51
|
Yu W, Shi Q, Wu C, Shen X, Chen L, Xu J. Promoter hypermethylation influences the suppressive role of long non-coding RNA MEG3 in the development of multiple myeloma. Exp Ther Med 2020; 20:637-645. [PMID: 32537021 DOI: 10.3892/etm.2020.8723] [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] [Received: 01/12/2019] [Accepted: 12/31/2019] [Indexed: 12/12/2022] Open
Abstract
Methylation is a fundamental regulator of gene transcription. Long non-coding RNA maternally expressed 3 (MEG3) inhibits cell proliferation in various types of cancer. However, the molecular mechanisms of MEG3 methylation in the regulation of multiple myeloma (MM) are unknown. In the present study, MEG3 upregulation was negatively associated with the International Staging System (ISS) status of the bone marrow samples of 39 patients with MM. MEG3 overexpression in an MM cell line resulted in elevated p53 expression. Furthermore, the results of methylation-specific PCR revealed that the abnormal methylation status of the MEG3 promoter region was present in eight of the 39 bone marrow samples collected. Treatment of the MM cell line with the DNA methylation inhibitor 5-Aza-2'-deoxycytidine (5-Aza-CdR) resulted in tumor cell proliferation inhibition, apoptosis induction and G0/G1 cell cycle arrest. Furthermore, 5-Aza-CdR decreased aberrant hypermethylation of the MEG3 promoter and increased the expression of MEG3. However, 5-Aza-CdR exerted no effect on p53 expression. To the best of our knowledge, the present study is the first to report that the demethylation reagent 5-Aza-CdR may serve as a therapeutic agent in MM by upregulating MEG3 expression. However, the mechanism of action was independent of p53 expression.
Collapse
Affiliation(s)
- Wenjun Yu
- Department of Geriatric Medicine, Geriatric Hospital of Nanjing Medical University, Jiangsu Province Geriatric Institute, Nanjing, Jiangsu 210000, P.R. China.,Department of Hematology, First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, Nanjing, Jiangsu 210000, P.R. China
| | - Qinglin Shi
- Department of Hematology, First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, Nanjing, Jiangsu 210000, P.R. China
| | - Chao Wu
- Department of Hematology, First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, Nanjing, Jiangsu 210000, P.R. China
| | - Xuxing Shen
- Department of Hematology, First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, Nanjing, Jiangsu 210000, P.R. China
| | - Lijuan Chen
- Department of Hematology, First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, Nanjing, Jiangsu 210000, P.R. China
| | - Jiaren Xu
- Department of Geriatric Medicine, Geriatric Hospital of Nanjing Medical University, Jiangsu Province Geriatric Institute, Nanjing, Jiangsu 210000, P.R. China
| |
Collapse
|
52
|
Tao P, Yang B, Zhang H, Sun L, Wang Y, Zheng W. The overexpression of lncRNA MEG3 inhibits cell viability and invasion and promotes apoptosis in ovarian cancer by sponging miR-205-5p. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2020; 13:869-879. [PMID: 32509057 PMCID: PMC7270692] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Accepted: 07/22/2019] [Indexed: 06/11/2023]
Abstract
PURPOSE Ovarian cancer is a common and fatal cancer in women. The long non-coding RNA (lncRNA) MEG3 was reported to affect the cellular processes of ovarian cancer, but the mechanisms remain unclear. Here, we aimed to explore the potential regulatory mechanism of MEG3 in ovarian cancer. MATERIALS AND METHODS A reverse transcription-quantitative polymerase chain reaction (RT-qPCR) was conducted to analyze the expression levels of MEG3 and miR-205-5p in tissues and cell lines. An MTT assay was utilized to determine the cell viability of ovarian cancer SKOV-3 and OVCAR-8 cells. A flow cytometry analysis was employed to disclose the ovarian cancer cell apoptosis. The migration and invasion of SKOV-3 and OVCAR-8 cells were examined using a Transwell assay. A bioinformatics analysis indicated miR-205-5p as a direct target of MEG3, and a luciferase reporter assay was conducted to validate the interaction between MEG3 and miR-205-5p. RESULTS MEG3 was significantly down-regulated, while miR-205-5p was up-regulated in ovarian cancer tissues and cell lines. The overexpression of MEG3 and the knockdown of miR-205-5p inhibited cell viability, migration and invasion but promoted the apoptosis rate in ovarian cancer cells. MiR-205-5p was identified as a downstream gene of MEG3 and is negatively regulated by MEG3. The introduction of miR-205-5p reversed the up-regulation of MEG3-mediated suppression effects on cell viability, migration and invasion and increased cell apoptosis in ovarian cancer cells. CONCLUSION The overexpression of lncRNA MEG3 inhibits cell proliferation and cell invasion and promotes apoptosis in ovarian cancer by sponging miR-205-5p.
Collapse
Affiliation(s)
- Pingping Tao
- Department of Obstetrics and Gynecology, Pudong New Area People’s Hospital Affiliated to Shanghai Health UniversityNo. 490, Chuanhuan South Road, Pudong New District, Shanghai, China
| | - Binlie Yang
- Department of Obstetrics and Gynecology, Pudong New Area People’s Hospital Affiliated to Shanghai Health UniversityNo. 490, Chuanhuan South Road, Pudong New District, Shanghai, China
| | - Huiya Zhang
- Department of Gynecology, Shaoxing People’s Hospital, Shaoxing Hospital, Zhejiang University, School of MedicineNo. 568 Zhongxing North Road, Yuecheng District, Shaoxing 201299, Zhejiang, China
| | - Liyan Sun
- Department of Obstetrics and Gynecology, Pudong New Area People’s Hospital Affiliated to Shanghai Health UniversityNo. 490, Chuanhuan South Road, Pudong New District, Shanghai, China
| | - Yungen Wang
- Department of Gynecology, Shaoxing People’s Hospital, Shaoxing Hospital, Zhejiang University, School of MedicineNo. 568 Zhongxing North Road, Yuecheng District, Shaoxing 201299, Zhejiang, China
| | - Weiping Zheng
- Department of Gynecology, Shaoxing People’s Hospital, Shaoxing Hospital, Zhejiang University, School of MedicineNo. 568 Zhongxing North Road, Yuecheng District, Shaoxing 201299, Zhejiang, China
| |
Collapse
|
53
|
Bekric D, Neureiter D, Ritter M, Jakab M, Gaisberger M, Pichler M, Kiesslich T, Mayr C. Long Non-Coding RNAs in Biliary Tract Cancer-An Up-to-Date Review. J Clin Med 2020; 9:jcm9041200. [PMID: 32331331 PMCID: PMC7231154 DOI: 10.3390/jcm9041200] [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: 03/03/2020] [Revised: 03/31/2020] [Accepted: 04/07/2020] [Indexed: 02/07/2023] Open
Abstract
The term long non-coding RNA (lncRNA) describes non protein-coding transcripts with a length greater than 200 base pairs. The ongoing discovery, characterization and functional categorization of lncRNAs has led to a better understanding of the involvement of lncRNAs in diverse biological and pathological processes including cancer. Aberrant expression of specific lncRNA species was demonstrated in various cancer types and associated with unfavorable clinical characteristics. Recent studies suggest that lncRNAs are also involved in the development and progression of biliary tract cancer, a rare disease with high mortality and limited therapeutic options. In this review, we summarize current findings regarding the manifold roles of lncRNAs in biliary tract cancer and give an overview of the clinical and molecular consequences of aberrant lncRNA expression as well as of underlying regulatory functions of selected lncRNA species in the context of biliary tract cancer.
Collapse
Affiliation(s)
- Dino Bekric
- Institute of Physiology and Pathophysiology, Paracelsus Medical University, 5020 Salzburg, Austria; (D.B.); (M.R.); (M.J.); (M.G.); (T.K.)
| | - Daniel Neureiter
- Institute of Pathology, Paracelsus Medical University/Salzburger Landeskliniken (SALK), 5020 Salzburg, Austria;
- Cancer Cluster Salzburg, 5020 Salzburg, Austria
| | - Markus Ritter
- Institute of Physiology and Pathophysiology, Paracelsus Medical University, 5020 Salzburg, Austria; (D.B.); (M.R.); (M.J.); (M.G.); (T.K.)
- Ludwig Boltzmann Institute for Arthritis and Rehabilitation, Paracelsus Medical University, 5020 Salzburg, Austria
- Gastein Research Institute, Paracelsus Medical University, 5020 Salzburg, Austria
| | - Martin Jakab
- Institute of Physiology and Pathophysiology, Paracelsus Medical University, 5020 Salzburg, Austria; (D.B.); (M.R.); (M.J.); (M.G.); (T.K.)
| | - Martin Gaisberger
- Institute of Physiology and Pathophysiology, Paracelsus Medical University, 5020 Salzburg, Austria; (D.B.); (M.R.); (M.J.); (M.G.); (T.K.)
- Ludwig Boltzmann Institute for Arthritis and Rehabilitation, Paracelsus Medical University, 5020 Salzburg, Austria
- Gastein Research Institute, Paracelsus Medical University, 5020 Salzburg, Austria
| | - Martin Pichler
- Research Unit of Non-Coding RNAs and Genome Editing, Division of Clinical Oncology, Department of Medicine, Comprehensive Cancer Center Graz, Medical University of Graz, 8036 Graz, Austria;
| | - Tobias Kiesslich
- Institute of Physiology and Pathophysiology, Paracelsus Medical University, 5020 Salzburg, Austria; (D.B.); (M.R.); (M.J.); (M.G.); (T.K.)
- Department of Internal Medicine I, Paracelsus Medical University/Salzburger Landeskliniken (SALK), 5020 Salzburg, Austria
| | - Christian Mayr
- Institute of Physiology and Pathophysiology, Paracelsus Medical University, 5020 Salzburg, Austria; (D.B.); (M.R.); (M.J.); (M.G.); (T.K.)
- Department of Internal Medicine I, Paracelsus Medical University/Salzburger Landeskliniken (SALK), 5020 Salzburg, Austria
- Correspondence:
| |
Collapse
|
54
|
Xu S, Gong Y, Yin Y, Xing H, Zhang N. The multiple function of long noncoding RNAs in osteosarcoma progression, drug resistance and prognosis. Biomed Pharmacother 2020; 127:110141. [PMID: 32334375 DOI: 10.1016/j.biopha.2020.110141] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2020] [Revised: 03/22/2020] [Accepted: 03/30/2020] [Indexed: 12/16/2022] Open
Abstract
Osteosarcoma is a bone tumor prevalent in children and young adults. LncRNAs are a family of non-protein-coding transcripts longer than 200 nucleotides. The tumor-related pathological functions of lncRNAs include proliferation, migration, and chemotherapy resistance, all of which have been widely acknowledged in research on osteosarcoma. In addition, compelling evidence suggests that lncRNAs could serve as diagnostic indicators, prognostic biomarkers, and targets for disease treatment. In this review, we systematically summarize how lncRNAs regulate tumorigenesis, invasion and therapeutic resistance. By deepening our knowledge of the relationship between lncRNAs and osteosarcoma, we hope to translate research findings into clinical applications as soon as possible.
Collapse
Affiliation(s)
- Shengjie Xu
- Department of Cardiology, Sir Run Run Shaw Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Yingchao Gong
- Department of Cardiology, Sir Run Run Shaw Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Ying Yin
- Department of Gastroenterology, Affiliated Zhongda Hospital of Southeast University, Nanjing, China
| | - Hongyuan Xing
- Department of Orthopedics, 2nd Affiliated Hospital, School of Medicine, Zhejiang University, Zhejiang, China
| | - Ning Zhang
- Department of Orthopedics, 2nd Affiliated Hospital, School of Medicine, Zhejiang University, Zhejiang, China.
| |
Collapse
|
55
|
Zuo S, Wu L, Wang Y, Yuan X. Long Non-coding RNA MEG3 Activated by Vitamin D Suppresses Glycolysis in Colorectal Cancer via Promoting c-Myc Degradation. Front Oncol 2020; 10:274. [PMID: 32219064 PMCID: PMC7078156 DOI: 10.3389/fonc.2020.00274] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Accepted: 02/17/2020] [Indexed: 01/04/2023] Open
Abstract
Colorectal cancer (CRC), a common tumor, is characterized by a high mortality rate. Long non-coding RNA maternally expressed gene 3 (MEG3) serves a regulatory role in the carcinogenesis and progression of several types of cancer; however, its role in CRC remains largely unknown. The aim of this study was to explore the regulatory role and mechanism(s) of MEG3 in CRC. The Warburg effect or aerobic glycolysis is characteristic of the metabolism of tumor cells. To determine the effect of MEG3 on glycolysis of CRC cells, we used an XF analyzer to perform glycolysis stress test assays and found that overexpression of MEG3 significantly inhibited glycolysis, glycolytic capacity, as well as lactate production in CRC cells, whereas knockdown of MEG3 produced the opposite effect. Mechanistically, overexpression of MEG3 induced ubiquitin-dependent degradation of c-Myc and inhibited c-Myc target genes involved in the glycolysis pathway such as lactate dehydrogenase A, pyruvate kinase muscle 2, and hexokinase 2. Moreover, we found that MEG3 can be activated by vitamin D and vitamin D receptor (VDR). Clinical data demonstrated that MEG3 was positively associated with serum vitamin D concentrations in patients with CRC. We found that 1,25(OH)2D3 treatment increased MEG3 expression, and knockdown of VDR abolished the effect of MEG3 on glycolysis. These results indicate that vitamin D-activated MEG3 suppresses aerobic glycolysis in CRC cells via degradation of c-Myc. Thus, vitamin D may have therapeutic value in the treatment of CRC.
Collapse
Affiliation(s)
- Siyu Zuo
- Department of Anatomy, Histology, and Embryology, Nanjing Medical University, Nanjing, China
| | - Lei Wu
- Department of Clinical Medicine, First Clinical Medicine College, Nanjing Medical University, Nanjing, China
| | - Yi Wang
- Department of Anatomy, Histology, and Embryology, Nanjing Medical University, Nanjing, China
| | - Xiaoqin Yuan
- Department of Anatomy, Histology, and Embryology, Nanjing Medical University, Nanjing, China.,Key Laboratory for Aging and Disease, Nanjing Medical University, Nanjing, China
| |
Collapse
|
56
|
Long noncoding RNA NEAT1 suppresses hepatocyte proliferation in fulminant hepatic failure through increased recruitment of EZH2 to the LATS2 promoter region and promotion of H3K27me3 methylation. Exp Mol Med 2020; 52:461-472. [PMID: 32157157 PMCID: PMC7156754 DOI: 10.1038/s12276-020-0387-z] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2019] [Revised: 12/17/2019] [Accepted: 12/27/2019] [Indexed: 01/06/2023] Open
Abstract
Fulminant hepatic failure (FHF) refers to the rapid development of severe acute liver injury with impaired synthetic function and encephalopathy in people with normal liver or well-compensated liver disease. This study aimed to investigate the function of long noncoding RNA (lncRNA) nuclear-enriched abundant transcript 1 (NEAT1) on the proliferation and apoptosis of hepatocytes in FHF. Our results revealed that lncRNA NEAT1 was upregulated in cell and animal models of FHF induced by D-galactosamine (D-GalN)/lipopolysaccharide (LPS). Overexpression of lncRNA NEAT1 resulted in elevated hepatocyte apoptosis and impaired large tumor-suppressor kinase 2 (LATS2) expression and proliferation. Functional analysis revealed that knockdown of lncRNA NEAT1 inhibited hepatocyte apoptosis and induced proliferation both in vitro and in vivo. RNA immunoprecipitation and chromatin immunoprecipitation assays demonstrated that lncRNA NEAT1 recruited enhancer of zeste homolog 2 (EZH2) to the LATS2 promoter and repressed LATS2 expression. Furthermore, ectopic expression of LATS2 increased proliferation and inhibited hepatocyte apoptosis by regulating the Hippo/Yes-associated protein (YAP) signaling pathway. Taken together, our findings indicate that lncRNA NEAT1 might serve as a novel target for FHF therapy due to its regulation of H3K27me3 methylation-dependent promotion of LATS2. A long noncoding RNA molecule, one that does not encode the synthesis of protein, is implicated in acute liver failure (AHF) and might offer a new target for drugs to treat the condition. AHF can be induced by various factors, including viruses, drugs, alcohol abuse, and inherited traits. Ke Cheng, Yujun Zhao and colleagues at Central South University in Changsha, China investigated the role of this RNA, called NEAT1, in cell and animal models of AHF. They identified increased production of NEAT1, which suppressed liver cell proliferation and promoted liver cell death. They also uncovered molecular details of the mechanisms underlying these effects, in which the RNA altered the production and regulatory modification of certain proteins. Further research should investigate the therapeutic possibilities of interfering with NEAT1 activity.
Collapse
|
57
|
Xiang Y, Zhang Y, Xia Y, Zhao H, Liu A, Chen Y. LncRNA MEG3 targeting miR-424-5p via MAPK signaling pathway mediates neuronal apoptosis in ischemic stroke. Aging (Albany NY) 2020; 12:3156-3174. [PMID: 32065781 PMCID: PMC7066902 DOI: 10.18632/aging.102790] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2019] [Accepted: 01/12/2020] [Indexed: 02/07/2023]
Abstract
Emerging evidence suggests that long non-coding RNAs (lncRNAs) are significant regulators in the pathological process of ischemic stroke (IS). However, little is known about lncRNAs and their roles in IS. In this study, we aimed to screen out differentially expressed lncRNAs and revealed the underlying mechanisms in IS. The results of bioinformatic analysis showed that lncRNA MEG3 and Sema3A were over-expressed in IS samples, while miR-424-5p was lower-expressed. Correlation between MEG3/miR-424-5p, and miR-424-5p/Sema3A were predicted with miRanda and TargetScan, and verified by dual luciferase assay. Inhibition of MEG3 remarkably increased the expression of miR-424-5p and decreased the expression of Sema3A, which also led to in an increased cell viability and decreased cellular apoptosis in oxygen-glucose deprivation and reoxygenation (OGD/R) model, as well as an activated MAPK signaling pathways. Consistently, MEG3 was upregulated in MCAO mice, knockdown of MEG3 reduced the infarct volume and improved neurobehavioral outcomes in rats following MCAO. In conclusion, it was demonstrated that MEG3 accelerated the process of IS by suppressing miR-424-5p, which targeted Sema3A and the activated MAPK pathway. These results might provide useful information for exploring the potential therapeutic targets in IS.
Collapse
Affiliation(s)
- Yanxiao Xiang
- Department of Pharmacy, Qilu Hospital of Shandong University, Jinan 250012, Shandong, China.,School of Medicine, Shandong University, Jinan 250100, Shandong, China.,Department of Emergency Medicine and Chest Pain Center, Qilu Hospital of Shandong University, Jinan 250012, Shandong, China
| | - Yayun Zhang
- Department of Orthopedics, Qilu Hospital of Shandong University, Jinan 250012, Shandong, China
| | - Yanni Xia
- Department of Operating Room, Qilu Hospital of Shandong University, Jinan 250012, Shandong, China
| | - Hua Zhao
- Department of Orthopedics, Qilu Hospital of Shandong University, Jinan 250012, Shandong, China
| | - Anchang Liu
- Department of Pharmacy, Qilu Hospital of Shandong University, Jinan 250012, Shandong, China
| | - Yuguo Chen
- School of Medicine, Shandong University, Jinan 250100, Shandong, China.,Department of Emergency Medicine and Chest Pain Center, Qilu Hospital of Shandong University, Jinan 250012, Shandong, China.,Clinical Research Center for Emergency and Critical Care Medicine of Shandong Province, Institute of Emergency and Critical Care Medicine of Shandong University, Qilu Hospital of Shandong University, Jinan 250012, Shandong, China.,Key Laboratory of Emergency and Critical Care Medicine of Shandong Province, Key Laboratory of Cardiopulmonary-Cerebral Resuscitation of Shandong Province, Qilu Hospital of Shandong University, Jinan 250012, Shandong, China.,The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese Ministry of Health and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Qilu Hospital of Shandong University, Jinan 250012, Shandong, China
| |
Collapse
|
58
|
circRNA_0000140 suppresses oral squamous cell carcinoma growth and metastasis by targeting miR-31 to inhibit Hippo signaling pathway. Cell Death Dis 2020; 11:112. [PMID: 32041942 PMCID: PMC7010827 DOI: 10.1038/s41419-020-2273-y] [Citation(s) in RCA: 148] [Impact Index Per Article: 29.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2019] [Revised: 01/13/2020] [Accepted: 01/14/2020] [Indexed: 11/08/2022]
Abstract
Oral squamous cell carcinoma (OSCC) is one of the most common malignancies and has a poor prognosis. Circular RNA (circRNA) has been increasingly recognized as a crucial contributor to carcinogenesis. circRNA_0000140 has been aberrantly expressed in OSCC, but its role in tumor growth and metastasis remains largely unclear. Sanger sequencing, actinomycin D, and RNase R treatments were used to confirm head-to-tail junction sequences and the stability of circ_0000140. In vitro cell activities, including proliferation, migration, invasion, and apoptosis, were determined by colony formation, transwell, and flow cytometry assays. The expression levels of circ_0000140, Hippo signaling pathway, and serial epithelial–mesenchymal transition (EMT) markers were measured by quantitative real-time PCR, western blotting, immunofluorescence, and immunohistochemistry. Dual luciferase reporter assays and Argonaute 2-RNA immunoprecipitation assays were performed to explore the interplay among circ_0000140, miR-31, and LATS2. Subcutaneous tumor growth was observed in nude mice, in which in vivo metastasis was observed following tail vein injection of OSCC cells. circ_0000140 is derived from exons 7 to 10 of the KIAA0907 gene. It was down-regulated in OSCC tissues and cell lines, and correlated negatively with poor prognostic outcomes in OSCC patients. Gain-of-function experiments demonstrated that circ_0000140 enhancement suppressed cell proliferation, migration, and invasion, and facilitated cell apoptosis in vitro. In xenograft mouse models, overexpression of circ_0000140 was able to repress tumor growth and lung metastasis. Furthermore, mechanistic studies showed that circ_0000140 could bind with miR-31 and up-regulate its target gene LATS2, thus affecting OSCC cellular EMT. Our findings demonstrated the roles of circ_0000140 in OSCC tumorigenesis as well as in metastasis, and circ_0000140 exerts its tumor-suppressing effect through miR-31/LATS2 axis of Hippo signaling pathway in OSCC.
Collapse
|
59
|
Pellecchia S, Sepe R, Decaussin-Petrucci M, Ivan C, Shimizu M, Coppola C, Testa D, Calin GA, Fusco A, Pallante P. The Long Non-Coding RNA Prader Willi/Angelman Region RNA5 ( PAR5) Is Downregulated in Anaplastic Thyroid Carcinomas Where It Acts as a Tumor Suppressor by Reducing EZH2 Activity. Cancers (Basel) 2020; 12:cancers12010235. [PMID: 31963578 PMCID: PMC7017000 DOI: 10.3390/cancers12010235] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Revised: 01/09/2020] [Accepted: 01/14/2020] [Indexed: 12/13/2022] Open
Abstract
Anaplastic thyroid carcinoma (ATC) represents one the most aggressive neoplasias in humans, and, nowadays, limited advances have been made to extend the survival and reduce the mortality of ATC. Thus, the identification of molecular mechanism underlying its progression is needed. Here, we evaluated the long non-coding RNA (lncRNA) expression profile of nine ATC in comparison with five normal thyroid tissues by a lncRNA microarray. By this analysis, we identified 19 upregulated and 28 downregulated lncRNAs with a fold change >1.1 or <−1.1 and p-value < 0.05, in ATC samples. Some of them were subsequently validated by qRT-PCR. Then, we investigated the role of the lncRNA Prader Willi/Angelman region RNA5 (PAR5), drastically and specifically downregulated in ATC. The restoration of PAR5 reduces proliferation and migration rates of ATC-derived cell lines indicating that its downregulation contributes to thyroid cancer progression. Our results suggest that PAR5 exerts its anti-oncogenic role by impairing Enhancer of Zeste Homolog 2 (EZH2) oncogenic activity since we demonstrated that PAR5 interacts with it in thyroid cancer cell lines, reducing EZH2 protein levels and its binding on the E-cadherin promoter, relieving E-cadherin from the negative regulation by EZH2. Consistently, EZH2 is overexpressed in ATC, but not in differentiated thyroid carcinomas. The results reported here define a tumor suppressor role for PAR5 in undifferentiated thyroid neoplasias, further highlighting the pivotal role of lncRNAs in thyroid carcinogenesis.
Collapse
Affiliation(s)
- Simona Pellecchia
- Institute for Experimental Endocrinology and Oncology (IEOS) “G. Salvatore”, National Research Council (CNR), via S. Pansini, 5-80131 Naples, Italy; (S.P.); (R.S.)
- Department of Molecular Medicine and Medical Biotechnology (DMMBM), University of Naples “Federico II” via S. Pansini, 5-80131 Naples, Italy
| | - Romina Sepe
- Institute for Experimental Endocrinology and Oncology (IEOS) “G. Salvatore”, National Research Council (CNR), via S. Pansini, 5-80131 Naples, Italy; (S.P.); (R.S.)
- Department of Molecular Medicine and Medical Biotechnology (DMMBM), University of Naples “Federico II” via S. Pansini, 5-80131 Naples, Italy
| | - Myriam Decaussin-Petrucci
- Service d’Anatomie et Cytologie Pathologiques, Centre de Biologie Sud, Groupement Hospitalier Lyon Sud, Universite Lyon 1, 69495 Pierre Bénite, France;
| | - Cristina Ivan
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (C.I.); (M.S.); (G.A.C.)
- Center for RNA Interference and Non-Coding RNAs, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Masayoshi Shimizu
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (C.I.); (M.S.); (G.A.C.)
- Center for RNA Interference and Non-Coding RNAs, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Carmela Coppola
- Scientific Directorate, Istituto Nazionale Tumori di Napoli, IRCCS “G. Pascale”, via M. Semmola, 80131 Naples, Italy;
| | - Domenico Testa
- Otorhinolaryngology, Head and Neck Surgery Unit, Department of Mental and Physical Health and Preventive Medicine, University of Campania “Luigi Vanvitelli”, via S. Pansini, 5-80131 Naples, Italy;
| | - George Adrian Calin
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (C.I.); (M.S.); (G.A.C.)
- Center for RNA Interference and Non-Coding RNAs, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Alfredo Fusco
- Institute for Experimental Endocrinology and Oncology (IEOS) “G. Salvatore”, National Research Council (CNR), via S. Pansini, 5-80131 Naples, Italy; (S.P.); (R.S.)
- Department of Molecular Medicine and Medical Biotechnology (DMMBM), University of Naples “Federico II” via S. Pansini, 5-80131 Naples, Italy
- Correspondence: (A.F.); (P.P.)
| | - Pierlorenzo Pallante
- Institute for Experimental Endocrinology and Oncology (IEOS) “G. Salvatore”, National Research Council (CNR), via S. Pansini, 5-80131 Naples, Italy; (S.P.); (R.S.)
- Correspondence: (A.F.); (P.P.)
| |
Collapse
|
60
|
Dong HT, Liu Q, Zhao T, Yao F, Xu Y, Chen B, Wu Y, Zheng X, Jin F, Li J, Xing P. Long Non-coding RNA LOXL1-AS1 Drives Breast Cancer Invasion and Metastasis by Antagonizing miR-708-5p Expression and Activity. MOLECULAR THERAPY. NUCLEIC ACIDS 2019; 19:696-705. [PMID: 31945728 PMCID: PMC6965509 DOI: 10.1016/j.omtn.2019.12.016] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/29/2019] [Revised: 12/07/2019] [Accepted: 12/13/2019] [Indexed: 12/17/2022]
Abstract
LOXL1-AS1, a recently characterized long non-coding RNA (lncRNA), has been reported to modulate tumor progression in several types of cancer. However, the expression and role of LOXL1-AS1 in breast cancer remain unclear. In this study, we sought to identify novel lncRNA regulators engaged in breast cancer metastasis. To this end, we examined 42 cancer-related lncRNAs between MCF7 (with low metastatic potential) and MDA-MB-231 (with high metastatic potential) cells. These lncRNAs have been found to affect the invasiveness of several cancer types, but they are still undefined in breast cancer. Among the 42 candidates, LOXL1-AS1 is significantly increased in MDA-MB-231 cells relative to MCF7 cells. We also show that LOXL1-AS1 is upregulated in breast cancer tissues and cells compared to noncancerous counterparts. Increased LOXL1-AS1 expression is correlated with tumor stage and lymph node metastasis in breast cancer patients. Biologically, overexpression of LOXL1-AS1 enhances and knockdown of LOXL1-AS1 suppresses breast cancer cell migration and invasion. In vivo studies demonstrate that depletion of LOXL1-AS1 inhibits breast cancer metastasis. Mechanistically, LOXL1-AS1 sponges miR-708-5p to increase nuclear factor κB (NF-κB) activity. LOXL1-AS1 can also interact with EZH2 protein to enhance EZH2-mediated transcriptional repression of miR-708-5p. Rescue experiments indicate that co-expression of miR-708-5p attenuates LOXL1-AS1-induced invasiveness in breast cancer. In addition, there is a negative correlation between LOXL1-AS1 and miR-708-5p expression in breast cancer specimens. Overall, LOXL1-AS1 upregulation facilitates breast cancer invasion and metastasis by blocking miR-708-5p expression and activity. LOXL1-AS1 serves as a potential therapeutic target for breast cancer treatment.
Collapse
Affiliation(s)
- Hui-Ting Dong
- Department of Breast Surgery, the First Affiliated Hospital of China Medical University, Shenyang, China
| | - Qun Liu
- Department of Breast Surgery, the First Affiliated Hospital of China Medical University, Shenyang, China
| | - Tingting Zhao
- Department of Breast Surgery, the First Affiliated Hospital of China Medical University, Shenyang, China
| | - Fan Yao
- Department of Breast Surgery, the First Affiliated Hospital of China Medical University, Shenyang, China
| | - Yingying Xu
- Department of Breast Surgery, the First Affiliated Hospital of China Medical University, Shenyang, China
| | - Bo Chen
- Department of Breast Surgery, the First Affiliated Hospital of China Medical University, Shenyang, China
| | - Yunfei Wu
- Department of Breast Surgery, the First Affiliated Hospital of China Medical University, Shenyang, China
| | - Xinyu Zheng
- Department of Breast Surgery, the First Affiliated Hospital of China Medical University, Shenyang, China
| | - Feng Jin
- Department of Breast Surgery, the First Affiliated Hospital of China Medical University, Shenyang, China
| | - Jiguang Li
- Department of Breast Surgery, the First Affiliated Hospital of China Medical University, Shenyang, China
| | - Peng Xing
- Department of Breast Surgery, the First Affiliated Hospital of China Medical University, Shenyang, China.
| |
Collapse
|
61
|
Gong M, Luo C, Meng H, Li S, Nie S, Jiang Y, Wan Y, Li H, Cheng W. Upregulated LINC00565 Accelerates Ovarian Cancer Progression By Targeting GAS6. Onco Targets Ther 2019; 12:10011-10022. [PMID: 31819497 PMCID: PMC6875503 DOI: 10.2147/ott.s227758] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2019] [Accepted: 11/05/2019] [Indexed: 12/25/2022] Open
Abstract
Background Long noncoding RNAs (lncRNAs) have been identified to participate in tumorigenesis. However, the underlying mechanisms of differentially expressed lncRNAs engaged in diseases remain indistinct and need further exploration. Methods Raw data files downloaded from TCGA and GEO dataset were used to analyze the differentially expressed lncRNAs and LINC00565 was picked out as the potential oncogene. qRT-PCR was used to analyze the LINC00565 level in ovarian tissues and cell lines. Subsequently, the selected ovarian tumor cells were then transfected with LINC00565 siRNA by Lipofectamine 2000 and the cell cycle was detected by flow cytometry. Effect of LINC00565 on tumor growth and cell cycle was verified by tumor formation assay in nude mice. The mechanism of LINC00565 involving in cell cycle regulation was further explored by Western blot. Results In this research, we discovered that LINC00565, a novel lncRNA, was highly expressed in ovarian cancer (OC). LINC00565 expression level was negatively associated with outcomes of OC patients. Further analysis showed that LINC00565 expression was closely correlated to tumor size, FIGO stage, but not related to other clinical features. In vitro experiments indicated that knockdown of LINC00565 significantly inhibited proliferative, invasive and migratory abilities of ovarian cancer cells. Besides, knockdown of LINC00565 can induce cell cycle arrest in G0/G1 phase. In addition, in vivo assay showed that low expression of LINC00565 inhibited the growth of OC. Further study found that LINC00565 knockdown markedly downregulated the protein expressions of CyclinD1, CyclinE1 and CDK4, but upregulated the expression of P16 and P21. Subsequently, we confirmed that LINC00565 promoted the progression of OC via upregulating GAS6, which has been confirmed to promote tumor progression. Conclusion In summary, our study firstly reported that the LINC00565 functioned as an oncogene to promote the progression of OC by interacting with GAS6.
Collapse
Affiliation(s)
- Mi Gong
- Department of Gynecology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, People's Republic of China.,Department of Gynecology, The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, Huai'an 223300, People's Republic of China
| | - Chengyan Luo
- Department of Gynecology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, People's Republic of China
| | - Huangyang Meng
- Department of Gynecology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, People's Republic of China
| | - Siyue Li
- Department of Gynecology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, People's Republic of China
| | - Sipei Nie
- Department of Gynecology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, People's Republic of China
| | - Yi Jiang
- Department of Gynecology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, People's Republic of China
| | - Yicong Wan
- Department of Gynecology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, People's Republic of China
| | - Huijian Li
- State Key Laboratory of Reproductive Medicine, Institute of Toxicology, Nanjing Medical University, Nanjing, Jiangsu 211166, People's Republic of China.,Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu 211166, People's Republic of China.,Department of Gynecology, Wuxi Maternal and Child Health Hospital, Wuxi, Jiangsu 214002, People's Republic of China
| | - Wenjun Cheng
- Department of Gynecology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, People's Republic of China
| |
Collapse
|
62
|
Begolli R, Sideris N, Giakountis A. LncRNAs as Chromatin Regulators in Cancer: From Molecular Function to Clinical Potential. Cancers (Basel) 2019; 11:E1524. [PMID: 31658672 PMCID: PMC6826483 DOI: 10.3390/cancers11101524] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2019] [Revised: 09/28/2019] [Accepted: 10/06/2019] [Indexed: 12/15/2022] Open
Abstract
During the last decade, high-throughput sequencing efforts in the fields of transcriptomics and epigenomics have shed light on the noncoding part of the transcriptome and its potential role in human disease. Regulatory noncoding RNAs are broadly divided into short and long noncoding transcripts. The latter, also known as lncRNAs, are defined as transcripts longer than 200 nucleotides with low or no protein-coding potential. LncRNAs form a diverse group of transcripts that regulate vital cellular functions through interactions with proteins, chromatin, and even RNA itself. Notably, an important regulatory aspect of these RNA species is their association with the epigenetic machinery and the recruitment of its regulatory apparatus to specific loci, resulting in DNA methylation and/or post-translational modifications of histones. Such epigenetic modifications play a pivotal role in maintaining the active or inactive transcriptional state of chromatin and are crucial regulators of normal cellular development and tissue-specific gene expression. Evidently, aberrant expression of lncRNAs that interact with epigenetic modifiers can cause severe epigenetic disruption and is thus is closely associated with altered gene function, cellular dysregulation, and malignant transformation. Here, we survey the latest breakthroughs concerning the role of lncRNAs interacting with the epigenetic machinery in various forms of cancer.
Collapse
Affiliation(s)
- Rodiola Begolli
- Department of Biochemistry and Biotechnology, University of Thessaly, Biopolis, 41500 Larissa, Greece.
| | - Nikos Sideris
- Department of Biochemistry and Biotechnology, University of Thessaly, Biopolis, 41500 Larissa, Greece.
| | - Antonis Giakountis
- Department of Biochemistry and Biotechnology, University of Thessaly, Biopolis, 41500 Larissa, Greece.
- B.S.R.C "Alexander Fleming", 34 Fleming str, 16672 Vari, Greece.
| |
Collapse
|
63
|
Long non-coding RNA MEG3 inhibits chondrogenic differentiation of synovium-derived mesenchymal stem cells by epigenetically inhibiting TRIB2 via methyltransferase EZH2. Cell Signal 2019; 63:109379. [PMID: 31376524 DOI: 10.1016/j.cellsig.2019.109379] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2019] [Revised: 07/26/2019] [Accepted: 07/27/2019] [Indexed: 01/05/2023]
Abstract
Osteoarthritis (OA) is a highly prevalent skeletal disease. Mesenchymal stem cell-derived cartilage tissue engineering is a clinical method used for OA treatment. Investigations on the molecular regulatory mechanisms of the chondrogenic differentiation of synovium-derived mesenchymal stem cells(SMSCs) will help promote its clinical applications. In this study, bioinformatics analysis from three different databases indicated that the long non-coding RNA (lncRNA) MEG3 may regulate the chondrogenic differentiation of SMSCs by targeting TRIB2. We then performed assays and found that both knockdown of MEG3 or overexpression of TRIB2 can stimulate the chondrogenic differentiation of SMSCs and increase Col2A1 and aggrecan expression. Knockdown of MEG3 can induce the expression of TRIB2; conversely, overexpression of MEG3 can inhibit the expression of TRIB2. Futhermore, knockdown of the TRIB2 can rescue the MEG3 silencing-mediated promotion of chondrogenic differentiation. Moreover, RNA immunoprecipitation(RIP) and RNA pull-down assays demonstrated that MEG3 can interact with EZH2, thus recruiting it to induce H3K27me3, which promotes the methylation of TRIB2 by binding with the promoter of TRIB2 in SMSCs. Additionally, EZH2 silencing significantly rescued the MEG3 overexpression-mediated inhibition of TRIB2 expression and chondrogenic differentiation of SMSCs. Taken together, these data indicated that MEG3 regulates chondrogenic differentiation by inhibiting TRIB2 expression through EZH2-mediated H3K27me3.
Collapse
|
64
|
Maternally expressed gene 3 (MEG3): A tumor suppressor long non coding RNA. Biomed Pharmacother 2019; 118:109129. [PMID: 31326791 DOI: 10.1016/j.biopha.2019.109129] [Citation(s) in RCA: 115] [Impact Index Per Article: 19.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2019] [Revised: 06/10/2019] [Accepted: 06/12/2019] [Indexed: 12/12/2022] Open
Abstract
Maternally expressed gene 3 (MEG3) is a long non-coding RNA (lncRNA) located on chromosome 14q32.3. Direct sequencing experiments have shown monoallelic expression of this lncRNA. Several studies have shown down-regulation of this lncRNA in human cancers. In some cases, hypermethylation of the promoter region has been suggested as the underlying mechanism. Functional studies have shown that this lncRNA controls expression of several tumor suppressor genes and oncogenes among them are p53, RB, MYC and TGF-β. Through regulation of Wnt-β-catenin pathway, it also affects epithelial-mesenchymal transition. In vitro studies have demonstrated contribution of MEG3 in defining response to chemotherapeutic agents such as paclitaxel, cisplatin and oxaliplatin. Certain polymorphisms within MEG3 are implicated in cancer risk (rs7158663, rs4081134 and rs11160608) or therapeutic response of cancer patients (rs10132552). Taken together, this lncRNA is regarded as a putative cancer biomarker and treatment target. In the current review, several aspects of the participation of MEG3 in carcinogenesis are discussed.
Collapse
|
65
|
Jiang D, Zhang Y, Yang L, Lu W, Mai L, Guo H, Liu X. Retracted
: Long noncoding RNA HCG22 suppresses proliferation and metastasis of bladder cancer cells by regulation of PTBP1. J Cell Physiol 2019; 235:1711-1722. [DOI: 10.1002/jcp.29090] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2019] [Accepted: 06/27/2019] [Indexed: 12/31/2022]
Affiliation(s)
- Dong Jiang
- Department of Urology The Fifth Affiliated Hospital of Sun Yat‐sen University Zhuhai China
| | - Yongyu Zhang
- Department of Interventional Radiology The Fifth Affiliated Hospital of Sun Yat‐sen University Zhuhai China
| | - Lewei Yang
- Department of Radiotherapy for Abdominal Neoplasms The Fifth Affiliated Hospital of Sun Yat‐sen University Zhuhai China
| | - Wuzhu Lu
- Department of Ultrasound The Fifth Affiliated Hospital of Sun Yat‐sen University Zhuhai China
| | - Lei Mai
- Department of Gastroenterology The Fifth Affiliated Hospital of Sun Yat‐sen University Zhuhai China
| | - Huixue Guo
- Department of Gastroenterology The Fifth Affiliated Hospital of Sun Yat‐sen University Zhuhai China
| | - Xialei Liu
- Department of Hepatobiliary Surgery The Fifth Affiliated Hospital of Sun Yat‐sen University Zhuhai China
| |
Collapse
|
66
|
Zhang J, Gao Y. Long non-coding RNA MEG3 inhibits cervical cancer cell growth by promoting degradation of P-STAT3 protein via ubiquitination. Cancer Cell Int 2019; 19:175. [PMID: 31320837 PMCID: PMC6615085 DOI: 10.1186/s12935-019-0893-z] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2018] [Accepted: 06/26/2019] [Indexed: 01/09/2023] Open
Abstract
Background Maternally expressed 3 (MEG3) plays an important role in cervical cancer development, but its exact role remains unclear. Here, we explored the specific regulatory mechanism of MEG3 and its downstream proteins in cervical cancer cells. Methods The effect of MEG3 on tumor formation ability of cervical cancer cells was determined in nude mice. The direct binding of MEG3 to phosphorylated signal transducer and activator of transcription 3 (P-STAT3) was detected by RNA pull-down and RNA-binding protein immunoprecipitation (RIP) assays. Cycloheximide (CHX)-chase and ubiquitination assays were performed to determine the regulatory effect of MEG3 on P-STAT3 ubiquitination. Clone formation assay and flow cytometry were used to evaluate the effect of the MEG3-STAT3 regulatory axis on cell proliferation and apoptosis. Results In vivo tumor formation experiments showed that MEG3 inhibited the tumor formation ability of cervical cancer cells. RNA pull-down and RIP assays demonstrated that MEG3 bound directly to P-STAT3 protein. CHX-chase and ubiquitination assay results showed that MEG3 promoted P-STAT3 degradation via ubiquitination. Clone formation assay and flow cytometry analysis results revealed that the inhibitory effect of MEG3 on P-STAT3 promoted apoptosis and inhibited proliferation of cervical cancer cells. Conclusion MEG3 binds to P-STAT3 in cervical cancer cells, resulting in P-STAT3 ubiquitination and degradation and apoptosis and inhibition of proliferation of tumor cells. The in-depth elaboration of the MEG3-STAT3 regulatory axis in cervical cancer may clarify the mechanism of action of MEG3 and provide new ideas for cervical cancer treatment.
Collapse
Affiliation(s)
- Jun Zhang
- 1Department of Obstetrics and Gynecology, The Second Clinical Medical College (Shenzhen People's Hospital), Jinan University, Shenzhen, 518020 People's Republic of China
| | - Yali Gao
- 2Department of Ophthalmology, The Second Clinical Medical College (Shenzhen People's Hospital), Jinan University, Shenzhen, 518020 People's Republic of China
| |
Collapse
|
67
|
Long noncoding RNAs as potential biomarkers and therapeutic targets in gallbladder cancer: a systematic review and meta-analysis. Cancer Cell Int 2019; 19:169. [PMID: 31297033 PMCID: PMC6599267 DOI: 10.1186/s12935-019-0891-1] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2019] [Accepted: 06/23/2019] [Indexed: 02/06/2023] Open
Abstract
Background Mounting evidence has shown that long noncoding RNAs (lncRNAs) can play a substantial role in gallbladder cancer (GBC) development as tumor promotors or suppressors, and their abnormal expression is relevant to GBC patient outcomes. We completed this systematic review and meta-analysis to explore the clinical significance and mechanisms of lncRNAs in GBC. Methods We conducted a comprehensive literature search and selected eligible records according to the inclusion and exclusion criteria. Hazard ratios (HRs) and odds ratios (ORs) were extracted or calculated to estimate the relationships of high lncRNA expression with GBC patient survival and clinical outcomes. Results Eighteen studies were identified as eligible for this systematic review and meta-analysis. Heterogeneity among HRs of overall survival (OS) was notably high (I2 = 86.2%, p < 0.001). Subgroup analysis suggested that overexpression of lncRNAs in a group that is upregulated in GBC showed a significant association with poor OS (HR = 2.454, 95% CI 2.004–3.004, I2 = 0%). Conversely, overexpression of lncRNAs in a downregulated group was markedly related to good OS (HR = 0.371, 95% CI 0.267–0.517, I2 = 0%). High expression levels of lncRNA AFAP1-AS1, MALAT1 and ROR were positively correlated with tumor size. Expression of lncRNA LET, LINC00152 and HEGBC exhibited a positive correlation with high T status. LncRNA LINC00152, HEGBC, MALAT1 and ROR showed a marked correlation with positive lymph node metastasis (LNM), while lncRNA GCASPC, MEG3, LET and UCA1 had the opposite effect. High expression levels of lncRNA HEGBC, PAGBC, PVT1 and UCA1 predicted high tumor node metastasis (TNM) stages, while lncRNA LET, GCASPC and MEG3 indicated low TNM stages. We also summarized the mechanisms of lncRNAs in GBC. Conclusion Aberrant expression of several lncRNAs was indicative of the prognosis of GBC patients, and lncRNAs showed promise as biomarkers and therapeutic targets for GBC.
Collapse
|
68
|
Ubiquitination and Long Non-coding RNAs Regulate Actin Cytoskeleton Regulators in Cancer Progression. Int J Mol Sci 2019; 20:ijms20122997. [PMID: 31248165 PMCID: PMC6627692 DOI: 10.3390/ijms20122997] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Revised: 06/16/2019] [Accepted: 06/17/2019] [Indexed: 12/15/2022] Open
Abstract
Actin filaments are a major component of the cytoskeleton in eukaryotic cells and play an important role in cancer metastasis. Dynamics and reorganization of actin filaments are regulated by numerous regulators, including Rho GTPases, PAKs (p21-activated kinases), ROCKs (Rho-associated coiled-coil containing kinases), LIMKs (LIM domain kinases), and SSH1 (slingshot family protein phosphate 1). Ubiquitination, as a ubiquitous post-transcriptional modification, deceases protein levels of actin cytoskeleton regulatory factors and thereby modulates the actin cytoskeleton. There is increasing evidence showing cytoskeleton regulation by long noncoding RNAs (lncRNAs) in cancer metastasis. However, which E3 ligases are activated for the ubiquitination of actin-cytoskeleton regulators involved in tumor metastasis remains to be fully elucidated. Moreover, it is not clear how lncRNAs influence the expression of actin cytoskeleton regulators. Here, we summarize physiological and pathological mechanisms of lncRNAs and ubiquitination control mediators of actin cytoskeleton regulators which that are involved in tumorigenesis and tumor progression. Finally, we briefly discuss crosstalk between ubiquitination and lncRNA control mediators of actin-cytoskeleton regulators in cancer.
Collapse
|
69
|
Li J, Jiang X, Li C, Liu Y, Kang P, Zhong X, Cui Y. LncRNA-MEG3 inhibits cell proliferation and invasion by modulating Bmi1/RNF2 in cholangiocarcinoma. J Cell Physiol 2019; 234:22947-22959. [PMID: 31119760 DOI: 10.1002/jcp.28856] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2019] [Revised: 05/05/2019] [Accepted: 05/07/2019] [Indexed: 12/15/2022]
Abstract
Cholangiocarcinoma (CCA) is a mortal cancer with gradually increasing incidences all over the world, whereas effective diagnosis and treatment for this disease are still lacking. As a classical long noncoding RNA (lncRNA), maternally expressed gene 3 (MEG3) has been reported to exhibit pivotal regulatory roles in the occurrence and development of various digestive system tumors. Nevertheless, the clinical relevance and biological function of MEG3 in CCA remain largely unclear. In this study, MEG3 expression was significantly downregulated in both CCA tissues and cells in comparison with that in nontumor controls, respectively, and this downexpression was prominently associated with advanced TNM stage, lymph node invasion, and poor survival. Moreover, decreased MEG3 was an independent forecaster of poor prognosis for CCA patients. Functionally, MEG3 overexpression inhibited CCA growth in vitro and in vivo. Enhanced MEG3 also suppressed migration and invasion of CCLP-1 and QBC939 cells by reversing epithelial-mesenchymal transition (EMT) process. On the contrary, the proliferation, metastasis, and EMT were facilitated via knocking down MEG3. In addition, the expression of B lymphoma Mo-MLV insertion region 1 (Bmi1) and RING finger protein 2 was impacted by gain or loss of MEG3, furthermore, the malignant processes induced by MEG3 knockdown were rescued by means of silencing Bmi1. These data suggested that MEG3 caused tumor suppressive effects partly through mediating polycomb repressive complex 1. Our findings elucidate that MEG3 exerts critical functions in CCA development and likely acts as a promising tumor indicator or intervention target for CCA.
Collapse
Affiliation(s)
- Jinglin Li
- Department of HPB Surgery, The 2nd Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Xingming Jiang
- Department of HPB Surgery, The 2nd Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Chunlong Li
- Department of HPB Surgery, The 2nd Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Yueping Liu
- Department of HPB Surgery, The 2nd Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Pengcheng Kang
- Department of HPB Surgery, The 2nd Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Xiangyu Zhong
- Department of HPB Surgery, The 2nd Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Yunfu Cui
- Department of HPB Surgery, The 2nd Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China
| |
Collapse
|
70
|
Cui Z, Luo Z, Lin Z, Shi L, Hong Y, Yan C. Long non-coding RNA TTN-AS1 facilitates tumorigenesis of papillary thyroid cancer through modulating the miR-153-3p/ZNRF2 axis. J Gene Med 2019; 21:e3083. [PMID: 30811764 DOI: 10.1002/jgm.3083] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Revised: 01/28/2019] [Accepted: 02/09/2019] [Indexed: 12/27/2022] Open
Abstract
BACKGROUND Long non-coding RNAs (lncRNAs) are crucial modulators in the tumorigenesis of numerous cancers, including papillary thyroid cancer (PTC). However, it is unclear whether lncRNA TTN antisense RNA 1 (TTN-AS1) can regulate PTC progression. The present study aimed to reveal the mechanism and function of TTN-AS1 in PTC. METHODS TTN-AS1 expression in 92 pairs PTC tissues and four PTC cells was measured via a quantitative reverse transcriptase-polymerase chain reaction assay. The relationship of TTN-AS1 expression and clinical pathological features of PTC patients was analyzed using a chi-squared test. The biofunction of TTN-AS1 in PTC was identified by loss or gain-of-function assays. Based on bioinformatics analysis and mechanism experiments, the molecular mechanism of TTN-AS1 was analyzed and identified. RESULTS A high level of TTN-AS1 was observed in PTC tissues and cells. The expression level of TTN-AS1 is possibly associated with lymphatic metastasis, TNM stage and the overall survival of PTC patients. Functionally, TTN-AS1 knockdown inhibited cell proliferation, migration, invasion and epithelial-mesenchymal transition in PTC, whereas overexpression of TTN-AS1 led to the opposite results. Mechanistically, TTN-AS1 acted as a competing endogenous RNA by sponging microRNA-153-3p (miR-153-3p) to elevate zinc and ring finger 2 (ZNRF2) expression. Additionally, a high level of TTN-AS1 in PTC was closely correlated with the activity of the phosphoinositide 3-kinase (PI3K)/Akt/mechanistic target of rapamycin (mTOR) pathway. CONCLUSIONS The findings obtained in the present study indicate that TTN-AS1 facilitated PTC progression by regulating the miR-153-3p/ZNRF2 axis and activating the PI3K/Akt/mTOR pathway.
Collapse
Affiliation(s)
- Zhenghui Cui
- Obstetrical Department, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Zhiyan Luo
- Department of Ultrasound, the Second Affiliated Hospital, School of Medical, Zhejiang University, Hangzhou, Zhejiang, China
| | - Zimei Lin
- Department of Ultrasound, the Second Affiliated Hospital, School of Medical, Zhejiang University, Hangzhou, Zhejiang, China
| | - Liuhong Shi
- Department of Ultrasound, the Second Affiliated Hospital, School of Medical, Zhejiang University, Hangzhou, Zhejiang, China
| | - Yurong Hong
- Department of Ultrasound, the Second Affiliated Hospital, School of Medical, Zhejiang University, Hangzhou, Zhejiang, China
| | - Caoxin Yan
- Department of Ultrasound, the Second Affiliated Hospital, School of Medical, Zhejiang University, Hangzhou, Zhejiang, China
| |
Collapse
|
71
|
Wang M, Liu Y, Li C, Zhang Y, Zhou X, Lu C. Long noncoding RNA OIP5-AS1 accelerates the ox-LDL mediated vascular endothelial cells apoptosis through targeting GSK-3β via recruiting EZH2. Am J Transl Res 2019; 11:1827-1834. [PMID: 30972206 PMCID: PMC6456540] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Accepted: 01/08/2019] [Indexed: 06/09/2023]
Abstract
An increasing amount of research is demonstrating the role of long noncoding RNAs (lncRNAs) in human cardiovascular disease, and in particular, atherosclerosis. To date, the mechanism through which lncRNA OIP5-AS1 regulates the oxidative low-density lipoprotein (ox-LDL)-mediated endothelial cell apoptosis is still unclear. Results from this study found that lncRNA OIP5-AS1 was significantly over-expressed in the human umbilical vein endothelial cells (HUVECs) administered with ox-LDL. The silencing of OIP5-AS1 inhibited apoptosis and promoted proliferation via inducing G0/G1 cycle arrest. Chromatin immunoprecipitate (ChIP) revealed that lncRNA OIP5-AS1 reduced GSK-3β expression through recruiting EZH2, a critical element of the Polycomb Repressive Complex 2 (PRC2) complex that directly bind with the GSK-3β promoter region. Rescue experiments validated that GSK-3β could eliminate the effect of OIP5-AS1 on HUVECs. Overall, these findings suggest that lncRNA OIP5-AS1 accelerates ox-LDL mediated vascular endothelial cell apoptosis through targeting GSK-3β via recruiting EZH2, providing potential therapeutic strategies for atherosclerosis.
Collapse
Affiliation(s)
- Minghui Wang
- Tianjin Medical UniversityTianjin 300070, China
- Department of Cardiology, Tianjin Chest HospitalTianjin 300222, China
| | - Yujie Liu
- Department of Cardiology, Tianjin Chest HospitalTianjin 300222, China
| | - Chao Li
- Department of Cardiology, Tianjin Chest HospitalTianjin 300222, China
| | - Ying Zhang
- Department of Cardiology, Tianjin Chest HospitalTianjin 300222, China
| | - Xiujun Zhou
- Department of Cardiology, Tianjin Chest HospitalTianjin 300222, China
| | - Chengzhi Lu
- Department of Cardiology, Tianjin First Center HospitalTianjin 300192, China
| |
Collapse
|
72
|
Guo X, Wei Y, Wang Z, Liu W, Yang Y, Yu X, He J. LncRNA LINC00163 upregulation suppresses lung cancer development though transcriptionally increasing TCF21 expression. Am J Cancer Res 2018; 8:2494-2506. [PMID: 30662806 PMCID: PMC6325476] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2018] [Accepted: 11/12/2018] [Indexed: 06/09/2023] Open
Abstract
Long noncoding RNAs (lncRNAs) have been illustrated as vital molecules in regulating human cancer by emerging evidence. LINC00163 is a novel lncRNA without functional definition. In this study, we investigated its role in the tumorigenesis of lung cancer. The results showed that LINC00163 level was significantly downregulated in lung cancer tissues and cell lines by bioinformatics and qRT-PCR analyses. Notably, we observed that LINC00163 expression was lower in metastatic tissues than in non-metastatic cases. Furthermore, higher expression of LINC00163 in patients with lung cancer predicted better prognosis. Gain-of-function assays illustrated that upregulation of LINC00163 dramatically suppressed the proliferation, migration and invasion of lung cancer cells in vitro, whereas promoting apoptosis. Consistently, LINC00163 overexpression impaired tumor propagation in vivo. Mechanical study revealed that LINC00163 recruited ARID1A to the promoter of TCF21 and initiated its expression. In conclusion, we concluded that LINC00163/ARID1A/TCF21 regulatory loop modulated the development of lung cancer, providing a new insight on the mechanism underlying lung cancer progression.
Collapse
Affiliation(s)
- Xiaotong Guo
- Department of Thoracic Surgical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijing 100021, China
| | - Youlei Wei
- Department of Thoracic Surgery, The Second Affiliated Hospital of Harbin Medical UniversityHarbin 150086, China
| | - Zhe Wang
- Department of Thoracic Surgical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital & Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical CollegeShenzhen 518116, China
| | - Wenyi Liu
- Department of Thoracic Surgical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital & Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical CollegeShenzhen 518116, China
| | - Yikun Yang
- Department of Thoracic Surgical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital & Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical CollegeShenzhen 518116, China
| | - Xin Yu
- Department of Thoracic Surgical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital & Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical CollegeShenzhen 518116, China
| | - Jie He
- Department of Thoracic Surgical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijing 100021, China
| |
Collapse
|
73
|
Han Y, Chen M, Wang A, Fan X. STAT3-induced upregulation of lncRNA CASC11 promotes the cell migration, invasion and epithelial-mesenchymal transition in hepatocellular carcinoma by epigenetically silencing PTEN and activating PI3K/AKT signaling pathway. Biochem Biophys Res Commun 2018; 508:472-479. [PMID: 30503497 DOI: 10.1016/j.bbrc.2018.11.092] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Accepted: 11/14/2018] [Indexed: 10/27/2022]
Abstract
Accumulating evidence suggest that long noncoding RNAs (lncRNAs) are dysregulated in various tumors and serve as crucial regulators in biological processes. Based on The Cancer Genome Atlas (TCGA) database, upregulation of CASC11 was associated with the low overall survival rate of patients with Hepatocellular carcinoma (HCC). However, the function and mechanism of lncRNA CASC11 in the progression of HCC remain unclear. Therefore, we further analyzed the expression pattern and biological role of CASC11 in HCC. CASC11 was found to be overexpressed in HCC tissues and cell lines and predicted a poor prognosis. Loss of CASC11 function efficiently suppressed cell migration, invasion and epithelial-mesenchymal transition (EMT). The mechanism which led to the upregulation of CASC11 was investigated. CASC11 was found to be activated by the transcription factor STAT3. Mechanically, the enhancer of zeste homolog 2 (EZH2) was found to be a binding partner of CASC11. Moreover, CASC11 epigenetically silenced PTEN by binding with EZH2. Finally, rescue assays were conducted to make confirmation. The present results revealed that CASC11 may be potential therapeutic target in HCC.
Collapse
Affiliation(s)
- Yidi Han
- Department of Second Liver Disease, Qingdao Sixth People's Hospital, No.9, Fushun Road, Sifang District, Qingdao City, Shandong, 266000, China
| | - Meizhu Chen
- Department of Blood Purification, Qingdao Sixth People's Hospital, No.9, Fushun Road, Sifang District, Qingdao City, Shandong, 266000, China
| | - Aili Wang
- Medical Laboratory, Qingdao Sixth People's Hospital, No. 9, Fushun Road, Sifang District, Qingdao, Shandong, 266000, China
| | - Xiaoping Fan
- Department of Second Liver Disease, Qingdao Sixth People's Hospital, No.9, Fushun Road, Sifang District, Qingdao, Shandong, 266000, China.
| |
Collapse
|