251
|
Wang L, Du L, Duan W, Yan S, Xie Y, Wang C. Overexpression of long noncoding RNA NORAD in colorectal cancer associates with tumor progression. Onco Targets Ther 2018; 11:6757-6766. [PMID: 30349308 PMCID: PMC6188072 DOI: 10.2147/ott.s176354] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Purpose The aim of this study was to elucidate the role and clinical significance of long noncoding RNA-activated by DNA damage (NORAD) in colorectal cancer (CRC). Methods Sixty pairs of tumorous and adjacent nontumorous tissues derived from CRC patients were subjected to quantitative real-time polymerase chain reaction to determine the expression level of NORAD. The serum levels of NORAD expression were also measured in an independent cohort of CRC patients as well as patients with benign diseases and healthy controls. Comparative analyses were performed to investigate the relationships between NORAD levels in tissues and clinicopathological features of CRC. Receiver operating characteristic (ROC) curve analysis was used to assess the diagnostic value of NORAD in patients with CRC. Furthermore, the potential functions of NORAD in the development of CRC were explored in vitro, using the HCT116 and SW1116 CRC cell lines. Result NORAD expression was significantly upregulated in the tumorous tissues of CRC patients (P<0.001) compared to the adjacent nontumorous tissues. Higher NORAD expression was associated with advanced CRC. Moreover, serum levels supported that NORAD could distinguish CRC patients from healthy controls and patients with benign diseases, indicating a potential diagnostic role in CRC. The ROC curve analysis showed a diagnostic efficacy with area under the curve of 0.800 (95% CI: 0.737–0.853). Mechanistic investigations indicated that NORAD silencing reduced CRC cell proliferation, migration, and invasion. Conclusion NORAD may serve as a novel predictor in CRC and may be a potential target for future therapy.
Collapse
Affiliation(s)
- Lili Wang
- Department of Clinical Laboratory, Qilu Hospital of Shandong University, Jinan, Shandong 250012, People's Republic of China
| | - Lutao Du
- Department of Clinical Laboratory, The Second Hospital of Shandong University, Jinan, Shandong 250033, People's Republic of China,
| | - Weili Duan
- Department of Clinical Laboratory, The Second Hospital of Shandong University, Jinan, Shandong 250033, People's Republic of China,
| | - Suzhen Yan
- Department of Clinical Laboratory, The Second Hospital of Shandong University, Jinan, Shandong 250033, People's Republic of China,
| | - Yujiao Xie
- Department of Clinical Laboratory, The Second Hospital of Shandong University, Jinan, Shandong 250033, People's Republic of China,
| | - Chuanxin Wang
- Department of Clinical Laboratory, The Second Hospital of Shandong University, Jinan, Shandong 250033, People's Republic of China,
| |
Collapse
|
252
|
Ma J, Wu K, Liu K, Miao R. Effects of MALAT1 on proliferation and apo- ptosis of human non-small cell lung cancer A549 cells in vitro and tumor xenograft growth in vivo by modulating autophagy. Cancer Biomark 2018; 22:63-72. [PMID: 29439314 DOI: 10.3233/cbm-170917] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
OBJECTIVE To explore the ability of MALAT1 to influence non-small cell lung cancer (NSCLC) A549 cells in vitro and tumor xenograft growth in vivo by modulating autophagy. METHODS LncRNA MALAT-1 in normal HBE cells and human NSCLC cells was measured. A549 cells were treated with si-MALAT-1, negative control and si-MALAT-1 + rapamycin. The mRNA levels of MALAT-1, P62 and LC3 was determined by the qRT-PCR and the protein levels of autophagy-related proteins by the western blotting. The CCK8 assay was performed for cell proliferation, the scratch test for cell migration, the Transwell assay for cell invasion, and the flow cytometry for cell cycle and apoptosis. Tumor xenograft in nude mice is performed to test tumorigenesis of the transfected A549 cells. RESULTS The expression level of MALAT-1 in A549, SPC-A-1 and NCI-H460 cells was increased compared to HBE cells. And A549 with a high expression level of MALAT-1 were selected for cell transfection. si-MALAT-1 decreased cell proliferation, migration, invasion, and LC3-II/LC3-I ratio, reduced cell cycle progression, and increased cell apoptosis and P62 protein expression. No significant difference was found between A549 cells and A549 cells transfected with si-MALAT-1 + RAPA, A549 cells transfected with NC and A549 cells transfected with si-MALAT-1 + RAPA. Nude mice injected with A549 cells transfected with si-MALAT-1 had smallest tumor on size and weight among other nude mice. CONCLUSION Downregulation of MALAT1 may promote apoptosis and suppress proliferation, migration and invasion of human NSCLC A549 cells by inhibiting autophagy, thereby suppressing the development of NSCLC.
Collapse
Affiliation(s)
- Jun Ma
- Thoracic Surgery Department, 1st Affiliated Hospital of Sun Yat-Sen University, Guangzhou 510080, Guangdong, China
| | - Kaiming Wu
- Colorectal Surgery Department, 1st Affiliated Hospital of Sun Yat-Sen University, Guangzhou 510080, Guangdong, China
| | - Kuanzhi Liu
- Department of Anaesthesiology, 1st Affiliated Hospital of Sun Yat-Sen University, Guangzhou 510080, Guangdong, China
| | - Rong Miao
- Physical Exam. Center, The Eastern Hospital of 1st Affiliated Hospital of Sun Yat-Sen University, Guangzhou 510700, Guangdong, China
| |
Collapse
|
253
|
Wang Y, Wang Y, Liu F. A 44-gene set constructed for predicting the prognosis of clear cell renal cell carcinoma. Int J Mol Med 2018; 42:3105-3114. [PMID: 30272265 PMCID: PMC6202093 DOI: 10.3892/ijmm.2018.3899] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2018] [Accepted: 09/07/2018] [Indexed: 12/14/2022] Open
Abstract
Clear cell renal cell carcinoma (ccRCC) is the most frequent type of renal cell carcinoma (RCC). The present study aimed to examine prognostic markers and construct a prognostic prediction system for ccRCC. The mRNA sequencing data of ccRCC was downloaded from The Cancer Genome Atlas (TCGA) database, and the GSE40435 dataset was obtained from the Gene Expression Omnibus database. Using the Limma package, the differentially expressed genes (DEGs) in the TCGA dataset and GSE40435 dataset were obtained, respectively, and the overlapped DEGs were selected. Subsequently, Cox regression analysis was applied for screening prognosis-associated genes. Following visualization of the co-expression network using Cytoscape software, the network modules were examined using the GraphWeb tool. Functional annotation for genes in the network was performed using the clusterProfiler package. Finally, a prognostic prediction system was constructed through Bayes discriminant analysis and confirmed with the GSE29609 validation dataset. The results revealed a total of 263 overlapped DEGs and 161 prognosis-associated genes. Following construction of the co-expression network, 16 functional terms and three pathways were obtained for genes in the network. In addition, red, yellow (Involving chemokine ligand 10 (CXCL10), CD27 molecule (CD27) and runt-related transcription factor 3 (RUNX3)], green (Involving angiopoietin-like 4 (ANGPTL4), stannio-calcin 2 (STC2), and sperm associated antigen 4 (SPAG4)], and cyan modules were extracted from the co-expression network. Additionally, the prognostic prediction system involving 44 signature genes, including ANGPTL4, STC2, CXCL10, SPAG4, CD27, matrix metalloproteinase (MMP9) and RUNX3, was identified and confirmed. In conclusion, the 44-gene prognostic prediction system involving ANGPTL4, STC2, CXCL10, SPAG4, CD27, MMP9 and RUNX3 may be utilized for predicting the prognosis of patients with ccRCC.
Collapse
Affiliation(s)
- Yonggang Wang
- Department of Urology, China‑Japan Union Hospital of Jilin University, Changchun, Jilin 130033, P.R. China
| | - Yao Wang
- Department of Urology, China‑Japan Union Hospital of Jilin University, Changchun, Jilin 130033, P.R. China
| | - Feng Liu
- Department of Urology, China‑Japan Union Hospital of Jilin University, Changchun, Jilin 130033, P.R. China
| |
Collapse
|
254
|
Chen X, Zeng K, Xu M, Hu X, Liu X, Xu T, He B, Pan Y, Sun H, Wang S. SP1-induced lncRNA-ZFAS1 contributes to colorectal cancer progression via the miR-150-5p/VEGFA axis. Cell Death Dis 2018; 9:982. [PMID: 30250022 PMCID: PMC6155123 DOI: 10.1038/s41419-018-0962-6] [Citation(s) in RCA: 159] [Impact Index Per Article: 22.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2018] [Revised: 06/24/2018] [Accepted: 07/17/2018] [Indexed: 02/07/2023]
Abstract
Increasing long non-coding RNAs (lncRNAs) have been reported to play key roles in the development and progression of various malignancies. ZNFX1 antisense RNA1 (ZFAS1) has been reported to be aberrant expression and suggested as a tumor suppressor or oncogene in many cancers. However, the biological role and underlying molecular mechanism of ZFAS1, especially the miRNA sponge role of which in CRC remain largely unknown. We found that ZFAS1 expression was higher in CRC tissues, where it was associated with poor overall survival (OS), we also showed that ZFAS1 upregulation was induced by nuclear transcription factor SP1. Moreover, ZFAS1 and VEGFA are both targets of miR-150-5p, while ZFAS1 binds to miR-150-5p in an AGO2-dependent manner. Additionally, ZFAS1 upregulation markedly promoted as well as ZFAS1 knockdown significantly suppressed CRC cell proliferation, migration, invasion and angiogenesis, and the inhibitory effect caused by ZFAS1 knockdown could be reversed by antagomiR-150-5p. Lastly, we demonstrated that ZFAS1 knockdown inhibited EMT process and inactivated VEGFA/VEGFR2 and downstream Akt/mTOR signaling pathway in CRC. Our data demonstrated that SP1-induced ZFAS1 contributed to CRC progression by upregulating VEGFA via competitively binding to miR-150-5p, which acts as a tumor suppressor by targeting VEGFA in CRC.
Collapse
Affiliation(s)
- Xiaoxiang Chen
- General Clinical Research Center, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210006, Jiangsu, China
- Medical College, Southeast University, Nanjing, 210009, Jiangsu, China
| | - Kaixuan Zeng
- General Clinical Research Center, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210006, Jiangsu, China
- Medical College, Southeast University, Nanjing, 210009, Jiangsu, China
| | - Mu Xu
- General Clinical Research Center, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210006, Jiangsu, China
| | - Xiuxiu Hu
- General Clinical Research Center, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210006, Jiangsu, China
- Medical College, Southeast University, Nanjing, 210009, Jiangsu, China
| | - Xiangxiang Liu
- General Clinical Research Center, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210006, Jiangsu, China
| | - Tao Xu
- General Clinical Research Center, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210006, Jiangsu, China
| | - Bangshun He
- General Clinical Research Center, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210006, Jiangsu, China
| | - Yuqin Pan
- General Clinical Research Center, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210006, Jiangsu, China
| | - Huiling Sun
- General Clinical Research Center, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210006, Jiangsu, China
| | - Shukui Wang
- General Clinical Research Center, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210006, Jiangsu, China.
| |
Collapse
|
255
|
Abstract
SIGNIFICANCE The emerging connections between an increasing number of long noncoding RNAs (lncRNAs) and oncogenic hallmarks provide a new twist to tumor complexity. Recent Advances: In the present review, we highlight specific lncRNAs that have been studied in relation to tumorigenesis, either as participants in the neoplastic process or as markers of pathway activity or drug response. These transcripts are typically deregulated by oncogenic or tumor-suppressing signals or respond to microenvironmental conditions such as hypoxia. CRITICAL ISSUES Among these transcripts are lncRNAs sufficiently divergent between mouse and human genomes that may contribute to biological differences between species. FUTURE DIRECTIONS From a translational standpoint, knowledge about primate-specific lncRNAs may help explain the reason behind the failure to reproduce the results from mouse cancer models in human cell-based systems. Antioxid. Redox Signal. 29, 922-935.
Collapse
Affiliation(s)
- Xue Wu
- 1 Department of Medicine, Indiana University School of Medicine , Indianapolis, Indiana.,2 Department of Microbiology and Immunology, Indiana University School of Medicine , Indianapolis, Indiana
| | - Oana M Tudoran
- 1 Department of Medicine, Indiana University School of Medicine , Indianapolis, Indiana.,3 Department of Functional Genomics and Experimental Pathology, The Oncology Institute "Prof. Dr. I. Chiricuta," Cluj-Napoca, Romania
| | - George A Calin
- 4 Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center , Houston, Texas.,5 Center for RNA Interference and Non-Coding RNA, The University of Texas MD Anderson Cancer Center , Houston, Texas
| | - Mircea Ivan
- 1 Department of Medicine, Indiana University School of Medicine , Indianapolis, Indiana.,2 Department of Microbiology and Immunology, Indiana University School of Medicine , Indianapolis, Indiana
| |
Collapse
|
256
|
Ji D, Jiang C, Zhang L, Liang N, Jiang T, Yang B, Liang H. LncRNA CRNDE promotes hepatocellular carcinoma cell proliferation, invasion, and migration through regulating miR-203/ BCAT1 axis. J Cell Physiol 2018; 234:6548-6560. [PMID: 30230527 DOI: 10.1002/jcp.27396] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2018] [Accepted: 08/17/2018] [Indexed: 12/27/2022]
Abstract
OBJECTIVE To investigate the impact of long noncodingRNA (lncRNA) colorectal neoplasia differentially expressed (CRNDE) on hepatocellular cancer (HCC) cell propagation, invasion, and migration by mediating miR-203/ BCAT1 axis. METHODS Microarray analysis was based on 25 pairs of HCC cancerous tissues and adjacent tissues. The expression levels of CRNDE, miR-203, and BCAT1 in HCC tissues were analyzed by quantitative real-time polymerase chain reaction (qRT-PCR). The liver cell line L-02 and HCC cell lines HepG2 and Huh-7 were utilized to assess the regulatory effects of CRNDE and miR-203 on HCC progression in vitro. Western blot was used to qualify BCAT1 protein expression level. Cell proliferation and apoptosis were evaluated using CCK-8 and flow cytometry analysis, whereas cell invasion and migration assay were performed by the Transwell assay. The relationship among CRNDE, miR-203, and BCAT1 was validated by dual luciferase assay. Tumor Xenograft study was established to verify the pathological effect of CRNDE on HCC development in vivo. RESULTS The expression levels of the CRNDE and BCAT1 were upregulated in HCC tissues and cells, whereas miR-203 was downregulated in HCC. Knockdown of CRNDE or miR-203 overexpression would inhibit HCC cell propagation and metastasis, and induced cell apoptosis. Moreover, miR-203 was negatively correlated with CRNDE, the same as miR-203 with BCAT1. Dual luciferase assay showed that miR-203 was an inhibitory target of CRNDE, and BCAT1 was directly targeted by miR-203 as well. CONCLUSION LncRNA CRNDE could enhance HCC tumorgenesis by sponging miR-203 and mediating BCAT1. LncRNA CRNDE might facilitate HCC cell propagation, invasiveness, and migration through regulating miR-203/ BCAT1 axis.
Collapse
Affiliation(s)
- Degang Ji
- Department of Hepatopancreatobiliary Surgery, China-Japan Union Hospital of Jilin University, Changchun, Jilin, China
| | - Chengwei Jiang
- Department of Pathology, China-Japan Union Hospital of Jilin University, Changchun, Jilin, China
| | - Lirong Zhang
- Department of Pathology, China-Japan Union Hospital of Jilin University, Changchun, Jilin, China
| | - Na Liang
- Office of Surgical Nursing, Changchun Medical College, Changchun, Jilin, China
| | - Tiechao Jiang
- Department of Cardiovascular Medicine, China-Japan Union Hospital of Jilin University, Changchun, Jilin, China
| | - Bin Yang
- Department of Breast Surgery, China-Japan Union Hospital of Jilin University, Changchun, Jilin, China
| | - Haiying Liang
- Department of Blood transfusion, China-Japan Union Hospital of Jilin University, Changchun, Jilin, China
| |
Collapse
|
257
|
Wang C, Wang G, Zhang Z, Wang Z, Ren M, Wang X, Li H, Yu Y, Liu J, Cai L, Li Y, Zhang D, Zhang C. The downregulated long noncoding RNA DHRS4-AS1 is protumoral and associated with the prognosis of clear cell renal cell carcinoma. Onco Targets Ther 2018; 11:5631-5646. [PMID: 30254456 PMCID: PMC6141115 DOI: 10.2147/ott.s164984] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
BACKGROUND Long noncoding RNAs (lncRNAs) have been identified as important factors in cancer biology and are deregulated in many cancers. The present study aimed to determine the expression and roles of lncRNA DHRS4-AS1 in the progression of clear cell renal cell carcinoma (ccRCC). METHODS AND RESULTS Using high-throughput RNA-sequencing data of ccRCC tumors from the Cancer Genome Atlas project, we identified lncRNA DHRS4-AS1 as significantly associated with ccRCC patients' overall survival. We confirmed the downregulation of DHRS4-AS1 in ccRCC by assessing its expression levels in a cohort of 52 tumor and paired non-tumor samples. In addition, we found that low DHRS4-AS1 expression was significantly associated with a high tumor node metastasis stage, lymph node metastasis, advanced pathological grade and poor prognosis. Furthermore, DHRS4-AS1 overexpression inhibited the progression of cell cycles of ccRCC in vitro. These data indicate that DHRS4-AS1 functions by preventing the proliferation and invasion, inhibiting the cell cycle progression and promoting the apoptosis of ccRCC cells. CONCLUSION Taken together, our findings identify the role of DHRS4-AS1 as a tumor inhibitor in ccRCC for the first time, demonstrating that DHRS4-AS1 is a potential prognostic biomarker that could potentially be applied in ccRCC therapy.
Collapse
Affiliation(s)
- Changlin Wang
- Department of Urology, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China,
| | - Gang Wang
- Department of Urology, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China,
| | - Zijian Zhang
- Department of Urology, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China,
| | - Zichun Wang
- Department of Urology, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China,
| | - Minghua Ren
- Department of Urology, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China,
| | - Xiaoxiong Wang
- Department of Neurosurgery, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China,
| | - Haoming Li
- Department of Urology, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China,
| | - Yipeng Yu
- Department of Urology, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China,
| | - Jing Liu
- Department of Urology, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China,
| | - Licheng Cai
- Department of Urology, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China,
| | - Yong Li
- Department of PET/CT, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Daming Zhang
- Department of Neurosurgery, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China,
| | - Cheng Zhang
- Department of Urology, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China,
| |
Collapse
|
258
|
Wang Z, Katsaros D, Biglia N, Shen Y, Fu Y, Loo LWM, Jia W, Obata Y, Yu H. High expression of long non-coding RNA MALAT1 in breast cancer is associated with poor relapse-free survival. Breast Cancer Res Treat 2018; 171:261-271. [PMID: 29845475 PMCID: PMC6488226 DOI: 10.1007/s10549-018-4839-2] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2018] [Accepted: 05/25/2018] [Indexed: 12/14/2022]
Abstract
PURPOSE Metastasis-associated lung adenocarcinoma transcript 1 (MALAT1) has been identified as a prognostic marker for the metastasis of early-stage non-small cell lung cancer (NSCLCs). We studied MALAT1 expression in breast cancer in relation to disease features and patient survival. METHODS Quantitative reverse transcriptase-polymerase chain reaction (qRT-PCR) was used to measure MALAT1 expression in tumor samples of 509 breast cancer patients. Hazards ratios (HRs) and 95% confidence intervals (CIs) were calculated to assess the association between MALAT1 expression and breast cancer survival using the Cox proportional hazards regression model, and the analysis was adjusted for age at surgery, tumor grade, disease stage, and hormone receptor status. Meta-analysis of multiple microarray datasets from online databases and our own study was performed to evaluate the association of MALAT1 with breast cancer survival. RESULTS Patients with low-grade or ER-positive tumors had higher expression of MALAT1 compared to those with high-grade (p = 0.013) or ER-negative (p = 0.0002) tumors. Patients with PR-positive tumors also had higher MALAT1 expression than those with PR-negative tumors (p < 0.0001). In patients with positive hormone receptors or low tumor grade, tumors with high MALAT1 expression were more likely to recur. Survival analysis showed that patients with high expression of MALAT1 had a twofold increase in risk of relapse (p = 0.0083) compared to those with low expression. This association remained significant after adjustment for age at surgery, disease stage, tumor grade, and hormone receptor status. Meta-analysis showed that high MALAT1 expression was associated with poor relapse-free survival in patients with hormone receptor-positive tumors (HR 1.44, 95% CI 1.08-1.92). CONCLUSIONS High expression of lncRNA MALAT1 is associated with breast cancer relapse and may play a role in tumor progression.
Collapse
Affiliation(s)
- Zhanwei Wang
- Cancer Epidemiology Program, University of Hawaii Cancer Center, 701 Ilalo Street, Honolulu, HI, 96813, USA
| | - Dionyssios Katsaros
- Department of Surgical Sciences, Gynecology, AOU Città della Salute, University of Torino School of Medicine, Turin, Italy
| | - Nicoletta Biglia
- Department of Surgical Sciences, Division of Obstetrics and Gynecology, University of Torino School of Medicine, Mauriziano Hospital, Turin, Italy
| | - Yi Shen
- Cancer Epidemiology Program, University of Hawaii Cancer Center, 701 Ilalo Street, Honolulu, HI, 96813, USA
| | - Yuanyuan Fu
- Cancer Epidemiology Program, University of Hawaii Cancer Center, 701 Ilalo Street, Honolulu, HI, 96813, USA
- Department of Molecular Biosciences & Bioengineering, University of Hawaii at Manoa, Honolulu, HI, USA
| | - Lenora W M Loo
- Cancer Epidemiology Program, University of Hawaii Cancer Center, 701 Ilalo Street, Honolulu, HI, 96813, USA
| | - Wei Jia
- Cancer Epidemiology Program, University of Hawaii Cancer Center, 701 Ilalo Street, Honolulu, HI, 96813, USA
| | - Yuki Obata
- College of Pharmacy, Kinjo Gakuin University, Nagoya, Aichi, Japan
| | - Herbert Yu
- Cancer Epidemiology Program, University of Hawaii Cancer Center, 701 Ilalo Street, Honolulu, HI, 96813, USA.
| |
Collapse
|
259
|
Wang Q, Wu G, Zhang Z, Tang Q, Zheng W, Chen X, Chen F, Li Q, Che X. Long non-coding RNA HOTTIP promotes renal cell carcinoma progression through the regulation of the miR-615/IGF-2 pathway. Int J Oncol 2018; 53:2278-2288. [PMID: 30226576 DOI: 10.3892/ijo.2018.4539] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2017] [Accepted: 07/27/2018] [Indexed: 11/06/2022] Open
Abstract
Emerging evidence has indicated that long non‑coding RNA (lncRNA) HOXA transcript at the distal tip (HOTTIP) regulates cell growth, differentiation, apoptosis and cancer progression. However, the expression and function of HOTTIP in the progression of renal cell carcinoma (RCC) remain largely unknown. In this study, we investigated the role of the lncRNA HOTTIP in RCC. The expression levels of HOTTIP in RCC tissues and cell lines were determined by RT‑qPCR. The association between HOTTIP expression and clinicopathological characteristics and prognosis was analyzed in patients with RCC from the TCGA database. Loss‑of‑ function assays were designed and conducted to verify the oncogenic function of HOTTIP in RCC progression. Luciferase assay was performed to explore the mechanisms of the miRNA‑lncRNA sponge. The results revealed that HOTTIP expression was upregulated in RCC. An increased HOTTIP expression in RCC was associated with a larger tumor size and a higher clinical stage, lymph node metastasis and vascular invasion. Additionally, patients RCC with a high HOTTIP expression had a significantly shorter overall survival (OS) and disease‑free survival (DFS). HOTTIP knockdown significantly inhibited cell proliferation, migration and invasion, and increased the apoptosis of RCC cells in vitro. Mechanistic analyses revealed that HOTTIP functioned as a competing endogenous RNA (ceRNA) for hsa‑miR‑615‑3p, and led to the derepression of its endogenous target, insulin‑like growth factor-2 (IGF‑2), which is a protein hormone that exerts a stimulatory effect on tumor cell growth. miR‑615 inhibition reversed the suppressive effects of HOTTIP knockdown on RCC cell progression. HOTTIP regulated IGF‑2 expression in a miR‑615‑dependent manner in RCC cells. In addition, IGF‑2 expression was significantly upregulated in the RCC specimens and a positive association between the expression of HOTTIP and IGF‑2 in RCC tissues was detected. The effect of HOTTIP was abolished by the siRNA‑mediated silencing of IGF-2 in RCC cells. On the whole, this study demonstrates, for the first time, at least to the best of our knowledge, that the HOTTIP/miR‑615/IGF‑2 axis plays an important role in RCC progression and potentially contributes to the improvement of RCC diagnosis and therapy.
Collapse
Affiliation(s)
- Qifei Wang
- Department of Urology, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning 116011, P.R. China
| | - Guangzhen Wu
- Department of Urology, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning 116011, P.R. China
| | - Zhiwei Zhang
- Department of Urology, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning 116011, P.R. China
| | - Qizhen Tang
- Department of Urology, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning 116011, P.R. China
| | - Wei Zheng
- Department of Urology, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning 116011, P.R. China
| | - Xiaochi Chen
- Department of Urology, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning 116011, P.R. China
| | - Feng Chen
- Department of Urology, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning 116011, P.R. China
| | - Quanlin Li
- Department of Urology, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning 116011, P.R. China
| | - Xiangyu Che
- Department of Urology, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning 116011, P.R. China
| |
Collapse
|
260
|
Lei L, Chen J, Huang J, Lu J, Pei S, Ding S, Kang L, Xiao R, Zeng Q. Functions and regulatory mechanisms of metastasis‐associated lung adenocarcinoma transcript 1. J Cell Physiol 2018; 234:134-151. [PMID: 30132842 DOI: 10.1002/jcp.26759] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2018] [Accepted: 04/26/2018] [Indexed: 12/19/2022]
Affiliation(s)
- Li Lei
- Department of Dermatology, Xiangya Hospital Central South University Changsha Hunan China
- Department of Hunan Key Laboratory of Skin Cancer and Psoriasis Xiangya Hospital, Central South University Changsha Hunan China
| | - Jing Chen
- Department of Dermatology Third Xiangya Hospital, Central South University Changsha Hunan China
| | - Jinhua Huang
- Department of Dermatology Third Xiangya Hospital, Central South University Changsha Hunan China
| | - Jianyun Lu
- Department of Dermatology Third Xiangya Hospital, Central South University Changsha Hunan China
| | - Shiyao Pei
- Department of Dermatology Third Xiangya Hospital, Central South University Changsha Hunan China
| | - Shu Ding
- Department of Dermatology Third Xiangya Hospital, Central South University Changsha Hunan China
| | - Liyang Kang
- Department of Dermatology Third Xiangya Hospital, Central South University Changsha Hunan China
| | - Rong Xiao
- Department of Dermatology Second Xiangya Hospital, Central South University Changsha Hunan China
| | - Qinghai Zeng
- Department of Dermatology Third Xiangya Hospital, Central South University Changsha Hunan China
| |
Collapse
|
261
|
Liu S, Yang Y, Wang W, Pan X. Long noncoding RNA TUG1 promotes cell proliferation and migration of renal cell carcinoma via regulation of YAP. J Cell Biochem 2018; 119:9694-9706. [PMID: 30132963 DOI: 10.1002/jcb.27284] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2018] [Accepted: 06/27/2018] [Indexed: 12/25/2022]
Abstract
OBJECTIVES Recently, long noncoding RNAs (lncRNAs) have captured much attention for their important roles in human diseases. Deregulation of lncRNA taurine-upregulated gene 1 (TUG1) has been reported to regulate cancer progression in many cancer types. However, how TUG1 contributes to renal cell carcinoma (RCC) remains elusive; we were eager to resolve the questions. METHODS Tumor tissues and the matched adjacent normal tissues were collected from patients with RCC. Messenger RNA (mRNA) levels of TUG1, yes-associated protein (YAP), and microRNA (miR)-9 levels were determined by reverse transcription quantitative polymerase chain reaction (RT-qPCR). The regulation of YAP by TUG1 was investigated using Western blot analysis, RT-qPCR, and immunofluorescence. The oncogenic roles of TUG1 and YAP were studied using a cell proliferation assay and a wound healing assay. The interaction of TUG1-miR-9-YAP was analyzed in RCC cell lines. RESULTS In the current study, we observed a positive correlation between TUG1 expression and YAP expression in RCC using the Gene Expression Omnibus database and tumor tissues collected from 58 patients with RCC. The TUG1 elevation enhanced YAP expression but did not alter the Hippo-signaling pathway activity or YAP protein distribution in cells. In addition, we found that TUG1 could bind to miR-9; therefore, TUG1 could positively control YAP expression via downregulation of miR-9 level. Furthermore, we observed that inhibition of cell proliferation and cell migration induced by TUG1 silencing could be reversed by overexpression of YAP in RCC cell lines. CONCLUSIONS Our findings indicated a pivotal role of TUG1 in driving RCC progression via regulation of miR-9/YAP, suggesting a potential therapeutic targeting role of TUG1 in RCC.
Collapse
Affiliation(s)
- Shan Liu
- Department of Pediatrics, The First Affiliated Hospital of Henan University of Science and Technology, Luoyang, China
| | - Yantong Yang
- Cancer Institute, Henan University of Science and Technology, Luoyang, China.,Department of Gastrointestinal Oncology Surgery, The First Affiliated Hospital of Henan University of Science and Technology, Luoyang, China
| | - Weiwei Wang
- Department of Pediatrics, The First Affiliated Hospital of Henan University of Science and Technology, Luoyang, China
| | - Xiaoyue Pan
- Department of Pediatrics, The First Affiliated Hospital of Henan University of Science and Technology, Luoyang, China
| |
Collapse
|
262
|
Li J, Cui Z, Li H, Lv X, Gao M, Yang Z, Bi Y, Zhang Z, Wang S, Zhou B, Yin Z. Clinicopathological and prognostic significance of long noncoding RNA MALAT1 in human cancers: a review and meta-analysis. Cancer Cell Int 2018; 18:109. [PMID: 30093838 PMCID: PMC6080354 DOI: 10.1186/s12935-018-0606-z] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2018] [Accepted: 07/31/2018] [Indexed: 12/11/2022] Open
Abstract
Background The aberrant regulation of MALAT1 has been indicated to be involved in various carcinogenic pathways contributing to the tumourigenesis and progression of cancers. The current meta-analysis summarized the research advances of MALAT1 functions and analyzed its prognostic value among multiple types of cancers. Methods Eligible studies were identified through retrieving the PubMed, Web of Science, and CNKI databases, up to Mar 1, 2018. 28 studies of 5436 patients and 36 studies of 3325 patients were enrolled in the meta-analysis to evaluate the association of MALAT1 expression with survival outcomes and clinical parameters. Results The results demonstrated that over-expression of MALAT1 may predict lymph node metastasis (pooled OR = 2.335, 95% CI 1.606–3.395, P = 0.000) and distant metastasis (pooled OR = 2.456, 95% CI 1.407–4.286, P = 0.002). Moreover, MALAT1 was also related with tumour size (pooled OR = 1.875, 95% CI 1.257–2.795, P = 0.002) and TNM stage (pooled OR = 2.034, 95% CI 1.111–3.724, P = 0.021). Additionally, elevated MALAT1 expression could predict poor OS (pooled HR = 2.298, 95% CI 1.953–2.704, P = 0.000), DFS (pooled HR = 2.036, 95% CI 1.240–3.342, P = 0.005), RFS (pooled HR = 2.491, 95% CI 1.505–4.123, P = 0.000), DSS (pooled HR = 2.098, 95% CI 1.372–3.211, P = 0.001) and PFS (pooled HR = 1.842, 95% CI 1.138–2.983, P = 0.013) in multivariate model. Importantly, subgroup analyses disclosed that increased MALAT1 expression had a poor OS among different cancer types (Estrogen-dependent cancer: pooled HR = 2.656, 95% CI 1.560–4.523; urological cancer: pooled HR = 1.952, 95% CI 1.189–3.204; glioma: pooled HR = 2.315, 95% CI 1.643–3.263; digestive cancer: pooled HR = 2.451, 95% CI 1.862–3.227). Conclusions The present findings demonstrated that MALAT1 may be a novel biomarker for predicting survival outcome, lymph node metastasis and distant metastasis. Electronic supplementary material The online version of this article (10.1186/s12935-018-0606-z) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Juan Li
- 1Department of Epidemiology, School of Public Health, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang, 110122 People's Republic of China.,2Key Laboratory of Cancer Etiology and Intervention, University of Liaoning Province, Shenyang, 110122 People's Republic of China
| | - Zhigang Cui
- 3School of Nursing, China Medical University, Shenyang, 110122 China
| | - Hang Li
- 1Department of Epidemiology, School of Public Health, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang, 110122 People's Republic of China.,2Key Laboratory of Cancer Etiology and Intervention, University of Liaoning Province, Shenyang, 110122 People's Republic of China
| | - Xiaoting Lv
- 1Department of Epidemiology, School of Public Health, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang, 110122 People's Republic of China.,2Key Laboratory of Cancer Etiology and Intervention, University of Liaoning Province, Shenyang, 110122 People's Republic of China
| | - Min Gao
- 1Department of Epidemiology, School of Public Health, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang, 110122 People's Republic of China.,2Key Laboratory of Cancer Etiology and Intervention, University of Liaoning Province, Shenyang, 110122 People's Republic of China
| | - Zitai Yang
- 1Department of Epidemiology, School of Public Health, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang, 110122 People's Republic of China.,2Key Laboratory of Cancer Etiology and Intervention, University of Liaoning Province, Shenyang, 110122 People's Republic of China
| | - Yanhong Bi
- 1Department of Epidemiology, School of Public Health, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang, 110122 People's Republic of China.,2Key Laboratory of Cancer Etiology and Intervention, University of Liaoning Province, Shenyang, 110122 People's Republic of China
| | - Ziwei Zhang
- 1Department of Epidemiology, School of Public Health, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang, 110122 People's Republic of China.,2Key Laboratory of Cancer Etiology and Intervention, University of Liaoning Province, Shenyang, 110122 People's Republic of China
| | - Shengli Wang
- 1Department of Epidemiology, School of Public Health, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang, 110122 People's Republic of China.,2Key Laboratory of Cancer Etiology and Intervention, University of Liaoning Province, Shenyang, 110122 People's Republic of China
| | - Baosen Zhou
- 1Department of Epidemiology, School of Public Health, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang, 110122 People's Republic of China.,2Key Laboratory of Cancer Etiology and Intervention, University of Liaoning Province, Shenyang, 110122 People's Republic of China
| | - Zhihua Yin
- 1Department of Epidemiology, School of Public Health, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang, 110122 People's Republic of China.,2Key Laboratory of Cancer Etiology and Intervention, University of Liaoning Province, Shenyang, 110122 People's Republic of China
| |
Collapse
|
263
|
Rafiee A, Riazi-Rad F, Havaskary M, Nuri F. Long noncoding RNAs: regulation, function and cancer. Biotechnol Genet Eng Rev 2018; 34:153-180. [PMID: 30071765 DOI: 10.1080/02648725.2018.1471566] [Citation(s) in RCA: 92] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Long noncoding RNAs (lncRNAs) are non-protein-coding RNA transcripts that exert a key role in many cellular processes and have potential toward addressing disease etiology. Here, we review existing noncoding RNA classes and then describe a variety of mechanisms and functions by which lncRNAs regulate gene expression such as chromatin remodeling, genomic imprinting, gene transcription and post-transcriptional processing. We also examine several lncRNAs that contribute significantly to pathogenesis, oncogenesis, tumor suppression and cell cycle arrest of diverse cancer types and also give a summary of the pathways that lncRNAs might be involved in.
Collapse
Affiliation(s)
- Aras Rafiee
- a Department of Biology , Central Tehran Branch, Islamic Azad University , Tehran , Iran
| | - Farhad Riazi-Rad
- b Immunology Department , Pasteur institute of Iran , Tehran , Iran
| | - Mohammad Havaskary
- c Young Researchers Club, Central Tehran Branch, Islamic Azad University , Tehran , Iran
| | - Fatemeh Nuri
- d Department of Biology , Central Tehran Branch, Islamic Azad University , Tehran , Iran
| |
Collapse
|
264
|
Kulkarni P, Dasgupta P, Bhat NS, Shahryari V, Shiina M, Hashimoto Y, Majid S, Deng G, Saini S, Tabatabai ZL, Yamamura S, Tanaka Y, Dahiya R. Elevated miR-182-5p Associates with Renal Cancer Cell Mitotic Arrest through Diminished MALAT-1 Expression. Mol Cancer Res 2018; 16:1750-1760. [PMID: 30037856 DOI: 10.1158/1541-7786.mcr-17-0762] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2017] [Revised: 05/03/2018] [Accepted: 07/03/2018] [Indexed: 12/23/2022]
Abstract
The molecular heterogeneity of clear cell renal carcinoma (ccRCC) makes prediction of disease progression and therapeutic response difficult. Thus, this report investigates the functional significance, mechanisms of action, and clinical utility of miR-182-5p and metastasis-associated lung adenocarcinoma transcript 1 (MALAT1/NEAT2), a long noncoding RNA (lncRNA), in the regulation of kidney cancer using human kidney cancer tissues as well as in vitro and in vivo model systems. Profiling of miR-182-5p and MALAT-1 in human renal cancer cells and clinical specimens was done by quantitative real-time PCR (qPCR). The biological significance was determined by series of in vitro and in vivo experiments. The interaction between miR-182-5p and MALAT-1 was investigated using luciferase reporter assays. In addition, the effects of miR-182-5p overexpression and MALAT-1 downregulation on cell-cycle progression were assessed in ccRCC cells. The data indicate that miR-182-5p is downregulated in ccRCC; the mechanism being CpG hypermethylation as observed from 5-Aza CdR treatment that decreased promoter methylation and expression of key methylation regulatory genes like DNMT1, DNMT3a, and DNMT3b Overexpression of miR-182-5p-inhibited cell proliferation, colony formation, apoptosis, and led to G2-M-phase cell-cycle arrest by directly targeting MALAT-1 Downregulation of MALAT-1 led to upregulation of p53, downregulation of CDC20, AURKA, drivers of the cell-cycle mitotic phase. Transient knockdown of MALAT-1 mimicked the effects of miR-182-5p overexpression. Finally, overexpression of miR-182-5p decreased tumor growth in mice, compared with controls; thus, demonstrating its antitumor effect in vivo Implications: This is the first study that offers new insight into role of miR-182-5p/MALAT-1 interaction on inhibition of ccRCC progression. Mol Cancer Res; 16(11); 1750-60. ©2018 AACR.
Collapse
Affiliation(s)
- Priyanka Kulkarni
- Department of Urology, Veterans Affairs Medical Center, San Francisco, California
- University of California San Francisco, San Francisco, California
| | - Pritha Dasgupta
- Department of Urology, Veterans Affairs Medical Center, San Francisco, California
- University of California San Francisco, San Francisco, California
| | - Nadeem S Bhat
- Department of Surgery, Miller School of Medicine, University of Miami, Miami, Florida
| | - Varahram Shahryari
- Department of Urology, Veterans Affairs Medical Center, San Francisco, California
- University of California San Francisco, San Francisco, California
| | - Marisa Shiina
- Department of Urology, Veterans Affairs Medical Center, San Francisco, California
- University of California San Francisco, San Francisco, California
| | - Yutaka Hashimoto
- Department of Urology, Veterans Affairs Medical Center, San Francisco, California
- University of California San Francisco, San Francisco, California
| | - Shahana Majid
- Department of Urology, Veterans Affairs Medical Center, San Francisco, California
- University of California San Francisco, San Francisco, California
| | - Guoren Deng
- Department of Urology, Veterans Affairs Medical Center, San Francisco, California
- University of California San Francisco, San Francisco, California
| | - Sharanjot Saini
- Department of Urology, Veterans Affairs Medical Center, San Francisco, California
- University of California San Francisco, San Francisco, California
| | - Z Laura Tabatabai
- Department of Urology, Veterans Affairs Medical Center, San Francisco, California
- University of California San Francisco, San Francisco, California
| | - Soichiro Yamamura
- Department of Urology, Veterans Affairs Medical Center, San Francisco, California
- University of California San Francisco, San Francisco, California
| | - Yuichiro Tanaka
- Department of Urology, Veterans Affairs Medical Center, San Francisco, California
- University of California San Francisco, San Francisco, California
| | - Rajvir Dahiya
- Department of Urology, Veterans Affairs Medical Center, San Francisco, California.
- University of California San Francisco, San Francisco, California
| |
Collapse
|
265
|
Xing Q, Huang Y, Wu Y, Ma L, Cai B. Integrated analysis of differentially expressed profiles and construction of a competing endogenous long non-coding RNA network in renal cell carcinoma. PeerJ 2018; 6:e5124. [PMID: 30038853 PMCID: PMC6054097 DOI: 10.7717/peerj.5124] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2018] [Accepted: 06/01/2018] [Indexed: 12/28/2022] Open
Abstract
Background Long non-coding RNAs (lncRNAs) play crucial roles in the initiation and progression of renal cell carcinoma (RCC) by competing in binding to miRNAs, and related competitive endogenous RNA (ceRNA) networks have been constructed in several cancers. However, the coexpression network has been poorly explored in RCC. Methods We collected RCC RNA expression profile data and relevant clinical features from The Cancer Genome Atlas (TCGA). A cluster analysis was explored to show different lncRNA expression patterns. Gene ontology (GO), Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analyses and gene set enrichment analysis (GSEA) were performed to analyze the functions of the intersecting mRNAs. Targetscan and miRanda bioinformatics algorithms were used to predict potential relationships among RNAs. Univariate Cox proportional hazards regression was conducted to determine the RNA expression levels and survival times. Results Bioinformatics analysis revealed that the expression profiles of hundreds of aberrantly expressed lncRNAs, miRNAs, and mRNAs were significantly changed between different stages of tumors and non-tumor groups. By combining the data predicted by databases with intersection RNAs, a ceRNA network consisting of 106 lncRNAs, 26 miRNAs and 69 mRNAs was established. Additionally, a protein interaction network revealed the main hub nodes (VEGFA, NTRK2, DLG2, E2F2, MYB and RUNX1). Furthermore, 63 lncRNAs, four miRNAs and 31 mRNAs were significantly associated with overall survival. Conclusion Our results identified cancer-specific lncRNAs and constructed a ceRNA network for RCC. A survival analysis related to the RNAs revealed candidate biomarkers for further study in RCC.
Collapse
Affiliation(s)
- Qianwei Xing
- Department of Urology, Affiliated Hospital of Nantong University, Nantong, Jiangsu, China
| | - Yeqing Huang
- Department of Urology, Affiliated Hospital of Nantong University, Nantong, Jiangsu, China
| | - You Wu
- Department of Urology, Affiliated Hospital of Nantong University, Nantong, Jiangsu, China
| | - Limin Ma
- Department of Urology, Affiliated Hospital of Nantong University, Nantong, Jiangsu, China
| | - Bo Cai
- Department of Urology, Affiliated Hospital of Nantong University, Nantong, Jiangsu, China
| |
Collapse
|
266
|
Peng F, Shi X, Meng Y, Dong B, Xu G, Hou T, Shi Y, Liu T. Long non-coding RNA HOTTIP is upregulated in renal cell carcinoma and regulates cell growth and apoptosis by epigenetically silencing of LATS2. Biomed Pharmacother 2018; 105:1133-1140. [PMID: 30021349 DOI: 10.1016/j.biopha.2018.06.081] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2018] [Revised: 06/14/2018] [Accepted: 06/14/2018] [Indexed: 12/31/2022] Open
Abstract
Renal cell carcinoma (RCC) is one of the most aggressive malignancies with increasing incidence worldwide and is characterized by dismal prognosis owing to a lack of early detection and prognostic biomarkers for this fatal disease. Accumulating studies demonstrated that abnormally expressed long non-coding RNAs (lncRNAs) are involved in tumorigenesis and progression. Specifically, HOTTIP is upregulated and exerts oncogenic properties in some cancers. However, its clinical significance, biological functions and molecular mechanisms in RCC have not been studied. In the current study, RT-qPCR was performed to quantify the relative expression of HOTTIP in RCC tissues and cells. Additionally, we explored its clinical value using Fisher's exact test. Moreover, cell growth and apoptosis altered by HOTTIP was evaluated in vitro and in vivo. Mechanistically, RNA immunoprecipitation (RIP) and chromatin immunoprecipitation (ChIP) analysis was used to determine its molecular mechanism in cell growth and apoptosis. As a result, upregulated HOTTIP is closely associated with unfavorable phenotypes in RCC patients. The mechanistic investigations showed that HOTTIP could bind to enhancer of zeste homolog 2 (EZH2) and lysine specific demethylase 1 (LSD1), thereby repressing LATS2 expression. Collectively, our study illustrates how HOTTIP plays an oncogenic role in RCC and may offer a potential therapeutic target for treating this fatal disease.
Collapse
Affiliation(s)
- Feifei Peng
- Department of Urology Surgery, Second Affiliated Hospital of Qiqihar Medical University, Heilongjiang Province, 161000, China
| | - Xiaoli Shi
- Department of Urology Surgery, Second Affiliated Hospital of Qiqihar Medical University, Heilongjiang Province, 161000, China
| | - Yin Meng
- Department of Urology Surgery, Second Affiliated Hospital of Qiqihar Medical University, Heilongjiang Province, 161000, China
| | - Bo Dong
- Department of Urology Surgery, Second Affiliated Hospital of Qiqihar Medical University, Heilongjiang Province, 161000, China
| | - Guangchi Xu
- Department of Urology Surgery, Second Affiliated Hospital of Qiqihar Medical University, Heilongjiang Province, 161000, China
| | - Tingting Hou
- Department of Urology Surgery, Second Affiliated Hospital of Qiqihar Medical University, Heilongjiang Province, 161000, China
| | - Yang Shi
- Department of Urology Surgery, Second Affiliated Hospital of Qiqihar Medical University, Heilongjiang Province, 161000, China
| | - Tao Liu
- Department of Urology Surgery, Second Affiliated Hospital of Qiqihar Medical University, Heilongjiang Province, 161000, China.
| |
Collapse
|
267
|
A Novel Model for Predicting Associations between Diseases and LncRNA-miRNA Pairs Based on a Newly Constructed Bipartite Network. COMPUTATIONAL AND MATHEMATICAL METHODS IN MEDICINE 2018; 2018:6789089. [PMID: 29853986 PMCID: PMC5960578 DOI: 10.1155/2018/6789089] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/21/2017] [Revised: 03/16/2018] [Accepted: 03/26/2018] [Indexed: 11/18/2022]
Abstract
Motivation Increasing studies have demonstrated that many human complex diseases are associated with not only microRNAs, but also long-noncoding RNAs (lncRNAs). LncRNAs and microRNA play significant roles in various biological processes. Therefore, developing effective computational models for predicting novel associations between diseases and lncRNA-miRNA pairs (LMPairs) will be beneficial to not only the understanding of disease mechanisms at lncRNA-miRNA level and the detection of disease biomarkers for disease diagnosis, treatment, prognosis, and prevention, but also the understanding of interactions between diseases and LMPairs at disease level. Results It is well known that genes with similar functions are often associated with similar diseases. In this article, a novel model named PADLMP for predicting associations between diseases and LMPairs is proposed. In this model, a Disease-LncRNA-miRNA (DLM) tripartite network was designed firstly by integrating the lncRNA-disease association network and miRNA-disease association network; then we constructed the disease-LMPairs bipartite association network based on the DLM network and lncRNA-miRNA association network; finally, we predicted potential associations between diseases and LMPairs based on the newly constructed disease-LMPair network. Simulation results show that PADLMP can achieve AUCs of 0.9318, 0.9090 ± 0.0264, and 0.8950 ± 0.0027 in the LOOCV, 2-fold, and 5-fold cross validation framework, respectively, which demonstrate the reliable prediction performance of PADLMP.
Collapse
|
268
|
Lin Q, Guan W, Ren W, Zhang L, Zhang J, Xu G. MALAT1 affects ovarian cancer cell behavior and patient survival. Oncol Rep 2018; 39:2644-2652. [PMID: 29693187 PMCID: PMC5983936 DOI: 10.3892/or.2018.6384] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2018] [Accepted: 04/12/2018] [Indexed: 12/16/2022] Open
Abstract
Epithelial ovarian cancer (EOC) is one of the most lethal malignancies of the female reproductive organs. Increasing evidence has revealed that long non‑coding RNAs (lncRNAs) participate in tumorigenesis. Metastasis associated lung adenocarcinoma transcript 1 (MALAT1) is an lncRNA and plays a role in various types of tumors. However, the function of MALAT1 on cellular behavior in EOC remains unclear. The current study explored the expression of MALAT1 in ovarian cancer tissues and in EOC cell lines. Quantitative RT‑PCR analysis revealed that the expression of MALAT1 was higher in human ovarian malignant tumor tissues and EOC cells than in normal ovarian tissues and non‑tumorous human ovarian surface epithelial cells, respectively. By analyzing the online database Kaplan‑Meier Plotter, MALAT1 was identified to be correlated with the overall survival (OS) and progression‑free survival (PFS) of patients with ovarian cancer. Furthermore, knockdown of MALAT1 by small interfering RNA (siRNA) significantly decreased EOC cell viability, migration, and invasion. Finally, dual‑luciferase reporter assays demonstrated that MALAT1 interacted with miR‑143‑3p, a miRNA that plays a role in EOC as demonstrated in our previous study. Inhibition of MALAT1 resulted in an increase of miR‑143‑3p expression, leading to a decrease of CMPK protein expression. In conclusion, our results indicated that MALAT1 was overexpressed in EOC. Silencing of MALAT1 decreased EOC cell viability and inhibited EOC cell migration and invasion. These data revealed that MALAT1 may serve as a new therapeutic target of human EOC.
Collapse
Affiliation(s)
- Qunbo Lin
- Center Laboratory, Jinshan Hospital, Fudan University, Shanghai 201508, P.R. China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, P.R. China
| | - Wencai Guan
- Center Laboratory, Jinshan Hospital, Fudan University, Shanghai 201508, P.R. China
| | - Weimin Ren
- Center Laboratory, Jinshan Hospital, Fudan University, Shanghai 201508, P.R. China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, P.R. China
| | - Lingyun Zhang
- Center Laboratory, Jinshan Hospital, Fudan University, Shanghai 201508, P.R. China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, P.R. China
| | - Jinguo Zhang
- Center Laboratory, Jinshan Hospital, Fudan University, Shanghai 201508, P.R. China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, P.R. China
| | - Guoxiong Xu
- Center Laboratory, Jinshan Hospital, Fudan University, Shanghai 201508, P.R. China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, P.R. China
| |
Collapse
|
269
|
Toraih EA, Ellawindy A, Fala SY, Al Ageeli E, Gouda NS, Fawzy MS, Hosny S. Oncogenic long noncoding RNA MALAT1 and HCV-related hepatocellular carcinoma. Biomed Pharmacother 2018; 102:653-669. [PMID: 29604585 DOI: 10.1016/j.biopha.2018.03.105] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2017] [Revised: 03/11/2018] [Accepted: 03/17/2018] [Indexed: 02/07/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is the second leading cause of cancer-related mortality worldwide. The oncogenic function of the long non-coding RNA; metastasis-associated lung adenocarcinoma transcript 1 (MALAT1) in HCC remains unclear. We aimed to evaluate MALAT1 serum expression profile in HCC and explore its relation to the clinicopathological features. Quantitative Real Time-Polymerase Chain Reaction was applied in 70 cohorts (30 HCC, 20 HCV, 20 controls). Further meta-analysis of clinical studies and in vitro validated experiments was employed. Serum MALAT1 showed area under the curve of 0.79 and 0.70 to distinguish patients with cancer from normal and cirrhotic individuals at fold change of 1.0 and 1.26, respectively. Expression level was significantly higher in males (P <0.001) and patients with massive ascites (P = 0.005). Correlation analysis showed positive correlation of MALAT1 with total bilirubin (r = 0.456, P <0.001) and AST (r = 0.280, P = 0.019), and negative correlation with the hemoglobin level (r = 0.312, P = 0.009). Meta-analysis showed that the over-expressed MALAT1 was linked to tumor number [Cohen's d = 0.450, 95% CI (0.21 to 0.68)], clinical stage [Cohen's d = 0.048, 95% CI (-0.83 to 0.74)], and AFP level [Cohen's d = 0.354, 95% CI (0.1 to 0.57)]. In silico data analysis and systematic review confirmed MALAT1 oncogenic function in cancer development and progression. In conclusion, circulatory MALAT1 might represent a putative non-invasive prognostic biomarker indicating worse liver failure score in HCV-related HCC patients with traditional markers. Large-scale verification is warranted in future studies.
Collapse
MESH Headings
- Adult
- Aged
- Aged, 80 and over
- Carcinogenesis/genetics
- Carcinogenesis/pathology
- Carcinoma, Hepatocellular/blood
- Carcinoma, Hepatocellular/genetics
- Carcinoma, Hepatocellular/virology
- Case-Control Studies
- Computer Simulation
- Demography
- Female
- Gene Expression Regulation, Neoplastic
- Hepacivirus/physiology
- Humans
- Liver Cirrhosis/diagnosis
- Liver Cirrhosis/genetics
- Liver Neoplasms/blood
- Liver Neoplasms/genetics
- Liver Neoplasms/pathology
- Liver Neoplasms/virology
- Male
- Middle Aged
- Prognosis
- Publication Bias
- RNA, Long Noncoding/blood
- RNA, Long Noncoding/genetics
- RNA, Long Noncoding/metabolism
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- Transcriptome/genetics
- alpha-Fetoproteins/metabolism
Collapse
Affiliation(s)
- Eman A Toraih
- Genetics Unit, Department of Histology and Cell Biology, Faculty of Medicine, Suez Canal University, Ismailia, Egypt; Center of Excellence of Molecular and Cellular Medicine, Suez Canal University, Ismailia, Egypt.
| | - Alia Ellawindy
- Genetics Unit, Department of Histology and Cell Biology, Faculty of Medicine, Suez Canal University, Ismailia, Egypt
| | - Salma Y Fala
- Faculty of Medicine, Suez Canal University, Ismailia, Egypt
| | - Essam Al Ageeli
- Department of Clinical Biochemistry (Medical Genetics), Faculty of Medicine, Jazan University, Jazan, P.O. 45142, Saudi Arabia
| | - Nawal S Gouda
- Department of Medical Microbiology and Immunology, Faculty of Medicine, Mansoura, Mansoura University, Egypt
| | - Manal S Fawzy
- Department of Medical Biochemistry, Faculty of Medicine, Suez Canal University, Ismailia, P.O. 41522, Egypt; Department of Biochemistry, Faculty of Medicine, Northern Border University, Arar, Saudi Arabia.
| | - Somaya Hosny
- Center of Excellence of Molecular and Cellular Medicine, Suez Canal University, Ismailia, Egypt; Department of Histology and Cell Biology, Faculty of Medicine, Suez Canal University, Ismailia, Egypt
| |
Collapse
|
270
|
Down-regulation of Long Noncoding RNA MALAT1 Protects Hippocampal Neurons Against Excessive Autophagy and Apoptosis via the PI3K/Akt Signaling Pathway in Rats with Epilepsy. J Mol Neurosci 2018; 65:234-245. [DOI: 10.1007/s12031-018-1093-3] [Citation(s) in RCA: 66] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2018] [Accepted: 05/21/2018] [Indexed: 12/13/2022]
|
271
|
Lu J, Lin Y, Li F, Ye H, Zhou R, Jin Y, Li B, Xiong X, Cheng N. MiR-205 suppresses tumor growth, invasion, and epithelial-mesenchymal transition by targeting SEMA4C in hepatocellular carcinoma. FASEB J 2018; 32:fj201800113R. [PMID: 29799789 DOI: 10.1096/fj.201800113r] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Growing evidence indicates that microRNAs are involved in tumorigenesis and progression of hepatocellular carcinoma (HCC). However, the functional mechanisms of miR-205 in HCC remain largely unknown. Here, we demonstrate that miR-205 expression was significantly down-regulated in HCC tissues and cell lines and was correlated with metastatic pathologic features and shorter disease-free and overall survival. Overexpression of miR-205 dramatically inhibited HCC cell proliferation, apoptosis, migration, invasion, epithelial-mesenchymal transition (EMT) in vitro, and tumor growth in vivo. We subsequently identified semaphorin 4C (SEMA4C) as a novel target of miR-205. Furthermore, high expression levels of SEMA4C were frequently found in HCC tissues and were associated with poor prognosis. Ectopic expression of SEMA4C restored the suppressive effect of overexpressed miR-205 on migration, invasion, and EMT. Taken together, our findings provide new insight into the critical role of miR-205 in regulating tumor growth, invasion, and EMT of HCC, suggesting miR-205 may serve as a promising therapeutic target and novel prognostic indicator for patients with HCC.-Lu, J., Lin, Y., Li, F., Ye, H., Zhou, R., Jin, Y., Li, B., Xiong, X., Cheng, N. MiR-205 suppresses tumor growth, invasion and epithelial-mesenchymal transition by targeting SEMA4C in hepatocellular carcinoma.
Collapse
Affiliation(s)
- Jiong Lu
- Department of Bile Duct Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Yixin Lin
- Department of Bile Duct Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Fuyu Li
- Department of Bile Duct Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Hui Ye
- Department of Bile Duct Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Rongxing Zhou
- Department of Bile Duct Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Yanwen Jin
- Department of Bile Duct Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Bei Li
- Department of Bile Duct Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Xianze Xiong
- Department of Bile Duct Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Nansheng Cheng
- Department of Bile Duct Surgery, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
272
|
Upregulation of the long noncoding RNA FOXD2-AS1 promotes carcinogenesis by epigenetically silencing EphB3 through EZH2 and LSD1, and predicts poor prognosis in gastric cancer. Oncogene 2018; 37:5020-5036. [PMID: 29789713 DOI: 10.1038/s41388-018-0308-y] [Citation(s) in RCA: 103] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2017] [Revised: 03/27/2018] [Accepted: 04/18/2018] [Indexed: 01/21/2023]
Abstract
Accumulating data indicate that long noncoding RNAs (lncRNAs) serve as important modulators in biological processes and are dysregulated in diverse tumors. The function of FOXD2-AS1 in gastric cancer (GC) progression and related biological mechanisms remain undefined. A comprehensive analysis identified that FOXD2-AS1 enrichment was upregulated markedly in GC and positively correlated with a large tumor size, a later pathologic stage, and a poor prognosis. Gene-set enrichment analysis (GSEA) in GEO datasets uncovered that cell cycle and DNA replication associated genes were enriched in patients with high FOXD2-AS1 expression. Loss of FOXD2-AS1 function inhibited cell growth via inhibiting the cell cycle in GC, whereas upregulation of FOXD2-AS1 expression promoted cancer progression. The enhancer of zeste homolog 2 (EZH2) and lysine (K)-specific demethylase 1A (LSD1) proteins were found to serve as binding partners of FOXD2-AS1 and mediators of FOXD2-AS1 function. Mechanically, FOXD2-AS1 promoted GC tumorigenesis partly through EZH2 and LSD1 mediated EphB3 downregulation. The present results revealed that FOXD2-AS1 acted as a tumor inducer in GC partly through EphB3 inhibition by direct interaction with EZH2 and LSD1, and may prove to be a potential biomarker of carcinogenesis.
Collapse
|
273
|
Crentsil VC, Liu H, Sellitti DF. Comparison of exosomal microRNAs secreted by 786-O clear cell renal carcinoma cells and HK-2 proximal tubule-derived cells in culture identifies microRNA-205 as a potential biomarker of clear cell renal carcinoma. Oncol Lett 2018; 16:1285-1290. [PMID: 30061948 DOI: 10.3892/ol.2018.8751] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2018] [Accepted: 04/30/2018] [Indexed: 01/12/2023] Open
Abstract
Previous reports have indicated that the abundance of specific microRNAs (miRNA) contained within the exosome/microvesicle compartment of patient biofluids may be useful in diagnosing specific types of cancer. In the present study, the 786-O cell line, which is derived from a clear cell renal cell carcinoma (ccRCC), was used as an in vitro ccRCC tumor model and the human renal proximal tubule cell line HK-2 was used as its normal renal tissue control to investigate the similarities of exosomal content of selected ccRCC miRNA biomarkers in the supernatant with the content of those markers in the cells themselves. A PCR array identified miRNA biomarkers of solid RCC tumors (miR-210, MiR-34a, miR-155-5p and miR-150-5p) that were increased by 2-8 fold in 786-O exosomes compared with the control. These were subsequently chosen for further investigation using TaqMan RT-qPCR in addition to miR-15a and miR-205, which were selected based on prior interest as RCC biomarkers. MiR-15a, -34a, -210 and -155 levels were significantly lower in exosomes when compared with that in whole cells but did not differ between the HK-2 and 786-O cells in either the cytoplasmic, exosome or exosome-free supernatant fractions. By contrast, cytoplasmic miR-150 and miR-205 exhibited significant differences in concentration between the two cell lines. In addition, the cytoplasmic content of miR-150 and miR-205 was mirrored in the exosomal content of these miRNAs. Furthermore, the difference in exosomal miR-205 content was statistically significant. The present study indicated that measurements of the exosomal content of miR-205 and possibly miR-150, but not those of the other examined miRNAs, are proportional to their respective contents in the cells that secreted them. These findings suggest that in vitro RCC systems may be useful in identifying miRNAs with sufficiently high levels of exportation into exosomes; and with sufficiently different expression levels between tumor and normal cells to serve as ccRCC biomarkers in vivo.
Collapse
Affiliation(s)
- Victor C Crentsil
- Department of Medicine, Uniformed Services University of The Health Sciences, Bethesda, MD 20814-4799, USA.,Department of Anatomy, Physiology and Genetics, Uniformed Services University of The Health Sciences, Bethesda, MD 20814-4799, USA
| | - Hui Liu
- Department of Medicine, Uniformed Services University of The Health Sciences, Bethesda, MD 20814-4799, USA.,Department of Anatomy, Physiology and Genetics, Uniformed Services University of The Health Sciences, Bethesda, MD 20814-4799, USA
| | - Donald F Sellitti
- Department of Medicine, Uniformed Services University of The Health Sciences, Bethesda, MD 20814-4799, USA.,Department of Anatomy, Physiology and Genetics, Uniformed Services University of The Health Sciences, Bethesda, MD 20814-4799, USA
| |
Collapse
|
274
|
Zhang W, Cai X, Yu J, Lu X, Qian Q, Qian W. Exosome-mediated transfer of lncRNA RP11‑838N2.4 promotes erlotinib resistance in non-small cell lung cancer. Int J Oncol 2018; 53:527-538. [PMID: 29845246 PMCID: PMC6017264 DOI: 10.3892/ijo.2018.4412] [Citation(s) in RCA: 73] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2018] [Accepted: 05/04/2018] [Indexed: 12/31/2022] Open
Abstract
Currently, resistance to tyrosine kinase inhibitors, such as erlotinib, has become a major obstacle for improving the clinical outcome of patients with metastatic and advanced-stage non-small cell lung cancer (NSCLC). While cell behavior can be modulated by long non-coding RNAs (lncRNAs), the roles of lncRNAs within extracellular vesicles (exosomes) are largely unknown. To this end, in this study, the involvement and regulatory functions of potential lncRNAs wrapped by exosomes during the development of chemoresistance in human NSCLC were investigated. Erlotinib-resistant cell lines were established by grafting HCC827 and HCC4006 cells into mice and which were treated with erlotinib. After one treatment course, xenografted NSCLC cells were isolated and transplanted into nude mice again followed by erlotinib treatment. This process was repeated until 4th generation xenografts were isolated and confirmed to be erlotinib-resistant NSCLC cells. lncRNA microarray assays followed by RT-qPCR were then performed which identified that lncRNA RP11-838N2.4 was upregulated in erlotinib-resistant cells when compared to normal NSCLC cells. Furthermore, bioinformatics analysis and chromatin immunoprecipitation revealed that forkhead box protein O1 (FOXO1) could bind to the promoter region of lncRNA RP11-838N2.4, resulting in its silencing through the recruitment of histone deacetylase. Functional experiments demonstrated that the knockdown of lncRNA RP11-838N2.4 potently promoted erlotinib-induced cytotoxicity. Furthermore, extracellular lncRNA RP11-838N2.4 could be incorporated into exosomes and transmitted to sensitive cells, thus disseminating erlotinib resistance. Treatment-sensitive cells with exosomes containing lncRNA RP11-838N2.4 induced erlotinib resistance, while the knockdown of lncRNA RP11-838N2.4 abrogated this effect. In addition, the serum expression levels of exosomal lncRNA RP11-838N2.4 were upregulated in patients exhibiting resistance to erlotinib treatment. On the whole, exosomal lncRNA RP11-838N2.4 may serve as a therapeutic target for patients with NSCLC.
Collapse
Affiliation(s)
- Wei Zhang
- Department of Lung Disease, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong 250011, P.R. China
| | - Xinrui Cai
- Department of Traditional Chinese Medicine, Shandong Academy of Occupational Health and Occupational Medicine, Shandong Academy of Medical Sciences, Jinan, Shandong 250062, P.R. China
| | - Jie Yu
- Department of Chinese Internal Medicine, Shandong University of Traditional Chinese Medicine, Jinan, Shandong 250355, P.R. China
| | - Xuxiang Lu
- Department of Chinese Internal Medicine, Shandong University of Traditional Chinese Medicine, Jinan, Shandong 250355, P.R. China
| | - Qiuhai Qian
- Department of Endocrinology, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong 250011, P.R. China
| | - Weibin Qian
- Department of Lung Disease, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong 250011, P.R. China
| |
Collapse
|
275
|
MicroRNA and transcriptome analysis in periocular Sebaceous Gland Carcinoma. Sci Rep 2018; 8:7531. [PMID: 29760516 PMCID: PMC5951834 DOI: 10.1038/s41598-018-25900-z] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2018] [Accepted: 04/26/2018] [Indexed: 12/16/2022] Open
Abstract
Sebaceous gland carcinoma (SGC) is a rare, but life-threatening condition with a predilection for the periocular region. Eyelid SGC can be broadly categorised into two subtypes, namely either nodular or pagetoid with the latter being more aggressive and requiring radical excision to save life. We have identified key altered microRNAs (miRNA) involved in SGC shared by both subtypes, hsa-miR-34a-5p and hsa-miR-16-5p. However, their gene targets BCL2 and MYC were differentially expressed with both overexpressed in pagetoid but unchanged in nodular suggesting different modes of action of these two miRNAs on BCL/MYC expression. Hsa-miR-150p is nodular-specifically overexpressed, and its target ZEB1 was significantly downregulated in nodular SGC suggesting a tumour suppressor role. Invasive pagetoid subtype demonstrated specific overexpression of hsa-miR-205 and downregulation of hsa-miR-199a. Correspondingly, miRNA gene targets, EZH2 (by hsa-miR-205) and CD44 (by hsa-miR-199a), were both overexpressed in pagetoid SGC. CD44 has been identified as a potential cancer stem cell marker in head and neck squamous cell carcinoma and its overexpression in pagetoid cells represents a novel treatment target. Aberrant miRNAs and their gene targets have been identified in both SGC subtypes, paving the way for better molecular understanding of these tumours and identifying new treatment targets.
Collapse
|
276
|
Ren GJ, Fan XC, Liu TL, Wang SS, Zhao GH. Genome-wide analysis of differentially expressed profiles of mRNAs, lncRNAs and circRNAs during Cryptosporidium baileyi infection. BMC Genomics 2018; 19:356. [PMID: 29747577 PMCID: PMC5946474 DOI: 10.1186/s12864-018-4754-2] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2017] [Accepted: 05/02/2018] [Indexed: 01/17/2023] Open
Abstract
Background Cryptosporidium baileyi is the most common Cryptosporidium species in birds. However, effective prevention measures and treatment for C. baileyi infection were still not available. Long non-coding RNAs (lncRNAs) and circular RNAs (circRNAs) play important roles in regulating occurrence and progression of many diseases and are identified as effective biomarkers for diagnosis and prognosis of several diseases. In the present study, the expression profiles of host mRNAs, lncRNAs and circRNAs associated with C. baileyi infection were investigated for the first time. Results The tracheal tissues of experimental (C. baileyi infection) and control chickens were collected for deep RNA sequencing, and 545,479,934 clean reads were obtained. Of them, 1376 novel lncRNAs were identified, including 1161 long intergenic non-coding RNAs (lincRNAs) and 215 anti-sense lncRNAs. A total of 124 lncRNAs were found to be significantly differentially expressed between the experimental and control groups. Additionally, 14,698 mRNAs and 9085 circRNAs were identified, and significantly different expressions were observed for 1317 mRNAs and 104 circRNAs between two groups. Bioinformatic analyses of gene ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway for their targets and source genes suggested that these dysregulated genes may be involved in the interaction between the host and C. baileyi. Conclusions The present study revealed the expression profiles of mRNAs, lncRNAs and circRNAs during C. baileyi infection for the first time, and sheds lights on the roles of lncRNAs and circRNAs underlying the pathogenesis of Cryptosporidium infection. Electronic supplementary material The online version of this article (10.1186/s12864-018-4754-2) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Guan-Jing Ren
- Department of Parasitology, College of Veterinary Medicine, Northwest A&F University, Yangling, 712100, China
| | - Xian-Cheng Fan
- Department of Parasitology, College of Veterinary Medicine, Northwest A&F University, Yangling, 712100, China
| | - Ting-Li Liu
- Department of Parasitology, College of Veterinary Medicine, Northwest A&F University, Yangling, 712100, China
| | - Sha-Sha Wang
- Department of Parasitology, College of Veterinary Medicine, Northwest A&F University, Yangling, 712100, China
| | - Guang-Hui Zhao
- Department of Parasitology, College of Veterinary Medicine, Northwest A&F University, Yangling, 712100, China.
| |
Collapse
|
277
|
Shenoda BB, Tian Y, Alexander GM, Aradillas-Lopez E, Schwartzman RJ, Ajit SK. miR-34a-mediated regulation of XIST in female cells under inflammation. J Pain Res 2018; 11:935-945. [PMID: 29773953 PMCID: PMC5947841 DOI: 10.2147/jpr.s159458] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Background Evidence is overwhelming for sex differences in pain, with women representing the majority of the chronic pain patient population. There is a need to explore novel avenues to elucidate this sex bias in the development of chronic inflammatory pain conditions. Complex regional pain syndrome (CRPS) is a chronic neuropathic pain disorder, and the incidence of CRPS is greater in women than in men by ~4:1. Since neurogenic inflammation is a key feature of CRPS, dysregulation of inflammatory responses can be a factor in predisposing women to chronic pain. Methods Our studies investigating alterations in circulating microRNAs (miRNAs) in whole blood from female CRPS patients showed significant differential expression of miRNAs between responders and poor responders to ketamine treatment. Several of these miRNAs are predicted to target the long noncoding RNA, X-inactive-specific transcript (XIST). XIST mediates X-chromosome inactivation and is essential for equalizing the expression of X-linked genes between females and males. Based on the well-established role in inflammatory process, we focused on miR-34a, one of the miRNAs predicted to target XIST, and downregulated in CRPS patients responding poorly to ketamine. Results Our in vitro and in vivo models of acute inflammation and data from patients with CRPS showed that miR-34a can regulate XIST under inflammation directly, and through pro-inflammatory transcription factor Yin-Yang 1 (YY1). XIST was significantly upregulated in a subset of CRPS patients responding poorly to ketamine. Conclusion Since dysregulation of XIST can result in genes escaping inactivation or reactivation in female cells, further investigations on the role of XIST in the predominance of chronic inflammatory and pain disorders in women is warranted.
Collapse
Affiliation(s)
- Botros B Shenoda
- Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, PA, USA
| | - Yuzhen Tian
- Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, PA, USA
| | | | - Enrique Aradillas-Lopez
- Neurology, Drexel University College of Medicine, Philadelphia, PA, USA.,Vincera Institute, Philadelphia, PA, USA
| | | | - Seena K Ajit
- Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, PA, USA
| |
Collapse
|
278
|
Amodio N, Raimondi L, Juli G, Stamato MA, Caracciolo D, Tagliaferri P, Tassone P. MALAT1: a druggable long non-coding RNA for targeted anti-cancer approaches. J Hematol Oncol 2018; 11:63. [PMID: 29739426 PMCID: PMC5941496 DOI: 10.1186/s13045-018-0606-4] [Citation(s) in RCA: 245] [Impact Index Per Article: 35.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2018] [Accepted: 04/26/2018] [Indexed: 02/07/2023] Open
Abstract
The deeper understanding of non-coding RNAs has recently changed the dogma of molecular biology assuming protein-coding genes as unique functional biological effectors, while non-coding genes as junk material of doubtful significance. In the last decade, an exciting boom of experimental research has brought to light the pivotal biological functions of long non-coding RNAs (lncRNAs), representing more than the half of the whole non-coding transcriptome, along with their dysregulation in many diseases, including cancer.In this review, we summarize the emerging insights on lncRNA expression and functional role in cancer, focusing on the evolutionary conserved and abundantly expressed metastasis-associated lung adenocarcinoma transcript 1 (MALAT1) that currently represents the best characterized lncRNA. Altogether, literature data indicate aberrant expression and dysregulated activity of MALAT1 in human malignancies and envision MALAT1 targeting as a novel treatment strategy against cancer.
Collapse
Affiliation(s)
- Nicola Amodio
- Department of Experimental and Clinical Medicine, Magna Graecia University, Viale Europa, 88100, Catanzaro, Italy.
| | - Lavinia Raimondi
- IRCSS Rizzoli Orthopedic Institute, Bologna, Italy
- Innovative Technology Platforms for Tissue Engineering, Theranostic and Oncology, Rizzoli Orthopedic Institute, Palermo, Italy
| | - Giada Juli
- Department of Experimental and Clinical Medicine, Magna Graecia University, Viale Europa, 88100, Catanzaro, Italy
| | - Maria Angelica Stamato
- Department of Experimental and Clinical Medicine, Magna Graecia University, Viale Europa, 88100, Catanzaro, Italy
| | - Daniele Caracciolo
- Department of Experimental and Clinical Medicine, Magna Graecia University, Viale Europa, 88100, Catanzaro, Italy
| | - Pierosandro Tagliaferri
- Department of Experimental and Clinical Medicine, Magna Graecia University, Viale Europa, 88100, Catanzaro, Italy
| | - Pierfrancesco Tassone
- Department of Experimental and Clinical Medicine, Magna Graecia University, Viale Europa, 88100, Catanzaro, Italy.
| |
Collapse
|
279
|
Liu S, Yan G, Zhang J, Yu L. Knockdown of Long Noncoding RNA (lncRNA) Metastasis-Associated Lung Adenocarcinoma Transcript 1 (MALAT1) Inhibits Proliferation, Migration, and Invasion and Promotes Apoptosis by Targeting miR-124 in Retinoblastoma. Oncol Res 2018; 26:581-591. [PMID: 28550678 PMCID: PMC7844671 DOI: 10.3727/096504017x14953948675403] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Evidence suggests that the long noncoding RNA (lncRNA) metastasis-associated lung adenocarcinoma transcript 1 (MALAT1) is upregulated in cancer tissues, and its elevated expression is associated with hyperproliferation. However, the underlying mechanisms regarding the role of MALAT1 in retinoblastoma (RB) remain unclear. This study aimed to explore the functional role of MALAT1 in RB by targeting miR-124. The results showed that the expression of MALAT1 was significantly higher in the Y79 cell line than in the ARPE-19 cell line (p < 0.01). Moreover, MALAT1 silence inhibited cell viability, migration, and invasion and promoted apoptosis in Y79 cells (p < 0.05, p < 0.01, or p < 0.001). miR-124 was upregulated by MALAT1 silence and hence was identified as a target of MALAT1 (p < 0.05 or p < 0.001). In addition, miR-124 suppression inhibited cell apoptosis and remarkably abolished the inhibitory effects of MALAT1 silence on cell viability, migration, and invasion (p < 0.05, p < 0.01, or p < 0.001). In addition, Slug was a target of miR-124 and regulated cell viability, migration, invasion, and apoptosis in Y79 cells (p < 0.05, p < 0.01, or p < 0.001). Further, Slug silence abolished miR-124 suppression-induced inactivation of the ERK/MAPK and Wnt/β-catenin pathways. Taken together, our data highlight the pivotal role of MALAT1 in RB. Moreover, the present study elucidated the MALAT1-miR-124-ERK/MAPK and Wnt/β-catenin signaling pathways in RB, which might provide a new approach for the treatment of RB.
Collapse
Affiliation(s)
- Shujun Liu
- *Department of Ophthalmology, Yantai Yuhuangding Hospital Affiliated to Qingdao University, Yantai, Shandong, P.R. China
| | - Guigang Yan
- *Department of Ophthalmology, Yantai Yuhuangding Hospital Affiliated to Qingdao University, Yantai, Shandong, P.R. China
| | - Junfu Zhang
- †Department of Ophthalmology, Weifang People’s Hospital, Weifang, Shandong, P.R. China
| | - Lianzhi Yu
- ‡Department of Physical Examination, Yantai Yuhuangding Hospital Affiliated to Qingdao University, Yantai, Shandong, P.R. China
| |
Collapse
|
280
|
Du Y, Kong C, Zhu Y, Yu M, Li Z, Bi J, Li Z, Liu X, Zhang Z, Yu X. Knockdown of SNHG15 suppresses renal cell carcinoma proliferation and EMT by regulating the NF-κB signaling pathway. Int J Oncol 2018; 53:384-394. [PMID: 29750422 DOI: 10.3892/ijo.2018.4395] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2017] [Accepted: 04/12/2018] [Indexed: 11/05/2022] Open
Abstract
Aberrant expression of long noncoding RNAs (lncRNAs) is associated with cancer tumorigenesis and progression. It has been suggested that lncRNAs may be potential clinical diagnostic and prognostic biomarkers, and therapeutic targets. In the present study, the expression levels of small nucleolar RNA host gene 15 (SNHG15) were significantly upregulated in renal cell carcinoma (RCC) tissues and cell lines compared with in adjacent tissues and a proximal tubule epithelial cell line, as determined by reverse transcription‑quantitative polymerase chain reaction. Subsequently, knockdown of SNHG15 expression with small interfering RNA inhibited RCC proliferation, invasion and migration, was determined by western blotting and Transwell assays. Furthermore, the present study suggested that SNHG15 may be involved in the nuclear factor‑κB signaling pathway, induce the epithelial‑mesenchymal transition process, and promote RCC invasion and migration.
Collapse
Affiliation(s)
- Yang Du
- Department of Urology, The First Hospital of China Medical University, Shenyang, Liaoning 110001, P.R. China
| | - Chuize Kong
- Department of Urology, The First Hospital of China Medical University, Shenyang, Liaoning 110001, P.R. China
| | - Yuyan Zhu
- Department of Urology, The First Hospital of China Medical University, Shenyang, Liaoning 110001, P.R. China
| | - Meng Yu
- Department of Reproductive Biology and Transgenic Animal China Medical University, Shenyang, Liaoning 110003, P.R. China
| | - Zeliang Li
- Department of Urology, The First Hospital of China Medical University, Shenyang, Liaoning 110001, P.R. China
| | - Jianbin Bi
- Department of Urology, The First Hospital of China Medical University, Shenyang, Liaoning 110001, P.R. China
| | - Zhenhua Li
- Department of Urology, The First Hospital of China Medical University, Shenyang, Liaoning 110001, P.R. China
| | - Xiankui Liu
- Department of Urology, The First Hospital of China Medical University, Shenyang, Liaoning 110001, P.R. China
| | - Zhe Zhang
- Department of Urology, The First Hospital of China Medical University, Shenyang, Liaoning 110001, P.R. China
| | - Xiuyue Yu
- Department of Urology, The First Hospital of China Medical University, Shenyang, Liaoning 110001, P.R. China
| |
Collapse
|
281
|
Zhao M, Wang S, Li Q, Ji Q, Guo P, Liu X. MALAT1: A long non-coding RNA highly associated with human cancers. Oncol Lett 2018; 16:19-26. [PMID: 29928382 PMCID: PMC6006327 DOI: 10.3892/ol.2018.8613] [Citation(s) in RCA: 64] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2015] [Accepted: 03/21/2017] [Indexed: 12/18/2022] Open
Abstract
Metastasis-associated lung adenocarcinoma transcript 1 (MALAT1), a well-known lncRNA associated with numerous diseases, particularly cancer, has received increased attention. The expression of MALAT1 was determined to be upregulated in numerous types of tumors and MALAT1 exhibited effects on tumor cell proliferation, migration, invasion and apoptosis. The abnormal expression of MALAT1 was identified in almost in every organ of the digestive system. MALAT1 performed an important role in the pathological alterations of organs that are associated with sex hormones and several reproductive system cancers. MALAT1 participates in molecular pathways. In the clinical application of MALAT1, MALAT1 was considered as a potential biomarker for the diagnosis and prediction of cancers, and may also serve as therapeutic target for treatment of specific tumors. This review summarizes the abnormal expression of MALAT1 in cancer, its significant effect on the primary features of cancer, as well as the underlying molecular mechanisms of MALAT1 in various cancers. According to studies on MALAT1, we introduce the upstream and downstream substances associated with the function of MALAT1. These reviewed studies promote the clinical application of MALAT1 in the aspect of diagnosis and treatment of different cancers, and may help point out new study directions for MALAT1.
Collapse
Affiliation(s)
- Miaomiao Zhao
- Department of Traditional Chinese Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, P.R. China.,Department of Oncology, Zhengzhou Traditional Chinese Medicine Hospital, Zhengzhou 450007, P.R. China
| | - Songpo Wang
- Department of Traditional Chinese Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, P.R. China
| | - Qi Li
- Department of Oncology, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, P.R. China
| | - Qing Ji
- Department of Oncology, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, P.R. China
| | - Piaoting Guo
- Department of Traditional Chinese Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, P.R. China
| | - Xiaowei Liu
- Department of Traditional Chinese Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, P.R. China
| |
Collapse
|
282
|
Li XJ. Long non-coding RNA nuclear paraspeckle assembly transcript 1 inhibits the apoptosis of retina Müller cells after diabetic retinopathy through regulating miR-497/brain-derived neurotrophic factor axis. Diab Vasc Dis Res 2018; 15:204-213. [PMID: 29383970 DOI: 10.1177/1479164117749382] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
BACKGROUND The role of long non-coding RNA in diabetic retinopathy, a serious complication of diabetes mellitus, has attracted increasing attention in recent years. The purpose of this study was to explore whether long non-coding RNA nuclear paraspeckle assembly transcript 1 was involved in the context of diabetic retinopathy and its underlying mechanisms. RESULTS Our results revealed that nuclear paraspeckle assembly transcript 1 was significantly downregulated in the retina of diabetes mellitus rats. Meanwhile, miR-497 was significantly increased in diabetes mellitus rats' retina and high glucose-treated Müller cells, but brain-derived neurotrophic factor was increased. We also found that high glucose-induced apoptosis of Müller cells was accompanied by the significant downregulation of nuclear paraspeckle assembly transcript 1 in vitro. Further study demonstrated that high glucose-promoted Müller cells apoptosis through downregulating nuclear paraspeckle assembly transcript 1 and downregulated nuclear paraspeckle assembly transcript 1 mediated this effect via negative regulating miR-497. Moreover, brain-derived neurotrophic factor was negatively regulated by miR-497 and associated with the apoptosis of Müller cells under high glucose. CONCLUSION Our results suggested that under diabetic conditions, downregulated nuclear paraspeckle assembly transcript 1 decreased the expression of brain-derived neurotrophic factor through elevating miR-497, thereby promoting Müller cells apoptosis and aggravating diabetic retinopathy.
Collapse
Affiliation(s)
- Xiu-Juan Li
- Department of Ophthalmology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
283
|
Liu Q, Li Y, Lv W, Zhang G, Tian X, Li X, Cheng H, Zhu C. UCA1 promotes cell proliferation and invasion and inhibits apoptosis through regulation of the miR129-SOX4 pathway in renal cell carcinoma. Onco Targets Ther 2018; 11:2475-2487. [PMID: 29760557 PMCID: PMC5937506 DOI: 10.2147/ott.s160192] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Background Renal cell carcinoma (RCC) is the most common cancer in kidney malignancies. UCA1 has been identified as an oncogenic lncRNA in multiple cancers, including RCC. However, the underlying molecular mechanism of UCA1 involved in RCC progression is far from being addressed. Methods Reverse-transcription quantitative polymerase chain reaction (RT-qPCR) assays were used to measure expressions of UCA1, miR129, and SOX4 mRNA. Western blot assays were employed to detect SOX4 protein expression. Cell proliferation, invasion, and apoptosis were assessed by CCK-8, Matrigel invasion, and annexin–fluorescein isothiocyanate (FITC) apoptosis-detection assays, respectively. The interaction between UCA1 and miR129 was demonstrated by luciferase, RNA pull-down, and RNA-immunoprecipitation (RIP) assays. Luciferase assays were also used to explore whether UCA1 was able to act as a molecular sponge of miR129 to affect the interplay of miR129 and SOX4. Results UCA1 expression was upregulated in RCC tissue and cells, and higher UCA1 expression was associated with advanced pathogenic status and poor prognosis of RCC patients. UCA1 knockdown suppressed proliferation and invasion and induced apoptosis in RCC cells. UCA1 inhibited miR129 expression by direct interaction in RCC cells. miR129 overexpression inhibited cell proliferation and invasion and promoted apoptosis. Moreover, miR129 downregulation abrogated UCA1 knockdown-mediated antiproliferation, anti-invasion, and proapoptosis effects in RCC cells. Furthermore, UCA1 acted as a ceRNA of miR129 to enhance target-gene SOX4 expression in RCC cells. Conclusion UCA1 promoted cell proliferation and invasion and inhibited apoptosis by regulating SOX4 via miR129 in RCC, offering a promising therapeutic target and prognosis marker for RCC patients.
Collapse
Affiliation(s)
- Qian Liu
- Department of Urology, Huaihe Hospital of Henan University, Kaifeng, Henan
| | - Yang Li
- Department of Urology, Huaihe Hospital of Henan University, Kaifeng, Henan
| | - Weiling Lv
- Department of Pharmacy, Huaihe Hospital of Henan University, Kaifeng, Henan, China
| | - Guangwei Zhang
- Department of Urology, Huaihe Hospital of Henan University, Kaifeng, Henan
| | - Xin Tian
- Department of Urology, Huaihe Hospital of Henan University, Kaifeng, Henan
| | - Xiaodong Li
- Department of Urology, Huaihe Hospital of Henan University, Kaifeng, Henan
| | - Hepeng Cheng
- Department of Urology, Huaihe Hospital of Henan University, Kaifeng, Henan
| | - Chaoyang Zhu
- Department of Urology, Huaihe Hospital of Henan University, Kaifeng, Henan
| |
Collapse
|
284
|
Singh AS, Heery R, Gray SG. In Silico and In Vitro Analyses of LncRNAs as Potential Regulators in the Transition from the Epithelioid to Sarcomatoid Histotype of Malignant Pleural Mesothelioma (MPM). Int J Mol Sci 2018; 19:ijms19051297. [PMID: 29701689 PMCID: PMC5983793 DOI: 10.3390/ijms19051297] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2018] [Revised: 04/23/2018] [Accepted: 04/24/2018] [Indexed: 02/06/2023] Open
Abstract
Malignant pleural mesothelioma (MPM) is a rare malignancy, with extremely poor survival rates. At present, treatment options are limited, with no second line chemotherapy for those who fail first line therapy. Extensive efforts are ongoing in a bid to characterise the underlying molecular mechanisms of mesothelioma. Recent research has determined that between 70–90% of our genome is transcribed. As only 2% of our genome is protein coding, the roles of the remaining proportion of non-coding RNA in biological processes has many applications, including roles in carcinogenesis and epithelial–mesenchymal transition (EMT), a process thought to play important roles in MPM pathogenesis. Non-coding RNAs can be separated loosely into two subtypes, short non-coding RNAs (<200 nucleotides) or long (>200 nucleotides). A significant body of evidence has emerged for the roles of short non-coding RNAs in MPM. Less is known about the roles of long non-coding RNAs (lncRNAs) in this disease setting. LncRNAs have been shown to play diverse roles in EMT, and it has been suggested that EMT may play a role in the aggressiveness of MPM histological subsets. In this report, using both in vitro analyses on mesothelioma patient material and in silico analyses of existing RNA datasets, we posit that various lncRNAs may play important roles in EMT within MPM, and we review the current literature regarding these lncRNAs with respect to both EMT and MPM.
Collapse
Affiliation(s)
- Anand S Singh
- Thoracic Oncology Research Group, Trinity Translational Medical Institute, St. James's Hospital, Dublin D08 W9RT, Ireland.
- MSc in Translational Oncology Program, Trinity College Dublin, Dublin 2, Ireland.
| | - Richard Heery
- Thoracic Oncology Research Group, Trinity Translational Medical Institute, St. James's Hospital, Dublin D08 W9RT, Ireland.
- MSc in Translational Oncology Program, Trinity College Dublin, Dublin 2, Ireland.
| | - Steven G Gray
- Thoracic Oncology Research Group, Trinity Translational Medical Institute, St. James's Hospital, Dublin D08 W9RT, Ireland.
- HOPE Directorate, St. James's Hospital, Dublin 8, Ireland.
- Department of Clinical Medicine, Trinity College Dublin, Dublin 8, Ireland.
- Labmed Directorate, St. James's Hospital, Dublin 8, Ireland.
| |
Collapse
|
285
|
Fragiadaki M, Zeidler MP. Ankyrin repeat and single KH domain 1 (ANKHD1) drives renal cancer cell proliferation via binding to and altering a subset of miRNAs. J Biol Chem 2018; 293:9570-9579. [PMID: 29695508 DOI: 10.1074/jbc.ra117.000975] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2017] [Revised: 04/09/2018] [Indexed: 12/21/2022] Open
Abstract
Clear cell renal cell carcinoma (ccRCC) represents the most common kidney cancer worldwide. Increased cell proliferation associated with abnormal microRNA (miRNA) regulation are hallmarks of carcinogenesis. Ankyrin repeat and single KH domain 1 (ANKHD1) is a highly conserved protein found to interact with core cancer pathways in Drosophila; however, its involvement in RCC is completely unexplored. Quantitative PCR studies coupled with large-scale genomics data sets demonstrated that ANKHD1 is significantly up-regulated in kidneys of RCC patients when compared with healthy controls. Cell cycle analysis revealed that ANKHD1 is an essential factor for RCC cell division. To understand the molecular mechanism(s) utilized by ANKHD1 to drive proliferation, we performed bioinformatics analyses that revealed that ANKHD1 contains a putative miRNA-binding motif. We screened 48 miRNAs with tumor-enhancing or -suppressing activities and found that ANKHD1 binds to and regulates three tumor-suppressing miRNAs (i.e. miR-29a, miR-205, and miR-196a). RNA-immunoprecipitation assays demonstrated that ANKHD1 physically interacts with its target miRNAs via a single K-homology domain, located in the C terminus of the protein. Functionally, we discovered that ANKHD1 positively drives ccRCC cell mitosis via binding to and suppressing mainly miR-29a and to a lesser degree via miR-196a/205, leading to up-regulation in proliferative genes such as CCDN1. Collectively, these data identify ANKHD1 as a new regulator of ccRCC proliferation via specific miRNA interactions.
Collapse
Affiliation(s)
- Maria Fragiadaki
- From the Academic Nephrology Unit, Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield S10 2RX, United Kingdom and .,the Bateson Centre, Departments of Biomedical Science, University of Sheffield, Sheffield S10 2TN, United Kingdom
| | - Martin P Zeidler
- the Bateson Centre, Departments of Biomedical Science, University of Sheffield, Sheffield S10 2TN, United Kingdom
| |
Collapse
|
286
|
Biswas S, Thomas AA, Chen S, Aref-Eshghi E, Feng B, Gonder J, Sadikovic B, Chakrabarti S. MALAT1: An Epigenetic Regulator of Inflammation in Diabetic Retinopathy. Sci Rep 2018; 8:6526. [PMID: 29695738 PMCID: PMC5916949 DOI: 10.1038/s41598-018-24907-w] [Citation(s) in RCA: 110] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2017] [Accepted: 04/12/2018] [Indexed: 12/16/2022] Open
Abstract
Despite possessing limited protein-coding potential, long non-coding RNAs (lncRNAs) have been implicated in a myriad of pathologic conditions. Most well documented in cancer, one prominent intergenic lncRNA known as MALAT1 is notorious for its role in impacting epigenetic mechanisms. In this study, we established a novel epigenetic paradigm for MALAT in diabetic retinopathy (DR) by employing siRNA-mediated MALAT1 knockdown in human retinal endothelial cells (HRECs), a Malat1 knockout animal model, vitreous humor from diabetic patients, pharmacological inhibitors for histone and DNA methylation, RNA immunoprecipitation, western blotting, and a unique DNA methylation array to determine glucose-related alterations in MALAT1. Our findings indicated that MALAT1 is capable of impacting the expressions of inflammatory transcripts through its association with components of the PRC2 complex in diabetes. Furthermore, the vitreous humors from diabetic patients revealed increased expressions of MALAT1, TNF-α, and IL-6. Intriguingly, our DNA methylation array demonstrated that transient high glucose exposure in HRECs does not contribute to significant methylation alterations at CpG sites across the MALAT1 gene. However, global inhibition of DNA methyltransferases induced significant increases in MALAT1 and associated inflammatory transcripts in HRECs. Our findings collectively demonstrate the importance of MALAT1 in inflammation and epigenetic regulation in DR.
Collapse
Affiliation(s)
- Saumik Biswas
- Department of Pathology and Laboratory Medicine, Western University, London, Ontario, Canada
| | - Anu Alice Thomas
- Department of Pathology and Laboratory Medicine, Western University, London, Ontario, Canada
| | - Shali Chen
- Department of Pathology and Laboratory Medicine, Western University, London, Ontario, Canada
| | - Erfan Aref-Eshghi
- Department of Pathology and Laboratory Medicine, Western University, London, Ontario, Canada
| | - Biao Feng
- Department of Pathology and Laboratory Medicine, Western University, London, Ontario, Canada
| | - John Gonder
- Department of Ophthalmology, Western University, London, Ontario, Canada
| | - Bekim Sadikovic
- Department of Pathology and Laboratory Medicine, Western University, London, Ontario, Canada
| | - Subrata Chakrabarti
- Department of Pathology and Laboratory Medicine, Western University, London, Ontario, Canada.
| |
Collapse
|
287
|
Liu X, Wu Q, Li L. Functional and therapeutic significance of EZH2 in urological cancers. Oncotarget 2018; 8:38044-38055. [PMID: 28410242 PMCID: PMC5514970 DOI: 10.18632/oncotarget.16765] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2016] [Accepted: 03/04/2017] [Indexed: 11/25/2022] Open
Abstract
The enhancer of zeste homolog 2 (EZH2) is a core subunit of the polycomb repressor complex 2 (PRC2), which is overexpressed in numerous cancers and mutated in several others. Notably, EZH2 acts not only a critical epigenetic repressor through its role in histone methylation, it is also an activator of gene expression, acting through multiple signaling pathways in distinct cancer types. Increasing evidence suggests that EZH2 is an oncogene and is central to initiation, growth and progression of urological cancers. In this review, we highlight the critical role of EZH2 as a master regulator of tumorigenesis in the prostate, bladder and the kidney through epigenetic control of transcription as well as a modulation of various critical signaling pathways. We also discuss the promise and challenges for EZH2 inhibitors as future anticancer therapeutics, some of which are currently in clinical trials.
Collapse
Affiliation(s)
- Xiaobing Liu
- Department of Urology, Second Affiliated Hospital, Third Military Medical University, Chongqing, China
| | - Qingjian Wu
- Department of Urology, Second Affiliated Hospital, Third Military Medical University, Chongqing, China
| | - Longkun Li
- Department of Urology, Second Affiliated Hospital, Third Military Medical University, Chongqing, China
| |
Collapse
|
288
|
Li M, Wang Y, Cheng L, Niu W, Zhao G, Raju JK, Huo J, Wu B, Yin B, Song Y, Bu R. Long non-coding RNAs in renal cell carcinoma: A systematic review and clinical implications. Oncotarget 2018; 8:48424-48435. [PMID: 28467794 PMCID: PMC5564659 DOI: 10.18632/oncotarget.17053] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2016] [Accepted: 03/20/2017] [Indexed: 12/27/2022] Open
Abstract
Renal cell carcinoma is one of the most common malignancy in adults, its prognosis is poor in an advanced stage and early detection is difficult due to the lack of molecular biomarkers. The identification of novel biomarkers for RCC is an urgent and meaningful project. Long non-coding RNA (lncRNA) is transcribed from genomic regions with a minimum length of 200 bases and limited protein-coding potential. Recently, lncRNAs have been greatly studied in a variety of cancer types. They participate in a wide variety of biological processes including cancer biology. In this review, we provide a new insight of the profiling of lncRNAs in RCC and their roles in renal carcinogenesis, with an emphasize on their potential in diagnosis, prognosis and potential roles in RCC therapy.
Collapse
Affiliation(s)
- Ming Li
- Department of Urology, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110004, P.R. China
| | - Ying Wang
- Department of Nuclear Medicine, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning 110001, P.R. China.,Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts 02114, USA
| | - Liang Cheng
- Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Wanting Niu
- Department of Orthopedics, Brigham and Women's Hospital, VA Boston Healthcare System, Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Guoan Zhao
- School of Network Education, Beijing University of Posts and Telecommunications, Hebei, Beijing 100088, P.R. China
| | - Jithin K Raju
- Department of Clinical Medicine, China Medical University, Shenyang, Liaoning 110122, P.R. China
| | - Jun Huo
- Department of Urology, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110004, P.R. China
| | - Bin Wu
- Department of Urology, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110004, P.R. China
| | - Bo Yin
- Department of Urology, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110004, P.R. China
| | - Yongsheng Song
- Department of Urology, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110004, P.R. China
| | - Renge Bu
- Department of Urology, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110004, P.R. China
| |
Collapse
|
289
|
Huo Y, Li Q, Wang X, Jiao X, Zheng J, Li Z, Pan X. MALAT1 predicts poor survival in osteosarcoma patients and promotes cell metastasis through associating with EZH2. Oncotarget 2018; 8:46993-47006. [PMID: 28388584 PMCID: PMC5564539 DOI: 10.18632/oncotarget.16551] [Citation(s) in RCA: 68] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2017] [Accepted: 03/06/2017] [Indexed: 12/30/2022] Open
Abstract
Osteosarcoma is the most common type of bone cancer, especially in children and young adults. Recently, long noncoding RNAs (lncRNAs) have emerged as new prognostic markers and gene regulators in several cancers, including osteosarcoma. In this study, we investigated the contributions of the lncRNA MALAT1 in osteosarcoma with a specific focus on its transcriptional regulation and its interaction with EZH2. Our results showed that MALAT1 was significantly increased in osteosarcoma specimens and cell lines. ROC curve analysis showed that MALAT1 had a higher area under the curve than alkaline phosphatase, and Kaplan-Meier survival analysis indicated that patients with high serum levels of MALAT1 showed reduced survival rate. Knockdown of MALAT1 decreased osteosarcoma cell invasion and promoted E-cadherin expression. Mechanistic investigations showed that MALAT1 was transcriptionally activated by TGF-β. Additionally, EZH2 is highly expressed and associated with the 3’ end region of lncRNA MALAT1 in osteosarcoma, and this association finally suppressed the expression of E-cadherin. Subsequently, our gain and loss function assay showed that MALAT1 overexpression promoted cell metastasis and decreased E-cadherin level, however, this effect was partially reversed by EZH2 knockdown. In conclusion, our work illuminates that lncRNA MALAT1 is a potential diagnostic and prognostic factor in osteosarcoma and further demonstrates how MALAT1 confers an oncogenic function. Thus, lncRNA MALAT1 may serve as a promising prognostic and therapeutic target for osteosarcoma patients.
Collapse
Affiliation(s)
- Yanqing Huo
- Department of Orthopedics, The Second Hospital of Shandong University, Jinan, 250133, Shandong Province, China
| | - Qingbo Li
- Department of Orthopedics, The Second Hospital of Shandong University, Jinan, 250133, Shandong Province, China
| | - Xiqian Wang
- Department of Orthopedics, The Second Hospital of Shandong University, Jinan, 250133, Shandong Province, China
| | - Xiejia Jiao
- Department of Orthopedics, The Second Hospital of Shandong University, Jinan, 250133, Shandong Province, China
| | - Jiachun Zheng
- Department of Orthopedics, The Second Hospital of Shandong University, Jinan, 250133, Shandong Province, China
| | - Zhiqiang Li
- Department of Orthopedics, The Second Hospital of Shandong University, Jinan, 250133, Shandong Province, China
| | - Xiaohan Pan
- Department of Health Management, The Second Hospital of Shandong University, Jinan, 250133, Shandong Province, China
| |
Collapse
|
290
|
Song S, Yu W, Lin S, Zhang M, Wang T, Guo S, Wang H. LncRNA ADPGK-AS1 promotes pancreatic cancer progression through activating ZEB1-mediated epithelial-mesenchymal transition. Cancer Biol Ther 2018; 19:573-583. [PMID: 29667486 DOI: 10.1080/15384047.2018.1423912] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
OBJECTIVE This study was conducted to investigate the effects of ADP dependent glucokinase antisense RNA 1 (ADPGK-AS1)/ miR-205-5p/ zinc finger E-box binding homeobox 1 (ZEB1) on PC cells. METHODS Differentially expressed lncRNAs and miRNAs in pancreatic cancer (PC) were identified by microarray analysis. In silico ceRNA analysis was conducted to find out the interactions among lncRNAs, miRNAs and mRNAs. Quantitative real-time PCR (qRT-PCR) was utilized to examine the expression of miR-205-5p and lncRNA ADPGK-AS1 in PC and non-cancerous cells. The association between miR-205-5p and ADPGK-AS1 as well as miR-205-5p and ZEB1 was determined by dual-luciferase reporter gene assay. After manipulating the expression of ADPGK-AS1, mir-205-5p and ZEB1 in PANC-1 and SW-1990 cells, cell proliferation, migration, invasion and apoptosis were respectively confirmed by cell counting kit-8 (CCK-8) assay, transwell assay and TUNEL. Western blot was applied to examine the expression of Epithelial-mesenchymal Transition-related proteins. In vivo experiment was conducted to further determine the effect of miR-205-5p/ZEB1 on tumorigenic ability of PC cells. RESULTS MiR-205-5p was low-expressed while ZEB1 and ADPGK-AS1 were high-expressed in PC tissues and cells compared with the normal. Dual-luciferase reporter gene assay proved that ADPGK-AS1 could directly target miR-205-5p and miR-205-5p could directly target ZEB1 3'UTR. The expression of MiR-205-5p was negatively correlated with proliferation, migration and invasion, and positively correlated with apoptosis rate of PC cells, while ZEB1 and ADPGK-AS1 had an inversed effect. Further in vitro and in vivo investigation indicated that epithelial-mesenchymal transition (EMT) could be restrained by miR-205-5p through targeting ZEB1. ADPGK-AS1 strongly promoted the tumorigenesis via downregulating miR-205-5p expression and induced the EMT process in vivo. CONCLUSION ADPGK-AS1 inhibited miR-205-5p and therefore promoted PC progression through activating ZEB1-induced EMT.
Collapse
Affiliation(s)
- Suzhen Song
- a Department of Internal Medicine , Shandong University of Traditional Chinese Medicine , Jinan , Shandong , China
| | - Weihua Yu
- b Department of Digestive Disease , the Qilu Second Hospital of Shandong University , Jinan , Shandong , China
| | - Sen Lin
- b Department of Digestive Disease , the Qilu Second Hospital of Shandong University , Jinan , Shandong , China
| | - Mingbao Zhang
- b Department of Digestive Disease , the Qilu Second Hospital of Shandong University , Jinan , Shandong , China
| | - Teng Wang
- b Department of Digestive Disease , the Qilu Second Hospital of Shandong University , Jinan , Shandong , China
| | - Shuang Guo
- b Department of Digestive Disease , the Qilu Second Hospital of Shandong University , Jinan , Shandong , China
| | - Hongbo Wang
- b Department of Digestive Disease , the Qilu Second Hospital of Shandong University , Jinan , Shandong , China
| |
Collapse
|
291
|
Li YL, Jin YF, Liu XX, Li HJ. A comprehensive analysis of Wnt/β-catenin signaling pathway-related genes and crosstalk pathways in the treatment of As 2O 3 in renal cancer. Ren Fail 2018; 40:331-339. [PMID: 29633893 PMCID: PMC6014489 DOI: 10.1080/0886022x.2018.1456461] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
We aimed to investigate the effect of As2O3 treatment on Wnt/β-catenin signaling pathway-related genes and pathways in renal cancer. Illumina-based RNA-seq of 786-O cells with or without As2O3 treatment was performed, and differentially expressed genes (DEGs) were identified using Cuffdiff software. TargetMine was utilized to perform Gene Ontology (GO) pathway and Disease Ontology enrichment analyses. Furthermore, TRANSFAC database and LPIA method were applied to select differentially expressed transcription factors (TFs) and pathways related to Wnt/β-catenin signaling pathway, respectively. Additionally, transcriptional regulatory and pathway crosstalk networks were constructed. In total, 1684 DEGs and 69 TFs were screened out. The 821 up-regulated DEGs were mainly enriched in 67 pathways, 70 GO terms, and 46 disease pathways, while only 1 pathway and 5 GO terms were enriched for 863 down-regulated DEGs. A total of 18 DEGs (4 up-regulated and 14 down-regulated genes) were involved in the Wnt/β-catenin signaling pathway. Among the 18 DEGs, 4 ones were TFs. Furthermore, 211 pathways were predicted to be linked to the Wnt/β-catenin signaling pathway. In conclusion, As2O3 may have a significant effect on the Wnt/β-catenin signaling pathway for renal cancer treatment. The potential key DEGs are expected to be used as therapeutic targets.
Collapse
Affiliation(s)
- Yan-Lei Li
- a Medical Examination Center , China-Japan Union Hospital of Jilin University , Changchun , China
| | - Yu-Fen Jin
- b Clinical Laboratory , The Second Hospital of Jilin University , Changchun , China
| | - Xiu-Xia Liu
- b Clinical Laboratory , The Second Hospital of Jilin University , Changchun , China
| | - Hong-Jun Li
- a Medical Examination Center , China-Japan Union Hospital of Jilin University , Changchun , China
| |
Collapse
|
292
|
Wu Q, Meng WY, Jie Y, Zhao H. LncRNA MALAT1 induces colon cancer development by regulating miR-129-5p/HMGB1 axis. J Cell Physiol 2018; 233:6750-6757. [PMID: 29226325 DOI: 10.1002/jcp.26383] [Citation(s) in RCA: 125] [Impact Index Per Article: 17.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2017] [Accepted: 12/04/2017] [Indexed: 01/08/2023]
Abstract
Recent studies have exhibited significant roles of lncRNAs in various tumors' development, including colon cancer. Our study focused on the biological roles of lncRNA MALAT1 in colon cancer. In our study, it was demonstrated that MALAT1 was upregulated in human colon cancer cell lines including Lovo, HCT116, SW480, and HT29 cells compared to the normal human intestinal epithelial HIEC cells. Moreover, we observed that miR-129-5p was downregulated in colon cancer cells with a significant increase of HMGB1 expression. Inhibition of MALAT1 can inhibit the proliferation of colon cancer SW480 and HCT116 cells and next, bioinformatics analysis was used to predict the target microRNA of MALAT1. miR-129-5p was identified and confirmed as a direct regulator of MALAT1 and it was shown that miR-129-5p mimics were able to restrain the progression of colon cancer cells. In addition, high motility group box protein 1 (HMGB1), was predicted as a mRNA target of miR-129-5p. Furthermore, we found that MALAT1 exerted its biological functions through regulating HMGB1 by sponging miR-129-5p in vitro. Silencing MALAT1 greatly inhibited HMGB1 expression which can be reversed by miR-129-5p inhibitors. It was indicated in our investigation that MALAT1 may serve as a competing endogenous lncRNA (ceRNA) to mediate HMGB1 by sponging miR-129-5p in colon cancer. Taken together, our results indicated that MALAT1/miR-129-5p/HMGB1 axis could be provided as an important prognostic biomarker in colon cancer development.
Collapse
Affiliation(s)
- Qiong Wu
- Department of Gastroenterology, Shanghai Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wen-Ying Meng
- Department of Gastroenterology, Shanghai Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ying Jie
- Department of Clinical Research Center, People's Hospital of Xuyi, Jiangsu Province, China
| | - Haijian Zhao
- Division of Pediatric Surgery, Department of General Surgery, Huai'an Second People's Hospital and The Affiliated Huai'an Hospital of Xuzhou Medical University, Jiangsu, China
| |
Collapse
|
293
|
Xue D, Lu H, Xu HY, Zhou CX, He XZ. Long noncoding RNA MALAT1 enhances the docetaxel resistance of prostate cancer cells via miR-145-5p-mediated regulation of AKAP12. J Cell Mol Med 2018; 22:3223-3237. [PMID: 29633510 PMCID: PMC5980122 DOI: 10.1111/jcmm.13604] [Citation(s) in RCA: 78] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2017] [Accepted: 02/13/2018] [Indexed: 01/04/2023] Open
Abstract
Our present work was aimed to study on the regulatory role of MALAT1/miR-145-5p/AKAP12 axis on docetaxel (DTX) sensitivity of prostate cancer (PCa) cells. The microarray data (GSE33455) to identify differentially expressed lncRNAs and mRNAs in DTX-resistant PCa cell lines (DU-145-DTX and PC-3-DTX) was retrieved from the Gene Expression Omnibus (GEO) database. QRT-PCR analysis was performed to measure MALAT1 expression in DTX-sensitive and DTX-resistant tissues/cells. The human DTX-resistant cell lines DU145-PTX and PC3-DTX were established as in vitro cell models, and the expression of MALAT1, miR-145-5p and AKAP12 was manipulated in DTX-sensitive and DTX-resistant cells. Cell viability was examined using MTT assay and colony formation methods. Cell apoptosis was assessed by TUNEL staining. Cell migration and invasion was determined by scratch test (wound healing) and Transwell assay, respectively. Dual-luciferase assay was applied to analyse the target relationship between lncRNA MALAT1 and miR-145-5p, as well as between miR-145-5p and AKAP12. Tumour xenograft study was undertaken to confirm the correlation of MALAT1/miR-145-5p/AKAP12 axis and DTX sensitivity of PCa cells in vivo. In this study, we firstly notified that the MALAT1 expression levels were up-regulated in clinical DTX-resistant PCa samples. Overexpressed MALAT1 promoted cell proliferation, migration and invasion but decreased cell apoptosis rate of PCa cells in spite of DTX treatment. We identified miR-145-5p as a target of MALAT1. MiR-145-5p overexpression in PC3-DTX led to inhibited cell proliferation, migration and invasion as well as reduced chemoresistance to DTX, which was attenuated by MALAT1. Moreover, we determined that AKAP12 was a target of miR-145-5p, which significantly induced chemoresistance of PCa cells to DTX. Besides, it was proved that MALAT1 promoted tumour cell proliferation and enhanced DTX-chemoresistance in vivo. There was an lncRNA MALAT1/miR-145-5p/AKAP12 axis involved in DTX resistance of PCa cells and provided a new thought for PCa therapy.
Collapse
Affiliation(s)
- Dong Xue
- Department of Urology, Third Affiliated Hospital, Suzhou University, Changzhou, Jiangsu, China
| | - Hao Lu
- Department of Urology, Third Affiliated Hospital, Suzhou University, Changzhou, Jiangsu, China
| | - Han-Yan Xu
- Department of Urology, Third Affiliated Hospital, Suzhou University, Changzhou, Jiangsu, China
| | - Cui-Xing Zhou
- Department of Urology, Third Affiliated Hospital, Suzhou University, Changzhou, Jiangsu, China
| | - Xiao-Zhou He
- Department of Urology, Third Affiliated Hospital, Suzhou University, Changzhou, Jiangsu, China
| |
Collapse
|
294
|
Li Y, Shen Z, Wang B, Ye C, Lai Z, Jiang H, Wang Z, Jiang K, Ye Y, Wang S. Long non-coding RNA GPR65-1 is up-regulated in gastric cancer and promotes tumor growth through the PTEN-AKT-slug signaling pathway. Cell Cycle 2018; 17:759-765. [PMID: 29336198 DOI: 10.1080/15384101.2018.1426414] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Increasing evidence has shown that abnormal expression of lncRNAs is involved in various biological behaviors and major cellular pathways of human cancers. However, the role of lncRNAs in the progression of gastric cancer has not been adequately investigated. Therefore, in this study, we investigated the expression levels of linc-GPR65-1 using Quantitative real-time PCR (qRT-PCR) and found that linc-GPR65-1 was significantly up-regulated in 50 gastric cancer tissues compared to the corresponding normal tissues. In addition, increased linc-GPR65-1 expression was associated with TNM stage (P = 0.037), tumor size (P = 0.024), distal metastasis (P = 0.023), and poor prognosis of gastric cancer patients. Moreover, functional assays indicated that decreased linc-GPR65-1 expression inhibited the aggressive phenotypes of gastric cancer cells, and enhanced linc-GPR65-1 expression resulted in the opposite phenomenon. Then, a cancer signaling phosphoantibody microarray was conducted to explore the potential mechanisms of linc-GPR65-1 in regulating gastric cancer progression and observed that linc-GPR65-1 could regulate the PTEN-AKT-slug signaling pathway. These data showed that linc-GPR65-1, regulating the PTEN-AKT-slug signaling pathway, might act as a tumor promoter and serve as a novel target for gastric cancer prevention and therapy.
Collapse
Affiliation(s)
- Yansen Li
- a Department of Gastroenterological Surgery , Peking University People's Hospital
| | - Zhanlong Shen
- a Department of Gastroenterological Surgery , Peking University People's Hospital.,b Laboratory of Surgical Oncology , Peking University People's Hospital
| | - Bo Wang
- a Department of Gastroenterological Surgery , Peking University People's Hospital.,b Laboratory of Surgical Oncology , Peking University People's Hospital
| | - Chunxiang Ye
- c Beijing Key Laboratory of Colorectal Cancer Diagnosis and Treatment Research , Peking University People's Hospital
| | - Zhiyong Lai
- a Department of Gastroenterological Surgery , Peking University People's Hospital
| | - Hongpeng Jiang
- a Department of Gastroenterological Surgery , Peking University People's Hospital
| | - Zhu Wang
- a Department of Gastroenterological Surgery , Peking University People's Hospital
| | - Kewei Jiang
- a Department of Gastroenterological Surgery , Peking University People's Hospital
| | - Yingjiang Ye
- a Department of Gastroenterological Surgery , Peking University People's Hospital.,b Laboratory of Surgical Oncology , Peking University People's Hospital
| | - Shan Wang
- a Department of Gastroenterological Surgery , Peking University People's Hospital.,b Laboratory of Surgical Oncology , Peking University People's Hospital
| |
Collapse
|
295
|
Xu D, Liu R, Meng L, Zhang Y, Lu G, Ma P. Long non-coding RNA ENST01108 promotes carcinogenesis of glioma by acting as a molecular sponge to modulate miR-489. Biomed Pharmacother 2018; 100:20-28. [DOI: 10.1016/j.biopha.2018.01.126] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2017] [Revised: 01/11/2018] [Accepted: 01/28/2018] [Indexed: 10/18/2022] Open
|
296
|
He X, Yan Q, Kuang G, Wang Y, Cao P, Ou C. Metastasis-associated lung adenocarcinoma transcript 1 regulates tumor progression: old wine in a new bottle. J Thorac Dis 2018; 10:S1088-S1091. [PMID: 29850189 PMCID: PMC5949406 DOI: 10.21037/jtd.2018.04.13] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2018] [Accepted: 03/30/2018] [Indexed: 12/31/2022]
Affiliation(s)
- Xiaoyun He
- Department of Hematology, Xiangya Hospital, Central South University, Changsha 410008, China
- Department of Endocrinology, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Qijia Yan
- Department of Pathology, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Gaoyan Kuang
- The First Affiliated Hospital of Hunan University of Chinese Medicine, Changsha 410007, China
| | - Yixuan Wang
- Department of Pathology, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Pengfei Cao
- Department of Hematology, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Chunlin Ou
- Department of Pathology, Xiangya Hospital, Central South University, Changsha 410008, China
| |
Collapse
|
297
|
Jiao D, Li Z, Zhu M, Wang Y, Wu G, Han X. LncRNA MALAT1 promotes tumor growth and metastasis by targeting miR-124/foxq1 in bladder transitional cell carcinoma (BTCC). Am J Cancer Res 2018; 8:748-760. [PMID: 29736319 PMCID: PMC5934564] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2016] [Accepted: 11/29/2016] [Indexed: 06/08/2023] Open
Abstract
Mounting evidence shows that the long non-coding RNA MALAT1 plays a pivotal role in tumorigenesis and metastasis, but the functional significance of MALAT1 in bladder transitional cell carcinoma (BTCC) remains unclear. MALAT1 expression was measured in 56 BTCC patients and 2 BTCC cell lines by real-time PCR. The effects of MALAT1 on BTCC cells were investigated by over-expression approaches in vitro and in vivo. Insights of the mechanism of competitive endogenous RNAs (ceRNAs) were validated through bioinformatic analysis and luciferase assay. MALAT1 up-regulation positively correlated with advanced clinical pathological stage and shorter survival of BTCC patients. Furthermore, MALAT1 over-expression promoted proliferation, migration and invasion of BTCC cells in vitro and in vivo. Particularly, MALAT1 may function as a ceRNA to sponge miR-124, thus modulating the derepression of foxq1, miR-124 target gene, in post-transcriptional levels. The positive MALAT1/foxq1 interaction was confirmed by bivariate correlation analysis, and this positive correlation was of great significance in BTCC tumor growth and metastasis, also accompanied by EMT changes. Overall, this ceRNA regulatory network concerning MALAT1 and the positive MALAT1/foxq1 correlation benefit a better understanding of BTCC pathogenesis and promote the feasibility of lncRNA-directed therapy against this disease.
Collapse
Affiliation(s)
- Dechao Jiao
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University Zhengzhou 450052, Henan, China
| | - Zongming Li
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University Zhengzhou 450052, Henan, China
| | - Ming Zhu
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University Zhengzhou 450052, Henan, China
| | - Yanli Wang
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University Zhengzhou 450052, Henan, China
| | - Gang Wu
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University Zhengzhou 450052, Henan, China
| | - Xinwei Han
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University Zhengzhou 450052, Henan, China
| |
Collapse
|
298
|
Jiang LT, Wan CH, Guo QH, Yang SJ, Wu JD, Cai J. Long Noncoding RNA Metastasis-Associated Lung Adenocarcinoma Transcript 1 (MALAT1) Promotes Renal Cell Carcinoma Progression via Sponging miRNA-429. Med Sci Monit 2018; 24:1794-1801. [PMID: 29588438 PMCID: PMC5887685 DOI: 10.12659/msm.909450] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Background It is well known that long noncoding RNA (lncRNA) metastasis-associated lung adenocarcinoma transcript 1 (MALAT1) is closely correlated with the tumorigenesis of multiple cancers, including renal cell carcinoma (RCC). However, the potential functional mechanism is still elusive. Material/Methods In our present research, quantitative real-time polymerase chain reaction (qRT-PCR) was performed for the measurement of MALAT1 and miR-429. CCK-8 assay and Transwell assay were performed for the proliferation, migration, and invasion abilities of RCC cells. Dual-luciferase reporter assay was performed to validate the interaction within MALAT1 and miR-429. Results Data found that MALAT1 was overexpressed in RCC clinical samples and cell lines. Moreover, loss-of-functional experiments showed that MALAT1 knockdown suppress the proliferation, migration, and invasion abilities of RCC cells. RT-PCR showed that miR-429 expression was downregulated in RCC cell lines, which was negatively correlated with that of MALAT1. Bioinformatics analysis suggested that miR-429 had complementary binding sequences with MALAT1, which was confirmed by dual-luciferase reporter assay. Conclusions In summary, our results concluded that MALAT1 functioned as an oncogene in RCC by sponging miR-429, acting as its competing endogenous RNA (ceRNA).
Collapse
Affiliation(s)
- Lin-Tao Jiang
- Department of Emergency and Trauma Surgery, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China (mainland)
| | - Chun-Hua Wan
- Department of Anesthesiology, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China (mainland)
| | - Qing-Hao Guo
- Department of Emergency and Trauma Surgery, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China (mainland)
| | - Shi-Jiang Yang
- Department of Emergency and Trauma Surgery, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China (mainland)
| | - Jing-Dong Wu
- Department of Anesthesiology, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China (mainland)
| | - Jun Cai
- Department of Emergency and Trauma Surgery, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China (mainland)
| |
Collapse
|
299
|
Kim SH, Kim SH, Yang WI, Kim SJ, Yoon SO. Association of the long non-coding RNA MALAT1 with the polycomb repressive complex pathway in T and NK cell lymphoma. Oncotarget 2018; 8:31305-31317. [PMID: 28412742 PMCID: PMC5458209 DOI: 10.18632/oncotarget.15453] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2016] [Accepted: 01/31/2017] [Indexed: 01/16/2023] Open
Abstract
Recently, various long non-coding RNAs (lncRNAs) have been reported to have significant therapeutic or prognostic value. However, the expression of lncRNAs has not been investigated in T and NK cell lymphoma. Thus, we evaluated the biological and prognostic role of lncRNAs related to the polycomb repressive complex (PRC) and PRC markers in tissue samples and cell lines of T and NK cell lymphoma. Among the tested lncRNAs, MALAT1 was most highly expressed in clinical samples and cell lines. High expression of MALAT1 as well as BMI1 was related to poor prognosis in patients with mature T cell lymphoma. In the tissue samples, BMI1 expression showed a positive correlation with EZH2, SUZ12, H3K27me3, and MALAT1. Multiple linear regression analysis showed that BMI1 expression was independently associated with H3K27me3. Direct binding of MALAT1 to the PRC2 components (EZH2 and SUZ12) was observed in a T cell lymphoma cell line; however, no direct binding of MALAT1 with H3K27me3 and BMI1 (a PRC1 component) was observed. In T and NK cell lymphomas, MALAT1 was related to poor prognosis. MALAT1 directly binds to EZH2 and SUZ12, and BMI1 activation may be induced possibly through H3K27me3.
Collapse
Affiliation(s)
- Soo Hee Kim
- Department of Pathology, Yonsei University College of Medicine, Seoul, Korea.,Anatomic Pathology Reference Lab, Seegene Medical Foundation, Seoul, Korea
| | - Se Hoon Kim
- Department of Pathology, Yonsei University College of Medicine, Seoul, Korea
| | - Woo Ick Yang
- Department of Pathology, Yonsei University College of Medicine, Seoul, Korea
| | - Soo Jeong Kim
- Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Korea
| | - Sun Och Yoon
- Department of Pathology, Yonsei University College of Medicine, Seoul, Korea
| |
Collapse
|
300
|
Chen J, Chen Y, Gu L, Li X, Gao Y, Lyu X, Chen L, Luo G, Wang L, Xie Y, Duan J, Peng C, Ma X. LncRNAs act as prognostic and diagnostic biomarkers in renal cell carcinoma: a systematic review and meta-analysis. Oncotarget 2018; 7:74325-74336. [PMID: 27527868 PMCID: PMC5342056 DOI: 10.18632/oncotarget.11101] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2016] [Accepted: 07/19/2016] [Indexed: 12/28/2022] Open
Abstract
We conducted a systematic review and meta-analysis to investigate the clinical values, including clinicopathology, prognosis, and diagnosis of different long non-coding RNAs (lncRNAs) in renal cell carcinoma (RCC). A total of 14 eligible studies, including 10 on clinicopathological features, 11 on prognosis, and 3 on diagnosis were identified. Results revealed that metastasis-associated lung adenocarcinoma transcript 1(MALAT1) expression was associated with tumor stage (odds ratio [OR], 3.46; 95% confidence interval [CI], 1.63-7.36; p=0.001). The high expression of MALAT1 could be considered a biomarker of the early detection of lymph node metastasis and predictor of poor survival in RCC patients, who likely manifested short overall survival (OS; hazard ratio [HR], 2.97; 95% CI, 1.68-5.28; p<0.001). For diagnostic value, the pooled result showed that lncRNA maintained a sensitivity of 0.89 and specificity of 0.91 in RCC diagnosis, The area under the curve of 0.94 (95% CI, 0.92-0.96) for lncRNA in RCC diagnosis also indicated a significant advantage over other biomarkers. Our systematic review and meta-analysis demonstrated that lncRNAs could be considered biomarkers to detect lymph node metastasis and distant metastasis in early stages. LncRNAs could function as potential prognostic markers in RCC. LncRNAs could also display high accuracy for RCC diagnosis.
Collapse
Affiliation(s)
- Jianwen Chen
- Department of Urology/State Key Laboratory of Kidney Diseases, Chinese People's Liberation Army General Hospital, Beijing, China
| | - Yalei Chen
- Department of Cardiology, Beijing Anzhen Hospital affiliated to Capital Medical University, Beijing, China
| | - Liangyou Gu
- Department of Urology/State Key Laboratory of Kidney Diseases, Chinese People's Liberation Army General Hospital, Beijing, China
| | - Xintao Li
- Department of Urology/State Key Laboratory of Kidney Diseases, Chinese People's Liberation Army General Hospital, Beijing, China
| | - Yu Gao
- Department of Urology/State Key Laboratory of Kidney Diseases, Chinese People's Liberation Army General Hospital, Beijing, China
| | - Xiangjun Lyu
- Department of Urology/State Key Laboratory of Kidney Diseases, Chinese People's Liberation Army General Hospital, Beijing, China
| | - Luyao Chen
- Department of Urology/State Key Laboratory of Kidney Diseases, Chinese People's Liberation Army General Hospital, Beijing, China
| | - Guoxiong Luo
- Department of Urology/State Key Laboratory of Kidney Diseases, Chinese People's Liberation Army General Hospital, Beijing, China
| | - Lei Wang
- Department of Urology/State Key Laboratory of Kidney Diseases, Chinese People's Liberation Army General Hospital, Beijing, China
| | - Yongpeng Xie
- Department of Urology/State Key Laboratory of Kidney Diseases, Chinese People's Liberation Army General Hospital, Beijing, China
| | - Junyao Duan
- Department of Urology/State Key Laboratory of Kidney Diseases, Chinese People's Liberation Army General Hospital, Beijing, China
| | - Cheng Peng
- Department of Urology/State Key Laboratory of Kidney Diseases, Chinese People's Liberation Army General Hospital, Beijing, China
| | - Xin Ma
- Department of Urology/State Key Laboratory of Kidney Diseases, Chinese People's Liberation Army General Hospital, Beijing, China
| |
Collapse
|