1
|
Qiu X, Gao Q, Wang J, Zhang Z, Tao L. The microbiota-m 6A-metabolism axis: Implications for therapeutic strategies in gastrointestinal cancers. Biochim Biophys Acta Rev Cancer 2025; 1880:189317. [PMID: 40222422 DOI: 10.1016/j.bbcan.2025.189317] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Revised: 04/06/2025] [Accepted: 04/06/2025] [Indexed: 04/15/2025]
Abstract
Gastrointestinal (GI) cancers remain a leading cause of cancer-related mortality worldwide, with metabolic reprogramming recognized as a central driver of tumor progression and therapeutic resistance. Among the key regulatory layers, N6-methyladenosine (m6A) RNA modification-mediated by methyltransferases (writers such as METTL3/14), RNA-binding proteins (readers like YTHDFs and IGF2BPs), and demethylases (erasers including FTO and ALKBH5), plays a pivotal role in controlling gene expression and metabolic flux in the tumor context. Concurrently, the gut microbiota profoundly influences GI tumorigenesis and immune evasion by modulating metabolite availability and remodeling the tumor microenvironment. Recent evidence has uncovered a bidirectional crosstalk between microbial metabolites and m6A methylation: microbiota-derived signals dynamically regulate m6A deposition on metabolic and immune transcripts, while m6A modifications, in turn, regulate the stability and translation of key mRNAs such as PD-L1 and FOXP3. This reciprocal interaction forms self-reinforcing epigenetic circuits that drive tumor plasticity, immune escape, and metabolic adaptation. In this review, we dissect the molecular underpinnings of the microbiota-m6A-metabolism axis in GI cancers and explore its potential to inform novel strategies in immunotherapy, metabolic intervention, and microbiome-guided precision oncology.
Collapse
Affiliation(s)
- Xiuxiu Qiu
- Department of Oncology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China
| | - Qi Gao
- Institute of Acupuncture and Moxibustion, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Jiahui Wang
- Cancer Institute, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China
| | - Zhanxia Zhang
- Cancer Institute, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China
| | - Li Tao
- Department of Oncology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China.
| |
Collapse
|
2
|
Li P, Chu D, Ding G, Qin D, Bu Y, Tian B. IGF2BP3 suppresses ferroptosis in lung adenocarcinoma by m6A-dependent regulation of TFAP2A to transcriptionally activate SLC7A11/GPX4. Mol Cell Biochem 2025; 480:2361-2375. [PMID: 39026029 DOI: 10.1007/s11010-024-05068-z] [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: 05/27/2024] [Accepted: 06/30/2024] [Indexed: 07/20/2024]
Abstract
Ferroptosis is recently discovered as an important player in the initiation, proliferation, and progression of human tumors. Insulin-like growth factor 2 mRNA-binding protein 3 (IGF2BP3) has been reported as an oncogene in multiple types of cancers, including lung adenocarcinoma (LUAD). However, little research has been designed to investigate the regulation of IGF2BP3 on ferroptosis in LUAD. qRT-PCR and western blot were used to measure the mRNA and protein expression of IGF2BP3 and transcription factor AP-2 alpha (TFAP2A). CCK-8 assay was performed to determine cell viability. DCFH-DA and C11-BODIPY staining were used to detect the levels of intracellular reactive oxygen species (ROS) and lipid ROS. The corresponding assay kits were used to analyze the levels of malondialdehyde (MDA) and glutathione (GSH). SRAMP website and m6A RNA immunoprecipitation (Me-RIP) were used to predict and confirm the m6A modification of TFAP2A. RIP experiments were conducted to confirm the binding of IGF2BP3 and TFAP2A. RNA stability assay was performed using actinomycin D. Chromatin immunoprecipitation (ChIP) and dual-luciferase reporter experiments were performed to confirm the interaction between TFAP2A and cystine/glutamate antiporter solute carrier family 7 member 11 (SLC7A11) or glutathione peroxidase 4 (GPX4). Mice xenotransplant model was also constructed to explore the effect of IGF2BP3 on LUAD tumor growth and ferroptosis. IGF2BP3 and TFAP2A were both highly expressed in LUAD. IGF2BP3 or TFAP2A knockdown induced ferroptosis by aggravating erastin-induced cell viability suppression, increasing the production of intracellular ROS, lipid ROS, and MDA, and decreasing GSH synthesis, GSH/GSSG ratio, and cystine uptake. Mechanistically, IGF2BP3 stabilized TFAP2A expression via m6A modification. Moreover, sh-IGF2BP3-mediated ferroptosis was significantly abated by TFAP2A overexpression. Furthermore, TFAP2A binds to the promoters of SLC7A11 and GPX4 to promote their transcription. Also, IGF2BP3 depletion suppressed LUAD tumor growth by inducing ferroptosis in mice. IGF2BP3 suppresses ferroptosis in LUAD by m6A-dependent regulation of TFAP2A to promote the transcription of SLC7A11 and GPX4. Our findings suggest that targeting IGF2BP3/TFAP2A/SLC7A11/GPX4 axis might be a potential therapeutic choice to increase ferroptosis sensitivity in LUAD.
Collapse
Affiliation(s)
- Pengpeng Li
- Tumor Treatment Center, The Fifth Affiliated Hospital of Zhengzhou University, No. 3 Kangfuqian Street, Erqi District, Zhengzhou, 450052, China
| | - Dan Chu
- Department of Respiratory Medicine, The First Affiliated Hospital of Zhengzhou University, No. 1 Jianshe East Road, Erqi District, Zhengzhou, 450052, China.
| | - Guangcheng Ding
- Tumor Treatment Center, The Fifth Affiliated Hospital of Zhengzhou University, No. 3 Kangfuqian Street, Erqi District, Zhengzhou, 450052, China
| | - Dehua Qin
- Tumor Treatment Center, The Fifth Affiliated Hospital of Zhengzhou University, No. 3 Kangfuqian Street, Erqi District, Zhengzhou, 450052, China
| | - Yajing Bu
- Tumor Treatment Center, The Fifth Affiliated Hospital of Zhengzhou University, No. 3 Kangfuqian Street, Erqi District, Zhengzhou, 450052, China
| | - Bi Tian
- Tumor Treatment Center, The Fifth Affiliated Hospital of Zhengzhou University, No. 3 Kangfuqian Street, Erqi District, Zhengzhou, 450052, China.
| |
Collapse
|
3
|
Zeng F, Chen L, Li J, Yu W, Sa N, Zhang K, Qu C, Wen D. A pan-cancer analysis reveals the oncogenic and immunological role of insulin-like growth factor 2 mRNA-binding protein family members. Discov Oncol 2025; 16:323. [PMID: 40088376 PMCID: PMC11910485 DOI: 10.1007/s12672-025-02077-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/16/2024] [Accepted: 03/05/2025] [Indexed: 03/17/2025] Open
Abstract
PURPOSE To investigate the expression and clinical significance of insulin-like growth factor 2 mRNA-binding protein family members (IGF2BPs) in pan-cancer and evaluate their potential as targets for tumor immunotherapy. METHODS Based on data from the cancer genome atlas (TCGA) database, pan-cancer analysis was conducted to examine the clinical significance of IGF2BPs expression in twenty-two tumors. RESULTS Differential expression analysis showed high expression of IGF2BPs in most tumor tissues. Survival and mutation analyses suggested that the overexpression of IGF2BPs was associated with poor prognosis and mutation status of certain tumors. Methylation analysis revealed the methylation levels of IGF2BP1/2/3 in certain tumors were intricately linked to their mRNA expression, patient prognosis, and immune cell infiltration. Enrichment analysis indicated that abnormal expression of IGF2BPs was associated with various common tumor-related pathways in different tumors, including AMPK, Hippo, PI3K-Akt, EMT, and p53. In addition, immune correlation analysis revealed that IGF2BPs were closely related to immunotherapy-related indicators (immune cell infiltration, major histocompatibility complex (MHC), immune checkpoints, tumor mutation burden (TMB), and microsatellite instability (MSI)) in some tumors. Drug sensitivity analysis indicated that IGF2BPs were sensitive to some common chemotherapeutic drugs (alvocidib, dasatinib, trametinib, and selumetinib). CONCLUSION IGF2BPs exhibit significantly high expression in most tumors and are associated with prognosis, pathological stage, mutational status, methylation levels, and the relevant indicators of immunotherapy sensitivity in multiple tumors. Moreover, IGF2BPs may play an oncogenic role by activating common signaling pathways. Therefore, IGF2BPs may be potential prognostic markers for tumor therapy and targets for immunotherapy and drug therapy.
Collapse
Affiliation(s)
- Fuling Zeng
- Department of Laboratory Medicine, Shenzhen Guangming District People's Hospital, Shenzhen, 518000, Guangdong, China
| | - Liuyan Chen
- Department of Pathophysiology, School of Medicine, Jinan University, Guangzhou, 510632, Guangdong, China
| | - Jing Li
- Department of Pathophysiology, School of Medicine, Jinan University, Guangzhou, 510632, Guangdong, China
| | - Wenna Yu
- College of Pharmacy, Jinan University, Guangzhou, 510632, Guangdong, China
| | - Niya Sa
- Department of Pathophysiology, School of Medicine, Jinan University, Guangzhou, 510632, Guangdong, China
| | - Keke Zhang
- Department of Pathophysiology, School of Medicine, Jinan University, Guangzhou, 510632, Guangdong, China
| | - Chen Qu
- Department of Pathophysiology, School of Medicine, Jinan University, Guangzhou, 510632, Guangdong, China.
| | - Daolin Wen
- Department of Laboratory Medicine, Shenzhen Guangming District People's Hospital, Shenzhen, 518000, Guangdong, China.
| |
Collapse
|
4
|
Zhang Z, Gao Y, Qian Y, Wei B, Jiang K, Sun Z, Zhang F, Yang M, Baldi S, Yu X, Zuo Y, Ren S. The Lyn/RUVBL1 Complex Promotes Colorectal Cancer Liver Metastasis by Regulating Arachidonic Acid Metabolism Through Chromatin Remodeling. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2406562. [PMID: 39665272 PMCID: PMC11792055 DOI: 10.1002/advs.202406562] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Revised: 11/26/2024] [Indexed: 12/13/2024]
Abstract
Liver metastasis is a common cause of death in colorectal cancer (CRC) patients, but epigenetic remodeling and metabolic reprogramming for CRC liver metastasis remain unclear. The study revealed that the Lyn/RUVBL1 complex is highly expressed in CRC and is closely correlated with liver metastasis. On the one hand, ATAC-seq and HiCut suggested that Lyn/RUVBL1 regulates the expression of TRIB3 through the POL II-mediated chromatin conformation of TRIB3 and thus the expression of β-catenin. This promotes the proliferation and migration of CRC through β-catenin-mediated upregulation of MMP9 and VEGF. On the other hand, metabolomics revealed that Lyn/RUVBL1 regulates the expression of PGE2 through the enzyme COX2, thereby promoting arachidonic acid (AA) metabolism. CUT-Tag showed that Lyn/RUVBL1 silencing reduces the H3K27ac level in the COX2 promoter. Then, it is found that COX2 is regulated by the transcription factor FOXA1. Lyn/RUVBL1 modulates AA metabolism by regulating the chromatin accessibility of FOXA1. AA metabolism promotes the metastasis of CRC by affecting β-catenin nuclear translocation and upregulating MMP9 and VEGF. These findings suggest that the Lyn/RUVBL1 complex mediates epigenetic remodeling to regulate the metabolic reprogramming of AA, highlighting its role in promoting the metastasis of CRC.
Collapse
Affiliation(s)
- Zhenyu Zhang
- Department of General SurgeryThe Second Hospital of Dalian Medical UniversityDalian116023China
- Department of Clinical BiochemistryCollege of Laoratory Medicine, Dalian Medical UniversityDalian116044China
| | - Yina Gao
- Department of General SurgeryThe Second Hospital of Dalian Medical UniversityDalian116023China
- Department of Clinical BiochemistryCollege of Laoratory Medicine, Dalian Medical UniversityDalian116044China
| | - Yuanyuan Qian
- Department of Clinical BiochemistryCollege of Laoratory Medicine, Dalian Medical UniversityDalian116044China
| | - Bowen Wei
- Department of Clinical BiochemistryCollege of Laoratory Medicine, Dalian Medical UniversityDalian116044China
| | - Kexin Jiang
- Department of Clinical BiochemistryCollege of Laoratory Medicine, Dalian Medical UniversityDalian116044China
| | - Zhiwei Sun
- Department of General SurgeryThe Second Hospital of Dalian Medical UniversityDalian116023China
| | - Feifan Zhang
- Department of General SurgeryThe Second Hospital of Dalian Medical UniversityDalian116023China
| | - Mingming Yang
- Department of Clinical BiochemistryCollege of Laoratory Medicine, Dalian Medical UniversityDalian116044China
| | - Salem Baldi
- Department of Clinical BiochemistryCollege of Laoratory Medicine, Dalian Medical UniversityDalian116044China
| | - Xiaoqi Yu
- Department of Clinical BiochemistryCollege of Laoratory Medicine, Dalian Medical UniversityDalian116044China
| | - Yunfei Zuo
- Department of Clinical BiochemistryCollege of Laoratory Medicine, Dalian Medical UniversityDalian116044China
| | - Shuangyi Ren
- Department of General SurgeryThe Second Hospital of Dalian Medical UniversityDalian116023China
| |
Collapse
|
5
|
Zhao Y, Li J, Dian M, Bie Y, Peng Z, Zhou Y, Zhou B, Hao W, Wang X. Role of N6-methyladenosine methylation in nasopharyngeal carcinoma: current insights and future prospective. Cell Death Discov 2024; 10:490. [PMID: 39695216 DOI: 10.1038/s41420-024-02266-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Revised: 11/22/2024] [Accepted: 12/06/2024] [Indexed: 12/20/2024] Open
Abstract
Nasopharyngeal carcinoma (NPC) is a distinct type of head and neck squamous cell carcinoma prevalent in Southern China, Southeast Asia, and North Africa. Despite advances in treatment options, the prognosis for advanced NPC remains poor, underscoring the urgent need to explore its underlying mechanisms and develop novel therapeutic strategies. Epigenetic alterations have been shown to play a key role in NPC progression. Recent studies indicate that dysregulation of RNA modifications in NPC specifically affects tumor-related transcripts, influencing various oncogenic processes. This review provides a comprehensive overview of altered RNA modifications and their regulators in NPC, with a focus on m6A and its regulatory mechanisms. We discuss how m6A RNA modification influences gene expression and affects NPC initiation and progression at the molecular level, analyzing its impact on cancer-related biological functions. Understanding these modifications could reveal new biomarkers and therapeutic targets for NPC, offering promising directions for future research and precision medicine.
Collapse
Affiliation(s)
- YaYan Zhao
- Department of Oncology, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, China
- Cancer Research Institute of Integrated Traditional Chinese and Western Medicine, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, China
| | - Jie Li
- Department of Oncology, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, China
- Cancer Research Institute of Integrated Traditional Chinese and Western Medicine, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, China
| | - MeiJuan Dian
- Department of Thoracic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - YaNan Bie
- School of Basic Medical Sciences, Guangdong Pharmaceutical University, Guangzhou, China
| | - ZhiTao Peng
- Cancer Research Institute, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Ying Zhou
- Cancer Research Institute, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - BingQian Zhou
- Department of Oncology, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, China
- Cancer Research Institute of Integrated Traditional Chinese and Western Medicine, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, China
| | - WeiChao Hao
- Department of Oncology, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, China.
- Cancer Research Institute of Integrated Traditional Chinese and Western Medicine, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, China.
| | - XiCheng Wang
- Department of Oncology, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, China.
- Cancer Research Institute of Integrated Traditional Chinese and Western Medicine, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, China.
| |
Collapse
|
6
|
Fakhar M, Gul M, Li W. Interactive Structural Analysis of KH3-4 Didomains of IGF2BPs with Preferred RNA Motif Having m 6A Through Dynamics Simulation Studies. Int J Mol Sci 2024; 25:11118. [PMID: 39456902 PMCID: PMC11508745 DOI: 10.3390/ijms252011118] [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: 08/04/2024] [Revised: 10/02/2024] [Accepted: 10/11/2024] [Indexed: 10/28/2024] Open
Abstract
m6A modification is the most common internal modification of messenger RNA in eukaryotes, and the disorder of m6A can trigger cancer progression. The GGACU is considered the most frequent consensus sequence of target transcripts which have a GGAC m6A core motif. Newly identified m6A 'readers' insulin-like growth factor 2 mRNA-binding proteins modulate gene expression by binding to the m6A binding sites of target mRNAs, thereby affecting various cancer-related processes. The dynamic impact of the methylation at m6A within the GGAC motif on human IGF2BPs has not been investigated at the structural level. In this study, through in silico analysis, we mapped IGF2BPs binding sites for the GGm6AC RNA core motif of target mRNAs. Subsequent molecular dynamics simulation analysis at 400 ns revealed that only the KH4 domain of IGF2BP1, containing the 503GKGG506 motif and its periphery residues, was involved in the interaction with the GGm6AC backbone. Meanwhile, the methyl group of m6A is accommodated by a shallow hydrophobic cradle formed by hydrophobic residues. Interestingly, in IGF2BP2 and IGF2BP3 complexes, the RNA was observed to shift from the KH4 domain to the KH3 domain in the simulation at 400 ns, indicating a distinct dynamic behavior. This suggests a conformational stabilization upon binding, likely essential for the functional interactions involving the KH3-4 domains. These findings highlight the potential of targeting IGF2BPs' interactions with m6A modifications for the development of novel oncological therapies.
Collapse
Affiliation(s)
- Muhammad Fakhar
- Institute for Advanced Study, Shenzhen University, Shenzhen 518060, China; (M.F.); (M.G.)
- College of Civil and Transportation Engineering, Shenzhen University, Shenzhen 518060, China
| | - Mehreen Gul
- Institute for Advanced Study, Shenzhen University, Shenzhen 518060, China; (M.F.); (M.G.)
- College of Civil and Transportation Engineering, Shenzhen University, Shenzhen 518060, China
| | - Wenjin Li
- Institute for Advanced Study, Shenzhen University, Shenzhen 518060, China; (M.F.); (M.G.)
| |
Collapse
|
7
|
Ding P, Wu H, Wu J, Li T, He J, Ju Y, Liu Y, Li F, Deng H, Gu R, Zhang L, Guo H, Tian Y, Yang P, Meng N, Li X, Guo Z, Meng L, Zhao Q. N6-methyladenosine modified circPAK2 promotes lymph node metastasis via targeting IGF2BPs/VEGFA signaling in gastric cancer. Oncogene 2024; 43:2548-2563. [PMID: 39014193 DOI: 10.1038/s41388-024-03099-w] [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: 03/10/2024] [Revised: 07/02/2024] [Accepted: 07/05/2024] [Indexed: 07/18/2024]
Abstract
Circular RNAs (circRNAs) have emerged as key regulators of cancer occurrence and progression, as well as promising biomarkers for cancer diagnosis and prognosis. However, the potential mechanisms of circRNAs implicated in lymph node (LN) metastasis of gastric cancer remain unclear. Herein, we identify a novel N6-methyladenosine (m6A) modified circRNA, circPAK2, which is significantly upregulated in gastric cancer tissues and metastatic LN tissues. Functionally, circPAK2 enhances the migration, invasion, lymphangiogenesis, angiogenesis, epithelial-mesenchymal transition (EMT), and metastasis of gastric cancer in vitro and in vivo. Mechanistically, circPAK2 is exported by YTH domain-containing protein 1 (YTHDC1) from the nucleus to the cytoplasm in an m6A methylation-dependent manner. Moreover, increased cytoplasmic circPAK2 interacts with Insulin-Like Growth Factor 2 mRNA-Binding Proteins (IGF2BPs) and forms a circPAK2/IGF2BPs/VEGFA complex to stabilize VEGFA mRNA, which contributes to gastric cancer vasculature formation and aggressiveness. Clinically, high circPAK2 expression is positively associated with LN metastasis and poor prognosis in gastric cancer. This study highlights m6A-modified circPAK2 as a key regulator of LN metastasis of gastric cancer, thus supporting circPAK2 as a promising therapeutic target and prognostic biomarker for gastric cancer.
Collapse
Affiliation(s)
- Ping'an Ding
- The Third Department of Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
- Hebei Key Laboratory of Precision Diagnosis and Comprehensive Treatment of Gastric Cancer, Shijiazhuang, Hebei, China
- Big data analysis and mining application for precise diagnosis and treatment of gastric cancer Hebei Provincial Engineering Research Center, Shijiazhuang, Hebei, China
| | - Haotian Wu
- The Third Department of Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
- Hebei Key Laboratory of Precision Diagnosis and Comprehensive Treatment of Gastric Cancer, Shijiazhuang, Hebei, China
- Big data analysis and mining application for precise diagnosis and treatment of gastric cancer Hebei Provincial Engineering Research Center, Shijiazhuang, Hebei, China
| | - Jiaxiang Wu
- The Third Department of Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
- Hebei Key Laboratory of Precision Diagnosis and Comprehensive Treatment of Gastric Cancer, Shijiazhuang, Hebei, China
- Big data analysis and mining application for precise diagnosis and treatment of gastric cancer Hebei Provincial Engineering Research Center, Shijiazhuang, Hebei, China
| | - Tongkun Li
- The Third Department of Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
- Hebei Key Laboratory of Precision Diagnosis and Comprehensive Treatment of Gastric Cancer, Shijiazhuang, Hebei, China
- Big data analysis and mining application for precise diagnosis and treatment of gastric cancer Hebei Provincial Engineering Research Center, Shijiazhuang, Hebei, China
| | - Jinchen He
- The Third Department of Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
- Hebei Key Laboratory of Precision Diagnosis and Comprehensive Treatment of Gastric Cancer, Shijiazhuang, Hebei, China
- Big data analysis and mining application for precise diagnosis and treatment of gastric cancer Hebei Provincial Engineering Research Center, Shijiazhuang, Hebei, China
| | - Yingchao Ju
- Hebei Key Laboratory of Precision Diagnosis and Comprehensive Treatment of Gastric Cancer, Shijiazhuang, Hebei, China
- Big data analysis and mining application for precise diagnosis and treatment of gastric cancer Hebei Provincial Engineering Research Center, Shijiazhuang, Hebei, China
- Animal Center of the Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Yueping Liu
- Hebei Key Laboratory of Precision Diagnosis and Comprehensive Treatment of Gastric Cancer, Shijiazhuang, Hebei, China
- Big data analysis and mining application for precise diagnosis and treatment of gastric cancer Hebei Provincial Engineering Research Center, Shijiazhuang, Hebei, China
- Department of Pathology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Fang Li
- Hebei Key Laboratory of Precision Diagnosis and Comprehensive Treatment of Gastric Cancer, Shijiazhuang, Hebei, China
- Big data analysis and mining application for precise diagnosis and treatment of gastric cancer Hebei Provincial Engineering Research Center, Shijiazhuang, Hebei, China
- Department of Pathology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Huiyan Deng
- Hebei Key Laboratory of Precision Diagnosis and Comprehensive Treatment of Gastric Cancer, Shijiazhuang, Hebei, China
- Big data analysis and mining application for precise diagnosis and treatment of gastric cancer Hebei Provincial Engineering Research Center, Shijiazhuang, Hebei, China
- Department of Pathology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Renjun Gu
- School of Chinese Medicine & School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
- Department of Gastroenterology and Hepatology, Jinling Hospital, Medical School of Nanjing University, Nanjing, Jiangsu, China
| | - Lilong Zhang
- Department of General Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Honghai Guo
- The Third Department of Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
- Hebei Key Laboratory of Precision Diagnosis and Comprehensive Treatment of Gastric Cancer, Shijiazhuang, Hebei, China
- Big data analysis and mining application for precise diagnosis and treatment of gastric cancer Hebei Provincial Engineering Research Center, Shijiazhuang, Hebei, China
| | - Yuan Tian
- The Third Department of Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
- Hebei Key Laboratory of Precision Diagnosis and Comprehensive Treatment of Gastric Cancer, Shijiazhuang, Hebei, China
- Big data analysis and mining application for precise diagnosis and treatment of gastric cancer Hebei Provincial Engineering Research Center, Shijiazhuang, Hebei, China
| | - Peigang Yang
- The Third Department of Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
- Hebei Key Laboratory of Precision Diagnosis and Comprehensive Treatment of Gastric Cancer, Shijiazhuang, Hebei, China
- Big data analysis and mining application for precise diagnosis and treatment of gastric cancer Hebei Provincial Engineering Research Center, Shijiazhuang, Hebei, China
| | - Ning Meng
- Department of General Surgery, Shijiazhuang People's Hospital, Shijiazhuang, Hebei, China
| | - Xiaolong Li
- Department of General Surgery, Baoding Central Hospital, Baoding, Hebei, China
| | - Zhenjiang Guo
- General Surgery Department, Hengshui People's Hospital, Hengshui, Hebei, China
| | - Lingjiao Meng
- Research Center and Tumor Research Institute, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, China.
| | - Qun Zhao
- The Third Department of Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, China.
- Hebei Key Laboratory of Precision Diagnosis and Comprehensive Treatment of Gastric Cancer, Shijiazhuang, Hebei, China.
- Big data analysis and mining application for precise diagnosis and treatment of gastric cancer Hebei Provincial Engineering Research Center, Shijiazhuang, Hebei, China.
| |
Collapse
|
8
|
Chen L, Hui L, Li J. The multifaceted role of insulin-like growth factor binding protein 7. Front Cell Dev Biol 2024; 12:1420862. [PMID: 39081862 PMCID: PMC11286461 DOI: 10.3389/fcell.2024.1420862] [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: 04/24/2024] [Accepted: 07/01/2024] [Indexed: 08/02/2024] Open
Abstract
Insulin-like growth factor binding protein 7 (IGFBP7) serves as a crucial extracellular matrix protein, exerting pivotal roles in both physiological and pathological processes. This comprehensive review meticulously delineates the structural attributes of IGFBP7, juxtaposing them with other members within the IGFBP families, and delves into the expression patterns across various tissues. Furthermore, the review thoroughly examines the multifaceted functions of IGFBP7, encompassing its regulatory effects on cell proliferation, apoptosis, and migration, elucidating the underlying mechanistic pathways. Moreover, it underscores the compelling roles in tumor progression, acute kidney injury, and reproductive processes. By rigorously elucidating the diverse functionalities and regulatory networks of IGFBP7 across various physiological and pathological contexts, this review aims to furnish a robust theoretical framework and delineate future research trajectories for leveraging IGFBP7 in disease diagnosis, therapeutic interventions, and pharmaceutical innovations.
Collapse
Affiliation(s)
| | | | - Jun Li
- Department of Immunology, Center of Immunomolecular Engineering, Innovation and Practice Base for Graduate Students Education, Zunyi Medical University, Zunyi, China
| |
Collapse
|
9
|
Zang Y, Tian Z, Wang D, Li Y, Zhang W, Ma C, Liao Z, Gao W, Qian L, Xu X, Jia J, Liu Z. METTL3-mediated N 6-methyladenosine modification of STAT5A promotes gastric cancer progression by regulating KLF4. Oncogene 2024; 43:2338-2354. [PMID: 38879589 PMCID: PMC11271408 DOI: 10.1038/s41388-024-03085-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Revised: 06/02/2024] [Accepted: 06/07/2024] [Indexed: 07/21/2024]
Abstract
N6-methyladenosine (m6A) is the predominant post-transcriptional RNA modification in eukaryotes and plays a pivotal regulatory role in various aspects of RNA fate determination, such as mRNA stability, alternative splicing, and translation. Dysregulation of the critical m6A methyltransferase METTL3 is implicated in tumorigenesis and development. Here, this work showed that METTL3 is upregulated in gastric cancer tissues and is associated with poor prognosis. METTL3 methylates the A2318 site within the coding sequence (CDS) region of STAT5A. IGF2BP2 recognizes and binds METTL3-mediated m6A modification of STAT5A through its GXXG motif in the KH3 and KH4 domains, leading to increased stability of STAT5A mRNA. In addition, both METTL3 and IGF2BP2 are positively correlated with STAT5A in human gastric cancer tissue samples. Helicobacter pylori infection increased the expression level of METTL3 in gastric cancer cells, thereby leading to the upregulation of STAT5A. Functional studies indicated that STAT5A overexpression markedly enhances the proliferation and migration of GC cells, whereas STAT5A knockdown has inhibitory effects. Further nude mouse experiments showed that STAT5A knockdown effectively inhibits the growth and metastasis of gastric cancer in vivo. Moreover, as a transcription factor, STAT5A represses KLF4 transcription by binding to its promoter region. The overexpression of KLF4 can counteract the oncogenic impact of STAT5A. Overall, this study highlights the crucial role of m6A in gastric cancer and provides potential therapeutic targets for gastric cancer.
Collapse
Affiliation(s)
- Yichen Zang
- Department of Biochemistry and Molecular Biology, Key Laboratory for Experimental Teratology of Chinese Ministry of Education, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Zhuangfei Tian
- Department of Biochemistry and Molecular Biology, Key Laboratory for Experimental Teratology of Chinese Ministry of Education, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Dandan Wang
- Department of Microbiology, Key Laboratory for Experimental Teratology of Chinese Ministry of Education, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Yaxuan Li
- Department of Biochemistry and Molecular Biology, Key Laboratory for Experimental Teratology of Chinese Ministry of Education, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Wenhui Zhang
- School of Clinical Medicine, Qingdao University, Qingdao, China
| | - Cunying Ma
- Department of Biochemistry and Molecular Biology, Key Laboratory for Experimental Teratology of Chinese Ministry of Education, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Zhenzhi Liao
- Department of Biochemistry and Molecular Biology, Key Laboratory for Experimental Teratology of Chinese Ministry of Education, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Wenrong Gao
- Department of Microbiology, Key Laboratory for Experimental Teratology of Chinese Ministry of Education, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Lilin Qian
- Department of Biochemistry and Molecular Biology, Key Laboratory for Experimental Teratology of Chinese Ministry of Education, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Xia Xu
- Department of Biochemistry and Molecular Biology, Key Laboratory for Experimental Teratology of Chinese Ministry of Education, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Jihui Jia
- Department of Microbiology, Key Laboratory for Experimental Teratology of Chinese Ministry of Education, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Zhifang Liu
- Department of Biochemistry and Molecular Biology, Key Laboratory for Experimental Teratology of Chinese Ministry of Education, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China.
| |
Collapse
|
10
|
Cai Y, Wang Y, Mao B, You Q, Guo X. Targeting insulin-like growth factor 2 mRNA-binding proteins (IGF2BPs) for the treatment of cancer. Eur J Med Chem 2024; 268:116241. [PMID: 38382391 DOI: 10.1016/j.ejmech.2024.116241] [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: 12/14/2023] [Revised: 02/05/2024] [Accepted: 02/14/2024] [Indexed: 02/23/2024]
Abstract
Insulin-like growth factor 2 mRNA-binding proteins (IMPs, IGF2BPs) are RNA-binding proteins that regulate a variety of biological processes. In recent years, several studies have found that IGF2BPs play multiple roles in various biological processes, especially in cancer, and speculated on their mechanism of anticancer effect. In addition, targeting IGF2BPs or their downstream target gene has also received extensive attention as an effective treatment for different types of cancer. In this review, we summarized the recent progress on the role of IGF2BPs in cancers and their structural characteristics. We focused on describing the development of inhibitors targeting IGF2BPs and the prospects for further applications.
Collapse
Affiliation(s)
- Yuanqian Cai
- State Key Laboratory of Natural Medicines, and Jiang Su Key Laboratory of Drug, Design and Optimization, China Pharmaceutical University, Nanjing, 210009, China; Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, China
| | - Yingzhe Wang
- State Key Laboratory of Natural Medicines, and Jiang Su Key Laboratory of Drug, Design and Optimization, China Pharmaceutical University, Nanjing, 210009, China; Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, China
| | - Bingjie Mao
- State Key Laboratory of Natural Medicines, and Jiang Su Key Laboratory of Drug, Design and Optimization, China Pharmaceutical University, Nanjing, 210009, China; Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, China
| | - Qidong You
- State Key Laboratory of Natural Medicines, and Jiang Su Key Laboratory of Drug, Design and Optimization, China Pharmaceutical University, Nanjing, 210009, China; Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, China.
| | - Xiaoke Guo
- State Key Laboratory of Natural Medicines, and Jiang Su Key Laboratory of Drug, Design and Optimization, China Pharmaceutical University, Nanjing, 210009, China; Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, China.
| |
Collapse
|
11
|
Wang J, Zheng F, Wang D, Yang Q. Regulation of ULK1 by WTAP/IGF2BP3 axis enhances mitophagy and progression in epithelial ovarian cancer. Cell Death Dis 2024; 15:97. [PMID: 38286802 PMCID: PMC10824720 DOI: 10.1038/s41419-024-06477-0] [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: 12/04/2023] [Revised: 01/15/2024] [Accepted: 01/16/2024] [Indexed: 01/31/2024]
Abstract
There is a pressing need for innovative therapeutic strategies for patients with epithelial ovarian cancer (EOC). Previous studies have shown that UNC-51-like kinase 1 (ULK1), a serine/threonine kinase, is crucial in regulating cellular autophagy and mitophagy across various tumor types. However, the clinical implications, biological functions, and potential mechanisms of ULK1 in EOC remain poorly understood. This study demonstrates that ULK1 expression is upregulated in EOC tissue samples and EOC cell lines, with increased ULK1 expression correlating with poor prognosis. Functionally, overexpressed ULK1 enhances the proliferation and migration abilities of EOC cells both in vitro and in vivo. Mechanistically, ULK1 was identified as an m6A target of WTAP. WTAP-mediated m6A modification of ULK1 enhanced its mRNA stability in an IGF2BP3-dependent manner, leading to elevated ULK1 expression and enhanced mitophagy in EOC. In summary, our research reveals that the WTAP/IGF2BP3-ULK1 axis significantly influences protective mitophagy in EOC, contributing to its progression. Therefore, the regulatory mechanisms and biological function of ULK1 identify it as a potential molecular target for therapeutic intervention in EOC.
Collapse
Affiliation(s)
- Jiao Wang
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, 110004, China
| | - Fei Zheng
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, 110004, China
| | - Dandan Wang
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, 110004, China
| | - Qing Yang
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, 110004, China.
| |
Collapse
|