1
|
Chen S, Zhang Z, Zhang B, Huang Q, Liu Y, Qiu Y, Long X, Wu M, Zhang Z. CircCDK14 Promotes Tumor Progression and Resists Ferroptosis in Glioma by Regulating PDGFRA. Int J Biol Sci 2022; 18:841-857. [PMID: 35002529 PMCID: PMC8741855 DOI: 10.7150/ijbs.66114] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Accepted: 12/02/2021] [Indexed: 12/28/2022] Open
Abstract
CircRNAs have garnered significant interest in recent years due to their regulation in human tumorigenesis, yet, the function of most glioma-related circRNAs remains unclear. In this study, using RNA-Seq, we screened differentially regulated circRNAs in glioma, in comparison to non-tumor brain tissue. Loss- and gain-of-function strategies were used to assess the effect of circCDK14 on tumor progression both in vitro and in vivo. Luciferase reporter, RNA pull-down and fluorescence in situ hybridization assays were carried out to validate interactions between circCDK14 and miR-3938 as well as miR-3938 and PDGFRA. Transmission electron microscopic observation of mitochondria, iron and reactive oxygen species assays were employed for the detection of circCDK14 effect on glioma cells' sensitivity to erastin-induced ferroptosis (Fp). Our findings indicated that circCDK14 was overexpressed in glioma tissues and cell lines, and elevated levels of circCDK14 induced poor prognosis of glioma patients. CircCDK14 promotes the migration, invasion and proliferation of glioma cells in vitro as well as tumorigenesis in vivo. An evaluation of the underlying mechanism revealed that circCDK14 sponged miR-3938 to upregulate oncogenic gene PDGFRA expression. Moreover, we also found that circCDK14 reduced glioma cells' sensitivity to Fp by regulating PDGFRA expression. In conclusion, circCDK14 induces tumor in glioma and increases malignant tumor behavior via the miR-3938/PDGFRA axis. Hence, the miR-3938/PDGFRA axis may be an excellent candidate of anti-glioma therapy.
Collapse
Affiliation(s)
- Simin Chen
- School of Basic Medical Science, Central South University, Changsha 410013, Hunan, China.,Department of Clinical Laboratory, Yueyang Central Hospital, Yueyang 414000, Hunan, China
| | - Zhaoyu Zhang
- School of Basic Medical Science, Central South University, Changsha 410013, Hunan, China.,Cancer Research Institute, Central South University, Changsha 410013, Hunan, China
| | - Baoxin Zhang
- Armed Police Hospital of Hunan Province, Changsha 410013, Hunan, China
| | - Qing Huang
- School of Basic Medical Science, Central South University, Changsha 410013, Hunan, China.,Department of Clinical Laboratory, Yueyang Central Hospital, Yueyang 414000, Hunan, China
| | - Yi Liu
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha 410013, Hunan, China
| | - Yi Qiu
- School of Basic Medical Science, Central South University, Changsha 410013, Hunan, China.,Cancer Research Institute, Central South University, Changsha 410013, Hunan, China
| | - Xinmiao Long
- School of Basic Medical Science, Central South University, Changsha 410013, Hunan, China.,Cancer Research Institute, Central South University, Changsha 410013, Hunan, China
| | - Minghua Wu
- Cancer Research Institute, Central South University, Changsha 410013, Hunan, China
| | - Zuping Zhang
- School of Basic Medical Science, Central South University, Changsha 410013, Hunan, China
| |
Collapse
|
2
|
Du C, Zhang J, Zhang L, Zhang Y, Wang Y, Li J. Hsa_circRNA_102229 facilitates the progression of triple-negative breast cancer via regulating the miR-152-3p/PFTK1 pathway. J Gene Med 2021; 23:e3365. [PMID: 34031947 PMCID: PMC8459279 DOI: 10.1002/jgm.3365] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Revised: 05/17/2021] [Accepted: 05/18/2021] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND Increasing evidence has suggested that circular RNAs (circRNAs) may act as an important regulatory factor in tumor progression. However, how circRNAs exert their functions in triple-negative breast cancer (TNBC) remains not clearly understood. METHODS First, circRNA microarrays were conducted to identify aberrantly expressed circRNAs in TNBC tissues. Kaplan-Meier survival analysis was conducted to calculate the correlation between the level of hsa_circRNA_102229 and outcomes of patients with TNBC. The effect of hsa_circRNA_102229 and serine/threonine-protein kinase PFTAIRE 1 (PFTK1) on TNBC cells was clarified by cell counting kit-8, transwell and wound healing assays, as well as by a flow cytometry. The molecular mechanism of hsa_circRNA_102229 was clarified through bioinformatics, a dual-luciferase reporter assay, western blotting, fluorescence in situ hybridization and real-time polymerase chain reaction. Tumor xenograft experiments were performed to analyze growth and metastasis of TNBC in vivo. RESULTS In TNBC tissues and cells, hsa_circ_102229 was remarkably up-regulated. Patients with TNBC presenting high hsa_circ_102229 exhibited poor prognosis. Moreover, hsa_circ_102229 could promote the migration, proliferation and invasion, whereas it inhibited the apoptosis of TNBC cells. Furthermore, hsa_circ_102229 directly targeted miR-152-3p and could regulate the expression of PFTK1 by targeting miR-152-3p. Rescue assays suggested that hsa_circ_102229 may exert its function in TNBC cells by regulating PFTK1. Additionally, knockdown of hsa_circ_102229 slowed down TNBC tumorigenesis and lung metastasis in a tumor xenograft animal model. CONCLUSIONS Hsa_circ_102229 might serve as a competing endogenous RNA (ceRNA) to modulate PFTK1 expression via regulating miR-152-3p to affect the functions of TNBC cells. Hsa_circ_102229 acts as a newly discovered biomarker for TNBC treatment.
Collapse
Affiliation(s)
- Chuang Du
- Department of Breast SurgeryThe First Affiliated Hospital of Zhengzhou UniversityZhengzhou CityHenan ProvinceChina
| | - Jianhua Zhang
- Department of Breast SurgeryThe First Affiliated Hospital of Zhengzhou UniversityZhengzhou CityHenan ProvinceChina
| | - Linfeng Zhang
- Department of Breast SurgeryThe First Affiliated Hospital of Zhengzhou UniversityZhengzhou CityHenan ProvinceChina
| | - Yingying Zhang
- Department of Breast SurgeryThe First Affiliated Hospital of Zhengzhou UniversityZhengzhou CityHenan ProvinceChina
| | - Yan Wang
- Department of Breast SurgeryThe First Affiliated Hospital of Zhengzhou UniversityZhengzhou CityHenan ProvinceChina
| | - Jingruo Li
- Department of Breast SurgeryThe First Affiliated Hospital of Zhengzhou UniversityZhengzhou CityHenan ProvinceChina
| |
Collapse
|
3
|
Endo Y, Fujimoto M, Ito N, Takahashi Y, Kitago M, Gotoh M, Hiraoka N, Yoshida T, Kitagawa Y, Kanai Y, Arai E. Clinicopathological impacts of DNA methylation alterations on pancreatic ductal adenocarcinoma: prediction of early recurrence based on genome-wide DNA methylation profiling. J Cancer Res Clin Oncol 2021; 147:1341-1354. [PMID: 33635431 PMCID: PMC8021514 DOI: 10.1007/s00432-021-03541-6] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Accepted: 01/19/2021] [Indexed: 12/21/2022]
Abstract
PURPOSE The present study was conducted to clarify the clinicopathological impacts of DNA methylation alterations on pancreatic ductal adenocarcinoma (PDAC). METHODS Genome-wide DNA methylation screening was performed using the Infinium HumanMethylation450 BeadChip, and DNA methylation quantification was verified using pyrosequencing. We analyzed fresh-frozen tissues from an initial cohort (17 samples of normal control pancreatic tissue [C] from 17 patients without PDAC, and 34 samples of non-cancerous pancreatic tissue [N] and 82 samples of cancerous tissue [T] both obtained from 82 PDAC patients) and formalin-fixed paraffin-embedded T samples from 34 patients in a validation cohort. RESULTS The DNA methylation profiles of N samples tended to differ from those of C samples, and 91,907 probes showed significant differences in DNA methylation levels between C and T samples. Epigenetic clustering of T samples was significantly correlated with a larger tumor diameter and early recurrence (ER), defined as relapse within 6 months after surgery. Three marker CpG sites, applicable to formalin-fixed paraffin-embedded surgically resected materials regardless of their tumor cell content, were identified for prediction of ER. The sensitivity and specificity for detection of patients belonging to the ER group using a panel combining these three marker CpG sites, including a CpG site in the CDK14 gene, were 81.8% and 71.7% and 88.9% and 70.4% in the initial and validation cohorts, respectively. CONCLUSION These findings indicate that DNA methylation alterations may have a clinicopathological impact on PDAC. Application of our criteria will ultimately allow prediction of ER after surgery to improve the outcome of PDAC patients.
Collapse
Affiliation(s)
- Yutaka Endo
- Department of Pathology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan
- Department of Surgery, Keio University School of Medicine, Tokyo, 160-8582, Japan
| | - Mao Fujimoto
- Department of Pathology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan
| | - Nanako Ito
- Department of Pathology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan
| | - Yoriko Takahashi
- Bioscience Department, Solution Knowledge Center, Mitsui Knowledge Industry Co., Ltd., Tokyo, 105-6215, Japan
| | - Minoru Kitago
- Department of Surgery, Keio University School of Medicine, Tokyo, 160-8582, Japan
| | - Masahiro Gotoh
- Fundamental Innovative Oncology Core Center, National Cancer Center Research Institute, Tokyo, 104-0045, Japan
| | - Nobuyoshi Hiraoka
- Department of Pathology and Clinical Laboratory, National Cancer Center Hospital, Tokyo, 104-0045, Japan
| | - Teruhiko Yoshida
- Fundamental Innovative Oncology Core Center, National Cancer Center Research Institute, Tokyo, 104-0045, Japan
| | - Yuko Kitagawa
- Department of Surgery, Keio University School of Medicine, Tokyo, 160-8582, Japan
| | - Yae Kanai
- Department of Pathology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan
| | - Eri Arai
- Department of Pathology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan.
| |
Collapse
|
4
|
Jiang M, Chen Q, Zhao X, Teng Y, Yin C, Yue W. Downregulation of PFTK1 Inhibits Migration and Invasion of Non-Small Cell Lung Cancer. Onco Targets Ther 2020; 13:9281-9289. [PMID: 33061417 PMCID: PMC7519878 DOI: 10.2147/ott.s265540] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Accepted: 08/20/2020] [Indexed: 01/24/2023] Open
Abstract
Background PFTK1, a novel cyclin-dependent kinase, plays pivotal roles in tumorigenesis. Cell motility and invasiveness could be enhanced by PFTK1 in various tumors. However, the function of PFTK1 in NSCLC metastasis remains unclear. In this study, the potential role of PFTK1 in NSCLC metastasis was determined. Materials and Methods In this study, the potential function of PFTK1 in lung cancer patients was analyzed with the Kaplan–Meier plotter database. RNA interference-mediated knockdown of PFTK1 was established in two NSCLC cell lines (H1299 and 95C) to explore the role of PFTK1 in NSCLC. The efficacy of downregulation of PFTK1 was examined by Western blot and immunofluorescence. The role of PFTK1 in cell migration and invasion ability was detected by wound healing and transwell assays. The protein levels in lung cancer cells were determined by Western blot. Immunofluorescence analysis was used to evaluate the structure of filamentous actin. Results Overexpression of PFTK1 was associated with the poor survival prognosis in NSCLC patients. PFTK1 knockdown cells were constructed successfully. Suppression of PFTK1 significantly inhibited the cell migration and invasion in H1299 and 95C cells. Notably, after PFTK1 downregulation, the epithelial–mesenchymal transition (EMT) markers vimentin, ZEB1 and β-catenin were obviously decreased. Additionally, immunofluorescence analysis indicated that PFTK1 downregulation remarkably induced filamentous actin depolymerization. Conclusion In summary, PFTK1 could significantly promote lung cancer metastasis through changing EMT progress and modulating intracellular cytoskeleton F-actin expression. Taken together, our findings indicated that PFTK1 might serve as a novel therapeutic target for the inhibition of NSCLC progression.
Collapse
Affiliation(s)
- Mei Jiang
- Central Laboratory, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing 100026, People's Republic of China
| | - Qi Chen
- Central Laboratory, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing 100026, People's Republic of China
| | - Xiaoting Zhao
- Central Laboratory, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing 100026, People's Republic of China
| | - Yu Teng
- Central Laboratory, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing 100026, People's Republic of China
| | - Chenghong Yin
- Departments of Internal Medicine, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing 100026, People's Republic of China
| | - Wentao Yue
- Central Laboratory, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing 100026, People's Republic of China
| |
Collapse
|
5
|
Zhen Y, Nan Y, Guo S, Zhang L, Li G, Yue S, Liu X. Knockdown of NEAT1 repressed the malignant progression of glioma through sponging miR-107 and inhibiting CDK14. J Cell Physiol 2018; 234:10671-10679. [PMID: 30480816 DOI: 10.1002/jcp.27727] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2018] [Accepted: 10/22/2018] [Indexed: 01/03/2023]
Abstract
Aberrant expressions of long noncoding RNAs (lncRNAs) contribute to carcinogenesis via regulating tumor suppressors or oncogenes. LncRNA nuclear paraspeckle assembly transcript 1 (NEAT1) has been recognized as an oncogene to promote tumor progression of many cancers. However, the function of NEAT1 in glioma remains poorly discovered. Currently, we focused on the role of NEAT1 in glioma. Here, we found that NEAT1 was greatly upregulated in glioma cells compared with normal human astrocytes (NHAs). Meanwhile, miR-107 was significantly downregulated in glioma cell lines. Then, we observed that knockdown of NEAT1 suppressed the growth and invasion of glioma cells including U251 and SW1783 cells. Reversely, overexpression of NEAT1 dramatically induced glioma cell survival, increased cell colony formation, and promoted cell invasion ability. Subsequently, bioinformatics analysis was performed to predict the correlation between NEAT1 and miR-107. Moreover, it was revealed that NEAT1 could modulate miR-107 via serving as an endogenous sponge of miR-107. The direct binding correlation between NEAT1 and miR-107 was validated in our study. In addition, cyclin dependent kinase 14 (CDK14) was predicted as an messenger RNA target of miR-107 and the association between them was confirmed in our research. Moreover, we implied that NEAT1 demonstrated its biological functions via regulating miR-107 and CDK14 in vivo. In summary, our findings indicated that NEAT1/miR-107/CDK14 axis participated in glioma development. NEAT1 could act as a significant prognostic biomarker in glioma progression.
Collapse
Affiliation(s)
- Yingwei Zhen
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yang Nan
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China
| | - Shewei Guo
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Longzhou Zhang
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Ge Li
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Saichao Yue
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Xianzhi Liu
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
6
|
Tome-Garcia J, Erfani P, Nudelman G, Tsankov AM, Katsyv I, Tejero R, Bin Zhang, Walsh M, Friedel RH, Zaslavsky E, Tsankova NM. Analysis of chromatin accessibility uncovers TEAD1 as a regulator of migration in human glioblastoma. Nat Commun 2018; 9:4020. [PMID: 30275445 PMCID: PMC6167382 DOI: 10.1038/s41467-018-06258-2] [Citation(s) in RCA: 59] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2017] [Accepted: 08/21/2018] [Indexed: 12/17/2022] Open
Abstract
The intrinsic drivers of migration in glioblastoma (GBM) are poorly understood. To better capture the native molecular imprint of GBM and its developmental context, here we isolate human stem cell populations from GBM (GSC) and germinal matrix tissues and map their chromatin accessibility via ATAC-seq. We uncover two distinct regulatory GSC signatures, a developmentally shared/proliferative and a tumor-specific/migratory one in which TEAD1/4 motifs are uniquely overrepresented. Using ChIP-PCR, we validate TEAD1 trans occupancy at accessibility sites within AQP4, EGFR, and CDH4. To further characterize TEAD’s functional role in GBM, we knockout TEAD1 or TEAD4 in patient-derived GBM lines using CRISPR-Cas9. TEAD1 ablation robustly diminishes migration, both in vitro and in vivo, and alters migratory and EMT transcriptome signatures with consistent downregulation of its target AQP4. TEAD1 overexpression restores AQP4 expression, and both TEAD1 and AQP4 overexpression rescue migratory deficits in TEAD1-knockout cells, implicating a direct regulatory role for TEAD1–AQP4 in GBM migration. The intrinsic drivers of glioblastoma (GBM) migration are still poorly understood. Here the authors purify GBM stem cells (GSCs) from patients and profile chromatin accessibility in these cells, identifying TEAD1 as a regulator of migration in human glioblastoma.
Collapse
Affiliation(s)
- Jessica Tome-Garcia
- Department of Pathology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.,Department of Neuroscience and The Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Parsa Erfani
- Department of Pathology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.,Department of Neuroscience and The Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - German Nudelman
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | | | - Igor Katsyv
- Department of Genetics and Genomic Sciences and Icahn Institute for Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Rut Tejero
- Department of Neuroscience and The Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Bin Zhang
- Department of Genetics and Genomic Sciences and Icahn Institute for Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Martin Walsh
- Department of Pharmacological Sciences, Center for RNA Biology and Medicine, New York, NY, 10029, USA
| | - Roland H Friedel
- Department of Neuroscience and The Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Elena Zaslavsky
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Nadejda M Tsankova
- Department of Pathology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA. .,Department of Neuroscience and The Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.
| |
Collapse
|
7
|
Li Q, Zhou L, Wang M, Wang N, Li C, Wang J, Qi L. MicroRNA-613 impedes the proliferation and invasion of glioma cells by targeting cyclin-dependent kinase 14. Biomed Pharmacother 2018; 98:636-642. [PMID: 29289838 DOI: 10.1016/j.biopha.2017.12.044] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2017] [Revised: 12/05/2017] [Accepted: 12/13/2017] [Indexed: 12/11/2022] Open
Abstract
Increasing evidence has suggested that microRNAs (miRNAs) are critical regulators of tumorigenesis. MicroRNA-613 (miR-613) has recently been reported as a novel tumor-related miRNA that plays an important role in multiple cancers. However, the expression and functional significance of miR-613 in glioma remains unclear. In this study, we aimed to investigate the biological function of miR-613 in glioma. We found that miR-613 expression was frequently downregulated in glioma tissues and cell lines compared with normal controls. Overexpression of miR-613 impeded proliferation and colony formation and induced cell cycle arrest in G0/G1 phase, and also inhibited the invasive ability of glioma cells. By contrast, miR-613 inhibition had the opposite effects. Bioinformatic analysis and dual-luciferase reporter assays showed that miR-613 directly targets the 3'-untranslated region of cyclin-dependent kinase 14 (CDK14). Real-time quantitative PCR and Western blot analysis showed that CDK14 expression is negatively regulated by miR-613. In addition, miR-613 expression was inversely correlated with CDK14 expression in clinical glioma tissues. Moreover, overexpression of miR-613 decreased the protein expression of β-catenin and inhibited the activation of Wnt signaling. Importantly, the antitumor effects of miR-613 were significantly reversed by CDK14 overexpression. Overall, our results show that miR-613 inhibits glioma cell proliferation and invasion by downregulating CDK14, suggesting that miR-613 and CDK14 may serve as potential therapeutic targets for the treatment of glioma.
Collapse
Affiliation(s)
- Qi Li
- Department of Neurosurgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China.
| | - Lei Zhou
- Department of Ultrasonography, Xi'an No. 4 Hospital, Xi'an, Shaanxi 710004, China
| | - Maode Wang
- Department of Neurosurgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Ning Wang
- Department of Neurosurgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Chuankun Li
- Department of Neurosurgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Jia Wang
- Department of Neurosurgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Lei Qi
- Department of Neurosurgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| |
Collapse
|
8
|
Li Y, Zhai Y, Song Q, Zhang H, Cao P, Ping J, Liu X, Guo B, Liu G, Song J, Zhang Y, Yang A, Yan H, Yang L, Cui Y, Ma Y, Xing J, Shen X, Liu T, Zhang H, An J, Bei JX, Jia W, Kang L, Liu L, Yuan D, Hu Z, Shen H, Lu L, Wang X, Li H, He F, Zhang H, Zhou G. Genome-Wide Association Study Identifies a New Locus at 7q21.13 Associated with Hepatitis B Virus-Related Hepatocellular Carcinoma. Clin Cancer Res 2017; 24:906-915. [PMID: 29246937 DOI: 10.1158/1078-0432.ccr-17-2537] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2017] [Revised: 10/20/2017] [Accepted: 12/05/2017] [Indexed: 12/17/2022]
Abstract
Purpose: Hepatocellular carcinoma (HCC) is one of the most common cancers worldwide. In China, chronic hepatitis B virus (HBV) infection remains the major risk factor for HCC. In this study, we performed a genome-wide association study (GWAS) among Chinese populations to identify novel genetic loci contributing to susceptibility to HBV-related HCC.Experimental Design: GWAS scan is performed in a collection of 205 HBV-related HCC trios (each trio includes an affected proband and his/her both parents), and 355 chronic HBV carriers with HCC (cases) and 360 chronic HBV carriers without HCC (controls), followed by two rounds of replication studies totally consisting of 3,796 cases and 2,544 controls.Results: We identified a novel association signal within the CDK14 gene at 7q21.13 (index rs10272859, OR = 1.28, P = 9.46 × 10-10). Furthermore, we observed that the at-risk rs10272859[G] allele was significantly associated with higher mRNA expression levels of CDK14 in liver tissues. Chromosome conformation capture assays in liver cells confirmed that a physical interaction exists between the promoter region of CDK14 and the risk-associated SNPs in strong linkage disequilibrium with the index rs10272859 at 7q21.13. This index rs10272859 also showed significant association with the survival of HCC patients.Conclusions: Our findings highlight a novel locus at 7q21.13 conferring both susceptibility and prognosis to HBV-related HCC, and suggest the CDK14 gene to be the functional target of the 7q21.13 locus. Clin Cancer Res; 24(4); 906-15. ©2017 AACR.
Collapse
Affiliation(s)
- Yuanfeng Li
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, Beijing Institute of Radiation Medicine, Beijing, P.R. China.,National Engineering Research Center for Protein Drugs, Beijing, P.R. China.,National Center for Protein Sciences at Beijing, Beijing, P.R. China
| | - Yun Zhai
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, Beijing Institute of Radiation Medicine, Beijing, P.R. China.,National Engineering Research Center for Protein Drugs, Beijing, P.R. China.,National Center for Protein Sciences at Beijing, Beijing, P.R. China
| | - Qingfeng Song
- Affiliated Cancer Hospital of Guangxi Medical University, Nanning, Guangxi, P.R. China
| | - Haitao Zhang
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, Beijing Institute of Radiation Medicine, Beijing, P.R. China.,National Engineering Research Center for Protein Drugs, Beijing, P.R. China.,National Center for Protein Sciences at Beijing, Beijing, P.R. China
| | - Pengbo Cao
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, Beijing Institute of Radiation Medicine, Beijing, P.R. China.,National Engineering Research Center for Protein Drugs, Beijing, P.R. China.,National Center for Protein Sciences at Beijing, Beijing, P.R. China
| | - Jie Ping
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, Beijing Institute of Radiation Medicine, Beijing, P.R. China.,National Engineering Research Center for Protein Drugs, Beijing, P.R. China.,National Center for Protein Sciences at Beijing, Beijing, P.R. China
| | - Xinyi Liu
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, Beijing Institute of Radiation Medicine, Beijing, P.R. China.,National Engineering Research Center for Protein Drugs, Beijing, P.R. China.,National Center for Protein Sciences at Beijing, Beijing, P.R. China
| | - Bingqian Guo
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, Beijing Institute of Radiation Medicine, Beijing, P.R. China.,National Engineering Research Center for Protein Drugs, Beijing, P.R. China.,National Center for Protein Sciences at Beijing, Beijing, P.R. China
| | - Guanjun Liu
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, Beijing Institute of Radiation Medicine, Beijing, P.R. China.,National Engineering Research Center for Protein Drugs, Beijing, P.R. China.,National Center for Protein Sciences at Beijing, Beijing, P.R. China
| | - Jin Song
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, Beijing Institute of Radiation Medicine, Beijing, P.R. China.,National Engineering Research Center for Protein Drugs, Beijing, P.R. China.,National Center for Protein Sciences at Beijing, Beijing, P.R. China
| | - Ying Zhang
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, Beijing Institute of Radiation Medicine, Beijing, P.R. China.,National Engineering Research Center for Protein Drugs, Beijing, P.R. China.,National Center for Protein Sciences at Beijing, Beijing, P.R. China
| | - Aiqing Yang
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, Beijing Institute of Radiation Medicine, Beijing, P.R. China.,National Engineering Research Center for Protein Drugs, Beijing, P.R. China.,National Center for Protein Sciences at Beijing, Beijing, P.R. China
| | - Hongbo Yan
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, Beijing Institute of Radiation Medicine, Beijing, P.R. China.,National Engineering Research Center for Protein Drugs, Beijing, P.R. China.,National Center for Protein Sciences at Beijing, Beijing, P.R. China
| | - Liang Yang
- Affiliated Cancer Hospital of Guangxi Medical University, Nanning, Guangxi, P.R. China
| | - Ying Cui
- Affiliated Cancer Hospital of Guangxi Medical University, Nanning, Guangxi, P.R. China
| | - Yilong Ma
- Affiliated Cancer Hospital of Guangxi Medical University, Nanning, Guangxi, P.R. China
| | - Jinliang Xing
- State Key Laboratory of Cancer Biology, Experimental Teaching Center of Basic Medicine, Fourth Military Medical University, Xi'an, Shaanxi, P.R. China
| | - Xizhong Shen
- Department of Gastroenterology, Zhongshan Hospital of Fudan University, Shanghai, P.R. China
| | - Taotao Liu
- Department of Gastroenterology, Zhongshan Hospital of Fudan University, Shanghai, P.R. China
| | - Hongxin Zhang
- Department of Pain Treatment, Tangdu Hospital, Fourth Military Medical University, Xi'an, Shaanxi, P.R. China
| | - Jiaze An
- Department of Hepatobiliary Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, P.R. China
| | - Jin-Xin Bei
- State Key Laboratory of Oncology in Southern China, Guangzhou, Guangdong, P.R. China
| | - Weihua Jia
- State Key Laboratory of Oncology in Southern China, Guangzhou, Guangdong, P.R. China.,Department of Experimental Research, Sun Yat-Sen University Cancer Center, Guangzhou, Guangdong, P.R. China
| | - Longli Kang
- Key Laboratory for Molecular Genetic Mechanisms and Intervention Research on High Altitude Disease of Tibet Autonomous Region, School of Medicine, Xizang Minzu University, Xianyang, Shaanxi, P.R. China.,Key Laboratory of High Altitude Environment and Genes Related to Diseases of Tibet Autonomous Region, School of Medicine, Xizang Minzu University, Xianyang, Shaanxi, P.R. China
| | - Lijun Liu
- Key Laboratory for Molecular Genetic Mechanisms and Intervention Research on High Altitude Disease of Tibet Autonomous Region, School of Medicine, Xizang Minzu University, Xianyang, Shaanxi, P.R. China.,Key Laboratory of High Altitude Environment and Genes Related to Diseases of Tibet Autonomous Region, School of Medicine, Xizang Minzu University, Xianyang, Shaanxi, P.R. China
| | - Dongya Yuan
- Key Laboratory for Molecular Genetic Mechanisms and Intervention Research on High Altitude Disease of Tibet Autonomous Region, School of Medicine, Xizang Minzu University, Xianyang, Shaanxi, P.R. China.,Key Laboratory of High Altitude Environment and Genes Related to Diseases of Tibet Autonomous Region, School of Medicine, Xizang Minzu University, Xianyang, Shaanxi, P.R. China
| | - Zhibin Hu
- Department of Epidemiology and Biostatistics, Ministry of Education (MOE) Key Laboratory of Modern Toxicology, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu, P.R. China
| | - Hongbing Shen
- Department of Epidemiology and Biostatistics, Ministry of Education (MOE) Key Laboratory of Modern Toxicology, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu, P.R. China
| | - Lei Lu
- Department of Surgical Oncology, Bayi Hospital Affiliated Nanjing University of Chinese Medicine, Nanjing, Jiangsu, P.R. China
| | - Xuan Wang
- Department of Surgical Oncology, Bayi Hospital Affiliated Nanjing University of Chinese Medicine, Nanjing, Jiangsu, P.R. China
| | - Hua Li
- Department of Oncology, Chengdu Military General Hospital, Chengdu, Sichuan, P.R. China
| | - Fuchu He
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, Beijing Institute of Radiation Medicine, Beijing, P.R. China. .,National Engineering Research Center for Protein Drugs, Beijing, P.R. China.,National Center for Protein Sciences at Beijing, Beijing, P.R. China
| | - Hongxing Zhang
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, Beijing Institute of Radiation Medicine, Beijing, P.R. China. .,National Engineering Research Center for Protein Drugs, Beijing, P.R. China.,National Center for Protein Sciences at Beijing, Beijing, P.R. China
| | - Gangqiao Zhou
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, Beijing Institute of Radiation Medicine, Beijing, P.R. China. .,National Engineering Research Center for Protein Drugs, Beijing, P.R. China.,National Center for Protein Sciences at Beijing, Beijing, P.R. China
| |
Collapse
|
9
|
Mao Y, Jia Y, Zhu H, Wang W, Jin Q, Huang F, Zhang S, Li X. High expression of PFTK1 in cancer cells predicts poor prognosis in colorectal cancer. Mol Med Rep 2017; 16:224-230. [PMID: 28498444 DOI: 10.3892/mmr.2017.6560] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2016] [Accepted: 02/15/2017] [Indexed: 11/05/2022] Open
Abstract
The serine/threonine-protein kinase PFTAIRE 1 (PFTK1) is a member of the cyclin‑dependent kinase family that is highly expressed in several malignant tumors, including hepatocellular carcinoma, esophageal, breast and gastric cancers, and glioma. It contributes to tumor progression and influences tumor prognosis. However, the expression and clinicopathological significance of PFTK1 in human colorectal cancer (CRC) remain to be elucidated. The present study aimed to examine the expression of PFTK1 and to evaluate the clinical significance of its expression in human CRC. Reverse transcription‑quantitative polymerase chain reaction was performed on 10 fresh CRC and 10 surrounding normal tissue samples to detect and compare the expression of PFTK1 mRNA in CRC and normal colorectal tissues. Immunohistochemistry was performed on 179 CRC tissue specimens and 47 control samples of normal colorectal lesions to characterize the expression of PFTK1 protein. Kaplan‑Meier overall survival (OS) rate and Cox regression analyses were performed to evaluate the prognosis of patients with CRC. The expression of PFTK1 mRNA in CRC tissues (1.433±0.168) was significantly higher compared with normal tissues (0.853±0.107; t=1.97 ('t' was the value obtained from quantification of the mRNA data, following a paired t‑test), P=0.008). High PFTK1 expression in cancerous cells was detected in 92 of the CRC specimens (51.40%), and high levels of PFTK1 were associated with tumor node metastasis (TNM) stage (P=0.042), tumor classification (P=0.022) and preoperative carcinoembryonic antigen (CEA) level (P<0.001). Kaplan‑Meier OS rate and Cox regression analysis revealed that high PFTK1 expression level (hazard ratio (HR)=1.999; P=0.019) was an independent prognostic factor of CRC patients. The degree of differentiation (HR, 0.368, P=0.003), TNM classification (HR, 2.118, P=0.001) and preoperative CEA level (HR, 2.302, P=0.003) were also predictors of the prognosis of patients with CRC. The present study suggested that PFTK1 may be a potential anticancer target and prognostic marker in patients with CRC.
Collapse
Affiliation(s)
- Youjun Mao
- Department of Vascular Surgery, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215000, P.R. China
| | - Yuqing Jia
- Department of General Surgery, Friendliness Hospital, Yangzhou, Jiangsu 225009, P.R. China
| | - Huijun Zhu
- Department of Pathology, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, P.R. China
| | - Wei Wang
- Department of Pathology, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, P.R. China
| | - Qin Jin
- Department of Pathology, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, P.R. China
| | - Fang Huang
- Department of Pathology, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, P.R. China
| | - Shu Zhang
- Department of Pathology, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, P.R. China
| | - Xiaoqiang Li
- Department of Vascular Surgery, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215000, P.R. China
| |
Collapse
|
10
|
Liu MH, Shi SM, Li K, Chen EQ. Knockdown of PFTK1 Expression by RNAi Inhibits the Proliferation and Invasion of Human Non-Small Lung Adenocarcinoma Cells. Oncol Res 2017; 24:181-7. [PMID: 27458099 PMCID: PMC7838604 DOI: 10.3727/096504016x14635761799038] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
PFTK1 (PFTAIRE protein kinase 1), also named CDK14 (cyclin-dependent kinase 14), is a member of the cell division cycle 2 (CDC2)-related protein kinase family. It is highly expressed in several malignant tumors. However, the role of PFTK1 in the progression of non-small cell lung cancer (NSCLC) is still elusive. In this study, we aimed to explore the expression and function of PFTK1 in NSCLC cells. Our results showed that PFTK1 was significantly upregulated in human NSCLC cell lines. Silencing the expression of PFTK1 inhibited the proliferation of NSCLC cells. In addition, silencing the expression of PFTK1 endowed NSCLC cells with decreased migration and invasion abilities, as well as epithelial-mesenchymal transition (EMT) progress in A549 cells. A mechanistic study showed that knockdown of PFTK1 inhibited the expression of β-catenin, cyclin D1, and c-Myc in A549 cells. In summary, we report that small interfering RNA (siRNA)-PFTK1 might inhibit the proliferation and invasion of NSCLC cells by suppressing the Wnt/β-catenin signaling pathway. Therefore, PFTK1 may represent a novel therapeutic target for the treatment of NSCLC.
Collapse
Affiliation(s)
- Mei-Han Liu
- Department of Ultrasonography, China-Japan Union Hospital of Jilin University, Changchun, Jilin Province, China
| | | | | | | |
Collapse
|
11
|
Structure and inhibitor specificity of the PCTAIRE-family kinase CDK16. Biochem J 2017; 474:699-713. [PMID: 28057719 PMCID: PMC5317395 DOI: 10.1042/bcj20160941] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2016] [Revised: 01/03/2017] [Accepted: 01/05/2016] [Indexed: 12/15/2022]
Abstract
CDK16 (also known as PCTAIRE1 or PCTK1) is an atypical member of the cyclin-dependent kinase (CDK) family that has emerged as a key regulator of neurite outgrowth, vesicle trafficking and cancer cell proliferation. CDK16 is activated through binding to cyclin Y via a phosphorylation-dependent 14-3-3 interaction and has a unique consensus substrate phosphorylation motif compared with conventional CDKs. To elucidate the structure and inhibitor-binding properties of this atypical CDK, we screened the CDK16 kinase domain against different inhibitor libraries and determined the co-structures of identified hits. We discovered that the ATP-binding pocket of CDK16 can accommodate both type I and type II kinase inhibitors. The most potent CDK16 inhibitors revealed by cell-free and cell-based assays were the multitargeted cancer drugs dabrafenib and rebastinib. An inactive DFG-out binding conformation was confirmed by the first crystal structures of CDK16 in separate complexes with the inhibitors indirubin E804 and rebastinib, respectively. The structures revealed considerable conformational plasticity, suggesting that the isolated CDK16 kinase domain was relatively unstable in the absence of a cyclin partner. The unusual structural features and chemical scaffolds identified here hold promise for the development of more selective CDK16 inhibitors and provide opportunity to better characterise the role of CDK16 and its related CDK family members in various physiological and pathological contexts.
Collapse
|
12
|
Sharma R, Fedorenko I, Spence PT, Sondak VK, Smalley KSM, Koomen JM. Activity-Based Protein Profiling Shows Heterogeneous Signaling Adaptations to BRAF Inhibition. J Proteome Res 2016; 15:4476-4489. [PMID: 27934295 DOI: 10.1021/acs.jproteome.6b00613] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Patients with BRAF V600E mutant melanoma are typically treated with targeted BRAF kinase inhibitors, such as vemurafenib and dabrafenib. Although these drugs are initially effective, they are not curative. Most of the focus to date has been upon genetic mechanisms of acquired resistance; therefore, we must better understand the global signaling adaptations that mediate escape from BRAF inhibition. In the current study, we have used activity-based protein profiling (ABPP) with ATP-analogue probes to enrich kinases and other enzyme classes that contribute to BRAF inhibitor (BRAFi) resistance in four paired isogenic BRAFi-naïve/resistant cell line models. Our analysis showed these cell line models, which also differ in their PTEN status, have considerable heterogeneity in their kinase ATP probe uptake in comparing both naïve cells and adaptations to chronic drug exposure. A number of kinases including FAK1, SLK, and TAOK2 had increased ATP probe uptake in BRAFi resistant cells, while KHS1 (M4K5) and BRAF had decreased ATP probe uptake in the BRAFi-resistant cells. Gene ontology (GO) enrichment analysis revealed BRAFi resistance is associated with a significant enhancement in ATP probe uptake in proteins implicated in cytoskeletal organization and adhesion, and decreases in ATP probe uptake in proteins associated with cell metabolic processes. The ABPP approach was able to identify key phenotypic mediators critical for each BRAFi resistant cell line. Together, these data show that common phenotypic adaptations to BRAF inhibition can be mediated through very different signaling networks, suggesting considerable redundancy within the signaling of BRAF mutant melanoma cells.
Collapse
Affiliation(s)
- Ritin Sharma
- Molecular Oncology, ‡Tumor Biology, §Cutaneous Oncology, and ∥The Chemical Biology and Molecular Medicine Program, Moffitt Cancer Center & Research Institute , 12902 Magnolia Drive, Tampa, Florida 33612, United States
| | - Inna Fedorenko
- Molecular Oncology, ‡Tumor Biology, §Cutaneous Oncology, and ∥The Chemical Biology and Molecular Medicine Program, Moffitt Cancer Center & Research Institute , 12902 Magnolia Drive, Tampa, Florida 33612, United States
| | - Paige T Spence
- Molecular Oncology, ‡Tumor Biology, §Cutaneous Oncology, and ∥The Chemical Biology and Molecular Medicine Program, Moffitt Cancer Center & Research Institute , 12902 Magnolia Drive, Tampa, Florida 33612, United States
| | - Vernon K Sondak
- Molecular Oncology, ‡Tumor Biology, §Cutaneous Oncology, and ∥The Chemical Biology and Molecular Medicine Program, Moffitt Cancer Center & Research Institute , 12902 Magnolia Drive, Tampa, Florida 33612, United States
| | - Keiran S M Smalley
- Molecular Oncology, ‡Tumor Biology, §Cutaneous Oncology, and ∥The Chemical Biology and Molecular Medicine Program, Moffitt Cancer Center & Research Institute , 12902 Magnolia Drive, Tampa, Florida 33612, United States
| | - John M Koomen
- Molecular Oncology, ‡Tumor Biology, §Cutaneous Oncology, and ∥The Chemical Biology and Molecular Medicine Program, Moffitt Cancer Center & Research Institute , 12902 Magnolia Drive, Tampa, Florida 33612, United States
| |
Collapse
|
13
|
Cyclin Y regulates the proliferation, migration, and invasion of ovarian cancer cells via Wnt signaling pathway. Tumour Biol 2016; 37:10161-75. [PMID: 26831658 DOI: 10.1007/s13277-016-4818-3] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2015] [Accepted: 01/07/2016] [Indexed: 01/12/2023] Open
Abstract
This study is designated to investigate the roles of cyclin Y (CCNY) and Wnt signaling pathway in regulating ovarian cancer (OC) cell proliferation, migration, and invasion. Quantitative real-time PCR (qRT-PCR), Western blot, MTT assay, cell scratch, and transwell test were used in our study, and transplanted tumor model was constructed on nude mice. C-Myc, cyclin D1, PFTK1, ki67, OGT, and β-catenin protein expressions in tumor tissues were detected. CCNY was significantly upregulated in OC cell lines and tissues (both P < 0.05); significant association was observed between CCNY expression and clinicopathological stage, lymph node metastasis (LNM) (P < 0.05); and the CCNY expression in stages III to IV was higher than that in stages I to II, and patients with LNM had higher CCNY expression when compared with those in patients without LNM (P < 0.05); expressions of c-Myc, cyclin D, PFTK1, ki67, and OGT were upregulated in OC tissues compared with ovarian benign tissues, suggesting that these expressions were significantly different between the two groups (P < 0.05); CCNY significantly exacerbated proliferation, migration, and invasion of A2780 cells; c-Myc and cyclin D1 protein expressions increased as the expression of CCNY increased (P < 0.001); β-catenin expressions in A2780 cells with over-expression of CCNY were significantly increased in the nucleus, but significantly decreased in the cytoplasm (both P < 0.05); high expressions of CCNY exacerbated the proliferation of A2780 cells in nude mice and significantly increased c-Myc, cyclin D1, PFTK1, ki67, and OGT protein expressions in tumor tissues which were transplanted into nude mice (P < 0.01). CCNY might exacerbate the proliferation, migration, and invasion of OC cells via activating the Wnt signaling pathway. Thus, this study provides a theoretical foundation for the development of therapeutic drugs that are able to cure OC by targeting CCNY.
Collapse
|