1
|
Xu H, Ye Y. LINC00488 stimulates the progression of esophageal cancer by targeting microRNA-485-5p. Oncol Lett 2020; 21:86. [PMID: 33376519 PMCID: PMC7751374 DOI: 10.3892/ol.2020.12347] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Accepted: 10/15/2020] [Indexed: 11/21/2022] Open
Abstract
Esophageal cancer is the eighth most prevalent malignancy in the world and China has a high incidence of esophageal cancer. Previous studies have identified that LINC00488 is an oncogene; however, its role in esophageal cancer remains unclear. The present study detected the expression and biological functions of LINC00488 in the progression of esophageal cancer. LINC00488 levels in 45 esophageal cancer and matched paracancerous tissues were detected. The association between LINC00488 level, clinical indexes and overall survival rate of patients with esophageal cancer was analyzed. Using Cell Counting Kit-8, Transwell and wound healing assays, the influence of LINC00488 on the biological functions of OE19 and OE33 cells were assessed. The target gene of LINC00488, microRNA-485-5p (miRNA-485-5p), was predicted using bioinformatics databases. In addition, the role of miRNA-485-5p in the progression of esophageal cancer was evaluated using rescue experiments. LINC00488 was upregulated in esophageal cancer tissues and cell lines. A high level of LINC00488 was associated with lymphatic and distant metastasis and poor prognosis in patients with esophageal cancer. Silencing LINC00488 attenuated the viability, migration and wound healing of OE19 and OE33 cells. miRNA-485-5p was downregulated in esophageal cancer and low expression levels predicted a poor prognosis in these patients. In addition, miRNA-485-5p level was negatively correlated with that of LINC00488. Rescue experiments showed that knockdown of miRNA-485-5p reversed the attenuated proliferation and migration of esophageal cancer cells with LINC00488-knockdown. In conclusion, LINC00488 aggravated the malignant progression of esophageal cancer by targeting miRNA-485-5p.
Collapse
Affiliation(s)
- Hongbo Xu
- Department of Cardiothoracic Surgery, Lu'an Affiliated Hospital of Anhui Medical University, Lu'an, Anhui 237000, P.R. China
| | - Yan Ye
- Department of Cardiothoracic Surgery, Lu'an Affiliated Hospital of Anhui Medical University, Lu'an, Anhui 237000, P.R. China
| |
Collapse
|
2
|
Wang Z, Lu Y, Sheng B, Ding Y, Cheng X. Long-noncoding RNA LINC00461 promotes proliferation and invasion of nonsmall cell lung cancer cells via targeting miR-518a-3p/WDR1 pathway. J Recept Signal Transduct Res 2020; 42:80-87. [PMID: 33233986 DOI: 10.1080/10799893.2020.1850786] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Long noncoding RNAs (lncRNAs) are a class of RNAs participating in many biological processes such as imprinting, alternative splicing and RNA decay. Recently, lncRNAs have drawn a great deal of attention for their critical role in cancer progression. LINC00461, a newly identified lncRNA, has been reported to be significantly overexpressed in breast cancer and markedly expedited breast cancer progression. However, the specific role of LINC00461 in nonsmall cell lung cancer (NSCLC) remains unknown. In this study, we for the first time showed the biological functions of LINC00461 in NSCLC. Our results demonstrated that LINC00461 was significantly up-regulated in NSCLC tissues and cell lines. Furthermore, knockdown of LINC00461 inhibited NSCLC cell proliferation and invasion in vitro as well as suppressed tumor growth and metastasis in vivo. We also performed luciferase reporter assays and found that LINC00461 functioned as a sponge for miR-518a-3p and WDR1 was a target of miR-518a-3p. Taken together, we suggested an essential role of LINC00461/miR-518a-3p/WDR1 axis in NSCLC, which could be used as a potential therapeutic target for NSCLC treatment.
Collapse
Affiliation(s)
- Zuopei Wang
- Department of Thoracic Surgery, Shanghai Pudong New area People's Hospital, Shanghai, China
| | - Yi Lu
- Department of Thoracic Surgery, Shanghai Pudong New area People's Hospital, Shanghai, China
| | - Bo Sheng
- Department of Thoracic Surgery, Shanghai Pudong New area People's Hospital, Shanghai, China
| | - Yi Ding
- Department of Thoracic Surgery, Shanghai Pudong New area People's Hospital, Shanghai, China
| | - Xiaoke Cheng
- Department of Scientific Research, Shanghai Pudong New area People's Hospital, Shanghai, China
| |
Collapse
|
3
|
Shi W, Huang Q, Xie J, Wang H, Yu X, Zhou Y. CKS1B as Drug Resistance-Inducing Gene-A Potential Target to Improve Cancer Therapy. Front Oncol 2020; 10:582451. [PMID: 33102238 PMCID: PMC7545642 DOI: 10.3389/fonc.2020.582451] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2020] [Accepted: 08/28/2020] [Indexed: 12/18/2022] Open
Abstract
Cancer is a threat to human health and life. Although previously centered on chemical drug treatments, cancer treatment has entered an era of precision targeted therapy. Targeted therapy entails precise guidance, allowing the selective killing of cancer cells and thereby reducing damage to healthy tissues. Therefore, the need to explore potential targets for tumor treatment is vital. Cyclin-dependent kinase regulatory subunit 1B (CKS1B), a member of the conserved cyclin kinase subunit 1 (CKS1) protein family, plays an essential role in cell cycling. A large number of studies have shown that CKS1B is associated with the pathogenesis of many human cancers and closely related to drug resistance. Here, we describe the current understanding of the cellular functions of CKS1B and its underlying mechanisms, summarize a recent study of CKS1B as a target for cancer treatment and discuss the potential of CKS1B as a therapeutic target.
Collapse
Affiliation(s)
- Wenwen Shi
- Key Laboratory of Molecular Target and Clinical Pharmacology, The State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and The Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Qiudi Huang
- Key Laboratory of Molecular Target and Clinical Pharmacology, The State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and The Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Jiacui Xie
- Key Laboratory of Molecular Target and Clinical Pharmacology, The State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and The Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - He Wang
- Center of Cancer Research, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Xiyong Yu
- Key Laboratory of Molecular Target and Clinical Pharmacology, The State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and The Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Yi Zhou
- Key Laboratory of Molecular Target and Clinical Pharmacology, The State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and The Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
4
|
Zuo Z, Ji S, He L, Zhang Y, Peng Z, Han J. LncRNA TTN-AS1/miR-134-5p/PAK3 axis regulates the radiosensitivity of human large intestine cancer cells through the P21 pathway and AKT/GSK-3β/β-catenin pathway. Cell Biol Int 2020; 44:2284-2292. [PMID: 32749739 DOI: 10.1002/cbin.11436] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Revised: 07/24/2020] [Accepted: 08/01/2020] [Indexed: 01/09/2023]
Abstract
Radiotherapy is an important adjuvant treatment for large intestine cancer even though it does not cause any response in many patients. The present study aimed to investigate the effects of the TTN antisense RNA 1 (TTN-AS1) long noncoding RNA (lncRNA) on radiotherapy dynamics of large intestine cancer cells and to explore the underlying molecular mechanisms. TTN-AS1 expression was evaluated by reverse-transcription quantitative polymerase chain reaction, western blot, and cellular immunofluorescence, and flow cytometry analysis was used to measure apoptosis. Radiotherapy was simulated in vitro by exposing cancer cells to X-ray. TTN-AS1 was highly expressed in large intestine cancer cells after an X-ray exposition for 24 hr. TTN-AS1 knockdown improved the radiosensitivity of large intestine cancer cells and promoted apoptosis by increasing Bax/Bcl2 protein expression and the active-caspase 3/caspase 3 ratios following X-ray treatment. In addition, TTN-AS1 negatively regulated miR-134-5p expression, and miR-134-5p-mimic transfection decreased PAK3 protein expression in large intestine cancer cells. Importantly, TTN-AS1 promoted PAK3 and P21 protein expression in HT29 cells after X-ray treatment. Moreover, the knockdown of P21 protein expression improved radiosensitivity and promoted X-ray-induced apoptosis of HT29 cells. Finally, PAK3 knockdown expression decreased the p-AKT/AKT and p-GSK-3β/GSK-3β ratios and promoted the β-catenin transfer from the nucleus to the cytoplasm. These data suggest that the TTN-AS1 lncRNA promoted resistance to radiotherapy of large intestine cancer cells by increasing PAK3 expression via miR-134-5p inhibition, and this may be related to the P21 and AKT/GSK-3β/β-catenin pathway.
Collapse
Affiliation(s)
- Zhenkui Zuo
- Department of Proctology, Henan Provincial Hospital of Traditional Chinese Medicine, The Second Hospital Affiliated to Henan University of Chinese Medicine, Zhengzhou, Henan, China
| | - Shuling Ji
- Department of Proctology, Henan Provincial Hospital of Traditional Chinese Medicine, The Second Hospital Affiliated to Henan University of Chinese Medicine, Zhengzhou, Henan, China
| | - Lulu He
- Department of Proctology, Henan Provincial Hospital of Traditional Chinese Medicine, The Second Hospital Affiliated to Henan University of Chinese Medicine, Zhengzhou, Henan, China
| | - Yage Zhang
- Department of Nephropathy, Henan Provincial Hospital of Traditional Chinese Medicine, The Second Hospital Affiliated to Henan University of Chinese Medicine, Zhengzhou, Henan, China
| | - Zining Peng
- Department of Nephropathy, Henan Provincial Hospital of Traditional Chinese Medicine, The Second Hospital Affiliated to Henan University of Chinese Medicine, Zhengzhou, Henan, China
| | - Jiarui Han
- Department of Nephropathy, Henan Provincial Hospital of Traditional Chinese Medicine, The Second Hospital Affiliated to Henan University of Chinese Medicine, Zhengzhou, Henan, China
| |
Collapse
|
5
|
Ou M, Li X, Zhao S, Cui S, Tu J. Long non-coding RNA CDKN2B-AS1 contributes to atherosclerotic plaque formation by forming RNA-DNA triplex in the CDKN2B promoter. EBioMedicine 2020; 55:102694. [PMID: 32335370 PMCID: PMC7184162 DOI: 10.1016/j.ebiom.2020.102694] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2019] [Revised: 02/04/2020] [Accepted: 02/12/2020] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Atherosclerosis involves a slow process of plaque formation on the walls of arteries, and comprises a leading cause of cardiovascular disease. Long non-coding RNAs (lncRNAs) have been implicated in the pathogenesis of atherosclerosis. In this study, we aim to explore the possible involvement of lncRNA 'cyclin-dependent kinase inhibitor 2B antisense noncoding RNA' (CDKN2B-AS1) and CDKN2B in the progression of atherosclerosis. METHODS Initially, we quantified the expression of CDKN2B-AS1 in atherosclerotic plaque tissues and, in THP-1 macrophage-derived, and human primary macrophage (HPM)-derived foam cells. Next, we established a mouse model of atherosclerosis using apolipoprotein E knockout (ApoE-/-) mice, where lipid uptake, lipid accumulation, and macrophage reverse cholesterol transport (mRCT) were assessed, in order to explore the contributory role of CDKN2B-AS1 to the progression of atherosclerosis. RIP and ChIP assays were used to identify interactions between CDKN2B-AS1, CCCTC-binding factor (CTCF), enhancer of zeste homologue 2 (EZH2), and CDKN2B. Triplex formation was determined by RNA-DNA pull-down and capture assay as well as EMSA experiment. FINDINGS CDKN2B-AS1 showed high expression levels in atherosclerosis, whereas CDKN2B showed low expression levels. CDKN2B-AS1 accelerated lipid uptake and intracellular lipid accumulation whilst attenuating mRCT in THP-1 macrophage-derived foam cells, HPM-derived foam cells, and in the mouse model. EZH2 and CTCF were found to bind to the CDKN2B promoter region. An RNA-DNA triplex formed by CDKN2B-AS1 and CDKN2B promoter was found to recruit EZH2 and CTCF in the CDKN2B promoter region and consequently inhibit CDKN2B transcription by accelerating histone methylation. INTERPRETATION The results demonstrated that CDKN2B-AS1 promotes atherosclerotic plaque formation and inhibits mRCT in atherosclerosis by regulating CDKN2B promoter, and thereby could be a potential therapeutic target for atherosclerosis.
Collapse
Affiliation(s)
- Minghui Ou
- Department of Vascular Surgery, Qingdao Municipal Hospital, Qingdao 266011, PR China
| | - Xia Li
- Department of Ultrasound, Qingdao Municipal Hospital, Qingdao 266011, PR China
| | - Shibo Zhao
- Department of Vascular Surgery, Qingdao Municipal Hospital, Qingdao 266011, PR China
| | - Shichao Cui
- Department of Vascular Surgery, Qingdao Municipal Hospital, Qingdao 266011, PR China
| | - Jie Tu
- Department of Science and Education, Qingdao Municipal Hospital, No. 1, Jiaozhou Road, Shibei District, Qingdao 266011, Shandong Province, PR China.
| |
Collapse
|
6
|
Wang M, Wang L, He X, Zhang J, Zhu Z, Zhang M, Li X. lncRNA CCAT2 promotes radiotherapy resistance for human esophageal carcinoma cells via the miR‑145/p70S6K1 and p53 pathway. Int J Oncol 2019; 56:327-336. [PMID: 31789385 DOI: 10.3892/ijo.2019.4929] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Accepted: 09/09/2019] [Indexed: 11/06/2022] Open
Abstract
The long non‑coding RNA colon cancer‑associated transcript 2 (CCAT2) is abnormally expressed in various types of malignant tumor tissues and considered to be an oncogene, including for esophageal cancer (EC). Radiotherapy is an important and widely used cancer treatment. However, some patients with EC do not respond to radiotherapy. This study was designed to investigate effects of CCAT2 expression on radiotherapy dynamics for EC cells and to explore underlying molecular mechanisms. Reverse transcription‑quantitative PCR was used to measure CCAT2 expression in EC tissues, normal esophageal mucosa, EC cells and normal human esophageal epithelial cells. TUNEL assays were used to assess the effect of CCAT2 on X‑ray‑induced apoptosis of EC cells. Protein expression was detected by western blot. CCAT2 was highly expressed in EC tissues and EC cells, and was negatively associated with radiotherapy efficacy in patients with EC. In vitro, knockdown of CCAT2 enhanced radiosensitivity of EC cells and promoted apoptosis by increasing Bax/Bcl2 and active‑caspase 3/caspase 3 following X‑ray treatment. In addition, CCAT2 negatively regulated miR‑145 and P70 ribosomal protein S6 kinase 1 (p70S6K1) expression, and inhibited phosphorylation of Akt, ERK and p70S6K1 in EC cells. After X‑ray treatment, CCAT2 negatively regulated protein levels of p53, P21 and c‑Myc. These results showed that CCAT2 promoted the radiotherapy resistance of EC cells via negative regulation of the miR‑145/p70S6K1 and the p53 signaling pathways and associated elements may be potential targets for improving the sensitivity of EC radiotherapy.
Collapse
Affiliation(s)
- Ming Wang
- Department of Radiation Therapy, Cangzhou Central Hospital, Cangzhou, Hebei 061001, P.R. China
| | - Liang Wang
- Department of Digestive Endoscopy, Cangzhou Central Hospital, Cangzhou, Hebei 061001, P.R. China
| | - Xiang He
- Department of CT Diagnosis, Cangzhou Central Hospital, Cangzhou, Hebei 061001, P.R. China
| | - Jinhua Zhang
- Department of Pediatrics, Mengcun County Hospital, Cangzhou, Hebei 061400, P.R. China
| | - Zhongcheng Zhu
- Department of Radiation Therapy, Cangzhou Central Hospital, Cangzhou, Hebei 061001, P.R. China
| | - Mingyun Zhang
- Department of Radiation Therapy, Cangzhou Central Hospital, Cangzhou, Hebei 061001, P.R. China
| | - Xingde Li
- Department of Digestive Endoscopy, Cangzhou Central Hospital, Cangzhou, Hebei 061001, P.R. China
| |
Collapse
|
7
|
Li W, Huang K, Wen F, Cui G, Guo H, He Z, Zhao S. LINC00184 silencing inhibits glycolysis and restores mitochondrial oxidative phosphorylation in esophageal cancer through demethylation of PTEN. EBioMedicine 2019; 44:298-310. [PMID: 31201145 PMCID: PMC6606559 DOI: 10.1016/j.ebiom.2019.05.055] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Revised: 05/27/2019] [Accepted: 05/28/2019] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Total lesion glycolysis has been reported to be a satisfactory predictor of survival in patients with locally advanced esophageal cancer (EC). The aim of the present study is to investigate the function of long intergenic non-protein coding RNA 184 (LINC00184) on the EC cell glycolysis and mitochondrial oxidative phosphorylation (OXPHOS). METHODS The expression of LINC00184 was determined to be highly expressed and PTEN was poorly expressed in EC tissues and cells by RT-qPCR. In order to evaluate the effects of LINC00184 on cellular process in vitro and in vivo, gain- and loss-of-function approaches were performed to alter the expression of LINC00184 and PTEN in EC cells. RESULTS Silencing of LINC00184 was observed to inhibit the proliferation, migration, invasion, colony formation, and glycolysis of EC cells and tumour growth, while the mitochondrial OXPHOS was restored. By recruiting DNMT1, LINC00184 enhanced the promoter methylation of PTEN. Inhibition of PTEN promoter methylation suppressed EC glycolysis, whereas, improved mitochondrial OXPHOS. Mechanically, LINC00184 modulated glycolysis and mitochondrial OXPHOS in EC cells through induction of the Akt phosphorylation. After blockage of Akt signaling pathway by an Akt inhibitor, LY294002, the regulatory effects of LINC00184 on the glycolysis and mitochondrial OXPHOS of EC cells were reversed. CONCLUSION Taken together, the LINC00184/PTEN/Akt axis mediates glycolysis and mitochondrial OXPHOS in EC cells. This study highlighted a potential intervention target for treating EC.
Collapse
Affiliation(s)
- Weihao Li
- Department of Thoracic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, PR China
| | - Kai Huang
- Department of Thoracic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, PR China
| | - Fengbiao Wen
- Department of Thoracic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, PR China
| | - Guanghui Cui
- Department of Thoracic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, PR China
| | - Haizhou Guo
- Department of Thoracic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, PR China
| | - Zhanfeng He
- Department of Thoracic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, PR China
| | - Song Zhao
- Department of Thoracic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, PR China.
| |
Collapse
|
8
|
Zhou Y, Wang R, Xu T, Xie P, Zhang Y, Zhang A, Wang X, Yang C, Yang H, Zhu S. Prognostic Value of Long Noncoding RNA CRNDE as a Novel Biomarker in Solid Cancers: An Updated Systematic Review and Meta-Analysis. J Cancer 2019; 10:2386-2396. [PMID: 31258743 PMCID: PMC6584336 DOI: 10.7150/jca.31088] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Accepted: 04/13/2019] [Indexed: 12/14/2022] Open
Abstract
Background: Long noncoding RNA colorectal neoplasia differentially expressed (CRNDE) has been reported to exhibit a potential oncogenic role in the development of human cancers. However, the clinical value of CRNDE expression in various cancers still remains unclear. Herein, we conducted a meta-analysis to investigate the association between CRNDE and clinical outcomes in solid cancers. Methods: A systematic search was performed though the PubMed, EMBASE, Web of Science, Ovid, Cochrane library, CNKI and WanFang databases for eligible studies on clinical values of CRNDE in solid cancers. The pooled hazard ratios (HRs) or odd ratios (ORs) with 95% confidence intervals (CIs) were used to evaluate the link between CRNDE and clinical outcomes. Results: A total of 3690 patients from 20 studies (including 2 studies have 2 cohorts, respectively) were included. The results suggested that elevated CRNDE expression predicted a poor overall survival (OS) for in 13 types of solid cancers (HR=1.46, 95% CI: 1.33-1.58, P<0.001) with no heterogeneity (I2=21.8%, P=0.19). Subgroup analysis indicated a significant association between high CRNDE expression and shorter OS in the studies with digestive system cancers (HR=1.42, 95% CI: 1.28-1.55, P<0.001), qRT-PCR method (HR=1.45, 95% CI: 1.30-1.59, P<0.001), sample size >100 (HR=1.44, 95% CI: 1.32-1.57, P<0.001), and NOS>7 (HR= 1.50, 95% CI: 1.23-1.78, P<0.001). Furthermore, the pooled results showed that CRNDE was an independent prognostic factor for OS in cancer patients (HR=1.37, 95% CI: 1.22-1.52, P<0.001). In addition, we also revealed that CRNDE was positively related to tumor size (OR=2.10, 95%CI: 1.68-2.63, P<0.001), TNM stage (OR=2.86, 95%CI: 2.29-3.56, P<0.001), lymph node metastasis (LNM) (OR=3.21, 95%CI: 2.01-5.13, P<0.001), and distant metastasis (OR=4.36, 95%CI: 2.36-8.07, P<0.001). Although the probable evidences of publication bias were found in the studies with OS, tumor size, TNM stage or LNM, the trim and fill analysis confirmed the reliability of these results was not affected. Conclusion: Elevated CRNDE expression was associated with larger tumor size, advanced TNM stage, worse LNM and distant metastasis, and shorter OS, suggesting that CRNDE may act as an independent prognostic biomarker in solid cancers.
Collapse
Affiliation(s)
- Yu Zhou
- Sichuan Provincial Key Laboratory for Human Disease Gene Study, Institute of Laboratory Medicine, Hospital of University of Electronic Science and Technology of China and Sichuan Provincial People's Hospital, Chengdu 610072, China
| | - Rui Wang
- Organ Transplant Center, Hospital of University of Electronic Science and Technology of China and Sichuan Provincial People's Hospital, Chengdu 610072, China
| | - Tian Xu
- Organ Transplant Center, Hospital of University of Electronic Science and Technology of China and Sichuan Provincial People's Hospital, Chengdu 610072, China
| | - Ping Xie
- Sichuan Provincial Key Laboratory for Human Disease Gene Study, Institute of Laboratory Medicine, Hospital of University of Electronic Science and Technology of China and Sichuan Provincial People's Hospital, Chengdu 610072, China
| | - Yun Zhang
- Organ Transplant Center, Hospital of University of Electronic Science and Technology of China and Sichuan Provincial People's Hospital, Chengdu 610072, China
| | - Aifeng Zhang
- Renal Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Xiaojie Wang
- Renal Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Chong Yang
- Organ Transplant Center, Hospital of University of Electronic Science and Technology of China and Sichuan Provincial People's Hospital, Chengdu 610072, China
| | - Hongji Yang
- Organ Transplant Center, Hospital of University of Electronic Science and Technology of China and Sichuan Provincial People's Hospital, Chengdu 610072, China
| | - Shikai Zhu
- Organ Transplant Center, Hospital of University of Electronic Science and Technology of China and Sichuan Provincial People's Hospital, Chengdu 610072, China.,Renal Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
9
|
Zhang S, Jiang H, Xu Z, Jiang Y, She Y, Huang X, Feng S, Chen W, Chen S, Chen Y, Qiu G, Zhong S. The resistance of esophageal cancer cells to paclitaxel can be reduced by the knockdown of long noncoding RNA DDX11-AS1 through TAF1/TOP2A inhibition. Am J Cancer Res 2019; 9:2233-2248. [PMID: 31720085 PMCID: PMC6834486 DOI: pmid/31720085] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Accepted: 08/18/2019] [Indexed: 02/05/2023] Open
Abstract
Esophageal cancer (EC) is one of the most common malignancies in the world. The currently used chemotherapeutic drug for the treatment of EC is paclitaxel (PTX), the efficacy of which is affected by the development of drug resistance. The present study aims to define the role of the long noncoding RNA (lncRNA) DDX11-AS1 in the progression of EC with the involvement of PTX-resistant EC cells. First, EC and adjacent normal tissue samples were collected from 82 patients with EC, after which the expression levels of DDX11-AS1, TOP2A and TAF1 were determined. The results showed that DDX11-AS1, TOP2A and TAF1 were highly expressed in EC tissues, and there was a positive correlation between the expression levels of DDX11-AS1 and TOP2A. A PTX-resistant EC cell line was constructed. Next, we evaluated the effects of DDX11-AS1 and TOP2A on the resistance of EC cells to PTX, and the regulatory relationships between DDX11-AS1, TOP2A and TAF1 were investigated. DDX11-AS1 could promote TOP2A transcription via TAF1, and the knockdown of TOP2A or DDX11-AS1 could increase the sensitivity of EC cells to PTX. The effect of DDX11-AS1 on the growth of PTX-inhibited tumors was confirmed using a tumor formation assay in nude mice. It was verified that knocking down DDX11-AS1 reduced the expression level of TOP2A and inhibited tumor growth. In conclusion, our findings suggest that DDX11-AS1 knockdown results in reduced resistance of EC cells to PTX by inhibiting TOP2A transcription via TAF1. Therefore, DDX11-AS1 knockdown could be a promising therapeutic strategy for EC.
Collapse
Affiliation(s)
- Shuyao Zhang
- Department of Pharmacy, Guangzhou Red Cross Hospital Affiliated of Ji-Nan University Medical CollegeGuangzhou 510220, Guangdong Province, P. R. China
- Clinical Pharmacy Research Center, Shantou University Medical CollegeShantou 515031, Guangdong Province, P. R. China
| | - Hong Jiang
- Department of Nursing, Guangzhou Red Cross Hospital Affiliated of Ji-Nan University Medical CollegeGuangzhou 510220, Guangdong Province, P. R. China
| | - Zhe Xu
- Department of Urology, Cancer Hospital of Shantou University Medical CollegeShantou 515031, Guangdong Province, P. R. China
| | - Yi Jiang
- Department of Digestive Oncology, Cancer Hospital of Shantou University Medical CollegeShantou 515031, Guangdong Province, P. R. China
| | - Yuqi She
- Clinical Pharmacy Research Center, Shantou University Medical CollegeShantou 515031, Guangdong Province, P. R. China
| | - Xiaoting Huang
- Clinical Pharmacy Research Center, Shantou University Medical CollegeShantou 515031, Guangdong Province, P. R. China
| | - Shanna Feng
- Clinical Pharmacy Research Center, Shantou University Medical CollegeShantou 515031, Guangdong Province, P. R. China
| | - Wanying Chen
- Clinical Pharmacy Research Center, Shantou University Medical CollegeShantou 515031, Guangdong Province, P. R. China
| | - Shuang Chen
- Clinical Pharmacy Research Center, Shantou University Medical CollegeShantou 515031, Guangdong Province, P. R. China
| | - Yun Chen
- Clinical Pharmacy Research Center, Shantou University Medical CollegeShantou 515031, Guangdong Province, P. R. China
| | - Guodong Qiu
- Clinical Pharmacy Research Center, Shantou University Medical CollegeShantou 515031, Guangdong Province, P. R. China
| | - Shilong Zhong
- Clinical Pharmacy Research Center, Shantou University Medical CollegeShantou 515031, Guangdong Province, P. R. China
- Guangdong Provincial Key Laboratory of Coronary Heart Disease Prevention, Guangdong Cardiovascular Institute, Guangdong General Hospital, Guangdong Academy of Medical SciencesGuangzhou 510080, Guangdong Province, P. R. China
| |
Collapse
|
10
|
Wang M, Li Y, Yang Y, Liu X, Zang M, Li Y, Yang K, Yang W, Zhang S. Long non‑coding RNA DLX6‑AS1 is associated with malignant progression and promotes proliferation and invasion in esophageal squamous cell carcinoma. Mol Med Rep 2018; 19:1942-1950. [PMID: 30592268 DOI: 10.3892/mmr.2018.9786] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2018] [Accepted: 09/10/2018] [Indexed: 11/06/2022] Open
Abstract
Despite being one of the most prevalent and fatal types of cancer worldwide, the biological details of esophageal squamous cell carcinoma (ESCC) remain unknown. Recent studies have demonstrated the crucial roles of long non‑coding RNAs (lncRNAs) in diverse biological processes including cancer initiation, progression and metastasis. The aim of the present study was to assess the expression profile of distal‑less homeobox 6 antisense RNA 1 (DLX6‑AS1) in ESCC tissues and its contributions to ESCC cell proliferation, apoptosis and invasion. The expression of DLX6‑AS1 in a series of ESCC samples and paired adjacent noncancerous tissues was evaluated by reverse transcription‑quantitative polymerase chain reaction. Cell proliferation, apoptosis, wound healing and Transwell invasion assays were performed to evaluate the roles of DLX6‑AS1 in the ESCC cell lines EC109 and KYSE30 transfected with DLX6‑AS1 small interfering RNA (siRNA). Compared with the paired adjacent noncancerous tissues, DLX6‑AS1 expression was upregulated in the ESCC tissues and significantly associated with differentiation status, Tumor‑Node‑Metastasis stage, distant metastasis, and lymph node metastasis. Knockdown of DLX6‑AS1 significantly suppressed cell proliferation, invasion and migration abilities, and enhanced the apoptotic rate in the two ESCC cell lines. Furthermore, western blot assays revealed that silencing DLX6‑AS1 partly influenced the epithelial‑mesenchymal transition process in ESCC cells. These results imply that the oncogenic function of DLX6‑AS1 may be a novel candidate target for treating human ESCC.
Collapse
Affiliation(s)
- Mingchen Wang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, P.R. China
| | - Yuan Li
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, P.R. China
| | - Yixin Yang
- School of Clinical Medicine, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, P.R. China
| | - Xiaosa Liu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, P.R. China
| | - Mingxi Zang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, P.R. China
| | - Yuebai Li
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, P.R. China
| | - Kunpeng Yang
- Department of Chest Surgery, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450001, P.R. China
| | - Wanguang Yang
- Department of Emergency, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450001, P.R. China
| | - Shanfeng Zhang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, P.R. China
| |
Collapse
|
11
|
Fanelli GN, Gasparini P, Coati I, Cui R, Pakula H, Chowdhury B, Valeri N, Loupakis F, Kupcinskas J, Cappellesso R, Fassan M. LONG-NONCODING RNAs in gastroesophageal cancers. Noncoding RNA Res 2018; 3:195-212. [PMID: 30533569 PMCID: PMC6257886 DOI: 10.1016/j.ncrna.2018.10.001] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2018] [Revised: 10/26/2018] [Accepted: 10/26/2018] [Indexed: 02/07/2023] Open
Abstract
Despite continuing improvements in multimodal therapies, gastro-esophageal malignances remain widely prevalent in the population and is characterized by poor overall and disease-free survival rates. Due to the lack of understanding about the pathogenesis and absence of reliable markers, gastro-esophageal cancers are associated with delayed diagnosis. The increasing understanding about cancer's molecular landscape in the recent years, offers the possibility of identifying 'targetable' molecular events and in particular facilitates novel treatment strategies and development of biomarkers for early stage diagnosis. At least 98% of our genome is actively transcribed into non-coding RNAs encompassing long non-coding RNAs (lncRNAs) constituted of transcripts longer than 200 nucleotides with no protein-coding capacity. Many studies have demonstrated that lncRNAs are functional genomic elements playing pivotal roles in main oncogenic processes. LncRNA can act at multiple levels developing a complex molecular network that can modulate directly or indirectly the expression of genes involved in tumorigenesis. In this review, we focus on lncRNAs as emerging players in gastro-esophageal carcinogenesis and critically assess their potential as reliable noninvasive biomarkers and in next generation targeted therapies.
Collapse
Affiliation(s)
- Giuseppe Nicolò Fanelli
- Department of Medicine (DIMED), Surgical Pathology & Cytopathology Unit, University of Padua, Padua, PD, Italy
- Department of Oncologic Pathology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Pierluigi Gasparini
- Department of Cancer Biology and Genetics, College of Medicine, The Ohio State University Comprehensive Cancer Center, Columbus, OH, USA
| | - Irene Coati
- Department of Medicine (DIMED), Surgical Pathology & Cytopathology Unit, University of Padua, Padua, PD, Italy
| | - Ri Cui
- Department of Cancer Biology and Genetics, College of Medicine, The Ohio State University Comprehensive Cancer Center, Columbus, OH, USA
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Hubert Pakula
- Department of Oncologic Pathology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Basudev Chowdhury
- Department of Oncologic Pathology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Nicola Valeri
- Division of Molecular Pathology, The Institute of Cancer Research, London, UK
- Department of Medicine, The Royal Marsden NHS Trust, London, UK
| | - Fotios Loupakis
- Oncology Unit, Istituto Oncologico Veneto, IOV-IRCCS, Padua, PD, Italy
| | - Juozas Kupcinskas
- Department of Gastroenterology and Institute for Digestive Research, Lithuanian University of Health Sciences, Kaunas, Lithuania
| | - Rocco Cappellesso
- Department of Medicine (DIMED), Surgical Pathology & Cytopathology Unit, University of Padua, Padua, PD, Italy
| | - Matteo Fassan
- Department of Medicine (DIMED), Surgical Pathology & Cytopathology Unit, University of Padua, Padua, PD, Italy
| |
Collapse
|
12
|
Yang L, Sun K, Chu J, Qu Y, Zhao X, Yin H, Ming L, Wan J, He F. Long non-coding RNA FTH1P3 regulated metastasis and invasion of esophageal squamous cell carcinoma through SP1/NF-kB pathway. Biomed Pharmacother 2018; 106:1570-1577. [PMID: 30119232 DOI: 10.1016/j.biopha.2018.07.129] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2018] [Revised: 07/18/2018] [Accepted: 07/24/2018] [Indexed: 01/28/2023] Open
Abstract
AIMS:: Recent research showed that Long non-protein coding RNA ferritin heavy chain 1 pseudogene 3 (FTH1P3) plays a crucial role in the course of tumor formation. The present study was aimed to explore its role in esophageal squamous cell carcinoma (ESCC). MAIN METHODS Quantitative reverse transcriptase polymerase chain reaction (qRT-PCR) was used to examine the expression levels of FTH1P3, mRNA SP1 and NF-kB in ESCC samples and cell lines. The impact of FTH1P3 knockdown was evaluated by WST-1 assays, colony formation assays, scratch wound assays, migration and invasion assays. KEY FINDINGS FTH1P3 was significantly upregulated in ESCC tissues and cells (P < 0.001). Knockdown of FTH1P3 notably decreased the proliferation, migration, and invasion capacity of ESCC cells. Silencing of FTH1P3 decreased the expression of specificity protein 1 (Sp1) and NF-kB (p65) in EC9706 and EC1. SIGNIFICANCE FTH1P3 plays a crucial role in ESCC tumorigenesis, and can be used as a potential therapeutic target for ESCC.
Collapse
Affiliation(s)
- Lijun Yang
- Department of Medical Laboratory, The First Affiliated Hospital, Zhengzhou University, Zhengzhou, Henan Province, China.
| | - Kaiyan Sun
- Department of Medical Laboratory, The First Affiliated Hospital, Zhengzhou University, Zhengzhou, Henan Province, China.
| | - Jie Chu
- Department of Medical Laboratory, The First Affiliated Hospital, Zhengzhou University, Zhengzhou, Henan Province, China.
| | - Yunhui Qu
- Department of Medical Laboratory, The First Affiliated Hospital, Zhengzhou University, Zhengzhou, Henan Province, China.
| | - Xue Zhao
- Institute of Medical and Pharmaceutical Science, Zhengzhou University, Zhengzhou, Henan Province, China.
| | - Huiqing Yin
- Department of Medical Laboratory, The First Affiliated Hospital, Zhengzhou University, Zhengzhou, Henan Province, China.
| | - Liang Ming
- Department of Medical Laboratory, The First Affiliated Hospital, Zhengzhou University, Zhengzhou, Henan Province, China.
| | - Junhu Wan
- Department of Medical Laboratory, The First Affiliated Hospital, Zhengzhou University, Zhengzhou, Henan Province, China.
| | - Fucheng He
- Department of Medical Laboratory, The First Affiliated Hospital, Zhengzhou University, Zhengzhou, Henan Province, China.
| |
Collapse
|
13
|
Wang Y, Hou Z, Li D. Long noncoding RNA UCA1 promotes anaplastic thyroid cancer cell proliferation via miR‑135a‑mediated c‑myc activation. Mol Med Rep 2018; 18:3068-3076. [PMID: 30015867 DOI: 10.3892/mmr.2018.9276] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2017] [Accepted: 05/08/2018] [Indexed: 11/05/2022] Open
Abstract
The long non‑coding RNA (lncRNA), urothelial carcinoma‑associated 1 (UCA1), has been demonstrated to be dysregulated and serves a role in the progression of several cancer types. However, the exact effects and underlying molecular mechanisms of UCA1 in anaplastic thyroid cancer (ATC) remain unknown. The aim of the present study was to investigate the detailed function and the mechanism of UCA1 in the regulation of ATC cell progression. The present study identified that the expression levels of UCA1, in ATC cell lines and tissues, were significantly upregulated compared with normal human thyroid cell line and adjacent non‑cancerous tissues, respectively. UCA1 knockdown significantly inhibited ATC cell viability, proliferation, migration and invasion and the expression level of c‑myc proto‑oncogene (c‑myc) in vitro, and suppressed ATC tumor growth in vivo. In addition, using luciferase assays, it was confirmed that miR‑135a directly bound to UCA1 and the 3' untranslated region of c‑myc, and UCA1 competed with c‑myc for miR‑135a binding. miR‑135a inhibition may upregulate c‑myc expression, however, the upregulation of c‑myc may be partially reduced by short hairpin UCA1. The present results illustrated that UCA1 promoted ATC cell proliferation through acting as a competing endogenous RNA by binding miR‑135a. In conclusion, in the present study, UCA1 served as an oncogenic long non‑coding RNA promoting ATC cell proliferation and may be a potential target for human ATC treatment.
Collapse
Affiliation(s)
- Yonghui Wang
- Department of Breast Surgery, Weifang People's Hospital, Weifang, Shandong 261041, P.R. China
| | - Zhengguang Hou
- Department of Breast Surgery, Linyi Central Hospital, Linyi, Shandong 276400, P.R. China
| | - Dong Li
- Department of No. 1 Endocrinology, Affiliated Hospital of Jining Medical University, Jining, Shandong 272029, P.R. China
| |
Collapse
|
14
|
Li D, Cui C, Chen J, Hu Z, Wang Y, Hu D. Long non‑coding RNA UCA1 promotes papillary thyroid cancer cell proliferation via miR‑204‑mediated BRD4 activation. Mol Med Rep 2018; 18:3059-3067. [PMID: 30015945 DOI: 10.3892/mmr.2018.9246] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2018] [Accepted: 06/19/2018] [Indexed: 11/06/2022] Open
Abstract
Long non‑coding RNA (lncRNA) urothelial carcinoma‑associated 1 (UCA1) has been used in tumor development and progression in many types of cancer. However, the function and mechanism underlying the action of UCA1 in papillary thyroid cancer (PTC) remains unclear. Therefore, these topics were investigated in the present study by in vitro and in vivo experiments. It was demonstrated that the expression level of UCA1 was more significantly upregulated in PTC cell lines and tissues when compared with the immortal human thyroid follicular cell line and adjacent normal tissues, respectively. UCA1 knockdown significantly inhibited PTC cell viability, colony formation and the bromodomain containing 4 (BRD4) expression level in vitro, and retarded PTC tumor growth in vivo. In the previous study, microRNA (miR)‑204 inhibited thyroid cancer progression and was regulated by UCA1 in other types of cancer. In addition, by conducting dual luciferase reporter assays, it was confirmed that miR‑204 directly binds to UCA1 and the 3'‑untranslated region of BRD4. Furthermore, UCA1 competed with BRD4 for miR‑204 binding. miR‑204 knockdown enhanced BRD4 expression, which can be partially restored by short hairpin‑UCA1. The results of the present study illustrated that UCA1 promotes PTC progression by acting as a competing endogenous RNA by sponging miR‑204. In conclusion, UCA1 may be regarded as an oncogenic lncRNA, promoting PTC cell proliferation, and be a potential target for human PTC treatment.
Collapse
Affiliation(s)
- Dong Li
- Department of Endocrinology 1, Affiliated Hospital of Jining Medical University, Jining, Shandong 272029, P.R. China
| | - Chuanyou Cui
- Department of Breast, Thyroid and Hernia Surgery, Liaocheng People's Hospital, Liaocheng, Shandong 252000, P.R. China
| | - Jing Chen
- Department of Endocrinology 1, Affiliated Hospital of Jining Medical University, Jining, Shandong 272029, P.R. China
| | - Zhifang Hu
- Department of Endocrinology 1, Affiliated Hospital of Jining Medical University, Jining, Shandong 272029, P.R. China
| | - Yonghui Wang
- Department of Breast Surgery, Weifang People's Hospital, Weifang, Shandong 261041, P.R. China
| | - Dongyu Hu
- Department of Oncology, Affiliated Hospital of Jining Medical University, Jining, Shandong 272029, P.R. China
| |
Collapse
|
15
|
Hahne JC, Valeri N. Non-Coding RNAs and Resistance to Anticancer Drugs in Gastrointestinal Tumors. Front Oncol 2018; 8:226. [PMID: 29967761 PMCID: PMC6015885 DOI: 10.3389/fonc.2018.00226] [Citation(s) in RCA: 59] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2018] [Accepted: 05/31/2018] [Indexed: 12/12/2022] Open
Abstract
Non-coding RNAs are important regulators of gene expression and transcription. It is well established that impaired non-coding RNA expression especially the one of long non-coding RNAs and microRNAs is involved in a number of pathological conditions including cancer. Non-coding RNAs are responsible for the development of resistance to anticancer treatments as they regulate drug resistance-related genes, affect intracellular drug concentrations, induce alternative signaling pathways, alter drug efficiency via blocking cell cycle regulation, and DNA damage response. Furthermore, they can prevent therapeutic-induced cell death and promote epithelial-mesenchymal transition (EMT) and elicit non-cell autonomous mechanisms of resistance. In this review, we summarize the role of non-coding RNAs for different mechanisms resulting in drug resistance (e.g., drug transport, drug metabolism, cell cycle regulation, regulation of apoptotic pathways, cancer stem cells, and EMT) in the context of gastrointestinal cancers.
Collapse
Affiliation(s)
- Jens C. Hahne
- Division of Molecular Pathology, The Institute of Cancer Research, London, United Kingdom
| | - Nicola Valeri
- Division of Molecular Pathology, The Institute of Cancer Research, London, United Kingdom
- Department of Medicine, The Royal Marsden NHS Trust, London, United Kingdom
| |
Collapse
|
16
|
Zong W, Ju S, Jing R, Cui M. Long non-coding RNA-mediated regulation of signaling pathways in gastric cancer. ACTA ACUST UNITED AC 2018; 56:1828-1837. [PMID: 29804098 DOI: 10.1515/cclm-2017-1139] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2017] [Accepted: 04/23/2018] [Indexed: 12/18/2022]
Abstract
Abstract
Gastric cancer (GC) is one of the most common cancers globally. Because of the high frequency of tumor recurrence, or metastasis, after surgical resection, the prognosis of patients with GC is poor. Therefore, exploring the mechanisms underlying GC is of great importance. Recently, accumulating evidence has begun to show that dysregulated long non-coding RNAs (lncRNAs) participate in the progression of GC via several typical signaling pathways, such as the AKT and MAPK signaling pathways. Moreover, the interactions between lncRNAs and microRNAs appear to represent a novel mechanism in the pathogenesis of GC. This review provides a synopsis of the latest research relating to lncRNAs and associated signaling pathways in GC.
Collapse
Affiliation(s)
- Wei Zong
- Department of Laboratory Medicine , Affiliated Hospital of Nantong University , Nantong , P.R. China
| | - Shaoqing Ju
- Department of Laboratory Medicine , Affiliated Hospital of Nantong University , Nantong , P.R. China
| | - Rongrong Jing
- Department of Laboratory Medicine , Affiliated Hospital of Nantong University , No. 20, Xisi Road , Nantong 226001 , P.R. China
| | - Ming Cui
- Department of Laboratory Medicine , Affiliated Hospital of Nantong University , No. 20, Xisi Road , Nantong 226001 , P.R. China , Phone: 0086-513-85052105
| |
Collapse
|
17
|
Wu Y, Chen X, Liang Y, Li J, Zhang K, Dai L, Guan X, Wang K, Bai Y. Overexpression of long non-coding RNA SOX2OT promotes esophageal squamous cell carcinoma growth. Cancer Cell Int 2018; 18:76. [PMID: 29849506 PMCID: PMC5970475 DOI: 10.1186/s12935-018-0570-7] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2018] [Accepted: 05/12/2018] [Indexed: 11/10/2022] Open
Abstract
Background SOX2 overlapping transcript (SOX2OT) has been reported to be an important lncRNA in various cancers. SOX2 is embedded in an intron of the SOX2OT gene. But the role of SOX2OT in esophageal squamous cell carcinoma (ESCC) and the association between SOX2OT and SOX2 remain unclear. Methods Quantitative PCR (qPCR) was used to detect the expression of SOX2OT and SOX2 in ESCC tissues and cells. The isoforms of SOX2OT were identified by PCR and confirmed by sequencing. CCK-8 and Edu assays were performed to investigate the effects of SOX2OT on cell growth. The relationship between SOX2OT and SOX2 was explored by luciferase reporter assay. Results Both SOX2OT and SOX2 were upregulated in ESCC tissues and cells. SOX2OT expression was positively associated with SOX2 expression in ESCC tissues. NR_004053 was one of the major SOX2OT transcripts aberrantly expressed in ESCC tissues and cells. Overexpression of SOX2OT (NR_004053) promoted ESCC cell growth, antagonized the effect of DDP and increased cell proliferation ratio. Ectopic expression of SOX2 could increase the luciferase activity of SOX2OT-pGL3/Basic and SOX2OT expression, while overexpression of SOX2OT (NR_004053) had no effect on SOX2 expression. Conclusion Our study demonstrates that the major isoform of SOX2OT in ESCC, SOX2OT (NR_004053) contributes to cell growth. SOX2 promotes SOX2OT expression at transcriptional level.
Collapse
Affiliation(s)
- Yuanyuan Wu
- 1Department of Medical Genetics, College of Basic Medical Science, Third Military Medical University or Army Medical University, Chongqing, China
| | - Xuedan Chen
- 1Department of Medical Genetics, College of Basic Medical Science, Third Military Medical University or Army Medical University, Chongqing, China
| | - Yan Liang
- 1Department of Medical Genetics, College of Basic Medical Science, Third Military Medical University or Army Medical University, Chongqing, China
| | - Juan Li
- 1Department of Medical Genetics, College of Basic Medical Science, Third Military Medical University or Army Medical University, Chongqing, China
| | - Kun Zhang
- 2Department of Pathgenic Biology, College of Basic Medical Science, Third Military Medical University or Army Medical University, Chongqing, China
| | - Limeng Dai
- 1Department of Medical Genetics, College of Basic Medical Science, Third Military Medical University or Army Medical University, Chongqing, China
| | - Xingying Guan
- 1Department of Medical Genetics, College of Basic Medical Science, Third Military Medical University or Army Medical University, Chongqing, China
| | - Kai Wang
- 1Department of Medical Genetics, College of Basic Medical Science, Third Military Medical University or Army Medical University, Chongqing, China
| | - Yun Bai
- 1Department of Medical Genetics, College of Basic Medical Science, Third Military Medical University or Army Medical University, Chongqing, China
| |
Collapse
|
18
|
Huang J, Li Y, Lu Z, Che Y, Sun S, Mao S, Lei Y, Zang R, Li N, Sun N, He J. Long non-coding RNA GAS5 is induced by interferons and plays an antitumor role in esophageal squamous cell carcinoma. Cancer Med 2018; 7:3157-3167. [PMID: 29745062 PMCID: PMC6051207 DOI: 10.1002/cam4.1524] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2018] [Revised: 04/02/2018] [Accepted: 04/05/2018] [Indexed: 01/01/2023] Open
Abstract
The long non-coding RNA GAS5 has been reported as a tumor suppressor in many cancers. However, its functions and mechanisms remain largely unknown in esophageal squamous cell carcinoma (ESCC). In this study, we found that GAS5 was over-expressed in ESCC tissue compared with that in normal esophageal tissue in a public database. Functional studies showed that GAS5 could inhibit ESCC cell proliferation, migration and invasion in vitro. Further analysis revealed that GAS5 was regulated by interferon (IFN) responses via the JAK-STAT pathway. Moreover, as an IFN-stimulated gene (ISG), GAS5 was a positive regulator of IFN responses. The feedback loop between GAS5 and the IFN signaling pathway plays an important antitumor role in ESCC, thus providing novel potential therapeutic targets.
Collapse
Affiliation(s)
- Jianbing Huang
- Department of Thoracic Surgery, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yuan Li
- Department of Thoracic Surgery, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Zhiliang Lu
- Department of Thoracic Surgery, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yun Che
- Department of Thoracic Surgery, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Shouguo Sun
- Department of Thoracic Surgery, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Shuangshuang Mao
- Department of Thoracic Surgery, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yuanyuan Lei
- Department of Thoracic Surgery, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Ruochuan Zang
- Department of Thoracic Surgery, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Ning Li
- Department of Thoracic Surgery, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Nan Sun
- Department of Thoracic Surgery, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jie He
- Department of Thoracic Surgery, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
19
|
Li Y, Jing F, Ding Y, He Q, Zhong Y, Fan C. Long noncoding RNA CCAT1 polymorphisms are associated with the risk of colorectal cancer. Cancer Genet 2018; 222-223:13-19. [PMID: 29666003 DOI: 10.1016/j.cancergen.2018.02.003] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2017] [Revised: 01/18/2018] [Accepted: 02/13/2018] [Indexed: 12/27/2022]
Abstract
Colorectal cancer associated transcript 1 (CCAT1) is a novel long noncoding RNA, whose overexpression is evident in both early phase of tumorigenesis and later disease stages in colorectal cancer (CRC). No study has explored the relationship between CCAT1 polymorphisms and CRC risk. In the present study, a case-control study was conducted to investigate the association between CCAT1 polymorphisms and CRC risk in Chinese population. We identified that CCAT1 rs67085638 polymorphism was associated with an increased risk of CRC (OR = 1.72, 95%CI = 1.14-2.58, P = 0.009 in heterozygote codominant model; OR = 1.67, 95%CI = 1.13-2.47, P = 0.010 in dominant model). Moreover, CCAT1 rs7013433 polymorphism was associated with late clinical stage (OR = 1.82, 95%CI = 1.16-2.86, P = 0.009 in heterozygote codominant model; OR = 1.72, 95%CI = 1.13-2.63, P = 0.012 in dominant model). Our finding proposed a link between CCAT1 polymorphisms with CRC risk as well as different clinical stages.
Collapse
Affiliation(s)
- Yingjun Li
- Department of Public Health, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Fangyuan Jing
- Department of Public Health, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Ye Ding
- Department of Public Health, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Qingfang He
- Department of Chronic Non-Communicable Diseases Control and Prevention, Zhejiang Provincial Center for Disease Control and Prevention, Hangzhou, Zhejiang, China
| | - Yaohong Zhong
- Department of Public Health, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Chunhong Fan
- Department of Public Health, Hangzhou Medical College, Hangzhou, Zhejiang, China.
| |
Collapse
|
20
|
Liang Y, Li Y, Song X, Zhang N, Sang Y, Zhang H, Liu Y, Chen B, Zhao W, Wang L, Guo R, Yu Z, Yang Q. Long noncoding RNA LINP1 acts as an oncogene and promotes chemoresistance in breast cancer. Cancer Biol Ther 2018; 19:120-131. [PMID: 29293402 PMCID: PMC5790352 DOI: 10.1080/15384047.2017.1394543] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2017] [Revised: 10/06/2017] [Accepted: 10/15/2017] [Indexed: 01/06/2023] Open
Abstract
Recent studies have shown that long non-coding RNAs (lncRNAs) are involved in a number of biological processes; however, further study is still warranted to comprehensively reveal their functions. In this study, we showed that the lncRNA in non-homologous end joining (NHEJ) pathway 1 (LINP1) was related to breast cancer cell proliferation, metastasis and chemoresistance. Loss- and gain-of function studies were used to assess the role of LINP1 in promoting breast cancer progression. LINP1 knockdown mitigated breast cancer cell growth by inducing G1-phase cell cycle arrest and apoptosis. LINP1 also promoted breast cancer cell metastasis and influenced the expression of epithelial-mesenchymal transition-related markers. We identified p53 as a regulator of LINP1, and LINP1 overexpression could restore the metastatic effects of p53. Furthermore, LINP1 was upregulated in doxorubicin- and 5-fluorouracil-resistant cells and induced chemoresistance. We also observed that LINP1 enrichment played a critical functional role in chemoresistance by inhibiting chemotherapeutics-induced apoptosis. Moreover, LINP1 in tumors was associated with lower overall survival and disease-free survival. In conclusion, LINP1 may serve as a potential oncogene and chemoresistance-related regulator of breast cancer cells, suggesting that LINP1 might be a potent therapeutic target and might reduce chemoresistance in breast cancer.
Collapse
Affiliation(s)
- Yiran Liang
- Department of Breast Surgery, Qilu Hospital, Shandong University, Jinan, Shandong, P.R. China
| | - Yaming Li
- Department of Breast Surgery, Qilu Hospital, Shandong University, Jinan, Shandong, P.R. China
| | - Xiaojin Song
- Department of Breast Surgery, Qilu Hospital, Shandong University, Jinan, Shandong, P.R. China
| | - Ning Zhang
- Department of Breast Surgery, Qilu Hospital, Shandong University, Jinan, Shandong, P.R. China
| | - Yuting Sang
- Department of Breast Surgery, Qilu Hospital, Shandong University, Jinan, Shandong, P.R. China
| | - Hanwen Zhang
- Department of Breast Surgery, Qilu Hospital, Shandong University, Jinan, Shandong, P.R. China
| | - Ying Liu
- Department of Breast Surgery, Qilu Hospital, Shandong University, Jinan, Shandong, P.R. China
| | - Bing Chen
- Pathology Tissue Bank, Qilu Hospital, Shandong University, Jinan, Shandong, P.R. China
| | - Wenjing Zhao
- Pathology Tissue Bank, Qilu Hospital, Shandong University, Jinan, Shandong, P.R. China
| | - Lijuan Wang
- Pathology Tissue Bank, Qilu Hospital, Shandong University, Jinan, Shandong, P.R. China
| | - Renbo Guo
- Department of Urology, Shandong Cancer Hospital Affiliated to Shandong University, Jinan, Shandong, P.R. China
| | - Zhigang Yu
- Department of Breast Surgery, The Second Hospital of Shandong University, Jinan, Shandong, P.R. China
| | - Qifeng Yang
- Department of Breast Surgery, Qilu Hospital, Shandong University, Jinan, Shandong, P.R. China
| |
Collapse
|
21
|
Alaei S, Sadeghi B, Najafi A, Masoudi-Nejad A. LncRNA and mRNA integration network reconstruction reveals novel key regulators in esophageal squamous-cell carcinoma. Genomics 2018; 111:76-89. [PMID: 29317304 DOI: 10.1016/j.ygeno.2018.01.003] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2017] [Revised: 01/05/2018] [Accepted: 01/05/2018] [Indexed: 12/19/2022]
Abstract
Many experimental and computational studies have identified key protein coding genes in initiation and progression of esophageal squamous cell carcinoma (ESCC). However, the number of researches that tried to reveal the role of long non-coding RNAs (lncRNAs) in ESCC has been limited. LncRNAs are one of the important regulators of cancers which are transcribed dominantly in the genome and in various conditions. The main goal of this study was to use a systems biology approach to predict novel lncRNAs as well as protein coding genes associated with ESCC and assess their prognostic values. By using microarray expression data for mRNAs and lncRNAs from a large number of ESCC patients, we utilized "Weighted Gene Co-expression Network Analysis" (WGCNA) method to make a big coding-non-coding gene co-expression network, and discovered important functional modules. Gene set enrichment and pathway analysis revealed major biological processes and pathways involved in these modules. After selecting some protein coding genes involved in biological processes and pathways related to cancer, we used "LncTar", a computational tool to predict potential interactions between these genes and lncRNAs. By combining interaction results with Pearson correlations, we introduced some novel lncRNAs with putative key regulatory roles in the network. Survival analysis with Kaplan-Meier estimator and Log-rank test statistic confirmed that most of the introduced genes are associated with poor prognosis in ESCC. Overall, our study reveals novel protein coding genes and lncRNAs associated with ESCC, along with their predicted interactions. Based on the promising results of survival analysis, these genes can be used as good estimators of patients' survival, or even can be analyzed further as new potential signatures or targets for the therapy of ESCC disease.
Collapse
Affiliation(s)
- Shervin Alaei
- Laboratory of Systems Biology and Bioinformatics (LBB), Institute of Biochemistry and Biophysics, University of Tehran, Tehran, Iran
| | - Balal Sadeghi
- Food Hygiene and Public Health Department, Faculty of Veterinary Medicine, Shahid Bahonar University of Kerman, Kerman, Iran
| | - Ali Najafi
- Molecular Biology Research Center, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Ali Masoudi-Nejad
- Laboratory of Systems Biology and Bioinformatics (LBB), Institute of Biochemistry and Biophysics, University of Tehran, Tehran, Iran.
| |
Collapse
|
22
|
Liang Y, Wu Y, Chen X, Zhang S, Wang K, Guan X, Yang K, Li J, Bai Y. A novel long noncoding RNA linc00460 up-regulated by CBP/P300 promotes carcinogenesis in esophageal squamous cell carcinoma. Biosci Rep 2017; 37:BSR20171019. [PMID: 28939763 PMCID: PMC5964888 DOI: 10.1042/bsr20171019] [Citation(s) in RCA: 71] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2017] [Revised: 08/30/2017] [Accepted: 09/19/2017] [Indexed: 12/12/2022] Open
Abstract
Esophageal cancer is one of the leading causes of cancer-related mortality because of poor prognosis. Long noncoding RNAs (lncRNAs) have been gradually demonstrated to play critical roles in cancer development. We identified a novel long noncoding RNA named linc00460 by microarray analysis using esophageal squamous cell carcinoma (ESCC) clinical samples, which has not been studied before. Our research indicated that linc00460 was overexpressed in the majority of tumor tissues and ESCC cell lines. Linc00460 expression was positively correlated with ESCC TNM stage, lymph node metastasis, and predicted poor prognosis. In vitro experiments showed that linc00460 depletion suppressed ESCC cell growth through regulating cell proliferation and cell cycle; in additional, linc00460 depletion accelerated ESCC cell apoptosis. We further revealed that linc00460 overexpression was manipulated by transcriptional co-activator CBP/P300 through histone acetylation. Given the high expression and important biological functions of linc00460, we suggest that linc00460 works as an oncogene and might be a valuable prognostic biomarker for ESCC diagnosis and treatment.
Collapse
Affiliation(s)
- Yan Liang
- Department of Medical Genetics, College of Basic Medical Science, Third Military Medical University, Gaotanyan Street, Shapingba District, Chongqing, China
| | - Yuanyuan Wu
- Department of Medical Genetics, College of Basic Medical Science, Third Military Medical University, Gaotanyan Street, Shapingba District, Chongqing, China
| | - Xuedan Chen
- Department of Medical Genetics, College of Basic Medical Science, Third Military Medical University, Gaotanyan Street, Shapingba District, Chongqing, China
| | - Shixin Zhang
- Department of Cardiothoracic Surgery, Southwest Hospital, Third Military Medical University, Gaotanyan Street, Shapingba District, Chongqing, China
| | - Kai Wang
- Department of Medical Genetics, College of Basic Medical Science, Third Military Medical University, Gaotanyan Street, Shapingba District, Chongqing, China
| | - Xingying Guan
- Department of Medical Genetics, College of Basic Medical Science, Third Military Medical University, Gaotanyan Street, Shapingba District, Chongqing, China
| | - Kang Yang
- Department of Cardiothoracic Surgery, Southwest Hospital, Third Military Medical University, Gaotanyan Street, Shapingba District, Chongqing, China
| | - Juan Li
- Department of Medical Genetics, College of Basic Medical Science, Third Military Medical University, Gaotanyan Street, Shapingba District, Chongqing, China
| | - Yun Bai
- Department of Medical Genetics, College of Basic Medical Science, Third Military Medical University, Gaotanyan Street, Shapingba District, Chongqing, China
| |
Collapse
|
23
|
Li Z, Zhou Y, Tu B, Bu Y, Liu A, Kong J. Long noncoding RNA MALAT1 affects the efficacy of radiotherapy for esophageal squamous cell carcinoma by regulating Cks1 expression. J Oral Pathol Med 2017; 46:583-590. [PMID: 27935117 DOI: 10.1111/jop.12538] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/28/2016] [Indexed: 12/13/2022]
Abstract
OBJECTIVE Long noncoding RNA metastasis-associated lung adenocarcinoma transcript 1 (MALAT1) has been well studied in the progression of many malignancies. However, its association with the radioresistance of tumors has not been well understood yet. This study tried to explore the role of MALAT1 in regulating the radiosensitivity of esophageal cancer (EC), especially esophageal squamous cell carcinoma (ESCC), involving its regulation on Cks1 expression. METHODS KYSE150 cells were subcutaneously inoculated into nude mice to establish ESCC xenografts. Real-time PCR and Western blot analysis were performed to detect the expression of MALAT1 and Cks1 in irradiated xenografts and cells. Functional analysis was performed in both EC9706 and KYSE150 cells via the transfection of corresponding plasmids or small interfering RNAs (siRNAs). Irradiation-induced damage was examined by the detection of cell viability and apoptosis using MTT and TUNEL assays, respectively. RESULTS Both MALAT1 and Cks1 were downregulated in irradiated xenografts and cells. Cks1FER1L4 showed significant downregulation. Overexpression of MALAT1 inhibited irradiation-induced decrease in cell viability, increase in apoptosis, and downregulation of Cks1. Cks1 expression was also downregulated by MALAT1 siRNA, while Cks1 siRNA strongly recovered MALAT1-induced radioresistance in vitro. Moreover, better tumor growth, accompanied by Cks1 upregulation, was observed in KYSE150 xenografts with MALAT1 overexpression, especially under radiation treatment. CONCLUSION MALAT1 acted as one positive regulator of the radioresistance of ESCC, at least partly due to its promotion on Cks1 expression. Furthermore, MALAT1-targeted therapies showed great potential in enhancing the radiotherapeutic effect on ESCC.
Collapse
Affiliation(s)
- Zhijun Li
- Department of Radiation Oncology, Inner Mongolia Autonomous Region Peoples Hospital, Hohhot, China
| | - Yang Zhou
- Department of Radiation Oncology, Inner Mongolia Autonomous Region Peoples Hospital, Hohhot, China
| | - Bo Tu
- Department of Radiation Oncology, Inner Mongolia Autonomous Region Peoples Hospital, Hohhot, China
| | - Yu Bu
- Department of Radiation Oncology, Inner Mongolia Autonomous Region Peoples Hospital, Hohhot, China
| | - Aqiu Liu
- Department of Radiation Oncology, Inner Mongolia Autonomous Region Peoples Hospital, Hohhot, China
| | - Jianmin Kong
- Department of Radiation Oncology, Inner Mongolia Autonomous Region Peoples Hospital, Hohhot, China
| |
Collapse
|
24
|
Huang ZL, Chen RP, Zhou XT, Zhan HL, Hu MM, Liu B, Wu GD, Wu LF. Long non-coding RNA MEG3 induces cell apoptosis in esophageal cancer through endoplasmic reticulum stress. Oncol Rep 2017; 37:3093-3099. [PMID: 28405686 DOI: 10.3892/or.2017.5568] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2016] [Accepted: 03/27/2017] [Indexed: 02/05/2023] Open
Abstract
Long non-coding RNAs (lncRNAs) play important roles in diverse biological processes, such as cell growth, apoptosis and migration. Although downregulation of lncRNA MEG3 has been identified in several cancers, little is known about its role in esophageal squamous cell carcinoma (ESCC). The aim of the present study was to detect MEG3 expression in clinical ESCC tissues, investigate its biological functions and the endoplasmic reticulum (ER) stress-relative mechanism. MEG3 expression levels were detected by qRT-PCR in both tumor tissues and adjacent non-tumor tissues from 28 ESCC patients. PcDNA3.1-MEG3 recombinant plasmids were constructed and transfected to EC109 cells. Cell growth was analyzed by CCK-8 assay. Cell apoptosis was analyzed by fluorescence microscope and Annexin V/PI assay. The protein expression was determined by western blot analysis. The results showed that MEG3 decreased significantly in ESCC tissues relative to adjacent normal tissues. PcDNA3.1-MEG3 plasmids were successfully constructed and the expression level of MEG3 significantly increased after MEG3 transfection to EC109 cells. Ectopic expression of MEG3 inhibited EC109 cell proliferation and induced apoptosis in vitro. MEG3 overexpression increased the expression of ER stress‑related proteins (GRP78, IRE1, PERK, ATF6, CHOP and cleaved‑caspase-3). Our results first demonstrate that MEG3 is downregulated in ESCC tissues. MEG3 was able to inhibit cell growth and induced apoptosis in EC109 cells, most probably via activation of the ER stress pathway.
Collapse
Affiliation(s)
- Zhen-Lun Huang
- Department of Gastroenterology, Second Affiliated Hospital, Shantou University Medical College, Shantou, Guangdong 515041, P.R. China
| | - Rui-Pei Chen
- Department of Gastroenterology, Second Affiliated Hospital, Shantou University Medical College, Shantou, Guangdong 515041, P.R. China
| | - Xiao-Tao Zhou
- Department of Gastroenterology, Second Affiliated Hospital, Shantou University Medical College, Shantou, Guangdong 515041, P.R. China
| | - Hao-Lian Zhan
- Department of Gastroenterology, Second Affiliated Hospital, Shantou University Medical College, Shantou, Guangdong 515041, P.R. China
| | - Min-Min Hu
- Department of Gastroenterology, Second Affiliated Hospital, Shantou University Medical College, Shantou, Guangdong 515041, P.R. China
| | - Bin Liu
- Department of Neurosurgery, Second Affiliated Hospital, Shantou University Medical College, Shantou, Guangdong 515041, P.R. China
| | - Guan-Di Wu
- First Clinical Medical College, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Ling-Fei Wu
- Department of Gastroenterology, Second Affiliated Hospital, Shantou University Medical College, Shantou, Guangdong 515041, P.R. China
| |
Collapse
|
25
|
Xia B, Hou Y, Chen H, Yang S, Liu T, Lin M, Lou G. Long non-coding RNA ZFAS1 interacts with miR-150-5p to regulate Sp1 expression and ovarian cancer cell malignancy. Oncotarget 2017; 8:19534-19546. [PMID: 28099946 PMCID: PMC5386703 DOI: 10.18632/oncotarget.14663] [Citation(s) in RCA: 69] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2016] [Accepted: 11/12/2016] [Indexed: 12/22/2022] Open
Abstract
We reported that long non-coding RNA ZFAS1 was upregulated in epithelial ovarian cancer tissues, and was negatively correlated to the overall survival rate of patients with epithelial ovarian cancer in this study. While depletion of ZFAS1 inhibited proliferation, migration, and development of chemoresistance, overexpression of ZFAS1 exhibited an even higher proliferation rate, migration activity, and chemoresistance in epithelial ovarian cancer cell lines. We further found miR-150-5p was a potential target of ZFAS1, which was downregulated in epithelial ovarian cancer tissue. MiR-150-5p subsequently inhibited expression of transcription factor Sp1, as evidence by luciferase assays. Inhibition of miR-150-5p rescued the suppressed proliferation and migration induced by depletion of ZFAS1 in epithelial ovarian cancer cells, at least in part. Taken together, our findings revealed a critical role of ZFAS1/miR-150-5p/Sp1 axis in promoting proliferation rate, migration activity, and development of chemoresistance in epithelial ovarian cancer. And ZFAS1/miR-150-5p may serve as novel markers and therapeutic targets of epithelial ovarian cancer.
Collapse
Affiliation(s)
- Bairong Xia
- Department of Gynecology, the Affiliated Tumor Hospital, Harbin Medical University, Harbin, China
| | - Yan Hou
- Department of Biostatistics, Public Health School, Harbin Medical University, Harbin, China
| | - Hong Chen
- Department of Gynecology, the Affiliated Tumor Hospital, Harbin Medical University, Harbin, China
| | - Shanshan Yang
- Department of Gynecology, the Affiliated Tumor Hospital, Harbin Medical University, Harbin, China
| | - Tianbo Liu
- Department of Gynecology, the Affiliated Tumor Hospital, Harbin Medical University, Harbin, China
| | - Mei Lin
- Department of Gynecology, the Affiliated Tumor Hospital, Harbin Medical University, Harbin, China
| | - Ge Lou
- Department of Gynecology, the Affiliated Tumor Hospital, Harbin Medical University, Harbin, China
| |
Collapse
|
26
|
Wu J, Wang D. Long noncoding RNA TCF7 promotes invasiveness and self-renewal of human non-small cell lung cancer cells. Hum Cell 2016; 30:23-29. [DOI: 10.1007/s13577-016-0147-5] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2016] [Accepted: 09/22/2016] [Indexed: 01/16/2023]
|