1
|
Nie H, Fang S, Zhou R, Jia Y, Zhou J, Ning Y, Yu Y, Hong Y, Xu F, Zhao Q, Nie J, Wang F. Upregulation of RIG-I is Critical for Responsiveness to IFN-α Plus Anti-PD-1 in Colorectal Cancer. Cancer Med 2025; 14:e70802. [PMID: 40116486 PMCID: PMC11926914 DOI: 10.1002/cam4.70802] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2025] [Revised: 03/08/2025] [Accepted: 03/11/2025] [Indexed: 03/23/2025] Open
Abstract
BACKGROUNDS Immunotherapy is a promising and effective approach that has achieved significant curative effects in colorectal cancer (CRC). Recently, retinoic acid-inducible gene I (RIG-I) has been shown to play a critical role in tumor immunity. However, the correlation between RIG-I and immunotherapy in CRC remains unclear. METHODS RIG-I expression was measured in CRC and normal samples based on analysis of the public databases, a tissue microarray, and CRC cell lines. The correlation between RIG-I and immune microenvironment was explored using well-established biological algorithms and in vitro and in vivo experiments. RESULTS We discovered that RIG-I expression was downregulated in CRC compared with normal samples. The bioinformatic algorithms indicated that high RIG-I-expressing samples showed a positive correlation with IFN-α response and enrichment of antitumor immune cells, especially CD8+ T cells. Furthermore, knockdown of RIG-I expression efficiently reduced the cell death, STAT1 phosphorylation, and CXCL10/11 expression induced by IFN-α in CRC cells. Finally, an in vivo study showed that the infiltration of CD3+ CD8+ T cells was significantly decreased in the RIG-I knockout group. An animal model further confirmed that the inhibition of tumor growth induced by IFN-α plus anti-PD-1 therapy was dependent on RIG-I expression. CONCLUSION RIG-I is a promising biomarker for CRC immunotherapy, which provides a novel concept for combinatorial immunotherapy.
Collapse
Affiliation(s)
- Haihang Nie
- Department of GastroenterologyZhongnan Hospital of Wuhan UniversityWuhanChina
- Hubei Provincial Clinical Research Center for Intestinal and Colorectal DiseasesWuhanChina
- Hubei Key Laboratory of Intestinal and Colorectal DiseasesZhongnan Hospital of Wuhan UniversityWuhanChina
| | - Shilin Fang
- Department of Infectious DiseasesZhongnan Hospital of Wuhan UniversityWuhanChina
| | - Rui Zhou
- Department of GastroenterologyZhongnan Hospital of Wuhan UniversityWuhanChina
- Hubei Provincial Clinical Research Center for Intestinal and Colorectal DiseasesWuhanChina
- Hubei Key Laboratory of Intestinal and Colorectal DiseasesZhongnan Hospital of Wuhan UniversityWuhanChina
| | - Yifan Jia
- Department of PainRenmin Hospital of Wuhan UniversityWuhanChina
| | - Jingkai Zhou
- Department of GastroenterologyZhongnan Hospital of Wuhan UniversityWuhanChina
- Hubei Provincial Clinical Research Center for Intestinal and Colorectal DiseasesWuhanChina
- Hubei Key Laboratory of Intestinal and Colorectal DiseasesZhongnan Hospital of Wuhan UniversityWuhanChina
| | - Yumei Ning
- Department of GastroenterologyZhongnan Hospital of Wuhan UniversityWuhanChina
- Hubei Provincial Clinical Research Center for Intestinal and Colorectal DiseasesWuhanChina
- Hubei Key Laboratory of Intestinal and Colorectal DiseasesZhongnan Hospital of Wuhan UniversityWuhanChina
| | - Yali Yu
- Department of GastroenterologyZhongnan Hospital of Wuhan UniversityWuhanChina
- Hubei Provincial Clinical Research Center for Intestinal and Colorectal DiseasesWuhanChina
- Hubei Key Laboratory of Intestinal and Colorectal DiseasesZhongnan Hospital of Wuhan UniversityWuhanChina
| | - Yuntian Hong
- Department of GastroenterologyZhongnan Hospital of Wuhan UniversityWuhanChina
- Hubei Provincial Clinical Research Center for Intestinal and Colorectal DiseasesWuhanChina
- Hubei Key Laboratory of Intestinal and Colorectal DiseasesZhongnan Hospital of Wuhan UniversityWuhanChina
| | - Fei Xu
- Department of GastroenterologyZhongnan Hospital of Wuhan UniversityWuhanChina
- Hubei Provincial Clinical Research Center for Intestinal and Colorectal DiseasesWuhanChina
- Hubei Key Laboratory of Intestinal and Colorectal DiseasesZhongnan Hospital of Wuhan UniversityWuhanChina
| | - Qiu Zhao
- Department of GastroenterologyZhongnan Hospital of Wuhan UniversityWuhanChina
- Hubei Provincial Clinical Research Center for Intestinal and Colorectal DiseasesWuhanChina
- Hubei Key Laboratory of Intestinal and Colorectal DiseasesZhongnan Hospital of Wuhan UniversityWuhanChina
| | - Jiayan Nie
- Department of GastroenterologyZhongnan Hospital of Wuhan UniversityWuhanChina
- Hubei Provincial Clinical Research Center for Intestinal and Colorectal DiseasesWuhanChina
- Hubei Key Laboratory of Intestinal and Colorectal DiseasesZhongnan Hospital of Wuhan UniversityWuhanChina
| | - Fan Wang
- Department of GastroenterologyZhongnan Hospital of Wuhan UniversityWuhanChina
- Hubei Provincial Clinical Research Center for Intestinal and Colorectal DiseasesWuhanChina
- Hubei Key Laboratory of Intestinal and Colorectal DiseasesZhongnan Hospital of Wuhan UniversityWuhanChina
| |
Collapse
|
2
|
Sun Y, Liu Y, Jiang L, Zhong C. m5C methylation modification may be an accomplice in colorectal cancer escaping from anti-tumor effects of innate immunity-type I/III interferon. Front Immunol 2025; 15:1512353. [PMID: 39867908 PMCID: PMC11757137 DOI: 10.3389/fimmu.2024.1512353] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Accepted: 12/19/2024] [Indexed: 01/28/2025] Open
Abstract
Colorectal cancer (CRC) is one of the most prevalent malignant tumors in the world, and its occurrence and development are closely related to the complex immune regulatory mechanisms. As the first barrier of the body's defense, innate immunity plays a key role in tumor immune surveillance and anti-tumor response, in which type I/III interferon (IFN) is an important mediator with significant antiviral and anti-tumor functions. 5-methylcytosine (m5C) modification of RNA is a key epigenetic regulation that promotes the expression of CRC oncogenes and immune-related genes. It can enhance the proliferation, migration, and invasion of tumor cells by affecting mRNA stability, translation efficiency, and nuclear export. In addition, m5C modification modulates the activity of innate immune signaling pathways and inhibits interferon production and function, further helping tumor cells evade immune surveillance. However, there are insufficient elucidations on the interaction between m5C modification and innate immunity in CRC. In this study, the mechanism of interferon I/III in colorectal cancer was systematically reviewed and explored. This work focused on how m5C modification promotes tumor immune escape by affecting the interferon signaling pathway, thereby providing new diagnostic markers and therapeutic targets for clinical use, and enhancing the immunotherapy efficacy.
Collapse
Affiliation(s)
- Yiqi Sun
- Surgery of Traditional Chinese Medicine Department, Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Yunfei Liu
- Department of Anesthesiology, Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Lu Jiang
- Department of Anesthesiology, Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Chao Zhong
- Traditional Chinese Medicine Department of Orthopaedic and Traumatic, Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, China
| |
Collapse
|
3
|
Li J, Han T, Wang X, Wang Y, Chen X, Chen W, Yang Q. Identification of prognostic immune-related lncRNA signature predicting the overall survival for colorectal cancer. Sci Rep 2023; 13:1333. [PMID: 36693898 PMCID: PMC9873726 DOI: 10.1038/s41598-023-28305-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2022] [Accepted: 01/17/2023] [Indexed: 01/25/2023] Open
Abstract
Long non-coding RNA (lncRNA) is an important regulator of gene expression and serves a fundamental role in immune regulation. The present study aimed to develop a novel immune-related lncRNA signature to assess the prognosis of patients with colorectal cancer (CRC). Transcriptome data and clinical information of patients with CRC were downloaded from The Cancer Genome Atlas (TCGA) and UCSC Xena platforms. Immune-related mRNAs were extracted from the Molecular Signatures Database (MSigDB), and the immune-related lncRNAs were identified based on correlation analysis. Then, univariate, Lasso and multivariate Cox regression were applied to construct an immune-related lncRNA signature, and CRC patients were divided into high- and low-risk groups according to the median risk score. Finally, we evaluated the signature from the perspectives of clinical outcome, clinicopathological parameters, tumor-infiltrating immune cells (TIICs), immune status, tumor mutation burden (TMB) and immunotherapy responsiveness. In total, 272 immune-related lncRNAs were identified, five of which were applied to construct an immune-related lncRNA signature. The signature divided patients with CRC into low- and high-risk groups, the prognosis of patients in the high-risk group were significantly poorer than those in low-risk group, and the results were further confirmed in external validation cohort. Furthermore, the high-risk group showed aggressive clinicopathological characteristics, specific TIIC and immune function status, and low sensitivity to immunotherapy. The immune-related lncRNA signature could be exploited as a promising biomarker for predicting the prognosis and immune status of patients with CRC.
Collapse
Affiliation(s)
- Jianxin Li
- Department of General Surgery (Gastrointestinal Surgery), The Affiliated Hospital of Southwest Medical University, 25 Taiping Street, Luzhou, 646000, Sichuan, People's Republic of China
| | - Ting Han
- Department of General Surgery (Gastrointestinal Surgery), The Affiliated Hospital of Southwest Medical University, 25 Taiping Street, Luzhou, 646000, Sichuan, People's Republic of China
| | - Xin Wang
- Department of General Surgery (Gastrointestinal Surgery), The Affiliated Hospital of Southwest Medical University, 25 Taiping Street, Luzhou, 646000, Sichuan, People's Republic of China
| | - Yinchun Wang
- Department of General Surgery (Gastrointestinal Surgery), The Affiliated Hospital of Southwest Medical University, 25 Taiping Street, Luzhou, 646000, Sichuan, People's Republic of China
| | - Xuan Chen
- Department of General Surgery (Gastrointestinal Surgery), The Affiliated Hospital of Southwest Medical University, 25 Taiping Street, Luzhou, 646000, Sichuan, People's Republic of China
| | - Wangsheng Chen
- Department of General Surgery (Gastrointestinal Surgery), The Affiliated Hospital of Southwest Medical University, 25 Taiping Street, Luzhou, 646000, Sichuan, People's Republic of China
| | - Qingqiang Yang
- Department of General Surgery (Gastrointestinal Surgery), The Affiliated Hospital of Southwest Medical University, 25 Taiping Street, Luzhou, 646000, Sichuan, People's Republic of China.
| |
Collapse
|
4
|
Song J, Zhao W, Zhang X, Tian W, Zhao X, Ma L, Cao Y, Yin Y, Zhang X, Deng X, Lu D. Mutant RIG-I enhances cancer-related inflammation through activation of circRIG-I signaling. Nat Commun 2022; 13:7096. [PMID: 36402769 PMCID: PMC9675819 DOI: 10.1038/s41467-022-34885-3] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Accepted: 11/10/2022] [Indexed: 11/21/2022] Open
Abstract
RIG-I/DDX58 plays a key role in host innate immunity. However, its therapeutic potential for inflammation-related cancers remains to be explored. Here we identify frameshift germline mutations of RIG-I occurring in patients with colon cancer. Accordingly, Rig-ifs/fs mice bearing a frameshift mutant Rig-i exhibit increased susceptibility to colitis-related colon cancer as well as enhanced inflammatory response to chemical, virus or bacteria. In addition to interruption of Rig-i mRNA translation, the Rig-i mutation changes the secondary structure of Rig-i pre-mRNA and impairs its association with DHX9, consequently inducing a circular RNA generation from Rig-i transcript, thereby, designated as circRIG-I. CircRIG-I is frequently upregulated in colon cancers and its upregulation predicts poor outcome of colon cancer. Mechanistically, circRIG-I interacts with DDX3X, which in turn stimulates MAVS/TRAF5/TBK1 signaling cascade, eventually activating IRF3-mediated type I IFN transcription and aggravating inflammatory damage. Reciprocally, all-trans retinoic acid acts as a DHX9 agonist, ameliorates immunopathology through suppression of circRIG-I biogenesis. Collectively, our results provide insight into mutant RIG-I action and propose a potential strategy for the treatment of colon cancer.
Collapse
Affiliation(s)
- Jia Song
- grid.11135.370000 0001 2256 9319Department of Geriatric Dentistry, Peking University School and Hospital of Stomatology, Beijing, 100081 P.R. China ,grid.11135.370000 0001 2256 9319Institute of Systems Biomedicine, School of Basic Medical Sciences, NHC Key Laboratory of Medical Immunology, Beijing Key Laboratory of Tumor Systems Biology, Peking University Health Science Center, Beijing, 100191 P.R. China ,grid.11135.370000 0001 2256 9319National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, NMPA Key Laboratory for Dental Materials, Beijing Laboratory of Biomedical Materials & Beijing Key Laboratory of Digital Stomatology, Peking University School and Hospital of Stomatology, Beijing, 100081 P.R. China
| | - Wei Zhao
- grid.415954.80000 0004 1771 3349Department of Clinical Laboratory, China-Japan Friendship Hospital, Beijing, 100029 P.R. China
| | - Xin Zhang
- grid.11135.370000 0001 2256 9319Institute of Systems Biomedicine, School of Basic Medical Sciences, NHC Key Laboratory of Medical Immunology, Beijing Key Laboratory of Tumor Systems Biology, Peking University Health Science Center, Beijing, 100191 P.R. China
| | - Wenyu Tian
- grid.11135.370000 0001 2256 9319Institute of Systems Biomedicine, School of Basic Medical Sciences, NHC Key Laboratory of Medical Immunology, Beijing Key Laboratory of Tumor Systems Biology, Peking University Health Science Center, Beijing, 100191 P.R. China
| | - Xuyang Zhao
- grid.11135.370000 0001 2256 9319Institute of Systems Biomedicine, School of Basic Medical Sciences, NHC Key Laboratory of Medical Immunology, Beijing Key Laboratory of Tumor Systems Biology, Peking University Health Science Center, Beijing, 100191 P.R. China
| | - Liang Ma
- grid.415954.80000 0004 1771 3349Department of Clinical Laboratory, China-Japan Friendship Hospital, Beijing, 100029 P.R. China
| | - Yongtong Cao
- grid.415954.80000 0004 1771 3349Department of Clinical Laboratory, China-Japan Friendship Hospital, Beijing, 100029 P.R. China
| | - Yuxin Yin
- grid.11135.370000 0001 2256 9319Institute of Systems Biomedicine, School of Basic Medical Sciences, NHC Key Laboratory of Medical Immunology, Beijing Key Laboratory of Tumor Systems Biology, Peking University Health Science Center, Beijing, 100191 P.R. China
| | - Xuehui Zhang
- grid.11135.370000 0001 2256 9319National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, NMPA Key Laboratory for Dental Materials, Beijing Laboratory of Biomedical Materials & Beijing Key Laboratory of Digital Stomatology, Peking University School and Hospital of Stomatology, Beijing, 100081 P.R. China ,grid.11135.370000 0001 2256 9319Department of Dental Materials & Dental Medical Devices Testing Center, Peking University School and Hospital of Stomatology, Beijing, 100081 P.R. China
| | - Xuliang Deng
- grid.11135.370000 0001 2256 9319Department of Geriatric Dentistry, Peking University School and Hospital of Stomatology, Beijing, 100081 P.R. China ,grid.11135.370000 0001 2256 9319Department of Dental Materials & Dental Medical Devices Testing Center, Peking University School and Hospital of Stomatology, Beijing, 100081 P.R. China
| | - Dan Lu
- grid.11135.370000 0001 2256 9319Institute of Systems Biomedicine, School of Basic Medical Sciences, NHC Key Laboratory of Medical Immunology, Beijing Key Laboratory of Tumor Systems Biology, Peking University Health Science Center, Beijing, 100191 P.R. China
| |
Collapse
|
5
|
Lu L, Wang H, Fang J, Zheng J, Liu B, Xia L, Li D. Overexpression of OAS1 Is Correlated With Poor Prognosis in Pancreatic Cancer. Front Oncol 2022; 12:944194. [PMID: 35898870 PMCID: PMC9309611 DOI: 10.3389/fonc.2022.944194] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2022] [Accepted: 06/21/2022] [Indexed: 11/17/2022] Open
Abstract
Background OAS1 expression in pancreatic cancer has been confirmed by many studies. However, the prognostic value and mechanism of OAS1 in pancreatic cancer have not been analyzed. Methods The RNA-seq in pancreatic cancer were obtained by UCSC XENA and GEO database. In addition, immunohistochemical validation and analysis were performed using samples from the 900th hospital. The prognosis of OAS1 was evaluated by timeROC package, Cox regression analysis, and Kaplan-Meier survival curves. Then, the main functional and biological signaling pathways enrichment and its relationship with the abundance of immune cells were analyzed by bioinformatics. Results OAS1 was highly expressed in pancreatic cancer compared with normal pancreatic tissue. High OAS1 expression was associated with poor overall survival (p<0.05). The OAS1 was significantly correlated to TNM staging (p=0.014). The timeROC analysis showed that the AUC of OAS1 was 0.734 for 3-year OS. In addition, the expression of OAS1 was significantly correlated with the abundance of a variety of immune markers. GSEA showed that enhanced signaling pathways associated with OAS1 include Apoptosis, Notch signaling pathway, and P53 signaling pathway. Conclusions OAS1 is a valuable prognostic factor in pancreatic cancer. Moreover, it may be a potential immunotherapeutic target.
Collapse
Affiliation(s)
- Lingling Lu
- Fuzong Clinical Medical College of Fujian Medical University, Fuzhou, China
| | - Huaxiang Wang
- Fuzong Clinical Medical College of Fujian Medical University, Fuzhou, China
- Department of Hepatobiliary and Pancreatic Surgery, Taihe Hospital, Affiliated Hospital of Hubei University of Medicine, Shiyan, China
| | - Jian Fang
- Department of Hepatobiliary Medicine, The Third Affiliated People's Hospital of Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Jiaolong Zheng
- Department of Hepatobiliary Disease, The 900th Hospital of the People’s Liberation Army Joint Logistics Support Force, Fuzhou, China
| | - Bang Liu
- Fuzong Clinical Medical College of Fujian Medical University, Fuzhou, China
| | - Lei Xia
- Department of Hepatobiliary Medicine, The Third Affiliated People's Hospital of Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Dongliang Li
- Fuzong Clinical Medical College of Fujian Medical University, Fuzhou, China
- Department of Hepatobiliary Disease, The 900th Hospital of the People’s Liberation Army Joint Logistics Support Force, Fuzhou, China
- *Correspondence: Dongliang Li,
| |
Collapse
|
6
|
Wang H, Zhou Y, Zhang Y, Fang S, Zhang M, Li H, Xu F, Liu L, Liu J, Zhao Q, Wang F. Subtyping of microsatellite stability colorectal cancer reveals guanylate binding protein 2 (GBP2) as a potential immunotherapeutic target. J Immunother Cancer 2022; 10:jitc-2021-004302. [PMID: 35383115 PMCID: PMC8984016 DOI: 10.1136/jitc-2021-004302] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/03/2022] [Indexed: 12/12/2022] Open
Abstract
Backgrounds Proficient-mismatch-repair or microsatellite stability (pMMR/MSS) colorectal cancer (CRC) has limited efficacy for immune checkpoint blockade (ICB) therapy and its underlying mechanism remains unclear. Guanylate binding protein 2 (GBP2) is a member of the GTPase family and is crucial to host immunity against pathogens. However, the correlations between GBP2 and immunosurveillance and immunotherapy for pMMR/MSS CRC have not been reported. Methods Unsupervised clustering was employed to classify immune class and non-immune class in 1424 pMMR/MSS patients from six independent public datasets. This binary classification was validated using immune cells or response related signatures. The correlation between GBP2 and immune microenvironment was explored using well-established biological algorithms, multiplex immunohistochemistry (mIHC), in vitro and in vivo experiments. Results We classified 1424 pMMR/MSS CRC patients into two classes, ‘immune’ and ‘non-immune’, and GBP2 was identified as a gene of interest. We found that lower GBP2 expression was correlated with poor prognosis and metastasis. GBP2 expression was also upregulated in the immune class and highly associated with interferon-γ (IFN-γ) signaling pathway and CD8 +T cell infiltration using gene set enrichment analysis, gene ontology analysis, single-cell sequencing and mIHC. Moreover, reduced GBP2 expression inhibited the antigen processing and presentation machinery and CXCL10/11 expression in MSS CRC cells on IFN-γ stimulation. A Transwell assay revealed that deletion of GBP2 in murine MSS CRC cells reduced CD8 +T cell migration. Mechanistically, GBP2 promoted signal transducer and transcription activator 1 (STAT1) phosphorylation by competing with SHP1 for binding to STAT1 in MSS CRC cells. Finally, an unsupervised subclass mapping (SubMap) algorithm showed that pMMR/MSS patients with high GBP2 expression may correlate with a favorable response to anti-PD-1 therapy. We further confirmed that GBP2 knockout reduced CD8 +T cell infiltration and blunted the efficacy of PD-1 blockade in tumor-bearing mice. Conclusions Our study reveals that pMMR/MSS CRC is immunogenically heterogeneous and that GBP2 is a promising target for combinatorial therapy with ICB.
Collapse
Affiliation(s)
- Haizhou Wang
- Department of Gastroenterology, Zhongnan Hospital of Wuhan University, Wuhan, China.,Hubei Clinical Center and Key Lab of Intestinal and Colorectal Diseases, Wuhan, China
| | - Yabo Zhou
- Department of Immunology & National Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences (CAMS) & Peking Union Medical College, Beijing, China
| | - Yangyang Zhang
- Department of Gastroenterology, Zhongnan Hospital of Wuhan University, Wuhan, China.,Hubei Clinical Center and Key Lab of Intestinal and Colorectal Diseases, Wuhan, China
| | - Shilin Fang
- Department of Pain, Renmin Hospital of Wuhan University, Wuhan, China
| | - Meng Zhang
- Department of Gastroenterology, Zhongnan Hospital of Wuhan University, Wuhan, China.,Hubei Clinical Center and Key Lab of Intestinal and Colorectal Diseases, Wuhan, China
| | - Haiou Li
- Department of Gastroenterology, Zhongnan Hospital of Wuhan University, Wuhan, China.,Hubei Clinical Center and Key Lab of Intestinal and Colorectal Diseases, Wuhan, China
| | - Fei Xu
- Department of Gastroenterology, Zhongnan Hospital of Wuhan University, Wuhan, China.,Hubei Clinical Center and Key Lab of Intestinal and Colorectal Diseases, Wuhan, China
| | - Lan Liu
- Department of Gastroenterology, Zhongnan Hospital of Wuhan University, Wuhan, China.,Hubei Clinical Center and Key Lab of Intestinal and Colorectal Diseases, Wuhan, China
| | - Jing Liu
- Department of Gastroenterology, Zhongnan Hospital of Wuhan University, Wuhan, China .,Hubei Clinical Center and Key Lab of Intestinal and Colorectal Diseases, Wuhan, China
| | - Qiu Zhao
- Department of Gastroenterology, Zhongnan Hospital of Wuhan University, Wuhan, China .,Hubei Clinical Center and Key Lab of Intestinal and Colorectal Diseases, Wuhan, China
| | - Fan Wang
- Department of Gastroenterology, Zhongnan Hospital of Wuhan University, Wuhan, China .,Hubei Clinical Center and Key Lab of Intestinal and Colorectal Diseases, Wuhan, China
| |
Collapse
|
7
|
MUC6 expression is a preferable prognostic marker for invasive mucinous adenocarcinoma of the lung. Histochem Cell Biol 2022; 157:671-684. [PMID: 35353213 DOI: 10.1007/s00418-022-02093-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/15/2022] [Indexed: 11/04/2022]
Abstract
Gastric gland mucin consists of core protein MUC6 with residues heavily glycosylated by unique O-glycans carrying α1,4-linked N-acetylglucosamine (αGlcNAc). αGlcNAc-glycosylated MUC6 protein is seen in normal gastric and duodenal glands. Decreased αGlcNAc glycosylation on MUC6-positive tumor cells is often observed in premalignant lesions of the stomach, pancreas, and bile duct, and decreased MUC6 expression is seen in invasive cancer of these organs. Lung cancer (LC) is the most common cause of cancer death worldwide. Recently, the adenocarcinoma subtype has become the most common histological subtype of LC, and one of its invasive forms is invasive mucinous adenocarcinoma (IMA). Currently, prognostic markers of LC IMA are unknown. Here, we analyzed MUC5AC, MUC6, and αGlcNAc expression in 54 IMA LC cases. MUC5AC was positively expressed in 50 (93%), MUC6 in 38 (70%), and αGlcNAc in 19 (35%). Each expression level was scored from 0 to 3. The αGlcNAc expression score was significantly decreased relative to MUC6 (P < 0.001). Interestingly, disease-free survival was significantly higher in MUC6-positive than MUC6-negative cases based on the log-rank test (P = 0.021). For in vitro analysis, we ectopically expressed MUC6 in A549 cells, derived from LC and harboring a KRAS mutation. MUC6-expressing A549 cells showed significantly lower proliferation, motility, and invasiveness than control cells. Finally, F-actin staining in MUC6-expressing cells revealed a decrease or loss of filopodia associated with decreased levels of FSCN transcripts, which encodes an actin-bundling protein fascin1 necessary for cell migration. We conclude that MUC6 expression is a preferable prognostic biomarker in IMA LC.
Collapse
|
8
|
Ahmad HI, Jabbar A, Mushtaq N, Javed Z, Hayyat MU, Bashir J, Naseeb I, Abideen ZU, Ahmad N, Chen J. Immune Tolerance vs. Immune Resistance: The Interaction Between Host and Pathogens in Infectious Diseases. Front Vet Sci 2022; 9:827407. [PMID: 35425833 PMCID: PMC9001959 DOI: 10.3389/fvets.2022.827407] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Accepted: 02/17/2022] [Indexed: 11/13/2022] Open
Abstract
The immune system is most likely developed to reduce the harmful impact of infections on the host homeostasis. This defense approach is based on the coordinated activity of innate and adaptive immune system components, which detect and target infections for containment, killing, or expulsion by the body's defense mechanisms. These immunological processes are responsible for decreasing the pathogen burden of an infected host to maintain homeostasis that is considered to be infection resistance. Immune-driven resistance to infection is connected with a second, and probably more important, defensive mechanism: it helps to minimize the amount of dysfunction imposed on host parenchymal tissues during infection without having a direct adverse effect on pathogens. Disease tolerance is a defensive approach that relies on tissue damage control systems to prevent infections from causing harm to the host. It also uncouples immune-driven resistance mechanisms from immunopathology and disease, allowing the body to fight infection more effectively. This review discussed the cellular and molecular processes that build disease tolerance to infection and the implications of innate immunity on those systems. In addition, we discuss how symbiotic relationships with microbes and their control by particular components of innate and adaptive immunity alter disease tolerance to infection.
Collapse
Affiliation(s)
- Hafiz Ishfaq Ahmad
- Department of Animal Breeding and Genetics, University of Veterinary and Animal Sciences, Lahore, Pakistan
- *Correspondence: Hafiz Ishfaq Ahmad
| | - Abdul Jabbar
- Department of Clinical Medicine, Faculty of Veterinary Science, University of Veterinary and Animal Sciences, Lahore, Pakistan
| | - Nadia Mushtaq
- Department of Biological Sciences, Faculty of Fisheries and Wildlife, University of Veterinary and Animal Sciences, Lahore, Pakistan
| | - Zainab Javed
- Institute of Pharmaceutical Sciences, Faculty of Biosciences, University of Veterinary and Animal Sciences, Lahore, Pakistan
| | - Muhammad Umar Hayyat
- Institute of Pharmaceutical Sciences, Faculty of Biosciences, University of Veterinary and Animal Sciences, Lahore, Pakistan
| | - Javaria Bashir
- Department of Medical Sciences, Sharif Medical and Dental Hospital, Lahore, Pakistan
| | - Iqra Naseeb
- Institute of Microbiology, Faculty of Veterinary Science, University of Veterinary and Animal Sciences, Lahore, Pakistan
| | - Zain Ul Abideen
- Department of Zoology, Ghazi University, Dera Ghazi Khan, Pakistan
| | - Nisar Ahmad
- Department of Livestock Management, University of Veterinary and Animal Sciences, Pattoki, Pakistan
| | - Jinping Chen
- Guangdong Key Laboratory of Animal Conservation and Resource Utilization, Guangdong Public Laboratory of Wild Animal Conservation and Utilization, Institute of Zoology, Guangdong Academy of Sciences, Guangzhou, China
- Jinping Chen
| |
Collapse
|
9
|
Yang W, Wei X, Jiao Y, Bai Y, Sam WN, Yan Q, Sun X, Li G, Ma J, Wei W, Tian D, Zheng F. STAT3/HIF-1α/fascin-1 axis promotes RA FLSs migration and invasion ability under hypoxia. Mol Immunol 2021; 142:83-94. [PMID: 34971867 DOI: 10.1016/j.molimm.2021.12.004] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Revised: 10/15/2021] [Accepted: 12/01/2021] [Indexed: 12/24/2022]
Abstract
Rheumatoid arthritis (RA) synovium was identified as "tumor-like" tissues because of the hypoxic microenvironment, significant cell proliferation, and invasion phenotypes. It was reported that hypoxia promoted tumor aggressiveness via up-regulated expression of fascin-1 in cancer. However, the role of fascin-1 in RA synovial hyperplasia and joint injury progression remains unknown. In the current study, we first identified that both fascin-1 and HIF-1α were highly expressed in the RA synovium, in which they were widely colocalized, compared to osteoarthritis(OA). As well, levels of fascin-1 in RA fibroblast-like synoviocytes(FLSs) were found significantly higher than those in OA FLSs. Further, it was demonstrated that the mRNA and protein levels of fascin-1 in RA FLSs were up-regulated in hypoxia (3 % O2) and experimental hypoxia induced by cobalt chloride. Mechanistically, the HIF-1α-mediated hypoxia environment activated the gene expression of the fascin-1 protein, which in turn promoted the migration and invasion of RA FLSs. Accordingly, the restoration of FLSs migration and invasion was observed following siRNA-mediated silencing of fascin-1 and HIF-1α expression. Notably, under the experimental hypoxia, we found that the expression levels of fascin-1, HIF-1α, and p-STAT3 were increased in a time-dependent manner, and fascin-1and HIF-1α expressions were dependent on p-STAT3. Our results indicated that hypoxia-induced fascin-1 up-regulation promoted RA FLSs migration and invasion through the STAT3/HIF-1α/fascin-1 axis, which might represent a novel therapeutic target for the treatment of RA.
Collapse
Affiliation(s)
- Wang Yang
- Department of Clinical Immunology, School of Medical Laboratory, Tianjin Medical University, Tianjin, China
| | - Xinyue Wei
- Department of Clinical Immunology, School of Medical Laboratory, Tianjin Medical University, Tianjin, China
| | - Yachong Jiao
- Department of Clinical Laboratory, The Third Hospital of Hebei Medical University, Hebei, China
| | - Yingyu Bai
- Laboratory for Mechanisms and Therapies of Heart Diseases, School of Pharmacy, Tianjin Medical University, Tianjin, China
| | - Wilfried Noel Sam
- Department of Clinical Immunology, School of Medical Laboratory, Tianjin Medical University, Tianjin, China
| | - Qiushuang Yan
- Department of Clinical Immunology, School of Medical Laboratory, Tianjin Medical University, Tianjin, China
| | - Xuguo Sun
- Department of Clinical Immunology, School of Medical Laboratory, Tianjin Medical University, Tianjin, China
| | - Guangping Li
- Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular Disease, Department of Cardiology, Tianjin Institute of Cardiology, The Second Hospital of Tianjin Medical University, Tianjin, China
| | - Jun Ma
- Department of Health Statistics, College of Public Health, Tianjin Medical University, Tianjin, China.
| | - Wei Wei
- Department of Rheumatology, General Hospital, Tianjin Medical University, Tianjin, China.
| | - Derun Tian
- Department of Clinical Laboratory Diagnostics, Tianjin Medical University, Tianjin, China.
| | - Fang Zheng
- Department of Clinical Immunology, School of Medical Laboratory, Tianjin Medical University, Tianjin, China.
| |
Collapse
|
10
|
Chathuranga K, Weerawardhana A, Dodantenna N, Lee JS. Regulation of antiviral innate immune signaling and viral evasion following viral genome sensing. Exp Mol Med 2021; 53:1647-1668. [PMID: 34782737 PMCID: PMC8592830 DOI: 10.1038/s12276-021-00691-y] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Revised: 06/15/2021] [Accepted: 09/07/2021] [Indexed: 02/07/2023] Open
Abstract
A harmonized balance between positive and negative regulation of pattern recognition receptor (PRR)-initiated immune responses is required to achieve the most favorable outcome for the host. This balance is crucial because it must not only ensure activation of the first line of defense against viral infection but also prevent inappropriate immune activation, which results in autoimmune diseases. Recent studies have shown how signal transduction pathways initiated by PRRs are positively and negatively regulated by diverse modulators to maintain host immune homeostasis. However, viruses have developed strategies to subvert the host antiviral response and establish infection. Viruses have evolved numerous genes encoding immunomodulatory proteins that antagonize the host immune system. This review focuses on the current state of knowledge regarding key host factors that regulate innate immune signaling molecules upon viral infection and discusses evidence showing how specific viral proteins counteract antiviral responses via immunomodulatory strategies.
Collapse
Affiliation(s)
- Kiramage Chathuranga
- College of Veterinary Medicine, Chungnam National University, Daejeon, 34134, Korea
| | - Asela Weerawardhana
- College of Veterinary Medicine, Chungnam National University, Daejeon, 34134, Korea
| | - Niranjan Dodantenna
- College of Veterinary Medicine, Chungnam National University, Daejeon, 34134, Korea
| | - Jong-Soo Lee
- College of Veterinary Medicine, Chungnam National University, Daejeon, 34134, Korea.
| |
Collapse
|
11
|
Liao KC, Chuo V, Fagg WS, Modahl CM, Widen S, Garcia-Blanco MA. The RNA binding protein Quaking represses splicing of the Fibronectin EDA exon and downregulates the interferon response. Nucleic Acids Res 2021; 49:10034-10045. [PMID: 34428287 PMCID: PMC8464043 DOI: 10.1093/nar/gkab732] [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] [Received: 12/07/2020] [Revised: 08/06/2021] [Accepted: 08/17/2021] [Indexed: 01/03/2023] Open
Abstract
Quaking (QKI) controls RNA metabolism in many biological processes including innate immunity, where its roles remain incompletely understood. To illuminate these roles, we performed genome scale transcriptome profiling in QKI knockout cells with or without poly(I:C) transfection, a double-stranded RNA analog that mimics viral infection. Analysis of RNA-sequencing data shows that QKI knockout upregulates genes induced by interferons, suggesting that QKI is an immune suppressor. Furthermore, differential splicing analysis shows that QKI primarily controls cassette exons, and among these events, we noted that QKI silences splicing of the extra domain A (EDA) exon in fibronectin (FN1) transcripts. QKI knockout results in elevated production and secretion of FN1-EDA protein, which is a known activator of interferons. Consistent with an upregulation of the interferon response in QKI knockout cells, our results show reduced production of dengue virus-2 and Japanese encephalitis virus in these cells. In conclusion, we demonstrate that QKI downregulates the interferon system and attenuates the antiviral state.
Collapse
Affiliation(s)
- Kuo-Chieh Liao
- Programme in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore 169857, Singapore
| | - Vanessa Chuo
- Programme in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore 169857, Singapore
| | - W Samuel Fagg
- Transplant Division, Department of Surgery, University of Texas Medical Branch, Galveston, TX 77555, USA.,Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Cassandra M Modahl
- Department of Biological Sciences, National University of Singapore, Singapore 119077, Singapore
| | - Steven Widen
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Mariano A Garcia-Blanco
- Programme in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore 169857, Singapore.,Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, TX 77555, USA.,Department of Internal Medicine, University of Texas Medical Branch, Galveston, TX 77555, USA.,Institute for Human Infections and Immunity, University of Texas Medical Branch, Galveston, TX 77555, USA
| |
Collapse
|
12
|
Actin Cytoskeleton Dynamics and Type I IFN-Mediated Immune Response: A Dangerous Liaison in Cancer? BIOLOGY 2021; 10:biology10090913. [PMID: 34571790 PMCID: PMC8469949 DOI: 10.3390/biology10090913] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Revised: 09/06/2021] [Accepted: 09/09/2021] [Indexed: 12/13/2022]
Abstract
Simple Summary Actin cytoskeleton is a dynamic subcellular component critical for maintaining cell shape and for elaborating response to any stimulus converging on the cell. Cytoskeleton constantly interfaces with diverse cellular components and affects a wide range of processes important in homeostasis and disease. What has been clearly demonstrated to date is that pathogens modify and use host cytoskeleton to their advantage. What is now emerging is that in sterile conditions, when a chronic inflammation occurs as in cancer, the subversion of tissue homeostasis induces an alarm status which mimics infection. This activates cellular players similar to those that solve an infection, but their persistence may pave the way for tumor progression. Understanding molecular mechanisms engaged by cytoskeleton to induce this viral mimicry could improve our knowledge of processes governing tumor progression and resistance to therapy. Abstract Chronic viral infection and cancer are closely inter-related and are both characterized by profound alteration of tissue homeostasis. The actin cytoskeleton dynamics highly participate in tissue homeostasis and act as a sensor leading to an immune-mediated anti-cancer and anti-viral response. Herein we highlight the crucial role of actin cytoskeleton dynamics in participating in a viral mimicry activation with profound effect in anti-tumor immune response. This still poorly explored field understands the cytoskeleton dynamics as a platform of complex signaling pathways which may regulate Type I IFN response in cancer. This emerging network needs to be elucidated to identify more effective anti-cancer strategies and to further advance the immuno-oncology field which has revolutionized the cancer treatment. For a progress to occur in this exciting arena we have to shed light on actin cytoskeleton related pathways and immune response. Herein we summarize the major findings, considering the double sword of the immune response and in particular the role of Type I IFN pathways in resistance to anti-cancer treatment.
Collapse
|
13
|
Hu F, Wang Q, Yang Z, Zhang Z, Liu X. Network-based identification of biomarkers for colon adenocarcinoma. BMC Cancer 2020; 20:668. [PMID: 32680494 PMCID: PMC7367377 DOI: 10.1186/s12885-020-07157-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2020] [Accepted: 07/09/2020] [Indexed: 12/15/2022] Open
Abstract
Background As one of the most common cancers with high mortality in the world, we are still facing a huge challenge in the prevention and treatment of colon cancer. With the rapid development of high throughput technologies, new biomarkers identification for colon cancer has been confronted with the new opportunities and challenges. Methods We firstly constructed functional networks for each sample of colon adenocarcinoma (COAD) by using a sample-specific network (SSN) method which can construct individual-specific networks based on gene expression profiles of a single sample. The functional genes and interactions were identified from the functional networks, respectively. Results Classification and subtyping were used to test the function of the functional genes and interactions. The results of classification showed that the functional genes could be used as diagnostic biomarkers. The subtypes displayed different mechanisms, which were shown by the functional and pathway enrichment analysis for the representative genes of each subtype. Besides, subtype-specific molecular patterns were also detected, such as subtype-specific clinical and mutation features. Finally, 12 functional genes and 13 functional edges could serve as prognosis biomarkers since they were associated with the survival rate of COAD. Conclusions In conclusion, the functional genes and interactions in the constructed functional network could be used as new biomarkers for COAD.
Collapse
Affiliation(s)
- Fuyan Hu
- Department of Statistics, School of Science, Wuhan University of Technology, 122 Luoshi Road, Wuhan, China
| | - Qing Wang
- Department of Traditional Chinese Medicine of Wuhan Puren Hospital, Affiliated Hospital of Wuhan University of Science and Technology, Benxi Street 1#, Qingshan District, Wuhan, Hubei, P.R. China
| | - Zhiyuan Yang
- College of Life Information Science & Instrument Engineering, Hangzhou Dianzi University, Hangzhou, People's Republic of China
| | - Zeng Zhang
- Department of Statistics, School of Science, Wuhan University of Technology, 122 Luoshi Road, Wuhan, China
| | - Xiaoping Liu
- School of Mathematics and Statistics, Shandong University, Weihai, 264209, China.
| |
Collapse
|
14
|
Biology and Therapeutic Targets of Colorectal Serrated Adenocarcinoma; Clues for a Histologically Based Treatment against an Aggressive Tumor. Int J Mol Sci 2020; 21:ijms21061991. [PMID: 32183342 PMCID: PMC7139914 DOI: 10.3390/ijms21061991] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Revised: 03/04/2020] [Accepted: 03/09/2020] [Indexed: 02/06/2023] Open
Abstract
Serrated adenocarcinoma (SAC) is a tumor recognized by the WHO as a histological subtype accounting for around 9% of colorectal carcinomas. Compared to conventional carcinomas, SACs are characterized by a worse prognosis, weak development of the immune response, an active invasive front and a frequent resistance to targeted therapy due to a high occurrence of KRAS or BRAF mutation. Nonetheless, several high-throughput studies have recently been carried out unveiling the biology of this cancer and identifying potential molecular targets, favoring a future histologically based treatment. This review revises the current evidence, aiming to propose potential molecular targets and specific treatments for this aggressive tumor.
Collapse
|
15
|
Gao W, An C, Xue X, Zheng X, Niu M, Zhang Y, Liu H, Zhang C, Lu Y, Cui J, Zhao Q, Wen S, Thorne RF, Zhang X, Wu Y, Wang B. Mass Spectrometric Analysis Identifies AIMP1 and LTA4H as FSCN1-Binding Proteins in Laryngeal Squamous Cell Carcinoma. Proteomics 2019; 19:e1900059. [PMID: 31287215 DOI: 10.1002/pmic.201900059] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2019] [Revised: 06/29/2019] [Indexed: 12/24/2022]
Abstract
Dysregulation of fascin actin-bundling protein 1 (FSCN1) enhances cell proliferation, invasion, and motility in laryngeal squamous cell carcinoma (LSCC), while the mechanism remains unclear. Here, co-immunoprecipitation and mass spectrometry is utilized to identify potential FSCN1-binding proteins. Functional annotation of FSCN1-binding proteins are performed by Gene Ontology and Kyoto Encyclopedia of Genes and Genomes pathway analysis. Furthermore, the protein-protein interaction network of FSNC1-binding proteins is constructed and the interactions between FSCN1 and novel identified interacting proteins AIMP1 and LTA4H are validated. Moreover, the expression and functional role of AIMP1 and LTA4H in LSCC are investigated. A total of 123 proteins are identified as potential FSCN1-binding proteins, and functional annotation shows that FSCN1-binding proteins are significantly enriched in carcinogenic processes, such as filopodium assembly-regulation and GTPase activity. Co-IP/western blotting and immunofluorescence confirm that AIMP1 and LTA4H bind and colocalize with FSCN1. Furthermore, both AIMP1 and LTA4H are upregulated in LSCC tissues, and knockdown of AIMP1 or LTA4H inhibits LSCC cell proliferation, migration, and invasion. Collectively, the identification of FSCN1-binding partners enhances understanding of the mechanism of FSCN1-mediated malignant phenotypes, and these findings indicate that FSCN1 binds to AIMP1 and LTA4H might promote the progression of LSCC.
Collapse
Affiliation(s)
- Wei Gao
- Shanxi Key Laboratory of Otorhinolaryngology, Head and Neck Cancer, Shanxi Medical University, Taiyuan, 030001, Shanxi, China.,Department of Otolaryngology Head & Neck Surgery, The First Hospital of Shanxi Medical University, Taiyuan, 030001, Shanxi, China.,Otolaryngology Head & Neck Surgery Research Institute, Shanxi Medical University, Taiyuan, 030001, Shanxi, China.,The Key Scientific and Technological Innovation Platform for Precision Diagnosis and Treatment of Head and Neck Cancer, Taiyuan, 030001, Shanxi, China
| | - Changming An
- Department of Head and Neck Surgery Cancer Hospital, National Cancer Center, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100021, China
| | - Xuting Xue
- Shanxi Key Laboratory of Otorhinolaryngology, Head and Neck Cancer, Shanxi Medical University, Taiyuan, 030001, Shanxi, China.,Department of Otolaryngology Head & Neck Surgery, The First Hospital of Shanxi Medical University, Taiyuan, 030001, Shanxi, China.,Otolaryngology Head & Neck Surgery Research Institute, Shanxi Medical University, Taiyuan, 030001, Shanxi, China.,The Key Scientific and Technological Innovation Platform for Precision Diagnosis and Treatment of Head and Neck Cancer, Taiyuan, 030001, Shanxi, China
| | - Xiwang Zheng
- Shanxi Key Laboratory of Otorhinolaryngology, Head and Neck Cancer, Shanxi Medical University, Taiyuan, 030001, Shanxi, China.,Department of Otolaryngology Head & Neck Surgery, The First Hospital of Shanxi Medical University, Taiyuan, 030001, Shanxi, China.,Otolaryngology Head & Neck Surgery Research Institute, Shanxi Medical University, Taiyuan, 030001, Shanxi, China.,The Key Scientific and Technological Innovation Platform for Precision Diagnosis and Treatment of Head and Neck Cancer, Taiyuan, 030001, Shanxi, China
| | - Min Niu
- Shanxi Key Laboratory of Otorhinolaryngology, Head and Neck Cancer, Shanxi Medical University, Taiyuan, 030001, Shanxi, China.,Department of Otolaryngology Head & Neck Surgery, The First Hospital of Shanxi Medical University, Taiyuan, 030001, Shanxi, China.,Otolaryngology Head & Neck Surgery Research Institute, Shanxi Medical University, Taiyuan, 030001, Shanxi, China.,The Key Scientific and Technological Innovation Platform for Precision Diagnosis and Treatment of Head and Neck Cancer, Taiyuan, 030001, Shanxi, China
| | - Yuliang Zhang
- Shanxi Key Laboratory of Otorhinolaryngology, Head and Neck Cancer, Shanxi Medical University, Taiyuan, 030001, Shanxi, China.,Department of Otolaryngology Head & Neck Surgery, The First Hospital of Shanxi Medical University, Taiyuan, 030001, Shanxi, China.,Otolaryngology Head & Neck Surgery Research Institute, Shanxi Medical University, Taiyuan, 030001, Shanxi, China.,The Key Scientific and Technological Innovation Platform for Precision Diagnosis and Treatment of Head and Neck Cancer, Taiyuan, 030001, Shanxi, China
| | - Hongliang Liu
- Shanxi Key Laboratory of Otorhinolaryngology, Head and Neck Cancer, Shanxi Medical University, Taiyuan, 030001, Shanxi, China.,Department of Otolaryngology Head & Neck Surgery, The First Hospital of Shanxi Medical University, Taiyuan, 030001, Shanxi, China.,Otolaryngology Head & Neck Surgery Research Institute, Shanxi Medical University, Taiyuan, 030001, Shanxi, China.,The Key Scientific and Technological Innovation Platform for Precision Diagnosis and Treatment of Head and Neck Cancer, Taiyuan, 030001, Shanxi, China
| | - Chunming Zhang
- Shanxi Key Laboratory of Otorhinolaryngology, Head and Neck Cancer, Shanxi Medical University, Taiyuan, 030001, Shanxi, China.,Department of Otolaryngology Head & Neck Surgery, The First Hospital of Shanxi Medical University, Taiyuan, 030001, Shanxi, China.,Otolaryngology Head & Neck Surgery Research Institute, Shanxi Medical University, Taiyuan, 030001, Shanxi, China.,The Key Scientific and Technological Innovation Platform for Precision Diagnosis and Treatment of Head and Neck Cancer, Taiyuan, 030001, Shanxi, China
| | - Yan Lu
- Department of Otolaryngology Head & Neck Surgery, The First Hospital, Jinzhou Medical University, Jinzhou, 121001, Liaoning, China
| | - Jiajia Cui
- Shanxi Key Laboratory of Otorhinolaryngology, Head and Neck Cancer, Shanxi Medical University, Taiyuan, 030001, Shanxi, China.,Department of Otolaryngology Head & Neck Surgery, The First Hospital of Shanxi Medical University, Taiyuan, 030001, Shanxi, China.,Otolaryngology Head & Neck Surgery Research Institute, Shanxi Medical University, Taiyuan, 030001, Shanxi, China.,The Key Scientific and Technological Innovation Platform for Precision Diagnosis and Treatment of Head and Neck Cancer, Taiyuan, 030001, Shanxi, China
| | - Qinli Zhao
- Shanxi Key Laboratory of Otorhinolaryngology, Head and Neck Cancer, Shanxi Medical University, Taiyuan, 030001, Shanxi, China.,Department of Otolaryngology Head & Neck Surgery, The First Hospital of Shanxi Medical University, Taiyuan, 030001, Shanxi, China
| | - Shuxin Wen
- Shanxi Key Laboratory of Otorhinolaryngology, Head and Neck Cancer, Shanxi Medical University, Taiyuan, 030001, Shanxi, China.,Department of Otolaryngology Head & Neck Surgery, The First Hospital of Shanxi Medical University, Taiyuan, 030001, Shanxi, China.,Otolaryngology Head & Neck Surgery Research Institute, Shanxi Medical University, Taiyuan, 030001, Shanxi, China.,The Key Scientific and Technological Innovation Platform for Precision Diagnosis and Treatment of Head and Neck Cancer, Taiyuan, 030001, Shanxi, China
| | - Rick F Thorne
- Translational Research Institute, Henan Provincial People's Hospital, School of Medicine, Henan University, Zhengzhou, 450053, Henan, China.,School of Environmental and Life Sciences, The University of Newcastle, Callaghan, NSW, 2308, Australia
| | - Xudong Zhang
- School of Biomedical Sciences and Pharmacy, The University of Newcastle, Callaghan, NSW, 2308, Australia
| | - Yongyan Wu
- Shanxi Key Laboratory of Otorhinolaryngology, Head and Neck Cancer, Shanxi Medical University, Taiyuan, 030001, Shanxi, China.,Department of Otolaryngology Head & Neck Surgery, The First Hospital of Shanxi Medical University, Taiyuan, 030001, Shanxi, China.,Otolaryngology Head & Neck Surgery Research Institute, Shanxi Medical University, Taiyuan, 030001, Shanxi, China.,The Key Scientific and Technological Innovation Platform for Precision Diagnosis and Treatment of Head and Neck Cancer, Taiyuan, 030001, Shanxi, China
| | - Binquan Wang
- Shanxi Key Laboratory of Otorhinolaryngology, Head and Neck Cancer, Shanxi Medical University, Taiyuan, 030001, Shanxi, China.,Department of Otolaryngology Head & Neck Surgery, The First Hospital of Shanxi Medical University, Taiyuan, 030001, Shanxi, China.,Otolaryngology Head & Neck Surgery Research Institute, Shanxi Medical University, Taiyuan, 030001, Shanxi, China.,The Key Scientific and Technological Innovation Platform for Precision Diagnosis and Treatment of Head and Neck Cancer, Taiyuan, 030001, Shanxi, China
| |
Collapse
|
16
|
Chen Y, Bi F, An Y, Yang Q. Identification of pathological grade and prognosis-associated lncRNA for ovarian cancer. J Cell Biochem 2019; 120:14444-14454. [PMID: 31034644 DOI: 10.1002/jcb.28704] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2018] [Revised: 03/02/2019] [Accepted: 03/15/2019] [Indexed: 12/13/2022]
Abstract
Ovarian carcinoma (OC) is one of the most common malignant tumors in female genitals. In recent years, the therapeutic effect of OC has been significantly improved through the application of effective chemotherapy regimen. However, the 5-year survival rate is also lower than 30% due to high rate of relapse. So, it is needed to screen reliable predictive and prognostic markers of OC. Ovarian cancer gene expression data and corresponding clinical data used were downloaded from Gene Expression Omnibus database. Weighted gene expression network analysis (WGCNA) and Cox proportional hazards regression (PHR) were used to screen Pathological Grade and Prognosis-associated long noncoding RNA (lncRNA). Kaplan-Meier analysis and receiver operating characteristic curves analysis were performed to evaluate the predictive ability of the selected lncRNA. Gene Ontology (GO) enrichment and Gene Set Enrichment Analysis (GSEA) enrichment analysis methods were used to explore the possible mechanisms of the selected lncRNA affecting the development of OC. Five reliably lncRNAs (LINC00664, LINC00667, LINC01139, LINC01419, and LOC286437) was identified through a series of bioinformatics methods. In testing cohorts, we found that the five lncRNAs in predicting the risk of OC recurrence is robustness, and multivariate Cox PHR analysis indicate that the five lncRNAs is an independent risk factor for OC recurrence. Moreover, GO and GSEA enrichment analysis showed that the five lncRNAs are involved in multiple ovarian cancer occurrence mechanism. In summary, all these findings indicated that the five lncRNAs can effectively predict the risk of recurrence of ovarian cancer.
Collapse
Affiliation(s)
- Ying Chen
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Fangfang Bi
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Yuanyuan An
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Qing Yang
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, China
| |
Collapse
|