1
|
Torner B, Géczi D, Klekner Á, Balogh I, Penyige A, Birkó Z. Construction of a miRNA Panel for Differentiating Lung Adenocarcinoma Brain Metastases and Glioblastoma. Cancers (Basel) 2025; 17:581. [PMID: 40002176 PMCID: PMC11853152 DOI: 10.3390/cancers17040581] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Revised: 01/31/2025] [Accepted: 02/04/2025] [Indexed: 02/27/2025] Open
Abstract
BACKGROUND/OBJECTIVES Brain metastases (BM) are the most common type of intracranial malignant tumor and are associated with high mortality. More than 50% of BM cases originate from lung cancer, and lung adenocarcinoma (LUAD) is most commonly associated with the development of BM (25%). The differential diagnosis of solitary BM and glioblastoma (GBM)-one of the most aggressive and fatal malignant brain tumors-remains a considerable challenge. Given the major role of microRNAs (miRNAs) in regulating gene expression, their clinical potential as biomarkers for tumor diagnosis and prognosis offers significant promise. METHODS Next-generation RNA Sequencing (RNA-seq) was used to assess the miRNA expression profiles of 6 LUAD-BM, 6 GBM, and 6 control (non-tumoral brain tissue samples) human brain tissue samples. miRNAs exhibiting the most significant differential expression in LUAD-BM patients in comparison to both control subjects and GBM patients were selected for validation through RT-qPCR. RESULTS The analysis of RNA-seq data revealed the presence of 229 differentially expressed miRNAs in the comparison between LUAD-BM and control samples and 46 in the comparison between LU-AD-BM and GBM samples. Eight miRNAs were selected for further analysis, four of which were upregulated and four downregulated, based on the significant differences in their expression levels observed between the LUAD-BM samples and the other two groups, as confirmed with the Mann-Whitney U test. Functional enrichment analysis was also conducted based on a miRNA-centered target analysis performed using the miRNet tool. To assess the diagnostic potential of these differentially expressed miRNAs, we performed a receiver operating characteristic (ROC) curve analysis. CONCLUSIONS A panel of eight miRNAs was identified in human brain tissue samples, exhibiting high accuracy in distinguishing LUAD-BM from both GBM and control samples.
Collapse
Affiliation(s)
- Bernadett Torner
- Department of Human Genetics, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary; (B.T.); (D.G.); (I.B.); (A.P.)
| | - Dóra Géczi
- Department of Human Genetics, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary; (B.T.); (D.G.); (I.B.); (A.P.)
| | - Álmos Klekner
- Department of Neurosurgery, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary;
| | - István Balogh
- Department of Human Genetics, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary; (B.T.); (D.G.); (I.B.); (A.P.)
- Division of Clinical Genetics, Department of Laboratory Medicine, University of Debrecen Clinical Center, 4032 Debrecen, Hungary
| | - András Penyige
- Department of Human Genetics, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary; (B.T.); (D.G.); (I.B.); (A.P.)
| | - Zsuzsanna Birkó
- Department of Human Genetics, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary; (B.T.); (D.G.); (I.B.); (A.P.)
| |
Collapse
|
2
|
Liu CC, Chen L, Cai YW, Chen YF, Liu YM, Zhou YJ, Shao ZM, Yu KD. Targeting EMSY-mediated methionine metabolism is a potential therapeutic strategy for triple-negative breast cancer. Cell Rep Med 2024; 5:101396. [PMID: 38290515 PMCID: PMC10897545 DOI: 10.1016/j.xcrm.2024.101396] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Revised: 10/19/2023] [Accepted: 01/05/2024] [Indexed: 02/01/2024]
Abstract
Cancer stem cells (CSCs) are the most intractable subpopulation of triple-negative breast cancer (TNBC) cells, which have been associated with a high risk of relapse and poor prognosis. However, eradication of CSCs continues to be difficult. Here, we integrate the multiomics data of a TNBC cohort (n = 360) to identify vital markers of CSCs. We discover that EMSY, inducing a BRCAness phenotype, is preferentially expressed in breast CSCs, promotes ALDH+ cells enrichment, and is positively correlated with poor relapse-free survival. Mechanistically, EMSY competitively binds to the Jmjc domain, which is critical for KDM5B enzyme activity, to reshape methionine metabolism, and to promote CSC self-renewal and tumorigenesis in an H3K4 methylation-dependent manner. Moreover, EMSY accumulation in TNBC cells sensitizes them to PARP inhibitors against bulk cells and methionine deprivation against CSCs. These findings indicate that clinically relevant eradication of CSCs could be achieved with a strategy that targets CSC-specific vulnerabilities in amino acid metabolism.
Collapse
Affiliation(s)
- Cui-Cui Liu
- Key Laboratory of Breast Cancer in Shanghai, Department of Breast Surgery, Fudan University Shanghai Cancer Center and Cancer Institute, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, P.R. China
| | - Lie Chen
- Key Laboratory of Breast Cancer in Shanghai, Department of Breast Surgery, Fudan University Shanghai Cancer Center and Cancer Institute, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, P.R. China
| | - Yu-Wen Cai
- Key Laboratory of Breast Cancer in Shanghai, Department of Breast Surgery, Fudan University Shanghai Cancer Center and Cancer Institute, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, P.R. China
| | - Yu-Fei Chen
- Key Laboratory of Breast Cancer in Shanghai, Department of Breast Surgery, Fudan University Shanghai Cancer Center and Cancer Institute, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, P.R. China
| | - Yi-Ming Liu
- Key Laboratory of Breast Cancer in Shanghai, Department of Breast Surgery, Fudan University Shanghai Cancer Center and Cancer Institute, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, P.R. China
| | - Yu-Jie Zhou
- Key Laboratory of Breast Cancer in Shanghai, Department of Breast Surgery, Fudan University Shanghai Cancer Center and Cancer Institute, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, P.R. China
| | - Zhi-Ming Shao
- Key Laboratory of Breast Cancer in Shanghai, Department of Breast Surgery, Fudan University Shanghai Cancer Center and Cancer Institute, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, P.R. China
| | - Ke-Da Yu
- Key Laboratory of Breast Cancer in Shanghai, Department of Breast Surgery, Fudan University Shanghai Cancer Center and Cancer Institute, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, P.R. China.
| |
Collapse
|
3
|
Awadia S, Sitto M, Ram S, Ji W, Liu Y, Damani R, Ray D, Lawrence TS, Galban CJ, Cappell SD, Rehemtulla A. The adapter protein FADD provides an alternate pathway for entry into the cell cycle by regulating APC/C-Cdh1 E3 ubiquitin ligase activity. J Biol Chem 2023; 299:104786. [PMID: 37146968 PMCID: PMC10248554 DOI: 10.1016/j.jbc.2023.104786] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Revised: 04/11/2023] [Accepted: 04/25/2023] [Indexed: 05/07/2023] Open
Abstract
The E3 ubiquitin ligase APC/C-Cdh1 maintains the G0/G1 state, and its inactivation is required for cell cycle entry. We reveal a novel role for Fas-associated protein with death domain (FADD) in the cell cycle through its function as an inhibitor of APC/C-Cdh1. Using real-time, single-cell imaging of live cells combined with biochemical analysis, we demonstrate that APC/C-Cdh1 hyperactivity in FADD-deficient cells leads to a G1 arrest despite persistent mitogenic signaling through oncogenic EGFR/KRAS. We further show that FADDWT interacts with Cdh1, while a mutant lacking a consensus KEN-box motif (FADDKEN) fails to interact with Cdh1 and results in a G1 arrest due to its inability to inhibit APC/C-Cdh1. Additionally, enhanced expression of FADDWT but not FADDKEN, in cells arrested in G1 upon CDK4/6 inhibition, leads to APC/C-Cdh1 inactivation and entry into the cell cycle in the absence of retinoblastoma protein phosphorylation. FADD's function in the cell cycle requires its phosphorylation by CK1α at Ser-194 which promotes its nuclear translocation. Overall, FADD provides a CDK4/6-Rb-E2F-independent "bypass" mechanism for cell cycle entry and thus a therapeutic opportunity for CDK4/6 inhibitor resistance.
Collapse
Affiliation(s)
- Sahezeel Awadia
- Department of Radiation Oncology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Merna Sitto
- Department of Radiation Oncology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Sundaresh Ram
- Department of Radiology and Biomedical Engineering, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Wenbin Ji
- Department of Radiation Oncology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Yajing Liu
- Department of Radiation Oncology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Raheema Damani
- Department of Biomedical Engineering, University of Alabama, Birmingham, Alabama, USA
| | - Dipankar Ray
- Department of Radiation Oncology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Theodore S Lawrence
- Department of Radiation Oncology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Craig J Galban
- Department of Radiology and Biomedical Engineering, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Steven D Cappell
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, USA
| | - Alnawaz Rehemtulla
- Department of Radiation Oncology, University of Michigan Medical School, Ann Arbor, Michigan, USA.
| |
Collapse
|
4
|
Wu N, Song H, Ren Y, Tao S, Li S. DGUOK-AS1 promotes cell proliferation in cervical cancer via acting as a ceRNA of miR-653-5p. Cell Biochem Funct 2020; 38:870-879. [PMID: 32283566 DOI: 10.1002/cbf.3506] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2019] [Revised: 01/08/2020] [Accepted: 01/14/2020] [Indexed: 12/16/2022]
Abstract
Cervical cancer (CC) holds the second highest incidence and is the fourth dominating cause of cancer-induced death in women. It has been widely accepted that long noncoding RNAs (lncRNAs) are implicated in pathological and physiological activities of CC. However, the research of lncRNAs is still in the initial stage. The biological function of lncRNA deoxyguanosine kinase antisense RNA 1 (DGUOK-AS1) in human cancers has not been reported yet. We found that DGUOK-AS1 was aberrantly upregulated in cervical squamous cell carcinoma and endocervical adenocarcinoma (CESC) tissues through TCGA database. Real-time quantitative polymerase chain reaction (RT-qPCR) also verified the high expression of DGUOK-AS1 in CC cell lines. Loss-of-function assays indicated that DGUOK-AS1 silence repressed CC cell growth. In addition, dual-luciferase reporter and RNA immunoprecipitation (RIP) experiments validated the binding relation between miR-653-5p and DGUOK-AS1 or EMSY. Results of the rescue assays elucidated that EMSY overexpression or miR-653-5p downregulation reversed the suppressive function of DGUOK-AS1 knockdown on cell growth and DNA repair in CC. To sum up, this research highlighted that DGUOK-AS1 could promote CC cell proliferation via serving as a ceRNA of miR-653-5p to release EMSY, which might inspire us to discover novel strategies for CC treatment. SIGNIFICANCE OF THE STUDY: DGUOK-AS1 knockdown hinders proliferation of CC cells. DGUOK-AS1 sequesters miR-653-5p to elevate EMSY in CC. EMSY is required for DGUOK-AS1 to induce cell proliferation and repress DNA damage in CC.
Collapse
Affiliation(s)
- Nanchang Wu
- Department of Gynecologic Oncology, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Honglin Song
- Department of Gynecologic Oncology, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Yaoyao Ren
- Department of Gynecologic Oncology, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Shan Tao
- Department of Gynecologic Oncology, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Shiting Li
- Department of Gynecologic Oncology, Guangxi Medical University Cancer Hospital, Nanning, China
| |
Collapse
|
5
|
Network Pharmacology Study on the Pharmacological Mechanism of Cinobufotalin Injection against Lung Cancer. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2020; 2020:1246742. [PMID: 32148531 PMCID: PMC7048923 DOI: 10.1155/2020/1246742] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/03/2019] [Revised: 12/17/2019] [Accepted: 01/07/2020] [Indexed: 02/06/2023]
Abstract
Cinobufotalin injection, extracted from the skin of Chinese giant salamander or black sable, has good clinical effect against lung cancer. However, owing to its complex composition, the pharmacological mechanism of cinobufotalin injection has not been fully clarified. This study aimed to explore the mechanism of action of cinobufotalin injection against lung cancer using network pharmacology and bioinformatics. Compounds of cinobufotalin injection were determined by literature retrieval, and potential therapeutic targets of cinobufotalin injection were screened from Swiss Target Prediction and STITCH databases. Lung-cancer-related genes were summarized from GeneCards, OMIM, and DrugBank databases. The pharmacological mechanism of cinobufotalin injection against lung cancer was determined by enrichment analysis of gene ontology and Kyoto Encyclopedia of Genes and Genomes, and protein-protein interaction network was constructed. We identified 23 compounds and 506 potential therapeutic targets of cinobufotalin injection, as well as 70 genes as potential therapeutic targets of cinobufotalin injection in lung cancer by molecular docking. The antilung cancer effect of cinobufotalin injection was shown to involve cell cycle, cell proliferation, antiangiogenesis effect, and immune inflammation pathways, such as PI3K-Akt, VEGF, and the Toll-like receptor signaling pathway. In network analysis, the hub targets of cinobufotalin injection against lung cancer were identified as VEGFA, EGFR, CCND1, CASP3, and AKT1. A network diagram of “drug-compounds-target-pathway” was constructed through network pharmacology to elucidate the pharmacological mechanism of the antilung cancer effect of cinobufotalin injection, which is conducive to guiding clinical medication.
Collapse
|
6
|
Orhan C, Bulut P, Dalay N, Ersen E, Buyru N. Downregulation of TCEAL7 expression induces CCND1 expression in non-small cell lung cancer. Mol Biol Rep 2019; 46:5251-5256. [PMID: 31321645 DOI: 10.1007/s11033-019-04982-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2019] [Accepted: 07/15/2019] [Indexed: 02/02/2023]
Abstract
Transcription Elongation Factor A-like 7 (TCEAL7) was first reported as a candidate tumor suppressor gene because of its inactivation in ovarian cancer as a result of promoter methylation. Down-regulation of the TCEAL7 gene expression was also associated with other cancers such as endometrial, breast, brain, prostate, gastric cancers, glioblastoma and linked to tumor phenotypes and clinical outcomes. However, there is no report in the literature investigating the role of TCEAL7 in non-small cell lung cancer. Cyclin D1 is an important molecule in the transition from G1 to S phase of the cell cycle, and is frequently deregulated in cancers. Cylin D1 (CCND1) gene is amplified or overexpressed in a variety of tumors. In our previous study we reported that CCND1 over-expression was not associated with amplification in non-small cell lung cancer. Recently, it has been reported that TCEAL7 regulates CCND1 expression through myc-binding E-box sequences. The aim of this study was to investigate the expression of TCEAL7 gene in non-small cell lung cancer and to determine its effect on the CCND1 expression level. For this purpose, expression levels of TCEAL7 and CCND1 genes were investigated in 50 patients with non-small cell lung cancer by quantitative real time polymerase chain reaction (qRT-PCR). TCEAL7 was under-expressed (68%) in non-small cell lung cancer tumor tissues while CCND1 was over-expressed (42%). The TCEAL7 levels negatively correlated with increased CCND1 expression (p = 0.002).
Collapse
Affiliation(s)
- Ceren Orhan
- Department of Medical Biology, Cerrahpasa Medical Faculty, Istanbul University-Cerrahpasa, Cerrahpasa Street, Kocamustafapasa, Fatih, 34098, Istanbul, Turkey
| | - Pelin Bulut
- Department of Medical Biology, Cerrahpasa Medical Faculty, Istanbul University-Cerrahpasa, Cerrahpasa Street, Kocamustafapasa, Fatih, 34098, Istanbul, Turkey
| | - Nejat Dalay
- Department of Medical Biology, Cerrahpasa Medical Faculty, Istanbul University-Cerrahpasa, Cerrahpasa Street, Kocamustafapasa, Fatih, 34098, Istanbul, Turkey
| | - Ezel Ersen
- Department of Chest Surgery, Cerrahpasa Medical Faculty, Istanbul University-Cerrahpasa, Kocamustafapasa, Fatih, Istanbul, Turkey
| | - Nur Buyru
- Department of Medical Biology, Cerrahpasa Medical Faculty, Istanbul University-Cerrahpasa, Cerrahpasa Street, Kocamustafapasa, Fatih, 34098, Istanbul, Turkey.
| |
Collapse
|
7
|
Cheng X, Wang X, Wu Z, Tan S, Zhu T, Ding K. CORO1C expression is associated with poor survival rates in gastric cancer and promotes metastasis in vitro. FEBS Open Bio 2019; 9:1097-1108. [PMID: 30974047 PMCID: PMC6551501 DOI: 10.1002/2211-5463.12639] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2019] [Revised: 03/13/2019] [Accepted: 04/08/2019] [Indexed: 12/21/2022] Open
Abstract
Coronin-like actin-binding protein 1C (CORO1C) is a member of the WD repeat protein family that regulates actin-dependent processes by assembling F-actin. CORO1C was previously reported to promote metastasis in breast cancer and lung squamous cell carcinoma. Here, we investigated the role of CORO1C in gastric cancer. Higher expression levels of CORO1C were detected in gastric cancer tissues as compared with normal gastric tissues. In addition, CORO1C levels were found to be positively correlated with lymph node metastasis in gastric cancer patients. The expression levels of CORO1C were higher in stage III-IV gastric cancer patients (80.8%) than in stage I-II gastric cancer patients(57.1%). Gastric cancer patients positive for CORO1C expression showed lower relapse-free survival and overall survival rates. Knockdown of CORO1C dramatically suppressed total cell number, cell viability, cell colony formation, cell mitosis and cell metastasis, and promoted apoptosis of gastric cancer cells. Furthermore, cyclin D1 and vimentin were found to be positively regulated by CORO1C. As cyclin D1 and vimentin play an oncogenic role in gastric cancer, CORO1C may exert its tumor-promoting activity through these proteins.
Collapse
Affiliation(s)
- Xiao Cheng
- Department of Pathology, Anhui Medical University, Hefei, China
| | - Xiaonan Wang
- Laboratory of Pathogenic Microbiology and Immunology, Anhui Medical University, Hefei, China
| | - Zhengsheng Wu
- Department of Pathology, Anhui Medical University, Hefei, China
| | - Sheng Tan
- Hefei National Laboratory for Physical Sciences at Microscale, University of Science and Technology of China, Hefei, China
| | - Tao Zhu
- Hefei National Laboratory for Physical Sciences at Microscale, University of Science and Technology of China, Hefei, China
| | - Keshuo Ding
- Department of Pathology, Anhui Medical University, Hefei, China
| |
Collapse
|
8
|
Xiang Q, Tang J, Luo Q, Xue J, Tao Y, Jiang H, Tian J, Fan C. In vitro study of anti-ER positive breast cancer effect and mechanism of 1,2,3,4-6-pentyl-O-galloyl-beta-d-glucose (PGG). Biomed Pharmacother 2019; 111:813-820. [DOI: 10.1016/j.biopha.2018.12.062] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2018] [Revised: 11/30/2018] [Accepted: 12/14/2018] [Indexed: 01/16/2023] Open
|
9
|
Identification of beta-arrestin-1 as a diagnostic biomarker in lung cancer. Br J Cancer 2018; 119:580-590. [PMID: 30078843 PMCID: PMC6162208 DOI: 10.1038/s41416-018-0200-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2018] [Revised: 07/04/2018] [Accepted: 07/06/2018] [Indexed: 01/12/2023] Open
Abstract
Background Distinguishing lung adenocarcinoma (ADC) from squamous cell carcinoma (SCC) has a tremendous therapeutic implication. Sometimes, the commonly used immunohistochemistry (IHC) markers fail to discriminate between them, urging for the identification of new diagnostic biomarkers. Methods We performed IHC on tissue microarrays from two cohorts of lung cancer patients to analyse the expression of beta-arrestin-1, beta-arrestin-2 and clinically used diagnostic markers in ADC and SCC samples. Logistic regression models were applied for tumour subtype prediction. Parallel reaction monitoring (PRM)-based mass spectrometry was used to quantify beta-arrestin-1 in plasma from cancer patients and healthy donors. Results Beta-arrestin-1 expression was significantly higher in ADC versus SCC samples. Beta-arrestin-1 displayed high sensitivity, specificity and negative predictive value. Its usefulness in an IHC panel was also shown. Plasma beta-arrestin-1 levels were considerably higher in lung cancer patients than in healthy donors and were higher in patients who later experienced a progressive disease than in patients showing complete/partial response following EGFR inhibitor therapy. Conclusions Our data identify beta-arrestin-1 as a diagnostic marker to differentiate ADC from SCC and indicate its potential as a plasma biomarker for non-invasive diagnosis of lung cancer. Its utility to predict response to EGFR inhibitors is yet to be confirmed.
Collapse
|
10
|
Liu Y, Wang LL, Chen S, Zong ZH, Guan X, Zhao Y. LncRNA ABHD11-AS1 promotes the development of endometrial carcinoma by targeting cyclin D1. J Cell Mol Med 2018; 22:3955-3964. [PMID: 29799152 PMCID: PMC6050509 DOI: 10.1111/jcmm.13675] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2017] [Accepted: 04/06/2018] [Indexed: 12/15/2022] Open
Abstract
To investigate the expression, role and mechanism of action of long non-coding RNA (lncRNA) ABHD11-AS1 in endometrial carcinoma. The expression of lncRNA ABHD11-AS1 was quantified by qRT-PCR in human endometrial carcinoma (n = 89) and normal endometrial tissues (n = 27). LncRNA ABHD11-AS1 was stably overexpressed or knocked-down in endometrial carcinoma cell lines to examine the cellular phenotype and expression of related molecules. Compared to normal endometrial tissue, lncRNA ABHD11-AS1 was significantly overexpressed in endometrial carcinoma. Overexpression of lncRNA ABHD11-AS1 promoted the proliferation, G1-S progression, invasion and migration of endometrial cancer cells; inhibited apoptosis; up-regulated cyclin D1, CDK1, CDK2, CDK4, Bcl-xl and VEGFA; and down-regulated p16, while ABHD11-AS1 down-regulation has the opposite effect. RNA pull down demonstrated that lncRNA ABHD11-AS1 binds directly to cyclin D1. Knockdown of cyclin D1 can reverse the effect of ABHD11-AS1. Overexpression of lncRNA ABHD11-AS1 increased the tumorigenicity and up-regulated cyclin D1 in an in vivo model of endometrial cancer in nude mice. LncRNA ABHD11-AS1 functions as an oncogene to promote cell proliferation and invasion in endometrial carcinoma by positively targeting cyclin D1.
Collapse
Affiliation(s)
- Yao Liu
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Guangzhou Medical University, Key laboratory for Major Obstetric Diseases of Guangdong Province, and Key Laboratory of Reproduction and Genetics of Guangdong Higher Education Institute in Guangdong Province, Guangzhou, 510150, China
| | - Li-Li Wang
- Department of Gynecology, The First Affiliated Hospital of China Medical University, Shenyang, 110001, China
| | - Shuo Chen
- Department of Gynecology, The First Affiliated Hospital of China Medical University, Shenyang, 110001, China
| | - Zhi-Hong Zong
- Department of Biochemistry and Molecular Biology, College of Basic Medicine, China Medical University, Shenyang, 100013, China
| | - Xue Guan
- Department of Gynecology, The First Affiliated Hospital of China Medical University, Shenyang, 110001, China
| | - Yang Zhao
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Guangzhou Medical University, Key laboratory for Major Obstetric Diseases of Guangdong Province, and Key Laboratory of Reproduction and Genetics of Guangdong Higher Education Institute in Guangdong Province, Guangzhou, 510150, China
| |
Collapse
|
11
|
LncRNA NEAT1 enhances the radio-resistance of cervical cancer via miR-193b-3p/CCND1 axis. Oncotarget 2017; 9:2395-2409. [PMID: 29416780 PMCID: PMC5788648 DOI: 10.18632/oncotarget.23416] [Citation(s) in RCA: 77] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2017] [Accepted: 12/04/2017] [Indexed: 12/18/2022] Open
Abstract
LncRNAs have become a hot topic in various cancer-related researches. Radio-resistance is a great threat for cancer therapy. However, how lncRNAs affect the radio-resistance in cervical cancer is masked. As for our paper, it was discovered that NEAT1 was highly expressed in cervical cancer tissues and non-sensitive tissues as well as radio-resistant cell lines. And the overexpression of NEAT1 accelerated proliferation, while the knockdown of NEAT1 had the opposite result. The effect of NEAT1 on cell proliferation was dependent on the dose of ionizing radiation. And the silence of NEAT1 also caused cell cycle arrest in G0/G1 phase, and triggered more apoptosis, indicating the oncogenic role of NEAT1 in cervical cancer. Next, mechanistic assays affirmed that NEAT1 could function as a ceRNA to regulate cyclin D1 through sponging miR-193b-3p in cervical cancer. Rescue assays were employed to validate that miR-193b-3p and cyclin D1 could inhibit NEAT1-mediated suppressive effect on proliferation, and its stimulative effect on cell cycle arrest and apoptosis. In general, this article disclosed that NEAT1 could facilitate the radio-resistance of cervical cancer via competitively binding miR-193b-3p to up-regulate the expression of cyclin D1.
Collapse
|