1
|
Xu W, Li H, Wang Z, Kang Y, Zheng L, Liu Y, Xu P, Li Z. LINC00152: Potential driver oncogene in pan-cancer. WILEY INTERDISCIPLINARY REVIEWS. RNA 2024; 15:e1851. [PMID: 38702938 DOI: 10.1002/wrna.1851] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Revised: 03/26/2024] [Accepted: 04/02/2024] [Indexed: 05/06/2024]
Abstract
Long noncoding RNAs (lncRNA) are a class of non-coding RNAs greater than 200 bp in length with limited peptide-coding function. The transcription of LINC00152 is derived from chromosome 2p11.2. Many studies prove that LINC00152 influences the progression of various tumors via promoting the tumor cells malignant phenotype, chemoresistance, and immune escape. LINC00152 is regulated by multiple transcription factors and DNA hypomethylation. In addition, LINC00152 participates in the regulation of complex molecular signaling networks through epigenetic regulation, protein interactions, and competitive endogenous RNA (ceRNA). Here, we provide a systematic review of the upstream regulatory factors of LINC00152 expression level in different types of tumors. In addition, we revisit the main functions and mechanisms of LINC00152 as driver oncogene and biomarker in pan-cancer. This article is categorized under: RNA in Disease and Development > RNA in Disease RNA Methods > RNA Analyses in Cells RNA Interactions with Proteins and Other Molecules > RNA-Protein Complexes.
Collapse
Affiliation(s)
- Wei Xu
- NHC Key Laboratory of Carcinogenesis, National Clinical Research Center for Geriatric Disorders, Key Laboratory of Carcinogenesis, Chinese Ministry of Health, Department of oncology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Cancer Research Institute, School of Basic Medical Science, Central South University, Changsha, Hunan, China
| | - Huiting Li
- NHC Key Laboratory of Carcinogenesis, National Clinical Research Center for Geriatric Disorders, Key Laboratory of Carcinogenesis, Chinese Ministry of Health, Department of oncology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Cancer Research Institute, School of Basic Medical Science, Central South University, Changsha, Hunan, China
| | - Ziyao Wang
- Department of Thoracic Surgery, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Yan Kang
- NHC Key Laboratory of Carcinogenesis, National Clinical Research Center for Geriatric Disorders, Key Laboratory of Carcinogenesis, Chinese Ministry of Health, Department of oncology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Cancer Research Institute, School of Basic Medical Science, Central South University, Changsha, Hunan, China
| | - Luojie Zheng
- NHC Key Laboratory of Carcinogenesis, National Clinical Research Center for Geriatric Disorders, Key Laboratory of Carcinogenesis, Chinese Ministry of Health, Department of oncology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Cancer Research Institute, School of Basic Medical Science, Central South University, Changsha, Hunan, China
| | - Yiping Liu
- NHC Key Laboratory of Carcinogenesis, National Clinical Research Center for Geriatric Disorders, Key Laboratory of Carcinogenesis, Chinese Ministry of Health, Department of oncology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Ping Xu
- Department of Respiratory and Critical Care Medicine, Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
| | - Zheng Li
- NHC Key Laboratory of Carcinogenesis, National Clinical Research Center for Geriatric Disorders, Key Laboratory of Carcinogenesis, Chinese Ministry of Health, Department of oncology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Cancer Research Institute, School of Basic Medical Science, Central South University, Changsha, Hunan, China
| |
Collapse
|
2
|
A review on the role of LINC00152 in different disorders. Pathol Res Pract 2023; 241:154274. [PMID: 36563561 DOI: 10.1016/j.prp.2022.154274] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Revised: 12/06/2022] [Accepted: 12/08/2022] [Indexed: 12/15/2022]
Abstract
LINC00152 is an important lncRNA in human disorders. It is mainly regarded as a tumor-promoting lncRNA. Mechanistically, LINC00152 serves as a molecular sponge for miR-143a-3p, miR-125a-5p, miR-139, miR-215, miR-193a/b-3p, miR-16-5p, miR-206, miR-195, miR-138, miR-185-5p, miR-103, miR-612, miR-150, miR-107, miR-205-5p and miR-153-3p. In addition, it can regulate activity of mTOR, EGFR/PI3K/AKT, ERK/MAPK, Wnt/β-Catenin, EGFR, NF-κB, HIF-1 and PTEN. In this review, we provide a concise but comprehensive explanation about the role of LINC00152 in tumor development and progression as well as its role in the pathology of non-malignant conditions with the aim of facilitating the clinical implementation of this lncRNA as a diagnostic or prognostic tumor marker and therapeutic target.
Collapse
|
3
|
Zhu H, Xu X, Zheng E, Ni J, Jiang X, Yang M, Zhao G. LncRNA RP11‑805J14.5 functions as a ceRNA to regulate CCND2 by sponging miR‑34b‑3p and miR‑139‑5p in lung adenocarcinoma. Oncol Rep 2022; 48:161. [PMID: 35866595 PMCID: PMC9350987 DOI: 10.3892/or.2022.8376] [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: 11/20/2020] [Accepted: 07/08/2021] [Indexed: 11/05/2022] Open
Abstract
Lung adenocarcinoma (LUAD) is the most common lung cancer with high incidence. The prognosis of LUAD is poor due to its aggressive behavior. Long non‑coding RNAs (lncRNAs) have been reported as a key modulator on LUAD progression. Therefore, the present study aimed to clarify the molecular mechanism of lncRNAs in LUAD development. The expression of lncRNA RP11‑805J14.5 (RP11‑805J14.5) in LUAD tissues and cells was quantified based on the data in The Cancer Genome Atlas (TCGA). Cell viability was determined using Cell Counting Kit‑8 method. Apoptotic cells were sorted and determined by flow cytometry. Cell migration and invasion abilities were detected by the Transwell assay. Luciferase reporter experiment and RNA pull‑down assay were utilized to determine the interactions between RP11‑805J14.5, microRNA (miR)‑34b‑3p, miR‑139‑5p, and cyclin D2 (CCND2). A xenograft tumor was established to determine tumor growth in vivo. RP11‑805J14.5 was highly expressed in LUAD and associated with poor survival of LUAD patients. Knockdown of RP11‑805J14.5 suppressed LUAD cell growth, invasion, migration and tumor growth, indicating that RP11‑805J14.5 is an important regulator of LUAD. Our study demonstrated that the regulation of RP11‑805J14.5 on LUAD was mediated by CCND2 whose expression was regulated by sponging miR‑34b‑3p and miR‑139‑5p. The expression of RP11‑805J14.5 was increased in LUAD, and the knockdown of RP11‑805J14.5 expression suppressed LUAD cell growth, invasion and migration by downregulating CCND2 by sponging miR‑34b‑3p and miR‑139‑5p, indicating that RP11‑805J14.5 could be a prospective target for LUAD therapy.
Collapse
Affiliation(s)
- Huangkai Zhu
- Medical School of Ningbo University, Ningbo, Zhejiang 315211, P.R. China
| | - Xiang Xu
- Department of Thoracic Surgery, Hwa Mei Hospital, University of Chinese Academy of Sciences, Ningbo, Zhejiang 315010, P.R. China
| | - Enkuo Zheng
- Department of Thoracic Surgery, Hwa Mei Hospital, University of Chinese Academy of Sciences, Ningbo, Zhejiang 315010, P.R. China
| | - Junjun Ni
- Department of Thoracic Surgery, Hwa Mei Hospital, University of Chinese Academy of Sciences, Ningbo, Zhejiang 315010, P.R. China
| | - Xu Jiang
- Department of Thoracic Surgery, Hwa Mei Hospital, University of Chinese Academy of Sciences, Ningbo, Zhejiang 315010, P.R. China
| | - Minglei Yang
- Medical School of Ningbo University, Ningbo, Zhejiang 315211, P.R. China
| | - Guofang Zhao
- Medical School of Ningbo University, Ningbo, Zhejiang 315211, P.R. China
| |
Collapse
|
4
|
Li S, Yao W, Liu R, Gao L, Lu Y, Zhang H, Liang X. Long non-coding RNA LINC00152 in cancer: Roles, mechanisms, and chemotherapy and radiotherapy resistance. Front Oncol 2022; 12:960193. [PMID: 36033524 PMCID: PMC9399773 DOI: 10.3389/fonc.2022.960193] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Accepted: 07/20/2022] [Indexed: 11/13/2022] Open
Abstract
Long non-coding RNA LINC00152 (cytoskeleton regulator, or LINC00152) is an 828-bp lncRNA located on chromosome 2p11.2. LINC00152 was originally discovered during research on hepatocarcinogenesis and has since been regarded as a crucial oncogene that regulates gene expression in many cancer types. LINC00152 is aberrantly expressed in various cancers, including gastric, breast, ovarian, colorectal, hepatocellular, and lung cancer, and glioma. Several studies have indicated that LINC00152 is correlated with cell proliferation, apoptosis, migration, invasion, cell cycle, epithelial-mesenchymal transition (EMT), chemotherapy and radiotherapy resistance, and tumor growth and metastasis. High LINC00152 expression in most tumors is significantly associated with poor patient prognosis. Mechanistic analysis has demonstrated that LINC00152 can serve as a competing endogenous RNA (ceRNA) by sponging miRNA, regulating the abundance of the protein encoded by a particular gene, or modulating gene expression at the epigenetic level. LINC00152 can serve as a diagnostic or prognostic biomarker, as well as a therapeutic target for most cancer types. In the present review, we discuss the roles and mechanisms of LINC00152 in human cancer, focusing on its functions in chemotherapy and radiotherapy resistance.
Collapse
Affiliation(s)
- Shuang Li
- Cancer Center, Department of Affiliated People’ Radiation Oncology, Zhejiang Provincial People’s Hospital, Affiliated People’s Hospital, Hangzhou Medical College, Hangzhou, China
- Graduate Department, Jinzhou Medical University, Jinzhou, China
| | - Weiping Yao
- Cancer Center, Department of Affiliated People’ Radiation Oncology, Zhejiang Provincial People’s Hospital, Affiliated People’s Hospital, Hangzhou Medical College, Hangzhou, China
- Graduate Department, Bengbu Medical College, Bengbu, China
| | - Ruiqi Liu
- Cancer Center, Department of Affiliated People’ Radiation Oncology, Zhejiang Provincial People’s Hospital, Affiliated People’s Hospital, Hangzhou Medical College, Hangzhou, China
- Graduate Department, Bengbu Medical College, Bengbu, China
| | - Liang Gao
- Cancer Center, Department of Medical Oncology, Zhejiang Provincial People’s Hospital, Affiliated People’s Hospital, Hangzhou Medical College, Hangzhou, China
| | - Yanwei Lu
- Cancer Center, Department of Affiliated People’ Radiation Oncology, Zhejiang Provincial People’s Hospital, Affiliated People’s Hospital, Hangzhou Medical College, Hangzhou, China
| | - Haibo Zhang
- Cancer Center, Department of Affiliated People’ Radiation Oncology, Zhejiang Provincial People’s Hospital, Affiliated People’s Hospital, Hangzhou Medical College, Hangzhou, China
- *Correspondence: Xiaodong Liang, ; Haibo Zhang,
| | - Xiaodong Liang
- Cancer Center, Department of Affiliated People’ Radiation Oncology, Zhejiang Provincial People’s Hospital, Affiliated People’s Hospital, Hangzhou Medical College, Hangzhou, China
- Graduate Department, Jinzhou Medical University, Jinzhou, China
- *Correspondence: Xiaodong Liang, ; Haibo Zhang,
| |
Collapse
|
5
|
Potentials of long non-coding RNAs as biomarkers of colorectal cancer. Clin Transl Oncol 2022; 24:1715-1731. [PMID: 35581419 DOI: 10.1007/s12094-022-02834-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Accepted: 04/04/2022] [Indexed: 02/08/2023]
Abstract
Colorectal cancer (CRC) is the third most common malignant tumor worldwide and the fourth major cause of cancer-related death, with high morbidity and increased mortality year by year. Although significant progress has been made in the therapy strategies for CRC, the great difficulty in early diagnosis, feeble susceptibility to radiotherapy and chemotherapy, and high recurrence rates have reduced therapeutic efficacy resulting in poor prognosis. Therefore, it is urgent to understand the pathogenesis of CRC and unravel novel biomarkers to improve the early diagnosis, treatment and prediction of CRC recurrence. Long non-coding RNAs (lncRNAs) are non-coding RNAs with a length of more than 200 nucleotides, which are abnormally expressed in tumor tissues and cell lines, activating or inhibiting specific genes through multiple mechanisms including transcription and translation. A growing number of studies have shown that lncRNAs are important regulators of microRNAs (miRNAs, miRs) expression in CRC and may be promising biomarkers and potential therapeutic targets in the research field of CRC. This review mainly summarizes the potential application value of lncRNAs as novel biomarkers in CRC diagnosis, radiotherapy, chemotherapy and prognosis. Additionally, the significance of lncRNA SNHGs family and lncRNA-miRNA networks in regulating the occurrence and development of CRC is mentioned, aiming to provide some insights for understanding the pathogenesis of CRC and developing new diagnostic and therapeutic strategies.
Collapse
|
6
|
Wang J, Han X, Yuan Y, Gu H, Liao X, Jiang M. The Value of Dysregulated LncRNAs on Clinicopathology and Survival in Non-Small-Cell Lung Cancer: A Systematic Review and Meta-Analysis. Front Genet 2022; 13:821675. [PMID: 35450214 PMCID: PMC9016135 DOI: 10.3389/fgene.2022.821675] [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: 11/24/2021] [Accepted: 02/22/2022] [Indexed: 11/13/2022] Open
Abstract
Background: There is growing evidence that a number of lncRNAs are involved in the pathogenesis of non-small-cell lung cancer (NSCLC). However, studies on lncRNA expression in NSCLC patients are far from conclusive. Therefore, we performed a systematic review of such studies to collect and examine the evidence on the potential role of lncRNAs in the development of NSCLC. Methods: We systematically searched seven literature databases to identify all published studies that evaluated the expression of one or more lncRNAs in human samples with NSCLC (cases) and without NSCLC (controls) from January 1, 1995 to May 24, 2021. Quality assessment of studies was conducted by using the “Quality in Prognosis Studies” (QUIPS) tool, and the heterogeneity across studies was analyzed with the I-squared statistic and chi-square-based Q-tests. Either fixed or random-effect meta-analysis was performed to summarize effect size to investigate the association between lncRNA expression and overall survival (OS), disease-free survival (DFS), progression-free survival (PFS), and clinicopathological features. The R statistical software program was used to conduct standard meta-analysis. Results: We finally obtained 48 studies with 5,211 patients included in this review after screening. Among the 48 lncRNAs, 38 lncRNAs were consistently upregulated, and 10 were deregulated in patients with NSCLC compared with the control groups. The upregulated lncRNAs were positively associated with histological type: study number (n) = 18, odds ratio (OR) = 0.78, 95% CI: 0.65–0.95 and OR = 1.30, 95% CI: 1.08–1.57, p < 0.01; TNM stages: n = 20, OR = 0.41, 95% CI: 0.29–0.57 and OR = 2.44, 95% CI: 1.73–3.44, p < 0.01; lymph node metastasis: n = 29, OR = 0.49, 95% CI: 0.34–0.71 and OR = 2.04, 95% CI: 1.40–2.96, p < 0.01; differentiation grade: n = 6, OR = 0.61, 95% CI: 0.38–0.99 and OR = 1.63, 95% CI: 1.01–2.64, p < 0.01; distant metastasis: n = 9, OR = 0.37, 95% CI: 0.26–0.53 and OR = 2.72, 95% CI: 1.90–3.90, p < 0.01; tumor size: n = 16, OR = 0.52, 95% CI: 0.43–0.64 and OR = 1.92, 95% CI: 1.57–2.34, p < 0.01; and overall survival [n = 38, hazard ratio (HR) = 1.79, 95% CI = 1.59–2.02, p < 0.01]. Especially, five upregulated lncRNAs (linc01234, ZEB1-AS1, linc00152, PVT1, and BANCR) were closely associated with TNM Ⅲa stage (n = 5, OR = 4.07, 95% CI: 2.63–6.28, p < 0.01). However, 10 deregulated lncRNAs were not significantly associated with the pathogenesis and overall survival in NSCLC in the meta-analysis (p ≥ 0.05). Conclusion: This systematic review suggests that the upregulated lncRNAs could serve as biomarkers for predicting promising prognosis of NSCLC. The prognostic value of downregulated lncRNA in NSCLC needs to be further explored. Systematic Review Registration: (http://www.crd.york.ac.uk/PROSPERO).identifier CRD42021240635.
Collapse
Affiliation(s)
- Juan Wang
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| | - Xu Han
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| | - Ye Yuan
- The Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Hao Gu
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| | - Xing Liao
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| | - Miao Jiang
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| |
Collapse
|
7
|
Yuan X, Dong Z, Shen S. LncRNA GACAT3: A Promising Biomarker and Therapeutic Target in Human Cancers. Front Cell Dev Biol 2022; 10:785030. [PMID: 35127682 PMCID: PMC8811307 DOI: 10.3389/fcell.2022.785030] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Accepted: 01/05/2022] [Indexed: 12/24/2022] Open
Abstract
Long non-coding RNAs (lncRNAs) are a class of functional RNA molecules that do not encode proteins and are composed of more than 200 nucleotides. LncRNAs play important roles in epigenetic and gene expression regulation. The oncogenic lncRNA GACAT3 was recently discovered to be dysregulated in many tumors. Aberrant expression of GACAT3 contributes to clinical characteristics and regulates multiple oncogenic processes. The association of GACAT3 with a variety of tumors makes it a promising biomarker for diagnosis, prognosis, and targeted therapy. In this review, we integrate the current understanding of the pathological features, biological functions, and molecular mechanisms of GACAT3 in cancer. Additionally, we provide insight into the utility of GACAT3 as an effective diagnostic and prognostic marker for specific tumors, which offers novel opportunities for targeted therapeutic intervention.
Collapse
Affiliation(s)
- Xin Yuan
- Department of Infectious Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Zihui Dong
- Department of Infectious Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Precision Medicine Center, Gene Hospital of Henan Province, The First Affifiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Shen Shen
- Department of Infectious Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Precision Medicine Center, Gene Hospital of Henan Province, The First Affifiliated Hospital of Zhengzhou University, Zhengzhou, China
- *Correspondence: Shen Shen,
| |
Collapse
|
8
|
Ye L, Jin W. Identification of lncRNA-associated competing endogenous RNA networks for occurrence and prognosis of gastric carcinoma. J Clin Lab Anal 2021; 35:e24028. [PMID: 34704289 PMCID: PMC8649378 DOI: 10.1002/jcla.24028] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2021] [Revised: 09/13/2021] [Accepted: 09/14/2021] [Indexed: 12/24/2022] Open
Abstract
Background Gastric cancer (GC) is one of the common digestive malignancies worldwide and causes a severe public health issue. So far, the underlying mechanisms of GC are largely unclear. Thus, we aim to identify the long non‐coding RNA (lncRNA)‐associated competing endogenous RNA (ceRNA) for GC. Methods TCGA database was downloaded and used for the identification of differentially expressed (DE) lncRNAs, miRNAs, and mRNAs, respectively. Then, the ceRNA network was constructed via multiple online datasets and approaches. In addition, various in vitro assays were carried out to validate the effect of certain hub lncRNAs. Results We constructed a ceRNA network, including 76 lncRNAs, 18 miRNAs, and 159 mRNAs, which involved multiple critical pathways. Next, univariate and multivariate analysis demonstrated 11 lncRNAs, including LINC02731, MIR99AHG, INHBA‐AS1, CCDC144NL‐AS1, VLDLR‐AS1, LIFR‐AS1, A2M‐AS1, LINC01537, and LINC00702, and were associated with OS, and nine of those lncRNAs were considered as hub lncRNAs involved in the sub‐ceRNA network. The in vitro assay indicated two lncRNAs, INHBA‐AS1 and CCDC144NL‐AS1, which were positively related to the GC aggressive features, including proliferation, invasion, and migration. Conclusions We identified nine hub lncRNAs and the associated ceRNA network related to the prognosis of GC, and then validated two out of them as promising oncogenes in GC.
Collapse
Affiliation(s)
- Lianmin Ye
- Department of Intensive Care, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Wumin Jin
- Department of Reproductive Medicine Centre, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
9
|
Guo L, Ding L, Tang J. Identification of a competing endogenous RNA axis "SVIL-AS1/miR-103a/ICE1" associated with chemoresistance in lung adenocarcinoma by comprehensive bioinformatics analysis. Cancer Med 2021; 10:6022-6034. [PMID: 34264003 PMCID: PMC8419767 DOI: 10.1002/cam4.4132] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Revised: 06/18/2021] [Accepted: 06/19/2021] [Indexed: 12/19/2022] Open
Abstract
Background Chemotherapy is an important treatment for lung cancer. The molecular mechanism of lung adenocarcinoma (LUAD) chemoresistance is not completely understood. Methods Weighted gene co‐expression network analysis (WGCNA) was applied to screen the modules related to chemosensitivity using the data of LUAD patients receiving chemotherapy in The Cancer Genome Atlas database. GDCRNATools package was used to establish competing endogenous RNA (ceRNA) network based on the key chemotherapy‐related module. Kaplan–Meier and risk models were used to analyze the influence of genes in the ceRNA network on the prognosis of LUAD patients receiving chemotherapy. Cell counting kit‐8, reverse transcription‐quantitative PCR, and dual‐luciferase reporter assay were used to detect the effects of abnormal expression of genes in the ceRNA network on the proliferation and IC50 of cisplatin (DDP)‐resistant LUAD cells, and the targeting relationship of genes in the ceRNA network. The signaling pathways and functions of ICE1 in LUAD were analyzed by LinkOmics and CancerSEA databases, and validated by Western blot. Results Midnightblue module was the only WGCNA module positively correlated with chemosensitivity, in which the function of genes was related to cancer progression. SVIL‐AS1/miR‐103a/ICE1 was constructed based on midnightblue module. High expression of SVIl‐AS1 and ICE1 corresponded to a favorable prognosis. High expression of miR‐103a corresponded to a dismal prognosis. SVIl‐AS1 was downregulated in DDP‐resistant LUAD cells. SVIL‐AS1 overexpression retarded the proliferation and DDP resistance of DDP‐resistant LUAD cell. miR‐103a was sponged by SVIL‐AS1 and directly targeted ICE1. miR‐103a overexpression and ICE1 knockdown overturned the suppressive effect of SVIL‐AS1 overexpression on cell proliferation and DDP resistance. Further bioinformatics analysis and experimental verification showed that SVIL‐AS1/miR‐103a‐3p/ICE1 axis can enhance DNA damage caused by chemotherapeutic agents. Conclusions SVIL‐AS1 inhibited chemoresistance by acting as a sponge for miR‐103a and upregulating ICE1 expression, which may be a potential therapeutic target for chemotherapy in LUAD.
Collapse
Affiliation(s)
- Lili Guo
- Department of Medical Oncology, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing Chest Hospital, Capital Medical University, Beijing, China
| | - Lina Ding
- Key Laboratory of Henan Province for Drug Quality and Evaluation, School of Pharmaceutical Sciences, Ministry of Education of China, Zhengzhou University, Zhengzhou, P.R. China
| | - Junfang Tang
- Department of Medical Oncology, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing Chest Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
10
|
Chen H, Zheng J, Yan L, Zhou X, Jiang P, Yan F. Super-enhancer-associated long noncoding RNA RP11-569A11.1 inhibited cell progression and metastasis by regulating IFIT2 in colorectal cancer. J Clin Lab Anal 2021; 35:e23780. [PMID: 33942366 PMCID: PMC8183909 DOI: 10.1002/jcla.23780] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Revised: 03/24/2021] [Accepted: 03/25/2021] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Recent studies have revealed that super-enhancer-associated long noncoding RNAs (SE-LncRNAs) act pivotal roles in carcinogenesis. This study aimed to report the identification of a novel SE-LncRNA, RP11-569A11.1, and its functional role in colorectal cancer (CRC) progression. METHODS Arraystar human SE-LncRNA microarray was performed to detect differentially expressed SE-LncRNAs in CRC tissues. RT-qPCR was conducted to detect the expression level of RP11-569A11.1 in CRC tissues and cells. The ROC curve was used to analyze the sensitivity and specificity of RP11-569A11.1 in CRC diagnosis. CCK-8 assay, colony formation assay, flow cytometry assay, and transwell assay were used to study the function of RP11-569A11.1. RNA-seq array was performed to analyze the potential downstream target gene of RP11-569A11.1. Western blot assay was conducted to measure the protein level of interferon-induced protein with tetratricopeptide repeat 2 (IFIT2). RESULTS A total of 23 (15 up- and 8 downregulated) significantly expressed SE-LncRNAs were identified in CRC tissues. The top 8 upregulated SE-LncRNAs were RP11-893F2.9, PTCSC1, RP11-803D5.4, AC005592.2, LINC00152, LINC01232, AC017002.1, and RP4-673M15.1, and the top 8 downregulated SE-LncRNAs were RP11-569A11.1, RP11-245G13.2, RP11-556N21.1, U91328.19, AX748340, CTD-2337J16.1, CATG00000108830.1, and RP11-670E13.2. Of which, RP11-569A11.1 was found to be significantly downregulated in CRC tissues and cells. ROC curve analysis showed the area under the curve (AUC) of 0.77 [95% confidence interval (CI), 0.660-0.884, p < 0.001], and the diagnostic sensitivity and specificity were 74.29% and 71.43%, respectively. Functionally, overexpression of RP11-569A11.1 inhibited CRC cell proliferation, migration and invasion, and induced cell apoptosis, while knockdown of RP11-569A11.1 generated an opposite effect. Mechanistically, RP11-569A11.1 positively regulated IFIT2 expression in CRC cells. CONCLUSION RP11-569A11.1 inhibited CRC tumorigenesis by IFIT2-dependent and could serve as a promising diagnostic biomarker in CRC.
Collapse
Affiliation(s)
- Huanhuan Chen
- Department of Clinical Laboratory, The Affiliated Cancer Hospital of Nanjing Medical University, Jiangsu Cancer Hospital and Jiangsu Institute of Cancer Research, Nanjing, China
| | - Junyu Zheng
- Department of Clinical Laboratory, The Affiliated Cancer Hospital of Nanjing Medical University, Jiangsu Cancer Hospital and Jiangsu Institute of Cancer Research, Nanjing, China
| | - Linping Yan
- Department of Clinical Laboratory, The Affiliated Cancer Hospital of Nanjing Medical University, Jiangsu Cancer Hospital and Jiangsu Institute of Cancer Research, Nanjing, China
| | - Xin Zhou
- Department of Clinical Laboratory, The Affiliated Cancer Hospital of Nanjing Medical University, Jiangsu Cancer Hospital and Jiangsu Institute of Cancer Research, Nanjing, China
| | - Pan Jiang
- Department of Clinical Laboratory, The Affiliated Cancer Hospital of Nanjing Medical University, Jiangsu Cancer Hospital and Jiangsu Institute of Cancer Research, Nanjing, China
| | - Feng Yan
- Department of Clinical Laboratory, The Affiliated Cancer Hospital of Nanjing Medical University, Jiangsu Cancer Hospital and Jiangsu Institute of Cancer Research, Nanjing, China
| |
Collapse
|
11
|
Ye S, Lu Y, Ru Y, Wu X, Zhao M, Chen J, Xu M, Huang Q, Wang Y, Shi S, Bu S, Xi Y. LncRNAs GACAT3 and LINC00152 regulate each other through miR-103 and are associated with clinicopathological characteristics in colorectal cancer. J Clin Lab Anal 2020; 34:e23378. [PMID: 32462718 PMCID: PMC7521261 DOI: 10.1002/jcla.23378] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2019] [Revised: 11/29/2019] [Accepted: 12/04/2019] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Long non-coding RNAs (lncRNAs) perform pivotal regulatory roles in tumor development. Our previous work revealed that the lncRNA gastric cancer-associated transcript 3 (GACAT3) was significantly overexpressed and associated with tumor size and metastasis in gastric cancer. METHODS Total RNAs were extracted from colorectal cancer (CRC) and reverse transcribed, and then quantitative real-time PCR (qRT-PCR) was conducted. Cell counting was performed to assess the effect of GACAT3 on CRC cell line proliferation. Bioinformatics prediction, dual luciferase assay, miRNA mimics, siRNAs, and transfection experiments were applied to determine whether GACAT3 and LINC00152 are reciprocally regulated by miR-103. The relationship between their expression levels and clinicopathological factors of patients was explored. A receiver operating characteristic (ROC) curve was used to assess the potential diagnostic value of GACAT3 and LINC00152. RESULTS GACAT3 was identified to be highly expressed in CRC tissues and associated with cell proliferation. Furthermore, we demonstrated that GACAT3 acted as a competing endogenous RNA of LINC00152 and they were both regulated by miR-103. Moreover, analysis of clinicopathological characteristics revealed that GACAT3 and LINC00152 were positively correlated with the depth of invasion, TNM stage, lymph node metastasis, and CA19-9 level. Importantly, a combination of GACAT3 and LINC00152 showed a superior diagnostic capacity compared with the use of the two molecules alone. CONCLUSION Our work shows that GACAT3 and LINC00152 are both overexpressed in CRC and they act as a ceRNA network. Therefore, our data suggest that GACAT3 and LINC00152 may be a promising potential diagnostic biomarker for CRC.
Collapse
Affiliation(s)
- Shazhou Ye
- Diabetes Research Center, Zhejiang Provincial Key Laboratory of Pathophysiology, Institute of Biochemistry and Molecular Biology, School of Medicine, Ningbo University, Ningbo, China
| | - Yicong Lu
- Diabetes Research Center, Zhejiang Provincial Key Laboratory of Pathophysiology, Institute of Biochemistry and Molecular Biology, School of Medicine, Ningbo University, Ningbo, China
| | - Yuqing Ru
- Diabetes Research Center, Zhejiang Provincial Key Laboratory of Pathophysiology, Institute of Biochemistry and Molecular Biology, School of Medicine, Ningbo University, Ningbo, China
| | - Xiaoyue Wu
- Diabetes Research Center, Zhejiang Provincial Key Laboratory of Pathophysiology, Institute of Biochemistry and Molecular Biology, School of Medicine, Ningbo University, Ningbo, China
| | - Ming Zhao
- Diabetes Research Center, Zhejiang Provincial Key Laboratory of Pathophysiology, Institute of Biochemistry and Molecular Biology, School of Medicine, Ningbo University, Ningbo, China
| | - Jiayi Chen
- Diabetes Research Center, Zhejiang Provincial Key Laboratory of Pathophysiology, Institute of Biochemistry and Molecular Biology, School of Medicine, Ningbo University, Ningbo, China
| | - Mingjun Xu
- Diabetes Research Center, Zhejiang Provincial Key Laboratory of Pathophysiology, Institute of Biochemistry and Molecular Biology, School of Medicine, Ningbo University, Ningbo, China
| | - Qin Huang
- Department of Endocrinology, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Yibo Wang
- Diabetes Research Center, Zhejiang Provincial Key Laboratory of Pathophysiology, Institute of Biochemistry and Molecular Biology, School of Medicine, Ningbo University, Ningbo, China
| | - Shanping Shi
- Diabetes Research Center, Zhejiang Provincial Key Laboratory of Pathophysiology, Institute of Biochemistry and Molecular Biology, School of Medicine, Ningbo University, Ningbo, China
| | - Shizhong Bu
- Diabetes Research Center, Zhejiang Provincial Key Laboratory of Pathophysiology, Institute of Biochemistry and Molecular Biology, School of Medicine, Ningbo University, Ningbo, China
| | - Yang Xi
- Diabetes Research Center, Zhejiang Provincial Key Laboratory of Pathophysiology, Institute of Biochemistry and Molecular Biology, School of Medicine, Ningbo University, Ningbo, China
| |
Collapse
|