1
|
Xu X, Zhao Y, Duan G, Du B. Downregulation of MIAT reduces the proliferation and migratory and invasive abilities of retinoblastoma cells by sponging miR-665 and regulating LASP1. Exp Ther Med 2021; 22:1342. [PMID: 34630696 DOI: 10.3892/etm.2021.10777] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Accepted: 04/28/2021] [Indexed: 12/21/2022] Open
Abstract
Long non-coding RNAs (lncRNAs) can function as onco-lncRNAs in several types of human cancer, including retinoblastoma (Rb). The present study investigated the potential role and regulatory mechanism of the lncRNA myocardial infarction-associated transcript (MIAT) in Rb. To do so, the expression levels of MIAT, microRNA (miR)-665, and LIM and SH3 protein 1 (LASP1) in Rb tissues from patients or Rb cells were analysed using reverse transcription quantitative PCR. The interactions between miR-665 and MIAT/LASP1 were confirmed by the dual-luciferase reporter assay. MTT, Transwell (to assess migration and invasion) and western blotting assays were used to explore the functions of the MIAT/miR-665/LASP1 axis on Rb progression in vitro. The results of the present study indicated that MIAT targeted miR-665. In Rb tissues and cell lines, high expression of MIAT was observed, whereas miR-665 was downregulated in Rb tissues. Furthermore, the proliferation and migratory and invasive abilities of Rb Y79 and HXO-RB44 cells were decreased following MIAT downregulation or miR-665 overexpression. In addition, LASP1 was identified as a target gene of miR-665. Both the decreased expression of miR-665 and the elevated expression of LASP1 reversed the suppressive effects of MIAT knockdown on the proliferation and migratory and invasive abilities of Y79 cells. Furthermore, MIAT silencing attenuated the development of Rb by regulating the miR-665/LASP1 axis. Taken together, these findings suggested that MIAT may be considered as a possible therapeutic target for Rb.
Collapse
Affiliation(s)
- Xiabing Xu
- Department of Ophthalmology, No. 215 Hospital of Shaanxi Nuclear Industry, Xianyang, Shaanxi 712000, P.R. China
| | - Yadong Zhao
- Department of Ophthalmology, No. 215 Hospital of Shaanxi Nuclear Industry, Xianyang, Shaanxi 712000, P.R. China
| | - Gang Duan
- Department of Ophthalmology, No. 215 Hospital of Shaanxi Nuclear Industry, Xianyang, Shaanxi 712000, P.R. China
| | - Bo Du
- Department of Ophthalmology, No. 215 Hospital of Shaanxi Nuclear Industry, Xianyang, Shaanxi 712000, P.R. China
| |
Collapse
|
2
|
lncRNA cytoskeleton regulator RNA (CYTOR): Diverse functions in metabolism, inflammation and tumorigenesis, and potential applications in precision oncology. Genes Dis 2021; 10:415-429. [DOI: 10.1016/j.gendis.2021.08.012] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Accepted: 08/20/2021] [Indexed: 12/19/2022] Open
|
3
|
Abstract
Non-small cell lung cancer (NSCLC) accounts for about 85% of all lung cancer cases. The pathogenesis of NSCLC involves complex gene networks that include different types of non-coding RNAs, such as long non-coding RNAs (lncRNAs). The role of lncRNAs in NSCLC is gaining an increasing interest as their function is being explored in various human cancers. Recently, a new oncogenic lncRNA, LINC00152 (cytoskeleton regulator RNA (CYTOR)), has been identified in different tumor types. In NSCLC, the high expression of LINC00152 in tumor tissue and peripheral blood samples has been shown to be associated with worse prognoses of NSCLC patients. Overexpression of LINC00152 has been confirmed to promote the proliferation, invasion, and migration of NSCLC cells in vitro, as well as increase tumor growth in vivo. This review discusses the role of LINC00152 in NSCLC.
Collapse
Affiliation(s)
- Hong Yu
- Cell Biology Laboratory, Jilin Province Institute of Cancer Prevention and Treatment, Jilin Cancer Hospital, Changchun 130012, China
| | - Shu-Bin Li
- Department of Internal Medicine, Southern Branch of Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 102600, China
| |
Collapse
|
4
|
Gao S, Chu Q, Liu X, Zhao X, Qin L, Li G, Liu Q. Long Noncoding RNA HEIH Promotes Proliferation, Migration and Invasion of Retinoblastoma Cells Through miR-194-5p/WEE1 Axis. Onco Targets Ther 2020; 13:12033-12041. [PMID: 33262604 PMCID: PMC7695688 DOI: 10.2147/ott.s268942] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Accepted: 10/12/2020] [Indexed: 12/31/2022] Open
Abstract
Background Abnormally expressed long noncoding RNA (lncRNA) high expression in hepatocellular carcinoma (HEIH) has been implicated in many types of human cancer, and plays crucial roles in tumor development and progression. However, little is known about its function in retinoblastoma. Methods qRT-PCR was used to determine the expression levels of HEIH, miR-194-5p and WEE1 in retinoblastoma tissues and cell lines. The trypan blue exclusion method, colony formation assay, wound-healing assay and transwell invasion assay were performed to evaluate the effects of HEIH, miR-194-5p and WEE1 on cell proliferation, migration and invasion. Bioinformatics analysis, dual-luciferase reporter assay and Western blot were employed to investigate the regulatory relationship among HEIH, miR-194-5p and WEE1. Results We found that HEIH was up-regulated in retinoblastoma tissues and cell lines. Furthermore, high level of HEIH was associated with TNM stage, optic nerve invasion and choroidal invasion of patients with retinoblastoma. Functional studies showed that HEIH knockdown significantly suppressed retinoblastoma cell proliferation, migration and invasion. Mechanistically, HEIH promoted retinoblastoma progression by serving as a sponge of miR-194-5p to regulate WEE1 expression. Conclusion Our work suggests that HEIH acts as an oncogenic lncRNA to promote retinoblastoma proliferation and metastasis, providing a new insight into the retinoblastoma treatment.
Collapse
Affiliation(s)
- Sheng Gao
- Department of Ophthalmology, Nanjing Pukou Central Hospital, Nanjing 211800, People's Republic of China
| | - Qingxia Chu
- Department of Ophthalmology, Nanjing Pukou Central Hospital, Nanjing 211800, People's Republic of China
| | - Xia Liu
- Department of Ophthalmology, Nanjing Pukou Central Hospital, Nanjing 211800, People's Republic of China
| | - Xia Zhao
- Department of Ophthalmology, Tangshan Eye Hospital, Tangshan 063000, People's Republic of China
| | - Libao Qin
- Department of Ophthalmology, Nanjing Pukou Central Hospital, Nanjing 211800, People's Republic of China
| | - Guoliang Li
- Department of Ophthalmology, Nanjing Pukou Central Hospital, Nanjing 211800, People's Republic of China
| | - Qinghuai Liu
- Department of Ophthalmology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, People's Republic of China
| |
Collapse
|
5
|
Wang Y, Xin D, Zhou L. LncRNA LINC00152 Increases the Aggressiveness of Human Retinoblastoma and Enhances Carboplatin and Adriamycin Resistance by Regulating MiR-613/Yes-Associated Protein 1 (YAP1) Axis. Med Sci Monit 2020; 26:e920886. [PMID: 32541644 PMCID: PMC7315805 DOI: 10.12659/msm.920886] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Long noncoding RNA (lncRNA) acts as key regulator in human cancers, including retinoblastoma. However, the function of LINC00152 remains largely unknown in retinoblastoma. Thus, this study aimed to explore the role and molecular mechanisms of LINC00152 in retinoblastoma. MATERIAL AND METHODS The real-time quantitative polymerase chain reaction (RT-qPCR) was used to quantify the expression levels of LINC00152, miR-613 and yes-associated protein 1 (YAP1). The target genes of LINC00152 and miR-613 were identified by dual-luciferase reporter analysis, RNA immunoprecipitation (RIP) and RNA pulldown assays. The viability, apoptosis, and invasion of retinoblastoma cells were assessed by Cell Counting Kit-8, flow cytometry, and Transwell assays, respectively. In addition, western blot was used to test the protein expression in retinoblastoma cells or tissues. Cell sensitivity to carboplatin and adriamycin was analyzed by computing IC₅₀ value. The effects of LINC00152 silencing in vivo were measured by a xenograft experiment. RESULTS LINC00152 was obviously upregulated, while miR-613 was decreased in retinoblastoma tissues and cells. MiR-613, a target of LINC00152, was negatively regulated by LINC00152. Functional experiment further illustrated that silencing of LINC00152 evidently repressed proliferation, invasion, and autophagy while reinforced apoptosis of retinoblastoma cells, besides, retinoblastoma cells were more sensitive to carboplatin and adriamycin after knockdown of LINC00152. Importantly, knockdown of LINC00152-induced effects on retinoblastoma cells could be overturned by introducing miR-613 inhibitor. Downregulation of miR-613 abolished silencing of YAP1-effects on proliferation, apoptosis, invasion, autophagy, and chemoresistance of retinoblastoma cells. The results of the xenograft experiment indicated that LINC00152 silencing impeded tumor growth in vivo. CONCLUSIONS Mechanistically, LINC00152 enhanced the aggressiveness of retinoblastoma and boosted carboplatin and adriamycin resistance by regulating YAP1 by sponging miR-613 in human retinoblastoma.
Collapse
Affiliation(s)
- Ying Wang
- Department of Ophthalmology, Ningbo Eye Hospital, Ningbo, Zhejiang, China (mainland)
| | - Danli Xin
- Department of Ophthalmology, Ningbo Eye Hospital, Ningbo, Zhejiang, China (mainland)
| | - Lei Zhou
- Department of Ophthalmology, Ningbo Eye Hospital, Ningbo, Zhejiang, China (mainland)
| |
Collapse
|
6
|
Zhang S, Long J, Hu Y. Long noncoding RNA LINC00205 enhances the malignant characteristics of retinoblastoma by acting as a molecular sponge of microRNA-665 and consequently increasing HMGB1 expression. Biochem Biophys Res Commun 2020; 526:396-403. [PMID: 32223925 DOI: 10.1016/j.bbrc.2020.03.083] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Accepted: 03/15/2020] [Indexed: 10/24/2022]
Abstract
Long intergenic non-protein-coding RNA 00205 (LINC00205) has been found to play crucial roles in hepatocellular carcinoma progression. In this study, we aimed to determine the expression pattern of LINC00205 in retinoblastoma (RB), to identify its functions in RB progression in detail, and to reveal the underlying mechanisms. Herein, we showed that LINC00205 is highly expressed in RB tissues and cell lines. The LINC00205 upregulation correlated with adverse clinicopathological parameters and shorter overall survival in patients with RB. LINC00205 depletion decreased the proliferative, migratory, and invasive abilities; promoted the apoptosis of RB cells in vitro; and impeded the tumor growth of RB cells in vivo. Mechanism investigation revealed that LINC00205 can act as a competing endogenous RNA by sponging microRNA-665 (miR-665) in RB cells, thereby upregulating miR-665's target: high-mobility group box 1 (HMGB1). Finally, rescue experiments confirmed that enhancing the miR-665-HMGB1 axis output attenuated the influence of the LINC00205 knockdown on RB cells. To sum up, the newly identified LINC00205-miR-665-HMGB1 pathway was systematically studied and may be validated as a potential target for RB diagnosis, prognosis, and therapy.
Collapse
Affiliation(s)
- Shu Zhang
- Department of Ophthalmology, The First People's Hospital of Jinzhou (Yangtze University Affiliated First People's Hospital), Hubei, 434000, PR China
| | - Jian Long
- Department of Oncology, Jingzhou Central Hospital, The Second Clinical Medical College of Yangtze University, Hubei, 434020, PR China.
| | - Yin Hu
- Department of Ophthalmology, Xiangyang Central Hospital, Hubei, 441021, PR China.
| |
Collapse
|
7
|
Shi Y, Sun H. Down-regulation of lncRNA LINC00152 Suppresses Gastric Cancer Cell Migration and Invasion Through Inhibition of the ERK/MAPK Signaling Pathway. Onco Targets Ther 2020; 13:2115-2124. [PMID: 32210577 PMCID: PMC7074822 DOI: 10.2147/ott.s217452] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2019] [Accepted: 02/19/2020] [Indexed: 12/25/2022] Open
Abstract
Purpose The aim of this study was to explore the regulatory role and mechanism of long noncoding RNA LINC00152 in gastric cancer (GC) cells. Methods LINC00152 expression in GC tissues and cells was detected by reverse transcription-polymerase chain reaction (qRT-PCR). MKN45 and MGC-803 cells were selected and assigned into different groups after transfection with si-LINC00152, activated ERK/MAPK signaling pathway (SA), or negative control. Cell proliferation, apoptosis, cycle, migration and invasion were assessed by CCK-8, flow cytometry, Transwell assay and Scratch test, respectively. Western blot analysis was conducted to detect the expression of E-cadherin, N-cadherin and ERK/MAPK signaling pathway protein. Results Compared with the normal tissues, higher expression of LINC00152 was found in GC tissues and LINC00152 was remarkably correlative with clinical stage and lymphatic metastasis. LINC00152 expression in GC cells was higher than that in GES-1 cells. Compared with the NC group, the cell proliferation rate, cells in G2/M phase, migration and invasion abilities as well as the expression of N-cadherin and p-ERK-1/2 were significantly decreased, and the expression of E-cadherin, cells in G0/G1 phase and cell apoptosis rate were significantly increased in the si-LINC00152-1 group. ERK/MAPK signaling pathway activator SA could reverse the biological role of LINC00152 in GC cells. Conclusion These results demonstrated that the interference of LINC00152 expression may inhibit the invasion and migration of GC cells by inhibiting the ERK/MAPK signaling pathway.
Collapse
Affiliation(s)
- Yan Shi
- Department of Hyperbaric Oxygen, Dongying People's Hospital, Dongying, Shandong, 257091, People's Republic of China
| | - Huihui Sun
- Department of Gastroenterology, Jinan First People's Hospital, Jinan, Shandong 250011, People's Republic of China
| |
Collapse
|
8
|
Abstract
Retinoblastoma (Rb) is the most common ocular pediatric malignancy that arises from the retina and is caused by a mutation of the two alleles of the tumor suppressor gene, RB1. Although early detection provides the opportunity of controlling the primary tumor with effective therapies, metastatic activity is fatal. Non-coding RNAs (ncRNAs) have emerged as important modifiers of a plethora of biological mechanisms including those involved in cancer. They are classified into short and long ncRNAs according to their length. Deregulation of all these molecules has also been shown to play a critical role in Rb pathogenesis and progression. It is believed that ncRNAs can provide new insights into novel regulatory mechanisms associated with clinical pathological characteristics, facilitating the development of therapeutic alternatives for the treatment of Rb. In this review, we describe a variety of ncRNAs, which capable of regulating the most likely candidate genes involved in the tumorigenesis of Rb, could prove useful in analyzing different aspects of this cancer.
Collapse
Affiliation(s)
- Meropi Plousiou
- Biosciences Laboratory, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Meldola, Italy
| | - Ivan Vannini
- Biosciences Laboratory, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Meldola, Italy
| |
Collapse
|
9
|
Han N, Zuo L, Chen H, Zhang C, He P, Yan H. Long non-coding RNA homeobox A11 antisense RNA (HOXA11-AS) promotes retinoblastoma progression via sponging miR-506-3p. Onco Targets Ther 2019; 12:3509-3517. [PMID: 31123409 PMCID: PMC6511239 DOI: 10.2147/ott.s195404] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Accepted: 03/16/2019] [Indexed: 12/13/2022] Open
Abstract
Background: Long non-coding RNA homeobox A11 antisense RNA (HOXA11-AS) has been reported to be involved in initiation and development of multiple cancers. However, the detailed biological roles and underlying molecular mechanism of HOXA11-AS remain unclear in retinoblastoma (RB). Herein, the goals of this study were to explore the biological function and the potential mechanism of HOXA11-AS in RB. Materials and methods: The expression of HOXA11-AS in RB tissues and cell lines was detected using real-time PCR (qRT-PCR). Cell proliferation, cycle arrest and apoptosis were measured using a cell counting kit 8 and flow cytometry. The target miRNAs of HOXA11-AS was predicted by Starbase2.0 software and was confirmed using a dual-luciferase reporter assay. Result: We found that HOXA11-AS expression was markedly elevated in RB tissues and cell lines compared to normal retina tissues and human retinal epithelial cells, respectively. Functional analysis showed that knockdown of HOXA11-AS in RB cells significantly suppressed cell proliferation, and induced cell cycle arrest at G1/G0 phase and promoted cell apoptosis. We also found that HOXA11-AS could serve as a competing endogenous RNA (ceRNA) that inhibited miR-506-3p expression, which regulated its downstream target NIMA-related kinase 6 (NEK6) in RB. In addition, miR-506-3p inhibitors partially reversed the effect of HOXA11-AS depletion on proliferation, cycle arrest and apoptosis in RB cells. Conclusion: Taken together, these findings demonstrated that HOXA11-AS could promote RB progression by sponging miR-506-3p, suggesting that HOXA-11-AS might be a potential therapeutic target for RB.
Collapse
Affiliation(s)
- Ning Han
- Department of Ophthalmology, The Second Hospital of Jilin University, Changchun, Jilin 130041, People's Republic of China
| | - Ling Zuo
- Department of Ophthalmology, The Second Hospital of Jilin University, Changchun, Jilin 130041, People's Republic of China
| | - Han Chen
- Department of Ophthalmology, The Second Hospital of Jilin University, Changchun, Jilin 130041, People's Republic of China
| | - Chunxia Zhang
- Department of Ophthalmology, The Second Hospital of Jilin University, Changchun, Jilin 130041, People's Republic of China
| | - Pei He
- Department of Ophthalmology, The Second Hospital of Jilin University, Changchun, Jilin 130041, People's Republic of China
| | - Hongtao Yan
- Department of Ophthalmology, The Second Hospital of Jilin University, Changchun, Jilin 130041, People's Republic of China
| |
Collapse
|
10
|
Yu F, Pang G, Zhao G. RETRACTED: ANRIL acts as onco-lncRNA by regulation of microRNA-24/c-Myc, MEK/ERK and Wnt/β-catenin pathway in retinoblastoma. Int J Biol Macromol 2019; 128:583-592. [PMID: 30703428 DOI: 10.1016/j.ijbiomac.2019.01.157] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2018] [Revised: 01/16/2019] [Accepted: 01/28/2019] [Indexed: 12/19/2022]
Abstract
This article has been retracted: please see Elsevier Policy on Article Withdrawal (http://www.elsevier.com/locate/withdrawalpolicy). This article has been retracted at the request of the Authors. Current research findings in the authors' laboratory are in conflict with previous experimental results published in this paper. It was found that the data reported in figures 4 and 5 were not able to be replicated and so therefore the authors have decided that it is best for the scientific record to retract this paper. All the authors agree with the retraction. The authors apologise for any confusion this paper might have caused readers.
Collapse
Affiliation(s)
- Feifei Yu
- Department of Ophthalmology, The Affiliated Qingdao Hiser Hospital of Qingdao University, Qingdao 266033, Shandong, China
| | - Guolong Pang
- Department of Ophthalmology, Taian City Central Hospital, Taian 271000, Shandong, China
| | - Guiqiu Zhao
- Department of Ophthalmology, The Affiliated Hospital of Qingdao University, Qingdao 266003, Shandong, China.
| |
Collapse
|
11
|
Wang L, Yang D, Tian R, Zhang H. NEAT1 promotes retinoblastoma progression via modulating miR-124. J Cell Biochem 2019; 120:15585-15593. [PMID: 31038819 PMCID: PMC6767513 DOI: 10.1002/jcb.28825] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2018] [Revised: 01/19/2019] [Accepted: 01/24/2019] [Indexed: 12/20/2022]
Abstract
The long noncoding RNA nuclear-enriched abundant transcript 1 (NEAT1) is reportedly involved in the initiation and progression of cancers of several types. However, the role, expression status, and the detailed mechanism of NEAT1 in retinoblastoma (RB) yet need to be unraveled. We explored the role and the mechanism of NEAT1 activity in RB. Our data show enhanced NEAT1 expression in RB-affected tissues compared with the corresponding control. Functional experiments reveal that a NEAT1 knockdown in RB cells significantly inhibits proliferation, cycle progression, and facilitates apoptosis and caspase-3 and -9 activities. Besides that, miR-124 was predicted to be a target of NEAT1 and its reduced expression, as well as the inverse correlation of NEAT1 with miR-124, was observed in RB-affected tissues. Further, luciferase and RNA immunoprecipitation (RIP) assays confirmed the interaction between NEAT1 and miR-124. Rescue experiments confirmed that the inhibition of miR-124 could reverse the effect of NEAT1 on RB cell proliferation, cycle arrest, apoptosis, and caspase-3 and -9 activities. Thus, NEAT1 promotes RB progression by sponging miR-124, providing a therapeutic target for RB.
Collapse
Affiliation(s)
- Lufei Wang
- Department of Ophthalmology, The Second Hospital of Jilin University, Changchun, Jilin, People's Republic of China
| | - Defeng Yang
- Department of Ophthalmology, The Second Hospital of Jilin University, Changchun, Jilin, People's Republic of China
| | - Rui Tian
- Department of Ophthalmology, The Second Hospital of Jilin University, Changchun, Jilin, People's Republic of China
| | - Hui Zhang
- Department of Ophthalmology, The Second Hospital of Jilin University, Changchun, Jilin, People's Republic of China
| |
Collapse
|
12
|
Wu XZ, Cui HP, Lv HJ, Feng L. Knockdown of lncRNA PVT1 inhibits retinoblastoma progression by sponging miR-488-3p. Biomed Pharmacother 2019; 112:108627. [PMID: 30797143 DOI: 10.1016/j.biopha.2019.108627] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2018] [Revised: 01/12/2019] [Accepted: 01/24/2019] [Indexed: 12/31/2022] Open
Abstract
Emerging evidence suggests that long non-coding RNAs (lncRNAs) play a regulatory role in the pathogenesis and progression of retinoblastoma (RB). lncRNA plasmacytoma variant translocation 1 (PVT1) is highly expressed in a plenty of tumors, and is believed to serve as an oncogene. However, the expression, roles, and action mechanisms of PVT1 in the carcinogenesis and progression of RB are still largely unknown. In this study, we found that PVT1 was upregulated in RB tissues and cell lines. PVT1 levels correlated with optic nerve invasion, and intraocular international retinoblastoma classify (IIRC) stage. In addition, the results demonstrated that patients with RB who showed higher expression of PVT1 had worse overall survivals. In WERI-Rb1 and Y79 cells, PVT1 silencing significantly inhibited cell proliferation, migration, invasion, and cell cycle progression and induced cell apoptosis in vitro. Moreover, in vivo xenograft assay indicated that PVT1 knockdown suppressed the tumor volume and tumor weight. The analysis of the mechanisms of action revealed that the reduction of PVT1 inhibited the expression of notch2 by upregulating miR-488-3p. In general, our results demonstrated that PVT1 may be a novel biomarker for prognosis and a new target for the treatment of RB.
Collapse
Affiliation(s)
- Xue-Zhi Wu
- Department of Ophthalmology, Zhumadian Central Hospital, Henan Province, 463000, China.
| | - Hong-Pei Cui
- Department of Ophthalmology, Henan Eye Hospital & Henan Eye Institute, Henan Provincial People's Hospital, Henan Province, 450003, China
| | - Hai-Jiang Lv
- Henan University of Traditional Chinese Medicine, Zhengzhou, Henan Province, 450046, China
| | - Lei Feng
- Department of Ophthalmology, The Third Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, Henan Province, 450008, China
| |
Collapse
|