1
|
Yang H, Xiong R, Zhang R, Sun S, Pan Y, Zhao Q, Bie J, Luo Y, Song G, Liu K. LINC01305 and LAD1 Co-Regulate CTTN and N-WASP Phosphorylation, Mediating Cytoskeletal Reorganization to Promote ESCC Metastasis. Mol Carcinog 2025; 64:756-768. [PMID: 39835575 PMCID: PMC11890417 DOI: 10.1002/mc.23885] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Revised: 12/29/2024] [Accepted: 01/07/2025] [Indexed: 01/22/2025]
Abstract
Esophageal squamous cell carcinoma (ESCC) is prone to metastasis and is a leading cause of mortality. The cytoskeleton is closely related to cell morphology and movement; however, little research has been conducted on ESCC metastasis. In this study, we found that the anchoring filament protein ladinin 1 (LAD1) specifically binds to LINC01305 for co-regulating the level of modulating cortactin proteins (CTTN) and neuronal Wiskott-Aldrich syndrome protein (N-WASP) phosphorylation, which mediates cytoskeletal reorganization and affects the metastasis of ESCC cells. Additionally, LINC01305 and LAD1 jointly promoted the epithelial-mesenchymal transition (EMT) process by activating the phosphoinositide-3-kinase-protein kinase B (PI3K/AKT) signaling pathway. Moreover, LINC01305 and LAD1 were related to the late clinical stage and lymph node metastasis of ESCC. Our study demonstrated that LINC01305 and LAD1 are major determinants of ESCC dissemination and revealed a novel molecular mechanism of cytoskeletal reorganization that controls ESCC metastasis. Trial Registration: N/A.
Collapse
Grants
- This work was supported by the National Institutes of the National Natural Science Foundation of China (82203851), the Sichuan Science and Technology Program (2024NSFSC1973, 2023YFS0473, MZGC20240072, MZGC20240071), the Nanchong Science and Technology Program (22SXQT0336, 20SXQT0328, 22SXQT0087, 22SXQT0340, 20SXQT0181), the Sichuan Medical Association Science and Technology Program (S2024009), and the North Sichuan Medical College Science and Technology Program (CBY24-KP03, CBY22-QDA01).
Collapse
Affiliation(s)
- Hang Yang
- Institute of Tissue Engineering and Stem Cells, Beijing Anzhen Nanchong Hospital of Capital Medical University, Nanchong Central Hospital, The Second Clinical Medical College of North Sichuan Medical CollegeNanchongChina
- Institute of Basic Medicine and Forensic Medicine, North Sichuan Medical CollegeNanchongChina
| | - Rong Xiong
- Institute of Tissue Engineering and Stem Cells, Beijing Anzhen Nanchong Hospital of Capital Medical University, Nanchong Central Hospital, The Second Clinical Medical College of North Sichuan Medical CollegeNanchongChina
| | - Ruolan Zhang
- Institute of Tissue Engineering and Stem Cells, Beijing Anzhen Nanchong Hospital of Capital Medical University, Nanchong Central Hospital, The Second Clinical Medical College of North Sichuan Medical CollegeNanchongChina
| | - Shan Sun
- Institute of Tissue Engineering and Stem Cells, Beijing Anzhen Nanchong Hospital of Capital Medical University, Nanchong Central Hospital, The Second Clinical Medical College of North Sichuan Medical CollegeNanchongChina
- Institute of Basic Medicine and Forensic Medicine, North Sichuan Medical CollegeNanchongChina
| | - Yingjie Pan
- Institute of Tissue Engineering and Stem Cells, Beijing Anzhen Nanchong Hospital of Capital Medical University, Nanchong Central Hospital, The Second Clinical Medical College of North Sichuan Medical CollegeNanchongChina
- Institute of Basic Medicine and Forensic Medicine, North Sichuan Medical CollegeNanchongChina
| | - Quanneng Zhao
- Institute of Tissue Engineering and Stem Cells, Beijing Anzhen Nanchong Hospital of Capital Medical University, Nanchong Central Hospital, The Second Clinical Medical College of North Sichuan Medical CollegeNanchongChina
| | - Jun Bie
- Institute of Tissue Engineering and Stem Cells, Beijing Anzhen Nanchong Hospital of Capital Medical University, Nanchong Central Hospital, The Second Clinical Medical College of North Sichuan Medical CollegeNanchongChina
| | - Yi Luo
- Institute of Tissue Engineering and Stem Cells, Beijing Anzhen Nanchong Hospital of Capital Medical University, Nanchong Central Hospital, The Second Clinical Medical College of North Sichuan Medical CollegeNanchongChina
| | - Guiqin Song
- Institute of Basic Medicine and Forensic Medicine, North Sichuan Medical CollegeNanchongChina
| | - Kang Liu
- Institute of Tissue Engineering and Stem Cells, Beijing Anzhen Nanchong Hospital of Capital Medical University, Nanchong Central Hospital, The Second Clinical Medical College of North Sichuan Medical CollegeNanchongChina
| |
Collapse
|
2
|
Shen J, Ding Y. Multifaceted roles of insulin‑like growth factor 2 mRNA binding protein 2 in human cancer (Review). Mol Med Rep 2025; 31:75. [PMID: 39886962 PMCID: PMC11795254 DOI: 10.3892/mmr.2025.13441] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Accepted: 01/07/2024] [Indexed: 02/01/2025] Open
Abstract
Insulin‑like growth factor 2 mRNA binding protein 2 (IGF2BP2) is an RNA binding protein that functions as an N6‑methyladenosine reader. It regulates various biological processes in human cancers by affecting the stability and expression of target RNA transcripts, including coding RNAs and non‑coding RNAs (ncRNAs). Numerous studies have shown that IGF2BP2 expression is aberrantly increased in various types of cancer and plays multifaceted roles in the development and progression of human cancers. In the present review, the clinical importance of IGF2BP2 is summarized and its involvement in the regulation of biological processes, including proliferation, metastasis, chemoresistance, metabolism, tumor immunity, stemness and cell death, in human cancers is discussed. The chemical compounds that have been developed as IGF2BP2 inhibitors are also detailed. As ncRNAs are now important potential therapeutic agents for cancer treatment, the microRNAs that have been reported to directly target and inhibit IGF2BP2 expression in cancers are also described. In summary, by reviewing the latest literature, the present study aimed to highlight the clinical importance and physiological functions of IGF2BP2 in human cancer, with a focus on the great potential of IGF2BP2 as a target for inhibitor development. The present review may inspire new ideas for future studies on IGF2BP2, which may serve as a specific therapeutic target in cancer.
Collapse
Affiliation(s)
- Jianan Shen
- Department of Pathology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu 210008, P.R. China
- School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu 210009, P.R. China
| | - Youxiang Ding
- Department of Pathology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu 210008, P.R. China
| |
Collapse
|
3
|
Xie X, Fang Z, Zhang H, Wang Z, Li J, Jia Y, Shang L, Cao F, Li F. The role of N(6)-methyladenosine (m6a) modification in cancer: recent advances and future directions. EXCLI JOURNAL 2025; 24:113-150. [PMID: 39967906 PMCID: PMC11830918 DOI: 10.17179/excli2024-7935] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Accepted: 12/18/2024] [Indexed: 02/20/2025]
Abstract
N(6)-methyladenosine (m6A) modification is the most abundant and prevalent internal modification in eukaryotic mRNAs. The role of m6A modification in cancer has become a hot research topic in recent years and has been widely explored. m6A modifications have been shown to regulate cancer occurrence and progression by modulating different target molecules. This paper reviews the recent research progress of m6A modifications in cancer and provides an outlook on future research directions, especially the development of molecularly targeted drugs. See also the graphical abstract(Fig. 1).
Collapse
Affiliation(s)
- Xiaozhou Xie
- Department of General Surgery, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Zhen Fang
- Department of Gastrointestinal Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Haoyu Zhang
- Department of General Surgery, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Zheng Wang
- Department of General Surgery, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Jie Li
- Department of General Surgery, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Yuchen Jia
- Department of General Surgery, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Liang Shang
- Department of Gastrointestinal Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Feng Cao
- Department of General Surgery, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Fei Li
- Department of General Surgery, Xuanwu Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
4
|
Liu X, Guo H, Kang M, Fu W, Li H, Ji H, Zhao J, Fang Y, Du M, Xue Y. Multi-step gene set analysis identified HTR3 family genes involving childhood acute lymphoblastic leukemia susceptibility. Arch Toxicol 2025; 99:299-307. [PMID: 39322821 DOI: 10.1007/s00204-024-03881-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Accepted: 09/19/2024] [Indexed: 09/27/2024]
Abstract
In our previous conventional genome-wide association study (GWAS), WWOX was a susceptibility gene associated with acute lymphoblastic leukemia (ALL) development. Nowadays, advancements in genetic association analyses promote an in-depth exploration of ALL genomics. We conducted a two-step enrichment analysis at both gene and pathway levels based on ALL GWAS data including 269 cases and 1039 controls of Chinese descent. The following functional prediction and experiments were used to evaluate the genetic biology of candidate variants and genes. The serotonin-activated cation-selective channel complex gene-set was a potential biological pathway involved in ALL occurrence. Of which, individuals carrying the T allele of rs33940208 exhibited a prominent reduced risk of ALL [odds ratio (OR) = 0.71, 95% confidence interval (CI) = 0.53 to 0.96, P = 2.81 × 10-2], whereas those with the A allele of rs6779545 demonstrated an increased risk (OR = 1.23, 95% CI = 1.01 to 1.51, P = 4.11 × 10-2). Notably, the two variants displayed a better prediction capability when combined, that the risk of developing childhood ALL increased by 131% in subjects with 2-4 risk alleles compared to those with 0-1 risk alleles (Ptrend = 2.05 × 10-3). In addition, the T allele of rs33940208 could reduce HTR3A mRNA level, while the A allele of rs6779545 increased HTR3D mRNA expression. In this study, we identified HTR3A rs33940208 and HTR3D rs6779545 as potential susceptibility loci for ALL in Chinese children. Future validation and functional research will elucidate the underlying molecular processes, refining preventive strategies for this disease.
Collapse
Affiliation(s)
- Xiao Liu
- Department of Biostatistics, Center for Global Health, School of Public Health, Nanjing Medical University, No. 101 Longmian Avenue, Nanjing, 211166, China
| | - Honghao Guo
- Department of Hematology, Nanjing Drum Tower Hospital, Clinical College of Nanjing Medical University, Nanjing, China
| | - Meiyun Kang
- Department of Hematology and Oncology, Children's Hospital of Nanjing Medical University, Nanjing, China
- Key Laboratory of Hematology, Nanjing Medical University, Nanjing, China
| | - Wenfeng Fu
- Department of Hematology and Oncology, Children's Hospital of Nanjing Medical University, Nanjing, China
- Key Laboratory of Hematology, Nanjing Medical University, Nanjing, China
| | - Huiqin Li
- Department of Genetic Toxicology and Environmental Genomics, The Key Laboratory of Modern Toxicology of Ministry of Education, Center for Global Health, Jiangsu Key Laboratory of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Hongsheng Ji
- Department of Biostatistics, Center for Global Health, School of Public Health, Nanjing Medical University, No. 101 Longmian Avenue, Nanjing, 211166, China
| | - Jiou Zhao
- Department of Hematology and Oncology, Children's Hospital of Nanjing Medical University, Nanjing, China
- Key Laboratory of Hematology, Nanjing Medical University, Nanjing, China
| | - Yongjun Fang
- Department of Hematology and Oncology, Children's Hospital of Nanjing Medical University, Nanjing, China.
- Key Laboratory of Hematology, Nanjing Medical University, Nanjing, China.
- Department of Hematology and Oncology, Children's Hospital of Nanjing Medical University, Key Laboratory of Hematology, Nanjing Medical University, No. 72 Guangzhou Road, Nanjing, 210008, China.
| | - Mulong Du
- Department of Biostatistics, Center for Global Health, School of Public Health, Nanjing Medical University, No. 101 Longmian Avenue, Nanjing, 211166, China.
- Department of Genetic Toxicology and Environmental Genomics, The Key Laboratory of Modern Toxicology of Ministry of Education, Center for Global Health, Jiangsu Key Laboratory of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, School of Public Health, Nanjing Medical University, Nanjing, China.
| | - Yao Xue
- Department of Hematology and Oncology, Children's Hospital of Nanjing Medical University, Nanjing, China.
- Key Laboratory of Hematology, Nanjing Medical University, Nanjing, China.
- Department of Hematology and Oncology, Children's Hospital of Nanjing Medical University, Key Laboratory of Hematology, Nanjing Medical University, No. 72 Guangzhou Road, Nanjing, 210008, China.
| |
Collapse
|
5
|
Jia Y, Liu S, Zhang M, Wu X, Chen X, Xing M, Hou X, Jiang W. The m6A reader IGF2BP2 promotes ESCC progression by stabilizing HDGF mRNA. J Cancer Res Ther 2024; 20:1173-1185. [PMID: 39206979 DOI: 10.4103/jcrt.jcrt_2272_23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2023] [Accepted: 02/02/2024] [Indexed: 09/04/2024]
Abstract
OBJECTIVE This study aimed to explore the role of IGF2BP2 in esophageal squamous cell carcinoma (ESCC) progression. MATERIALS AND METHODS The Cancer Genome Atlas (TCGA) dataset, transcriptome sequencing, and the Gene Expression Omnibus (GEO) dataset were used to detect the expression of m6A-associated genes in ESCC. The in vitro and in vivo assays were used to explore the role of IGF2BP2 in ESCC. RESULTS IGF2BP2 was significantly overexpressed in human ESCC specimens, which was confirmed by analyzing the GEO dataset. IGF2BP2 overexpression was correlated with poor prognosis in patients with ESCC. Altering the expression of IGF2BP2 influenced the proliferation, migration, and invasion of ESCC cells in vitro and tumorigenicity in vivo. IGF2BP2 could bind to and stabilize hepatoma-derived growth factor (HDGF) transcripts in ESCC in an m6A-dependent manner and promote HDGF expression. CONCLUSIONS These findings indicate that the novel IGF2BP2-HDGF axis is pivotal for ESCC cancer progression and can serve as a target for developing therapeutics.
Collapse
Affiliation(s)
- Yang Jia
- Department of Thoracic Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, No. 324, Jing Wu Road, Jinan, China
| | - Sujing Liu
- Department of Oncology, Shandong Provincial Third Hospital Shandong University, No. 12, Wu Ying Shan Zhong Road, Jinan, China
| | - Miao Zhang
- Department of Radiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, No. 324, Jing Wu Road, Jinan, China
| | - Xia Wu
- Department of Oncology, Shandong Provincial Third Hospital Shandong University, No. 12, Wu Ying Shan Zhong Road, Jinan, China
| | - Xiangyu Chen
- Department of Thoracic Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, No. 324, Jing Wu Road, Jinan, China
| | - Mengmeng Xing
- Department of Thoracic Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, No. 324, Jing Wu Road, Jinan, China
| | - Xianghui Hou
- Department of Thoracic Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, No. 324, Jing Wu Road, Jinan, China
| | - Wenpeng Jiang
- Department of Thoracic Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, No. 324, Jing Wu Road, Jinan, China
| |
Collapse
|
6
|
Ashrafizadeh M, Dai J, Torabian P, Nabavi N, Aref AR, Aljabali AAA, Tambuwala M, Zhu M. Circular RNAs in EMT-driven metastasis regulation: modulation of cancer cell plasticity, tumorigenesis and therapy resistance. Cell Mol Life Sci 2024; 81:214. [PMID: 38733529 PMCID: PMC11088560 DOI: 10.1007/s00018-024-05236-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Revised: 03/05/2024] [Accepted: 04/03/2024] [Indexed: 05/13/2024]
Abstract
The non-coding RNAs comprise a large part of human genome lack of capacity in encoding functional proteins. Among various members of non-coding RNAs, the circular RNAs (circRNAs) have been of importance in the pathogenesis of human diseases, especially cancer. The circRNAs have a unique closed loop structure and due to their stability, they are potential diagnostic and prognostic factors in cancer. The increasing evidences have highlighted the role of circRNAs in the modulation of proliferation and metastasis of cancer cells. On the other hand, metastasis has been responsible for up to 90% of cancer-related deaths in patients, requiring more investigation regarding the underlying mechanisms modulating this mechanism. EMT enhances metastasis and invasion of tumor cells, and can trigger resistance to therapy. The cells demonstrate dynamic changes during EMT including transformation from epithelial phenotype into mesenchymal phenotype and increase in N-cadherin and vimentin levels. The process of EMT is reversible and its reprogramming can disrupt the progression of tumor cells. The aim of current review is to understanding the interaction of circRNAs and EMT in human cancers and such interaction is beyond the regulation of cancer metastasis and can affect the response of tumor cells to chemotherapy and radiotherapy. The onco-suppressor circRNAs inhibit EMT, while the tumor-promoting circRNAs mediate EMT for acceleration of carcinogenesis. Moreover, the EMT-inducing transcription factors can be controlled by circRNAs in different human tumors.
Collapse
Affiliation(s)
- Milad Ashrafizadeh
- Department of Radiation Oncology, Shandong Provincial Key Laboratory of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University, Shandong Academy of Medical Sciences, Jinan, Shandong 250000, China
- Department of General Surgery and Integrated Chinese and Western Medicine, Institute of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Carson International Cancer Center, Shenzhen University General Hospital, Shenzhen University, Shenzhen, Guangdong, 518060, China
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Jingyuan Dai
- School of computer science and information systems, Northwest Missouri State University, Maryville, MO, 64468, USA.
| | - Pedram Torabian
- Cumming School of Medicine, Arnie Charbonneau Cancer Research Institute, University of Calgary, Calgary, AB, T2N 4Z6, Canada
- Department of Medical Sciences, University of Calgary, Calgary, AB, T2N 4Z6, Canada
| | - Noushin Nabavi
- Department of Urologic Sciences and Vancouver Prostate Centre, University of British Columbia, Vancouver, BC, V6H3Z6, Canada
| | - Amir Reza Aref
- Belfer Center for Applied Cancer Science, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
- Department of Translational Sciences, Xsphera Biosciences Inc. Boston, Boston, MA, USA
| | - Alaa A A Aljabali
- Faculty of Pharmacy, Department of Pharmaceutics and Pharmaceutical Technology, Yarmouk University, Irbid, Jordan
| | - Murtaza Tambuwala
- Lincoln Medical School, University of Lincoln, Brayford Pool Campus, Lincoln, LN6 7TS, UK.
- College of Pharmacy, Ras Al Khaimah Medical and Health Sciences University, Ras Al Khaimah, United Arab Emirates.
| | - Minglin Zhu
- Department of Cardiovascular Surgery, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China.
- Hubei Provincial Engineering Research Center of Minimally Invasive Cardiovascular Surgery, Wuhan, Hubei, 430071, China.
| |
Collapse
|
7
|
Li H, Cai L, Pan Q, Jiang X, Zhao J, Xiang T, Tang Y, Wang Q, He J, Weng D, Zhang Y, Liu Z, Xia J. N 6-methyladenosine-modified VGLL1 promotes ovarian cancer metastasis through high-mobility group AT-hook 1/Wnt/β-catenin signaling. iScience 2024; 27:109245. [PMID: 38439973 PMCID: PMC10910247 DOI: 10.1016/j.isci.2024.109245] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Revised: 11/30/2023] [Accepted: 02/13/2024] [Indexed: 03/06/2024] Open
Abstract
The main causes of death in patients with ovarian cancer (OC) are invasive lesions and the spread of metastasis. The present study aimed to explore the mechanisms that might promote OC metastasis. Here, we identified that VGLL1 expression was remarkably increased in metastatic OC samples. The role of VGLL1 in OC metastasis and tumor growth was examined by cell function assays and mouse models. Mechanistically level, METTL3-mediated N6-methyladenosine (m6A) modification contributed to VGLL1 upregulation in an IGF2BP2 recognition-dependent manner. Furthermore, VGLL1 directly interacts with TEAD4 and co-transcriptionally activates HMGA1. HMGA1 further activates Wnt/β-catenin signaling to enhance OC metastasis by promoting the epithelial-mesenchyme transition traits. Rescue assays indicated that the upregulation of HMGA1 was essential for VGLL1-induced metastasis. Collectively, these findings showed that the m6A-induced VGLL1/HMGA1/β-catenin axis might play a vital role in OC metastasis and tumor growth. VGLL1 might serve as a prognostic marker and therapeutic target against the metastasis of OC.
Collapse
Affiliation(s)
- Han Li
- Sun Yat-sen University Cancer Center; State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou 510060, China
- Department of Gynecology, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, China
| | - Liming Cai
- International Institute for Translational Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou 510006, Guangdong, China
| | - Qiuzhong Pan
- Sun Yat-sen University Cancer Center; State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou 510060, China
| | - Xingyu Jiang
- Sun Yat-sen University Cancer Center; State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou 510060, China
| | - Jingjing Zhao
- Sun Yat-sen University Cancer Center; State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou 510060, China
| | - Tong Xiang
- Sun Yat-sen University Cancer Center; State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou 510060, China
| | - Yan Tang
- Sun Yat-sen University Cancer Center; State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou 510060, China
| | - Qijing Wang
- Sun Yat-sen University Cancer Center; State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou 510060, China
| | - Jia He
- Sun Yat-sen University Cancer Center; State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou 510060, China
| | - Desheng Weng
- Sun Yat-sen University Cancer Center; State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou 510060, China
| | - Yanna Zhang
- Sun Yat-sen University Cancer Center; State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou 510060, China
| | - Zhongqiu Liu
- International Institute for Translational Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou 510006, Guangdong, China
| | - Jianchuan Xia
- Sun Yat-sen University Cancer Center; State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou 510060, China
| |
Collapse
|
8
|
Duan M, Liu H, Xu S, Yang Z, Zhang F, Wang G, Wang Y, Zhao S, Jiang X. IGF2BPs as novel m 6A readers: Diverse roles in regulating cancer cell biological functions, hypoxia adaptation, metabolism, and immunosuppressive tumor microenvironment. Genes Dis 2024; 11:890-920. [PMID: 37692485 PMCID: PMC10491980 DOI: 10.1016/j.gendis.2023.06.017] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Revised: 03/24/2023] [Accepted: 06/14/2023] [Indexed: 09/12/2023] Open
Abstract
m6A methylation is the most frequent modification of mRNA in eukaryotes and plays a crucial role in cancer progression by regulating biological functions. Insulin-like growth factor 2 mRNA-binding proteins (IGF2BP) are newly identified m6A 'readers'. They belong to a family of RNA-binding proteins, which bind to the m6A sites on different RNA sequences and stabilize them to promote cancer progression. In this review, we summarize the mechanisms by which different upstream factors regulate IGF2BP in cancer. The current literature analyzed here reveals that the IGF2BP family proteins promote cancer cell proliferation, survival, and chemoresistance, inhibit apoptosis, and are also associated with cancer glycolysis, angiogenesis, and the immune response in the tumor microenvironment. Therefore, with the discovery of their role as 'readers' of m6A and the characteristic re-expression of IGF2BPs in cancers, it is important to elucidate their mechanism of action in the immunosuppressive tumor microenvironment. We also describe in detail the regulatory and interaction network of the IGF2BP family in downstream target RNAs and discuss their potential clinical applications as diagnostic and prognostic markers, as well as recent advances in IGF2BP biology and associated therapeutic value.
Collapse
Affiliation(s)
- Meiqi Duan
- Department of General Surgery, The Fourth Affiliated Hospital of China Medical University, Shenyang, Liaoning 110032, China
| | - Haiyang Liu
- Department of General Surgery, The Fourth Affiliated Hospital of China Medical University, Shenyang, Liaoning 110032, China
| | - Shasha Xu
- Department of Gastroendoscopy, The Fourth Affiliated Hospital of China Medical University, Shenyang, Liaoning 110032, China
| | - Zhi Yang
- Department of General Surgery, The Fourth Affiliated Hospital of China Medical University, Shenyang, Liaoning 110032, China
| | - Fusheng Zhang
- Department of General Surgery, The Fourth Affiliated Hospital of China Medical University, Shenyang, Liaoning 110032, China
| | - Guang Wang
- Department of General Surgery, The Fourth Affiliated Hospital of China Medical University, Shenyang, Liaoning 110032, China
| | - Yutian Wang
- Department of General Surgery, The Fourth Affiliated Hospital of China Medical University, Shenyang, Liaoning 110032, China
| | - Shan Zhao
- Department of Rheumatology and Immunology, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning 110002, China
| | - Xiaofeng Jiang
- Department of General Surgery, The Fourth Affiliated Hospital of China Medical University, Shenyang, Liaoning 110032, China
| |
Collapse
|
9
|
Yang R, Wan J, Ma L, Zhou F, Yang Z, Li Z, Zhang M, Ming L. TMEM44-AS1 promotes esophageal squamous cell carcinoma progression by regulating the IGF2BP2-GPX4 axis in modulating ferroptosis. Cell Death Discov 2023; 9:431. [PMID: 38040698 PMCID: PMC10692126 DOI: 10.1038/s41420-023-01727-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Revised: 11/07/2023] [Accepted: 11/16/2023] [Indexed: 12/03/2023] Open
Abstract
The long non-coding RNA (lncRNA) TMEM44-AS1 is a novel lncRNA whose pro-carcinogenic role in gastric cancer and glioma has been demonstrated. However, its function in esophageal squamous cell carcinoma (ESCC) is unknown. In this study, we identified that TMEM44-AS1 was highly expressed in ESCC tissues and cells. Functionally, TMEM44-AS1 promoted ESCC cell proliferation, invasion and metastasis in vitro and in vivo. TMEM44-AS1 inhibited ferroptosis in ESCC cells, and ferroptosis levels were significantly increased after knockdown of TMEM44-AS1. Mechanistically, TMEM44-AS1 was positively correlated with GPX4 expression, and TMEM44-AS1 could bind to the RNA-binding protein IGF2BP2 to enhance the stability of GPX4 mRNA, thereby affecting ferroptosis and regulating the malignant progression of ESCC. In summary, this study reveals the TMEM44-AS1-IGF2BP2-GPX4 axis could influence cancer progression in ESCC. TMEM44-AS1 can be used as a potential treatment target against ESCC.
Collapse
Affiliation(s)
- Ruotong Yang
- Department of Clinical Laboratory, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Key Clinical Laboratory of Henan province, Zhengzhou, China
| | - Junhu Wan
- Department of Clinical Laboratory, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Key Clinical Laboratory of Henan province, Zhengzhou, China
| | - Liwei Ma
- Department of Clinical Laboratory, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Key Clinical Laboratory of Henan province, Zhengzhou, China
| | - Fuyou Zhou
- Thoracic Department, Anyang Tumor Hospital, Henan Key Medical Laboratory of Precise Prevention and Treatment of Esophageal Cancer, Anyang, China
| | - Zhengwu Yang
- Department of Clinical Laboratory, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Key Clinical Laboratory of Henan province, Zhengzhou, China
| | - Zhuofang Li
- Department of Clinical Laboratory, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Key Clinical Laboratory of Henan province, Zhengzhou, China
| | - Mingyuan Zhang
- Department of Clinical Laboratory, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Key Clinical Laboratory of Henan province, Zhengzhou, China
| | - Liang Ming
- Department of Clinical Laboratory, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.
- Key Clinical Laboratory of Henan province, Zhengzhou, China.
| |
Collapse
|
10
|
Xiong L, Tan J, Zhang R, Long Q, Xiong R, Liu Y, Liu Y, Tang J, Li Y, Feng G, Song G, Liu K. LINC01305 recruits basonuclin 1 to act on G-protein pathway suppressor 1 to promote esophageal squamous cell carcinoma. Cancer Sci 2023; 114:4314-4328. [PMID: 37705202 PMCID: PMC10637064 DOI: 10.1111/cas.15963] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Revised: 08/20/2023] [Accepted: 08/27/2023] [Indexed: 09/15/2023] Open
Abstract
EsophageaL squamous cell carcinoma (ESCC) is one of the most common and lethal tumors, however, its underlying molecular mechanisms are not completely understood and new therapeutic targets are needed. Here, we found that the transcription factor basonuclin 1 (BNC1) was significantly upregulated and closely related to the differentiation and metastasis of ESCC. Furthermore, BNC1, LINC01305, and G-protein pathway suppressor 1 (GPS1) had significant oncogenic roles in ESCC. In addition, in vivo experiments showed that knockdown of BNC1 indeed significantly inhibited the proliferation and metastasis of ESCC. We also revealed the molecular mechanism by which LINC01305 recruits BNC1 to the promoter of GPS1, and then GPS1 could mediate the JNK signaling pathway to promote the proliferation and metastases of ESCC. Taken together, we discovered the novel molecular mechanism by which LINC01305/BNC1 upregulates GPS1 expression to promote the development of ESCC, providing a new therapeutic target for ESCC.
Collapse
Affiliation(s)
- Li Xiong
- Institute of Tissue Engineering and Stem Cells, Nanchong Central Hospital, The Second Clinical Medical CollegeNorth Sichuan Medical CollegeNanchongChina
- Department of Laboratory MedicineNorth Sichuan Medical CollegeNanchongChina
| | - Jinsong Tan
- Institute of Tissue Engineering and Stem Cells, Nanchong Central Hospital, The Second Clinical Medical CollegeNorth Sichuan Medical CollegeNanchongChina
- Institute of Basic Medicine and Forensic MedicineNorth Sichuan Medical CollegeNanchongChina
| | - Ruolan Zhang
- Institute of Tissue Engineering and Stem Cells, Nanchong Central Hospital, The Second Clinical Medical CollegeNorth Sichuan Medical CollegeNanchongChina
- Department of Laboratory MedicineNorth Sichuan Medical CollegeNanchongChina
| | - Qiongxian Long
- Institute of Tissue Engineering and Stem Cells, Nanchong Central Hospital, The Second Clinical Medical CollegeNorth Sichuan Medical CollegeNanchongChina
| | - Rong Xiong
- Institute of Tissue Engineering and Stem Cells, Nanchong Central Hospital, The Second Clinical Medical CollegeNorth Sichuan Medical CollegeNanchongChina
- Department of Laboratory MedicineNorth Sichuan Medical CollegeNanchongChina
| | - Yanqun Liu
- Institute of Tissue Engineering and Stem Cells, Nanchong Central Hospital, The Second Clinical Medical CollegeNorth Sichuan Medical CollegeNanchongChina
| | - Yun Liu
- Institute of Basic Medicine and Forensic MedicineNorth Sichuan Medical CollegeNanchongChina
| | - Jiancai Tang
- Institute of Basic Medicine and Forensic MedicineNorth Sichuan Medical CollegeNanchongChina
| | - Yan Li
- Department of Anatomy and Physiology, College of Basic Medical Science, Songjiang Research Institute and Songjiang HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Gang Feng
- Institute of Tissue Engineering and Stem Cells, Nanchong Central Hospital, The Second Clinical Medical CollegeNorth Sichuan Medical CollegeNanchongChina
| | - Guiqin Song
- Institute of Basic Medicine and Forensic MedicineNorth Sichuan Medical CollegeNanchongChina
| | - Kang Liu
- Institute of Tissue Engineering and Stem Cells, Nanchong Central Hospital, The Second Clinical Medical CollegeNorth Sichuan Medical CollegeNanchongChina
- Department of Laboratory MedicineNorth Sichuan Medical CollegeNanchongChina
| |
Collapse
|
11
|
Liu X, Chen J, Chen W, Xu Y, Shen Y, Xu X. Targeting IGF2BP3 in Cancer. Int J Mol Sci 2023; 24:ijms24119423. [PMID: 37298373 DOI: 10.3390/ijms24119423] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Revised: 05/20/2023] [Accepted: 05/23/2023] [Indexed: 06/12/2023] Open
Abstract
RNA-binding proteins (RBPs) can regulate multiple pathways by binding to RNAs, playing a variety of functions, such as localization, stability, and immunity. In recent years, with the development of technology, researchers have discovered that RBPs play a key role in the N6-methyladenosine (m6A) modification process. M6A methylation is the most abundant form of RNA modification in eukaryotes, which is defined as methylation on the sixth N atom of adenine in RNA. Insulin-like growth factor 2 mRNA-binding protein 3 (IGF2BP3) is one of the components of m6A binding proteins, which plays an important role in decoding m6A marks and performing various biological functions. IGF2BP3 is abnormally expressed in many human cancers, often associated with poor prognosis. Here, we summarize the physiological role of IGF2BP3 in organisms and describe its role and mechanism in tumors. These data suggest that IGF2BP3 may be a valuable therapeutic target and prognostic marker in the future.
Collapse
Affiliation(s)
- Xin Liu
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Jiayu Chen
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Wenliang Chen
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Yangtao Xu
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Yang Shen
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Ximing Xu
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan 430060, China
| |
Collapse
|
12
|
Zheng X, Wu J, Song L, Huang B. ACSM3 suppresses proliferation and induces apoptosis and cell cycle arrest in acute myeloid leukemia cells via the regulation of IGF2BP2. Exp Ther Med 2023; 25:177. [PMID: 37006876 PMCID: PMC10061044 DOI: 10.3892/etm.2023.11876] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Accepted: 10/07/2022] [Indexed: 03/11/2023] Open
Abstract
Acyl-CoA medium-chain synthetase-3 (ACSM3) has been reported to be involved in the malignant progression of multiple types of human cancer. Nevertheless, the role of ACSM3 in acute myeloid leukemia (AML) and its exact mechanism of action are as yet undefined. In the present study, the expression levels of ACSM3 and IGF2 mRNA-binding protein 2 (IGF2BP2) were evaluated using the Gene Expression Profiling Interactive Analysis database and AML cells. The Cell Counting Kit-8 assay and 5-ethynyl-2'-deoxyuridine staining were employed for the estimation of the cell proliferative activity. Induction of apoptosis and the assessment of the cell cycle were measured using flow cytometry and western blotting, respectively. The interaction of ACSM3 with IGF2BP2 was confirmed using an RNA immunoprecipitation assay. mRNA stabilization of ACSM3 following actinomycin D treatment was evaluated using reverse transcription-quantitative PCR analysis. The data indicated that the expression levels of ACSM3 were significantly downregulated, whereas those of IGF2BP2 were upregulated in tissues and AML cells. Downregulation of ACSM3 expression was closely associated with poor overall survival of patients with AML. ACSM3 overexpression repressed cell proliferative activity and induced apoptosis and cell cycle arrest. IGF2BP2 downregulated ACSM3 expression by reducing the stability of ACSM3 mRNA. In addition, IGF2BP2 overexpression counteracted the effects of ACSM3 overexpression noted on proliferation, induction of apoptosis and cell cycle arrest of HL-60 cells. In conclusion, ACSM3 repressed the cell proliferative activity and facilitated induction of apoptosis and cell cycle arrest in AML cells by modulating the expression of IGF2BP2.
Collapse
Affiliation(s)
- Xin Zheng
- Department of Clinical Laboratory, Jianghan Oilfield General Hospital of Changjiang University, Qianjiang, Hubei 433124, P.R. China
| | - Jinjun Wu
- Department of Clinical Laboratory, Jianghan Oilfield General Hospital of Changjiang University, Qianjiang, Hubei 433124, P.R. China
| | - Linlan Song
- Department of Clinical Laboratory, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710004, P.R. China
| | - Bo Huang
- Department of Clinical Laboratory, The Affiliated Children Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710003, P.R. China
| |
Collapse
|
13
|
Yang Z, Wan J, Ma L, Li Z, Yang R, Yang H, Li J, Zhou F, Ming L. Long non-coding RNA HOXC-AS1 exerts its oncogenic effects in esophageal squamous cell carcinoma by interaction with IGF2BP2 to stabilize SIRT1 expression. J Clin Lab Anal 2022; 37:e24801. [PMID: 36510377 PMCID: PMC9833966 DOI: 10.1002/jcla.24801] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Revised: 10/28/2022] [Accepted: 11/01/2022] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Long non-coding RNA HOXC cluster antisense RNA 1 (HOXC-AS1) is a novel lncRNA whose cancer-promoting effect in gastric cancer and nasopharyngeal carcinoma has already been demonstrated. However, its functions in esophageal squamous cell carcinoma (ESCC) remains unknown. LncRNAs can interact with RNA-binding proteins (RBPs) and affect gene expression levels through post-transcriptional regulation. Insulin-like growth factor 2 mRNA-binding protein 2 (IGF2BP2) is a widely studied RBP, and sirtuin 1 also known as SIRT1 has been reported to be involved in cancer progression. METHODS Establishment of in vivo models, HE and immunohistochemistry staining verified the oncogenic effect of HOXC-AS1. The interaction relationship between HOXC-AS1, IGF2BP2 and SIRT1 was verified by RNA pulldown and RNA immunoprecipitation (RIP) assay. Relative expression and stability changes of genes were detected by qPCR and actinomycin D experiments. Finally, the effect of HOXC-AS1-IGF2BP2-SIRT1 axis on ESCC was verified by rescue experiments. RESULTS HOXC-AS1 is highly expressed in ESCC cells and plays oncogenic effects in vivo. qPCR showed the positive relationship between HOXC-AS1 and SIRT1 following HOXC-AS1 knockdown or overexpression. RNA-pulldown, mass spectrometry and RIP assay demonstrated that IGF2BP2 is an RBP downstream of HOXC-AS1. Then, RIP and qPCR showed that IGF2BP2 could bind to SIRT1 mRNA and knockdown IGF2BP2 resulted in decreased SIRT1 mRNA level. Finally, a series of rescue assay showed that the HOXC-AS1-IGF2BP2-SIRT1 axis can affect the function of ESCC. CONCLUSION LncRNA HOXC-AS1 acts as an oncogenic role in ESCC, which impacts ESCC progression by interaction with IGF2BP2 to stabilize SIRT1 expression.
Collapse
Affiliation(s)
- Zhengwu Yang
- Department of Clinical LaboratoryThe First Affiliated Hospital of Zhengzhou University, and the Key Clinical Laboratory of Henan ProvinceHenanChina
| | - Junhu Wan
- Department of Clinical LaboratoryThe First Affiliated Hospital of Zhengzhou University, and the Key Clinical Laboratory of Henan ProvinceHenanChina
| | - Liwei Ma
- Department of Clinical LaboratoryThe First Affiliated Hospital of Zhengzhou University, and the Key Clinical Laboratory of Henan ProvinceHenanChina
| | - Zhuofang Li
- Department of Clinical LaboratoryThe First Affiliated Hospital of Zhengzhou University, and the Key Clinical Laboratory of Henan ProvinceHenanChina
| | - Ruotong Yang
- Department of Clinical LaboratoryThe First Affiliated Hospital of Zhengzhou University, and the Key Clinical Laboratory of Henan ProvinceHenanChina
| | - Haijun Yang
- Henan Key Medical Laboratory of Precise Prevention and Treatment of Esophageal CancerAnyangChina
| | - Junkuo Li
- Department of PathologyAnyang Cancer Hospital, The Forth Affiliated Hospital of Henan University of Science and TechnologyHenanChina
| | - Fuyou Zhou
- Thoracic DepartmentAnyang Tumor Hospital, Henan Key Medical Laboratory of Precise Prevention and Treatment of Esophageal CancerAnyangChina
| | - Liang Ming
- Department of Clinical LaboratoryThe First Affiliated Hospital of Zhengzhou University, and the Key Clinical Laboratory of Henan ProvinceHenanChina
| |
Collapse
|
14
|
Liu A, Chen Y, Zhong D, Wang C, Yu M, Liu C, Yang Z, Chen W, Yin K. CircRNA AFF4 induced by KDM1A promotes osteogenic differentiation through FNDC5/Irisin pathway. Mol Med 2022; 28:134. [PMID: 36401176 PMCID: PMC9673395 DOI: 10.1186/s10020-022-00557-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Revised: 10/08/2022] [Accepted: 10/13/2022] [Indexed: 11/19/2022] Open
Abstract
Background Circular RNA (circ) AFF4 was documented to regulate osteogenesis but the underlying mechanism remains to be elucidated. The preliminary study showed that circ_AFF4 may promote osteogenesis via FNDC5/Irisin. Furthermore, the online prediction tool indicated the interaction of circ_AFF4, insulin-like growth factor-2 mRNA-binding protein 3 (IGF2BP3), FNDC5 and lysine (K)-specific demethylase 1 A (KDM1A). Therefore, this study aims to elucidate the relationships of KDM1A, circ_AFF4, IGF2BP3 and FNDC5/Irisin during osteogenesis. Methods The alkaline phosphatase (ALP) activities and osteogenic-related factors were determined using ALP and alizarin red S (ARS) staining, real-time quantitative PCR(RT-qPCR) and western blot. Immunoprecipitation (RIP), pull-down assay and fluorescence in situ hybridization (FISH) were used to examine the interactions among circ_AFF4/IGF2BP3/FNDC5. A mouse in vivo model was utilized to further confirm the regulatory effect on bone formation. Results Circ_AFF4 and KDM1A expression levels were increased during osteoinduction of BM-MSCs. Knockdown of circ_AFF4 and KDM1A significantly suppressed BM-MSC osteogenesis. We also proved that KDM1A directly bound to circ_AFF4 and FNDC5 promoter and induced circ_AFF4 and FNDC5 expression. Furthermore, circ_AFF4 enhanced the stability of FNDC5 by generating a circ_AFF4, IGF2BP3 and FNDC5 RNA-protein complex, and thereby induced Irisin and osteogenesis. The in vitro data was confirmed with in vivo model. Conclusion These findings elucidate that KDM1A induces circ_AFF4, which promotes promote osteogenesis via IGF2BP3. This study indicates that circ_AFF4 may potentially represent a critical therapeutic target for the diseases. Supplementary Information The online version contains supplementary material available at 10.1186/s10020-022-00557-7.
Collapse
Affiliation(s)
- Ansong Liu
- grid.412017.10000 0001 0266 8918The First Affiliated Hospital, Department of Orthopedics, Hengyang Medical School, University of South China, No. 69 Chuanshan Road, Hengyang, Hunan 421001 China
| | - Yong Chen
- grid.412017.10000 0001 0266 8918The First Affiliated Hospital, Department of Spine, Hengyang Medical School, University of South China, No. 69 Chuanshan Road, Hengyang, Hunan 421001 China
| | - Da Zhong
- grid.216417.70000 0001 0379 7164Department of Orthopedics, Xiangya Hospital, Central South University, No.87 Xiangya Road, 410008 Changsha, Hunan China
| | - Chenggong Wang
- grid.216417.70000 0001 0379 7164Department of Orthopedics, Xiangya Hospital, Central South University, No.87 Xiangya Road, 410008 Changsha, Hunan China
| | - Mi Yu
- grid.412017.10000 0001 0266 8918Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
| | - Chao Liu
- grid.412017.10000 0001 0266 8918The First Affiliated Hospital, Department of Orthopedics, Hengyang Medical School, University of South China, No. 69 Chuanshan Road, Hengyang, Hunan 421001 China
| | - Zhijun Yang
- grid.412017.10000 0001 0266 8918The First Affiliated Hospital, Department of Orthopedics, Hengyang Medical School, University of South China, No. 69 Chuanshan Road, Hengyang, Hunan 421001 China
| | - Wenkang Chen
- grid.412017.10000 0001 0266 8918The First Affiliated Hospital, Department of Orthopedics, Hengyang Medical School, University of South China, No. 69 Chuanshan Road, Hengyang, Hunan 421001 China
| | - Ke Yin
- grid.412017.10000 0001 0266 8918The First Affiliated Hospital, Department of Orthopedics, Hengyang Medical School, University of South China, No. 69 Chuanshan Road, Hengyang, Hunan 421001 China
| |
Collapse
|
15
|
Qiu L, Tao A, Liu F, Ge X, Li C. Potential prognostic value of a eight ferroptosis-related lncRNAs model and the correlative immune activity in oral squamous cell carcinoma. BMC Genom Data 2022; 23:80. [PMID: 36384476 PMCID: PMC9667687 DOI: 10.1186/s12863-022-01097-z] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Accepted: 11/02/2022] [Indexed: 11/17/2022] Open
Abstract
Background To investigate the prognostic value of ferroptosis-related long noncoding RNAs (lncRNAs) in oral squamous cell carcinoma (OSCC) and to construct a prognostic risk and immune activity model. Methods We obtained clinical and RNA-seq information on OSCC patient data in The Cancer Genome Atlas (TCGA) Genome Data Sharing (GDC) portal. Through a combination of a differential analysis, Pearson correlation analysis and Cox regression analysis, ferroptosis-related lncRNAs were identified, and a prognostic model was established based on these ferroptosis-related lncRNAs. The accuracy of the model was evaluated via analyses based on survival curves, receiver operating characteristic (ROC) curves, and clinical decision curve analysis (DCA). Univariate Cox and multivariate Cox regression analyses were performed to evaluate independent prognostic factors. Then, the infiltration and functional enrichment of immune cells in high- and low-risk groups were compared. Finally, certain small-molecule drugs that potentially target OSCC were predicted via use of the L1000FWD database. Results The prognostic model included 8 ferroptosis-related lncRNAs (FIRRE, LINC01305, AC099850.3, AL512274.1, AC090246.1, MIAT, AC079921.2 and LINC00524). The area under the ROC curve (AUC) was 0.726. The DCA revealed that the risk score based on the prognostic model was a better prognostic indicator than other clinical indicators. The multivariate Cox regression analysis showed that the risk score was an independent prognostic factor for OSCC. There were differences in immune cell infiltration, immune functions, m6A-related gene expression levels, and signal pathway enrichment between the high- and low-risk groups. Subsequently, several small-molecule drugs were predicted for use against differentially expressed ferroptosis-related genes in OSCC. Conclusions We constructed a new prognostic model of OSCC based on ferroptosis-related lncRNAs. The model is valuable for prognostic prediction and immune evaluation, laying a foundation for the study of ferroptosis-related lncRNAs in OSCC. Supplementary Information The online version contains supplementary material available at 10.1186/s12863-022-01097-z.
Collapse
|
16
|
Li Y, Niu C, Wang N, Huang X, Cao S, Cui S, Chen T, Huo X, Zhou R. The Role of m 6A Modification and m 6A Regulators in Esophageal Cancer. Cancers (Basel) 2022; 14:5139. [PMID: 36291923 PMCID: PMC9600289 DOI: 10.3390/cancers14205139] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Revised: 10/15/2022] [Accepted: 10/18/2022] [Indexed: 03/19/2025] Open
Abstract
N6-methyladenosine (m6A) modification, the most prevalent RNA modification, is involved in all aspects of RNA metabolism, including RNA processing, nuclear export, stability, translation and degradation. Therefore, m6A modification can participate in various physiological functions, such as tissue development, heat shock response, DNA damage response, circadian clock control and even in carcinogenesis through regulating the expression or structure of the gene. The deposition, removal and recognition of m6A are carried out by methyltransferases, demethylases and m6A RNA binding proteins, respectively. Aberrant m6A modification and the dysregulation of m6A regulators play critical roles in the occurrence and development of various cancers. The pathogenesis of esophageal cancer (ESCA) remains unclear and the five-year survival rate of advanced ESCA patients is still dismal. Here, we systematically reviewed the recent studies of m6A modification and m6A regulators in ESCA and comprehensively analyzed the role and possible mechanism of m6A modification and m6A regulators in the occurrence, progression, remedy and prognosis of ESCA. Defining the effect of m6A modification and m6A regulators in ESCA might be helpful for determining the pathogenesis of ESCA and providing some ideas for an early diagnosis, individualized treatment and improved prognosis of ESCA patients.
Collapse
Affiliation(s)
- Yuekao Li
- Department of Computed Tomography, The Fourth Hospital of Hebei Medical University, Shijiazhuang 050011, China
| | - Chaoxu Niu
- Department of Surgery, Shijiazhuang Ping’an Hospital, Shijiazhuang 050021, China
| | - Na Wang
- Hebei Provincial Cancer Institute, The Fourth Hospital of Hebei Medical University, Shijiazhuang 050011, China
| | - Xi Huang
- Hebei Provincial Cancer Institute, The Fourth Hospital of Hebei Medical University, Shijiazhuang 050011, China
| | - Shiru Cao
- Hebei Provincial Cancer Institute, The Fourth Hospital of Hebei Medical University, Shijiazhuang 050011, China
| | - Saijin Cui
- Hebei Provincial Cancer Institute, The Fourth Hospital of Hebei Medical University, Shijiazhuang 050011, China
| | - Tianyu Chen
- Hebei Provincial Cancer Institute, The Fourth Hospital of Hebei Medical University, Shijiazhuang 050011, China
| | - Xiangran Huo
- Hebei Provincial Cancer Institute, The Fourth Hospital of Hebei Medical University, Shijiazhuang 050011, China
| | - Rongmiao Zhou
- Hebei Provincial Cancer Institute, The Fourth Hospital of Hebei Medical University, Shijiazhuang 050011, China
| |
Collapse
|
17
|
Wu K, Chang F, Li W, Su T, Lei D. Role of IGF2BPs in head and neck squamous cell carcinoma. Front Oncol 2022; 12:1003808. [PMID: 36237306 PMCID: PMC9552850 DOI: 10.3389/fonc.2022.1003808] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Accepted: 09/07/2022] [Indexed: 12/24/2022] Open
Abstract
IGF2BPs belongs to a family of conserved RNA-bound oncoembryonic proteins that play a crucial part in various aspects of cell function, such as cell migration, morphology, metabolism, proliferation and differentiation. Recent studies have shown that IGF2BPs play a role as a member of m6A reader. m6A is the most abundant modification in RNA epigenetics, which is closely related to a family of RNA-binding proteins. These proteins are fell into three categories—writers, readers and erasers. In the present study, IGF2BPs play an important role in tumor metabolism, especially in head and neck squamous cell carcinoma (HNSCC) metabolism. In this paper, the basic structure of IGF2BPs, its role in the development of HNSCC, molecular mechanism, research progress and research prospect of IGF2BPs in HNSCC are reviewed, which will providing new ideas for further study of IGF2BPs.
Collapse
Affiliation(s)
- Kainan Wu
- Key Laboratory of Otolaryngology, NHFPC (Shandong University), Shandong, China
- Department of Otorhinolaryngology, Qilu Hospital, Shandong University, Shandong, China
| | - Fen Chang
- Key Laboratory of Otolaryngology, NHFPC (Shandong University), Shandong, China
- Department of Otorhinolaryngology, Qilu Hospital, Shandong University, Shandong, China
| | - Wenming Li
- Key Laboratory of Otolaryngology, NHFPC (Shandong University), Shandong, China
- Department of Otorhinolaryngology, Qilu Hospital, Shandong University, Shandong, China
| | - Tongdong Su
- Key Laboratory of Otolaryngology, NHFPC (Shandong University), Shandong, China
- Department of Otorhinolaryngology, Qilu Hospital, Shandong University, Shandong, China
| | - Dapeng Lei
- Key Laboratory of Otolaryngology, NHFPC (Shandong University), Shandong, China
- Department of Otorhinolaryngology, Qilu Hospital, Shandong University, Shandong, China
- *Correspondence: Dapeng Lei,
| |
Collapse
|
18
|
Liu X, Zhou L, Gao M, Dong S, Hu Y, Hu C. Signature of seven cuproptosis-related lncRNAs as a novel biomarker to predict prognosis and therapeutic response in cervical cancer. Front Genet 2022; 13:989646. [PMID: 36204323 PMCID: PMC9530991 DOI: 10.3389/fgene.2022.989646] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Accepted: 09/05/2022] [Indexed: 11/13/2022] Open
Abstract
Background: Given the high incidence and high mortality of cervical cancer (CC) among women in developing countries, identifying reliable biomarkers for the prediction of prognosis and therapeutic response is crucial. We constructed a prognostic signature of cuproptosis-related long non-coding RNAs (lncRNAs) as a reference for individualized clinical treatment. Methods: A total of seven cuproptosis-related lncRNAs closely related to the prognosis of patients with CC were identified and used to construct a prognostic signature via least absolute shrinkage and selection operator regression analysis in the training set. The predictive performance of the signature was evaluated by Kaplan-Meier (K-M) analysis, receiver operating characteristic (ROC) analysis, and univariate and multivariate Cox analyses. Functional enrichment analysis and single-sample gene set enrichment analysis were conducted to explore the potential mechanisms of the prognostic signature, and a lncRNA-microRNA-mRNA network was created to investigate the underlying regulatory relationships between lncRNAs and cuproptosis in CC. The associations between the prognostic signature and response to immunotherapy and targeted therapy were also assessed. Finally, the prognostic value of the signature was validated using the CC tissues with clinical information in my own center. Results: A prognostic signature was developed based on seven cuproptosis-related lncRNAs, including five protective factors (AL441992.1, LINC01305, AL354833.2, CNNM3-DT, and SCAT2) and two risk factors (AL354733.3 and AC009902.2). The ROC curves confirmed the superior predictive performance of the signature compared with conventional clinicopathological characteristics in CC. The ion transport-related molecular function and various immune-related biological processes differed significantly between the two risk groups according to functional enrichment analysis. Furthermore, we discovered that individuals in the high-risk group were more likely to respond to immunotherapy and targeted therapies including trametinib and cetuximab than those in the low-risk group. Finally, CC tissues with clinical data from my own center further verify the robustness of the seven-lncRNA risk signature. Conclusion: We generated a cuproptosis-related lncRNA risk signature that could be used to predict prognosis of CC patients. Moreover, the signature could be used to predict response to immunotherapy and chemotherapy and thus could assist clinicians in making personalized treatment plans for CC patients.
Collapse
Affiliation(s)
- Xinyu Liu
- Department of Obstetrics and Gynecology, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, China
- NHC Key Laboratory of Molecular Probes and Targeted Diagnosis and Therapy, Harbin Medical University, Harbin, China
| | - Lei Zhou
- Department of Orthopedics, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Minghui Gao
- Department of Obstetrics and Gynecology, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, China
- NHC Key Laboratory of Molecular Probes and Targeted Diagnosis and Therapy, Harbin Medical University, Harbin, China
| | - Shuhong Dong
- Department of Obstetrics and Gynecology, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, China
- NHC Key Laboratory of Molecular Probes and Targeted Diagnosis and Therapy, Harbin Medical University, Harbin, China
| | - Yanan Hu
- Department of Obstetrics and Gynecology, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, China
- NHC Key Laboratory of Molecular Probes and Targeted Diagnosis and Therapy, Harbin Medical University, Harbin, China
| | - Chunjie Hu
- Department of Obstetrics and Gynecology, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, China
- NHC Key Laboratory of Molecular Probes and Targeted Diagnosis and Therapy, Harbin Medical University, Harbin, China
| |
Collapse
|
19
|
Liu XS, Liu C, Zeng J, Zeng DB, Chen YJ, Tan F, Gao Y, Liu XY, Zhang Y, Zhang YH, Pei ZJ. Nucleophosmin 1 is a prognostic marker of gastrointestinal cancer and is associated with m6A and cuproptosis. Front Pharmacol 2022; 13:1010879. [PMID: 36188614 PMCID: PMC9515486 DOI: 10.3389/fphar.2022.1010879] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Accepted: 08/29/2022] [Indexed: 11/30/2022] Open
Abstract
Background: NPM1 is highly expressed in a variety of solid tumors and promotes tumor development. However, there are few comprehensive studies on NPM1 analysis in gastrointestinal cancer. Methods: We used bioinformatics tools to study the expression difference of NPM1 between gastrointestinal cancer and control group, and analyzed the relationship between its expression level and the diagnosis, prognosis, functional signaling pathway, immune infiltration, m6A and cuproptosis related genes of gastrointestinal cancer. At the same time, the expression difference of NPM1 between esophageal carcinoma (ESCA) samples and control samples was verified by in vitro experiments. Results: NPM1 was overexpressed in gastrointestinal cancer. In vitro experiments confirmed that the expression of NPM1 in ESCA samples was higher than that in normal samples. The expression of NPM1 has high accuracy in predicting the outcome of gastrointestinal cancer. The expression of NPM1 is closely related to the prognosis of multiple gastrointestinal cancers. Go and KEGG enrichment analysis showed that NPM1 co-expressed genes involved in a variety of biological functions. NPM1 expression is potentially associated with a variety of immune cell infiltration, m6A and cuproptosis related genes in gastrointestinal cancers. Conclusion: NPM1 can be used as a diagnostic and prognostic marker of gastrointestinal cancer, which is related to the immune cell infiltration and the regulation of m6A and cuproptosis.
Collapse
Affiliation(s)
- Xu-Sheng Liu
- Department of Nuclear Medicine, Taihe Hospital, Hubei University of Medicine, Shiyan, China
- *Correspondence: Xu-Sheng Liu, ; Zhi-Jun Pei,
| | - Chao Liu
- Medical Imaging Center, Taihe Hospital, Hubei University of Medicine, Shiyan, China
| | - Jing Zeng
- Department of Infection Control, Taihe Hospital, Hubei University of Medicine, Shiyan, China
| | - Dao-Bing Zeng
- Department of Nuclear Medicine, Taihe Hospital, Hubei University of Medicine, Shiyan, China
| | - Yi-Jia Chen
- Department of Nuclear Medicine, Taihe Hospital, Hubei University of Medicine, Shiyan, China
| | - Fan Tan
- Department of Nuclear Medicine, Taihe Hospital, Hubei University of Medicine, Shiyan, China
| | - Yan Gao
- Department of Nuclear Medicine, Taihe Hospital, Hubei University of Medicine, Shiyan, China
| | - Xiao-Yu Liu
- Department of Nuclear Medicine, Taihe Hospital, Hubei University of Medicine, Shiyan, China
| | - Yu Zhang
- Department of Nuclear Medicine, Taihe Hospital, Hubei University of Medicine, Shiyan, China
| | - Yao-Hua Zhang
- Department of Nuclear Medicine, Taihe Hospital, Hubei University of Medicine, Shiyan, China
| | - Zhi-Jun Pei
- Department of Nuclear Medicine, Taihe Hospital, Hubei University of Medicine, Shiyan, China
- Hubei Clinical Research Center for Precise Diagnosis and Treatment of Liver Cancer, Taihe Hospital, Hubei University of Medicine, Shiyan, China
- *Correspondence: Xu-Sheng Liu, ; Zhi-Jun Pei,
| |
Collapse
|
20
|
Fang Z, Mei W, Qu C, Lu J, Shang L, Cao F, Li F. Role of m6A writers, erasers and readers in cancer. Exp Hematol Oncol 2022; 11:45. [PMID: 35945641 PMCID: PMC9361621 DOI: 10.1186/s40164-022-00298-7] [Citation(s) in RCA: 133] [Impact Index Per Article: 44.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Accepted: 08/04/2022] [Indexed: 02/06/2023] Open
Abstract
The N(6)-methyladenosine (m6A) modification is the most pervasive modification of human RNAs. In recent years, an increasing number of studies have suggested that m6A likely plays important roles in cancers. Many studies have demonstrated that m6A is involved in the biological functions of cancer cells, such as proliferation, invasion, metastasis, and drug resistance. In addition, m6A is closely related to the prognosis of cancer patients. In this review, we highlight recent advances in understanding the function of m6A in various cancers. We emphasize the importance of m6A to cancer progression and look forward to describe future research directions.
Collapse
Affiliation(s)
- Zhen Fang
- Department of General Surgery, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Wentong Mei
- Department of General Surgery, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Chang Qu
- Department of General Surgery, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Jiongdi Lu
- Department of General Surgery, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Liang Shang
- Department of Gastrointestinal Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China.
| | - Feng Cao
- Department of General Surgery, Xuanwu Hospital, Capital Medical University, Beijing, China.
| | - Fei Li
- Department of General Surgery, Xuanwu Hospital, Capital Medical University, Beijing, China.
| |
Collapse
|
21
|
Chen J, Sun M, Huang L, Fang Y. The Long noncoding RNA LINC00200 Promotes the Malignant Progression of MYCN-Amplified Neuroblastoma via Binding to Insulin like growth factor 2 mRNA binding protein 3 (IGF2BP3) to Enhance the Stability of of Zic family member 2 (ZIC2) mRNA. Pathol Res Pract 2022; 237:154059. [DOI: 10.1016/j.prp.2022.154059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/09/2022] [Revised: 07/24/2022] [Accepted: 08/03/2022] [Indexed: 12/09/2022]
|
22
|
A Pleiotropic Role of Long Non-Coding RNAs in the Modulation of Wnt/β-Catenin and PI3K/Akt/mTOR Signaling Pathways in Esophageal Squamous Cell Carcinoma: Implication in Chemotherapeutic Drug Response. Curr Oncol 2022; 29:2326-2349. [PMID: 35448163 PMCID: PMC9031703 DOI: 10.3390/curroncol29040189] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Revised: 03/19/2022] [Accepted: 03/20/2022] [Indexed: 02/06/2023] Open
Abstract
Despite the availability of modern techniques for the treatment of esophageal squamous cell carcinoma (ESCC), tumor recurrence and metastasis are significant challenges in clinical management. Thus, ESCC possesses a poor prognosis and low five-year overall survival rate. Notably, the origin and recurrence of the cancer phenotype are under the control of complex cancer-related signaling pathways. In this review, we provide comprehensive knowledge about long non-coding RNAs (lncRNAs) related to Wnt/β-catenin and phosphatidylinositol-3-kinase (PI3K)/protein kinase B (Akt)/mammalian target of rapamycin (mTOR) signaling pathway in ESCC and its implications in hindering the efficacy of chemotherapeutic drugs. We observed that a pool of lncRNAs, such as HERES, TUG1, and UCA1, associated with ESCC, directly or indirectly targets various molecules of the Wnt/β-catenin pathway and facilitates the manifestation of multiple cancer phenotypes, including proliferation, metastasis, relapse, and resistance to anticancer treatment. Additionally, several lncRNAs, such as HCP5 and PTCSC1, modulate PI3K/Akt/mTOR pathways during the ESCC pathogenesis. Furthermore, a few lncRNAs, such as AFAP1-AS1 and LINC01014, block the efficiency of chemotherapeutic drugs, including cisplatin, 5-fluorouracil, paclitaxel, and gefitinib, used for ESCC treatment. Therefore, this review may help in designing a better therapeutic strategy for ESCC patients.
Collapse
|
23
|
Zhou L, Li H, Cai H, Liu W, Pan E, Yu D, He S. Upregulation of IGF2BP2 Promotes Oral Squamous Cell Carcinoma Progression That Is Related to Cell Proliferation, Metastasis and Tumor-Infiltrating Immune Cells. Front Oncol 2022; 12:809589. [PMID: 35299748 PMCID: PMC8921988 DOI: 10.3389/fonc.2022.809589] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Accepted: 01/31/2022] [Indexed: 12/21/2022] Open
Abstract
The strong invasive and metastatic abilities of oral squamous cell carcinoma (OSCC) cells in the early stage are the main reason for its poor prognosis. The early diagnosis and treatment of OSCC may reduce the metastasis rate and improve the survival rate. The aim of this study was to explore candidate biomarkers related to the prognosis and progression of OSCC. We performed weighted gene coexpression network analysis to identify key modules and genes associated with OSCC and intersected the differentially expressed genes (DEGs) in The Cancer Genome Atlas (TCGA)-OSCC and GSE30784 datasets. Next, we performed survival analysis and immunohistochemistry to screen and validate the hub gene insulin-like growth factor 2 (IGF2) mRNA binding protein 2 IGF2BP2. We also used TCGA pan-cancer data to verify that IGF2BP2 was expressed at high levels in a variety of cancers and was related to a poor prognosis in patients. Furthermore, we divided patients with OSCC into high and low expression groups based on the median expression level of IGF2BP2. Gene set enrichment analysis (GSEA) showed that IGF2BP2 led to a poor prognosis in OSCC by affecting cancer-related (epithelial-mesenchymal transition, glycolysis, cell cycle, etc.) and immune-related biological functions and pathways. Single-sample GSEA (ssGSEA), CIBERSORT, and xCell algorithms helped reveal that high IGF2BP2 expression was accompanied by a significant reduction in the immune score, stromal score, and microenvironment score and a decrease in the number of infiltrating CD8+ T cells in OSCC. In addition, silencing IGF2BP2 suppressed the proliferation, migration, and invasion of OSCC cells. In general, IGF2BP2 is a potential biomarker for the progression, immunotherapy response, and prognosis of OSCC.
Collapse
Affiliation(s)
- Lijie Zhou
- Department of Stomatology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Hongyu Li
- Department of Oral and Maxillofacial Surgery, Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, China
| | - Hongshi Cai
- Department of Oral and Maxillofacial Surgery, Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, China
| | - Wenhui Liu
- Department of Stomatology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Enjiu Pan
- Department of Stomatology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Dongsheng Yu
- Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, China.,Department of Oral Emergency, Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, China
| | - Shuai He
- Department of Stomatology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| |
Collapse
|