluorescence in situ hybridization detects frequent chromosome 9 deletions and aneuploidy in histologically normal urothelium of bladder cancer patients. Oncol Rep. 11:745–751. 2004.PubMed/NCBI

30 

van Kessel KE, Zuiverloon TC, Alberts AR, Boormans JL and Zwarthoff EC: Targeted therapies in bladder cancer: An overview of in vivo research. Nat Rev Urol. 12:681–694. 2015. View Article : Google Scholar : PubMed/NCBI

31 

Hautmann RE, Abol-Enein H, Davidsson T, Gudjonsson S, Hautmann SH, Holm HV, Lee CT, Liedberg F, Madersbacher S, Manoharan M, et al: ICUD-EAU international consultation on bladder cancer 2012: Urinary diversion. Eur Urol. 63:67–80. 2013. View Article : Google Scholar

32 

Liu J, Zhang Y, Yu C, Zhang P, Gu S, Wang G, Xiao H and Li S: Bergenin inhibits bladder cancer progression via activating the PPARγ/PTEN/Akt signal pathway. Drug Dev Res. 82:278–286. 2021. View Article : Google Scholar

33 

Li Y, Lu W, Chen D, Boohaker RJ, Zhai L, Padmalayam I, Wennerberg K, Xu B and Zhang W: KIFC1 is a novel potential therapeutic target for breast cancer. Cancer Biol Ther. 16:1316–1322. 2015. View Article : Google Scholar : PubMed/NCBI

34 

Wang J, Ma S, Ma R, Qu X, Liu W, Lv C, Zhao S and Gong Y: KIF2A silencing inhibits the proliferation and migration of breast cancer cells and correlates with unfavorable prognosis in breast cancer. BMC Cancer. 14:4612014. View Article : Google Scholar : PubMed/NCBI

35 

Huang X, Liu F, Zhu C, Cai J, Wang H, Wang X, He S, Liu C, Yao L, Ding Z, et al: Suppression of KIF3B expression inhibits human hepatocellular carcinoma proliferation. Dig Dis Sci. 59:795–806. 2014. View Article : Google Scholar :

36 

Xu H, Choe C, Shin SH, Park SW, Kim HS, Jung SH, Yim SH, Kim TM and Chung YJ: Silencing of KIF14 interferes with cell cycle progression and cytokinesis by blocking the p27(Kip1) ubiquitination pathway in hepatocellular carcinoma. Exp Mol Med. 46:e972014. View Article : Google Scholar : PubMed/NCBI

37 

Chen S, Han M, Chen W, He Y, Huang B, Zhao P, Huang Q, Gao L, Qu X and Li X: KIF1B promotes glioma migration and invasion via cell surface localization of MT1-MMP. Oncol Rep. 35:971–977. 2016. View Article : Google Scholar

38 

Thériault BL, Basavarajappa HD, Lim H, Pajovic S, Gallie BL and Corson TW: Transcriptional and epigenetic regulation of KIF14 overexpression in ovarian cancer. PLoS One. 9:e915402014. View Article : Google Scholar : PubMed/NCBI

39 

Zhang C, Zhu C, Chen H, Li L, Guo L, Jiang W and Lu SH: Kif18A is involved in human breast carcinogenesis. Carcinogenesis. 31:1676–1684. 2010. View Article : Google Scholar : PubMed/NCBI

40 

Maddika S, Sy SM and Chen J: Functional interaction between Chfr and Kif22 controls genomic stability. J Biol Chem. 284:12998–13003. 2009. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

December-2021
Volume 48 Issue 6

Print ISSN: 1107-3756
Online ISSN:1791-244X

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Li K, Li S, Tang S, Zhang M, Ma Z, Wang Q and Chen F: KIF22 promotes bladder cancer progression by activating the expression of CDCA3 Retraction in /10.3892/ijmm.2023.5295. Int J Mol Med 48: 211, 2021
APA
Li, K., Li, S., Tang, S., Zhang, M., Ma, Z., Wang, Q., & Chen, F. (2021). KIF22 promotes bladder cancer progression by activating the expression of CDCA3 Retraction in /10.3892/ijmm.2023.5295. International Journal of Molecular Medicine, 48, 211. https://doi.org/10.3892/ijmm.2021.5044
MLA
Li, K., Li, S., Tang, S., Zhang, M., Ma, Z., Wang, Q., Chen, F."KIF22 promotes bladder cancer progression by activating the expression of CDCA3 Retraction in /10.3892/ijmm.2023.5295". International Journal of Molecular Medicine 48.6 (2021): 211.
Chicago
Li, K., Li, S., Tang, S., Zhang, M., Ma, Z., Wang, Q., Chen, F."KIF22 promotes bladder cancer progression by activating the expression of CDCA3 Retraction in /10.3892/ijmm.2023.5295". International Journal of Molecular Medicine 48, no. 6 (2021): 211. https://doi.org/10.3892/ijmm.2021.5044