51
|
Zhao Z, Ju Q, Ji J, Li Y, Zhao Y. N6-Methyladenosine Methylation Regulator RBM15 is a Potential Prognostic Biomarker and Promotes Cell Proliferation in Pancreatic Adenocarcinoma. Front Mol Biosci 2022; 9:842833. [PMID: 35223996 PMCID: PMC8864094 DOI: 10.3389/fmolb.2022.842833] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2021] [Accepted: 01/24/2022] [Indexed: 12/21/2022] Open
Abstract
RNA binding motif protein 15 (RBM15) is a key regulatory factor involved in N6-methyladenosine (m6A) methylation. It has been reported that RBM15 plays an important role in the progress of laryngeal squamous cell carcinoma (LSCC), promoting LSCC migration and invasion. However, the role of RBM15 in human different cancers remains unknown. This study aims to analyze the prognostic value of RBM15, and to demonstrate the correlation between RBM15 expression and tumor immunity, as well as to provide clues for further mechanism research. The results showed that RBM15 was mutated or copy number varied in 25 types of cancer. RBM15 mRNA was abnormally up-regulated across various cancers. Survival analysis suggested high expression of RBM15 was associated with poor prognosis in many cancer types. Among these, it affected patients’ overall survival (OS) in 10 cancer types, disease-free interval (DFI) in 8 cancer types, progression-free interval (PFI) in 12 cancer types and disease-specific survival (DSS) in 7 cancer types. Importantly, in pancreatic adenocarcinoma (PAAD), overexpression of RBM15 is associated with patients’ OS, DFI, PFI, or DSS. In addition, RBM15 expression was positively correlated with immune infiltrating cells in kidney renal clear cell carcinoma (KIRC), brain lower grade glioma (LGG), and PAAD. Moreover, RBM15 expression showed a strong correlation with immune checkpoint markers in PAAD. Cell counting kit-8 (CCK-8) assay showed that knockdown of RBM15 significantly inhibited the proliferation of pancreatic cancer cells. PPI analysis showed USP10, USP24, SMG1, NRAS were closely connected with RBM15 alterations. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis showed that many biological processes (BP), cellular components (CC), molecular functions (MF), cancer related pathways including “sister chromatid cohesion”, “peptidyl-serine phosphorylation”, “cell division”, “nucleoplasm”, “nucleus”, “protein binding”, “protein serine/threonine kinase activity”, “T cell receptor signaling pathway”, “Cell cycle” were regulated by RBM15 alterations. Taken together, pan-cancer analysis of RBM15 suggested it may be served as a prognostic biomarker and immunotherapeutic target for PAAD.
Collapse
Affiliation(s)
- Zhiying Zhao
- School of Public Health, Qingdao University, Qingdao, China
| | - Qiang Ju
- Department of Blood Transfusion, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Jing Ji
- School of Public Health, Qingdao University, Qingdao, China
| | - Yutong Li
- School of Public Health, Qingdao University, Qingdao, China
| | - Yanjie Zhao
- School of Public Health, Qingdao University, Qingdao, China
- *Correspondence: Yanjie Zhao,
| |
Collapse
|
52
|
Huangfu L, Fan B, Wang G, Gan X, Tian S, He Q, Yao Q, Shi J, Li X, Du H, Gao X, Xing X, Ji J. Novel prognostic marker LINC00205 promotes tumorigenesis and metastasis by competitively suppressing miRNA-26a in gastric cancer. Cell Death Dis 2022; 8:5. [PMID: 35013132 PMCID: PMC8748761 DOI: 10.1038/s41420-021-00802-8] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Revised: 11/25/2021] [Accepted: 12/14/2021] [Indexed: 12/03/2022]
Abstract
Rapid proliferation and metastasis of gastric cancer (GC) resulted in a poor prognosis in the clinic. Previous studies elucidated that long non-coding RNA (LncRNA) LINC00205 was upregulated in various tumors and participated in tumor progression. The aim of our study was to investigate the regulating role of LINC00205 in tumorigenesis and metastasis of GC. Both public datasets and our data showed that the LINC00205 was highly expressed in GC tissues and several cell lines. Notably, GC patients with high level of LINC00205 had a poor prognosis in our cohort. Mechanistically, knockdown of LINC00205 by shRNAs suppressed GC cells proliferation, migration, invasion remarkably, and induced cell cycle arrest. Based on bioinformatics prediction, we found that LINC00205 might act as a competitive endogenous RNA (ceRNA) through targeting miR-26a. The level of miR-26a had negatively correlated with LINC00205 expression and was decreased among GC cell lines, tissues, and serum samples. Our results for the first time confirmed that miR-26a was a direct target of LINC00205 and might have the potential to become a plasma marker for clinical tumor diagnosis. Indeed, LINC00205 knockdown resulted in the dramatic promotion of miR-26a expression as well as inhibition of miR-26a potential downstream targets, such as HMGA2, EZH2, and USP15. These targets were essential for cell survival and epithelial-mesenchymal transition. Importantly, LINC00205 was able to remodel the miR-26a-mediated downstream silence, which identified a new mechanism of malignant transformation of GC cells. In conclusion, this study revealed the regulating role of the LINC00205/miR-26a axis in GC progression and provided a new potential therapeutic strategy for GC treatment.
Collapse
Affiliation(s)
- Longtao Huangfu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Division of Gastrointestinal Cancer Translational Research Laboratory, Peking University Cancer Hospital & Institute, Fu-Cheng Road, Beijing, 100142, China
| | - Biao Fan
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Gastrointestinal Cancer Center, Peking University Cancer Hospital & Institute, Fu-Cheng Road, Beijing, 100142, China
| | - Gangjian Wang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Division of Gastrointestinal Cancer Translational Research Laboratory, Peking University Cancer Hospital & Institute, Fu-Cheng Road, Beijing, 100142, China.,Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Gastrointestinal Cancer Center, Peking University Cancer Hospital & Institute, Fu-Cheng Road, Beijing, 100142, China
| | - Xuejun Gan
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Division of Gastrointestinal Cancer Translational Research Laboratory, Peking University Cancer Hospital & Institute, Fu-Cheng Road, Beijing, 100142, China.,Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Gastrointestinal Cancer Center, Peking University Cancer Hospital & Institute, Fu-Cheng Road, Beijing, 100142, China
| | - Shanshan Tian
- National Institute on Drug Dependence, Peking University, North Huayuan Road, Beijing, 100191, China
| | - Qifei He
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Division of Gastrointestinal Cancer Translational Research Laboratory, Peking University Cancer Hospital & Institute, Fu-Cheng Road, Beijing, 100142, China.,Department of Orthopedics, Shenzhen Institute of Translational Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen, 518025, China
| | - Qian Yao
- Department of Pathology, Peking University Cancer Hospital & Institute, Fu-Cheng Road, Beijing, 100142, China
| | - Jinyao Shi
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Division of Gastrointestinal Cancer Translational Research Laboratory, Peking University Cancer Hospital & Institute, Fu-Cheng Road, Beijing, 100142, China.,Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Gastrointestinal Cancer Center, Peking University Cancer Hospital & Institute, Fu-Cheng Road, Beijing, 100142, China
| | - Xiaomei Li
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Division of Gastrointestinal Cancer Translational Research Laboratory, Peking University Cancer Hospital & Institute, Fu-Cheng Road, Beijing, 100142, China
| | - Hong Du
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Division of Gastrointestinal Cancer Translational Research Laboratory, Peking University Cancer Hospital & Institute, Fu-Cheng Road, Beijing, 100142, China
| | - Xiangyu Gao
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Gastrointestinal Cancer Center, Peking University Cancer Hospital & Institute, Fu-Cheng Road, Beijing, 100142, China
| | - Xiaofang Xing
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Division of Gastrointestinal Cancer Translational Research Laboratory, Peking University Cancer Hospital & Institute, Fu-Cheng Road, Beijing, 100142, China.
| | - Jiafu Ji
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Division of Gastrointestinal Cancer Translational Research Laboratory, Peking University Cancer Hospital & Institute, Fu-Cheng Road, Beijing, 100142, China. .,Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Gastrointestinal Cancer Center, Peking University Cancer Hospital & Institute, Fu-Cheng Road, Beijing, 100142, China.
| |
Collapse
|
53
|
Wei P, Jiang J, Xiao M, Zeng M, Liu X, Zhao B, Chen F. The transcript ENST00000444125 of lncRNA LINC01503 promotes cancer stem cell properties of glioblastoma cells via reducing FBXW1 mediated GLI2 degradation. Exp Cell Res 2022; 412:113009. [PMID: 34990616 DOI: 10.1016/j.yexcr.2022.113009] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Revised: 12/22/2021] [Accepted: 01/02/2022] [Indexed: 11/04/2022]
Abstract
LINC010503 is a novel oncogenic lncRNA in multiple cancers. In this study, we further explored the expression of LINC010503 transcripts and their regulations on the glioblastoma (GBM) stem cell (GSC) properties. LINC01503 transcription patterns in GBM and normal brain tissues were compared using RNA-seq data from Genotype-Tissue Expression (GTEx) and The Cancer Genome Atlas (TCGA)-GBM. GBM cell lines (U251 and U87) were used as in vitro cell models for cellular and molecular studies. The results showed that ENST00000444125 was the dominant transcript of LINC01503 in both normal and tumor tissues. Its expression was significantly elevated in the tumor group and associated with poor survival outcomes. LINC01503 had both cytoplasmic and nuclear distribution. It positively modulated the expression of multiple GSC markers, including CD133, SOX2, NESTIN, ALDH1A1, and MSI1, and tumorsphere formation in U251 and U87 cells. RNA pull-down and RIP-qPCR assay confirmed an interaction between ENST00000444125 and GLI2. ENST00000444125 positively regulated the half-life of the GLI2 protein in GBM cells. ENST00000444125 overexpression reduced GLI2 ubiquitination and partially attenuated FBXW1 overexpression induced GLI2 ubiquitination. ENST00000444125 overexpression could activate Wnt/β-catenin signaling in GBM cells. However, these activating effects were remarkedly hampered when GLI2 was knocked down. In conclusion, this study revealed that LINC01503 might have isoform-specific dysregulation in GBM. Among the two major transcripts expressed in GBM cells, ENST00000444125 might be the major functional transcript. Its upregulation might enhance the GSC properties of GBM cells via reducing FBXW1-mediated proteasomal degradation of GLI2.
Collapse
Affiliation(s)
- Pan Wei
- Department of Neurosurgery, The First People(')s Hospital of LongQuanYi District, Chengdu, Sichuan, 610100, China
| | - Jing Jiang
- Department of Gastroenterology, The First People(')s Hospital of LongQuanYi District, Chengdu, Sichuan, 610100, China
| | - Ming Xiao
- Department of Neurosurgery, The First People(')s Hospital of LongQuanYi District, Chengdu, Sichuan, 610100, China
| | - Mengfei Zeng
- Department of Neurosurgery, The First People(')s Hospital of LongQuanYi District, Chengdu, Sichuan, 610100, China
| | - Xingzhi Liu
- Department of Neurosurgery, The First People(')s Hospital of LongQuanYi District, Chengdu, Sichuan, 610100, China
| | - Baihao Zhao
- Department of Neurosurgery, The First People(')s Hospital of LongQuanYi District, Chengdu, Sichuan, 610100, China
| | - Fang Chen
- Department of Neurosurgery, The First People(')s Hospital of LongQuanYi District, Chengdu, Sichuan, 610100, China.
| |
Collapse
|
54
|
Wang C, Chen D, Pan C, Wang C. Research progress of Bub3 gene in malignant tumors. Cell Biol Int 2021; 46:673-682. [PMID: 34882895 PMCID: PMC9303375 DOI: 10.1002/cbin.11740] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Revised: 11/28/2021] [Accepted: 12/04/2021] [Indexed: 12/09/2022]
Abstract
The spindle assembly checkpoint (SAC) is a highly conserved monitoring system that ensures a fidelity of chromosome segregation during mitosis. Bub3, a mitotic Checkpoint Protein, is a member of the Bub protein family, and an important factor in the SAC. Abnormal expression of Bub3 results in mitotic defects, defective spindle gate function, chromosomal instability and the development of aneuploidy cells. Aneuploidy is a state of abnormal karyotype that has long been considered as a marker of tumorigenesis. Karyotypic heterogeneity in tumor cells, known as "chromosomal instability" (CIN), can be used to distinguish cancerous cells from their normal tissue counterpart. In this review, we summarize the expression and clinical significance of Bub3 in a variety of tumors and suggest that it has potential in the treatment of cancer. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Chenyang Wang
- Department of Pathology, First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China.,1997-09, Woman, Han, Breast cancer
| | - Dating Chen
- Department of Pathology, First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Chenglong Pan
- Department of Pathology, First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Chunyan Wang
- Department of Pathology, First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China.,1974-07, Woman, Han, Breast cancer
| |
Collapse
|
55
|
EGR1 modulated LncRNA HNF1A-AS1 drives glioblastoma progression via miR-22-3p/ENO1 axis. Cell Death Dis 2021; 7:350. [PMID: 34772911 PMCID: PMC8590016 DOI: 10.1038/s41420-021-00734-3] [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: 06/04/2021] [Revised: 09/30/2021] [Accepted: 10/20/2021] [Indexed: 11/26/2022]
Abstract
Accumulating evidences revealed that long noncoding RNAs (lncRNAs) have been participated in cancer malignant progression, including glioblastoma multiforme (GBM). Despite much studies have found the precise biological role in the regulatory mechanisms of GBM, however the molecular mechanisms, particularly upstream mechanisms still need further elucidated. RT-QPCR, cell transfection, western blotting and bioinformatic analysis were executed to detect the expression of EGR1, HNF1A-AS1, miR-22-3p and ENO1 in GBM. Cell proliferation assay, colony formation assay, wound healing, migration and invasion assays were performed to detect the malignant characters of GBM cells. The molecular regulation mechanism was confirmed by luciferase reporter assay, ChIP and RIP. Finally, orthotopic mouse models were established to examine the effect of HNF1A-AS1 in vivo. In the current study, we analyzed clinical samples to show that the HNF1A-AS1 expression is upregulated and associated with poor patient survival in GBM. Functional studies revealed that HNF1A-AS1 knockdown markedly inhibits malignant phenotypes of GBM cells, whereas overexpression of HNF1A-AS1 exerts opposite effect. Mechanistically, the transcription factor EGR1 forced the HNF1A-AS1 expression by directly binding the promoter region of HNF1A-AS1. Furthermore, combined bioinformatics analysis with our mechanistic work, using luciferase reporter assays and RIP, we first demonstrated that HNF1A-AS1 functions as a competing endogenous RNA (ceRNA) with miR-22-3p to regulate ENO1 expression in GBM cells. HNF1A-AS1 directly binds to miR-22-3p and significantly inhibits miR-22-3p expression, while ENO1 expression was increased. miR-22-3p inhibitor offsets the HNF1A-AS1 silencing induced suppression in malignant behaviors of GBM cells. ENO1 was verified as a direct target of miR-22-3p and its expression levels was negatively with the prognosis in GBM patients. Taken together, our study illuminated the definite mechanism of HNF1A-AS1 in promoting GBM malignancy, and provided a novel therapeutic target for further clinical application.
Collapse
|
56
|
Saha SK, Islam SMR, Saha T, Nishat A, Biswas PK, Gil M, Nkenyereye L, El-Sappagh S, Islam MS, Cho SG. Prognostic role of EGR1 in breast cancer: a systematic review. BMB Rep 2021. [PMID: 34488929 PMCID: PMC8560464 DOI: 10.5483/bmbrep.2021.54.10.087] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
EGR1 (early growth response 1) is dysregulated in many cancers and exhibits both tumor suppressor and promoter activities, making it an appealing target for cancer therapy. Here, we used a systematic multiomics analysis to review the expression of EGR1 and its role in regulating clinical outcomes in breast cancer (BC). EGR1 expression, its promoter methylation, and protein expression pattern were assessed using various publicly available tools. COSMIC-based somatic mutations and cBioPortal-based copy number alterations were analyzed, and the prognostic roles of EGR1 in BC were determined using Prognoscan and Kaplan-Meier Plotter. We also used bc-GenEx-Miner to investigate the EGR1 co-expression profile. EGR1 was more often downregulated in BC tissues than in normal breast tissue, and its knockdown was positively correlated with poor survival. Low EGR1 expression levels were also associated with increased risk of ER+, PR+, and HER2-BCs. High positive correlations were observed among EGR1, DUSP1, FOS, FOSB, CYR61, and JUN mRNA expression in BC tissue. This systematic review suggested that EGR1 expression may serve as a prognostic marker for BC patients and that clinicopathological parameters influence its prognostic utility. In addition to EGR1, DUSP1, FOS, FOSB, CYR61, and JUN can jointly be considered prognostic indicators for BC.
Collapse
Affiliation(s)
- Subbroto Kumar Saha
- Department of Stem Cell and Regenerative Biotechnology, Konkuk University, Seoul 05029, Korea
| | - S. M. Riazul Islam
- Department of Computer Science and Engineering, Sejong University, Seoul 05006, Korea
| | - Tripti Saha
- Department of Stem Cell and Regenerative Biotechnology, Konkuk University, Seoul 05029, Korea
| | - Afsana Nishat
- Department of Microbiology & Cell Science, University of Florida, Gainesville, FL 32611, USA
| | - Polash Kumar Biswas
- Department of Stem Cell and Regenerative Biotechnology, Konkuk University, Seoul 05029, Korea
| | - Minchan Gil
- Department of Stem Cell and Regenerative Biotechnology, Konkuk University, Seoul 05029, Korea
| | - Lewis Nkenyereye
- Department of Computer and Information Security, Sejong University, Seoul 05006, Korea
| | - Shaker El-Sappagh
- Centro Singular de Investigación en Tecnoloxías Intelixentes (CiTIUS), Universidade de Santiago de Compostela, 15705 Santiago de Compostela, Spain
| | - Md. Saiful Islam
- School of Information and Communication Technology, Griffith University, QLD 4222, Australia
| | - Ssang-Goo Cho
- Department of Stem Cell and Regenerative Biotechnology, Konkuk University, Seoul 05029, Korea
| |
Collapse
|
57
|
Yang D, Ma J, Ma XX. CDKN2B-AS1 Promotes Malignancy as a Novel Prognosis-Related Molecular Marker in the Endometrial Cancer Immune Microenvironment. Front Cell Dev Biol 2021; 9:721676. [PMID: 34712660 PMCID: PMC8546264 DOI: 10.3389/fcell.2021.721676] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Accepted: 09/17/2021] [Indexed: 11/25/2022] Open
Abstract
The prognosis of patients with endometrial cancer (EC) is closely associated with immune cell infiltration. Although abnormal long non-coding RNA (lncRNA) expression is also linked to poor prognosis in patients with EC, the function and action mechanism of immune infiltration-related lncRNAs underlying the occurrence and development of EC remains unclear. In this study, we analyzed lncRNA expression using The Cancer Genome Atlas and clinical data and identified six lncRNAs as prognostic markers for EC, all of which are associated with the infiltration of immune cell subtypes, as illustrated by ImmLnc database and ssGSEA analysis. Real-time quantitative polymerase chain reaction showed that CDKN2B-AS1 was significantly overexpressed in EC, whereas its knockdown inhibited the proliferation and invasion of EC cells and the in vivo growth of transplanted tumors in nude mice. Finally, we constructed a competing endogenous RNA regulatory network and conducted Gene Ontology enrichment analysis to elucidate the potential molecular mechanism underlying CDKN2B-AS1 function. Overall, we identified molecular targets associated with immune infiltration and prognosis and provide new insights into the development of molecular therapies and treatment strategies against EC.
Collapse
Affiliation(s)
- Di Yang
- Department of Gynecology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, Shenyang, China
| | - Jian Ma
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Xiao-Xin Ma
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, China
| |
Collapse
|
58
|
Cai HJ, Zhuang ZC, Wu Y, Zhang YY, Liu X, Zhuang JF, Yang YF, Gao Y, Chen B, Guan GX. Development and validation of a ferroptosis-related lncRNAs prognosis signature in colon cancer. Bosn J Basic Med Sci 2021; 21:569-576. [PMID: 33714257 PMCID: PMC8381210 DOI: 10.17305/bjbms.2020.5617] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Accepted: 03/04/2021] [Indexed: 11/16/2022] Open
Abstract
Ferroptosis is a form of iron-dependent programmed cell death. Regulation of ferroptosis in tumor cells is a novel treatment modality. The present study aimed to investigate ferroptosis-related long non-coding RNAs (lncRNAs) and construct a prognostic model for colon adenocarcinoma (COAD). RNA- sequencing data and ferroptosis-related genes were obtained from The Cancer Genome Atlas database and FerrDb database. COAD patients were randomly assigned to training- and validation groups. The Least Absolute Shrinkage and Selection Operator regression and Cox regression model were used to determine and develop a predictive model. The model was corroborated using the validation group and the entire group. In total, 259 ferroptosis-related genes and 905 ferroptosis-related LncRNAs were obtained. Cox model revealed and constructed seven ferroptosis-related LncRNAs signature (LINC01503, AC004687.1, AC010973.2, AP001189.3, ARRDC1-AS1, OIP5-AS1, and NCK1-DT). Patients were assigned into two groups according to the median risk score. Kaplan-Meier survival curves showed that overall survival between high- and low-risk groups was statistically significant (P<0.01). Cox multivariate analysis seven ferroptosis-related LncRNAs signature was an independent risk factor for COAD outcomes (P<0.05). The relationship between seven ferroptosis-related LncRNAs and clinicopathological features was also examined. The principal component analysis showed a difference between high- and low-risk groups intuitively. With the aid of gene set enrichment analysis, the underlying mechanisms of seven ferroptosis-related LncRNAs were uncovered, including the MAPK signaling pathway, mTOR signaling pathway, and glutathione metabolism pathway. Finally, we established and validated seven ferroptosis-related lncRNAs signature for COAD patients to predict survival. These results may provide meaningful targets for future study.
Collapse
Affiliation(s)
- Hua-jun Cai
- Department of Colorectal Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Zhi-cheng Zhuang
- Department of Colorectal Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Yong Wu
- Department of Colorectal Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Yi-yi Zhang
- Department of Colorectal Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Xing Liu
- Department of Colorectal Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Jin-fu Zhuang
- Department of Colorectal Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Yuan-feng Yang
- Department of Colorectal Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Yuan Gao
- Department of Colorectal Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Bin Chen
- Department of Colorectal Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Guo-xian Guan
- Department of Colorectal Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| |
Collapse
|
59
|
Zhang W, Zheng M, Kong S, Li X, Meng S, Wang X, Wang F, Tang C, Ju S. Circular RNA hsa_circ_0007507 May Serve as a Biomarker for the Diagnosis and Prognosis of Gastric Cancer. Front Oncol 2021; 11:699625. [PMID: 34595108 PMCID: PMC8477006 DOI: 10.3389/fonc.2021.699625] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Accepted: 08/17/2021] [Indexed: 12/29/2022] Open
Abstract
PURPOSE The morbidity and mortality of gastric cancer (GC) remain high worldwide. In recent years, circular RNAs (circRNAs) have attracted widespread attention among cancer researchers due to the stable ring structure. The present work aims to find serum circRNA biomarkers that can be used in clinical applications and effective diagnosis. METHODS Hsa_circ_0007507 was extracted through circRNA sequencing. Exonuclease digestion assay, actinomycin D, agarose gel electrophoresis (AGE), and Sanger sequencing verified the potential of hsa_circ_0007507 as a biomarker. Besides, a real-time fluorescent quantitative polymerase chain reaction (RT-qPCR) was established to detect the level of expression of hsa_circ_0007507. Twenty cases of GC and the paired adjacent tissues were collected to verify its overexpression. Then, serum samples from 30 cases of colorectal cancer, 30 cases of thyroid cancer, and 30 cases of breast cancer were collected to verify their organ specificity. Additionally, serum samples from 80 healthy people, 62 gastritis patients, 31 intestinal metaplasia patients, and 100 GC patients were collected, and the diagnostic efficacy was evaluated through analysis of the receiver operating characteristic (ROC) curve. Furthermore, 16 post-operative GC samples, samples of 65 relapsed patients and 36 non-relapsed patients were collected to evaluate the prognosis of GC. RESULTS The level of expression of hsa_circ_0007507 in GC tissues was up-regulated (p = 0.0121), which was consistent with the results of circRNA sequencing. Exonuclease digestion assay and actinomycin D confirmed that hsa_circ_0007507 had a stable structure and a longer half-life. In the analysis of organ specificity experiments, serum hsa_circ_0007507 did not have specificity for patients with colorectal cancer (p = 0.5319), thyroid cancer (p = 0.5422), or breast cancer (p = 0.5178). Analysis of diagnostic efficacy indicated that the expression of hsa_circ_0007507 was significantly higher than that of normal people (p <0.0001); the area under the ROC (AUC) was 0.832 (95% CI: 0.771-0.892); the diagnostic power of hsa_circ_0007507 was higher than that of CEA (AUC = 0.765, 95% CI: 0.697-0.833) and CA199 (AUC = 0.587, 95% CI: 0.504-0.67). Through diagnosis using a combination of the three, GC patients could be distinguished from normal people (AUC = 0.849), and higher diagnostic efficiency could be achieved. The expression of serum hsa_circ_0007507 in GC patients significantly decreased after surgery (p = 0.001). Besides, the expression of serum hsa_circ_0007507 in patients with post-operative recurrence was significantly up-regulated again (p = 0.0139). CONCLUSIONS Serum hsa_circ_0007507 is differentially expressed in GC patients, post-operative GC patients, gastritis patients, intestinal metaplasia patients and relapsed patients, suggesting that serum hsa_circ_0007507 can be used as a new diagnostic and dynamic monitoring biomarker for GC.
Collapse
Affiliation(s)
- Weiwei Zhang
- Department of Laboratory Medicine, Affiliated Hospital of Nantong University, Nantong, China
- Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Nantong, China
| | - Ming Zheng
- Department of Laboratory Medicine, Affiliated Hospital of Nantong University, Nantong, China
- Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Nantong, China
- Nantong University School of Medicine, Nantong, China
| | - Shan Kong
- Department of Laboratory Medicine, Affiliated Hospital of Nantong University, Nantong, China
- Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Nantong, China
- Nantong University School of Medicine, Nantong, China
| | - Xian Li
- Department of Laboratory Medicine, Affiliated Hospital of Nantong University, Nantong, China
| | - Shuting Meng
- Department of Laboratory Medicine, Hai’an People’s Hospital, Hai’an, China
| | - Xudong Wang
- Department of Laboratory Medicine, Affiliated Hospital of Nantong University, Nantong, China
| | - Feng Wang
- Department of Laboratory Medicine, Affiliated Hospital of Nantong University, Nantong, China
| | - Chenxue Tang
- Department of Laboratory Medicine, Affiliated Hospital of Nantong University, Nantong, China
| | - Shaoqing Ju
- Department of Laboratory Medicine, Affiliated Hospital of Nantong University, Nantong, China
| |
Collapse
|
60
|
Liu X, Liu X, Du Y, Hu M, Tian Y, Li Z, Lv L, Zhang X, Liu Y, Zhou Y, Zhang P. DUSP5 promotes osteogenic differentiation through SCP1/2-dependent phosphorylation of SMAD1. STEM CELLS (DAYTON, OHIO) 2021; 39:1395-1409. [PMID: 34169608 PMCID: PMC8518947 DOI: 10.1002/stem.3428] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/27/2021] [Revised: 04/27/2021] [Accepted: 05/06/2021] [Indexed: 11/23/2022]
Abstract
Dual‐specificity phosphatases (DUSPs) are defined by their capability to dephosphorylate both phosphoserine/phosphothreonine (pSer/pThr) and phosphotyrosine (pTyr). DUSP5, a member of DUSPs superfamily, is located in the nucleus and plays crucially regulatory roles in the signaling pathway transduction. In our present study, we discover that DUSP5 significantly promotes osteogenic differentiation of mesenchymal stromal cells (MSCs) by activating SMAD1 signaling pathway. Mechanistically, DUSP5 physically interacts with the phosphatase domain of small C‐terminal phosphatase 1/2 (SCP1/2, SMAD1 phosphatases) by the linker region. In addition, we further confirm that DUSP5 activates SMAD1 signaling through a SCP1/2‐dependent manner. Specifically, DUSP5 attenuates the SCP1/2‐SMAD1 interaction by competitively binding to SCP1/2, which is responsible for the SMAD1 dephosphorylation, and thus results in the activation of SMAD1 signaling. Importantly, DUSP5 expression in mouse bone marrow MSCs is significantly reduced in ovariectomized (OVX) mice in which osteogenesis is highly passive, and overexpression of Dusp5 via tail vein injection reverses the bone loss of OVX mice efficiently. Collectively, this work demonstrates that the linker region of DUSP5 maybe a novel chemically modifiable target for controlling MSCs fate choices and for osteoporosis treatment.
Collapse
Affiliation(s)
- Xuejiao Liu
- Department of Prosthodontics, School and Hospital of Stomatology, Peking University, Beijing, People's Republic of China.,National Engineering Lab for Digital and Material Technology of Stomatology, National Clinical Diseases, Peking University School and Hospital of Stomatology, Peking University, Beijing, People's Republic of China
| | - Xuenan Liu
- Department of Prosthodontics, School and Hospital of Stomatology, Peking University, Beijing, People's Republic of China.,National Engineering Lab for Digital and Material Technology of Stomatology, National Clinical Diseases, Peking University School and Hospital of Stomatology, Peking University, Beijing, People's Republic of China
| | - Yangge Du
- Department of Prosthodontics, School and Hospital of Stomatology, Peking University, Beijing, People's Republic of China.,National Engineering Lab for Digital and Material Technology of Stomatology, National Clinical Diseases, Peking University School and Hospital of Stomatology, Peking University, Beijing, People's Republic of China
| | - Menglong Hu
- Department of Prosthodontics, School and Hospital of Stomatology, Peking University, Beijing, People's Republic of China.,National Engineering Lab for Digital and Material Technology of Stomatology, National Clinical Diseases, Peking University School and Hospital of Stomatology, Peking University, Beijing, People's Republic of China
| | - Yueming Tian
- Department of Prosthodontics, School and Hospital of Stomatology, Peking University, Beijing, People's Republic of China.,National Engineering Lab for Digital and Material Technology of Stomatology, National Clinical Diseases, Peking University School and Hospital of Stomatology, Peking University, Beijing, People's Republic of China
| | - Zheng Li
- Department of Prosthodontics, School and Hospital of Stomatology, Peking University, Beijing, People's Republic of China.,National Engineering Lab for Digital and Material Technology of Stomatology, National Clinical Diseases, Peking University School and Hospital of Stomatology, Peking University, Beijing, People's Republic of China
| | - Longwei Lv
- Department of Prosthodontics, School and Hospital of Stomatology, Peking University, Beijing, People's Republic of China.,National Engineering Lab for Digital and Material Technology of Stomatology, National Clinical Diseases, Peking University School and Hospital of Stomatology, Peking University, Beijing, People's Republic of China
| | - Xiao Zhang
- Department of Prosthodontics, School and Hospital of Stomatology, Peking University, Beijing, People's Republic of China.,National Engineering Lab for Digital and Material Technology of Stomatology, National Clinical Diseases, Peking University School and Hospital of Stomatology, Peking University, Beijing, People's Republic of China
| | - Yunsong Liu
- Department of Prosthodontics, School and Hospital of Stomatology, Peking University, Beijing, People's Republic of China.,National Engineering Lab for Digital and Material Technology of Stomatology, National Clinical Diseases, Peking University School and Hospital of Stomatology, Peking University, Beijing, People's Republic of China
| | - Yongsheng Zhou
- Department of Prosthodontics, School and Hospital of Stomatology, Peking University, Beijing, People's Republic of China.,National Engineering Lab for Digital and Material Technology of Stomatology, National Clinical Diseases, Peking University School and Hospital of Stomatology, Peking University, Beijing, People's Republic of China
| | - Ping Zhang
- Department of Prosthodontics, School and Hospital of Stomatology, Peking University, Beijing, People's Republic of China.,National Engineering Lab for Digital and Material Technology of Stomatology, National Clinical Diseases, Peking University School and Hospital of Stomatology, Peking University, Beijing, People's Republic of China
| |
Collapse
|
61
|
Chen L, Wu C, Wang H, Chen S, Ma D, Tao Y, Wang X, Luan Y, Wang T, Shi Y, Song G, Zhao Y, Dong X, Wang B. Analysis of Long Noncoding RNAs in Aila-Induced Non-Small Cell Lung Cancer Inhibition. Front Oncol 2021; 11:652567. [PMID: 34235076 PMCID: PMC8255921 DOI: 10.3389/fonc.2021.652567] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Accepted: 05/21/2021] [Indexed: 01/24/2023] Open
Abstract
Non-small cell lung cancer (NSCLC) has the highest morbidity and mortality among all carcinomas. However, it is difficult to diagnose in the early stage, and current therapeutic efficacy is not ideal. Although numerous studies have revealed that Ailanthone (Aila), a natural product, can inhibit multiple cancers by reducing cell proliferation and invasion and inducing apoptosis, the mechanism by which Aila represses NSCLC progression in a time-dependent manner remains unclear. In this study, we observed that most long noncoding RNAs (lncRNAs) were either notably up- or downregulated in NSCLC cells after treatment with Aila. Moreover, alterations in lncRNA expression induced by Aila were crucial for the initiation and metastasis of NSCLC. Furthermore, in our research, expression of DUXAP8 was significantly downregulated in NSCLC cells after treatment with Aila and regulated expression levels of EGR1. In conclusion, our findings demonstrate that Aila is a potent natural suppressor of NSCLC by modulating expression of DUXAP8 and EGR1.
Collapse
Affiliation(s)
- Lin Chen
- College of Clinical Medicine, College of Integrated Traditional Chinese and Western Medicine, Changchun University of Chinese Medicine, Changchun, China.,College of Animal Science, Jilin University, Changchun, China
| | - Cui Wu
- College of Clinical Medicine, College of Integrated Traditional Chinese and Western Medicine, Changchun University of Chinese Medicine, Changchun, China
| | - Heming Wang
- Department of Gastroenterology and Hepatology, Zhongshan Hospital, Fudan University, Shanghai, China.,Department of Gastroenterology and Hepatology, Zhongshan Hospital of Fudan University, Shanghai, China
| | - Sinuo Chen
- Department of Gastroenterology and Hepatology, Zhongshan Hospital, Fudan University, Shanghai, China.,Department of Gastroenterology and Hepatology, Zhongshan Hospital of Fudan University, Shanghai, China
| | - Danhui Ma
- Department of Gastroenterology and Hepatology, Zhongshan Hospital, Fudan University, Shanghai, China.,Department of Gastroenterology and Hepatology, Zhongshan Hospital of Fudan University, Shanghai, China
| | - Ye Tao
- Affiliated Hospital to Changchun University of Chinese Medicine, Changchun University of Chinese Medicine, Changchun, China
| | - Xingye Wang
- College of Clinical Medicine, College of Integrated Traditional Chinese and Western Medicine, Changchun University of Chinese Medicine, Changchun, China
| | - Yanhe Luan
- Affiliated Hospital to Changchun University of Chinese Medicine, Changchun University of Chinese Medicine, Changchun, China
| | - Tiedong Wang
- College of Animal Science, Jilin University, Changchun, China
| | - Yan Shi
- School of Pharmacy, Jilin University, Changchun, China
| | - Guangqi Song
- Department of Gastroenterology and Hepatology, Zhongshan Hospital, Fudan University, Shanghai, China.,Department of Gastroenterology and Hepatology, Zhongshan Hospital of Fudan University, Shanghai, China
| | - Yicheng Zhao
- College of Clinical Medicine, College of Integrated Traditional Chinese and Western Medicine, Changchun University of Chinese Medicine, Changchun, China
| | - Xijun Dong
- College of Clinical Medicine, College of Integrated Traditional Chinese and Western Medicine, Changchun University of Chinese Medicine, Changchun, China.,Affiliated Hospital to Changchun University of Chinese Medicine, Changchun University of Chinese Medicine, Changchun, China
| | - Bingmei Wang
- College of Clinical Medicine, College of Integrated Traditional Chinese and Western Medicine, Changchun University of Chinese Medicine, Changchun, China
| |
Collapse
|
62
|
Liu C, Hu C, Li J, Jiang L, Zhao C. Identification of Epithelial-Mesenchymal Transition-Related lncRNAs that Associated With the Prognosis and Immune Microenvironment in Colorectal Cancer. Front Mol Biosci 2021; 8:633951. [PMID: 33898515 PMCID: PMC8059639 DOI: 10.3389/fmolb.2021.633951] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Accepted: 02/19/2021] [Indexed: 12/23/2022] Open
Abstract
Background: The expression of long non-coding RNA (lncRNA) is associated with the epithelial-mesenchymal transition (EMT) in tumorigenicity, but the role of EMT-related lncRNA in colorectal cancer (CRC) remains unclear. Methods: The clinical data and gene expression profile of CRC patients were obtained from The Cancer Genome Atlas database. Differential expression analysis, Cox regression model, and Kaplan-Meier analysis were used to study the relationship between EMT-related lncRNAs and the prognosis of CRC. Functional analysis and unsupervised clustering analysis were performed to explore the influence of certain lncRNAs on CRC. Finally, Cytoscape was used to construct mRNA-lncRNA networks. Results: Two signatures incorporating six and ten EMT-related lncRNAs were constructed for predicting the overall survival (OS) and disease-free survival (DFS), respectively. Kaplan-Meier survival curves indicated that patients in the high-risk group had a poorer prognosis than those in the low-risk group. The results of the functional analysis suggested that the P53 and ECM-receptor pathways affect the prognosis of CRC, and AL591178.1 is a key prognostic EMT-related lncRNA, which is negatively related to immune cells, P53 pathway, and ECM-receptor pathway. Conclusion: Six OS-related and ten DFS-related EMT-related lncRNAs were correlated with the prognosis of CRC by potentially affecting the immune microenvironment, and AL591178.1 plays a key role as a prognostic factor.
Collapse
Affiliation(s)
- Chuan Liu
- Graduate College, China Medical University, Shenyang, China
| | - Chuan Hu
- Department of Spinal Surgery, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Jianyi Li
- Department of Spinal Surgery, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Liqing Jiang
- Graduate College, China Medical University, Shenyang, China
| | - Chengliang Zhao
- Department of Spinal Surgery, The Affiliated Hospital of Qingdao University, Qingdao, China
| |
Collapse
|
63
|
Wang B, Guo H, Yu H, Chen Y, Xu H, Zhao G. The Role of the Transcription Factor EGR1 in Cancer. Front Oncol 2021; 11:642547. [PMID: 33842351 PMCID: PMC8024650 DOI: 10.3389/fonc.2021.642547] [Citation(s) in RCA: 124] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Accepted: 02/25/2021] [Indexed: 12/12/2022] Open
Abstract
Early growth response factor 1 (EGR1) is a transcription factor that is mainly involved in the processes of tissue injury, immune responses, and fibrosis. Recent studies have shown that EGR1 is closely related to the initiation and progression of cancer and may participate in tumor cell proliferation, invasion, and metastasis and in tumor angiogenesis. Nonetheless, the specific mechanism whereby EGR1 modulates these processes remains to be elucidated. This review article summarizes possible mechanisms of action of EGR1 in tumorigenesis and tumor progression and may serve as a reference for clinical efficacy predictions and for the discovery of new therapeutic targets.
Collapse
Affiliation(s)
- Bin Wang
- Department of Neurosurgery, The First Hospital of Jilin University, Changchun, China
| | - Hanfei Guo
- Cancer Center, The First Hospital of Jilin University, Changchun, China
| | - Hongquan Yu
- Department of Neurosurgery, The First Hospital of Jilin University, Changchun, China
| | - Yong Chen
- Department of Neurosurgery, The First Hospital of Jilin University, Changchun, China
| | - Haiyang Xu
- Department of Neurosurgery, The First Hospital of Jilin University, Changchun, China
| | - Gang Zhao
- Department of Neurosurgery, The First Hospital of Jilin University, Changchun, China
| |
Collapse
|
64
|
Ma Z, Gao X, Shuai Y, Wu X, Yan Y, Xing X, Ji J. EGR1-mediated linc01503 promotes cell cycle progression and tumorigenesis in gastric cancer. Cell Prolif 2020; 54:e12922. [PMID: 33145887 PMCID: PMC7791171 DOI: 10.1111/cpr.12922] [Citation(s) in RCA: 60] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Revised: 08/27/2020] [Accepted: 09/18/2020] [Indexed: 12/11/2022] Open
Abstract
Objectives Long non‐coding RNAs (lncRNAs) are key mediators in various malignancies. Linc01503 was previously elucidated to promote gastric cancer (GC) cell invasion. However, the upstream mechanism of linc01503 and its involvement in GC cell cycle, apoptosis and tumorigenesis still remain unclear. Materials and Methods Bioinformatics analysis and quantitative reverse transcription polymerase chain reaction (qRT‐PCR) assays were implicated to detect linc01503 level in GC. The role of linc01503 was detected by in vitro functional assays and in vivo xenograft tumour models. The association between linc01503 and its upstream effector was identified by chromatin immunoprecipitation (ChIP) assays. The mechanistic model of linc01503 was clarified using subcellular separation, fluorescence in situ hybridization, RNA‐sequencing, RNA immunoprecipitation (RIP) and ChIP assays. Results Linc01503 was remarkably elevated in GC and tightly linked with the overall survival of patients with GC. The key transcription factor early growth response protein 1 (EGR1) critically activated the transcription of linc01503. Functionally, linc01503 knockdown resulted in the activation of apoptosis and G1/G0 phase arrest in GC. Mechanistically, linc01503 interacted with histone modification enzyme enhancer of zeste 2 (EZH2) and lysine (K)‐specific demethylase 1A (LSD1), thereby mediating the transcriptional silencing of dual‐specificity phosphatase 5 (DUSP5) and cyclin‐dependent kinase inhibitor 1A (CDKN1A) in GC. Conclusions EGR1‐activated linc01503 could epigenetically silence DUSP5/CDKN1A expression to mediate cell cycle progression and tumorigenesis, implicating it as a prospective target for GC therapeutics.
Collapse
Affiliation(s)
- Zhonghua Ma
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Division of Gastrointestinal Cancer Translational Research Laboratory, Peking University Cancer Hospital and Institute, Beijing, China
| | - Xiangyu Gao
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Division of Gastrointestinal Cancer Translational Research Laboratory, Peking University Cancer Hospital and Institute, Beijing, China
| | - You Shuai
- Department of Medical Oncology, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, China
| | - Xiaolong Wu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Division of Gastrointestinal Cancer Translational Research Laboratory, Peking University Cancer Hospital and Institute, Beijing, China
| | - Yan Yan
- Department of Endoscopy Center, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital and Institute, Beijing, China
| | - Xiaofang Xing
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Division of Gastrointestinal Cancer Translational Research Laboratory, Peking University Cancer Hospital and Institute, Beijing, China
| | - Jiafu Ji
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Division of Gastrointestinal Cancer Translational Research Laboratory, Peking University Cancer Hospital and Institute, Beijing, China
| |
Collapse
|