1
|
ZHANG HENG, YANG XIAO, GUO YUJIN, ZHAO HAIBO, JIANG PEI, YU QINGQING. The regulatory role of lncRNA in tumor drug resistance: refracting light through a narrow aperture. Oncol Res 2025; 33:837-849. [PMID: 40191723 PMCID: PMC11964869 DOI: 10.32604/or.2024.053882] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Accepted: 08/05/2024] [Indexed: 04/09/2025] Open
Abstract
As living conditions improve and diagnostic capabilities advance, the incidence of tumors has increased, with cancer becoming a leading cause of death worldwide. Surgery, chemotherapy, and radiotherapy are the most common treatments. Despite advances in treatment options, chemotherapy remains a routine first-line treatment for most tumors. Due to the continuous and extensive use of chemotherapy drugs, tumor resistance often develops, becoming a significant cause of treatment failure and poor prognosis. Recent research has increasingly focused on how long stranded non-coding RNAs (LncRNAs) influence the development of malignant tumors and drug resistance by regulating gene expression and other biological mechanisms during cell growth. Studies have demonstrated that variations in lncRNA expression levels, influenced by both interpatient variability and intratumoral genetic and epigenetic differences, are closely linked to tumor drug resistance. Therefore, this review advocates using lncRNA as a framework to investigate the regulation of genes associated with drug resistance, proposing lncRNA-targeted therapeutic strategies to potentially increase the efficacy of chemotherapy, improve patient outcomes, and guide future research directions.
Collapse
Affiliation(s)
- HENG ZHANG
- Department of Laboratory, Shandong Daizhuang Hospital, Jining, 272051, China
| | - XIAO YANG
- Department of Anesthesiology, Affiliated Hospital of Jining Medical University, Jining, 272000, China
| | - YUJIN GUO
- Department of Clinical Pharmacy, Jining No.1 People’s Hospital, Jining, 272002, China
| | - HAIBO ZHAO
- Department of Oncology, Jining No.1 People’s Hospital, Jining, 272002, China
| | - PEI JIANG
- Translational Pharmaceutical Laboratory, Jining No.1 People’s Hospital, Jining, 272002, China
| | - QING-QING YU
- Department of Clinical Pharmacy, Jining No.1 People’s Hospital, Jining, 272002, China
| |
Collapse
|
2
|
Cao S, Liu M, Guo Z, Li Y. Long noncoding RNA KCNMB2-AS1 acts as an oncogene in ovarian cancer. Acta Biochim Biophys Sin (Shanghai) 2023; 55:1844-1846. [PMID: 37710992 PMCID: PMC10679871 DOI: 10.3724/abbs.2023228] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Accepted: 08/15/2023] [Indexed: 09/16/2023] Open
Affiliation(s)
- Siyu Cao
- Department of Gynecologic OncologyFudan University Shanghai Cancer CenterShanghai200032China
| | - Min Liu
- Lab for Noncoding RNA and CancerSchool of Life SciencesShanghai UniversityShanghai200444China
| | - Ziyi Guo
- Lab for Noncoding RNA and CancerSchool of Life SciencesShanghai UniversityShanghai200444China
| | - Yanli Li
- Lab for Noncoding RNA and CancerSchool of Life SciencesShanghai UniversityShanghai200444China
| |
Collapse
|
3
|
Zhang Z, Wang X. Roles of long non-coding RNAs in digestive tract cancer and their clinical application. Zhejiang Da Xue Xue Bao Yi Xue Ban 2023; 52:451-459. [PMID: 37643979 PMCID: PMC10495243 DOI: 10.3724/zdxbyxb-2023-0169] [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: 04/13/2023] [Accepted: 08/02/2023] [Indexed: 08/24/2023]
Abstract
Long non-coding RNAs (lncRNAs) are strongly related to the occurrence and development of digestive tract cancer in human. Firstly, lncRNAs target and regulate the expression of downstream cancer genes to affect the growth, metastasis, apoptosis, metabolism and immune escape of cancer cells. Secondly, lncRNAs are considered to be important regulating factors for lipid metabolism in cancer, which is related to signaling pathways of adipogenesis and involved in the occurrence and development of digestive tract cancer. Finally, lncRNAs have application value in the diagnosis and treatment of digestive tract cancer. For example, lncRNAMALAT1 has been reported as a target for diagnosis and treatment of hepatocellular carcinoma. This article reviews current progress on the regulatory role of lncRNAs in digestive tract cancer, to provide references for the research and clinical application in the prevention and treatment of digestive tract cancer.
Collapse
Affiliation(s)
- Zhendong Zhang
- School of Medicine, Xizang Minzu University, Key Laboratory of High Altitude Hypoxia Environment and Life Health, Joint Laboratory for Research on Active Components and Pharmacological Mechanism of Tibetan Materia Medica of Tibetan Medical Research Center of Tibet, Xianyang 712082, Shaanxi Province, China.
| | - Xiaoping Wang
- School of Medicine, Xizang Minzu University, Key Laboratory of High Altitude Hypoxia Environment and Life Health, Joint Laboratory for Research on Active Components and Pharmacological Mechanism of Tibetan Materia Medica of Tibetan Medical Research Center of Tibet, Xianyang 712082, Shaanxi Province, China.
| |
Collapse
|
4
|
Guan J, Xu X, Qiu G, He C, Lu X, Wang K, Liu X, Li Y, Ling Z, Tang X, Liang Y, Tao X, Cheng B, Yang B. Cellular hierarchy framework based on single-cell/multi-patient sample sequencing reveals metabolic biomarker PYGL as a therapeutic target for HNSCC. J Exp Clin Cancer Res 2023; 42:162. [PMID: 37420300 DOI: 10.1186/s13046-023-02734-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Accepted: 06/04/2023] [Indexed: 07/09/2023] Open
Abstract
BACKGROUND A growing body of research has revealed the connection of metabolism reprogramming and tumor progression, yet how metabolism reprogramming affects inter-patient heterogeneity and prognosis in head and neck squamous cell carcinoma (HNSCC) still requires further explorations. METHODS A cellular hierarchy framework based on metabolic properties discrepancy, METArisk, was introduced to re-analyze the cellular composition from bulk transcriptomes of 486 patients through deconvolution utilizing single-cell reference profiles from 25 primary and 8 metastatic HNSCC sample integration of previous studies. Machine learning methods were used to identify the correlations between metabolism-related biomarkers and prognosis. The functions of the genes screened out in tumor progression, metastasis and chemotherapy resistance were validated in vitro by cellular functional experiments and in vivo by xenograft tumor mouse model. RESULTS Incorporating the cellular hierarchy composition and clinical properties, the METArisk phenotype divided multi-patient cohort into two classes, wherein poor prognosis of METArisk-high subgroup was associated with a particular cluster of malignant cells with significant activity of metabolism reprogramming enriched in metastatic single-cell samples. Subsequent analysis targeted for phenotype differences between the METArisk subgroups identified PYGL as a key metabolism-related biomarker that enhances malignancy and chemotherapy resistance by GSH/ROS/p53 pathway, leading to poor prognosis of HNSCC. CONCLUSION PYGL was identified as a metabolism-related oncogenic biomarker that promotes HNSCC progression, metastasis and chemotherapy resistance though GSH/ROS/p53 pathway. Our study revealed the cellular hierarchy composition of HNSCC from the cell metabolism reprogramming perspective and may provide new inspirations and therapeutic targets for HNSCC in the future.
Collapse
Affiliation(s)
- Jiezhong Guan
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-Sen University, Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, China
| | - Xi Xu
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Guo Qiu
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Chong He
- Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China
| | - Xiaoyue Lu
- Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China
| | - Kang Wang
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-Sen University, Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, China
| | - Xinyu Liu
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-Sen University, Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, China
| | - Yuanyuan Li
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-Sen University, Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, China
| | - Zihang Ling
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-Sen University, Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, China
| | - Xuan Tang
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-Sen University, Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, China
| | - Yujie Liang
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-Sen University, Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, China
| | - Xiaoan Tao
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-Sen University, Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, China.
| | - Bin Cheng
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-Sen University, Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, China.
| | - Bo Yang
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-Sen University, Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, China.
| |
Collapse
|
5
|
He XL, Lyu WY, Li XY, Zhao H, Qi L, Lu JJ. Identification of glycogen phosphorylase L as a potential target for lung cancer. Med Oncol 2023; 40:211. [PMID: 37347364 DOI: 10.1007/s12032-023-02069-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Accepted: 05/29/2023] [Indexed: 06/23/2023]
Abstract
Traditional Chinese medicine (TCM) has been widely used for cancer treatment. Identification of anti-cancer targets of TCM is the first and principal step in discovering molecular mechanisms of TCM as well as obtaining novel targets for cancer therapy. In this study, glycogen phosphorylase L (PYGL) was identified as one of the targeted proteins for several TCMs and was upregulated in various cancer types. The expression level of PYGL was positively correlated with the stage of lung cancer and the poor prognosis of patients. Meanwhile, knockdown of PYGL significantly inhibited proliferation and migration in lung cancer cells. In addition, PYGL was associated with spindle, kinetochore, and microtubule, the cellular components that are closely related to mitosis, in lung cancer. Moreover, PYGL was more susceptible to be upregulated by 144 mutated genes. Taken together, PYGL is a potential target for lung cancer treatment and its molecular mechanism probably influences the mitotic function of cells by regulating energy metabolism.
Collapse
Affiliation(s)
- Xin-Ling He
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, China
| | - Wen-Yu Lyu
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, China
| | - Xin-Yuan Li
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, China
| | - Hong Zhao
- The First Affiliated Hospital of Zhejiang, Chinese Medical University, Hangzhou, 310006, China
| | - Lu Qi
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, China.
- Department of Pathology, School of Basic Medical Sciences, Southern Medical University, No.1023 Shatai Road Baiyun District, Guangzhou, 510515, Guangdong, China.
| | - Jin-Jian Lu
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, China.
- Department of Pharmaceutical Sciences, Faculty of Health Sciences, University of Macau, Macao, China.
- Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, University of Macau, Macao, China.
| |
Collapse
|
6
|
lncRNA SSTR5-AS1 Predicts Poor Prognosis and Contributes to the Progression of Esophageal Cancer. DISEASE MARKERS 2023; 2023:5025868. [PMID: 36726845 PMCID: PMC9886483 DOI: 10.1155/2023/5025868] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Revised: 10/19/2022] [Accepted: 11/24/2022] [Indexed: 01/24/2023]
Abstract
Esophageal cancer (ESCA), as a common cancer worldwide, is a main cause of cancer-related mortality. Long noncoding RNAs (lncRNAs) have been shown in an increasing number of studies to be capable of playing an important regulatory function in human malignancies. Our study is aimed at delving into the prognostic value and potential function of lncRNA SSTR5-AS1 (SSTR5-AS1) in ESCA. The gene expression data of 182 ESCA samples from TCGA and 653 nontumor specimens from GTEx. The expressions of SSTR5-AS1 were analyzed. We investigated whether there was a correlation between the expression of SSTR5-AS1 and the clinical aspects of ESCA. In order to compare survival curves, the Kaplan-Meier method together with the log-rank test was utilized. The univariate and multivariate Cox regression models were used to analyze the data in order to determine the SSTR5-AS1 expression's significance as a prognostic factor in ESCA patients. In order to investigate the level of SSTR5-AS1 expression in ESCA cells, RT-PCR was utilized. CCK-8 trials served as a model for the loss-of-function tests. In this study, we found that the expressions of SSTR5-AS1 were increased in ESCA specimens compared with nontumor specimens. According to the ROC assays, high SSTR5-AS1 expression had an AUC value of 0.7812 (95% CI: 0.7406 to 0.8217) for ESCA. Patients who had a high level of SSTR5-AS1 expression had a lower overall survival rate than those who had a low level of SSTR5-AS1 expression. In addition, multivariate analysis suggested that SSTR5-AS1 was an independent predictor of overall survival for ESCA patients. Moreover, RT-PCR experiments indicated that SSTR5-AS1 expression was distinctly increased in three ESCA cells compared with HET1A cells. CCK-8 experiments indicated that silence of SSTR5-AS1 distinctly inhibited the proliferation of ESCA cells. Overall, ESCA patients with elevated SSTR5-AS1 had a worse chance of survival, suggesting it could be used as a prognostic and diagnostic biomarker for ESCA.
Collapse
|
7
|
Wu H, Zhao X, Zhu T, Rong D, Wang Y, Leng D, Wu D. A Glycosyltransferase-Related Signature for Predicting Overall Survival in Head and Neck Squamous Cell Carcinoma. Front Genet 2022; 13:856671. [PMID: 35899200 PMCID: PMC9311713 DOI: 10.3389/fgene.2022.856671] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Accepted: 05/30/2022] [Indexed: 01/02/2023] Open
Abstract
Background: Here, we establish a prognostic signature based on glycosyltransferase-related genes (GTRGs) for head and neck squamous cell carcinoma (HNSCC) patients. Methods: The prognostic signature of GTRGs was constructed via univariate and multivariate Cox analyses after obtaining the expression patterns of GTRGs from the TCGA. A nomogram based on the signature and clinical parameters was established to predict the survival of each HNSCC patient. Potential mechanisms were explored through gene set enrichment analysis (GSEA) and immune cell infiltration, immune checkpoints, immunotherapy, and tumor mutational burden (TMB) analyses. The expression differences and prognostic efficacy of the signature were verified through the gene expression omnibus (GEO) and several online databases. Results: The prognostic signature was constructed based on five glycosyltransferases (PYGL, ALG3, EXT2, FUT2, and KDELC1) and validated in the GSE65858 dataset. The pathways enriched in the high- and low-risk groups were significantly different. The high-risk group had higher tumor purity; lower infiltration of immune cells, such as CD8+ T cells and Tregs; higher cancer-associated fibroblast (CAF) infiltration; lower immune function; and lower checkpoint expression. The signature can also be applied to distinguish whether patients benefit from immunotherapy. In addition, the high-risk group had a higher TMB and more gene mutations, including those in TP53, CSMD1, CDKN2A, and MUC17. Conclusion: We propose a prognostic signature based on glycosyltransferases for HNSCC patients that may provide potential targets and biomarkers for the precise treatment of HNSCC.
Collapse
Affiliation(s)
- Huili Wu
- Department of Endodontics, The Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing, China
- Department of Oral and Maxillofacial Imaging, The Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing, China
- Jiangsu Province Key Laboratory of Oral Diseases, Nanjing, China
- Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing, China
| | - Xiao Zhao
- Department of Endodontics, The Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing, China
- Department of Oral and Maxillofacial Imaging, The Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing, China
- Jiangsu Province Key Laboratory of Oral Diseases, Nanjing, China
- Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing, China
| | - Tingting Zhu
- Department of Endodontics, The Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing, China
- Department of Oral and Maxillofacial Imaging, The Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing, China
- Jiangsu Province Key Laboratory of Oral Diseases, Nanjing, China
- Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing, China
| | - Di Rong
- Department of Endodontics, The Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing, China
- Department of Oral and Maxillofacial Imaging, The Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing, China
- Jiangsu Province Key Laboratory of Oral Diseases, Nanjing, China
- Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing, China
| | - Ying Wang
- Department of Endodontics, The Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing, China
- Department of Oral and Maxillofacial Imaging, The Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing, China
- Jiangsu Province Key Laboratory of Oral Diseases, Nanjing, China
- Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing, China
| | - Diya Leng
- Department of Endodontics, The Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing, China
- Department of Oral and Maxillofacial Imaging, The Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing, China
- Jiangsu Province Key Laboratory of Oral Diseases, Nanjing, China
- Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing, China
| | - Daming Wu
- Department of Endodontics, The Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing, China
- Department of Oral and Maxillofacial Imaging, The Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing, China
- Jiangsu Province Key Laboratory of Oral Diseases, Nanjing, China
- Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing, China
- *Correspondence: Daming Wu,
| |
Collapse
|
8
|
Jiang Q, Wang H, Yuan D, Qian X, Ma X, Yan M, Xing W. Circular_0086414 induces SPARC like 1 ( SPARCL1) production to inhibit esophageal cancer cell proliferation, invasion and glycolysis and induce cell apoptosis by sponging miR-1290. Bioengineered 2022; 13:12099-12114. [PMID: 35549806 PMCID: PMC9275914 DOI: 10.1080/21655979.2022.2073114] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
Circular RNA (circRNA) plays an important role in cancer progression. Here, we investigated the function of circ_0086414 in the malignant progression of esophageal cancer (EC). RNA expression of circ_0086414, microRNA-1290 (miR-1290), and SPARC like 1 (SPARCL1) was detected by quantitative real-time polymerase chain reaction. The protein levels of N-cadherin, E-cadherin, and SPARCL1 were checked by Western blotting analysis. Cell proliferation was investigated by 3-(4,5-Dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT), 5-Ethynyl-29-deoxyuridine (EdU), and cell colony formation assays. Cell invasion and apoptosis were analyzed by transwell invasion assay and flow cytometry analysis, respectively. Glycolysis was evaluated by analyzing glucose consumption and lactate production. In an xenograft mouse model, the effect of circ_0086414 on tumor tumorigenesis was investigated. The interactions among circ_0086414, miR-1290, and SPARCL1 were identified by dual-luciferase reporter and RNA pull-down assays. Results showed that circ_0086414 and SPARCL1 expression were significantly downregulated, while miR-1290 was upregulated in EC tissues and cells. EC patients with low circ_0086414 expression had a poor prognosis. Increasing circ_0086414 expression led to decreased EC cell proliferation, invasion and glycolysis and increased cell apoptosis, accompanied by a decrease of N-cadherin expression and an increase of E-cadherin expression. Also, the enforced expression of circ_0086414 delayed tumor tumorigenesis. Besides, circ_0086414 acted as a miR-1290 sponge and regulated EC cell processes by binding to the miRNA. MiR-1290 also participated in EC malignant progression through SPARCL1. Further, circ_0086414 stimulated SPARCL1 production by negatively regulating miR-1290. Thus, circ_0086414 inhibited EC cell malignancy through the miR-1290/SPARCL1 pathway, providing a reliable target for the therapy of EC.
Collapse
Affiliation(s)
- Qingfeng Jiang
- Department of Thoracic Surgery, The Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Haoran Wang
- Department of Thoracic Surgery, The Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Dongfeng Yuan
- Department of Thoracic Surgery, The Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Xin Qian
- Department of Thoracic Surgery, The Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Xiaochao Ma
- Department of Thoracic Surgery, The Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Ming Yan
- Department of Thoracic Surgery, The Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Wenqun Xing
- Department of Thoracic Surgery, The Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, Henan, China
| |
Collapse
|
9
|
Li P, Lv H, Wu Y, Xu K, Xu M, Ma Y. E2F transcription factor 1 is involved in the phenotypic modulation of esophageal squamous cell carcinoma cells via microRNA-375. Bioengineered 2021; 12:10047-10062. [PMID: 34699320 PMCID: PMC8809976 DOI: 10.1080/21655979.2021.1996510] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Revised: 10/15/2021] [Accepted: 10/16/2021] [Indexed: 12/28/2022] Open
Abstract
E2F family of transcription factors modulates multiple cellular functions associated with cell cycle and apoptosis. Here, we focused on the relevance of E2F1 to esophageal squamous cell carcinoma (ESCC) and identification of E2F1-mediated network in this study. Query of Gene Expression Omnibus database revealed that E2F1 was the core gene that was upregulated in ESCC. E2F1 downregulation inhibited ESCC cell activity. microRNA (miR)-375 was confirmed to be a downstream target of E2F1. E2F1 bound to miR-375 promoter and inhibited miR-375 transcription. Moreover, miR-375 inhibitor mitigated the repressive impacts of si-E2F1 on ESCC cells in part. Further study showed that sestrin 3 (SESN3) could interact with miR-375, and its knockdown annulled the stimulative effect of miR-375 inhibitor on ESCC development. Finally, E2F1 and SESN3 downregulation inhibited the phosphatidylinositol 3 kinase (PI3K)/AKT pathway activity in cells, while miR-375 inhibitor promoted PI3K/AKT pathway activation. These findings suggest that E2F1 inhibited miR-375 expression and promoted SESN3 expression to activate the PI3K/AKT pathway in ESCC.
Collapse
Affiliation(s)
- Pengfei Li
- Department of Thoracic Surgery, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, Shenyang, Liaoning, China
| | - Huina Lv
- Department of Ultrasound, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Yongkai Wu
- Department of Thoracic Surgery, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, Shenyang, Liaoning, China
| | - Ke Xu
- Department of Thoracic Surgery, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, Shenyang, Liaoning, China
| | - Min Xu
- Department of Thoracic Surgery, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, Shenyang, Liaoning, China
| | - Yegang Ma
- Department of Thoracic Surgery, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, Shenyang, Liaoning, China
| |
Collapse
|
10
|
Dong H, Wang M, Li Q. Exosomal miR-4488 and miR-1273g-5p inhibit the epithelial-mesenchymal transition of transforming growth factor β2-mediated retinal pigment epithelial cells by targeting ATP-binding cassette A4. Bioengineered 2021; 12:9693-9706. [PMID: 34592902 PMCID: PMC8810054 DOI: 10.1080/21655979.2021.1987068] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022] Open
Abstract
Exosomal microRNAs (miRNAs) have been shown to be involved in the regulation of many disease progression, including proliferative vitreoretinopathy (PVR). However, the roles of exosomal miR-4488 and miR-1273g-5p in PVR progression have not been demonstrated. Transforming growth factor β2 (TGF-β2)-induced ARPE-19 cells were used to stimulate the epithelial-mesenchymal transition (EMT) of cells. Exosomes derived from TGF-β2-induced ARPE-19 cells were identified by transmission electron microscopy and nanoparticle tracking analysis. The expression levels of miR-4488, miR-1273g-5p and ATP-binding cassette A4 (ABCA4) were measured by quantitative real-time PCR. The promotion levels of exosomes markers, EMT markers, apoptosis markers and ABCA4 were determined by western blot analysis. The migration, invasion and apoptosis of cells were determined by transwell assay, wound healing assay and flow cytometry. Our data showed that miR-4488 and miR-1273g-5p were lowly expressed in TGF-β2-induced ARPE-19 cells. Overexpressed exosomal miR-4488 and miR-1273g-5p could inhibit the EMT, migration, invasion, and promote apoptosis in TGF-β2-induced ARPE-19 cells. In addition, ABCA4 was a target of miR-4488 and miR-1273g-5p. Overexpressed ABCA4 also could reverse the negatively regulation of exosomal miR-4488 and miR-1273g-5p on the EMT, migration, and invasion of TGF-β2-induced ARPE-19 cells. In conclusion, our data showed that exosomal miR-4488 and miR-1273g-5p could inhibit TGF-β2-stimulated EMT in ARPE-19 cells through targeting ABCA4.
Collapse
Affiliation(s)
- Hongtao Dong
- Department of Ophthalmology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou City, 450052, Henan, China
| | - Menghua Wang
- Department of Ophthalmology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou City, 450052, Henan, China
| | - Qiuming Li
- Department of Ophthalmology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou City, 450052, Henan, China
| |
Collapse
|