1
|
Chen X, Wu Y, Xing Y, Zhong P. CENPF as a prognostic marker of glioma: unraveling the molecular mechanisms. J Cancer Res Clin Oncol 2025; 151:96. [PMID: 40019588 PMCID: PMC11870995 DOI: 10.1007/s00432-025-06144-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2025] [Accepted: 02/17/2025] [Indexed: 03/01/2025]
Abstract
OBJECTIVE Glioma is the dominant primary intracranial malignancy. The roles of CENPF and the CENPF - p53 axis in glioma remain elusive. This study uses bioinformatics and animal experiments to clarify the relationship between CENPF and p53 in glioma. CENPF affects spindle assembly and chromosomal segregation, while p53 is a tumor-suppressor gene. Their dysregulation may interact and impact glioma development. Our research aims to uncover the underlying molecular mechanisms, offering new perspectives for glioma diagnosis and treatment. METHOD Gene expression data from the Gene Expression Omnibus (GEO) database ( http://www.ncbi.nlm.nih.gov/geo/ ) were retrieved, specifically datasets GSE50161, GSE104291, and GSE12249. Volcano plots were generated to visualize differentially expressed genes (DEGs), and intersecting DEGs were identified using Venn diagrams. Weighted gene co-expression network analysis (WGCNA) was employed to construct and analyze the protein-protein interaction (PPI) network. Additionally, gene ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analyses were conducted. Gene set enrichment analysis (GSEA) was utilized for comprehensive GO and KEGG analyses of the entire genome. Comparative Toxicogenomics Database (CTD) analysis was performed, and TargetScan was used to identify miRNAs regulating central DEGs. An animal model of glioma was established and analyzed via Western blot. RESULT A total of 501 differentially expressed genes (DEGs) were identified, from which eight significant modules were generated and ten core genes were extracted. These core genes exhibited differential expression patterns between glioma tumor and non-tumor samples. Expression analysis revealed that the ten core genes associated with glioma (CENPF, PBK, ASPM, KIF2C, KIF20A, CDC20, TOP2A, NUSAP1, TTK, KIF23) were significantly upregulated in tumor tissues (P < 0.05). They are primarily enriched in protein signal transduction, coated membrane structures, AP-type membrane coat adaptor complexes, and chloride channel activity. KEGG pathway analysis indicated that these target genes were mainly involved in nicotine addiction, arginine and proline metabolism, beta-alanine metabolism, and histidine metabolism. The mouse model confirmed that CENPF and CDK-1 were highly expressed in glioma tissues, while p53, p21, and Caspase9 were downregulated, leading to inhibition of the apoptosis pathway and exacerbation of glioma progression. Overexpression of CENPF further suppressed key molecules in the p53-mediated apoptosis pathway. Conversely, low expression of CENPF activated these key molecules, inducing apoptosis in glioma cells. CONCLUSIONS CENPF exhibits elevated expression levels in glioma, potentially inhibiting cell apoptosis via the p53 signaling pathway, consequently contributing to the onset and progression of glioma.
Collapse
Affiliation(s)
- Xiuyang Chen
- Department of Neurological Care Unit, The Affliated Yantai Yuhuangding Hospital of Qingdao University, No. 20, Yuhuangding East Road, Zhifu District, Yantai, 264000, Shandong, People's Republic of China
| | - Yiwei Wu
- Department of Orthopaedic Surgery, Yantaishan Hospital, Yantai, 264000, Shandong, People's Republic of China
| | - Yining Xing
- Department of Neurological Care Unit, The Affliated Yantai Yuhuangding Hospital of Qingdao University, No. 20, Yuhuangding East Road, Zhifu District, Yantai, 264000, Shandong, People's Republic of China
- Department of Orthopaedic Surgery, Yantaishan Hospital, Yantai, 264000, Shandong, People's Republic of China
| | - Peng Zhong
- Department of Neurological Care Unit, The Affliated Yantai Yuhuangding Hospital of Qingdao University, No. 20, Yuhuangding East Road, Zhifu District, Yantai, 264000, Shandong, People's Republic of China.
| |
Collapse
|
2
|
Xie Z, Lin H, Wu Y, Yu Y, Liu X, Zheng Y, Wang X, Wu J, Xu M, Han Y, Zhang Q, Deng Y, Lin L, Linzhu Y, Qingyun L, Lin X, Huang Y, Chi P. USP4-mediated CENPF deubiquitylation regulated tumor metastasis in colorectal cancer. Cell Death Dis 2025; 16:81. [PMID: 39922805 PMCID: PMC11807140 DOI: 10.1038/s41419-025-07424-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Revised: 01/14/2025] [Accepted: 02/03/2025] [Indexed: 02/10/2025]
Abstract
Metastasis is a major challenge for colorectal cancer (CRC) treatment. In this study, we identified autophagy activation as a prognostic indicator in CRC and observed that the expression of key autophagy proteins is elevated in metastatic and recurrent cases. Our subsequent goal was to identify potential genes associated with the autophagy panel and assess their prognostic significance, biological roles, and mechanisms in CRC metastasis. Among the candidates, CENPF emerged as the top gene in our screening process. We found that CENPF expression was preferentially elevated in CRC tissues compared to adjacent normal tissues, with significantly higher levels in CRC patients with tumor recurrence. Furthermore, a multicenter cohort study demonstrated that upregulated CENPF expression was strongly associated with poorer disease-free survival in CRC. Functional experiments showed that CENPF knockdown inhibited CRC cell invasion and metastasis both in vitro and in vivo. Intriguingly, we found CENPF undergoes degradation in CRC via the ubiquitination-proteasome pathway. Mechanistically, we observed that USP4 interacted with and stabilized CENPF via deubiquitination. Furthermore, USP4-mediated CENPF upregulation was critical regulators of metastasis of CRC. Examination of clinical samples confirmed that USP4 expression positively correlates with CENPF protein expression, but not mRNA transcript levels. Taken together, this study describes a novel USP4-CENPF signaling axis which is crucial for CRC metastasis, potentially serving as a therapeutic target and a promising prognostic biomarker for CRC.
Collapse
Affiliation(s)
- Zhongdong Xie
- Department of Colorectal Surgery, Union Hospital, Fujian Medical University, Fuzhou, China
- Department of Colorectal Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Hanbin Lin
- Central Laboratory, Affiliated Hospital of Putian University, Putian, China
- Key Laboratory of Gastrointestinal Cancer (Fujian Medical University), Ministry of Education, Fuzhou, China
| | - Yuecheng Wu
- Key Laboratory of Gastrointestinal Cancer (Fujian Medical University), Ministry of Education, Fuzhou, China
| | - Yanan Yu
- Guilin Medical University, Guilin, China
| | - Xintong Liu
- Central Laboratory, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Yating Zheng
- Department of Colorectal Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Xiaojie Wang
- Department of Colorectal Surgery, Union Hospital, Fujian Medical University, Fuzhou, China
| | - Jiashu Wu
- Department of Science and Technology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Meifang Xu
- Department of Pathology, Union Hospital, Fujian Medical University, Fuzhou, China
| | - Yuting Han
- Key Laboratory of Gastrointestinal Cancer (Fujian Medical University), Ministry of Education, Fuzhou, China
| | - Qiongying Zhang
- Department of Pathology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Yu Deng
- Department of Colorectal Surgery, Union Hospital, Fujian Medical University, Fuzhou, China
| | - Lin Lin
- Department of Pathology, Union Hospital, Fujian Medical University, Fuzhou, China
| | - Yan Linzhu
- Department of Colorectal Surgery, Union Hospital, Fujian Medical University, Fuzhou, China
| | - Li Qingyun
- Department of Colorectal Surgery, Union Hospital, Fujian Medical University, Fuzhou, China
| | - Xinjian Lin
- Key Laboratory of Gastrointestinal Cancer (Fujian Medical University), Ministry of Education, Fuzhou, China.
| | - Ying Huang
- Department of Colorectal Surgery, Union Hospital, Fujian Medical University, Fuzhou, China.
| | - Pan Chi
- Department of Colorectal Surgery, Union Hospital, Fujian Medical University, Fuzhou, China.
| |
Collapse
|
3
|
Li J, Liu L, Zong G, Yang Z, Zhang D, Zhao B. Knockdown of CENPF induces cell cycle arrest and inhibits epithelial‑mesenchymal transition progression in glioma. Oncol Lett 2025; 29:61. [PMID: 39611064 PMCID: PMC11602827 DOI: 10.3892/ol.2024.14807] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Accepted: 10/02/2024] [Indexed: 11/30/2024] Open
Abstract
Gliomas are among the most common malignant tumors of the central nervous system. Despite surgical resection followed by postoperative radiotherapy and chemotherapy, their prognosis remains unfavorable. The present study aimed to assess new mechanisms and explore promising prognostic biomarkers for patients with glioma using comprehensive bioinformatics analysis and in vitro and in vivo assays. Overlapping differentially expressed genes were screened from The Cancer Genome Atlas, GSE111260 and GSE16011 samples for protein-protein interaction networks, a risk score model, gene mutation analysis and a nomogram to identify the prognostic hub genes. Subsequently, an immunoassay was performed to determine key genes. Functional and animal assays were then performed to assess the tumorigenesis of the key genes in glioma. Using bioinformatics analysis, centromere protein F (CENPF), kinesin superfamily member 20A, kinesin superfamily protein 4A and marker of proliferation Ki-67 were identified as potential prognostic biomarkers for patients with glioma. Furthermore, CENPF knockdown was demonstrated to suppress the proliferation and metastasis of glioma cells, and induce G2 arrest in the cell cycle. Moreover, CENPF knockdown was revealed to decrease Vimentin and increase E-cadherin levels in glioma cells, and significantly reduce the size and mass of tumors in vivo. Overall, the present study identified new clinical biomarkers and revealed that CENPF may promote glioma progression by regulating the epithelial-mesenchymal transition pathway. By elucidating the complexities of glioma and identifying prognostic biomarkers, the present research enables further improvement of patient outcomes and the advancement of precision medicine for this disease.
Collapse
Affiliation(s)
- Jia Li
- Department of Neurosurgery, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230601, P.R. China
- Department of Emergency Surgery, Fuyang Hospital Affiliated to Anhui Medical University, Fuyang, Anhui 236112, P.R. China
| | - Lei Liu
- Department of Emergency Surgery, Fuyang Hospital Affiliated to Anhui Medical University, Fuyang, Anhui 236112, P.R. China
| | - Gang Zong
- Department of Neurosurgery, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230601, P.R. China
| | - Zhihao Yang
- Department of Neurosurgery, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230601, P.R. China
| | - Deran Zhang
- Department of Neurosurgery, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230601, P.R. China
| | - Bing Zhao
- Department of Neurosurgery, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230601, P.R. China
| |
Collapse
|
4
|
Li J, Chen Q, Ni S, Dong X, Mi T, Xie Y, Yuan X, Luo X, Wang H. CENPF May Act as a Novel Marker and Highlight the Influence of Pericyte in Infantile Hemangioma. Angiology 2024:33197241262373. [PMID: 38898633 DOI: 10.1177/00033197241262373] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/21/2024]
Abstract
Infantile hemangioma (IH), a benign microvascular tumor, is marked by early and extensive proliferation of immature hemangioma endothelial cells (Hem-ECs) that naturally regress through differentiation into fibroblasts or adipocytes. However, a challenge persists, as the unique biological behavior of IH remains elusive, despite its general sensitivity to propranolol treatment. Recent evidence suggests that abnormal volume proliferation in IH is primarily attributed to the accumulation of hemangioma pericytes (Hem-Pericytes), in addition to Hem-ECs. Centromere protein F (CENPF) is involved in regulating mitotic processes and has been associated with malignant tumor cell proliferation. It is a key player in maintaining genomic stability during cell division. Our findings revealed specific expression of CENPF in Hem-Pericytes, with a proliferation index (PI) approximately half that of Ki67 (3.28 vs 6.97%) during the proliferative phase of IH. This index decreased rapidly in the involuting phase (P < .05), suggesting that the contribution of pericytes to IH development was comparable to that of Hem-ECs. Tumor expansion and shrinkage may be due to the proliferation, reduction, and differentiation of Hem-Pericytes. In conclusion, we speculate CENPF as a novel marker for clinical pathological diagnosis and a potential therapeutic target, fostering advancements in drug development.
Collapse
Affiliation(s)
- Jiwei Li
- Department of Dermatology, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China
- Department of Pathology, Kunming Children's Hospital, Kunming, China
| | - Qiang Chen
- Department of Dermatology, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China
- Department of Pediatric Surgery, Chongqing University Three Gorges Hospital, Chongqing, China
| | - Sili Ni
- Department of Dermatology, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Xiaobo Dong
- Department of Dermatology, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Tao Mi
- Department of Dermatology, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Yimin Xie
- Department of Pediatric Surgery, Chongqing University Three Gorges Hospital, Chongqing, China
| | - Xingang Yuan
- Department of Dermatology, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Xiaoyan Luo
- Department of Dermatology, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Hua Wang
- Department of Dermatology, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
5
|
Tang XH, Zhao TN, Guo L, Liu XY, Zhang WN, Zhang P. Cell-Cycle-related Protein Centromere Protein F Deficiency Inhibits Cervical Cancer Cell Growth by Inducing Ferroptosis Via Nrf2 Inactivation. Cell Biochem Biophys 2024; 82:997-1006. [PMID: 38536579 PMCID: PMC11344725 DOI: 10.1007/s12013-024-01251-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2023] [Accepted: 03/12/2024] [Indexed: 08/25/2024]
Abstract
Cervical cancer (CC) is one of the severe cancers that pose a threat to women's health and result in death. CENPF, the centromere protein F, plays a crucial role in mitosis by regulating numerous cellular processes, such as chromosome segregation during mitosis. According to bioinformatics research, CENPF serves as a master regulator that is upregulated and activated in cervical cancer. Nevertheless, the precise biological mechanism that CENPF operates in CC remains unclear. The aim of this study was to analyze the function of CENPF on cervical cancer and its mechanism. We conducted immunohistochemistry and western blot analysis to examine the expression levels of CENPF in both cervical cancer tissues and cells. To explore the hidden biological function of CENPF in cell lines derived from CC, we applied lentivirus transfection to reduce CENPF manifestation. CENPF's main role is to regulate ferroptosis which was assessed by analyzing Reactive Oxygen Species (ROS), malonaldehyde (MDA), etc. The vitro findings were further validated through a subcutaneous tumorigenic nude mouse model. Our research finding indicates that there is an apparent upregulation of CENPF in not merely tumor tissues but also cell lines in the carcinomas of the cervix. In vitro and vivo experimental investigations have demonstrated that the suppression of CENPF can impede cellular multiplication, migration, and invasion while inducing ferroptosis. The ferroptosis induced by CENPF inhibition in cervical cancer cell lines is likely mediated through the Nrf2/HO-1 pathway. The data herein come up with the opinion that CENPF may have a crucial role in influencing anti-cervical cancer effects by inducing ferroptosis via the triggering of the Nrf2/HO-1 signaling pathway.
Collapse
Affiliation(s)
- Xin Hui Tang
- Department of Gynecology, Qingdao Municipal Hospital, Qingdao, 266011, China
- Department of Obstetrics and Gynecology, Putuo People's Hospital, School of Medicine, Tongji University, Shanghai, 200060, China
| | - Tian Nan Zhao
- Department of Gynecology, Qingdao Municipal Hospital, Qingdao, 266011, China
- Dalian Medical University, School of Graduate, Dalian, 116000, China
| | - Li Guo
- Department of Gynecology, Qingdao Municipal Hospital, Qingdao, 266011, China
| | - Xin Yue Liu
- Department of Gynecology, Qingdao Municipal Hospital, Qingdao, 266011, China
- Department of Gynecology, Changzhi People's Hospital, Changzhi, 046000, China
| | - Wei Na Zhang
- Department of Gynecology, Qingdao Municipal Hospital, Qingdao, 266011, China.
| | - Ping Zhang
- Department of Gynecology, Qingdao Municipal Hospital, Qingdao, 266011, China.
| |
Collapse
|
6
|
Liu X, Guo L, Suo Y, Tang X, Zhu T, Zhao T, Zhang W, Zhang P. Cell Cycle-Related Centromere Protein F Deficiency Suppresses Ovarian Cancer Cell Growth by Inducing Ferroptosis. Gynecol Obstet Invest 2024; 89:424-436. [PMID: 38723616 DOI: 10.1159/000539235] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Accepted: 04/27/2024] [Indexed: 06/14/2024]
Abstract
OBJECTIVES This study aimed to investigate the involvement of the cell cycle-related protein centromere protein F (CENPF) in the development of ovarian cancer (OC) and explored its relationship with ferroptosis. DESIGN The databases were analysed to identify differential expression of cell cycle-related proteins between individuals with OC and normal individuals. Immunohistochemistry and statistical analysis were conducted on ovarian tissues obtained from 40 patients with epithelial OC and 20 normal individuals. In vitro experiments were performed using SKOV3 and HEY epithelial OC cell lines. PARTICIPANTS/MATERIALS, SETTING, METHODS The mRNA microarray dataset, consisting of GSE14001, GSE54388, GSE40595, and GSE14407, was downloaded from the Gene Expression Omnibus (GEO) database to investigate the genes associated with cell cycle regulation in OC cells. CENPF was selected as the subject of study through differential analysis.Assessed the expression of CENPF in both OC patients and normal ovarian tissues using immunohistochemistry. Lentivirus infection was employed to downregulate CENPF expression, and subsequent experiments including Cell Counting Kit-8 assay, cell cycle analysis, transwell assay, and wound-healing assay were conducted to investigate the effects of CENPF on proliferation, invasion, migration, and cell cycle regulation in OC cells. The reactive oxygen species (ROS) and the malondialdehyde (MDA) assays were performed to assess the involvement of CENPF in cellular redox reactions. Western blot analysis was conducted to examine the expression levels of ferroptosis-related proteins (GPX4, SLC7A11, DMT1, and protein 53 [p53]). RESULTS By querying and integrating cell cycle-related genes from the GEO database, in silico analyses using The Cancer Genome Atlas database combined with immunohistochemical studies, we discovered that CENPF is upregulated in OC tissues and is related to survival. Downregulation of CENPF inhibited biological function of OC cells, increased intracellular ROS and MDA levels, and downregulated the GPX4 protein and the SLC7A11/xCT protein, but upregulated the DMT1 protein and the tumour p53 expression to induce ferroptosis. LIMITATIONS This study did not investigate ferroptosis-related studies following CENPF overexpression, and the findings have not been validated in animal studies. CONCLUSIONS Our findings demonstrated that the deficiency of CENPF played a crucial anti-oncogenic role in the progression of OC through the mechanism of ferroptosis.
Collapse
Affiliation(s)
- Xinyue Liu
- Department of Gynecology, Qingdao Municipal Hospital, Qingdao, China,
- Department of Gynecology, Shanxi Provincial People' Hospital of Shanxi Medical University, Taiyuan, China,
- Department of Gynecology, Changzhi People's Hospital, Changzhi, China,
| | - Li Guo
- Department of Gynecology, Qingdao Municipal Hospital, Qingdao, China
| | - Yuping Suo
- Department of Gynecology, Shanxi Provincial People' Hospital of Shanxi Medical University, Taiyuan, China
| | - XinHui Tang
- Department of Gynecology, Qingdao Municipal Hospital, Qingdao, China
| | - Ting Zhu
- Department of Gynaecological Oncology, Qingdao Central Hospital, Qingdao, China
| | - Tiannan Zhao
- Department of Gynecology, Qingdao Municipal Hospital, Qingdao, China
| | - Weina Zhang
- Department of Gynecology, Qingdao Municipal Hospital, Qingdao, China
| | - Ping Zhang
- Department of Gynecology, Qingdao Municipal Hospital, Qingdao, China
| |
Collapse
|
7
|
Cai Y, Guo H, Zhou J, Zhu G, Qu H, Liu L, Shi T, Ge S, Qu Y. An alternative extension of telomeres related prognostic model to predict survival in lower grade glioma. J Cancer Res Clin Oncol 2023; 149:13575-13589. [PMID: 37515613 DOI: 10.1007/s00432-023-05155-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Accepted: 07/09/2023] [Indexed: 07/31/2023]
Abstract
OBJECTIVE The alternative extension of the telomeres (ALT) mechanism is activated in lower grade glioma (LGG), but the role of the ALT mechanism has not been well discussed. The primary purpose was to demonstrate the significance of the ALT mechanism in prognosis estimation for LGG patients. METHOD Gene expression and clinical data of LGG patients were collected from the Chinese Glioma Genome Atlas (CGGA) and the Cancer Genome Atlas (TCGA) cohort, respectively. ALT-related genes obtained from the TelNet database and potential prognostic genes related to ALT were selected by LASSO regression to calculate an ALT-related risk score. Multivariate Cox regression analysis was performed to construct a prognosis signature, and a nomogram was used to represent this signature. Possible pathways of the ALT-related risk score are explored by enrichment analysis. RESULT The ALT-related risk score was calculated based on the LASSO regression coefficients of 22 genes and then divided into high-risk and low-risk groups according to the median. The ALT-related risk score is an independent predictor of LGG (HR and 95% CI in CGGA cohort: 5.70 (3.79, 8.58); in TCGA cohort: 1.96 (1.09, 3.54)). ROC analysis indicated that the model contained ALT-related risk score was superior to conventional clinical features (AUC: 0.818 vs 0.729) in CGGA cohorts. The results in the TCGA cohort also shown a powerful ability of ALT-related risk score (AUC: 0.766 vs 0.691). The predicted probability and actual probability of the nomogram are consistent. Enrichment analysis demonstrated that the ALT mechanism was involved in the cell cycle, DNA repair, immune processes, and others. CONCLUSION ALT-related risk score based on the 22-gene is an important factor in predicting the prognosis of LGG patients.
Collapse
Affiliation(s)
- Yaning Cai
- Department of Neurosurgery, Tangdu Hospital, Fourth Military Medical University, No. 569 Xinsi Road, Xi'an 710038, China
| | - Hao Guo
- Department of Neurosurgery, Tangdu Hospital, Fourth Military Medical University, No. 569 Xinsi Road, Xi'an 710038, China
| | - JinPeng Zhou
- Department of Neurosurgery, Tangdu Hospital, Fourth Military Medical University, No. 569 Xinsi Road, Xi'an 710038, China
| | - Gang Zhu
- Department of Neurosurgery, Tangdu Hospital, Fourth Military Medical University, No. 569 Xinsi Road, Xi'an 710038, China
| | - Hongwen Qu
- Department of Neurosurgery, Tangdu Hospital, Fourth Military Medical University, No. 569 Xinsi Road, Xi'an 710038, China
| | - Lingyu Liu
- Department of Neurosurgery, Tangdu Hospital, Fourth Military Medical University, No. 569 Xinsi Road, Xi'an 710038, China
| | - Tao Shi
- Department of Neurosurgery, Tangdu Hospital, Fourth Military Medical University, No. 569 Xinsi Road, Xi'an 710038, China
| | - Shunnan Ge
- Department of Neurosurgery, Tangdu Hospital, Fourth Military Medical University, No. 569 Xinsi Road, Xi'an 710038, China.
| | - Yan Qu
- Department of Neurosurgery, Tangdu Hospital, Fourth Military Medical University, No. 569 Xinsi Road, Xi'an 710038, China.
| |
Collapse
|
8
|
Zheng Y, Li X, Deng S, Zhao H, Ye Y, Zhang S, Huang X, Bai R, Zhuang L, Zhou Q, Li M, Su J, Li R, Bao X, Zeng L, Chen R, Zheng J, Lin D, He C, Zhang J, Zuo Z. CSTF2 mediated mRNA N 6-methyladenosine modification drives pancreatic ductal adenocarcinoma m 6A subtypes. Nat Commun 2023; 14:6334. [PMID: 37816727 PMCID: PMC10564946 DOI: 10.1038/s41467-023-41861-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Accepted: 09/21/2023] [Indexed: 10/12/2023] Open
Abstract
N6-methyladenosine (m6A) modification of gene transcripts plays critical roles in cancer. Here we report transcriptomic m6A profiling in 98 tissue samples from 65 individuals with pancreatic ductal adenocarcinoma (PDAC). We identify 17,996 m6A peaks with 195 hyper-methylated and 93 hypo-methylated in PDAC compared with adjacent normal tissues. The differential m6A modifications distinguish two PDAC subtypes with different prognosis outcomes. The formation of the two subtypes is driven by a newly identified m6A regulator CSTF2 that co-transcriptionally regulates m6A installation through slowing the RNA Pol II elongation rate during gene transcription. We find that most of the CSTF2-regulated m6As have positive effects on the RNA level of host genes, and CSTF2-regulated m6As are mainly recognized by IGF2BP2, an m6A reader that stabilizes mRNAs. These results provide a promising PDAC subtyping strategy and potential therapeutic targets for precision medicine of PDAC.
Collapse
Affiliation(s)
- Yanfen Zheng
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Xingyang Li
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Shuang Deng
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Hongzhe Zhao
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Ying Ye
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Shaoping Zhang
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Xudong Huang
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Ruihong Bai
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Lisha Zhuang
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Quanbo Zhou
- Department of Pancreaticobiliary Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Mei Li
- Department of Pathology, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Jiachun Su
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Rui Li
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Xiaoqiong Bao
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Lingxing Zeng
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Rufu Chen
- Guangdong Provincial People's Hospital & Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Jian Zheng
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
- Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Medicine, Nanjing Medical University, Nanjing, China
- Affiliated Cancer Hospital and Institute of Guangzhou Medical University, Guangzhou, China
| | - Dongxin Lin
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China.
- Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Medicine, Nanjing Medical University, Nanjing, China.
- Department of Etiology and Carcinogenesis, National Cancer Center/National Clinical Research Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| | - Chuan He
- Department of Chemistry, The University of Chicago, Chicago, IL, USA.
- Howard Hughes Medical Institute, The University of Chicago, Chicago, IL, USA.
- Department of Biochemistry and Molecular Biology, and Institute for Biophysical Dynamics, The University of Chicago, Chicago, IL, USA.
| | - Jialiang Zhang
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China.
| | - Zhixiang Zuo
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China.
| |
Collapse
|
9
|
Gui Z, Tian Y, Yu T, Liu S, Liu C, Zhang L. Clinical implications and immune features of CENPN in breast cancer. BMC Cancer 2023; 23:851. [PMID: 37697245 PMCID: PMC10496242 DOI: 10.1186/s12885-023-11376-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Accepted: 09/05/2023] [Indexed: 09/13/2023] Open
Abstract
BACKGROUND A number of human diseases have been associated with Centromere protein N (CENPN), but its role in breast cancer is unclear. METHODS A pan-cancer database of Genotype Tissue Expression (GTEx) and the Cancer Genome Atlas (TCGA) were used to examine the expression of CENPN. Using TCGA clinical survival data and breast cancer specimens from our center for validation, the relationship between CENPN expression, breast cancer prognosis, and clinicopathological characteristics of patients was examined. Bioinformatics was utilized to conduct an enrichment study of CENPN. Additionally, the potential of CENPN as a predictive biomarker for immunotherapy success was confirmed by analyzing the co-expression of CENPN with immune-checkpoint related genes, reviewing the TCGA database, and evaluating the correlation between CENPN expression and immune cell infiltration. Using the CCK8 test and colony formation assay, CENPN was evaluated for its ability to inhibit breast cancer cell proliferation. Transwell assays and scratch tests were used to assess the impact of CENPN on breast cancer cell migration. RESULTS CENPN is found in a wide range of tumors, including breast cancer. Additional investigation revealed that CENPN was co-expressed with the majority of immune checkpoint-related genes, had the potential to serve as a predictive biomarker for immunotherapy effectiveness, and that high CENPN expression was linked to high Tregs and low CD8 + T cells and NK cells. Breast cancer cells' malignant characteristics, such as migration and cell proliferation, were inhibited by CENPN knockdown. CONCLUSIONS According to our findings, CENPN may be an oncogene in breast cancer, as well as a new therapeutic target for immune checkpoint inhibitors.
Collapse
Affiliation(s)
- Zhengwei Gui
- Department of Thyroid and Breast Surgery, Tongji Hospital of Tongji Medical College of Huazhong, University of Science and Technology, 1095 Jiefang Avenue, Wuhan City, 430030, Hubei Province, China
| | - Yao Tian
- Department of Thyroid and Breast Surgery, Tongji Hospital of Tongji Medical College of Huazhong, University of Science and Technology, 1095 Jiefang Avenue, Wuhan City, 430030, Hubei Province, China
| | - Tianyao Yu
- Department of Thyroid and Breast Surgery, Tongji Hospital of Tongji Medical College of Huazhong, University of Science and Technology, 1095 Jiefang Avenue, Wuhan City, 430030, Hubei Province, China
| | - Shiyang Liu
- Department of Thyroid and Breast Surgery, Tongji Hospital of Tongji Medical College of Huazhong, University of Science and Technology, 1095 Jiefang Avenue, Wuhan City, 430030, Hubei Province, China
| | - Chenguang Liu
- Department of Thyroid and Breast Surgery, Tongji Hospital of Tongji Medical College of Huazhong, University of Science and Technology, 1095 Jiefang Avenue, Wuhan City, 430030, Hubei Province, China
| | - Lin Zhang
- Department of Thyroid and Breast Surgery, Tongji Hospital of Tongji Medical College of Huazhong, University of Science and Technology, 1095 Jiefang Avenue, Wuhan City, 430030, Hubei Province, China.
| |
Collapse
|
10
|
Cao Z, Zeng L, Wang Z, Wen X, Zhang J. Integrated pan-cancer analysis of centromere protein F and experimental verification of its role and clinical significance in cholangiocarcinoma. Funct Integr Genomics 2023; 23:190. [PMID: 37247093 DOI: 10.1007/s10142-023-01108-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Revised: 05/12/2023] [Accepted: 05/16/2023] [Indexed: 05/30/2023]
Abstract
Centromere protein F (CENPF), a protein related to the cell cycle, is a key part of the kinetochore-centromere complex involved in cell division, differentiation, and proliferation. CENPF expression is upregulated in various types of cancer and plays a role in oncogenesis and tumor progression. However, the expression pattern, prognostic significance, and biological role of CENPF in these cancer types are poorly understood. Therefore, in this study, we conducted a pan-cancer analysis of the role of CENPF, which we considered a cut point, to investigate its utility as a prognostic and immunological indicator for malignancies, especially cholangiocarcinoma (CCA). Using systematic bioinformatics analysis, we investigated the expression patterns, prognostic relevance, molecular function, signaling pathways, and immune infiltration patterns of CENPF in the pan-cancer analysis. Western blot and immunohistochemistry staining assays were performed to evaluate the expression profiles of CENPF in CCA tissues and cell lines. Furthermore, Cell Counting Kit-8, colony formation, wound healing, and Transwell assays, as well as CCA xenograft mouse models, were employed to determine the role and function of CENPF in CCA. The results showed that CENPF expression was upregulated and strongly linked to a worse prognosis in most cancer types. CENPF expression was substantially associated with immune cell infiltration, tumor microenvironment, genes related to immune checkpoints, tumor mutational burden, microsatellite instability, and immunotherapy response in diverse malignancies. CENPF was considerably overexpressed in CCA tissues and cells. Functionally, inhibiting CENPF expression significantly reduced the proliferating, migrating, and invading abilities of CCA cells. CENPF expression also affects the prognosis of multiple malignancies, which is highly associated with immunotherapy response and tumor immune cell infiltration. In conclusion, CENPF may act as an oncogene and an immune infiltration-related biomarker and can accelerate tumor development in CCA.
Collapse
Affiliation(s)
- Zhenyu Cao
- Department of General Surgery, The Second Xiangya Hospital of Central South University, No. 139 Renminzhong Road, Changsha, 410012, China
| | - Liyun Zeng
- Department of General Surgery, The Second Xiangya Hospital of Central South University, No. 139 Renminzhong Road, Changsha, 410012, China
| | - Zicheng Wang
- Department of General Surgery, The Second Xiangya Hospital of Central South University, No. 139 Renminzhong Road, Changsha, 410012, China
| | - Xueyi Wen
- Department of General Surgery, The Second Xiangya Hospital of Central South University, No. 139 Renminzhong Road, Changsha, 410012, China
| | - Ju Zhang
- Department of General Surgery, The Second Xiangya Hospital of Central South University, No. 139 Renminzhong Road, Changsha, 410012, China.
| |
Collapse
|
11
|
Identification of crucial hub genes and potential molecular mechanisms in breast cancer by integrated bioinformatics analysis and experimental validation. Comput Biol Med 2022; 149:106036. [DOI: 10.1016/j.compbiomed.2022.106036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Revised: 08/14/2022] [Accepted: 08/20/2022] [Indexed: 11/24/2022]
|
12
|
Morin M, Moindjie H, Nahmias C. Le transport mitochondrial. Med Sci (Paris) 2022; 38:585-593. [DOI: 10.1051/medsci/2022085] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
La reprogrammation métabolique est l’un des marqueurs de la carcinogenèse. Au cœur de cette reprogrammation se trouvent les mitochondries qui produisent l’énergie sous forme de molécules d’ATP. La régulation spatio-temporelle de la production d’ATP, indispensable pour fournir l’énergie au bon endroit et au bon moment, est assurée par le transport intracellulaire des mitochondries. Les complexes Miro/TRAK présents à la surface des mitochondries se lient aux protéines motrices de la cellule (dynéine, kinésine, myosine) pour transporter les mitochondries le long du cytosquelette. Ces acteurs du transport mitochondrial sont souvent dérégulés dans le cancer. Nous présentons dans cette revue les mécanismes par lesquels le transport mitochondrial contribue à la migration, à la division cellulaire et à la réponse au stress des cellules cancéreuses. Décrypter ces mécanismes pourrait ouvrir la voie à de nouvelles approches thérapeutiques en oncologie.
Collapse
|
13
|
Chen H, Jiang Y, Liu R, Deng J, Chen Q, Chen L, Liang G, Chen X, Xu Z. Curcumin Derivative C66 Suppresses Pancreatic Cancer Progression through the Inhibition of JNK-Mediated Inflammation. Molecules 2022; 27:3076. [PMID: 35630552 PMCID: PMC9147000 DOI: 10.3390/molecules27103076] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Revised: 05/05/2022] [Accepted: 05/09/2022] [Indexed: 11/16/2022] Open
Abstract
Pancreatic adenocarcinoma is by far the deadliest type of cancer. Inflammation is one of the important risk factors in tumor development. However, it is not yet clear whether deterioration in pancreatic cancer patients is related to inflammation, as well as the underlying mechanism. In addition, JNK is abnormally activated in pancreatic cancer cells and the JNK inhibitor C66 reduces the inflammatory microenvironment in the tumor. Therefore, the aim of this study was to evaluate the role of C66 in the proliferation and migration of pancreatic cancer. Our results showed that various inflammatory cytokines, such as IL-1β, IL-6, IL-8, and IL-15, were more expressed in pancreatic cancer than in the matching normal tissue. Furthermore, C66, a curcumin analogue with good anti-inflammatory activity, inhibited the proliferation and migration of pancreatic cancer cells in a dose-dependent manner, and effectively inhibited the expression of the above inflammatory factors. Our previous research demonstrated that C66 prevents the inflammatory response by targeting JNK. Therefore, in this study, JNK activity in pancreatic cancer cells was investigated, revealing that JNK was highly activated, and the treatment with C66 inhibited the phosphorylation of JNK. Next, shJNK was used to knockdown JNK expression in pancreatic cancer cells to further confirm the role of JNK in the proliferation and migration of this tumor, as well as in the inflammatory tumor microenvironment (TME). The results demonstrated that JNK knockdown could significantly inhibit the proliferation and migration of pancreatic cancer. Moreover, the low JNK expression in pancreatic cancer cells significantly inhibited the expression of various inflammatory factors. These results indicated that C66 inhibited the progression of pancreatic cancer through the inhibition of JNK-mediated inflammation.
Collapse
Affiliation(s)
- Hongjin Chen
- Translational Medicine Research Center, Guizhou Medical University, Guiyang 550000, China
| | - Yuchen Jiang
- The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu 322000, China
- Clinical Pharmacy Center, Department of Pharmacy, Zhejiang Provincial People's Hospital, Affiliated People's Hospital Hangzhou Medical College, Hangzhou 310000, China
| | - Rongdiao Liu
- Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou 325000, China
| | - Jie Deng
- Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou 325000, China
| | - Qinbo Chen
- Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou 325000, China
| | - Lingfeng Chen
- School of Pharmacy, Hangzhou Medical College, Hangzhou 310000, China
| | - Guang Liang
- School of Pharmacy, Hangzhou Medical College, Hangzhou 310000, China
| | - Xiong Chen
- Department of Endocrinology, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou 325000, China
| | - Zheng Xu
- Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou 325000, China
| |
Collapse
|
14
|
Huang Y, Chen S, Xiao L, Qin W, Li L, Wang Y, Ma L, Yuan X. A Novel Prognostic Signature for Survival Prediction and Immune Implication Based on SARS-CoV-2–Related Genes in Kidney Renal Clear Cell Carcinoma. Front Bioeng Biotechnol 2022; 9:744659. [PMID: 35141213 PMCID: PMC8819071 DOI: 10.3389/fbioe.2021.744659] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Accepted: 12/31/2021] [Indexed: 12/28/2022] Open
Abstract
Kidney renal clear cell carcinoma (KIRC) is a common aggressive malignancy of the urinary system. COVID-19, a highly infectious and severe disease caused by SARS-CoV-2, has become a significant challenge for global public health. Cancer patients have been reported to be more vulnerable to SARS-CoV-2 infection and have a higher risk for serious complications than the general population. However, the correlation between KIRC and COVID-19 remains incompletely elucidated. In this study, we comprehensively investigated the expression and prognostic significance of 333 SARS-CoV-2 infection–related genes in KIRC using the TCGA dataset and identified 31 SARS-CoV-2–related differently expressed genes between KIRC and normal renal tissues. Based on these genes, we constructed and validated a 5-gene prognostic signature (including ACADM, CENPF, KDELC1, PLOD2, and TRMT1) to distinguish low- and high-risk KIRC patients of poor survival in TCGA and E-MTAB-1980 cohorts. Gene set enrichment analysis (GSEA) showed that some inflammatory/immune-related pathways were significantly enriched in the high-risk group. The ESTIMATE analysis indicated that patients in the high-risk group had higher stromal and immune cell scores, therefore lower tumor purity. Moreover, they presented higher proportions of macrophages M0, regulatory T cells (Tregs), and T follicular helper cells and higher expression of immune checkpoints CTLA-4, LAG-3, TIGIT, and PDCD1 than low-risk patients. Besides, we also developed a nomogram to expand clinical applicability, which exhibits excellent predictive accuracy for survival. In conclusion, we identified a novel prognostic signature and nomogram based on SARS-CoV-2–related genes as reliable prognostic predictors for KIRC patients and provided potential therapeutic targets for KIRC and COVID-19.
Collapse
Affiliation(s)
- Yongbiao Huang
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Sheng Chen
- Department of Hepatobiliary Surgery, Affiliated Hospital of Hebei University, Baoding, China
| | - Lingyan Xiao
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Wan Qin
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Long Li
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yali Wang
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Li Ma
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xianglin Yuan
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- *Correspondence: Xianglin Yuan,
| |
Collapse
|
15
|
Wang J, Liu X, Chu HJ, Li N, Huang LY, Chen J. Centromere Protein I (CENP-I) Is Upregulated in Gastric Cancer, Predicts Poor Prognosis, and Promotes Tumor Cell Proliferation and Migration. Technol Cancer Res Treat 2021; 20:15330338211045510. [PMID: 34617858 PMCID: PMC8723174 DOI: 10.1177/15330338211045510] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/09/2022] Open
Abstract
This study aimed to investigate the expression and cellular function of the centromeric family of proteins (CENPs), especially centromere protein I (CENP-I), in gastric cancer (GC) and identified its clinical significance and cellular functions. CENP-I expression in GC was studied by cDNA microarray, quantitative real-time PCR (qRT-PCR), and immunohistochemistry (IHC), and using datasets from The Cancer Genome Atlas (TCGA), UALCAN, and Gene Expression Omnibus (GEO) databases. Microarray and bioinformatic analyses identified upregulated CENP-A/E/F/H/I/K/P/W and HJURP in stomach adenocarcinoma (STAD), but not in signet ring cell carcinoma (SRCC). Significantly higher CENP-I mRNA expression was also confirmed in 40 pairs of GC tissues than in paired normal gastric tissues by qRT-PCR (P<.001). IHC showed that elevated CENP-I expression was associated with higher tumor stage, lymph node invasion, increased HER2-positive rate (36.7% vs 10.0%), and intestinal Lauren classification in 69 GC samples compared to paired paracancerous normal tissues. The survival of the high-CENP-I group members was poor compared with that of the low-CENP-I group (P = .0011). Cox univariate regression analysis identified tumor size (P = .008), HER2 status (P = .027), and CENP-I expression (P = .049) were independent prognostic factors of GC. The cellular function of CENP-I was studied in MKN45 and MKN28 GC cell lines in vitro. Cell proliferation, migration, and apoptosis were determined using CCK-8, transwell assay, TUNEL assay, and flow cytometry. Our results showed that CENP-I promoted GC cell proliferation, inhibited apoptosis, facilitated cell migration, and induced epithelial–mesenchymal transition (EMT), possibly by activating the AKT pathway. CENP-I expression was correlated with genetic signatures of the proliferative subtype of GC, characterized by intestinal Lauren classification, HER2 amplification, and TP53 mutation. In conclusion, this study revealed an elevated CENP-I expression in GC, which was associated with malignant features and poor prognosis of GC patients, and identified its function in modulating cell proliferation, apoptosis, and migration.
Collapse
Affiliation(s)
- Jiahui Wang
- Central Laboratory, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, Shandong 264000, China
| | - Xin Liu
- Central Laboratory, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, Shandong 264000, China
| | - Hong-Jin Chu
- Central Laboratory, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, Shandong 264000, China
| | - Ning Li
- Central Laboratory, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, Shandong 264000, China
| | - Liu-Ye Huang
- Department of Gastroenterology, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, Shandong 264000, China
| | - Jian Chen
- Medical Oncology Department, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, Shandong 264000, China
| |
Collapse
|