1
|
Lv J, Li S, Zhang H. UBTD2 protein molecules emerges as a key prognostic protein marker in glioma: Insights from integrated omics and machine learning analysis of GRM7, NCAPG, CEP55, and other biomarkers. Int J Biol Macromol 2025; 310:143473. [PMID: 40286967 DOI: 10.1016/j.ijbiomac.2025.143473] [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: 03/09/2025] [Revised: 04/16/2025] [Accepted: 04/22/2025] [Indexed: 04/29/2025]
Abstract
Glioma is a malignant brain tumor with poor prognosis, and there is an urgent need to find effective biomarkers for early diagnosis and treatment. The aim of this study was to explore the potential of UBTD2 as a key prognostic protein marker for gliomas through comprehensive omics and machine learning analysis, and to reveal its relationship with other biomarkers such as GRM7, NCAPG, CEP55, etc. In this study, relevant data were downloaded from a public database, and differential expression gene (DEG) analysis was used to identify the characteristic genes of glioma. Through gene expression standardization and Venn analysis, 80 overlapping genes were selected and enriched by gene ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG). Key genes were identified based on LASSO analysis, while SHAP models were used to evaluate key DEGs and their interactions that influence glioma prediction models. Further analysis of UBTD2, including gene set enrichment analysis (GSEA), cell culture experiments, transfection techniques and Westernblot, was performed to investigate its function in glioma cells. The results showed that UBTD2 was significantly upregulated in glioma cells and correlated with the expression of five key proteins. LASSO analysis identified key difference-expressed genes in gliomas, and SHAP analysis revealed the importance of these genes in predictive models. Further experiments showed that UBTD2 knockdown inhibited cell proliferation and invasion by up-regulating p53 expression, underscoring its potential function in gliomas.
Collapse
Affiliation(s)
- Jia Lv
- Department of neurosurgery, The Affiliated Huai'an No. 1 Hospital of Nanjing Medical University, Huai 'an 223300, Jiangsu Province, China.
| | - Shaoxun Li
- Department of neurosurgery, The Affiliated Huai'an No. 1 Hospital of Nanjing Medical University, Huai 'an 223300, Jiangsu Province, China
| | - Hongrui Zhang
- Department of neurosurgery, The Affiliated Huai'an No. 1 Hospital of Nanjing Medical University, Huai 'an 223300, Jiangsu Province, China
| |
Collapse
|
2
|
Biological functions and therapeutic potential of SHCBP1 in human cancer. Biomed Pharmacother 2023; 160:114362. [PMID: 36739763 DOI: 10.1016/j.biopha.2023.114362] [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: 11/28/2022] [Revised: 01/29/2023] [Accepted: 01/31/2023] [Indexed: 02/05/2023] Open
Abstract
The incidence of cancer is increasing globally, and it is the most common cause of death. The identification of novel cancer diagnostic and prognostic biomarkers is important for developing cancer treatment strategies and reducing mortality. SHCSH2 domain-binding protein 1 (SHCBP1) is a protein that specifically binds to the SH2 domain of Src homology-collagen. It participates in the regulation of a variety of signal transduction pathways and can activate a variety of signaling molecules to perform a series of physiological functions. SHCBP1 is expressed in a variety of human tissues, but its abnormal expression in various systems is associated with cancer. SHCBP1 is abnormally expressed in a variety of tumors, and plays roles in almost all aspects of cancer biology (such as cell proliferation, apoptosis prevention, invasion, and metastasis) through various possible mechanisms. Its expression level is related to the clinicopathological characteristics of patients. In addition, the SHCBP1 expression pattern is closely related to cancer type, stage, and other clinical variables. Therefore, SHCBP1 is a promising tumor biomarker for cancer diagnosis and prognosis and a potential therapeutic target. This article reviews the expression, biological functions, mechanisms, and potential clinical significance of SHCBP1 in various human tumors to provide a new theoretical basis for clinical molecular diagnosis, molecular targeted therapy, and scientific research on cancer.
Collapse
|
3
|
Ran R, Gong CY, Wang ZQ, Zhou WM, Zhang SB, Shi YQ, Ma CW, Zhang HH. Long non‑coding RNA PART1: dual role in cancer. Hum Cell 2022; 35:1364-1374. [PMID: 35864416 DOI: 10.1007/s13577-022-00752-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Accepted: 07/14/2022] [Indexed: 12/24/2022]
Abstract
Increasing evidence has shown that long non-coding RNAs (lncRNAs), which are non-coding endogenous single-stranded RNAs, play an essential role in various physiological and pathological processes through transcriptional interference, post-transcriptional regulation, and epigenetic modification. Moreover, lncRNAs, as oncogenes or tumor suppressor genes, play an important role in the occurrence and development of human cancers. Prostate androgen-regulated transcript 1 (PART1) was initially identified as a carcinogenic lncRNA in prostate adenomas. The upregulated expression of PART1 plays a tumor-promoting role in liver, prostate, lung cancers, and other tumors. In contrast, the expression of PART1 is downregulated in esophageal squamous cell carcinoma, glioma, and other tumors, which may inhibit the tumor. PART1 plays a dual role in cancer and regulates cell proliferation, apoptosis, invasion, and metastasis through a variety of potential mechanisms. These findings suggest that PART1 is a promising tumor biomarker and therapeutic target. This article reviews the biological functions, related mechanisms, and potential clinical significance of PART1 in a variety of human cancers.
Collapse
Affiliation(s)
- Rui Ran
- Lanzhou University Second Hospital, 82 Cuiying Men, Lanzhou, 730000, People's Republic of China.,Orthopaedics Key Laboratory of Gansu Province, Lanzhou, 730000, People's Republic of China
| | - Chao-Yang Gong
- Lanzhou University Second Hospital, 82 Cuiying Men, Lanzhou, 730000, People's Republic of China.,Orthopaedics Key Laboratory of Gansu Province, Lanzhou, 730000, People's Republic of China
| | - Zhi-Qiang Wang
- Lanzhou University Second Hospital, 82 Cuiying Men, Lanzhou, 730000, People's Republic of China.,Orthopaedics Key Laboratory of Gansu Province, Lanzhou, 730000, People's Republic of China
| | - Wen-Ming Zhou
- Lanzhou University Second Hospital, 82 Cuiying Men, Lanzhou, 730000, People's Republic of China.,Orthopaedics Key Laboratory of Gansu Province, Lanzhou, 730000, People's Republic of China
| | - Shun-Bai Zhang
- Lanzhou University Second Hospital, 82 Cuiying Men, Lanzhou, 730000, People's Republic of China.,Orthopaedics Key Laboratory of Gansu Province, Lanzhou, 730000, People's Republic of China
| | - Yong-Qiang Shi
- Lanzhou University Second Hospital, 82 Cuiying Men, Lanzhou, 730000, People's Republic of China.,Orthopaedics Key Laboratory of Gansu Province, Lanzhou, 730000, People's Republic of China
| | - Chun-Wei Ma
- Lanzhou University Second Hospital, 82 Cuiying Men, Lanzhou, 730000, People's Republic of China.,Orthopaedics Key Laboratory of Gansu Province, Lanzhou, 730000, People's Republic of China
| | - Hai-Hong Zhang
- Lanzhou University Second Hospital, 82 Cuiying Men, Lanzhou, 730000, People's Republic of China.
| |
Collapse
|
4
|
H19- and hsa-miR-338-3p-mediated NRP1 expression is an independent predictor of poor prognosis in glioblastoma. PLoS One 2021; 16:e0260103. [PMID: 34843522 PMCID: PMC8629300 DOI: 10.1371/journal.pone.0260103] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2021] [Accepted: 11/02/2021] [Indexed: 12/19/2022] Open
Abstract
Glioblastoma multiforme (GBM) is the most common and also the most invasive brain cancer. GBM progression is rapid and its prognosis is poor. Therefore, finding molecular targets in GBM is a critical goal that could also play important roles in clinical diagnostics and treatments to improve patient prognosis. We jointly analyzed the GSE103227, GSE103229, and TCGA databases for differentially expressed RNA species, obtaining 52 long non-coding RNAs (lncRNAs), 31 microRNAs (miRNAs), and 186 mRNAs, which were used to build a competing endogenous RNA network. Kaplan–Meier and receiver operating characteristic (ROC) analyses revealed five survival-related lncRNAs: H19, LINC01574, LINC01614, RNF144A-AS1, and OSMR-AS1. With multiple optimization mRNAs, we found the H19-hsa-miR-338-3P-NRP1 regulatory pathway. Additionally, we noted high NRP1 expression in GBM patients, and Kaplan–Meier and ROC analyses showed that NRP1 expression was associated with GBM prognosis. Cox analysis indicated that NRP1 is an independent prognostic factor in GBM patients. In conclusion, H19 and hsa-miR-338-3P regulate NRP1 expression, and this pathway plays an important role in GBM.
Collapse
|
5
|
Chen J, Meng E, Lin Y, Shen Y, Hu C, Zhou G, Yuan C. The Role of Tumor-related LncRNA PART1 in cancer. Curr Pharm Des 2021; 27:4152-4159. [PMID: 34225608 DOI: 10.2174/1381612827666210705161955] [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: 01/28/2021] [Accepted: 06/01/2021] [Indexed: 11/22/2022]
Abstract
BACKGROUND As we all know, long non-coding RNA (lncRNA) affects tumor progression, which has caused a great upsurge in recent years. It can also affect the growth, migration, and invasion of tumors. When we refer to the abnormal expression of lncRNA, we will find it associated with malignant tumors. In addition, lncRNA has been proved to be a key targeted gene for the treatment of some diseases. PART1, a member of lncRNA, has been reported as a regulator in the process of tumor occurrence and development. This study aims to reveal the biological functions, specific mechanisms, and clinical significance of PART1 in various tumor cells. METHODS Through the careful search of PUBMED, the mechanisms of the effect of PART1 on tumorigenesis and development are summarized. RESULTS On the one hand, the up-regulated expression of PART1 plays a tumor-promoting role in tumors, including lung cancer, prostate cancer, bladder cancer and so on. On the other hand, PART1 is down-regulated in gastric cancer, glioma and other tumors to play a tumor inhibitory role. In addition, PART1 regulates tumor growth mainly by targeting microRNA such as miR-635, directly regulating the expression of proteins such as FUS/EZH2, affecting signal pathways such as the Toll-like receptor pathway, or regulating immune cells. CONCLUSION PART1 is closely related to tumors by regulating a variety of molecular mechanisms. In addition, PART1 can be used as a clinical marker for the early diagnosis of tumors and plays an important role in tumor-targeted therapy.
Collapse
Affiliation(s)
- Jinlan Chen
- College of Medical Science, China Three Gorges University, Yichang 443002, China
| | - Enqing Meng
- College of Medical Science, China Three Gorges University, Yichang 443002, China
| | - Yexiang Lin
- College of Medical Science, China Three Gorges University, Yichang 443002, China
| | - Yujie Shen
- College of Medical Science, China Three Gorges University, Yichang 443002, China
| | - Chengyu Hu
- College of Medical Science, China Three Gorges University, Yichang 443002, China
| | - Gang Zhou
- College of Traditional Chinese Medicine, China Three Gorges University, Yichang, China
| | - Chengfu Yuan
- College of Medical Science, China Three Gorges University, Yichang 443002, China
| |
Collapse
|
6
|
Identification of ubiquitination-related genes in human glioma as indicators of patient prognosis. PLoS One 2021; 16:e0250239. [PMID: 33914773 PMCID: PMC8084191 DOI: 10.1371/journal.pone.0250239] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Accepted: 04/04/2021] [Indexed: 11/21/2022] Open
Abstract
Ubiquitination is a dynamic and reversible process of a specific modification of target proteins catalyzed by a series of ubiquitination enzymes. Because of the extensive range of substrates, ubiquitination plays a crucial role in the localization, metabolism, regulation, and degradation of proteins. Although the treatment of glioma has been improved, the survival rate of patients is still not satisfactory. Therefore, we explore the role of ubiquitin proteasome in glioma. Survival-related ubiquitination related genes (URGs) were obtained through analysis of the Genotype-Tissue Expression (GTEx) and the Cancer Genome Atlas (TCGA). Cox analysis was performed to construct risk model. The accuracy of risk model is verified by survival, Receiver operating characteristic (ROC) and Cox analysis. We obtained 36 differentially expressed URGs and found that 25 URGs were related to patient prognosis. We used the 25 URGs to construct a model containing 8 URGs to predict glioma patient risk by Cox analysis. ROC showed that the accuracy rate of this model is 85.3%. Cox analysis found that this model can be used as an independent prognostic factor. We also found that this model is related to molecular typing markers. Patients in the high-risk group were enriched in multiple tumor-related signaling pathways. In addition, we predicted TFs that may regulate the risk model URGs and found that the risk model is related to B cells, CD4 T cells, and neutrophils.
Collapse
|
7
|
Pan Z, Mo F, Liu H, Zeng J, Huang K, Huang S, Cao Z, Xu X, Xu J, Liu T, Huang J. LncRNA prostate androgen-regulated transcript 1 (PART 1) functions as an oncogene in osteosarcoma via sponging miR-20b-5p to upregulate BAMBI. ANNALS OF TRANSLATIONAL MEDICINE 2021; 9:488. [PMID: 33850885 PMCID: PMC8039658 DOI: 10.21037/atm-21-658] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Background Osteosarcoma (OS) is an aggressive bone cancer that most commonly affects adolescents and children. Emerging studies have shown that long noncoding RNA (lncRNA) performs essential roles in the occurrence and development of many tumors. Prostate androgen-regulated transcript 1 (PART 1) has been reported as a tumor oncogene; despite this, the mechanisms underlying its involvement in OS are unclear. Methods OS and paired normal tissue samples were obtained, and gene expressions were detected by real time-quantitative polymerase chain reaction (RT-qPCR). The functions of PART 1 in OS cell proliferation, invasion, and migration were determined by Cell Counting Kit-8 (CCK-8) and Transwell assays. Furthermore, the binding sites of PART 1 and miR-20b-5p as well as those between miR-20b-5p and bone morphogenic protein and activin membrane-bound inhibitor homolog (BAMBI) were verified by bioinformatics analysis and dual-luciferase reporter assay. Results Our study found obvious overexpression of PART 1 in OS tissues and cells. Furthermore, PART 1 overexpression facilitated OS cell proliferation, invasion, and migration. Further mechanistic investigations revealed that PART 1 could sponge to miR-20b-5p, which was expressed at a low level in OS tissues and cells. Importantly, miR-20b-5p overexpression inhibited OS cell proliferation, invasion, and migration. Additionally, BAMBI was confirmed as a downstream gene of miR-20b-5p, and its expression was reversely modulated by miR-20b-5p and positively modulated by PART 1. Rescue experiments suggested that BAMBI was involved in PART 1-mediated promotion of OS progression. Conclusions PART 1 serves as a competing endogenous RNA to promote OS tumorigenesis via its regulation of the miR-20b-5p/BAMBI axis, which may provide a promising therapeutic biomarkers for OS patients.
Collapse
Affiliation(s)
- Zhimin Pan
- Department of Orthopaedics, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Fengbo Mo
- Department of Orthopaedics, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Hucheng Liu
- Department of Orthopaedics, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Jin Zeng
- Department of Orthopaedics, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Kai Huang
- Department of Orthopedics, Zhabei Central Hospital of Jing'an District, Shanghai, China
| | - Sheng Huang
- Department of Orthopaedics, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Zhiyou Cao
- Department of Orthopaedics, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Xiaoyu Xu
- Department of Orthopaedics, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Jianyun Xu
- Department of Orthopaedics, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Tianmi Liu
- Department of Rehabilitation, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Jiang Huang
- Department of Orthopaedics, The First Affiliated Hospital of Nanchang University, Nanchang, China
| |
Collapse
|
8
|
Geng H, Guo M, Xu W, Zang X, Wu T, Teng F, Wang Y, Liu X, Wang X, Sun Q, Liang J. SHCBP1 Promotes Papillary Thyroid Carcinoma Carcinogenesis and Progression Through Promoting Formation of Integrin and Collagen and Maintaining Cell Stemness. Front Endocrinol (Lausanne) 2021; 11:613879. [PMID: 33716952 PMCID: PMC7953042 DOI: 10.3389/fendo.2020.613879] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Accepted: 12/31/2020] [Indexed: 01/08/2023] Open
Abstract
Papillary thyroid carcinoma (PTC) is the most common thyroid cancer with a rapidly increasing incidence globally. Bioinformatics analyses suggested that SHCBP1 (SHC SH2 Domain-Binding Protein 1) was significantly up-regulated in PTC tumor tissues, which was further confirmed by immunohistochemical staining and qPCR analyses in Xuzhou cohort. Moreover, the results indicated that the mRNA level of SHCBP1 was negatively associated with patients' disease-free survival rate, and further analysis reveals that patients with high SHCBP1 expression tend to have more lymph node metastasis. Afterward, MTT, colony formation, cell-cycle assay, FACS apoptosis assay, invasion, migration, as well as scratch assay were performed to study the phenotypes change of PTC cells after knocking down SHCBP1. The in vivo subcutaneous tumor model was developed to study the proliferation ability of PTC cells after SHCBP1 knockdown. We show that knock down of SHCBP1 significantly inhibits PTC cell proliferation, cell cycle, invasion and migration in vivo and in vitro. Western blot and qRT-PCR showed that knockdown of SHCBP1 could significantly reduce MYC, KLF4, CD44, ITGA6, ITGB1, ITGB5, and COL4A2 expression at both RNA and protein levels, which indicated that SHCBP1 might be involved in PTC carcinogenesis and progression through targeting formation of integrin and collagen and cell stemness pathways, and can be a potential diagnosis biomarker and therapeutic target for PTC.
Collapse
Affiliation(s)
- Houfa Geng
- Department of Endocrinology, Affiliated Hospital of Medical College of Southeast University and Xuzhou Central Hospital, Xuzhou, China
- Xuzhou Clinical School, Xuzhou Medical University, Xuzhou, China
- Xuzhou Clinical School, Nanjing Medical University, Xuzhou, China
| | - Mengzhe Guo
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, China
| | - Wei Xu
- Department of Endocrinology, Affiliated Hospital of Medical College of Southeast University and Xuzhou Central Hospital, Xuzhou, China
- Xuzhou Clinical School, Xuzhou Medical University, Xuzhou, China
- Xuzhou Clinical School, Nanjing Medical University, Xuzhou, China
| | - Xiu Zang
- Department of Endocrinology, Affiliated Hospital of Medical College of Southeast University and Xuzhou Central Hospital, Xuzhou, China
- Xuzhou Clinical School, Xuzhou Medical University, Xuzhou, China
- Xuzhou Clinical School, Nanjing Medical University, Xuzhou, China
| | - Tingting Wu
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, China
- Cancer Institute, Xuzhou Medical University, Xuzhou, China
| | - Fei Teng
- Department of Endocrinology, Affiliated Hospital of Medical College of Southeast University and Xuzhou Central Hospital, Xuzhou, China
- Xuzhou Clinical School, Xuzhou Medical University, Xuzhou, China
- Xuzhou Clinical School, Nanjing Medical University, Xuzhou, China
| | - Yu Wang
- Department of Endocrinology, Affiliated Hospital of Medical College of Southeast University and Xuzhou Central Hospital, Xuzhou, China
- Xuzhou Clinical School, Xuzhou Medical University, Xuzhou, China
- Xuzhou Clinical School, Nanjing Medical University, Xuzhou, China
| | - Xuekui Liu
- Department of Endocrinology, Affiliated Hospital of Medical College of Southeast University and Xuzhou Central Hospital, Xuzhou, China
- Xuzhou Clinical School, Xuzhou Medical University, Xuzhou, China
- Xuzhou Clinical School, Nanjing Medical University, Xuzhou, China
| | - Xiuli Wang
- Department of Endocrinology, Affiliated Hospital of Medical College of Southeast University and Xuzhou Central Hospital, Xuzhou, China
- Xuzhou Clinical School, Xuzhou Medical University, Xuzhou, China
- Xuzhou Clinical School, Nanjing Medical University, Xuzhou, China
| | - Qiang Sun
- Cancer Institute, Xuzhou Medical University, Xuzhou, China
| | - Jun Liang
- Department of Endocrinology, Affiliated Hospital of Medical College of Southeast University and Xuzhou Central Hospital, Xuzhou, China
- Xuzhou Clinical School, Xuzhou Medical University, Xuzhou, China
- Xuzhou Clinical School, Nanjing Medical University, Xuzhou, China
- Xuzhou Institute of Medical Science, Postgraduate Workstation of Soochow University, Xuzhou, China
| |
Collapse
|
9
|
Zhao Y, Zhang Q, Liu H, Wang N, Zhang X, Yang S. lncRNA PART1, manipulated by transcriptional factor FOXP2, suppresses proliferation and invasion in ESCC by regulating the miR‑18a‑5p/SOX6 signaling axis. Oncol Rep 2021; 45:1118-1132. [PMID: 33432363 PMCID: PMC7859983 DOI: 10.3892/or.2021.7931] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Accepted: 11/24/2020] [Indexed: 12/11/2022] Open
Abstract
An increasing number of studies have demonstrated that long non-coding (lnc)RNAs are associated with tumor invasion, metastasis and the prognosis of patients with a variety of different tumors. However, the roles of lncRNA prostate androgen regulated transcript 1 (PART1) in esophageal squamous cell carcinoma (ESCC) remain unknown. In the present study, reverse transcription-quantitative PCR was performed to investigate the levels of PART1, SRY-box transcription factor 6 (SOX6) and miR-18a-5p in ESCC tissues and cells. The functions of PART1 in ESCC were demonstrated using Cell Counting Kit-8 and Matrigel assays. Promoter activity and dual-luciferase reporter assays, RNA immunoprecipitation and western blot analyses were also used to determine the potential mechanisms of PART1 in ESCC cell lines. It was found that PART1 and SOX6 were both downregulated in ESCC tissues and cells, and their low expression levels were associated with TNM stage, lymph node metastasis and poor prognosis in patients with ESCC. Forkhead box protein P2 (FOXP2) exhibited low expression level in ESCC tissues, and its expression was positively correlated with PART1 expression level in ESCC tissues. FOXP2 was found to bind to the promoter region of PART1 to regulate its expression in ESCC cells. Functionally, PART1 overexpression suppressed cell proliferation and invasion, whereas PART1 downregulation promoted cell proliferation and invasion in the ESCC cell lines. Mechanistically, PART1 functions as a competing endogenous (ce)RNA by sponging miR-18a-5p, resulting in the upregulation of the downstream target gene, SOX6, coupled with the inactivation of the β-catenin/c-myc signaling axis, to suppress ESCC cell proliferation and invasion. In conclusion, data from the present study unveil a potential ceRNA regulatory pathway, in which PART1 affects SOX6 expression level by sponging miR-18a-5p, to ultimately suppress ESCC development and progression.
Collapse
Affiliation(s)
- Yan Zhao
- Department of Medical Oncology, Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, Henan 450008, P.R. China
| | - Qing Zhang
- School of Life Sciences, Zhengzhou University, Zhengzhou, Henan 450001, P.R. China
| | - Hongtao Liu
- School of Life Sciences, Zhengzhou University, Zhengzhou, Henan 450001, P.R. China
| | - Ning Wang
- Henan Academy of Medical and Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Xiaosan Zhang
- Department of Medical Oncology, Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, Henan 450008, P.R. China
| | - Shujun Yang
- Department of Medical Oncology, Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, Henan 450008, P.R. China
| |
Collapse
|
10
|
Yin D, Liu L, Shi Z, Zhang L, Yang Y. Ropivacaine Inhibits Cell Proliferation, Migration and Invasion, Whereas Induces Oxidative Stress and Cell Apoptosis by circSCAF11/miR-145-5p Axis in Glioma. Cancer Manag Res 2020; 12:11145-11155. [PMID: 33173347 PMCID: PMC7648566 DOI: 10.2147/cmar.s274975] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Accepted: 09/23/2020] [Indexed: 12/12/2022] Open
Abstract
Background Glioma is a heterogeneous aggressive tumor. Ropivacaine, a widely used anesthetic, has been shown to repress the progression of multiple cancers, including glioma. In this study, the effects of ropivacaine on cell proliferation, migration, invasion and apoptosis in glioma were revealed. Methods The expression levels of circSCAF11 and miR-145-5p were detected by quantitative real-time polymerase chain reaction (qRT-PCR) in glioma tissues and cells. The expression levels of epithelial–mesenchymal transition (EMT)-related proteins were determined by Western blot. Oxidative stress was evaluated by the measurement of reactive oxygen species (ROS) and determination of mitochondrial 8-hydroxy-2-deoxyguanosine (8-OHdG) assay in glioma cells. Cell proliferation was determined by cell counting kit-8 (CCK-8) assay and cell colony formation assay. Cell apoptosis and metastasis were detected by flow cytometry analysis and transwell assay, respectively. The binding relationship between circSCAF11 and miR-145-5p was predicted by circular RNA Interactome and identified by dual-luciferase reporter assay and RNA immunoprecipitation assay. In vivo tumor formation assay was performed to reveal the effects between ropivacaine and circSCAF11 overexpression on tumorigenesis in vivo. Results CircSCAF11 expression was obviously upregulated and miR-145-5p was significantly downregulated in glioma tissues and cells compared with control groups. Ropivacaine treatment upregulated E-cadherin protein expression and repressed the protein expression of Vimentin. Functionally, ropivacaine exposure promoted ROS and 8-OHdG production and cell apoptosis, whereas inhibited cell proliferation, migration and invasion; however, these effects were hindered by circSCAF11 overexpression. Mechanistically, circSCAF11 was a sponge of miR-145-5p. In addition, ropivacaine was revealed to inhibit tumor growth in vivo by regulating circSCAF11 and miR-145-5p expression. Conclusion Ropivacaine suppressed glioma progression by regulating circSCAF11 and miR-145-5p, which might provide a theoretical foundation in glioma treatment.
Collapse
Affiliation(s)
- Danqin Yin
- Department of Anesthesiology, Danyang People's Hospital of Jiangsu, Danyang City, Jiangsu Province, People's Republic of China
| | - Li Liu
- Department of Anesthesiology, Tianjin Fourth Central Hospital, Tianjin City, People's Republic of China
| | - Zhengyuan Shi
- Department of Anesthesiology, Danyang People's Hospital of Jiangsu, Danyang City, Jiangsu Province, People's Republic of China
| | - Lihui Zhang
- Department of Anesthesiology, Hulunbeier Municipal People's Hospital (Hulunbuir Hospital Affiliated to Suzhou University), Hulunbeier City, Inner Mongolia Province, People's Republic of China
| | - Yan Yang
- Department of Anesthesiology, The First People's Hospital of Jiangxia District, Wuhan City, Hubei Province, People's Republic of China
| |
Collapse
|