1
|
Hu M, Zhang T, Ma Y, Wang H, Hou S. Unraveling lncRNA TRG-AS1: a novel biomarker for poor prognosis of gastric cancer and key to regulating malignant behaviors by targeting miR-873-5p. Hereditas 2025; 162:88. [PMID: 40442738 PMCID: PMC12121179 DOI: 10.1186/s41065-025-00459-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2025] [Accepted: 05/18/2025] [Indexed: 06/02/2025] Open
Abstract
BACKGROUND AND STUDY AIMS To alleviate patient stress and advance gastric cancer research, this study aims to investigate the potential association between aberrant expression of TRG-AS1 and gastric cancer, and to examine its potential impact on the biological behaviors of gastric cancer cells. MATERIALS AND METHODS To find out how TRG-AS1 is expressed in the tissues and cells of gastric cancer, real-time fluorescence quantitative PCR was employed. The connection between TRG-AS1 and pathological features, as well as its prognostic importance, were examined using the Chi-square test and Cox regression analysis. The dual luciferase reporting assay was utilized to confirm the targeting of TRG-AS1 and miR-873-5p. Transwell assay and CCK-8 test were used to identify the roles that TRG-AS1 plays in cell metastasis and proliferation, respectively. RESULTS Research has revealed a downregulation of TRG-AS1 in the tissues and cells, which is strongly correlated with the differentiation, TNM stage, lymph node metastasis, depth of invasion, and patient survival rate in gastric cancer. The binding sites exists between miR-873-5p and TRG-AS1, and TRG-AS1 has the ability to negatively control miR-873-5p by acting as a competitive endogenous RNA. When TRG-AS1 was overexpressed, the malignant behavioral activity of gastric cancer cells was significantly decreased; however, the inhibitory effect was reversed when miR-873-5p was overexpressed. CONCLUSIONS TRG-AS1 is a potential predictor of poor prognosis in gastric cancer patients and targets miR-873-5p to inhibit the progression of cancer.
Collapse
Affiliation(s)
- Miao Hu
- Cancer Treatment Center, Affiliated Hospital of Beihua University, Jilin, 132011, China
| | - Tiesong Zhang
- Department of Cardiothoracic Surgery, The Fourth Medical Center, Chinese PLA General Hospital, Beijing, 100048, China
| | - Yuzhen Ma
- Department of Endoscopic Diagnosis and Treatment, Daqing Oil Field General Hospital, Daqing, 163001, China
| | - Huiling Wang
- Department of Clinical Laboratory, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, People's Hospital of Henan University, Zhengzhou, 450003, China
| | - Suping Hou
- Pathology, Third Hospital of Hebei Medical University, No. 139, Ziqiang Road, Shijiazhuang, Hebei, 050051, China.
| |
Collapse
|
2
|
Bi Z, Zhang Y, Song XR, Zheng WH, Chen P, Qiu PF, Liu YB, Lu YJ, Song XG, Wang YS. Identification of biomarkers of shrinkage modes after neoadjuvant therapy in HER-2 positive breast cancer. Int J Surg 2025; 111:3677-3684. [PMID: 40143733 DOI: 10.1097/js9.0000000000002349] [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: 10/09/2024] [Accepted: 03/12/2025] [Indexed: 03/28/2025]
Abstract
PURPOSE A nomogram to predict shrinkage modes after neoadjuvant therapy (NAT) was constructed in HER-2 positive (HER2+) breast cancer. The value and mechanism of targeting long noncoding RNA (lncRNA) as efficacy prediction biomarker was also evaluated. METHODS All enrolled patients received six cycles of chemotherapy (Docetaxel + Carboplatin) and anti-HER-2 dual-targeted therapy (Trastuzumab + Pertuzumab) before surgery. According to pathological three-dimensional (3D) models of residual tumor from 71 HER2+ patients, shrinkage modes were divided into concentric shrinkage mode (CSM) and non-CSM (NCSM). LncRNAs in core biopsy tissues in the CSM and NCSM groups were selected by microarray and validated by RT-PCR. A nomogram was constructed to predict shrinkage modes after NAT in combination with clinical-pathological and transcriptome signatures. Cell proliferation was used CCK-8 and colony formation assay. PAPIS Kit was used to perform nuclear and cytoplasmic separation. The cell drug resistance assays were to explore the value of paclitaxel. The ChIRP-MS assay was to search RNA-binding proteins and verified by WB. Cell cycle analysis was carried out by flow cytometry. RESULTS Independent predictors of NCSM were lymph nodes downstaging after NAT, mammographic malignant calcification, hormone receptor expression, and RUVBL1-AS1 expression. A nomogram was constructed in combination with these predictors, which showed an area under the curve of 0.883, supporting the predictive power of the method. Overexpression of RUVBL1-AS1 inhibited HER2+ cells proliferation. Overexpression of RUVBL1-AS1 increased the number of cells in G1/S phase and decreased that of cells in G2 phase. RUVBL1-AS1 increased paclitaxel resistance and downregulated VCP expression. RUVBL1-AS1 affects cell cycle progression by downregulating VCP, resulting in the reduction of cells in G2/M phase, thereby weakening the sensitivity to paclitaxel. CONCLUSION The nomogram could accurately predict shrinkage modes after NAT, and may help guide the individualized selection of breast conserving surgery candidates after NAT. RUVBL1-AS1 might be a promising therapeutic target of paclitaxel-based chemotherapy inHER2+ breast cancer.
Collapse
Affiliation(s)
- Zhao Bi
- Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, People's Republic of China
| | - Yue Zhang
- Shanghai Pudong New Area Center for Disease Control and Prevention, Shanghai, People's Republic of China
| | - Xian-Rang Song
- Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, People's Republic of China
| | - Wen-Hao Zheng
- Rizhao Central Hospital, Rizhao, Shandong, People's Republic of China
| | - Peng Chen
- Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, People's Republic of China
| | - Peng-Fei Qiu
- Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, People's Republic of China
| | - Yan-Bing Liu
- Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, People's Republic of China
| | - Yong-Jin Lu
- Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, People's Republic of China
| | - Xing-Guo Song
- Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, People's Republic of China
| | - Yong-Sheng Wang
- Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, People's Republic of China
| |
Collapse
|
3
|
Yu S, Liang J, Liu L, Chen M, Chen C, Zhou D. AC129507.1 is a ferroptosis-related target identified by a novel mitochondria-related lncRNA signature that is involved in the tumor immune microenvironment in gastric cancer. J Transl Med 2025; 23:290. [PMID: 40050892 PMCID: PMC11887229 DOI: 10.1186/s12967-025-06287-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Accepted: 02/23/2025] [Indexed: 03/09/2025] Open
Abstract
BACKGROUND Gastric cancer (GC) is one of the most common malignancies. Previous studies have shown that mitochondrial metabolism is associated with malignancies. However, relevant research on mitochondria-related lncRNAs in GC is lacking. METHODS We integrated the corresponding information of patients with GC from The Cancer Genome Atlas (TCGA) database. Mitochondria-related lncRNAs were selected based on differential expression and a correlation analysis to construct a prognostic model. The mutation data were analyzed to distinguish differences in the tumor mutation burden (TMB). Single-sample gene set enrichment analysis (ssGSEA) was performed to evaluate immunological differences. A series of cell-based experiments were adopted to evaluate the biological behavior of GC. RESULTS A total of 1571 mitochondria-related lncRNAs were identified. A prognostic signature incorporating nine lncRNAs was built based on 293 suitable GC cases and could predict patient prognosis. The TMB and ssGSEA indicated that the low-risk group displayed increased immune function. The enrichment analysis indicated that the differentially expressed genes were enriched in metabolic functions. AC129507.1 was significantly upregulated in GC cells and associated with a poor prognosis, and its knockdown inhibited the proliferation and migration of GC cells. Mechanistically, silencing AC129507.1 led to abnormal glycolipid metabolism and oxidative stress, thus inducing ferroptosis. CONCLUSIONS Our nine-lncRNA risk signature could powerfully predict patient prognosis. AC129507.1 promoted the malignant phenotypes of GC cells. AC129507.1 could play a nonnegligible role in GC by promoting the formation of a immunosuppressive tumor microenvironment by inhibiting the initiation of ferroptosis, which needs to be further explored.
Collapse
Affiliation(s)
- Shanshan Yu
- Department of Surgical Oncology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310000, China
| | - Jinxiao Liang
- Department of Surgical Oncology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310000, China
| | - Lixiao Liu
- Department of Obstetrics and Gynecology, Ningbo City First Hospital, Ningbo University, Ningbo, China
| | - Ming Chen
- Department of Surgical Oncology, Children's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Cheng Chen
- Department of Surgical Oncology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310000, China
| | - Donghui Zhou
- Department of Surgical Oncology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310000, China.
| |
Collapse
|
4
|
Li Z, Chen L, Zhang G, Wang S, Xu E, Teng J, Xu J, Peng F, Min Q, Wang Z, Shao S, Zhao L, Shan B, Wang Y, Zhan Q, Liu X. Loss of MNX1 Sensitizes Tumors to Cytotoxic T Cells by Degradation of PD-L1 mRNA. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2403077. [PMID: 39912421 PMCID: PMC11947991 DOI: 10.1002/advs.202403077] [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: 03/23/2024] [Revised: 12/04/2024] [Indexed: 02/07/2025]
Abstract
Immune checkpoint blockade (ICB) therapy, targeting programmed cell death ligand-1 (PD-L1)/programmed cell death protein 1 (PD-1) axis and cytotoxic T-lymphocyte-associated protein 4 (CTLA-4), has exhibited amazing clinical outcomes in various types of cancers. However, only a small portion of patients benefit from ICB therapy, indicating that the mechanism underlying immune checkpoint is still unclear. Here, it is reported that motor neuron and pancreas homeobox 1 (MNX1), a homeobox domain-containing transcription factor, contributes to the tumor immune escape. MNX1 increases PD-L1 expression in cancer cells by stabilizing PD-L1 mRNA rather than activating transcription. Mechanistically, MNX1 exists in the cytoplasm of cancer cells and interacts with Y-box binding protein 1 (YBX1), a multifunctional DNA/RNA-binding protein, to enhance the binding of YBX1 to PD-L1 mRNA. MNX1 ablation activates cytotoxic T cell-mediated anti-tumor immunity and sensitizes CTLA-4 blockade therapy. Moreover, MNX1 also facilitates tumor progression in an immune-independent manner in cancer cells. In addition, MNX1 is upregulated by its adjacent long non-coding RNA MNX1-AS1 via HECT and RLD domain containing E3 ubiquitin protein ligase 2 (HERC2). Together, these results reveal MNX1 as a novel immune checkpoint regulator with promising therapeutic potential.
Collapse
Affiliation(s)
- Zhengzheng Li
- Institute of Cancer Stem CellDalian Medical UniversityDalian116044China
- Soochow University Cancer InstituteSuzhou215000China
| | - Lei Chen
- Institute of Cancer Stem CellDalian Medical UniversityDalian116044China
- Department of Pulmonary OncologyAffiliated Hospital of Guangdong Medical UniversityZhanjiang524001China
| | - Ge Zhang
- Department of ImmunologyCollege of Basic Medical SciencesDalian Medical UniversityDalian116044China
| | - Shuang Wang
- Institute of Cancer Stem CellDalian Medical UniversityDalian116044China
| | - Enhang Xu
- Institute of Cancer Stem CellDalian Medical UniversityDalian116044China
| | - Jinglei Teng
- Institute of Cancer Stem CellDalian Medical UniversityDalian116044China
| | - Jiancheng Xu
- Soochow University Cancer InstituteSuzhou215000China
| | - Fang Peng
- Department of Pathologythe Second Affiliated Hospital of Dalian Medical UniversityDalian116023China
| | - Qingjie Min
- Institute of Cancer Stem CellDalian Medical UniversityDalian116044China
| | - Zhuoya Wang
- Institute of Cancer Stem CellDalian Medical UniversityDalian116044China
| | - Shujuan Shao
- University Key Laboratory of Proteomics in Liaoning ProvinceDalian Medical UniversityDalian116044China
| | - Lianmei Zhao
- Research Centerthe Fourth Hospital of Hebei Medical UniversityShijiazhuang050011China
| | - Baoen Shan
- Research Centerthe Fourth Hospital of Hebei Medical UniversityShijiazhuang050011China
| | - Yang Wang
- Institute of Cancer Stem CellDalian Medical UniversityDalian116044China
| | - Qimin Zhan
- Institute of Cancer Stem CellDalian Medical UniversityDalian116044China
- Soochow University Cancer InstituteSuzhou215000China
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing)Laboratory of Molecular OncologyPeking University Cancer Hospital & InstituteBeijing100142China
| | - Xuefeng Liu
- Institute of Cancer Stem CellDalian Medical UniversityDalian116044China
| |
Collapse
|
5
|
Wang Q, Li D, Ma H, Li Z, Wu J, Qiao J, Liu J, Zhao J, Ma R, Tian L, Zhang L, Yang J, Wang J, Qin S, Su Z. Tumor cell-derived EMP1 is essential for cancer-associated fibroblast infiltration in tumor microenvironment of triple-negative breast cancer. Cell Death Dis 2025; 16:143. [PMID: 40016223 PMCID: PMC11868485 DOI: 10.1038/s41419-025-07464-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Revised: 02/06/2025] [Accepted: 02/19/2025] [Indexed: 03/01/2025]
Abstract
The role of epithelial membrane protein 1 (EMP1) in tumor microenvironment (TME) remodeling has not yet been elucidated. In addition, the biological function of EMP1 in triple-negative breast cancer (TNBC) is largely unclear. In this study, we examined the infiltration landscape of cell types in the TME of breast cancer, and found that EMP1 expression was positively correlated with stromal and microenvironmental scores. Infiltration analysis and immunohistochemical (IHC) staining of serial sections confirmed the critical role of EMP1 in cancer-associated fibroblast (CAF) infiltration. Cell co-culture assays, xenograft tumor experiments, loss-of-function, gain-of-function, RNA sequencing studies, and rescue assays were performed to confirm the role of EMP1 in CAF infiltration in vitro and in vivo. These findings revealed that EMP1 depletion in TNBC cells resulted in considerable inhibition of CAF infiltration in vivo and in vitro. Mechanistically, EMP1 knockdown induced a substantial decrease in IL6 secretion from TNBC through the NF-κB signaling pathway, hindering CAF proliferation and subsequently inhibiting TNBC progression and metastasis. These cumulative results indicate that EMP1 functions as an oncogene in TNBC by mediating the cell communication of TNBC and CAFs. Targeted inhibition of EMP1 by suppressing CAF infiltration is a promising strategy for TNBC treatment.
Collapse
Affiliation(s)
- Qi Wang
- Research Center for High-Altitude Medicine, Key Laboratory of High-Altitude Medicine, Ministry of Education, Laboratory for High Altitude Medicine of Qinghai Province, Key Laboratory of Application and Foundation for High Altitude Medicine Research in Qinghai Province (Qinghai-Utah Joint Research Key Lab for High-Altitude Medicine), Qinghai University, Xining, China
- Department of Pathology, Renmin Hospital, Hubei University of Medicine, Shiyan, Hubei, China
| | - Dandan Li
- Department of Pathology, Renmin Hospital, Hubei University of Medicine, Shiyan, Hubei, China
- Shiyan Key Laboratory of Comprehensive Prevention and Treatment of Oral Cancer, Department of Stomatology, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei, China
| | - Haixiu Ma
- Research Center for High-Altitude Medicine, Key Laboratory of High-Altitude Medicine, Ministry of Education, Laboratory for High Altitude Medicine of Qinghai Province, Key Laboratory of Application and Foundation for High Altitude Medicine Research in Qinghai Province (Qinghai-Utah Joint Research Key Lab for High-Altitude Medicine), Qinghai University, Xining, China
| | - Zengyan Li
- Experimental Animal Center & Institute of Clinical Medicine, Renmin Hospital, Hubei University of Medicine, Shiyan, Hubei, China
| | - Juan Wu
- Research Center for High-Altitude Medicine, Key Laboratory of High-Altitude Medicine, Ministry of Education, Laboratory for High Altitude Medicine of Qinghai Province, Key Laboratory of Application and Foundation for High Altitude Medicine Research in Qinghai Province (Qinghai-Utah Joint Research Key Lab for High-Altitude Medicine), Qinghai University, Xining, China
| | - Jinwan Qiao
- Research Center for High-Altitude Medicine, Key Laboratory of High-Altitude Medicine, Ministry of Education, Laboratory for High Altitude Medicine of Qinghai Province, Key Laboratory of Application and Foundation for High Altitude Medicine Research in Qinghai Province (Qinghai-Utah Joint Research Key Lab for High-Altitude Medicine), Qinghai University, Xining, China
| | - Jun Liu
- Research Center for High-Altitude Medicine, Key Laboratory of High-Altitude Medicine, Ministry of Education, Laboratory for High Altitude Medicine of Qinghai Province, Key Laboratory of Application and Foundation for High Altitude Medicine Research in Qinghai Province (Qinghai-Utah Joint Research Key Lab for High-Altitude Medicine), Qinghai University, Xining, China
| | - Jing Zhao
- Research Center for High-Altitude Medicine, Key Laboratory of High-Altitude Medicine, Ministry of Education, Laboratory for High Altitude Medicine of Qinghai Province, Key Laboratory of Application and Foundation for High Altitude Medicine Research in Qinghai Province (Qinghai-Utah Joint Research Key Lab for High-Altitude Medicine), Qinghai University, Xining, China
| | - Ronghua Ma
- Research Center for High-Altitude Medicine, Key Laboratory of High-Altitude Medicine, Ministry of Education, Laboratory for High Altitude Medicine of Qinghai Province, Key Laboratory of Application and Foundation for High Altitude Medicine Research in Qinghai Province (Qinghai-Utah Joint Research Key Lab for High-Altitude Medicine), Qinghai University, Xining, China
| | - Lin Tian
- Department of Pathology, Renmin Hospital, Hubei University of Medicine, Shiyan, Hubei, China
| | - Lei Zhang
- Department of Pathology, Renmin Hospital, Hubei University of Medicine, Shiyan, Hubei, China
| | - Jianye Yang
- Experimental Animal Center & Institute of Clinical Medicine, Renmin Hospital, Hubei University of Medicine, Shiyan, Hubei, China
| | - Jianing Wang
- Experimental Animal Center & Institute of Clinical Medicine, Renmin Hospital, Hubei University of Medicine, Shiyan, Hubei, China.
| | - Shanshan Qin
- Department of Pathology, Renmin Hospital, Hubei University of Medicine, Shiyan, Hubei, China.
- Shiyan Key Laboratory of Comprehensive Prevention and Treatment of Oral Cancer, Department of Stomatology, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei, China.
| | - Zhanhai Su
- Research Center for High-Altitude Medicine, Key Laboratory of High-Altitude Medicine, Ministry of Education, Laboratory for High Altitude Medicine of Qinghai Province, Key Laboratory of Application and Foundation for High Altitude Medicine Research in Qinghai Province (Qinghai-Utah Joint Research Key Lab for High-Altitude Medicine), Qinghai University, Xining, China.
| |
Collapse
|
6
|
Tang JY, Zhao ML, Zhou XM, Chai YQ, Yuan R, Lei YM, Zhuo Y. Engineering DNA Nanodevices with Multi-site Recognition and Multi-signal Output for Accurate Intracellular LncRNA Imaging. Anal Chem 2025; 97:3378-3386. [PMID: 39907677 DOI: 10.1021/acs.analchem.4c05353] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2025]
Abstract
Dynamic DNA nanodevices, known for their high programmability and controllability, are pivotal in intracellular biomarker imaging. However, these nanodevices often suffer from inadequate detection sensitivity and specificity due to limited cellular loading capacity and low signal feedback. Herein, we engineered an integrated multi-site recognition and multi-signal output of four-leaf clover dynamic DNA nanodevice (MEMORY) that enables sensitive and accurate intracellular long noncoding RNA (lncRNA) imaging. MEMORY features one fluorophore (FAM)-modified cross-shaped structure as spatial-confinement scaffolds loaded with four identical quenchers (BHQ1)-modified recognition probes (RPs), ensuring a low background signal initially. In the presence of target lncRNA, the multiple recognition sites of MEMORY facilitate hybridization with the target to selectively release the RPs, exposing the toehold region and outputting the green fluorescence (FAM) signal. Furthermore, the exposed toehold region can trigger efficient and rapid hybridization chain reaction (HCR) amplification, outputting the red fluorescence (Cy5) signal. MEMORY's multiple recognition sites increase the likelihood of target collisions, enhancing reaction efficiency, while its multi-signal output provides sequential feedback through FAM and Cy5, boosting overall signal intensity. With the lncRNA metastasis-related lung adenocarcinoma transcript 1 (MALAT1) as a detection model, MEMORY offers a linear detection range from 1 pM to 100 nM, with a limit of detection of 0.29 pM. We demonstrated that MEMORY can differentiate between normal and tumor cells based on intracellular MALAT1 imaging. This integrated DNA nanodevice will offer valuable tools for sensitive and accurate imaging of intracellular biomarkers.
Collapse
Affiliation(s)
- Jing-Yi Tang
- Key Laboratory of Luminescence Analysis and Molecular Sensing, Ministry of Education, Institute of Developmental Biology and Regenerative Medicine, College of Chemistry and Chemical Engineering, Southwest University, Chongqing 400715, China
| | - Mei-Ling Zhao
- Key Laboratory of Luminescence Analysis and Molecular Sensing, Ministry of Education, Institute of Developmental Biology and Regenerative Medicine, College of Chemistry and Chemical Engineering, Southwest University, Chongqing 400715, China
| | - Xue-Mei Zhou
- Key Laboratory of Luminescence Analysis and Molecular Sensing, Ministry of Education, Institute of Developmental Biology and Regenerative Medicine, College of Chemistry and Chemical Engineering, Southwest University, Chongqing 400715, China
| | - Ya-Qin Chai
- Key Laboratory of Luminescence Analysis and Molecular Sensing, Ministry of Education, Institute of Developmental Biology and Regenerative Medicine, College of Chemistry and Chemical Engineering, Southwest University, Chongqing 400715, China
| | - Ruo Yuan
- Key Laboratory of Luminescence Analysis and Molecular Sensing, Ministry of Education, Institute of Developmental Biology and Regenerative Medicine, College of Chemistry and Chemical Engineering, Southwest University, Chongqing 400715, China
| | - Yan-Mei Lei
- Key Laboratory of Luminescence Analysis and Molecular Sensing, Ministry of Education, Institute of Developmental Biology and Regenerative Medicine, College of Chemistry and Chemical Engineering, Southwest University, Chongqing 400715, China
| | - Ying Zhuo
- Key Laboratory of Luminescence Analysis and Molecular Sensing, Ministry of Education, Institute of Developmental Biology and Regenerative Medicine, College of Chemistry and Chemical Engineering, Southwest University, Chongqing 400715, China
| |
Collapse
|
7
|
Wu Y, Wang D, Zhao J, Guo J, Gao Z, Xu Q, Hu X. NRAV promoted the malignant progression of gastric cancer. Gene 2025; 937:149134. [PMID: 39615808 DOI: 10.1016/j.gene.2024.149134] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Revised: 11/21/2024] [Accepted: 11/27/2024] [Indexed: 12/06/2024]
Abstract
Gastric cancer (GC) has been ranked as the third incidence tumors globally. Long non-coding RNA (lncRNA) NRAV has been reported as a tumor-enhancer in the development of human cancers, whereas the function of NRAV in GC remains to be elucidated. The aim of this research was to explore the underlying function of NRAV in GC. Through comprehensive bioinformatics analysis, a significantly elevation of NRAV was found in both human GC tissues and cell lines, which indicated the poor prognosis of GC patients. Then, we conducted a series of functional experiments to illustrate the role of NRAV in GC. The results showed that the down-regulation of NRAV exhibited a significant inhibitory effect on GC cell proliferation and migration, while NRAV overexpression promoted GC cell proliferation and migration. Through xenograft mouse tumor model, the suppression of NRAV led to a reduction in the growth of tumor mice, whereas overexpression of NRAV notably enhanced tumor growth. Finally, EFHC1 was revealed as the potential target gene of NRAV. Overall, our findings indicated the promising application of NRAV as a therapeutic target and prognostic biomarker for GC.
Collapse
Affiliation(s)
- Yuchen Wu
- The Key Laboratory of Tumor Molecular Diagnosis and Individualized Medicine of Zhejiang Province, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou 310014, China; Bengbu Medical University, Bengbu 233030, China
| | | | - Jie Zhao
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou 310014, China
| | - Jinhui Guo
- Qingdao Medical College, Qingdao University, Qingdao 266071, China
| | - Zhenyu Gao
- Wenzhou Medical University, Wenzhou 325035, China
| | - Qiuran Xu
- The Key Laboratory of Tumor Molecular Diagnosis and Individualized Medicine of Zhejiang Province, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou 310014, China.
| | - Xiaoge Hu
- The Key Laboratory of Tumor Molecular Diagnosis and Individualized Medicine of Zhejiang Province, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou 310014, China.
| |
Collapse
|
8
|
Fu H, Wang Q, Li H, Li H, Li J, Liu Y, Dang F, Wang L, Zhang X, Yang Y, Du Y. LINC02987 suppression hepatocellular carcinoma progression by modulating autophagy via the miR-338-3p/ATG12 axis. Exp Cell Res 2025; 444:114398. [PMID: 39746597 DOI: 10.1016/j.yexcr.2024.114398] [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: 10/13/2024] [Revised: 12/06/2024] [Accepted: 12/25/2024] [Indexed: 01/04/2025]
Abstract
Hepatocellular carcinoma (HCC), the most common primary liver cancer, is marked by a high mortality rate, with the misregulation of long non-coding RNAs (LncRNAs) playing a key role in its development. Here, we studied the role of LINC02987 in HCC. We employed bioinformatics tools to identify LncRNAs and miRNAs that exhibit differential expression in HCC. Quantitative real-time reverse transcription PCR (RT-qPCR) and Western blot analysis were utilized to quantify gene and protein expression levels. The interaction between miR-338-3p and LINC02987 or ATG12 was confirmed through dual-luciferase reporter and RNA immunoprecipitation (RIP) assays. We observed that LINC02987 was overexpressed in HCC tumor tissues and cell lines. Silencing of LINC02987 led to a reduction in cell viability, diminished clonogenic potential, and attenuated invasive and migratory capabilities. Also, decreasing protein level and fluorescence intensity of the autophagy-associated LC3 I/II. In HCC, miR-338-3p expression was downregulated, while inversely correlates with the overexpression of the autophagy protein ATG12. Mimicking miR-338-3p suppresses the activity of both LINC02987 and ATG12, as evidenced by reduced luciferase signals in corresponding reporter assays. Mimicking miR-338-3p suppresses the activity of both LINC02987 and ATG12, as evidenced by reduced luciferase signals in reporter assays. Transfection with si-LINC02987 decreased ATG12 expression, an effect that was partially reversed by miR-338-3p knockdown. Inhibition of miR-338-3p or overexpression of ATG12 increased LC3 I/II protein levels. Our results indicate that LINC02987 sequesters miR-338-3p, leading to increased ATG12 and promoting autophagy in HCC cells. These results highlight the potential of LINC02987 as a therapeutic target for the treatment of HCC.
Collapse
Affiliation(s)
- Haiyan Fu
- Oncology Department the Third People's Hospital of Kunming Sixth Affiliated Hospital of Dali University, No.319 Wujing Road, Guandu Area, 650000, China; Yunnan Infectious Disease Clinical Medical Center, China.
| | - Qiuhong Wang
- Hepatobiliary and Pancreatic Surgery Department the Second Affiliated Hospital of Kunming Medical University, No. 374 Dianmian Avenue Wuhua Area, Kunming, 650101, China.
| | - Haiwen Li
- Oncology Department the Third People's Hospital of Kunming Sixth Affiliated Hospital of Dali University, No.319 Wujing Road, Guandu Area, 650000, China; Yunnan Infectious Disease Clinical Medical Center, China.
| | - Hongjuan Li
- Oncology Department the Third People's Hospital of Kunming Sixth Affiliated Hospital of Dali University, No.319 Wujing Road, Guandu Area, 650000, China; Yunnan Infectious Disease Clinical Medical Center, China.
| | - Jie Li
- Oncology Department the Third People's Hospital of Kunming Sixth Affiliated Hospital of Dali University, No.319 Wujing Road, Guandu Area, 650000, China; Yunnan Infectious Disease Clinical Medical Center, China.
| | - Yu Liu
- Department of Gastroenterology, Kunming Ganmei Hospital, No.504 Qingnian Road Xishan Area, 650100, China.
| | - Futao Dang
- Oncology Department the Third People's Hospital of Kunming Sixth Affiliated Hospital of Dali University, No.319 Wujing Road, Guandu Area, 650000, China; Yunnan Infectious Disease Clinical Medical Center, China.
| | - Lifeng Wang
- Oncology Department the Third People's Hospital of Kunming Sixth Affiliated Hospital of Dali University, No.319 Wujing Road, Guandu Area, 650000, China.
| | - Xuan Zhang
- Oncology Department the Third People's Hospital of Kunming Sixth Affiliated Hospital of Dali University, No.319 Wujing Road, Guandu Area, 650000, China.
| | - Yongrui Yang
- Oncology Department the Third People's Hospital of Kunming Sixth Affiliated Hospital of Dali University, No.319 Wujing Road, Guandu Area, 650000, China; Yunnan Infectious Disease Clinical Medical Center, China.
| | - Yingrong Du
- Oncology Department the Third People's Hospital of Kunming Sixth Affiliated Hospital of Dali University, No.319 Wujing Road, Guandu Area, 650000, China; Yunnan Infectious Disease Clinical Medical Center, China.
| |
Collapse
|
9
|
Xia L, Wang H, Du G, Cheng X, Zhang R, Yu H, Cheng M, Chen Y, Qin S, Leng W. Receptor accessory protein 6, a novel ferroptosis suppressor, drives oral squamous cell carcinoma by maintaining endoplasmic reticulum hemostasis. Int J Biol Macromol 2024; 283:137565. [PMID: 39566754 DOI: 10.1016/j.ijbiomac.2024.137565] [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: 08/13/2024] [Revised: 11/09/2024] [Accepted: 11/10/2024] [Indexed: 11/22/2024]
Abstract
Increasing evidence suggests a close association between endoplasmic-reticulum (ER) stress and ferroptosis. Receptor accessory protein 6 (REEP6) is known to play a crucial role in maintaining ER homeostasis. However, its involvement in ferroptosis remains unknown. In this study, we found that REEP6 was overexpressed, and its overexpression showed a significant association with tumor size and poor survival in OSCC patients. Besides, in vitro and in vivo assays together showed that REEP6 plays an oncogenic role in OSCC progression. The GO/KEGG, and GSEA analysis showed that REEP6 overexpression leads to the inactivation of ferroptosis signaling in OSCC. Moreover, REEP6 overexpression conferred resistance to RSL3, a ferroptosis inducer, whereas REEP6 knockdown sensitized OSCC cells to RSL3. Overexpression of REEP6 decrease the accumulation of iron ions, ROS production, but increase the number of mitochondrial cristae in OSCC cells. More importantly, we confirmed that REEP6 inhibited ferroptosis in OSCC cells by maintaining ER homeostasis via regulating ACSL4 expression. In addition, we identified promoter DNA hypomethylation as the underlying cause of REEP6 overexpression in OSCC. Taken together, REEP6 acts as a novel suppressor of ferroptosis, with its overexpression driven by promoter hypomethylation contributing to OSCC progression by ER stress-mediated ferroptosis via ACSL4.
Collapse
Affiliation(s)
- Lingyun Xia
- Department of Stomatology, Taihe Hospital, Hubei University of Medicine, Shiyan 442000, China; Institute of Oral Diseases, School of Dentistry, Hubei University of Medicine, Shiyan 442000, China
| | - Hongbing Wang
- Department of Stomatology, Taihe Hospital, Hubei University of Medicine, Shiyan 442000, China; Institute of Oral Diseases, School of Dentistry, Hubei University of Medicine, Shiyan 442000, China
| | - Gao Du
- Department of Stomatology, Taihe Hospital, Hubei University of Medicine, Shiyan 442000, China; Institute of Oral Diseases, School of Dentistry, Hubei University of Medicine, Shiyan 442000, China
| | - Xiaobo Cheng
- Department of Stomatology, Taihe Hospital, Hubei University of Medicine, Shiyan 442000, China; Institute of Oral Diseases, School of Dentistry, Hubei University of Medicine, Shiyan 442000, China
| | - Rui Zhang
- Department of Stomatology, Taihe Hospital, Hubei University of Medicine, Shiyan 442000, China; Institute of Oral Diseases, School of Dentistry, Hubei University of Medicine, Shiyan 442000, China
| | - Hedong Yu
- Department of Stomatology, Taihe Hospital, Hubei University of Medicine, Shiyan 442000, China; Institute of Oral Diseases, School of Dentistry, Hubei University of Medicine, Shiyan 442000, China
| | - Mumo Cheng
- Department of General Practice, Shanghai Baoshan District Wusong Central Hospital (Zhongshan Hospital Wusong Branch, Fudan University), Shanghai 200940, China
| | - Yongji Chen
- Department of Stomatology, Taihe Hospital, Hubei University of Medicine, Shiyan 442000, China; Institute of Oral Diseases, School of Dentistry, Hubei University of Medicine, Shiyan 442000, China.
| | - Shanshan Qin
- Department of Stomatology, Taihe Hospital, Hubei University of Medicine, Shiyan 442000, China; Institute of Oral Diseases, School of Dentistry, Hubei University of Medicine, Shiyan 442000, China.
| | - Weidong Leng
- Department of Stomatology, Taihe Hospital, Hubei University of Medicine, Shiyan 442000, China; Institute of Oral Diseases, School of Dentistry, Hubei University of Medicine, Shiyan 442000, China.
| |
Collapse
|
10
|
Zhang J, Zhu H, Li L, Gao Y, Yu B, Ma G, Jin X, Sun Y. New mechanism of LncRNA: In addition to act as a ceRNA. Noncoding RNA Res 2024; 9:1050-1060. [PMID: 39022688 PMCID: PMC11254507 DOI: 10.1016/j.ncrna.2024.06.002] [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: 04/07/2024] [Revised: 05/20/2024] [Accepted: 06/03/2024] [Indexed: 07/20/2024] Open
Abstract
Long non-coding RNAs (LncRNAs) are a class of RNA molecules with nucleic acid lengths ranging from 200 bp to 100 kb that cannot code for proteins, which are diverse and widely expressed in both animals and plants. Scholars have found that lncRNAs can regulate human physiological processes at the gene and protein levels, mainly through the regulation of epigenetic, transcriptional and post-transcriptional levels of genes and proteins, as well as in the immune response by regulating the expression of immune cells and inflammatory factors, and thus participate in the occurrence and development of a variety of diseases. From the downstream targets of lncRNAs, we summarize the new research progress of lncRNA mechanisms other than miRNA sponges in recent years, aiming to provide new ideas and directions for the study of lncRNA mechanisms.
Collapse
Affiliation(s)
- Jiahao Zhang
- Department of Toxicology, School of Public Health, Lanzhou University, Lanzhou, 730000, China
- Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou 730000, China
| | - Huike Zhu
- Department of Toxicology, School of Public Health, Lanzhou University, Lanzhou, 730000, China
| | - Linjing Li
- Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou 730000, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Yuting Gao
- Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou 730000, China
- College of Life Sciences, Northwest Normal University, Gansu Province, Lanzhou, 730070, China
| | - Boyi Yu
- Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou 730000, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Guorong Ma
- The First Clinical Medical College of Gansu University of Chinese Medicine Gansu Provincial Hospital, Lanzhou, 730000, China
| | - Xiaodong Jin
- Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou 730000, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Yingbiao Sun
- Department of Toxicology, School of Public Health, Lanzhou University, Lanzhou, 730000, China
| |
Collapse
|
11
|
Liu SS, Wang JK, Liu MS, Guo DF, Wen Q, Liang YH, Wang T, Zhang KH. ILF2 protein is a promising serum biomarker for early detection of gastric cancer. BMC Cancer 2024; 24:1447. [PMID: 39587551 PMCID: PMC11587746 DOI: 10.1186/s12885-024-13205-6] [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: 08/26/2024] [Accepted: 11/14/2024] [Indexed: 11/27/2024] Open
Abstract
BACKGROUND Our previous small-sample study indicated that serum levels of interleukin enhancer binding factor 2 (ILF2) may have the potential for gastric cancer (GC) detection. The present study was conducted to further validate the diagnostic value of serum ILF2 protein for GC. METHODS Serum specimens and clinical data were collected from patients with GC (n = 99) or benign gastric disease (BGD) (n = 49) and healthy controls (HC) (n = 51). Serum ILF2 levels were measured using enzyme-linked immunosorbent assay. The diagnostic performance of ILF2 was evaluated using the area under the receiver operating characteristic curve (AUC). The independence and synergy of ILF2 in GC diagnosis were analyzed by modeling with conventional blood indicators. RESULTS The median serum ILF2 level was higher in the GC group (227.8ng/mL) than in the BGD group (72.0ng/mL) and the HC group (56.8ng/mL) (p < 0.001), and no significant difference across GC subgroups. The AUCs of ILF2 were 0.915 (95%CI 0.873-0.957) for GC vs. HC, 0.854 (95%CI 0.793-0.915) for GC vs. BGD, 0.885 (95%CI 0.841-0.929) for GC vs. BGD + HC, and 0.888 (95% CI 0.830-0.945) for TNM I stage GC vs. BGD + HC, outperforming conventional blood indicators (corresponding AUCs ranging from 0.641 to 0.782). ILF2 was independent of and synergistic with conventional blood indicators in GC diagnosis, and a simple diagnostic model based on ILF2 and red blood cell count improved the diagnostic performance, with positive rates of approximately 90% in various subgroups of GC. CONCLUSIONS Serum ILF2 protein is a novel and potential serum biomarker for the detection of GC, especially for early GC.
Collapse
Affiliation(s)
- Shao-Song Liu
- Department of Gastroenterology, Jiangxi Provincial Key Laboratory of Digestive Diseases, Jiangxi Clinical Research Center for Gastroenterology, Digestive Disease Hospital, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, No 17, Yongwai Zheng Street, Nanchang, Jiangxi, 330006, China
| | - Jin-Ke Wang
- Department of Gastroenterology, Jiangxi Provincial Key Laboratory of Digestive Diseases, Jiangxi Clinical Research Center for Gastroenterology, Digestive Disease Hospital, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, No 17, Yongwai Zheng Street, Nanchang, Jiangxi, 330006, China
| | - Mao-Sheng Liu
- Department of Gastroenterology, Jiangxi Provincial Key Laboratory of Digestive Diseases, Jiangxi Clinical Research Center for Gastroenterology, Digestive Disease Hospital, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, No 17, Yongwai Zheng Street, Nanchang, Jiangxi, 330006, China
| | - Ding-Fan Guo
- Department of Gastroenterology, Jiangxi Provincial Key Laboratory of Digestive Diseases, Jiangxi Clinical Research Center for Gastroenterology, Digestive Disease Hospital, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, No 17, Yongwai Zheng Street, Nanchang, Jiangxi, 330006, China
| | - Qi Wen
- Department of Gastroenterology, Jiangxi Provincial Key Laboratory of Digestive Diseases, Jiangxi Clinical Research Center for Gastroenterology, Digestive Disease Hospital, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, No 17, Yongwai Zheng Street, Nanchang, Jiangxi, 330006, China
| | - Yun-Hui Liang
- Department of Gastroenterology, Jiangxi Provincial Key Laboratory of Digestive Diseases, Jiangxi Clinical Research Center for Gastroenterology, Digestive Disease Hospital, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, No 17, Yongwai Zheng Street, Nanchang, Jiangxi, 330006, China
| | - Ting Wang
- Department of Gastroenterology, Jiangxi Provincial Key Laboratory of Digestive Diseases, Jiangxi Clinical Research Center for Gastroenterology, Digestive Disease Hospital, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, No 17, Yongwai Zheng Street, Nanchang, Jiangxi, 330006, China.
| | - Kun-He Zhang
- Department of Gastroenterology, Jiangxi Provincial Key Laboratory of Digestive Diseases, Jiangxi Clinical Research Center for Gastroenterology, Digestive Disease Hospital, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, No 17, Yongwai Zheng Street, Nanchang, Jiangxi, 330006, China.
| |
Collapse
|
12
|
Wang Y, Shen K, Cheng Q, Zhou X, Liu K, Xiao J, Hu L. The long noncoding RNA ELFN1-AS1 promotes gastric cancer growth and metastasis by interacting with TAOK1 to inhibit the Hippo signaling pathway. Cell Death Discov 2024; 10:465. [PMID: 39528458 PMCID: PMC11555383 DOI: 10.1038/s41420-024-02235-5] [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: 10/26/2023] [Revised: 10/26/2024] [Accepted: 11/01/2024] [Indexed: 11/16/2024] Open
Abstract
Gastric cancer (GC) is a common digestive malignancy that causes numerous cancer-related deaths. Long noncoding RNAs (lncRNAs) play a crucial role in the development of various tumors, including GC. In this study, we revealed that ELFN1-AS1, a lncRNA with aberrantly high expression, contributes to the proliferation and metastasis of GC. Mechanically, ELFN1-AS1 plays an oncogenic role by binding to the protein kinase domain of thousand and one amino acid protein kinase (TAOK1), a tumor suppressor in GC, and disrupting the TAOK1-STK3 interaction, leading to decreased STK3 phosphorylation. This decrease is accompanied by attenuation of the Hippo kinase cascade, resulting in reduced YAP1 phosphorylation, a crucial effector of the Hippo signaling pathway. Subsequently, the reduced YAP1 phosphorylation promotes its nuclear translocation, thereby enhancing the expression of MYC, a downstream target of the pathway and well-known oncogene. Taken together, the ELFN1-AS1/TAOK1/STK3/YAP1 axis may promote GC progression and is a promising target for GC treatment.
Collapse
Affiliation(s)
- Yuanhang Wang
- Department of General Surgery, Yancheng Third People's Hospital, Affiliated Yancheng Hospital, School of Medicine, Southeast University, Yancheng, Jiangsu Province, China.
- Department of General Surgery, the First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China.
| | - Kuan Shen
- Department of General Surgery, the First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
- Department of General Surgery, Liyang People's Hospital, Liyang Branch Hospital of Jiangsu Province Hospital, Liyang, Jiangsu Province, China
| | - Quan Cheng
- Department of General Surgery, the First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Xinyi Zhou
- Department of General Surgery, the First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Kanghui Liu
- Department of General Surgery, the First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Jian Xiao
- Department of General Surgery, the First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Li Hu
- Department of General Surgery, the First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| |
Collapse
|
13
|
Cao H, Wang Z, Guo Q, Qin S, Li D. MIR194-2HG, a miRNA host gene activated by HNF4A, inhibits gastric cancer by regulating microRNA biogenesis. Biol Direct 2024; 19:95. [PMID: 39425187 PMCID: PMC11487860 DOI: 10.1186/s13062-024-00549-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: 05/23/2024] [Accepted: 10/14/2024] [Indexed: 10/21/2024] Open
Abstract
BACKGROUND MicroRNA host gene (MIRHG) lncRNA is a particular lncRNA subclass that can perform both typical and atypical lncRNA functions. The biological function of MIRHG lncRNA MIR194-2HG in cancer is poorly understood. METHODS Loss-of-function studies were performed in vivo and in vitro to reveal the biological function of MIR194-2HG in GC. MicroRNA PCR array, northern blotting, RNA sequencing, chromatin immunoprecipitation, and rescue assays were conducted to uncover the molecular mechanism of MIR194-2HG. RESULTS In this study, we reported an atypical lncRNA function of MIR194-2HG in GC. MIR194-2HG downregulation was clinically associated with malignant progression and poor prognosis in GC. Functional assays confirmed that MIR194-2HG knockdown significantly promoted GC proliferation and metastasis in vitro and in vivo. Mechanismically, MIR194-2HG was required for the biogenesis of miR-194 and miR-192, which were reported to be tumor-suppressor genes in GC. Moreover, hepatocyte nuclear factor HNF4A directly activated the transcription of MIR194-2HG and its derived miR-194 and miR-192. Meanwhile, BTF3L4 was proved to be a common target gene of miR-192 and miR-194. Rescue assay further confirmed that MIR194-2HG knockdown promotes GC progression through maintaining BTF3L4 overexpression in a miR-194/192-dependent manner. CONCLUSION The dysregulated MIR194-2HG/BTF3L4 axis is responsible for GC progression. Targeting HNF4A to inhibit miR-192/194 expression may be a promising strategy for overcoming GC.
Collapse
Affiliation(s)
- Hong Cao
- Department of Orthopaedic Surgery, Renmin Hospital, Hubei University of Medicine, Shiyan, China
| | - Zidi Wang
- Hubei Key Laboratory of Embryonic Stem Cell Research, School of Basic Medical Sciences, Hubei University of Medicine, Shiyan, Hubei, 442000, China
| | - Qiwei Guo
- Hubei Key Laboratory of Embryonic Stem Cell Research, School of Basic Medical Sciences, Hubei University of Medicine, Shiyan, Hubei, 442000, China
| | - Shanshan Qin
- Hubei Key Laboratory of Embryonic Stem Cell Research, School of Basic Medical Sciences, Hubei University of Medicine, Shiyan, Hubei, 442000, China.
- Laboratory of Tumor Biology, Academy of Bio-Medicine Research, Hubei University of Medicine, Shiyan, Hubei, P.R. China.
- Shiyan Key Laboratory of Natural Medicine Nanoformulation Research, Hubei University of Medicine, Shiyan, Hubei, 442000, China.
| | - Dandan Li
- Department of Orthopaedic Surgery, Renmin Hospital, Hubei University of Medicine, Shiyan, China.
- Hubei Key Laboratory of Embryonic Stem Cell Research, School of Basic Medical Sciences, Hubei University of Medicine, Shiyan, Hubei, 442000, China.
- Laboratory of Tumor Biology, Academy of Bio-Medicine Research, Hubei University of Medicine, Shiyan, Hubei, P.R. China.
- Shiyan Key Laboratory of Natural Medicine Nanoformulation Research, Hubei University of Medicine, Shiyan, Hubei, 442000, China.
| |
Collapse
|
14
|
Werner A, Kanhere A, Wahlestedt C, Mattick JS. Natural antisense transcripts as versatile regulators of gene expression. Nat Rev Genet 2024; 25:730-744. [PMID: 38632496 DOI: 10.1038/s41576-024-00723-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/07/2024] [Indexed: 04/19/2024]
Abstract
Long non-coding RNAs (lncRNAs) are emerging as a major class of gene products that have central roles in cell and developmental biology. Natural antisense transcripts (NATs) are an important subset of lncRNAs that are expressed from the opposite strand of protein-coding and non-coding genes and are a genome-wide phenomenon in both eukaryotes and prokaryotes. In eukaryotes, a myriad of NATs participate in regulatory pathways that affect expression of their cognate sense genes. Recent developments in the study of NATs and lncRNAs and large-scale sequencing and bioinformatics projects suggest that whether NATs regulate expression, splicing, stability or translation of the sense transcript is influenced by the pattern and degrees of overlap between the sense-antisense pair. Moreover, epigenetic gene regulatory mechanisms prevail in somatic cells whereas mechanisms dependent on the formation of double-stranded RNA intermediates are prevalent in germ cells. The modulating effects of NATs on sense transcript expression make NATs rational targets for therapeutic interventions.
Collapse
Affiliation(s)
| | | | | | - John S Mattick
- University of New South Wales, Sydney, New South Wales, Australia
| |
Collapse
|
15
|
Cui N, Ding F. Co-Expression Network Analysis and Molecular Docking Demonstrate That Diosgenin Inhibits Gastric Cancer Progression via SLC1A5/mTORC1 Pathway. Drug Des Devel Ther 2024; 18:3157-3173. [PMID: 39071813 PMCID: PMC11283265 DOI: 10.2147/dddt.s458613] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Accepted: 07/10/2024] [Indexed: 07/30/2024] Open
Abstract
Background Tumor-Node-Metastasis (TNM) stage of gastric cancer (GC) is one of the main factors affecting clinical outcome. The aim of this study was to explore the targets related to TNM stage of GC, and screening natural bioactive drug. Methods RNA sequencing data of the TCGA-STAD cohort were downloaded from UCSC database. Genes associated with TNM staging were identified by weighted gene co-expression network analysis (WGCNA). Univariate Cox regression, least absolute shrinkage and selection operator (LASSO), extreme gradient boosting (Xgboost), random forest (RF) and cytohubba plug-in of cytoscope were applied to screen hub genes. Natural bioactive ingredients were available from the HERB database. Molecular docking was used to evaluate the binding activity of active ingredients to the hub protein. CCK-8, flow cytometry, transwell and Western blot assays were used to analyze the effects of diosgenin on GC cells. Results 898 TNM-related genes were screened out through WGCNA. Three genes associated with GC progression/prognosis were identified, including nuclear receptor subfamily 3 group C member 2 (NR3C2), solute carrier family 1 member 5 (SLC1A5) and FAT atypical cadherin 1 (FAT1) based on the machine learning algorithms and hub co-expression network analysis. Diosgenin had good binding activity with SLC1A5. SLC1A5 was highly expressed in GC and was closely associated with tumor stage, overall survival and immune infiltration of GC patients. Diosgenin could inhibit cell viability and invasive ability, promote apoptosis and induce cell cycle arrest in G0/G1 phase. In addition, diosgenin promoted cleaved caspase 3 expression and inhibited Ki67, cyclin D1, p-S6K1, and SLC1A5 expression levels, while the mTORC1 activator (MHY1485) reversed this phenomenon. Conclusion For the first time, this work reports diosgenin may inhibit the activation of mTORC1 signaling through targeting SLC1A5, thereby inhibiting the malignant behaviors of GC cells.
Collapse
Affiliation(s)
- Ning Cui
- Department of Gastroenterology, Renmin Hospital of Wuhan University, Wuhan, People’s Republic of China
| | - Feng Ding
- Department of Gastroenterology, Zhongnan Hospital of Wuhan University, Wuhan, People’s Republic of China
| |
Collapse
|
16
|
Zhu J, Xu HN, Lin T, Xia ZJ. Silencing of cysteine and serine rich nuclear protein 1 inhibits apoptosis, senescence and collagen degradation in human-derived vaginal fibroblasts in response to oxidative stress or DNA damage. Exp Cell Res 2024; 440:114139. [PMID: 38908423 DOI: 10.1016/j.yexcr.2024.114139] [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: 05/24/2024] [Accepted: 06/19/2024] [Indexed: 06/24/2024]
Abstract
Pelvic organ prolapse (POP) is a group of diseases caused by extracellular matrix (ECM) degradation in pelvic supportive tissues. Cysteine and serine rich nuclear protein 1 (CSRNP1) is involved in cell proliferation and survival regulation, and reportedly facilitates collagen breakdown in human chondrocytes. The present study aimed to probe the effect of CSRNP1 on collagen metabolism in human-derived vaginal fibroblasts. High expression of CSRNP1 was found in POP patient-derived vaginal fibroblasts in comparison to normal-derived vaginal fibroblasts. Following functional experiments revealed that CSRNP1 overexpression led to proliferation inhibition, apoptosis and collagen degradation in normal vaginal fibroblasts. In line with this, silencing of CSRNP1 inhibited hydrogen peroxide (H2O2)-triggered apoptosis, ROS generation and collagen loss in normal vaginal fibroblasts. Silencing of CSRNP1 also reduced the expression of cell senescence markers p21 and γ-H2Ax (the histone H2Ax phosphorylated at Ser139), as well as curbed collagen breakdown in normal vaginal fibroblasts caused by a DNA damage agent etoposide. Transcriptomic analysis of vaginal fibroblasts showed that differentially expressed genes affected by CSRNP1 overexpression were mainly enriched in the Wnt signaling pathway. Treatment with a Wnt pathway inhibitor DKK1 blocked CSRNP1 knockdown-caused collagen deposition. Mechanistically, CSRNP1 was identified to be a target of Snail family transcriptional repressor 2 (SNAI2). Forced expression of CSRNP1 reversed the anti-apoptotic, anti-senescent and anti-collagen loss effects of SNAI2 in normal vaginal fibroblasts exposed to H2O2 or etoposide. Our study indicates that the SNAI2/CSRNP1 axis may be a key driver in POP progression, which provides a potential therapeutic strategy for POP.
Collapse
Affiliation(s)
- Jing Zhu
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, Liaoning, People's Republic of China; Department of Obstetrics and Gynecology, Maternity & Child care Center of Qinhuangdao, Qinhuangdao, Hebei, People's Republic of China
| | - Hai-Nan Xu
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, Liaoning, People's Republic of China
| | - Te Lin
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, Liaoning, People's Republic of China
| | - Zhi-Jun Xia
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, Liaoning, People's Republic of China.
| |
Collapse
|
17
|
Qin S, Liu Y, Zhang X, Huang P, Xia L, Leng W, Li D. lncRNA FGD5-AS1 is required for gastric cancer proliferation by inhibiting cell senescence and ROS production via stabilizing YBX1. J Exp Clin Cancer Res 2024; 43:188. [PMID: 38965605 PMCID: PMC11225384 DOI: 10.1186/s13046-024-03103-x] [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: 04/17/2024] [Accepted: 06/16/2024] [Indexed: 07/06/2024] Open
Abstract
BACKGROUND The vast majority of lncRNAs have low expression abundance, which greatly limits their functional range and impact. As a high expression abundance lncRNA, FGD5-AS1's non-ceRNA biological function in cancer is unclear. METHODS RNA-seq studies and chromatin immunoprecipitation (Chip) assays were performed to identify ZEB1-regulated lncRNAs. RNA sequencing, RNA pulldown, RNA Immunoprecipitation assays, and rescue assays were conducted to explore the molecular mechanisms of FGD5-AS1 in GC. RESULTS As one of the most abundant lncRNAs in cells, FGD5-AS1 has been shown to be transcriptionally activated by ZEB1, thus closely related to epithelial-mesenchymal transition (EMT) signaling. Clinical analysis showed that FGD5-AS1 overexpression was clinically associated with lymph node metastasis, and predicted poor survival in GC. Loss-of-function studies confirmed that FGD5-AS1 knockdown inhibited GC proliferation and induced cisplatin chemosensibility, cell senescence, and DNA damage in GC cells. Mechanismically, FGD5-AS1 is a YBX1-binding lncRNA due to its mRNA contains three adjacent structural motifs (UAAUCCCA, ACCAGCCU, and CAGUGAGC) that can be recognized and bound by YBX1. And this RNA-protein interaction prolonged the half-life of the YBX1 protein in GC. Additionally, a rescue assay showed that FGD5-AS1 promotes GC by repressing cell senescence and ROS production via YBX1. CONCLUSION FGD5-AS1 is a cellular high-abundant lncRNA that is transcriptionally regulated by ZEB1. FGD5-AS1 overexpression promoted GC progression by inhibiting cell senescence and ROS production through binding and stabilizing the YBX1 protein.
Collapse
Affiliation(s)
- Shanshan Qin
- Department of Stomatology, Taihe Hospital and Hubei Key Laboratory of Embryonic Stem Cell Research, School of Basic Medical Sciences, Hubei University of Medicine, Shiyan, Hubei, 442000, China.
- Laboratory of Tumor Biology, Academy of Bio-Medicine Research, Hubei University of Medicine, Shiyan, Hubei, P.R. China.
- Shiyan Key Laboratory of Natural Medicine Nanoformulation Research, Hubei University of Medicine, Shiyan, Hubei, 442000, China.
| | - Yue Liu
- Laboratory of Tumor Biology, Academy of Bio-Medicine Research, Hubei University of Medicine, Shiyan, Hubei, P.R. China
| | - Xiangang Zhang
- Laboratory of Tumor Biology, Academy of Bio-Medicine Research, Hubei University of Medicine, Shiyan, Hubei, P.R. China
| | - Pan Huang
- Laboratory of Tumor Biology, Academy of Bio-Medicine Research, Hubei University of Medicine, Shiyan, Hubei, P.R. China
| | - Lingyun Xia
- Department of Stomatology, Taihe Hospital and Hubei Key Laboratory of Embryonic Stem Cell Research, School of Basic Medical Sciences, Hubei University of Medicine, Shiyan, Hubei, 442000, China
| | - Weidong Leng
- Department of Stomatology, Taihe Hospital and Hubei Key Laboratory of Embryonic Stem Cell Research, School of Basic Medical Sciences, Hubei University of Medicine, Shiyan, Hubei, 442000, China.
| | - Dandan Li
- Department of Stomatology, Taihe Hospital and Hubei Key Laboratory of Embryonic Stem Cell Research, School of Basic Medical Sciences, Hubei University of Medicine, Shiyan, Hubei, 442000, China.
- Laboratory of Tumor Biology, Academy of Bio-Medicine Research, Hubei University of Medicine, Shiyan, Hubei, P.R. China.
- Shiyan Key Laboratory of Natural Medicine Nanoformulation Research, Hubei University of Medicine, Shiyan, Hubei, 442000, China.
| |
Collapse
|
18
|
Liu Y, Li Z, Zhang J, Liu W, Guan S, Zhan Y, Fang Y, Li Y, Deng H, Shen Z. DYNLL1 accelerates cell cycle via ILF2/CDK4 axis to promote hepatocellular carcinoma development and palbociclib sensitivity. Br J Cancer 2024; 131:243-257. [PMID: 38824222 PMCID: PMC11263598 DOI: 10.1038/s41416-024-02719-2] [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: 10/15/2023] [Revised: 05/06/2024] [Accepted: 05/10/2024] [Indexed: 06/03/2024] Open
Abstract
BACKGROUND Disorder of cell cycle represents as a major driver of hepatocarcinogenesis and constitutes an attractive therapeutic target. However, identifying key genes that respond to cell cycle-dependent treatments still facing critical challenges in hepatocellular carcinoma (HCC). Increasing evidence indicates that dynein light chain 1 (DYNLL1) is closely related to cell cycle progression and plays a critical role in tumorigenesis. In this study, we explored the role of DYNLL1 in the regulation of cell cycle progression in HCC. METHODS We analysed clinical specimens to assess the expression and predictive value of DYNLL1 in HCC. The oncogenic role of DYNLL1 was determined by gain or loss-of-function experiments in vitro, and xenograft tumour, liver orthotopic, and DEN/CCl4-induced mouse models in vivo. Mass spectrometry analysis, RNA sequencing, co-immunoprecipitation assays, and forward and reverse experiments were performed to clarify the mechanism by which DYNLL1 activates the interleukin-2 enhancer-binding factor 2 (ILF2)/CDK4 signalling axis. Finally, the sensitivity of HCC cells to palbociclib and sorafenib was assessed by apoptosis, cell counting kit-8, and colony formation assays in vitro, and xenograft tumour models and liver orthotopic models in vivo. RESULTS DYNLL1 was significantly higher in HCC tissues than that in normal liver tissues and closely related to the clinicopathological features and prognosis of patients with HCC. Importantly, DYNLL1 was identified as a novel hepatocarcinogenesis gene from both in vitro and in vivo evidence. Mechanistically, DYNLL1 could interact with ILF2 and facilitate the expression of ILF2, then ILF2 could interact with CDK4 mRNA and delay its degradation, which in turn activates downstream G1/S cell cycle target genes CDK4. Furthermore, palbociclib, a selective CDK4/6 inhibitor, represents as a promising therapeutic strategy for DYNLL1-overexpressed HCC, alone or particularly in combination with sorafenib. CONCLUSIONS Our work uncovers a novel function of DYNLL1 in orchestrating cell cycle to promote HCC development and suggests a potential synergy of CDK4/6 inhibitor and sorafenib for the treatment of HCC patients, especially those with increased DYNLL1.
Collapse
Affiliation(s)
- Yuechen Liu
- Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong Province, 510515, China
- Department of General Surgery & Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, The First School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong Province, 510515, China
| | - Zhenkang Li
- Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong Province, 510515, China
- Department of General Surgery & Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, The First School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong Province, 510515, China
| | - Jinchao Zhang
- Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong Province, 510515, China
- Department of General Surgery & Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, The First School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong Province, 510515, China
| | - Wei Liu
- Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong Province, 510515, China
- Department of General Surgery & Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, The First School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong Province, 510515, China
| | - Shenyuan Guan
- Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong Province, 510515, China
- Department of General Surgery & Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, The First School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong Province, 510515, China
| | - Yizhi Zhan
- Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong Province, 510515, China
- Department of General Surgery & Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, The First School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong Province, 510515, China
| | - Yuan Fang
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong Province, 510515, China
| | - Yongsheng Li
- Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong Province, 510515, China.
- Department of General Surgery & Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, The First School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong Province, 510515, China.
| | - Haijun Deng
- Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong Province, 510515, China.
- Department of General Surgery & Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, The First School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong Province, 510515, China.
| | - Zhiyong Shen
- Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong Province, 510515, China.
- Department of General Surgery & Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, The First School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong Province, 510515, China.
| |
Collapse
|
19
|
Li D, Xia L, Zhang X, Liu Y, Wang Z, Guo Q, Huang P, Leng W, Qin S. A new high-throughput screening methodology for the discovery of cancer-testis antigen using multi-omics data. COMPUTER METHODS AND PROGRAMS IN BIOMEDICINE 2024; 250:108193. [PMID: 38678957 DOI: 10.1016/j.cmpb.2024.108193] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Revised: 03/09/2024] [Accepted: 04/20/2024] [Indexed: 05/01/2024]
Abstract
BACKGROUND Cancer/testis antigens (CTAs), also known as tumor-specific antigens (TSAs) are specifically expressed in cancer cells and exhibit high immunogenicity, making them promising targets for immunotherapy and cancer vaccines. METHODS A new integrated high-throughput screening methodology for CTAs was proposed in this study through combining DNA methylation and RNA sequencing data. Briefly, the genes with increased transcript level and decreased DNA methylation were identified by multi-omics analysis. RNA sequencing studies in cell lines exposed to DNA methyltransferase (DNMT) inhibitors were performed to validate the inherent causal relationship between DNA hypomethylation and gene expression upregulation. RESULTS We proposed a new integrated high-throughput screening methodology for identification of CTAs using multi-omics analysis. In addition, we tested the feasibility of this method using gastric cancer (GC) as an example. In GC, we identified over 2000 primary candidate CTAs and ultimately identified 20 CTAs with significant tissue-specificity, including a testis-specific serine protease TESSP1/PRSS41. Integrated analysis confirmed that PRSS41 expression was reactivated in gastrointestinal cancers by promoter DNA hypomethylation at the CpG site (cg08104780). Additionally, DNA hypomethylation of PRSS41 predicted a poor prognosis in GC. CONCLUSION We propose a new high-throughput screening method for the identification of CTAs in cancer and validate its effectiveness. Our work emphasizes that serine protease PRSS41 is a novel TSA that is reactivated in GC due to promoter DNA hypomethylation.
Collapse
Affiliation(s)
- Dandan Li
- Department of Stomatology, Taihe Hospital and Hubei Key Laboratory of Embryonic Stem Cell Research, School of Basic Medical Sciences, Hubei University of Medicine, Shiyan 442000, Hubei, China; Laboratory of Tumor biology, Academy of Bio-Medicine Research, Hubei University of Medicine, Shiyan, Hubei, PR China; Shiyan Key Laboratory of Natural Medicine Nanoformulation Research, Hubei University of Medicine, Shiyan, Hubei 442000, China
| | - Lingyun Xia
- Department of Stomatology, Taihe Hospital and Hubei Key Laboratory of Embryonic Stem Cell Research, School of Basic Medical Sciences, Hubei University of Medicine, Shiyan 442000, Hubei, China
| | - Xiangang Zhang
- Department of Stomatology, Taihe Hospital and Hubei Key Laboratory of Embryonic Stem Cell Research, School of Basic Medical Sciences, Hubei University of Medicine, Shiyan 442000, Hubei, China; Laboratory of Tumor biology, Academy of Bio-Medicine Research, Hubei University of Medicine, Shiyan, Hubei, PR China
| | - Yue Liu
- Department of Stomatology, Taihe Hospital and Hubei Key Laboratory of Embryonic Stem Cell Research, School of Basic Medical Sciences, Hubei University of Medicine, Shiyan 442000, Hubei, China; Laboratory of Tumor biology, Academy of Bio-Medicine Research, Hubei University of Medicine, Shiyan, Hubei, PR China
| | - Zidi Wang
- Department of Stomatology, Taihe Hospital and Hubei Key Laboratory of Embryonic Stem Cell Research, School of Basic Medical Sciences, Hubei University of Medicine, Shiyan 442000, Hubei, China; Laboratory of Tumor biology, Academy of Bio-Medicine Research, Hubei University of Medicine, Shiyan, Hubei, PR China
| | - Qiwei Guo
- Department of Stomatology, Taihe Hospital and Hubei Key Laboratory of Embryonic Stem Cell Research, School of Basic Medical Sciences, Hubei University of Medicine, Shiyan 442000, Hubei, China; Laboratory of Tumor biology, Academy of Bio-Medicine Research, Hubei University of Medicine, Shiyan, Hubei, PR China
| | - Pan Huang
- Department of Stomatology, Taihe Hospital and Hubei Key Laboratory of Embryonic Stem Cell Research, School of Basic Medical Sciences, Hubei University of Medicine, Shiyan 442000, Hubei, China; Laboratory of Tumor biology, Academy of Bio-Medicine Research, Hubei University of Medicine, Shiyan, Hubei, PR China
| | - Weidong Leng
- Department of Stomatology, Taihe Hospital and Hubei Key Laboratory of Embryonic Stem Cell Research, School of Basic Medical Sciences, Hubei University of Medicine, Shiyan 442000, Hubei, China
| | - Shanshan Qin
- Department of Stomatology, Taihe Hospital and Hubei Key Laboratory of Embryonic Stem Cell Research, School of Basic Medical Sciences, Hubei University of Medicine, Shiyan 442000, Hubei, China; Laboratory of Tumor biology, Academy of Bio-Medicine Research, Hubei University of Medicine, Shiyan, Hubei, PR China; Shiyan Key Laboratory of Natural Medicine Nanoformulation Research, Hubei University of Medicine, Shiyan, Hubei 442000, China.
| |
Collapse
|
20
|
Ju Y, Fang S, Liu L, Ma H, Zheng L. The function of the ELF3 gene and its mechanism in cancers. Life Sci 2024; 346:122637. [PMID: 38614305 DOI: 10.1016/j.lfs.2024.122637] [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: 01/28/2024] [Revised: 04/01/2024] [Accepted: 04/10/2024] [Indexed: 04/15/2024]
Abstract
E74-like factor 3 (ELF3) is an important member of the E-twenty-six (ETS) transcription factor family. ELF3 is expressed in various types of cells and regulates a variety of biological behaviors, such as cell proliferation, differentiation, apoptosis, migration, and invasion, by binding to DNA to regulate the expression of other genes. In recent years, studies have shown that ELF3 plays an important role in the occurrence and development of many tumors and inflammation and immune related diseases. ELF3 has different functions and expression patterns in different tumors; it can function as a tumor suppressor gene or an oncogene, highlighting its dual effects of tumor promotion and inhibition. ELF3 also affects the levels of tumor immunity-related cytokines and is involved in the regulation and expression of multiple signaling pathways. In tumor therapy, ELF3 is a complex and multifunctional gene and has become a key focus of targeted treatment research. An in-depth study of the biological function of ELF3 can help to elucidate its role in biological processes and provide ideas and a basis for the development and clinical application of ELF3-related therapeutic methods. This review introduces the structure and physiological and cellular functions of the ELF3 gene, summarizes the mechanisms of action of ELF3 in different types of malignant tumors and its role in immune regulation, inflammation, etc., and discusses treatment methods for ELF3-related diseases, providing significant reference value for scholars studying the ELF3 gene and related diseases.
Collapse
Affiliation(s)
- Yiheng Ju
- Affiliated Hospital of Qingdao University, Qingdao, China
| | - Sheng Fang
- Yantai Penglai People's Hospital, Yantai, China
| | - Lei Liu
- Affiliated Hospital of Qingdao University, Qingdao, China
| | - Hui Ma
- Affiliated Hospital of Qingdao University, Qingdao, China.
| | - Longbo Zheng
- Affiliated Hospital of Qingdao University, Qingdao, China.
| |
Collapse
|
21
|
Qin S, Guo Q, Liu Y, Zhang X, Huang P, Yu H, Xia L, Leng W, Li D. A novel TGFbeta/TGILR axis mediates crosstalk between cancer-associated fibroblasts and tumor cells to drive gastric cancer progression. Cell Death Dis 2024; 15:368. [PMID: 38806480 PMCID: PMC11133402 DOI: 10.1038/s41419-024-06744-0] [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: 12/17/2023] [Revised: 05/12/2024] [Accepted: 05/14/2024] [Indexed: 05/30/2024]
Abstract
Transforming growth factor beta (TGFβ) signaling plays a critical role in tumorigenesis and metastasis. However, little is known about the biological function of TGFbeta-induced lncRNA in cancer. In this study, we discovered a novel TGFbeta-induced lncRNA, termed TGILR, whose function in cancer remains unknown to date. TGILR expression was directly activated by the canonical TGFbeta/SMAD3 signaling axis, and this activation is highly conserved in cancer. Clinical analysis showed that TGILR overexpression showed a significant correlation with lymph node metastasis and poor survival and was an independent prognostic factor in gastric cancer (GC). Depletion of TGILR caused an obvious inhibitory effect on GC cell proliferation, invasion, and epithelial-mesenchymal transition (EMT) in vitro and in vivo. More importantly, we demonstrated that TGFbeta signaling in GC was overactivated due to cancer-associated fibroblast (CAF) infiltration. Mechanistically, increased level of CAF-secreted TGFbeta activates TGFbeta signaling, leading to TGILR overexpression in GC cells. Meanwhile, TGILR overexpression inhibited the microRNA biogenesis of miR-1306 and miR-33a by interacting with TARBP2 and reducing its protein stability, thereby promoting GC progression via TCF4-mediated EMT signaling. In conclusion, CAF infiltration drives GC metastasis and EMT signaling through activating TGFbeta/TGILR axis. Targeted blocking of CAF-derived TGFbeta should be a promising anticancer strategy in GC.
Collapse
Affiliation(s)
- Shanshan Qin
- Department of Stomatology, Taihe Hospital and Hubei Key Laboratory of Embryonic Stem Cell Research, School of Basic Medical Sciences, Hubei University of Medicine, Shiyan, 442000, Hubei, China.
- Laboratory of Tumor biology, Academy of Bio-Medicine Research, Hubei University of Medicine, Shiyan, Hubei, P.R. China.
- Shiyan Key Laboratory of Natural Medicine Nanoformulation Research, Hubei University of Medicine, Shiyan, Hubei, 442000, China.
| | - Qiwei Guo
- Department of Stomatology, Taihe Hospital and Hubei Key Laboratory of Embryonic Stem Cell Research, School of Basic Medical Sciences, Hubei University of Medicine, Shiyan, 442000, Hubei, China
- Laboratory of Tumor biology, Academy of Bio-Medicine Research, Hubei University of Medicine, Shiyan, Hubei, P.R. China
| | - Yue Liu
- Department of Stomatology, Taihe Hospital and Hubei Key Laboratory of Embryonic Stem Cell Research, School of Basic Medical Sciences, Hubei University of Medicine, Shiyan, 442000, Hubei, China
- Laboratory of Tumor biology, Academy of Bio-Medicine Research, Hubei University of Medicine, Shiyan, Hubei, P.R. China
| | - Xiangang Zhang
- Department of Stomatology, Taihe Hospital and Hubei Key Laboratory of Embryonic Stem Cell Research, School of Basic Medical Sciences, Hubei University of Medicine, Shiyan, 442000, Hubei, China
- Laboratory of Tumor biology, Academy of Bio-Medicine Research, Hubei University of Medicine, Shiyan, Hubei, P.R. China
| | - Pan Huang
- Department of Stomatology, Taihe Hospital and Hubei Key Laboratory of Embryonic Stem Cell Research, School of Basic Medical Sciences, Hubei University of Medicine, Shiyan, 442000, Hubei, China
- Laboratory of Tumor biology, Academy of Bio-Medicine Research, Hubei University of Medicine, Shiyan, Hubei, P.R. China
| | - Hedong Yu
- Department of Stomatology, Taihe Hospital and Hubei Key Laboratory of Embryonic Stem Cell Research, School of Basic Medical Sciences, Hubei University of Medicine, Shiyan, 442000, Hubei, China
| | - Lingyun Xia
- Department of Stomatology, Taihe Hospital and Hubei Key Laboratory of Embryonic Stem Cell Research, School of Basic Medical Sciences, Hubei University of Medicine, Shiyan, 442000, Hubei, China
| | - Weidong Leng
- Department of Stomatology, Taihe Hospital and Hubei Key Laboratory of Embryonic Stem Cell Research, School of Basic Medical Sciences, Hubei University of Medicine, Shiyan, 442000, Hubei, China.
| | - Dandan Li
- Department of Stomatology, Taihe Hospital and Hubei Key Laboratory of Embryonic Stem Cell Research, School of Basic Medical Sciences, Hubei University of Medicine, Shiyan, 442000, Hubei, China.
- Laboratory of Tumor biology, Academy of Bio-Medicine Research, Hubei University of Medicine, Shiyan, Hubei, P.R. China.
- Shiyan Key Laboratory of Natural Medicine Nanoformulation Research, Hubei University of Medicine, Shiyan, Hubei, 442000, China.
| |
Collapse
|
22
|
Li D, Huang P, Xia L, Leng W, Qin S. Cancer-associated fibroblasts promote gastric cancer cell proliferation by paracrine FGF2-driven ribosome biogenesis. Int Immunopharmacol 2024; 131:111836. [PMID: 38479160 DOI: 10.1016/j.intimp.2024.111836] [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: 01/28/2024] [Revised: 03/05/2024] [Accepted: 03/08/2024] [Indexed: 04/10/2024]
Abstract
The cancer-associated fibroblast (CAF)-derived secretome plays critical roles in tumor progression by remodelling tumor microenvironment. Tumorigenesis is accompanied by the transformation of normal fibroblasts (NF) into CAF, leading to significant changes in their secretome. This work aims to identify the differential components of secretome between NFs and CAFs and reveal their functions in gastric cancer (GC). Firstly, our molecular typing studies and immune infiltration analysis showed that CAF infiltration level was increased and showed a significant association with clinical characteristics and poor prognosis of GC patients. Secondly, RNA-seq analysis revealed that a total of 1531 genes showed significant expression changes between NF and CAF. According to the annotation of the Human Protein Atlas (HPA) database, 147 genes encode secreted proteins, including FGF2. Particularly, the cell co-culture and RNA sequencing studies confirmed that exogenous recombinant FGF2 protein treatment promoted GC cell proliferation by enhancing ribosome biogenesis. The rescue assay showed that CAF-secreted FGF2 protein promotes GC cell growth and proliferation in a FGFR1-dependent manner. Our finding provides evidence that targeting blockade of CAF-derived FGF2 protein might be a promising treatment for GC.
Collapse
Affiliation(s)
- Dandan Li
- Department of Stomatology, Taihe Hospital and Hubei Key Laboratory of Embryonic Stem Cell Research, School of Basic Medical Sciences, Hubei University of Medicine, Shiyan, Hubei 442000, China; Laboratory of Tumor Biology, Academy of Bio-Medicine Research, Hubei University of Medicine, Shiyan, Hubei 442000, China; Shiyan Key Laboratory of Natural Medicine Nanoformulation Research, Hubei University of Medicine, Shiyan, Hubei 442000, China
| | - Pan Huang
- Department of Stomatology, Taihe Hospital and Hubei Key Laboratory of Embryonic Stem Cell Research, School of Basic Medical Sciences, Hubei University of Medicine, Shiyan, Hubei 442000, China; Laboratory of Tumor Biology, Academy of Bio-Medicine Research, Hubei University of Medicine, Shiyan, Hubei 442000, China
| | - Lingyun Xia
- Department of Stomatology, Taihe Hospital and Hubei Key Laboratory of Embryonic Stem Cell Research, School of Basic Medical Sciences, Hubei University of Medicine, Shiyan, Hubei 442000, China
| | - Weidong Leng
- Department of Stomatology, Taihe Hospital and Hubei Key Laboratory of Embryonic Stem Cell Research, School of Basic Medical Sciences, Hubei University of Medicine, Shiyan, Hubei 442000, China.
| | - Shanshan Qin
- Department of Stomatology, Taihe Hospital and Hubei Key Laboratory of Embryonic Stem Cell Research, School of Basic Medical Sciences, Hubei University of Medicine, Shiyan, Hubei 442000, China; Laboratory of Tumor Biology, Academy of Bio-Medicine Research, Hubei University of Medicine, Shiyan, Hubei 442000, China; Shiyan Key Laboratory of Natural Medicine Nanoformulation Research, Hubei University of Medicine, Shiyan, Hubei 442000, China.
| |
Collapse
|
23
|
Yin Z, Guo X, Liang X, Wang Z. FTO promotes gastric cancer progression by modulating MAP4K4 expression via demethylation in an m6A-dependent manner. Med Oncol 2024; 41:120. [PMID: 38643333 DOI: 10.1007/s12032-024-02369-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2024] [Accepted: 03/20/2024] [Indexed: 04/22/2024]
Abstract
Gastric cancer (GC) is a serious malignant tumour with a high mortality rate and a poor prognosis. Recently, emerging evidence has suggested that N6-methyladenosine (m6A) modification plays a crucial regulatory role in cancer progression. However, the exact role of m6A regulatory factors FTO in GC is unclear. First, the expression of m6A methylation-related regulatory factors in clinical samples and the clinical data of the corresponding patients were obtained from The Cancer Genome Atlas (TCGA-STAD) dataset, and correlation analysis between FTO expression and patient clinicopathological parameters was subsequently performed. qRT-PCR, immunohistochemistry (IHC) and western blotting (WB) were used to verify FTO expression in GC. CCK-8, EdU, flow cytometry and transwell assays were used to evaluate the effect of FTO on the behaviour of GC cells. Transcriptome sequencing and RNA immunoprecipitation analysis were used to explore the potential regulatory mechanisms mediated by FTO. FTO was highly expressed in GC tissues and cells, and high expression of FTO predicted a worse prognosis than low expression. Functionally, overexpression of FTO promoted the proliferation, migration and invasion of GC cells but inhibited cell apoptosis. Mechanistically, we found that FTO is upregulated in GC and promotes GC progression by modulating the expression of MAP4K4. Taken together, our findings provide new insights into the effects of FTO-mediated m6A demethylation and could lead to the development of new strategies for GC monitoring and aggressive treatment.
Collapse
Affiliation(s)
- Zhe Yin
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, People's Republic of China
- Department of Thoracic Surgery, Chongqing University Cancer Hospital, Chongqing Cancer Hospital, Chongqing, 400030, People's Republic of China
| | - Xiong Guo
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, People's Republic of China
| | - Xiaolong Liang
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, People's Republic of China
| | - Ziwei Wang
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, People's Republic of China.
| |
Collapse
|
24
|
Liu SS, Wan QS, Lv C, Wang JK, Jiang S, Cai D, Liu MS, Wang T, Zhang KH. Integrating trans-omics, cellular experiments and clinical validation to identify ILF2 as a diagnostic serum biomarker and therapeutic target in gastric cancer. BMC Cancer 2024; 24:465. [PMID: 38622522 PMCID: PMC11017608 DOI: 10.1186/s12885-024-12175-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: 01/26/2024] [Accepted: 03/24/2024] [Indexed: 04/17/2024] Open
Abstract
BACKGROUND Gastric cancer (GC) lacks serum biomarkers with clinical diagnostic value. Multi-omics analysis is an important approach to discovering cancer biomarkers. This study aimed to identify and validate serum biomarkers for GC diagnosis by cross-analysis of proteomics and transcriptomics datasets. METHODS A cross-omics analysis was performed to identify overlapping differentially expressed genes (DEGs) between our previous aptamer-based GC serum proteomics dataset and the GC tissue RNA-Seq dataset in The Cancer Genome Atlas (TCGA) database, followed by lasso regression and random forest analysis to select key overlapping DEGs as candidate biomarkers for GC. The mRNA levels and diagnostic performance of these candidate biomarkers were analyzed in the original and independent GC datasets to select valuable candidate biomarkers. The valuable candidate biomarkers were subjected to bioinformatics analysis to select those closely associated with the biological behaviors of GC as potential biomarkers. The clinical diagnostic value of the potential biomarkers was validated using serum samples, and their expression levels and functions in GC cells were validated using in vitro cell experiments. RESULTS Four candidate biomarkers (ILF2, PGM2L1, CHD7, and JCHAIN) were selected. Their mRNA levels differed significantly between tumor and normal tissues and showed different diagnostic performances for GC, with areas under the receiver operating characteristic curve (AUROCs) of 0.629-0.950 in the TCGA dataset and 0.736-0.840 in the Gene Expression Omnibus (GEO) dataset. In the bioinformatics analysis, only ILF2 (interleukin enhancer-binding factor 2) gene levels were associated with immune cell infiltration, some checkpoint gene expression, chemotherapy sensitivity, and immunotherapy response. Serum levels of ILF2 were higher in GC patients than in controls, with an AUROC of 0.944 for the diagnosis of GC, and it was also detected in the supernatants of GC cells. Knockdown of ILF2 by siRNA significantly reduced the proliferation and colony formation of GC cells. Overexpression of ILF2 significantly promotes the proliferation and colony formation of gastric cancer cells. CONCLUSIONS Trans-omics analysis of proteomics and transcriptomics is an efficient approach for discovering serum biomarkers, and ILF2 is a potential diagnostic biomarker and therapeutic target of gastric cancer.
Collapse
Affiliation(s)
- Shao-Song Liu
- Department of Gastroenterology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University; Jiangxi Institute of Gastroenterology & Hepatology, Nanchang, China, No 17, Yongwai Zheng Street, 330006, Nanchang, China
| | - Qin-Si Wan
- Department of Gastroenterology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University; Jiangxi Institute of Gastroenterology & Hepatology, Nanchang, China, No 17, Yongwai Zheng Street, 330006, Nanchang, China
| | - Cong Lv
- Department of Gastroenterology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University; Jiangxi Institute of Gastroenterology & Hepatology, Nanchang, China, No 17, Yongwai Zheng Street, 330006, Nanchang, China
| | - Jin-Ke Wang
- Department of Gastroenterology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University; Jiangxi Institute of Gastroenterology & Hepatology, Nanchang, China, No 17, Yongwai Zheng Street, 330006, Nanchang, China
| | - Song Jiang
- Department of Gastroenterology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University; Jiangxi Institute of Gastroenterology & Hepatology, Nanchang, China, No 17, Yongwai Zheng Street, 330006, Nanchang, China
| | - Dan Cai
- Department of Gastroenterology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University; Jiangxi Institute of Gastroenterology & Hepatology, Nanchang, China, No 17, Yongwai Zheng Street, 330006, Nanchang, China
| | - Mao-Sheng Liu
- Department of Gastroenterology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University; Jiangxi Institute of Gastroenterology & Hepatology, Nanchang, China, No 17, Yongwai Zheng Street, 330006, Nanchang, China
| | - Ting Wang
- Department of Gastroenterology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University; Jiangxi Institute of Gastroenterology & Hepatology, Nanchang, China, No 17, Yongwai Zheng Street, 330006, Nanchang, China
| | - Kun-He Zhang
- Department of Gastroenterology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University; Jiangxi Institute of Gastroenterology & Hepatology, Nanchang, China, No 17, Yongwai Zheng Street, 330006, Nanchang, China.
| |
Collapse
|
25
|
Xia L, Zhang T, Yao J, Lu K, Hu Z, Gu X, Chen Y, Qin S, Leng W. Fibromodulin overexpression drives oral squamous cell carcinoma via activating downstream EGFR signaling. iScience 2023; 26:108201. [PMID: 37965134 PMCID: PMC10641260 DOI: 10.1016/j.isci.2023.108201] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Revised: 09/01/2023] [Accepted: 10/11/2023] [Indexed: 11/16/2023] Open
Abstract
Accumulating evidence has shown that fibromodulin (FMOD) plays a pivotal role in tumorigenesis and metastasis. However, the biological function of FMOD in oral squamous cell carcinoma (OSCC) remains largely unclear to date. In this study, we confirmed that FMOD was overexpressed and showed a significant association with malignant progression and lymph node metastasis in OSCC. Depletion of FMOD inhibited OSCC proliferation and metastasis in vitro and in vivo. RNA sequencing, western blotting, and rescue assays verified that FMOD exerted oncogenic roles in OSCC via activation of EGFR signaling. In addition, FMOD was proved to be a putative target gene of miR-338-3p. Taken together, FMOD overexpression due to the reduced level of miR-338-3p promotes OSCC by activating EGFR signaling. Our findings provide direct evidence that targeting FMOD could be a promising therapeutic strategy for OSCC patients.
Collapse
Affiliation(s)
- Lingyun Xia
- Department of Stomatology, Taihe Hospital, Hubei University of Medicine, Shiyan 442000, P.R. China
- Institute of Oral Diseases, School of Dentistry, Hubei University of Medicine, Shiyan 442000, P.R. China
| | - Tianshu Zhang
- Department of Stomatology, Taihe Hospital, Hubei University of Medicine, Shiyan 442000, P.R. China
- Institute of Oral Diseases, School of Dentistry, Hubei University of Medicine, Shiyan 442000, P.R. China
| | - Juncheng Yao
- Department of Stomatology, Taihe Hospital, Hubei University of Medicine, Shiyan 442000, P.R. China
- Institute of Oral Diseases, School of Dentistry, Hubei University of Medicine, Shiyan 442000, P.R. China
| | - Kaitian Lu
- Department of Stomatology, Taihe Hospital, Hubei University of Medicine, Shiyan 442000, P.R. China
- Institute of Oral Diseases, School of Dentistry, Hubei University of Medicine, Shiyan 442000, P.R. China
| | - Ziqiu Hu
- Department of Stomatology, Taihe Hospital, Hubei University of Medicine, Shiyan 442000, P.R. China
- Institute of Oral Diseases, School of Dentistry, Hubei University of Medicine, Shiyan 442000, P.R. China
| | - Xinsheng Gu
- Department of Stomatology, Taihe Hospital, Hubei University of Medicine, Shiyan 442000, P.R. China
- Hubei Key Laboratory of Embryonic Stem Cell Research, School of Basic Medical Sciences, Hubei University of Medicine, Shiyan 442000, Hubei, China
| | - Yongji Chen
- Department of Stomatology, Taihe Hospital, Hubei University of Medicine, Shiyan 442000, P.R. China
- Hubei Key Laboratory of Embryonic Stem Cell Research, School of Basic Medical Sciences, Hubei University of Medicine, Shiyan 442000, Hubei, China
| | - Shanshan Qin
- Department of Stomatology, Taihe Hospital, Hubei University of Medicine, Shiyan 442000, P.R. China
- Hubei Key Laboratory of Embryonic Stem Cell Research, School of Basic Medical Sciences, Hubei University of Medicine, Shiyan 442000, Hubei, China
| | - Weidong Leng
- Department of Stomatology, Taihe Hospital, Hubei University of Medicine, Shiyan 442000, P.R. China
- Institute of Oral Diseases, School of Dentistry, Hubei University of Medicine, Shiyan 442000, P.R. China
| |
Collapse
|
26
|
Wang Y, Hong Z, Wei S, Ye Z, Chen L, Qiu C. Investigating the role of LncRNA PSMG3-AS1 in gastric cancer: implications for prognosis and therapeutic intervention. Cell Cycle 2023; 22:2161-2171. [PMID: 37946320 PMCID: PMC10732658 DOI: 10.1080/15384101.2023.2278942] [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: 02/27/2023] [Accepted: 10/30/2023] [Indexed: 11/12/2023] Open
Abstract
LncRNAs are widely linked to the complex development of gastric cancer, which is acknowledged worldwide as the third highest contributor to cancer-related deaths and the fifth most common form of cancer. The primary focus of this study is to examine the role of LncRNA PSMG3-AS1 in a group of individuals with gastric cancer. The results of our study indicate that PSMG3-AS1 is highly expressed in over 20 different types of cancer. Significantly, there was a clear association found between the expression of PSMG3-AS1 and a multitude of TMB and MSI tumors. PSMG3-AS1 exhibited significant upregulation in gastric cancer patients compared to healthy individuals within the gastric cancer cohort. The prognosis of gastric cancer patients is intrinsically associated with PSMG3-AS1, as confirmed by survival analysis and ROC curves. Furthermore, we created a disruption vector based on LncRNA PSMG3-AS1 and introduced it into AGS and MKN-45 cells, which are human gastric cancer cells. Significant decreases in the expression of the PSMG3-AS1 gene were noticed in both intervention groups compared to the NC group, reflecting the protein level expressions. Significantly, the proliferative and invasive capabilities of MKN-45 and AGS cells were notably reduced following transfection with PSMG3-AS1 siRNA. The results of our study indicate that disruption of the LncRNA PSMG3-AS1 gene may impact the CAV1/miR-451a signaling pathway, thereby leading to a reduction in the ability of gastric cancer cells to multiply and invade.
Collapse
Affiliation(s)
- Yi Wang
- Department of General Surgery, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian, China
- Department of Gastrointestinal Surgical Oncology, Clinical Oncology School of Fujian Medical University, Fuzhou, Fujian, China
| | - Zhongshi Hong
- Department of General Surgery, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian, China
| | - Shenghong Wei
- Department of Gastrointestinal Surgical Oncology, Clinical Oncology School of Fujian Medical University, Fuzhou, Fujian, China
| | - Zaisheng Ye
- Department of Gastrointestinal Surgical Oncology, Clinical Oncology School of Fujian Medical University, Fuzhou, Fujian, China
| | - Luchuan Chen
- Department of Gastrointestinal Surgical Oncology, Clinical Oncology School of Fujian Medical University, Fuzhou, Fujian, China
| | - Chengzhi Qiu
- Department of General Surgery, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian, China
| |
Collapse
|
27
|
Yao M, Mao X, Zhang Z, Xi Y, Gan H, Cui F, Shao S. Tumor-derived CircRNA_102191 promotes gastric cancer and facilitates M2 macrophage polarization. Cell Cycle 2023; 22:2003-2017. [PMID: 37872772 PMCID: PMC10761078 DOI: 10.1080/15384101.2023.2271341] [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: 04/03/2023] [Accepted: 09/24/2023] [Indexed: 10/25/2023] Open
Abstract
BACKGROUND Gastric cancer is a common malignant tumor of the digestive tract and the fourth leading cause of death from cancer-related diseases. In recent years, many studies have found that circular RNAs play an important role in cancer. Tumor-associated macrophages (TAMs) are also critical for tumor progression. OBJECTIVE This study examined the role of circRNA_102191 in gastric cancer progression. METHODS The relative mRNA levels were determined by qRT-PCR. Western blotting and ELISA were used to detect the protein levels. In vitro proliferation was assessed using CCK8 and clonogenic assays. The migration and invasion of cell lines were assessed by transwell-based assays. The interactions between molecules were detected using a luciferase reporter assay. M0 macrophages were induced with PMA. M1 macrophages were induced with LPS and IFN-γ, and M2 macrophages were induced with IL-4. RESULTS The expression of circRNA_102191 was enhanced significantly in gastric cancer cell lines and clinical tumor tissues. CircRNA_102191 promotes gastric cancer cell progression by regulating miR-493-3p and its downstream target gene XPR1. CircRNA_102191 can enhance the EMT process of gastric cancer cells by promoting the M2 polarization of macrophages. CONCLUSION CircRNA_102191 promotes the biological function of gastric cancer cells by regulating the miR-493-3p/XPR1 axis and M2 macrophage polarization.
Collapse
Affiliation(s)
- Min Yao
- Department of Urology, The Fourth Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu, China
- Department of Urology, The Affiliated Taizhou Second People's Hospital of Yangzhou University, Taizhou, Jiangsu, China
| | - Xuhua Mao
- Department of Clinical Laboratory, The Affiliated Yixing Hospital of Jiangsu University, Wuxi, Jiangsu, China
| | - Zherui Zhang
- School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, China
| | - Yue Xi
- School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, China
| | - Haining Gan
- School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, China
| | - Feilun Cui
- Department of Urology, The Fourth Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu, China
- Department of Urology, The Affiliated Taizhou Second People's Hospital of Yangzhou University, Taizhou, Jiangsu, China
| | - Shihe Shao
- School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, China
| |
Collapse
|
28
|
Li D, Xia L, Huang P, Wang Z, Guo Q, Huang C, Leng W, Qin S. Serine protease PRSS56, a novel cancer-testis antigen activated by DNA hypomethylation, promotes colorectal and gastric cancer progression via PI3K/AKT axis. Cell Biosci 2023; 13:124. [PMID: 37400936 DOI: 10.1186/s13578-023-01060-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Accepted: 05/27/2023] [Indexed: 07/05/2023] Open
Abstract
BACKGROUND Cancer/testis (CT) antigens/genes are usually overexpressed in cancers and exhibit high immunogenicity, making them promising targets for immunotherapy and cancer vaccines. The role of serine protease PRSS56 in cancers remains unknown to date. METHODS RNA sequencing studies were performed to screen CT genes in gastric cancer (GC) and colorectal cancer (CRC) cells exposed to DNA methyltransferase inhibitor 5-aza-2'-deoxycytidine (5-AZA-CdR). Bioinformatics analysis was conducted to analyze the correlation between PRSS56 expression and DNA methylation. Functional experiments were performed to explore the biological function of PRSS56 in GC and CRC. RESULTS In this study, we identified the testis-specific serine proteases PRSS56 as a novel CT antigen. PRSS56 was frequently overexpressed in various cancers, especially in gastrointestinal cancer. PRSS56 expression was negatively associated with promoter DNA methylation level, and positively associated with gene body methylation level. PRSS56 expression was significantly activated in colorectal and gastric cancer cells exposed to DNA methyltransferase inhibitors. Importantly, our finding highlights that the decreased methylation level of the CpG site cg10242318 in the PRSS56 promoter region resulted in its overexpression in GC and CRC. Additionally, functional assays verified that PRSS56 overexpression activated PI3K-AKT signaling in GC and CRC. CONCLUSION Serine protease PRSS56 is a novel CT antigen that is reactivated in cancers by promoter DNA hypomethylation. PRSS56 functions oncogenic roles in GC and CRC by activating of PI3K/AKT axis. Our results presented here represent the first data on the function of the serine protease PRSS56 in cancers.
Collapse
Affiliation(s)
- Dandan Li
- Department of Stomatology, Taihe Hospital and Hubei Key Laboratory of Embryonic Stem Cell Research, School of Basic Medical Sciences, Hubei University of Medicine, Shiyan, Hubei, 442000, P.R. China
- Laboratory of Tumor biology, Academy of Bio-Medicine Research, Hubei University of Medicine, Shiyan, Hubei, 442000, P.R. China
| | - Lingyun Xia
- Department of Stomatology, Taihe Hospital and Hubei Key Laboratory of Embryonic Stem Cell Research, School of Basic Medical Sciences, Hubei University of Medicine, Shiyan, Hubei, 442000, P.R. China
| | - Pan Huang
- Department of Stomatology, Taihe Hospital and Hubei Key Laboratory of Embryonic Stem Cell Research, School of Basic Medical Sciences, Hubei University of Medicine, Shiyan, Hubei, 442000, P.R. China
- Laboratory of Tumor biology, Academy of Bio-Medicine Research, Hubei University of Medicine, Shiyan, Hubei, 442000, P.R. China
| | - Zidi Wang
- Laboratory of Tumor biology, Academy of Bio-Medicine Research, Hubei University of Medicine, Shiyan, Hubei, 442000, P.R. China
| | - Qiwei Guo
- Laboratory of Tumor biology, Academy of Bio-Medicine Research, Hubei University of Medicine, Shiyan, Hubei, 442000, P.R. China
| | - Congcong Huang
- Department of Stomatology, Taihe Hospital and Hubei Key Laboratory of Embryonic Stem Cell Research, School of Basic Medical Sciences, Hubei University of Medicine, Shiyan, Hubei, 442000, P.R. China
- Laboratory of Tumor biology, Academy of Bio-Medicine Research, Hubei University of Medicine, Shiyan, Hubei, 442000, P.R. China
| | - Weidong Leng
- Department of Stomatology, Taihe Hospital and Hubei Key Laboratory of Embryonic Stem Cell Research, School of Basic Medical Sciences, Hubei University of Medicine, Shiyan, Hubei, 442000, P.R. China.
| | - Shanshan Qin
- Department of Stomatology, Taihe Hospital and Hubei Key Laboratory of Embryonic Stem Cell Research, School of Basic Medical Sciences, Hubei University of Medicine, Shiyan, Hubei, 442000, P.R. China.
- Laboratory of Tumor biology, Academy of Bio-Medicine Research, Hubei University of Medicine, Shiyan, Hubei, 442000, P.R. China.
| |
Collapse
|
29
|
Wang H, Ding Y, Zhu Q, Yu Z, Wang Q, Gong A, Xu M. LncRNA FAM83A-AS1 promotes epithelial-mesenchymal transition of pancreatic cancer cells via Hippo pathway. Cell Cycle 2023; 22:1514-1527. [PMID: 37245082 PMCID: PMC10281461 DOI: 10.1080/15384101.2023.2216507] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Revised: 03/20/2023] [Accepted: 04/01/2023] [Indexed: 05/29/2023] Open
Abstract
BACKGROUND Long non-coding RNAs (lncRNAs) have been proved to play a vital role in pancreatic cancer (PC). However, the role of lncRNA FAM83A-AS1 in PC remains unclear. In this study, we explored the biological function and underlying mechanism of FAM83A-AS1 in PC cells. METHODS The FAM83A-AS1 expression was assessed via public databases and validated by qRT-PCR. The biofunction and immune cell infiltration of FAM83A-AS1 were analyzed through GO, KEGG, GESA and ssGSEA. The migration, invasion and proliferation abilities of PC cells were examined by Transwell, wound healing, CCK8 and colony formation. The EMT and Hippo pathway markers were evaluated by western blot. RESULTS FAM83A-AS1 expression was higher in PC tissues and cells than normal. Additionally, FAM83A-AS1 was associated with poor prognosis of PC and involved in cadherin binding and immune infiltration. Subsequently, we proved FAM83A-AS1 overexpression enhanced the migration, invasion and proliferation abilities of PC cells, whereas FAM83A-AS1 downregulation inhibited those. Moreover, western blot results showed that FAM83A-AS1 knockdown increased the E-cadherin expression and decreased the expression of N-cadherin, β-catenin, Vimentin, Snail and Slug. On the contrary, FAM83A-AS1 upregulation results in the opposite effects. Besides, FAM83A-AS1 overexpression inhibited the expression of p-YAP, p-MOB1, p-Lats1, SAV1, MST1 and MST2 as well as the results of FAM83A-AS1 knockdown were opposite. CONCLUSION FAM83A-AS1 promoted EMT of PC cells via Hippo signaling inactivation and may be a potential diagnosis and prognosis target.
Collapse
Affiliation(s)
- Huizhi Wang
- Department of Gastroenterology, Affiliated Hospital of Jiangsu University, Jiangsu University, Zhenjiang, China
| | - Yuntao Ding
- Department of Gastroenterology, Affiliated Hospital of Jiangsu University, Jiangsu University, Zhenjiang, China
| | - Qiuming Zhu
- Department of Gastroenterology, Zhongshan Wusong Hospital Fudan University, Shanghai, China
| | - Zhengyue Yu
- Department of Gastroenterology, Affiliated Hospital of Jiangsu University, Jiangsu University, Zhenjiang, China
| | - Qi Wang
- Department of Gastroenterology, Affiliated Hospital of Jiangsu University, Jiangsu University, Zhenjiang, China
| | - Aihua Gong
- Department of Cell Biology, School of Medicine, Jiangsu University, Zhenjiang, China
| | - Min Xu
- Department of Gastroenterology, Affiliated Hospital of Jiangsu University, Jiangsu University, Zhenjiang, China
| |
Collapse
|
30
|
Zhang D, Jiang Z, Hu J, Sun X, Zheng Y, Shen Y. Comprehensively prognostic and immunological analysis of snail family transcriptional repressor 2 in pan-cancer and identification in pancreatic carcinoma. Front Immunol 2023; 14:1117585. [PMID: 37251370 PMCID: PMC10213725 DOI: 10.3389/fimmu.2023.1117585] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Accepted: 04/27/2023] [Indexed: 05/31/2023] Open
Abstract
Background Snail family transcriptional repressor 2 (SNAI2) is a transcription factor that induces epithelial to mesenchymal transition in neoplastic epithelial cells. It is closely related to the progression of various malignancies. However, the significance of SNAI2 in human pan-cancer is still largely unknown. Methods The Cancer Genome Atlas (TCGA), Genotype-Tissue Expression (GTEx), and Cancer Cell Line Encyclopedia (CCLE) databases were taken to examine the SNAI2 expression pattern in tissues and cancer cells. The link between SNAI2 gene expression levels and prognosis, as well as immune cell infiltration, was investigated using the Kaplan-Meier technique and Spearman correlation analysis. We also explored the expression and distribution of SNAI2 in various tumor tissues and cells by the THPA (Human Protein Atlas) database. We further investigated the relationship between SNAI2 expression levels and immunotherapy response in various clinical immunotherapy cohorts. Finally, the immunoblot was used to quantify the SNAI2 expression levels, and the proliferative and invasive ability of pancreatic cancer cells was determined by colony formation and transwell assays. Results We discovered heterogeneity in SNAI2 expression in different tumor tissues and cancer cell lines by exploring public datasets. The genomic alteration of SNAI2 existed in most cancers. Also, SNAI2 exhibits prognosis predictive ability in various cancers. SNAI2 was significantly correlated with immune-activated hallmarks, cancer immune cell infiltrations, and immunoregulators. It's worth noting that SNAI2 expression is significantly related to the effectiveness of clinical immunotherapy. SNAI2 expression was also found to have a high correlation with the DNA mismatch repair (MMR) genes and DNA methylation in many cancers. Finally, the knockdown of SNAI2 significantly weakened the proliferative and invasive ability of pancreatic cancer cells. Conclusion These findings suggested that SNAI2 could be used as a biomarker in human pan-cancer to detect immune infiltration and poor prognosis, which provides a new idea for cancer treatment.
Collapse
Affiliation(s)
- Dandan Zhang
- Department of General Surgery, the Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
- Jiangxi Key Laboratory of Molecular Medicine, the Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Zhenhong Jiang
- Jiangxi Key Laboratory of Molecular Medicine, the Second Affiliated Hospital of Nanchang University, Nanchang, China
- Department of Medical Genetics, the Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Jianping Hu
- Jiangxi Key Laboratory of Molecular Medicine, the Second Affiliated Hospital of Nanchang University, Nanchang, China
- Department of Medical Genetics, the Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Xiaoyun Sun
- Department of Medical Genetics, the Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Yan Zheng
- Jiangxi Key Laboratory of Molecular Medicine, the Second Affiliated Hospital of Nanchang University, Nanchang, China
- Department of Medical Genetics, the Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Yang Shen
- Jiangxi Key Laboratory of Molecular Medicine, the Second Affiliated Hospital of Nanchang University, Nanchang, China
- Department of Medical Genetics, the Second Affiliated Hospital of Nanchang University, Nanchang, China
| |
Collapse
|
31
|
Li D, Xia L, Huang P, Wang Z, Guo Q, Huang C, Leng W, Qin S. Heterogeneity and plasticity of epithelial-mesenchymal transition (EMT) in cancer metastasis: Focusing on partial EMT and regulatory mechanisms. Cell Prolif 2023:e13423. [PMID: 36808651 DOI: 10.1111/cpr.13423] [Citation(s) in RCA: 67] [Impact Index Per Article: 33.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 01/05/2023] [Accepted: 01/27/2023] [Indexed: 02/22/2023] Open
Abstract
Epithelial-mesenchymal transition (EMT) or mesenchymal-epithelial transition (MET) plays critical roles in cancer metastasis. Recent studies, especially those based on single-cell sequencing, have revealed that EMT is not a binary process, but a heterogeneous and dynamic disposition with intermediary or partial EMT states. Multiple double-negative feedback loops involved by EMT-related transcription factors (EMT-TFs) have been identified. These feedback loops between EMT drivers and MET drivers finely regulate the EMT transition state of the cell. In this review, the general characteristics, biomarkers and molecular mechanisms of different EMT transition states were summarized. We additionally discussed the direct and indirect roles of EMT transition state in tumour metastasis. More importantly, this article provides direct evidence that the heterogeneity of EMT is closely related to the poor prognosis in gastric cancer. Notably, a seesaw model was proposed to explain how tumour cells regulate themselves to remain in specific EMT transition states, including epithelial state, hybrid/intermediate state and mesenchymal state. Additionally, this article also provides a review of the current status, limitations and future perspectives of EMT signalling in clinical applications.
Collapse
Affiliation(s)
- Dandan Li
- Department of Stomatology, Taihe Hospital and Hubei Key Laboratory of Embryonic Stem Cell Research, School of Basic Medical Sciences, Hubei University of Medicine, Shiyan, China.,Laboratory of Tumor Biology, Academy of Bio-medicine Research, Hubei University of Medicine, Shiyan, China
| | - Lingyun Xia
- Department of Stomatology, Taihe Hospital and Hubei Key Laboratory of Embryonic Stem Cell Research, School of Basic Medical Sciences, Hubei University of Medicine, Shiyan, China
| | - Pan Huang
- Laboratory of Tumor Biology, Academy of Bio-medicine Research, Hubei University of Medicine, Shiyan, China
| | - Zidi Wang
- Laboratory of Tumor Biology, Academy of Bio-medicine Research, Hubei University of Medicine, Shiyan, China
| | - Qiwei Guo
- Laboratory of Tumor Biology, Academy of Bio-medicine Research, Hubei University of Medicine, Shiyan, China
| | - Congcong Huang
- Laboratory of Tumor Biology, Academy of Bio-medicine Research, Hubei University of Medicine, Shiyan, China
| | - Weidong Leng
- Department of Stomatology, Taihe Hospital and Hubei Key Laboratory of Embryonic Stem Cell Research, School of Basic Medical Sciences, Hubei University of Medicine, Shiyan, China
| | - Shanshan Qin
- Department of Stomatology, Taihe Hospital and Hubei Key Laboratory of Embryonic Stem Cell Research, School of Basic Medical Sciences, Hubei University of Medicine, Shiyan, China.,Laboratory of Tumor Biology, Academy of Bio-medicine Research, Hubei University of Medicine, Shiyan, China
| |
Collapse
|
32
|
Li D, Xia L, Huang P, Wang Z, Guo Q, Huang C, Leng W, Qin S. Cancer-associated fibroblast-secreted IGFBP7 promotes gastric cancer by enhancing tumor associated macrophage infiltration via FGF2/FGFR1/PI3K/AKT axis. Cell Death Dis 2023; 9:17. [PMID: 36681667 PMCID: PMC9867714 DOI: 10.1038/s41420-023-01336-x] [Citation(s) in RCA: 58] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Revised: 01/11/2023] [Accepted: 01/13/2023] [Indexed: 01/22/2023]
Abstract
We previously reported that IGFBP7 plays a role in maintaining mRNA stability of oncogenic lncRNA UBE2CP3 by RNA-RNA interaction in gastric cancer (GC). Clinical cohort studies had implied an oncogenic role of IGFBP7 in GC. However, the molecular mechanism of IGFBP7 in GC progression remains unknown. In this study, clinical analysis based on two independent cohorts showed that IGFBP7 was positively associated with poor prognosis and macrophage infiltration in GC. Loss-of-function studies confirmed the oncogenic properties of IGFBP7 in regulating GC cell proliferation and invasion. Mechanismly, IGFBP7 was highly expressed in cancer-associated fibroblasts (CAF) and mesenchymal cells, and was induced by epithelial-to-mesenchymal transition (EMT) signaling, since its expression was increased by TGF-beta treatment and reduced by overexpression of OVOL2 in GC. RNA sequencing, qRT-PCR, ELISA assay showed that IGFBP7 positively regulated FGF2 expression and secretion in GC. Transcriptome analysis revealed that FGFR1 was downregulated in M1 polarization but upregulated in M2 polarization. Exogenous recombinant IGFBP7 treatment in macrophages and GC cells further identified that IGFBP7 promotes tumor associated macrophage (TAM) polarization via FGF2/FGFR1/PI3K/AKT axis. Our finding here represented the first evidence that IGFBP7 promotes GC by enhancing TAM/M2 macrophage polarization through FGF2/FGFR1/PI3K/AKT axis.
Collapse
Affiliation(s)
- Dandan Li
- Department of Stomatology, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei, People's Republic of China.,Hubei Key Laboratory of Embryonic Stem Cell Research, School of Basic Medical Sciences, Hubei University of Medicine, Shiyan, Hubei, People's Republic of China.,Laboratory of Tumor Biology, Academy of Bio-medicine Research, Hubei University of Medicine, Shiyan, Hubei, People's Republic of China
| | - Lingyun Xia
- Department of Stomatology, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei, People's Republic of China
| | - Pan Huang
- Hubei Key Laboratory of Embryonic Stem Cell Research, School of Basic Medical Sciences, Hubei University of Medicine, Shiyan, Hubei, People's Republic of China.,Laboratory of Tumor Biology, Academy of Bio-medicine Research, Hubei University of Medicine, Shiyan, Hubei, People's Republic of China
| | - Zidi Wang
- Laboratory of Tumor Biology, Academy of Bio-medicine Research, Hubei University of Medicine, Shiyan, Hubei, People's Republic of China
| | - Qiwei Guo
- Laboratory of Tumor Biology, Academy of Bio-medicine Research, Hubei University of Medicine, Shiyan, Hubei, People's Republic of China
| | - Congcong Huang
- Hubei Key Laboratory of Embryonic Stem Cell Research, School of Basic Medical Sciences, Hubei University of Medicine, Shiyan, Hubei, People's Republic of China.,Laboratory of Tumor Biology, Academy of Bio-medicine Research, Hubei University of Medicine, Shiyan, Hubei, People's Republic of China
| | - Weidong Leng
- Department of Stomatology, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei, People's Republic of China.
| | - Shanshan Qin
- Department of Stomatology, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei, People's Republic of China. .,Hubei Key Laboratory of Embryonic Stem Cell Research, School of Basic Medical Sciences, Hubei University of Medicine, Shiyan, Hubei, People's Republic of China. .,Laboratory of Tumor Biology, Academy of Bio-medicine Research, Hubei University of Medicine, Shiyan, Hubei, People's Republic of China.
| |
Collapse
|
33
|
Yin Y, Huang C, Wang Z, Huang P, Qin S. Identification of cellular heterogeneity and key signaling pathways associated with vascular remodeling and calcification in young and old primate aortas based on single-cell analysis. Aging (Albany NY) 2022; 15:982-1003. [PMID: 36566020 PMCID: PMC10008505 DOI: 10.18632/aging.204442] [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: 09/30/2022] [Accepted: 12/09/2022] [Indexed: 12/24/2022]
Abstract
Aging of the vascular system is the main cause of many cardiovascular diseases. The structure and function of the blood vessel wall change with aging. To prevent age-related cardiovascular diseases, it is essential to understand the cellular heterogeneity of vascular wall and changes of cellular communication among cell subpopulations during aging. Here, using published single-cell RNA sequencing datasets of young and old monkey aortas, we analyzed the heterogeneity of vascular endothelial cells and smooth muscle cells in detail and identified a distinct endothelial cell subpopulation that involved in vascular remodeling and calcification. Moreover, cellular communication that changed with aging was analyzed and we identified a number of signaling pathways that associated with vascular aging. We found that EGF signaling pathway play an essential role in vascular remodeling and calcification of aged aortas. This work provided a better understanding of vascular aging and laid the foundation for prevention of age-related vascular pathologies.
Collapse
Affiliation(s)
- Yehu Yin
- Department of Stomatology, Taihe Hospital and Hubei Key Laboratory of Embryonic Stem Cell Research, School of Basic Medical Sciences, Hubei University of Medicine, Shiyan 442000, Hubei, P.R. China.,Institute of Medicine, Jishou University, Jishou 416000, P.R. China
| | - Congcong Huang
- Laboratory of Tumor Biology, Academy of Bio-Medicine Research, Hubei University of Medicine, Shiyan 442000, Hubei, P.R. China
| | - Zidi Wang
- Laboratory of Tumor Biology, Academy of Bio-Medicine Research, Hubei University of Medicine, Shiyan 442000, Hubei, P.R. China
| | - Pan Huang
- Laboratory of Tumor Biology, Academy of Bio-Medicine Research, Hubei University of Medicine, Shiyan 442000, Hubei, P.R. China
| | - Shanshan Qin
- Department of Stomatology, Taihe Hospital and Hubei Key Laboratory of Embryonic Stem Cell Research, School of Basic Medical Sciences, Hubei University of Medicine, Shiyan 442000, Hubei, P.R. China.,Laboratory of Tumor Biology, Academy of Bio-Medicine Research, Hubei University of Medicine, Shiyan 442000, Hubei, P.R. China
| |
Collapse
|