1
|
Guo D, Yao W, Du X, Dong J, Zhang X, Shen W, Zhu S. NEK2 promotes esophageal squamous cell carcinoma cell proliferation, migration and invasion through the Wnt/β-catenin signaling pathway. Discov Oncol 2023; 14:80. [PMID: 37233832 DOI: 10.1007/s12672-023-00692-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/04/2023] [Accepted: 05/11/2023] [Indexed: 05/27/2023] Open
Abstract
OBJECTIVES The NEK2 (never in mitosis gene A-related kinase 2), a serine/threonine kinase involved in chromosome instability and tumorigenesis. Hence, this study aimed to explore the molecular function of NEK2 in esophageal squamous cell carcinoma (ESCC). METHODS By available transcriptome datasets (GSE53625 cohort, GSE38129 cohort, and GSE21293 cohort), we analyzed the differentially expressed genes in invading and non-invading ESCC. Subsequently, we evaluated the association between NEK2 expression level and clinical outcomes through Kaplan-Meier analysis method. The quantitative real-time polymerase chain reaction (qRT-PCR) and western blotting (WB) analyses were performed to determine the expression levels of NEK2 mRNA and protein, respectively. We knocked down the NEK2 expression in ESCC cells (ECA109 and TE1), and evaluated the NEK2 biology function associated with ESCC cell proliferation, migration, invasion, and colony formation abilities. Finally, the downstream pathway of NEK2 was analyzed through Gene Set Enrichment Analysis (GSEA) and validated the regulatory mechanism of NEK2 on the potential pathway through WB. RESULTS We found that NEK2 was highly expressed in ESCC cells compared with human esophageal epithelial cells (HEEC) (P < 0.0001), and high NEK2 expression was remarkably associated with poor survival (P = 0.019). Knockdown of NEK2 showed the significant inhibitory effect for tumorigenesis, and suppressed the ESCC cells proliferation, migration, invasion, and formation of colonies abilities. Additionally, GSEA revealed that Wnt/β-catenin pathway was a downstream pathway of NEK2. WB results further validated the regulatory mechanism of NEK2 for Wnt/β-catenin signaling. CONCLUSIONS Our results indicated that NEK2 promotes ESCC cell proliferation, migration and invasion by activating the Wnt/β-catenin pathway. NEK2 could be a promising target for ESCC.
Collapse
Affiliation(s)
- Dong Guo
- Department of Radiation Oncology, Fourth Hospital of Hebei Medical University, Shijiazhuang, 050000, China
| | - Weinan Yao
- Department of Radiation Oncology, Fourth Hospital of Hebei Medical University, Shijiazhuang, 050000, China
| | - Xingyu Du
- Department of Radiation Oncology, Fourth Hospital of Hebei Medical University, Shijiazhuang, 050000, China
| | - Jing Dong
- Department of Radiation Oncology, Fourth Hospital of Hebei Medical University, Shijiazhuang, 050000, China
| | - Xueyuan Zhang
- Department of Radiation Oncology, Fourth Hospital of Hebei Medical University, Shijiazhuang, 050000, China
| | - Wenbin Shen
- Department of Radiation Oncology, Fourth Hospital of Hebei Medical University, Shijiazhuang, 050000, China
| | - Shuchai Zhu
- Department of Radiation Oncology, Fourth Hospital of Hebei Medical University, Shijiazhuang, 050000, China.
| |
Collapse
|
2
|
Huang X, Zhang G, Tang T, Gao X, Liang T. One shoot, three birds: Targeting NEK2 orchestrates chemoradiotherapy, targeted therapy, and immunotherapy in cancer treatment. Biochim Biophys Acta Rev Cancer 2022; 1877:188696. [PMID: 35157980 DOI: 10.1016/j.bbcan.2022.188696] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Revised: 02/02/2022] [Accepted: 02/04/2022] [Indexed: 12/16/2022]
Abstract
Combinational therapy has improved the cancer therapeutic landscape but is associated with a concomitant increase in adverse side reactions. Emerging evidence proposes that targeting one core target with multiple critical roles in tumors can achieve combined anti-tumor effects. This review focuses on NEK2, a member of serine/threonine kinases, with broad sequence identity to the mitotic regulator NIMA of the filamentous fungus Aspergillus nidulans. Elevated expression of NEK2 was initially found to promote tumorigeneses through abnormal regulation of the cell cycle. Subsequent studies report that NEK2 is overexpressed in a broad spectrum of tumor types and is associated with tumor progression and therapeutic resistance. Intriguingly, NEK2 has recently been revealed to mediate tumor immune escape by stabilizing the expression of PD-L1. Targeting NEK2 is thus becoming a promising approach for cancer treatment by orchestrating chemoradiotherapy, targeted therapy, and immunotherapy. It represents a novel strategy for inducing combined anti-cancer effects using a mono-agent.
Collapse
Affiliation(s)
- Xing Huang
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, Zhejiang, China; Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, Zhejiang, China; Zhejiang Clinical Research Center of Hepatobiliary and Pancreatic Diseases, Hangzhou 310003, Zhejiang, China; The Innovation Center for the Study of Pancreatic Diseases of Zhejiang Province, Hangzhou 310009, Zhejiang, China; Cancer Center, Zhejiang University, Hangzhou 310058, Zhejiang, China.
| | - Gang Zhang
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, Zhejiang, China; Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, Zhejiang, China; Zhejiang Clinical Research Center of Hepatobiliary and Pancreatic Diseases, Hangzhou 310003, Zhejiang, China; The Innovation Center for the Study of Pancreatic Diseases of Zhejiang Province, Hangzhou 310009, Zhejiang, China; Cancer Center, Zhejiang University, Hangzhou 310058, Zhejiang, China
| | - Tianyu Tang
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, Zhejiang, China; Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, Zhejiang, China; Zhejiang Clinical Research Center of Hepatobiliary and Pancreatic Diseases, Hangzhou 310003, Zhejiang, China; The Innovation Center for the Study of Pancreatic Diseases of Zhejiang Province, Hangzhou 310009, Zhejiang, China; Cancer Center, Zhejiang University, Hangzhou 310058, Zhejiang, China
| | - Xiang Gao
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, Zhejiang, China; Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, Zhejiang, China; Zhejiang Clinical Research Center of Hepatobiliary and Pancreatic Diseases, Hangzhou 310003, Zhejiang, China; The Innovation Center for the Study of Pancreatic Diseases of Zhejiang Province, Hangzhou 310009, Zhejiang, China; Cancer Center, Zhejiang University, Hangzhou 310058, Zhejiang, China
| | - Tingbo Liang
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, Zhejiang, China; Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, Zhejiang, China; Zhejiang Clinical Research Center of Hepatobiliary and Pancreatic Diseases, Hangzhou 310003, Zhejiang, China; The Innovation Center for the Study of Pancreatic Diseases of Zhejiang Province, Hangzhou 310009, Zhejiang, China; Cancer Center, Zhejiang University, Hangzhou 310058, Zhejiang, China.
| |
Collapse
|
3
|
Diez-Fraile A, De Ceulaer J, Derpoorter C, Spaas C, De Backer T, Lamoral P, Abeloos J, Lammens T. Tracking the Molecular Fingerprint of Head and Neck Cancer for Recurrence Detection in Liquid Biopsies. Int J Mol Sci 2022; 23:ijms23052403. [PMID: 35269544 PMCID: PMC8910330 DOI: 10.3390/ijms23052403] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Revised: 02/17/2022] [Accepted: 02/19/2022] [Indexed: 02/04/2023] Open
Abstract
The 5-year relative survival for patients with head and neck cancer, the seventh most common form of cancer worldwide, was reported as 67% in developed countries in the second decade of the new millennium. Although surgery, radiotherapy, chemotherapy, or combined treatment often elicits an initial satisfactory response, relapses are frequently observed within two years. Current surveillance methods, including clinical exams and imaging evaluations, have not unambiguously demonstrated a survival benefit, most probably due to a lack of sensitivity in detecting very early recurrence. Recently, liquid biopsy monitoring of the molecular fingerprint of head and neck squamous cell carcinoma has been proposed and investigated as a strategy for longitudinal patient care. These innovative methods offer rapid, safe, and highly informative genetic analysis that can identify small tumors not yet visible by advanced imaging techniques, thus potentially shortening the time to treatment and improving survival outcomes. In this review, we provide insights into the available evidence that the molecular tumor fingerprint can be used in the surveillance of head and neck squamous cell carcinoma. Challenges to overcome, prior to clinical implementation, are also discussed.
Collapse
Affiliation(s)
- Araceli Diez-Fraile
- Division of Oral and Maxillofacial Surgery, Department of Surgery, General Hospital Sint-Jan Brugge-Oostende A.V., 8000 Bruges, Belgium; (A.D.-F.); (J.D.C.); (C.S.); (T.D.B.); (P.L.); (J.A.)
| | - Joke De Ceulaer
- Division of Oral and Maxillofacial Surgery, Department of Surgery, General Hospital Sint-Jan Brugge-Oostende A.V., 8000 Bruges, Belgium; (A.D.-F.); (J.D.C.); (C.S.); (T.D.B.); (P.L.); (J.A.)
| | - Charlotte Derpoorter
- Department of Pediatric Hematology-Oncology and Stem Cell Transplantation, Ghent University Hospital, 9000 Ghent, Belgium;
- Department of Internal Medicine and Pediatrics, Ghent University, 9000 Ghent, Belgium
- Cancer Research Institute Ghent (C.R.I.G.), 9000 Ghent, Belgium
| | - Christophe Spaas
- Division of Oral and Maxillofacial Surgery, Department of Surgery, General Hospital Sint-Jan Brugge-Oostende A.V., 8000 Bruges, Belgium; (A.D.-F.); (J.D.C.); (C.S.); (T.D.B.); (P.L.); (J.A.)
| | - Tom De Backer
- Division of Oral and Maxillofacial Surgery, Department of Surgery, General Hospital Sint-Jan Brugge-Oostende A.V., 8000 Bruges, Belgium; (A.D.-F.); (J.D.C.); (C.S.); (T.D.B.); (P.L.); (J.A.)
| | - Philippe Lamoral
- Division of Oral and Maxillofacial Surgery, Department of Surgery, General Hospital Sint-Jan Brugge-Oostende A.V., 8000 Bruges, Belgium; (A.D.-F.); (J.D.C.); (C.S.); (T.D.B.); (P.L.); (J.A.)
| | - Johan Abeloos
- Division of Oral and Maxillofacial Surgery, Department of Surgery, General Hospital Sint-Jan Brugge-Oostende A.V., 8000 Bruges, Belgium; (A.D.-F.); (J.D.C.); (C.S.); (T.D.B.); (P.L.); (J.A.)
| | - Tim Lammens
- Department of Pediatric Hematology-Oncology and Stem Cell Transplantation, Ghent University Hospital, 9000 Ghent, Belgium;
- Department of Internal Medicine and Pediatrics, Ghent University, 9000 Ghent, Belgium
- Cancer Research Institute Ghent (C.R.I.G.), 9000 Ghent, Belgium
- Correspondence: ; Tel.: +32-9-332-2480
| |
Collapse
|
4
|
Chen Q, Li H, Yang L, Wen S, Huang X, Liu J, Guo X, Hu B, Li G, He M. Preparation of an anti-NEK2 monoclonal antibody and its application in liver cancer. BMC Biotechnol 2021; 21:62. [PMID: 34706700 PMCID: PMC8549277 DOI: 10.1186/s12896-021-00717-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Accepted: 09/22/2021] [Indexed: 12/12/2022] Open
Abstract
Background Never in mitosis gene-A (NIMA)-related expressed kinase 2 (NEK2) is a serine/threonine protein kinase regulated by the cell cycle. The purpose of this study was to obtain NEK2 protein to prepare an anti-NEK2 monoclonal antibody (mAb) and explore the application of the anti-NEK2 mAb of therapeutic and diagnostic in hepatocellular carcinoma (HCC). Results The NEK2 gene sequence was cloned from the normal liver cell line HL7702, and the full-length NEK2 gene sequence was cloned into the prokaryotic expression vector pET30a and transformed into Escherichia coli BL21 (DE3) cells. The recombinant fusion protein was obtained under optimized conditions and injected in BALB/c mice to prepare an anti-NEK2 mAb. By screening, we obtained a stable hybridoma cell line named 3A3 that could stably secrete anti-NEK2 mAb. Anti-NEK2 3A3 mAb was purified from ascites fluid. The isotype was IgG1, and the affinity constant (Kaff) was 6.0 × 108 L/mol. Western blot, indirect enzyme-linked immunosorbent assay (iELISA), immunofluorescence and immunocytochemical analyses showed that the mAb could specifically recognize the NEK2 protein. MTT assays showed that the mAb 3A3 could inhibit the proliferation of HCC cells. KEGG pathway analysis showed that NEK2 might affected pathways of the cell cycle. Moreover, NEK2-related genes were mainly enriched in the S and G2 phases and might act as tumor-promoting genes by regulating the S/G2 phase transition of HCC cells. Conclusions An anti-NEK2 mAb with high potency, high affinity and high specificity was prepared by prokaryotic expression system in this study and may be used in the establishment of ELISA detection kits and targeted treatment of liver cancer. Supplementary Information The online version contains supplementary material available at 10.1186/s12896-021-00717-3.
Collapse
Affiliation(s)
- Qiuli Chen
- School of Public Health, Guangxi Medical University, Nanning, 530021, China
| | - Hui Li
- School of Public Health, Guangxi Medical University, Nanning, 530021, China
| | - Lichao Yang
- School of Public Health, Guangxi Medical University, Nanning, 530021, China
| | - Sha Wen
- Laboratory Animal Center of Guangxi Medical University, Nanning, 530021, China
| | - Xuejing Huang
- Laboratory Animal Center of Guangxi Medical University, Nanning, 530021, China
| | - Jiajuan Liu
- Laboratory Animal Center of Guangxi Medical University, Nanning, 530021, China
| | - Xiaoping Guo
- Laboratory Animal Center of Guangxi Medical University, Nanning, 530021, China
| | - Bing Hu
- Laboratory Animal Center of Guangxi Medical University, Nanning, 530021, China
| | - Gang Li
- School of Public Health, Guangxi Medical University, Nanning, 530021, China.
| | - Min He
- School of Public Health, Guangxi Medical University, Nanning, 530021, China. .,Laboratory Animal Center of Guangxi Medical University, Nanning, 530021, China. .,Key Laboratory of High-Incidence-Tumor Prevention & Treatment (Guangxi Medical University), Ministry of Education, Nanning, 530021, China.
| |
Collapse
|
5
|
Chen D, Wang M, Guo Y, Wu W, Ji X, Dou X, Tang H, Zong Z, Zhang X, Xiong D. An aberrant DNA methylation signature for predicting the prognosis of head and neck squamous cell carcinoma. Cancer Med 2021; 10:5936-5947. [PMID: 34313009 PMCID: PMC8419750 DOI: 10.1002/cam4.4142] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Revised: 05/28/2021] [Accepted: 06/24/2021] [Indexed: 11/09/2022] Open
Abstract
Head and neck squamous cell carcinoma (HNSCC) is a common malignancy worldwide with a poor prognosis. DNA methylation is an epigenetic modification that plays a critical role in the etiology and pathogenesis of HNSCC. The current study aimed to develop a predictive methylation signature based on bioinformatics analysis to improve the prognosis and optimize therapeutic outcome in HNSCC. Clinical information and methylation sequencing data of patients with HNSCC were downloaded from The Cancer Genome Atlas database. The R package was used to identify differentially methylated genes (DMGs) between HNSCC and adjacent normal tissues. We identified 22 DMGs associated with 246 differentially methylated sites. Patients with HNSCC were classified into training and test groups. Cox regression analysis was used to build a risk score formula based on the five methylation sites (cg26428455, cg13754259, cg17421709, cg19229344, and cg11668749) in the training group. The Kaplan–Meier survival curves showed that the overall survival (OS) rates were significantly different between the high‐ and low‐risk groups sorted by the signature in the training group (median: 1.38 vs. 1.57 years, log‐rank test, p < 0.001). The predictive power was then validated in the test group (median: 1.34 vs. 1.75 years, log‐rank test, p < 0.001). The area under the receiver operating characteristic curve (area under the curve) based on the signature for predicting the 5‐year survival rates, was 0.7 in the training and 0.73 in test groups, respectively. The results of multivariate Cox regression analysis showed that the riskscore (RS) signature based on the five methylation sites was an independent prognostic tool for OS prediction in patients. In addition, a predictive nomogram model that incorporated the RS signature and patient clinical information was developed. The innovative methylation signature‐based model developed in our study represents a robust prognostic tool for guiding clinical therapy and predicting the OS in patients with HNSCC.
Collapse
Affiliation(s)
- Dayang Chen
- Medical Laboratory, Shenzhen Luohu People's Hospital, Shenzhen, China
| | - Mengmeng Wang
- Medical Laboratory, Shenzhen Luohu People's Hospital, Shenzhen, China.,School of Medicine, Anhui University of Science and Technology, Huainan, China
| | - Ying Guo
- Department of Clinical Laboratory, Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
| | - Wei Wu
- Medical Laboratory, Shenzhen Luohu People's Hospital, Shenzhen, China
| | - Xiang Ji
- Medical Laboratory, Shenzhen Luohu People's Hospital, Shenzhen, China
| | - Xiaowen Dou
- Medical Laboratory, Shenzhen Luohu People's Hospital, Shenzhen, China
| | - Huamei Tang
- Medical Laboratory, Shenzhen Luohu People's Hospital, Shenzhen, China
| | - Zengyan Zong
- Medical Laboratory, Shenzhen Luohu People's Hospital, Shenzhen, China.,School of Medicine, Anhui University of Science and Technology, Huainan, China
| | - Xiuming Zhang
- Medical Laboratory, Shenzhen Luohu People's Hospital, Shenzhen, China.,School of Medicine, Anhui University of Science and Technology, Huainan, China
| | - Dan Xiong
- Medical Laboratory, Shenzhen Luohu People's Hospital, Shenzhen, China.,School of Medicine, Anhui University of Science and Technology, Huainan, China
| |
Collapse
|