51
|
Yang Q, Wu MF, Zhu LH, Qiao LX, Zhao RB, Xia ZK. Long non-coding RNA Snhg3 protects against hypoxia/ischemia-induced neonatal brain injury. Exp Mol Pathol 2019; 112:104343. [PMID: 31751562 DOI: 10.1016/j.yexmp.2019.104343] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Revised: 11/16/2019] [Accepted: 11/17/2019] [Indexed: 01/10/2023]
Abstract
Hypoxic-ischemic brain damage (HIBD) is a major cause of morbidity and mortality in the preterm and term infant. However, the precise mechanism of HIBD remains largely elusive. As a newly discovered long non-coding RNA, small nucleolar RNA host gene 3 (Snhg3) has shown its important roles in cell apoptosis, proliferation, and disease development. In this study, we determined the role of Snhg3 in the pathogenesis of HIBD. Snhg3 expression was significantly down-regulated in the neonatal brain and primary hippocampal cells response to hypoxic/ischemic stress. Snhg3 overexpression protected against hypoxic/ischemic-induced brain injury in vivo and hippocampal cell injury in vitro. Snhg3 acted as the sponge of miR-196 in the hippocampal cells by regulating the expression of miR-196 target genes, XIAP and CAAP1. Moreover, Snhg3 overexpression decreased brain infarct size and ameliorated hypoxic-ischemic neonatal brain damage. This study suggests that Snhg3 is a potential target for the treatment of HIBD.
Collapse
Affiliation(s)
- Qing Yang
- Wuhu Hospital of Traditonal Chinese Medicine, Wuhu, China
| | - Ming-Fu Wu
- Alliliated Hospital of Yang Zhou University, Yang Zhou, China
| | - Li-Hua Zhu
- Jiangsu Health Vocational College, Nanjing, China
| | - Li-Xing Qiao
- Department of Pediatrics, Zhongda Hospital, Southeast University, Nanjing, China
| | - Rui-Bin Zhao
- Alliliated Hospital of Yang Zhou University, Yang Zhou, China.
| | - Zheng-Kun Xia
- Department of Pediatrics, Jinling Hospital, Nanjing University School of Medicine, Nanjing, China.
| |
Collapse
|
52
|
Lin X, Kapoor A, Gu Y, Chow MJ, Xu H, Major P, Tang D. Assessment of biochemical recurrence of prostate cancer (Review). Int J Oncol 2019; 55:1194-1212. [PMID: 31638194 PMCID: PMC6831208 DOI: 10.3892/ijo.2019.4893] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2019] [Accepted: 09/24/2019] [Indexed: 12/12/2022] Open
Abstract
The assessment of the risk of biochemical recurrence (BCR) is critical in the management of males with prostate cancer (PC). Over the past decades, a comprehensive effort has been focusing on improving risk stratification; a variety of models have been constructed using PC-associated pathological features and molecular alterations occurring at the genome, protein and RNA level. Alterations in RNA expression (lncRNA, miRNA and mRNA) constitute the largest proportion of the biomarkers of BCR. In this article, we systemically review RNA-based BCR biomarkers reported in PubMed according to the PRISMA guidelines. Individual miRNAs, mRNAs, lncRNAs and multi-gene panels, including the commercially available signatures, Oncotype DX and Prolaris, will be discussed; details related to cohort size, hazard ratio and 95% confidence intervals will be provided. Mechanistically, these individual biomarkers affect multiple pathways critical to tumorigenesis and progression, including epithelial-mesenchymal transition (EMT), phosphatase and tensin homolog (PTEN), Wnt, growth factor receptor, cell proliferation, immune checkpoints and others. This variety in the mechanisms involved not only validates their associations with BCR, but also highlights the need for the coverage of multiple pathways in order to effectively stratify the risk of BCR. Updates of novel biomarkers and their mechanistic insights are considered, which suggests new avenues to pursue in the prediction of BCR. Additionally, the management of patients with BCR and the potential utility of the stratification of the risk of BCR in salvage treatment decision making for these patients are briefly covered. Limitations will also be discussed.
Collapse
Affiliation(s)
- Xiaozeng Lin
- Department of Medicine, McMaster University, Hamilton, ON L8S 4K1, Canada
| | - Anil Kapoor
- The Research Institute of St. Joe's Hamilton, St. Joseph's Hospital, Hamilton, ON L8N 4A6, Canada
| | - Yan Gu
- Department of Medicine, McMaster University, Hamilton, ON L8S 4K1, Canada
| | - Mathilda Jing Chow
- Department of Medicine, McMaster University, Hamilton, ON L8S 4K1, Canada
| | - Hui Xu
- Department of Nephrology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
| | - Pierre Major
- Division of Medical Oncology, Department of Oncology, McMaster University, Hamilton, ON L8V 5C2, Canada
| | - Damu Tang
- Department of Medicine, McMaster University, Hamilton, ON L8S 4K1, Canada
| |
Collapse
|
53
|
Lei J, Fu Y, Zhuang Y, Zhang K, Lu D. LncRNA SNHG1 alleviates IL-1β-induced osteoarthritis by inhibiting miR-16-5p-mediated p38 MAPK and NF-κB signaling pathways. Biosci Rep 2019; 39:BSR20191523. [PMID: 31383786 PMCID: PMC6732361 DOI: 10.1042/bsr20191523] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2019] [Revised: 07/12/2019] [Accepted: 07/22/2019] [Indexed: 12/20/2022] Open
Abstract
Long non-coding RNA (LncRNA) small nucleolar RNA host gene 1 (SNHG1) has been reported in the occurrence and development of several diseases, but its biological role and mechanism in osteoarthritis (OA) remain to be illuminated. In the present research, we aimed to investigate the effect of SNHG1 on IL-1β-induced OA and its molecular mechanism. Results revealed that SNHG1 decreased the expression of MMPs, ADAMTs, collagen, and aggrecan, and ameliorates IL-1β-induced metabolic dysfunction in normal human chondrocytes-keen. In addition, SNHG1 inhibited the expressions of pro-inflammatory cytokines in chondrocytes, including NO, PGE2, IL-6, TNF-α, i-NOS, and COX-2. Furthermore, luciferase reporter assay demonstrated that SNHG1 could directly interact with miR-16-5p and suppressed miR-16-5p expression and activity. What is more, miR-16-5p overexpression reversed SNHG1-inhibited aberrant catabolism and inflammation triggered by IL-1β stimulation. Finally, SNHG1 inhibits the expression of miR-16-5p-mediated factors involved in p38MAPK and NF-κB signaling pathways, including ERK1/2, p-p38 and p-p65. Taken together, the results of our studies illuminate that SNHG1 alleviates the inflammation of IL-1β-induced OA through the activation of miR-16-5p-mediated p38MAPK and NF-κB signaling pathway. It suggested that SNHG1 may serve as a potential target for OA diagnosis and treatment.
Collapse
Affiliation(s)
- Jinlai Lei
- Department of Orthopaedic Trauma, Honghui Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi province 710054, China
| | - Yahui Fu
- Department of Orthopaedic Trauma, Honghui Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi province 710054, China
| | - Yan Zhuang
- Department of Orthopaedic Trauma, Honghui Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi province 710054, China
| | - Kun Zhang
- Department of Orthopaedic Trauma, Honghui Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi province 710054, China
| | - Daigang Lu
- Department of Orthopaedic Trauma, Honghui Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi province 710054, China
| |
Collapse
|
54
|
Liu S, Wang L, Li Y, Cui Y, Wang Y, Liu C. Long non-coding RNA CHRF promotes proliferation and mesenchymal transition (EMT) in prostate cancer cell line PC3 requiring up-regulating microRNA-10b. Biol Chem 2019; 400:1035-1045. [PMID: 30844757 DOI: 10.1515/hsz-2018-0380] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2018] [Accepted: 02/20/2019] [Indexed: 01/17/2023]
Abstract
Despite the advance of diagnosis and treatment for prostate cancer, the prognosis of metastatic prostate cancer is poor. We aimed to explore the functional role of long non-coding RNA cardiac hypertrophy-related factor (lncRNA CHRF) in prostate cancer cells (PC3) as well as the molecular mechanisms. LncRNA CHRF silence repressed cell number (%), down-regulated expression of cyclinD1, CDK4 and CDK6, and promoted apoptosis along with activation of the casapse-3 and caspase-9. LncRNA CHRF promoted mesenchymal transition (EMT), showing down-regulation of E-cadherin and up-regulation of N-cadherin, vimentin and ZEB1. Afterwards, we found miR-10b expression was positively correlated with lncRNA CHRF expression, and miR-10b inhibition could reverse the effects of lncRNA CHRF on PC3 and LNCaP cell proliferation and EMT. Finally, lncRNA CHRF was found to activate the GSK3β/AKT and NF-κB pathways via up-regulation of miR-10b. LncRNA CHRF silence repressed proliferation and EMT while promoted apoptosis in PC3 cells via positive regulation of miR-10b. The GSK3β/AKT and NF-κB pathways were activated by lncRNA CHRF, possibly through up-regulation of miR-10b.
Collapse
Affiliation(s)
- Shuang Liu
- School of Rehabilitation Medicine, Binzhou Medical University, No. 346 Guanhai Road, Yantai 264003, China.,Department of Urology, Yantai Yuhuangding Hospital, Yantai 264000, China
| | - Lin Wang
- Department of Urology, Yantai Yuhuangding Hospital, Yantai 264000, China
| | - Yongwei Li
- Department of Urology, Yantai Yuhuangding Hospital, Yantai 264000, China
| | - Yuanshan Cui
- Department of Urology, Yantai Yuhuangding Hospital, Yantai 264000, China
| | - Yongqiang Wang
- Department of Urology, Yantai Yuhuangding Hospital, Yantai 264000, China
| | - Chu Liu
- Department of Urology, Yantai Yuhuangding Hospital, No. 20 Yudong Road, Yantai 264000, China
| |
Collapse
|
55
|
Bai J, Xu J, Zhao J, Zhang R. lncRNA SNHG1 cooperated with miR-497/miR-195-5p to modify epithelial-mesenchymal transition underlying colorectal cancer exacerbation. J Cell Physiol 2019; 235:1453-1468. [PMID: 31276207 DOI: 10.1002/jcp.29065] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2019] [Accepted: 06/04/2019] [Indexed: 12/11/2022]
Abstract
Our study was intended to provide evidence for whether long noncoding RNA (lncRNA) SNHG1 would accelerate the epithelial-mesenchymal transition (EMT) course intrinsic in colorectal cancer (CRC) by sponging downstream miR-497-5p and miR-195-5p. We altogether collected 338 pairs of CRC and noncancerous tissues, and meanwhile purchased five CRC cell lines (i.e., SW480, HCT116, Lovo, CaCO-2, and HT29) and human embryo intestinal mucosal tissue-sourced cell line (i.e., CCC-HIE-2). The CRC cells as mentioned above were appraised regarding their potencies in proliferation, migration, and invasion, after being transfected with pcDNA3.1-SNHG1, si-SNHG1, miR-195-5p mimic/inhibitor, and miR-497-5p mimic/inhibitor. Eventually, we depended on reverse transcription-polymerase chain reaction to assess SNHG1, miR-497-5p, and miR-195-5p expressions, and the protein levels of EMT-specific molecules were determined on the strength of western blotting. It seemed that there was a high potential for highly expressed SNHG1 and lowly expressed miR-497/miR-195 to symbolize CRC patients' unfavorable prognosis (p < .05). Concurrently, CRC cells were detected with higher SNHG1 expression and lower miR-497/miR-195 expression than CCC-HIE-2 cells (p < .05). In addition, the EMT process of CRC cells was facilitated markedly against the contexts of overexpressed SNHG1 and underexpressed miR-497-5p/miR-195-5p. Intriguingly, the strength of miR-195-5p collaborating with miR-497-5p in affecting the activity of CRC cells seemed to overweigh that of miR-497/miR-195-5p alone. Besides, both miR-195-5p and miR-497-5p were subjected to in vivo and in vitro modification of SNHG1 (p < .05). Conclusively, application of lncRNA SNHG1 for treating CRC might be promising, given its dual modulation of miR-497 and miR-195 underlying CRC pathogenesis.
Collapse
Affiliation(s)
- Jinghui Bai
- Department of Internal Medicine, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Insititute, Shenyang, Liaoning, China
| | - Jian Xu
- Department of Colorectal Surgery, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Insititute, Shenyang, Liaoning, China
| | - Jian Zhao
- Department of Colorectal Surgery, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Insititute, Shenyang, Liaoning, China
| | - Rui Zhang
- Department of Colorectal Surgery, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Insititute, Shenyang, Liaoning, China
| |
Collapse
|
56
|
Zhu M, Li M, Zhou W, Yang Y, Li F, Zhang L, Ji G. Qianggan extract improved nonalcoholic steatohepatitis by modulating lncRNA/circRNA immune ceRNA networks. BMC COMPLEMENTARY AND ALTERNATIVE MEDICINE 2019; 19:156. [PMID: 31269941 PMCID: PMC6609373 DOI: 10.1186/s12906-019-2577-6] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/13/2019] [Accepted: 06/25/2019] [Indexed: 01/01/2023]
Abstract
BACKGROUND The traditional Chinese medicine prescription, Qianggan formula have been confirmed to be effective on non-alcoholic steatohepatitis (NASH), however, the underlying molecular mechanisms remain obscure. METHODS Thirty-six male C57BL/6 mice were randomly divided into three groups: normal chow diet group; methionine-and-choline-deficient diet (MCD) group, and Qianggan extract (QG) intervention group (0.4 g/kg daily) that fed with MCD. The efficacy of QG was biochemically and histologically evaluated. The expression profiles of messenger ribonucleic acids (mRNAs), long non-coding RNAs (lncRNAs) and circular RNAs (circRNAs) were examined using microarray and verified by RT-qPCR. RESULTS QG significantly improved the phenotypic characteristics of NASH, as serum alanine aminotransferase (ALT), aspartate aminotransferase (AST), alkaline phosphatase (ALP), and lactate dehydrogenase (LDH) levels and liver inflammatory cytokines were significantly decreased. By the cutoff of a 1.5-fold change and P < 0.05, 6193 mRNAs, 5692 lncRNAs and 4843 circRNAs were identified as differentially expressed between the MCD and normal groups, and 514 mRNAs, 1182 lncRNAs and 443 circRNAs were identified as differentially expressed between the QG and MCD groups. The intersections (244 mRNAs, 259 lncRNAs and 98 circRNAs) among the three groups were chosen for analysis. Gene Ontology (GO) terms and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment revealed that most overlapping mRNAs were related to immune functions such as natural-killer-cell-mediated cytotoxicity, intestinal immune network for IgA production, and T cell receptor signaling pathway. Pathway interactions, protein-protein interactions and molecular complex detection (MCODE) analysis identified numerous immune-related hub genes e.g. natural cytotoxicity triggering receptor 1(Ncr1), C-X-C motif chemokine ligand 9 (Cxcl9), Klra1, and Cd28. Finally, two lncRNAs (Sngh1 and Slc36a3os) and four circRNAs (circ_0009029, circ_0004572, circ_0009212 and circ_0009453) in competing endogenous RNA (ceRNA) networks were constructed by Cytoscape, and immune-related mRNAs (e.g., Cd28, Cd8a, Il15, and Klrk1) were involved in the ceRNA networks. CONCLUSIONS LncRNA and circRNA-associated immune ceRNA networks might be the targets of QG in alleviating NASH, and our work may provide valuable clues for exploring the mechanisms underlying the effect of QG.
Collapse
Affiliation(s)
- Mingzhe Zhu
- Institute of Digestive Diseases, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, No.725 South Wanping Road, Shanghai, 200032 China
- School of Public Health, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203 China
| | - Meng Li
- Institute of Digestive Diseases, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, No.725 South Wanping Road, Shanghai, 200032 China
| | - Wenjun Zhou
- Institute of Digestive Diseases, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, No.725 South Wanping Road, Shanghai, 200032 China
| | - Yang Yang
- Experiment Center for Science and Technology, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203 China
| | - Fenghua Li
- Experiment Center for Science and Technology, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203 China
| | - Li Zhang
- Institute of Digestive Diseases, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, No.725 South Wanping Road, Shanghai, 200032 China
| | - Guang Ji
- Institute of Digestive Diseases, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, No.725 South Wanping Road, Shanghai, 200032 China
| |
Collapse
|
57
|
Li Y, Guo S, Liu W, Jin T, Li X, He X, Zhang X, Su H, Zhang N, Duan C. Silencing of SNHG12 Enhanced the Effectiveness of MSCs in Alleviating Ischemia/Reperfusion Injuries via the PI3K/AKT/mTOR Signaling Pathway. Front Neurosci 2019; 13:645. [PMID: 31293373 PMCID: PMC6603177 DOI: 10.3389/fnins.2019.00645] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Accepted: 06/05/2019] [Indexed: 12/30/2022] Open
Abstract
Previous studies have reported that the long non-coding RNA SNHG12 (lncRNA SNHG12) plays a critical role in regulating the function of mesenchymal stem cells (MSCs); however, the effect of lncRNA SNHG12 on MSCs in injured brain tissue has rarely been reported. We studied the effect and mechanism of lncRNA SNHG12-modified mesenchymal stem cells (MSCs) in treating brain injuries caused by ischemia/reperfusion (I/R). I/R treated rat brain microvascular endothelial cells (BMECs) were co-cultured with MSCs or I/R pretreated MSCs. Next, BMEC proliferation was detected by using CCK-8 and EdU assays, and cell apoptosis was determined by using flow cytometry and the Hoechst staining method. Autophagy of BMECs was determined using immunofluorescence and expression of associated pathway proteins were measured by western blotting. Moreover, BMEC proliferation, apoptosis, and autophagy were also determined after the BMECs had been co-cultured with shSNHG12-MSCs. In addition, a rat model of middle cerebral artery occlusion (MCAO) was used to further confirm the findings obtained with cells. I/R treatment significantly decreased the proliferation of BMECs, but increased their levels of SNHG12 expression, apoptosis, and autophagy. However, co-culturing of BMECs with MSCs markedly alleviated the reduction in BMEC proliferation and the increases in BMEC apoptosis and autophagy, as well as the phosphorylation of PI3K, AKT, and mTOR proteins in BMECs that had been induced by I/R. Furthermore, shSNHG12 remarkably enhanced the effects of MSCs. In addition, an injection MSCs reduced the infarct areas and rates of cell apoptosis in MACO rats, and reduced the phosphorylation of PI3K, AKT, and mTOR proteins. Moreover, shSNHG12 enhanced the ameliorative effect of MSCs in treating brain injuries in the MACO rats. In conclusion, silencing of SNHG12 enhanced the effects of MSCs in reducing apoptosis and autophagy of BMECs by activating the PI3K/AKT/mTOR signaling pathway.
Collapse
Affiliation(s)
- Yuanzhi Li
- The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, Department of Neurosurgery, Zhujiang Hospital, Southern Medical University, Guangzhou, China.,Department of Neurosurgery, Affiliated Hengyang Hospital, Southern Medical University (Hengyang Central Hospital), Hengyang, China
| | - Shenquan Guo
- The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, Department of Neurosurgery, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Wenchao Liu
- The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, Department of Neurosurgery, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Tao Jin
- The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, Department of Neurosurgery, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Xifeng Li
- The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, Department of Neurosurgery, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Xuying He
- The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, Department of Neurosurgery, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Xin Zhang
- The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, Department of Neurosurgery, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Hengxian Su
- The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, Department of Neurosurgery, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Nan Zhang
- The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, Department of Neurosurgery, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Chuanzhi Duan
- The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, Department of Neurosurgery, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
58
|
Zheng S, Li M, Miao K, Xu H. SNHG1 contributes to proliferation and invasion by regulating miR-382 in breast cancer. Cancer Manag Res 2019; 11:5589-5598. [PMID: 31354360 PMCID: PMC6590395 DOI: 10.2147/cmar.s198624] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Accepted: 04/22/2019] [Indexed: 12/27/2022] Open
Abstract
Introduction: Long non-coding RNAs (lncRNAs) are key regulators in multiple cancers. lncRNA, SNHG1, was shown to be associated with tumorigenesis. However, little is known about the role SNHG1 plays in breast cancer. The aim of the study was to study the role and underlying mechanism of SNHG1 regulation in breast cancer. Methods: Quantitative real-time PCR was used to measure the levels of SNHG1, miR-382 and ZEB1 levels in breast cancer tissue or cells. The proliferation, colony formation, migration and invasion of breast cancer cells, under SNHG1 knockdown achieved by transfection of SNHG1-specific siRNAs, were assessed by Cell Counting Kit-8, colony forming, scratch wound and transwell assays. Bioinformatical analysis and luciferase assay were used to explore the interaction between SNHG1 and its potential miRNA target. Western blot was used to evaluate the expression of epithelial-to-mesenchymal transition (EMT) markers. MDA-MB-231 cells with or without SNHG1 knockdown were used to initiate tumor xenografts in vivo. Tumor growth and expression of SNHG1, miR-382-5p and EMT markers were evaluated. Results: SNHG1 upregulation was observed in breast cancer tissues and cells. Knockdown of SNHG1 attenuated breast cancer proliferation, colony formation, migration and invasion. A miRNA, miR-382-5p, was identified as the target of SNHG1. A reciprocal negative regulation was found between SNHG1 and miR-382-5p. SNHG1 knockdown attenuated EMT both in vitro and in vivo. miR-382-5p transfection reversed the tumor-promoting role by SNHG1. In vivo, SNHG1 knockdown decreased breast tumor growth. Conclusion: SNHG1 promotes breast cancer through the regulation of miR-382-5p and EMT markers. Our results report SNHG1 as a novel miRNA that govern the progression of breast cancer, providing a potential new therapeutic target in breast cancer.
Collapse
Affiliation(s)
- Shipeng Zheng
- Department of Breast Surgery, the First Affiliated Hospital of Zhengzhou University, Zhengzhou City, Henan Province 450052, China
| | - Mengquan Li
- Department of Breast Surgery, the First Affiliated Hospital of Zhengzhou University, Zhengzhou City, Henan Province 450052, China
| | - Keke Miao
- Department of Breast Surgery, the First Affiliated Hospital of Zhengzhou University, Zhengzhou City, Henan Province 450052, China
| | - Han Xu
- Department of Breast Surgery, the First Affiliated Hospital of Zhengzhou University, Zhengzhou City, Henan Province 450052, China
| |
Collapse
|
59
|
Sun L, Chu H, Li H, Liu Y. LncRNA SNHG1 correlates with higher T stage and worse overall survival, and promotes cell proliferation while reduces cell apoptosis in breast cancer. Transl Cancer Res 2019; 8:603-613. [PMID: 35116793 PMCID: PMC8798092 DOI: 10.21037/tcr.2019.03.20] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2018] [Accepted: 11/29/2018] [Indexed: 12/17/2022]
Abstract
Background The aim of this study was to investigate the correlation of long non-coding RNA (lncRNA) small nucleolar RNA host gene 1 (SNHG1) with the prognosis in breast cancer patients, and its effect on breast cancer cell proliferation and apoptosis. Methods A total of 178 breast cancer patients were consecutively recruited, then tumor tissue and the paired adjacent tissue were obtained during surgery for lncRNA SNHG1 determination by quantitative polymerase chain reaction (qPCR). LncRNA SNHG1 expression was also measured in breast cancer cell lines and normal breast epithelial cell line. Subsequently, negative control (NC) overexpression plasmids, lncRNA SNHG1 overexpression plasmids, NC short hairpin RNA (shRNA) plasmids and lncRNA SNHG1 shRNA plasmids were transfected into MDA-MB-453 cells as well as MCF7 cells, and cell proliferation and apoptosis were measured afterward. Results LncRNA SNHG1 expression in tumor tissue was increased compared with paired adjacent tissue, and it correlated with higher T stage and worse overall survival (OS) in breast cancer patients. LncRNA SNHG1 expression was also elevated in breast cancer cell lines compared with normal breast epithelial cell line. Cell Counting Kit-8 (CCK8) assay revealed that lncRNA SNHG1 overexpression promoted while lncRNA SNHG1 shRNA reduced cell proliferation, and Annexin V-fluorescein isothiocyanate/propidium iodide staining (AV/PI) assay illustrated that lncRNA SNHG1 overexpression decreased while lncRNA SNHG1 shRNA increased cell apoptosis rate. In addition, Western Blot assay disclosed that lncRNA SNHG1 overexpression downregulated while lncRNA SNHG1 shRNA upregulated pro-apoptotic marker (C-Caspase3) expression, and lncRNA SNHG1 overexpression increased while lncRNA SNHG1 shRNA decreased anti-apoptotic marker (p-P38) expression. Conclusions LncRNA SNHG1 is upregulated in tumor tissue and correlates with higher T stage and worse OS, and it promotes cell proliferation but inhibits cell apoptosis in breast cancer.
Collapse
Affiliation(s)
- Lin Sun
- Department of Thyroid and Breast Surgery, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan430014, China
| | - Huimin Chu
- Department of Thyroid and Breast Surgery, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan430014, China
| | - Hai Li
- Department of Thyroid and Breast Surgery, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan430014, China
| | - Yongjun Liu
- Department of Thyroid and Breast Surgery, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan430014, China
| |
Collapse
|
60
|
Thin KZ, Tu JC, Raveendran S. Long non-coding SNHG1 in cancer. Clin Chim Acta 2019; 494:38-47. [PMID: 30849309 DOI: 10.1016/j.cca.2019.03.002] [Citation(s) in RCA: 82] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2018] [Revised: 03/02/2019] [Accepted: 03/04/2019] [Indexed: 12/17/2022]
Abstract
OBJECTIVES Long non-coding RNAs (lncRNAs) consist of a cluster of RNAs having >200 nucleotides lacking protein-coding function. Recent studies indicate that lncRNAs are involved in various cellular processes and their aberrant expression may lead to tumour development and progression. They may also serve as oncogenes or tumour suppressor genes in other diseases. In this review, we emphasize current investigations involving clinical management, tumour progression and the molecular mechanism of SNHG1 in human cancer. MATERIALS AND METHODS We investigate and summarize recent studies regarding the biologic functions and mechanisms of lncRNA SNHG1 in tumorigenesis. Related studies were obtained through a systematic search of google scholar, PubMed, Embase and Cochrane Library. RESULTS SNHG1 is a novel oncogenic lncRNA aberrantly expressed in different diseases including colorectal, liver, lung, prostate, gastric and esophageal cancers as well as ischemic stroke, nasopharyngeal carcinoma, laryngeal squamous cell carcinoma, neuroblastoma, renal cell carcinoma and osteosarcoma. Upregulation of SNHG1 was significantly associated with advanced tumour stage, tumour size, TNM stage and decreased overall survival. Furthermore, aberrant expression of SNHG1 contributes to cell proliferation, metastasis, migration and invasion of cancer cells. CONCLUSION SNHG1 likely acts as a useful tumour biomarker for cancer diagnosis, prognosis and treatment.
Collapse
Affiliation(s)
- Khaing Zar Thin
- Department of Medical Laboratory Technology, University of Medical Technology, Yankin Hill Road, 19(th) Street, Patheingyi Township, Mandalay, Myanmar; Department & Program of Clinical Laboratory Medicine, Center for Gene Diagnosis, Zhongnan Hospital of Wuhan University, Wuhan 430071, China.
| | - Jian Cheng Tu
- Department & Program of Clinical Laboratory Medicine, Center for Gene Diagnosis, Zhongnan Hospital of Wuhan University, Wuhan 430071, China.
| | - Sudheesh Raveendran
- Department of Radiology & Nuclear Medicine, Zhongnan Hospital of Wuhan University, Wuchang, Hubei province, Wuhan 430071, China.
| |
Collapse
|
61
|
Li Z, Li X, Du X, Zhang H, Wu Z, Ren K, Han X. The Interaction Between lncRNA SNHG1 and miR-140 in Regulating Growth and Tumorigenesis via the TLR4/NF-κB Pathway in Cholangiocarcinoma. Oncol Res 2019; 27:663-672. [PMID: 30764893 PMCID: PMC7848332 DOI: 10.3727/096504018x15420741307616] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Cholangiocarcinoma (CCA) is the second most common primary hepatobiliary carcinoma. The long noncoding RNA (lncRNA) small nucleolar RNA host gene 1 (SNHG1) has been reported to contribute to the progression of multiple cancers. Nonetheless, the functions and hidden mechanism of SNHG1 remain unclear in CCA. In this study, the SNHG1 levels were boosted in CCA cell lines, and knockdown of SNHG1 repressed CCA cell proliferation and invasion in vitro. The data also demonstrated that miR-140 could act as a target of SNHG1 in CCA and inhibited CCA cell proliferation and invasion, whereas the inhibition effects were relieved by overexpression of SNHG1. In addition, Toll-like receptor 4 (TLR4), an NF-κB-activating signal, was identified to be a target of miR-140. SNHG1, as a competing endogenous RNA (ceRNA) for miR-140, enhanced TLR4 expression and activated NF-κB signaling, thereby regulating growth and tumorigenesis in CCA. Animal experiments further confirmed this conclusion. Collectively, these findings not only uncovered a key role of SNHG1/miR-140/TLR4/NF-κB signaling axis in CCA tumorigenesis and progression but also denoted the probable utilization of SNHG1 as a therapeutic target for CCA.
Collapse
Affiliation(s)
- Zhen Li
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, P.R. China
| | - Xin Li
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, P.R. China
| | - Xiao Du
- Beijing Genecast Biotechnology Co., Beijing, P.R. China
| | - Henghui Zhang
- Beijing Genecast Biotechnology Co., Beijing, P.R. China
| | - Zhengyang Wu
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, P.R. China
| | - Kewei Ren
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, P.R. China
| | - Xinwei Han
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, P.R. China
| |
Collapse
|
62
|
Liu Y, Chen Y, Tan L, Zhao H, Xiao N. Linc00299/miR-490-3p/AURKA axis regulates cell growth and migration in atherosclerosis. Heart Vessels 2019; 34:1370-1380. [PMID: 30734057 DOI: 10.1007/s00380-019-01356-7] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/05/2018] [Accepted: 02/01/2019] [Indexed: 12/16/2022]
Abstract
Long non-coding RNA (lncRNA) plays a crucial role in regulating various cellular processes in atherosclerosis. The present study identified the regulation of Linc00299, via miR-490-3p targeting Aurora kinase A (AURKA), on migration and proliferation of endothelial cells and vascular smooth muscle cells (VSMCs) during atherosclerosis. The expression of RNAs was assessed by real-time PCR. The proliferation, apoptosis and migration were detected using MTT assay, Annexin V/PI staining and Transwell system, respectively. Bindings of Linc00299/miR-490-3p and subsequent miR-490-3p/AURKA were verified by luciferase and biotin pull-down assays. The protein expression of AURKA was detected by Western blotting. Expressions of Linc00299 and miR-490-3p were upregulated and downregulated in atherosclerosis patients, respectively. Both Linc00299 knockdown and miR-490-3p overexpression suppressed cell proliferation, increased apoptosis and inhibited migration of VSMCs and HUVECs. Linc00299 directly bound to miR-490-3p which targeted AURKA. The regulation of Linc00299 on expression of AURKA and proliferation and migration of VSMCs were dependent on miR-490-3p. Atherosclerosis-increased Linc00299 acts as a sponge of miR-490-3p to upregulate AURKA, and as a result increases proliferation and migration in VSMCs and HUVECs. Our study reveals an important effect of Linc00299/miR-490-3p/AURKA axis on regulating cell proliferation and migration in atherosclerosis.
Collapse
Affiliation(s)
- Yong Liu
- Affiliated Hospital of Hebei University, No 212 Yuhua East Road, Baoding, 071000, Hebei, China
| | - Yaqing Chen
- Affiliated Hospital of Hebei University, No 212 Yuhua East Road, Baoding, 071000, Hebei, China
| | - Lili Tan
- Affiliated Hospital of Hebei University, No 212 Yuhua East Road, Baoding, 071000, Hebei, China
| | - Hongmei Zhao
- Affiliated Hospital of Hebei University, No 212 Yuhua East Road, Baoding, 071000, Hebei, China
| | - Nuan Xiao
- Affiliated Hospital of Hebei University, No 212 Yuhua East Road, Baoding, 071000, Hebei, China.
| |
Collapse
|
63
|
Li H, Xue Y, Ma J, Shao L, Wang D, Zheng J, Liu X, Yang C, He Q, Ruan X, Li Z, Liu Y. SNHG1 promotes malignant biological behaviors of glioma cells via microRNA-154-5p/miR-376b-3p- FOXP2- KDM5B participating positive feedback loop. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2019; 38:59. [PMID: 30728054 PMCID: PMC6364475 DOI: 10.1186/s13046-019-1063-9] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/10/2018] [Accepted: 01/27/2019] [Indexed: 01/22/2023]
Abstract
Background Long non-coding RNAs has been reported in tumorigenesis and play important roles in regulating malignant behavior of cancers, including glioma. Methods According to the TCGA database, we identified SNHG1, miRNA-154-5p and miR-376b-3p whose expression were significantly changed in the glioma samples. Furthermore, we investigated SNHG1, miRNA-154-5p and miR-376b-3p expression in clinical samples and glioma cell lines using qRT-PCR analysis and the correlation between them using RNA immunoprecipitation and dual-luciferase reporter. The underlying mechanisms of SNHG1 in glioma were also investigated using immunohistochemistry staining, Western blotting, chromatin immunoprecipitation, and RNA pulldown. Cell Counting Kit-8, transwell assays, and flow cytometry were used to investigate malignant biological behaviors. Results We have elucidated the potential molecular mechanism of long non-coding RNA SNHG1 regulating the malignant behavior of glioma cells by binding to microRNA-154-5p or miR-376b-3p. Moreover, our deep-going results showed that FOXP2 existed as a direct downstream target of both microRNA-154-5p and miR-376b-3p; FOXP2 increased promoter activities and enhanced the expression of the oncogenic gene KDM5B; and KDM5B also acts as a RNA-binding protein to maintain the stability of SNHG1. Conclusion Collectively, this study demonstrates that the SNHG1- microRNA-154-5p/miR-376b-3p- FOXP2- KDM5B feedback loop plays a pivotal role in regulating the malignant behavior of glioma cells. Graphical abstract ![]()
Electronic supplementary material The online version of this article (10.1186/s13046-019-1063-9) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Han Li
- Department of Neurosurgery, Shengjing Hospital of China Medical University, Shenyang, Liaoning, 110004, People's Republic of China.,Liaoning Clinical Medical Research Center in Nervous System Disease, Shenyang, Liaoning, 110004, People's Republic of China.,Key Laboratory of Neuro-oncology in Liaoning Province, Shenyang, Liaoning, 110004, People's Republic of China
| | - Yixue Xue
- Department of Neurobiology, College of Basic Medicine, China Medical University, Shenyang, Liaoning, 110122, People's Republic of China.,Key Laboratory of Cell Biology, Ministry of Public Health of China, China Medical University, Shenyang, Liaoning, 110122, People's Republic of China.,Key Laboratory of Medical Cell Biology, Ministry of Education of China, China Medical University, Shenyang, Liaoning, 110122, People's Republic of China
| | - Jun Ma
- Department of Neurobiology, College of Basic Medicine, China Medical University, Shenyang, Liaoning, 110122, People's Republic of China.,Key Laboratory of Cell Biology, Ministry of Public Health of China, China Medical University, Shenyang, Liaoning, 110122, People's Republic of China.,Key Laboratory of Medical Cell Biology, Ministry of Education of China, China Medical University, Shenyang, Liaoning, 110122, People's Republic of China
| | - Lianqi Shao
- Department of Neurobiology, College of Basic Medicine, China Medical University, Shenyang, Liaoning, 110122, People's Republic of China.,Key Laboratory of Cell Biology, Ministry of Public Health of China, China Medical University, Shenyang, Liaoning, 110122, People's Republic of China.,Key Laboratory of Medical Cell Biology, Ministry of Education of China, China Medical University, Shenyang, Liaoning, 110122, People's Republic of China
| | - Di Wang
- Department of Neurosurgery, Shengjing Hospital of China Medical University, Shenyang, Liaoning, 110004, People's Republic of China.,Liaoning Clinical Medical Research Center in Nervous System Disease, Shenyang, Liaoning, 110004, People's Republic of China.,Key Laboratory of Neuro-oncology in Liaoning Province, Shenyang, Liaoning, 110004, People's Republic of China
| | - Jian Zheng
- Department of Neurosurgery, Shengjing Hospital of China Medical University, Shenyang, Liaoning, 110004, People's Republic of China.,Liaoning Clinical Medical Research Center in Nervous System Disease, Shenyang, Liaoning, 110004, People's Republic of China.,Key Laboratory of Neuro-oncology in Liaoning Province, Shenyang, Liaoning, 110004, People's Republic of China
| | - Xiaobai Liu
- Department of Neurosurgery, Shengjing Hospital of China Medical University, Shenyang, Liaoning, 110004, People's Republic of China
| | - Chunqing Yang
- Department of Neurosurgery, Shengjing Hospital of China Medical University, Shenyang, Liaoning, 110004, People's Republic of China.,Liaoning Clinical Medical Research Center in Nervous System Disease, Shenyang, Liaoning, 110004, People's Republic of China.,Key Laboratory of Neuro-oncology in Liaoning Province, Shenyang, Liaoning, 110004, People's Republic of China
| | - Qianru He
- Department of Neurobiology, College of Basic Medicine, China Medical University, Shenyang, Liaoning, 110122, People's Republic of China.,Key Laboratory of Cell Biology, Ministry of Public Health of China, China Medical University, Shenyang, Liaoning, 110122, People's Republic of China.,Key Laboratory of Medical Cell Biology, Ministry of Education of China, China Medical University, Shenyang, Liaoning, 110122, People's Republic of China
| | - Xuelei Ruan
- Department of Neurobiology, College of Basic Medicine, China Medical University, Shenyang, Liaoning, 110122, People's Republic of China.,Key Laboratory of Cell Biology, Ministry of Public Health of China, China Medical University, Shenyang, Liaoning, 110122, People's Republic of China.,Key Laboratory of Medical Cell Biology, Ministry of Education of China, China Medical University, Shenyang, Liaoning, 110122, People's Republic of China
| | - Zhen Li
- Department of Neurosurgery, Shengjing Hospital of China Medical University, Shenyang, Liaoning, 110004, People's Republic of China.,Liaoning Clinical Medical Research Center in Nervous System Disease, Shenyang, Liaoning, 110004, People's Republic of China.,Key Laboratory of Neuro-oncology in Liaoning Province, Shenyang, Liaoning, 110004, People's Republic of China
| | - Yunhui Liu
- Department of Neurosurgery, Shengjing Hospital of China Medical University, Shenyang, Liaoning, 110004, People's Republic of China. .,Liaoning Clinical Medical Research Center in Nervous System Disease, Shenyang, Liaoning, 110004, People's Republic of China. .,Key Laboratory of Neuro-oncology in Liaoning Province, Shenyang, Liaoning, 110004, People's Republic of China.
| |
Collapse
|
64
|
Guo W, Huang J, Lei P, Guo L, Li X. LncRNA SNHG1 promoted HGC-27 cell growth and migration via the miR-140/ADAM10 axis. Int J Biol Macromol 2019; 122:817-823. [DOI: 10.1016/j.ijbiomac.2018.10.214] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2017] [Revised: 10/30/2018] [Accepted: 10/30/2018] [Indexed: 02/07/2023]
|
65
|
Zhang S, Song X. Long non-coding RNA SNHG1 promotes cell proliferation and invasion of hepatocellular carcinoma by acting as a molecular sponge to modulate miR-195. Arch Med Sci 2019; 16:386-394. [PMID: 32190150 PMCID: PMC7069425 DOI: 10.5114/aoms.2019.81311] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/17/2018] [Accepted: 12/09/2018] [Indexed: 01/22/2023] Open
Abstract
INTRODUCTION Although long non-coding RNA SNHG1 (lncRNA SNHG1) action on cell proliferation and invasion of hepatocellular carcinoma (HCC) cells has been reported, the effects of lncRNA SNHG1 on migration of HCC cells and the mechanisms are still unclear. The present study aimed to investigate the influence of lncRNA SNHG1 on metastasis in HCC cells and the possible mechanisms underlying this phenotype. MATERIAL AND METHODS Expression of lncRNA SNHG1 and miR-195 was determined using qRT-PCR in both HCC cell lines Huh7 and HepG2. Si-RNA was used to silence SNHG1 and miR-195 inhibitor was used to inhibit expression of miR-195. Luciferase reporter assay was conducted to confirm whether miR-195 was the direct binding target of SNHG1. RESULTS lncRNA SNHG1 was significantly up-regulated and miR-195 was significantly down-regulated in HCC cell lines. When transfected with si-SNHG1, migration and invasion of HCC cells, as well as expression of astrocyte elevated gene 1 (AEG-1) protein, were significantly inhibited compared with the control cells. Results of dual luciferase reporter assay showed that lncRNA SNHG1 acted as an endogenous sponge of miR-195. On the other hand, the expression of miR-195 in tumor tissue was much lower than that of miR-195 in the corresponding normal tissue. Furthermore, the correlation analysis showed a strong negative relationship between lncRNA SNHG1 and miR-195 expression in HCC tissues. CONCLUSIONS SNHG1 may promote cell invasion and migration in HCC cells by sponging miR-195. These results can provide deeper understanding of SNHG1 in hepatocellular cancer and give new potential targets for treatment of HCC.
Collapse
Affiliation(s)
- Shuai Zhang
- Department of Radiation Oncology, Hainan General Hospital, Haikou, China
| | - Xiaoding Song
- Clinical Laboratory, Hainan General Hospital, Haikou, China
| |
Collapse
|
66
|
Wu M, Huang Y, Chen T, Wang W, Yang S, Ye Z, Xi X. LncRNA MEG3 inhibits the progression of prostate cancer by modulating miR-9-5p/QKI-5 axis. J Cell Mol Med 2019; 23:29-38. [PMID: 30565858 PMCID: PMC6307767 DOI: 10.1111/jcmm.13658] [Citation(s) in RCA: 123] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2017] [Accepted: 04/01/2018] [Indexed: 01/02/2023] Open
Abstract
This study was designed to detecting the influences of lncRNA MEG3 in prostate cancer. Aberrant lncRNAs expression profiles of prostate cancer were screened by microarray analysis. The qRT-PCR and Western blot were employed to investigating the expression levels of lncRNA MEG3, miR-9-5p and QKI-5. The luciferase reporter assay was utilized to testifying the interactions relationship among these molecules. Applying CCK-8 assay, wound healing assay, transwell assay and flow cytometry in turn, the cell proliferation, migration and invasion abilities as well as apoptosis were measured respectively. LncRNA MEG3 was a down-regulated lncRNA in prostate cancer tissues and cells and could inhibit the expression of miR-9-5p, whereas miR-9-5p down-regulated QKI-5 expression. Overexpressed MEG3 and QKI-5 could decrease the abilities of proliferation, migration and invasion in prostate cancer cells effectively and increased the apoptosis rate. On the contrary, miR-9-5p mimics presented an opposite tendency in prostate cancer cells. Furthermore, MEG3 inhibited tumour growth and up-regulated expression of QKI-5 in vivo. LncRNA MEG3 was a down-regulated lncRNA in prostate cancer and impacted the abilities of cell proliferation, migration and invasion, and cell apoptosis rate, this regulation relied on regulating miR-9-5p and its targeting gene QKI-5.
Collapse
Affiliation(s)
- Meng Wu
- Department of UrologyThe Second Affiliated Hospital of Nanchang UniversityNanchangJiangxiChina
| | - Yawei Huang
- Department of UrologyThe Second Affiliated Hospital of Nanchang UniversityNanchangJiangxiChina
| | - Tongchang Chen
- Department of UrologyThe Second Affiliated Hospital of Nanchang UniversityNanchangJiangxiChina
| | - Weichao Wang
- Department of UrologyThe Second Affiliated Hospital of Nanchang UniversityNanchangJiangxiChina
| | - Shiguang Yang
- Department of UrologyThe Second Affiliated Hospital of Nanchang UniversityNanchangJiangxiChina
| | - Zhenfeng Ye
- Department of UrologyThe Second Affiliated Hospital of Nanchang UniversityNanchangJiangxiChina
| | - Xiaoqing Xi
- Department of UrologyThe Second Affiliated Hospital of Nanchang UniversityNanchangJiangxiChina
| |
Collapse
|
67
|
Zhen S, Li X. Application of CRISPR-Cas9 for Long Noncoding RNA Genes in Cancer Research. Hum Gene Ther 2019; 30:3-9. [PMID: 30045635 DOI: 10.1089/hum.2018.063] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Affiliation(s)
- Shuai Zhen
- Center for Translational Medicine, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
- Key Laboratory for Tumor Precision Medicine of Shaanxi Province, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Xu Li
- Center for Translational Medicine, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
- Key Laboratory for Tumor Precision Medicine of Shaanxi Province, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| |
Collapse
|
68
|
MiR-199a-3p inhibits proliferation and induces apoptosis in rheumatoid arthritis fibroblast-like synoviocytes via suppressing retinoblastoma 1. Biosci Rep 2018; 38:BSR20180982. [PMID: 30352835 PMCID: PMC6239273 DOI: 10.1042/bsr20180982] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2018] [Revised: 09/14/2018] [Accepted: 10/09/2018] [Indexed: 01/04/2023] Open
Abstract
Background Fibroblast-like synoviocytes (FLSs) that line the intimal synovium play a crucial role in the pathogenesis of rheumatoid arthritis (RA). miR-199a-3p is a highly conserved miRNA that has been shown to regulate a variety of growth behaviors in diverse cell types. However, the role of miR-199a-3p in RA-FLS is still unknown. Methods Here, we presented the first experimental evidence showing that miR-199a-3p was a critical regulator of RA-FLS function. Results miR-199a-3p expression was significantly reduced in RA-FLS compared with normal FLS. Ectopic expression of miR-199a-3p significantly inhibited RA-FLS proliferation and induced apoptosis, which was demonstrated by an increase in caspase-3 activity and Bax/Bcl-2 ratio. Our bioinformatics analysis identified Retinoblastoma 1 (RB1) gene to be a direct target of miR-199a-3p. In RA-FLS, miR-199a-3p directly targetted the 3′-UTR of RB1 mRNA and suppressed endogenous RB1 expression, whereas miR-199a-3p-resistant variant of RB1 was not affected. Silencing RB1 decreased cell proliferation and promoted apoptosis in RA-FLS, an effect comparable with miR-199a-3p overexpression. Enforced expression of RB1 partially restored cell proliferation and attenuated apoptosis in miR-199a-3p-overexpressing RA-FLSs. Conclusion In summary, miR-199a-3p is down-regulated in RA-FLS, and miR-199a-3p inhibits proliferation and induces apoptosis in RA-FLS, partially via targetting RB1. The miR-199a-3p/RB1 pathway may represent a new therapeutic target for RA.
Collapse
|
69
|
Yang X, Zi XH. LncRNA SNHG1 alleviates OGD induced injury in BMEC via miR-338/HIF-1α axis. Brain Res 2018; 1714:174-181. [PMID: 30414401 DOI: 10.1016/j.brainres.2018.11.003] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2018] [Revised: 11/02/2018] [Accepted: 11/03/2018] [Indexed: 02/07/2023]
Abstract
BACKGROUND Brain microvascular endothelial cell (BMEC) is an important therapeutic target for the inhibition of brain vascular dysfunction in ischemic stroke. Expression of long non-coding RNA SNHG1 is reportedly upregulated in BMEC after OGD. The present study aims to investigate the potential roles of SNHG1 in OGD-induced injury in BMEC. METHODS Mice primary brain microvascular endothelial cells (BMEC) were cultured under "normal" or "oxygen/glucose-deprived" (OGD) conditions. The expression of SNHG1 and miR-338 after OGD were examined by qPCR. shRNA against SNHG1 was used to knockdown SNHG1 in BMEC. MiR-338-3p mimic and inhibitor were used to change the expression of miR-338 in BMEC. The relationship between SNHG1 and miR-338, and the relationship between miR-338 and HIF-1α were clarified using RNA pull-down and luciferase reporter gene assays, respectively. RESULTS SNHG1 and miR-338 were upregulated in OGD induced BMEC. SNHG1 silence aggravated OGD-induced cell apoptosis by down-regulating Bcl-2, HIF-1α and VEGF-A, and upregulating caspase 3 activity and Bax. MiR-338 was upregulated in SNHG1-silenced BMEC. RNA pull-down assays showed that SNHG1 could be directly bound by miR-338. In addition, miR-338 overexpression reduced cell viability in OGD while miR-338 inhibition protected BMEC against OGD-induced injury. Furthermore, luciferase reporter assay showed that HIF-1α was a direct target of miR-338. CONCLUSIONS SNHG1 exerted protective effects against OGD induced injury via sponging miR-338, thus upregulating HIF-1α/VEGF-A in BMEC.
Collapse
Affiliation(s)
- Xia Yang
- Department of Neurology, The Third Xiangya Hospital of Central South University, Changsha 410013, PR China
| | - Xiao-Hong Zi
- Department of Neurology, The Third Xiangya Hospital of Central South University, Changsha 410013, PR China.
| |
Collapse
|
70
|
Chen Y, Gu M, Liu C, Wan X, Shi Q, Chen Q, Wang Z. Long noncoding RNA FOXC2-AS1 facilitates the proliferation and progression of prostate cancer via targeting miR-1253/EZH2. Gene 2018; 686:37-42. [PMID: 30389560 DOI: 10.1016/j.gene.2018.10.085] [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] [Received: 08/12/2018] [Revised: 10/17/2018] [Accepted: 10/29/2018] [Indexed: 12/11/2022]
Abstract
The vital roles of long noncoding RNAs (lncRNAs) in the cancers have been evidenced. However, there are still numerous unsolved queries for the molecular mechanism. This study tries to investigate the role of lncRNA FOXC2-AS1 in the human prostate cancer tumorigenesis. Results stated that lncRNA FOXC2-AS1 was ectopically up-regulated in prostate cancer tissue and cells. The over-expression of FOXC2-AS1 indicates the poor prognosis of prostate cancer patients. Functionally, the gain- and loss-of-functional experiments revealed that FOXC2-AS1 promoted the proliferation and tumor growth of prostate cancer cells in vitro and in vivo. Mechanically, we found that miR-1253 targeted FOXC2-AS1 at the 3'‑untranslated regions (UTR), which in turn bind the EZH2 mRNA 3-UTR. Luciferase reporter assay and rescue experiment confirmed the FOXC2-AS1/miR-1253/EZH2 pathway. In conclusion, we confirmed that lncRNA FOXC2-AS1 accelerated the tumor progression of prostate cancer cells by regulating the proliferation and tumor growth through miR-1253/EZH2 axis.
Collapse
Affiliation(s)
- Yanbo Chen
- Department of Urology, Shanghai Ninth People's Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Meng Gu
- Department of Urology, Shanghai Ninth People's Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Chong Liu
- Department of Urology, Shanghai Ninth People's Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Xiang Wan
- Department of Urology, Shanghai Ninth People's Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Qiling Shi
- Department of Urology, Shanghai Ninth People's Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Qi Chen
- Department of Urology, Shanghai Ninth People's Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, China.
| | - Zhong Wang
- Department of Urology, Shanghai Ninth People's Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, China.
| |
Collapse
|
71
|
Wang F, Liang S, Liu X, Han L, Wang J, Du Q. LINC00460 modulates KDM2A to promote cell proliferation and migration by targeting miR-342-3p in gastric cancer. Onco Targets Ther 2018; 11:6383-6394. [PMID: 30323616 PMCID: PMC6174301 DOI: 10.2147/ott.s169307] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Background Increasing evidence has shown that long non-coding RNAs (lncRNAs) play important roles in the occurrence and development of human cancers. LINC00460, a novel tumor-related lncRNA, has been reported to be involved in several types of human malignancies. However, the role of LINC00460 in gastric cancer (GC) is still unclear. The present study aimed at exploring the biological role of LINC00460 in GC and illuminating the potential molecular mechanisms. Methods In this study, qRT-PCR, western blotting, MTT assay, and Transwell invasion assay were used to conduct relevant experimental analysis. Results Here, we found that LINC00460 was highly expressed in GC tissues and cell lines. Moreover, LINC00460 overexpression was found to promote GC cell proliferation, migration and invasion, whereas LINC00460 down-regulation significantly inhibited these processes. Notably, we confirmed that LINC00460 could up-regulate KDM2A expression by competitively binding to miR-342-3p in GC cells. Furthermore, the suppressive effects of LINC00460 down-regulation on GC cell proliferation, migration and invasion were partially reversed by a miR-342-3p inhibitor. Conclusion In summary, our findings provide evidence for LINC00460 as a potential therapeutic target in GC.
Collapse
Affiliation(s)
- Fang Wang
- Department of Oncology, Affiliated Hospital of Jining Medical College, Jining 272029, China,
| | - Shaobo Liang
- Department of Gastroenterology, Central Hospital of Shanxian County, Heze, 274399, China
| | - Xiaowei Liu
- Department of Oncology, Affiliated Hospital of Jining Medical College, Jining 272029, China,
| | - Lei Han
- Department of Oncology, Affiliated Hospital of Jining Medical College, Jining 272029, China,
| | - Junye Wang
- Department of Oncology, Affiliated Hospital of Jining Medical College, Jining 272029, China,
| | - Qin Du
- Department of Oncology, Affiliated Hospital of Jining Medical College, Jining 272029, China,
| |
Collapse
|
72
|
Wang O, Huang Y, Wu H, Zheng B, Lin J, Jin P. LncRNA LOC728196/miR-513c axis facilitates glioma carcinogenesis by targeting TCF7. Gene 2018; 679:119-125. [PMID: 30179681 DOI: 10.1016/j.gene.2018.08.081] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2018] [Revised: 08/15/2018] [Accepted: 08/30/2018] [Indexed: 01/17/2023]
Abstract
Numerous long noncoding RNAs (lncRNAs) are reported to be dysregulated in glioma. However, how lncRNA participates in the process of glioma development and progression still remains elusive. Here, we identified a novel lncRNA LOC728196 highly expressed in glioma tissues. LOC728196 high expression predicts low survival rate in patients. Our data proved that LOC728196 knockdown repressed cellular growth, migration and invasion in vitro. Silencing LOC728196 led to impaired growth of glioma in vivo. Mechanistic studies further demonstrated that LOC728196 acts as the sponge for miR-513c to upregulate TCF7 expression. We observed a reciprocal inhibition between LOC728196 and miR-513c. Rescue assay showed that either inhibition of miR-513c or TCF7 overexpression restored the abilities of proliferation, migration and invasion in LOC728196-silenced glioma cells. Taken together, our study provides a comprehensive investigation on the role of LOC728196 in glioma progression and contributes to understanding the vital role of competing endogenous RNA (ceRNA).
Collapse
Affiliation(s)
- Ouyang Wang
- Department of Neurosurgery, Wenzhou People's Hospital, Wenzhou 325000, China
| | - Yuenuo Huang
- Department of Respiratory, Wenzhou Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medicine University, Wenzhou 325000, China
| | - Hao Wu
- Department of Neurosurgery, Wenzhou People's Hospital, Wenzhou 325000, China
| | - Buyi Zheng
- Department of Neurosurgery, Wenzhou People's Hospital, Wenzhou 325000, China
| | - Jie Lin
- Department of Neurosurgery, Wenzhou People's Hospital, Wenzhou 325000, China
| | - Pengcheng Jin
- Department of Neurosurgery, Wenzhou People's Hospital, Wenzhou 325000, China.
| |
Collapse
|
73
|
Small nucleolar RNA host gene 1: A new biomarker and therapeutic target for cancers. Pathol Res Pract 2018; 214:1247-1252. [PMID: 30107989 DOI: 10.1016/j.prp.2018.07.033] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2018] [Revised: 07/28/2018] [Accepted: 07/28/2018] [Indexed: 01/05/2023]
Abstract
OBJECTIVES Long non-coding RNAs (lncRNAs), a group of transcripts with length greater than 200 nucleotides, have been involved in multiple pathophysiological processes of the human body, especially in tumorigenesis and progression of cancers. The aberrant expression of lncRNAs processes crucial functions involved in proliferation, apoptosis and metastatic capacity of cancers. Recent studies have revealed that small nucleolar RNA host gene 1 (SNHG1), a long non-coding RNA transcribed from UHG, was located in chromosome 11. Aberrant expression of SNHG1 has been demonstrated to be associated with various sites of cancers such as glioma, esophageal cancer, gastric cancer and many others, and its deregulation could be related to survival and prognosis of cancer patients. Pertinent to clinical practice, SNHG1 might act as a prognostic biomarker for tumors and might even serve as potential target for therapy. In this review, we summarized current researches concerning the role of SNHG1 in tumor progression and discussed its mechanisms involved. MATERIALS AND METHODS In this review, we summarized and figured out recent studies concerning the expression and biological mechanisms of SNHG1in tumor development. The related studies were obtained through a systematic search of PubMed, Embase and Cochrane Library. RESULTS SNHG1 was a valuable cancer-related lncRNA that the expression level was up-regulation in a variety of malignancies, including glioma, esophageal cancer, lung cancer, gastric cancer, hepatocellular carcinoma, colorectal carcinoma, prostate cancer, cervical cancer, osteosarcoma, neuroblastoma, nasopharyngeal carcinoma. The aberrant expressions of SNHG1 have shown to contribute to proliferation, migration, and invasion of cancer cells. CONCLUSIONS SNHG1 represents promising novel biomarkers for various cancer types and have a great potential to be effectively used in clinical practice in the near future.
Collapse
|
74
|
Long Noncoding RNA SNHG1 Promotes Neuroinflammation in Parkinson's Disease via Regulating miR-7/NLRP3 Pathway. Neuroscience 2018; 388:118-127. [PMID: 30031125 DOI: 10.1016/j.neuroscience.2018.07.019] [Citation(s) in RCA: 131] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2018] [Revised: 07/08/2018] [Accepted: 07/11/2018] [Indexed: 12/19/2022]
Abstract
Parkinson's disease (PD) is the second most common neurodegenerative disorders. Neuroinflammation plays an important role in the pathogenesis of PD. Long noncoding RNA small nucleolar RNA host gene 1 (SNHG1) was elevated in the brain specimens of PD patients and MPP+-treated SH-SY5Y cells. The expression of mouse Snhg1 and miR-7 was firstly determined in lipopolysaccharide (LPS)-induced BV2 cells. The role and mechanism of SNHG1 in the neuroinflammation of PD were investigated using gain- and loss-of function approaches both in vitro and in vivo. Snhg1 expression was elevated, whereas miR-7 reduced in LPS-induced BV2 cells. Upregulation of Snhg1 elevated, and Snhg1 knockdown suppressed LPS-induced BV2 microglial activation and inflammation. miR-7 reversed, while anti-miR-7 further enhanced the effects of Snhg1 on BV2 cells. Furthermore, we found that Snhg1 functioned as a competing endogenous RNA for miR-7 to regulate nod-like receptor protein 3 (NLRP3) expression, leading to the activation of NLRP3 inflammasome. In the microglial culture supernatant transfer model, knockdown of Snhg1 or NLRP3 in LPS-stimulated BV2 cells inhibited primary neurons from apoptosis and elevated caspase-3 activity. Additionally, Snhg1 was increased in MPTP-induced PD mouse models. Downregulation of Snhg1 elevated miR-7 expression, suppressed the activation of microglia and NLRP3 inflammasome as well as dopaminergic neuron loss in the midbrain substantia nigra pars compacta in MPTP-treated mice. In conclusion, our study suggests that SNHG1 promotes neuroinflammation in the pathogenesis of PD via modulating miR-7/NLRP3 pathway.
Collapse
|
75
|
Tan H, Zhao L, Song R, Liu Y, Wang L. The long noncoding RNA SNHG1 promotes nucleus pulposus cell proliferation through regulating miR-326 and CCND1. Am J Physiol Cell Physiol 2018; 315:C21-C27. [PMID: 29466672 DOI: 10.1152/ajpcell.00220.2017] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Aberrant nucleus pulposus cell proliferation is implicated in the development of intervertebral disk degeneration (IDD). Recent studies have suggested that long noncoding RNAs (lncRNAs) can modulate cell proliferation in several pathological conditions. Here, we indicate that expression of SNHG1 was upregulated in IDD tissues compared with control tissues and that higher SNHG1 expression was associated with disk degeneration grade. In addition, we show that ectopic expression of SNHG1 promoted nucleus pulposus (NP) cell proliferation and increased the PCNA and cyclin D1 expression in NP cells. Ectopic expression of SNHG1 inhibited miR-326 expression in nucleus pulposus cells and promoted CCND1 expression, which is a direct target gene of SNHG1. Moreover, we demonstrate that expression of miR-326 was downregulated in IDD tissues compared with control tissues and that lower SNHG1 expression was associated with disk degeneration grade. Expression of miR-326 was negatively associated with SNHG1 expression in disk degeneration tissues. Overexpression of miR-326 inhibited NP cell growth and inhibited PCNA and cyclin D1 expression in NP cells. Furthermore, we show that overexpression of SNHG1 promoted nucleus pulposus cell proliferation through inhibiting miR-326 expression. These data shed novel light on the role of SNHG1 in the pathogenesis of IDD.
Collapse
Affiliation(s)
- Hongyu Tan
- Department of Orthopedics, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Liang Zhao
- Department of Orthopedics, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Ruipeng Song
- Department of Orthopedics, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yilin Liu
- Department of Orthopedics, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Limin Wang
- Department of Orthopedics, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
76
|
Zhang Y, Zhang R, Luo G, Ai K. Long noncoding RNA SNHG1 promotes cell proliferation through PI3K/AKT signaling pathway in pancreatic ductal adenocarcinoma. J Cancer 2018; 9:2713-2722. [PMID: 30087712 PMCID: PMC6072808 DOI: 10.7150/jca.26207] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2018] [Accepted: 06/02/2018] [Indexed: 01/02/2023] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) remains one of the most common causes of cancer-related death. Recently, long noncoding RNAs (lncRNAs) have emerged as significant regulators in numerous cancers, including PDAC. LncRNA small nucleolar RNA host gene 1 (SNHG1) has been reported in the development of several tumors, but the biological roles of it in PDAC remain to be illuminated. This study aims to investigate the function of lncRNA SNHG1, revealing its molecular mechanism and clinical significance in PDAC. Herein, we found that SNHG1 was highly expressed in PDAC tissues in comparison with adjacent noncancerous tissues, being closely related to tumor size and TNM stage. Functionally, silencing of SNHG1 could significantly inhibit cell proliferation, promote cell apoptosis, as well as alter cell cycle progression, whereas the contrary results could be presented in the overexpression of SNHG1. In addition, in vivo xenograft experiment also further confirmed the above results. Finally, an activator (740Y-P) and inhibitor (LY294002) of the PI3K/AKT signaling pathway were used in the western blot assays and the following rescue experiments, demonstrating that SNHG1 facilitates cell proliferation and tumorigenicity partly via the PI3K/AKT signaling pathway in PDAC. Hence, SNHG1 may be a prospective therapeutic target.
Collapse
Affiliation(s)
- Yalu Zhang
- Department of General Surgery, Tongji hospital, Tongji University School of Medicine, Shanghai 200065, China
| | - Rundong Zhang
- Department of General Surgery, Tongji hospital, Tongji University School of Medicine, Shanghai 200065, China
| | - Guopei Luo
- Department of Pancreas and Hepatobiliary Surgery, Pancreas Cancer Institute, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Kaixing Ai
- Department of General Surgery, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai 200433, China
| |
Collapse
|
77
|
Zhao S, Wang Y, Luo M, Cui W, Zhou X, Miao L. Long Noncoding RNA Small Nucleolar RNA Host Gene 1 (SNHG1) Promotes Renal Cell Carcinoma Progression and Metastasis by Negatively Regulating miR-137. Med Sci Monit 2018; 24:3824-3831. [PMID: 29874202 PMCID: PMC6018379 DOI: 10.12659/msm.910866] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Background Data on the expression of RCC tissues from the GEO database and patient survival data from TCGA were used to explore the prognostic significance of long noncoding RNA SNHG1. SNHG1 has been reported to participate in the development of several cancers, but, the underlying mechanism of SNHG1 in renal cell carcinoma (RCC) has not been reported. The purpose of our study was to investigate the potential function of SNHG1 in RCC. Material/Methods The expression of SNHG1 in 40 cases of RCC and adjacent normal tissues and 5 cell lines was detected by qRT-PCR. Cell proliferation, Transwell assay, and Western blotting assay were carried out to investigate the biological function of SNHG1. A rescue experiment was performed to verify that miR-137 can partly impede the effect of SNHG1 on renal cancer cells. Results SNHG1 was identified to be overexpressed in RCC tissues and RCC cell lines. High levels of SNHG1 were correlated with poor prognosis of RCC patients. Knockdown of SNHG1 suppressed the proliferation, invasion, and EMT capacity in RCC. Moreover, miR-137 abrogated the effect of SNHG1 on RCC. Conclusions SNHG1 is significantly upregulated in RCC and renal cancer cell lines. Overexpression of SNHG1 participates in RCC tumorigenesis by regulating miR-137.
Collapse
Affiliation(s)
- Shiyue Zhao
- Department of Nephrology, Second Hospital of Jilin University, Changchun, Jilin, China (mainland)
| | - Yangwei Wang
- Department of Nephrology, Second Hospital of Jilin University, Changchun, Jilin, China (mainland)
| | - Manyu Luo
- Department of Nephrology, Second Hospital of Jilin University, Changchun, Jilin, China (mainland)
| | - Wenpeng Cui
- Department of Nephrology, Second Hospital of Jilin University, Changchun, Jilin, China (mainland)
| | - Xiaoxi Zhou
- Department of Nephrology, Second Hospital of Jilin University, Changchun, Jilin, China (mainland)
| | - Lining Miao
- Department of Nephrology, Second Hospital of Jilin University, Changchun, Jilin, China (mainland)
| |
Collapse
|
78
|
Yang H, Wang S, Kang YJ, Wang C, Xu Y, Zhang Y, Jiang Z. Long non-coding RNA SNHG1 predicts a poor prognosis and promotes colon cancer tumorigenesis. Oncol Rep 2018; 40:261-271. [PMID: 29749530 PMCID: PMC6059747 DOI: 10.3892/or.2018.6412] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2017] [Accepted: 04/23/2018] [Indexed: 02/06/2023] Open
Abstract
Colon cancer is the main cause of cancer mortality worldwide. Its poor prognosis is mainly ascribed to high recurrence rates. Identifying novel prognostic biomarkers and therapeutic key points for management is crucial and important. Long non-coding RNAs (lncRNAs) are a class of RNAs, which have various roles in carcinogenicity and molecular mechanisms. The lncRNA small nucleolar RNA host gene 1 (SNHG1) contributes to the promotion of tumor development, however, the connections between SNHG1 and colon cancer are still unclear. The aim of the present study was to investigate the clinical significance, the biological functions, and the potential mechanism of SNHG1 in colon cancer. In the present study, we referred to the Oncomine database and used RT-qPCR to determine that SNHG1 expression was significantly higher both in colon cancer tissues and cancerous cell lines than in normal samples. Cell functional experiments were performed after knockdown of SNHG1, including Cell Counting Kit-8 assay, colony formation assay, Transwell® assay, and flow cytometric analyses of cell apoptosis, which suggested that SNHG1 stimulated colon cancer cell proliferation, promoted cell invasion and migration, and inhibited apoptosis. Immunohistochemical staining and western blotting experiments revealed that in colon cancer cells with SNHG1 knockdown, β-catenin, c-Myc and cyclin D1 protein levels were decreased, while E-cadherin was increased, which suggested that SNHG1 promoted colon cancer cell proliferation, migration and invasion through the Wnt/β-catenin signaling pathway. Our results indicated that SNHG1 and its interrelated components may be future therapeutic targets of carcinoma of the colon.
Collapse
Affiliation(s)
- Huan Yang
- Department of Gastroenterology, The First Affiliated Hospital, Chongqing Medical University, Chongqing 400016, P.R. China
| | - Shuang Wang
- Department of Gastroenterology, Chongqing People's Hospital, Chongqing 400016, P.R. China
| | - Yu-Jun Kang
- Department of Gastroenterology, Chongqing People's Hospital, Chongqing 400016, P.R. China
| | - Chuan Wang
- Department of Gastroenterology, Chongqing People's Hospital, Chongqing 400016, P.R. China
| | - Yongzhu Xu
- Chongqing Health Service Center, Chongqing 400000, P.R. China
| | - Yi Zhang
- Key Laboratory of Birth Defects and Reproductive Health, Chongqing Population and Family Planning of Chongqing Science and Technology Research Institute, Chongqing 400000, P.R. China
| | - Zheng Jiang
- Department of Gastroenterology, The First Affiliated Hospital, Chongqing Medical University, Chongqing 400016, P.R. China
| |
Collapse
|
79
|
Feeley KP, Edmonds MD. Hiding in Plain Sight: Rediscovering the Importance of Noncoding RNA in Human Malignancy. Cancer Res 2018; 78:2149-2158. [PMID: 29632135 DOI: 10.1158/0008-5472.can-17-2675] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2017] [Revised: 02/12/2018] [Accepted: 02/21/2018] [Indexed: 12/22/2022]
Abstract
At the time of its construction in the 1950s, the central dogma of molecular biology was a useful model that represented the current state of knowledge for the flow of genetic information after a period of prolific scientific discovery. Unknowingly, it also biased many of our assumptions going forward. Whether intentional or not, genomic elements not fitting into this paradigm were deemed unimportant and emphasis on the study of protein-coding genes prevailed for decades. The phrase "Junk DNA," first popularized in the 1960s, is still used with alarming frequency to describe the entirety of noncoding DNA. It has since become apparent that RNA molecules not coding for protein are vitally important in both normal development and human malignancy. Cancer researchers have been pioneers in determining noncoding RNA function and developing new technologies to study these molecules. In this review, we will discuss well known and newly emerging species of noncoding RNAs, their functions in cancer, and new technologies being utilized to understand their mechanisms of action in cancer. Cancer Res; 78(9); 2149-58. ©2018 AACR.
Collapse
Affiliation(s)
- Kyle P Feeley
- Department of Genetics, University of Alabama at Birmingham, Birmingham, Alabama
| | - Mick D Edmonds
- Department of Genetics, University of Alabama at Birmingham, Birmingham, Alabama.
| |
Collapse
|
80
|
Lan X, Liu X. LncRNA SNHG1 functions as a ceRNA to antagonize the effect of miR-145a-5p on the down-regulation of NUAK1 in nasopharyngeal carcinoma cell. J Cell Mol Med 2018; 23:2351-2361. [PMID: 29575772 PMCID: PMC6434074 DOI: 10.1111/jcmm.13497] [Citation(s) in RCA: 63] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2017] [Accepted: 11/15/2017] [Indexed: 12/24/2022] Open
Abstract
How lncRNA SNHG1 influences the aggressiveness of nasopharyngeal carcinoma cells as well as the underlying mechanism was studied. The lncRNA differences were analysed by GSE12452 gene microarray. The expression of SNHG1, MiR‐145‐5p and NUAK1 was identified by qRT‐PCR and western blot. Transfection was conducted to construct nasopharyngeal carcinoma cells with different expressions of SNHG1, miR‐145‐5p and NUAK1. Dual‐luciferase reporter assay was performed to explore the relationship between SNHG1, miR‐145‐5p and NUAK1. Wound‐healing assay and transwell invasion experiments were employed to study changes in cell migration capacity and cell invasion, respectively. Tumour xenografts were performed to observe lung metastasis of nude mice inoculated with transfected CNE cells. SNHG1 is highly expressed in nasopharyngeal carcinoma tissues and in cell lines. Down‐regulation of SNHG1 facilitated the expression of miR‐145‐5p and further suppressed the level of NAUK1 in CNE and HNE‐1 cells. Silencing of SNHG1, up‐regulation of miR‐145‐5p and inhibition of NAUK1 by relative transfection all attenuated the aggressiveness of CNE and HNE‐1 cells both in vivo and in vitro. Moreover, the impaired cell migration and invasion by SNHG1 siRNA could be rescued by cotransfection of miR‐145‐5p in CNE and HNE‐1 cells. LncRNA SNHG1 promoted the expression of NUAK1 by down‐regulating miR‐145‐5p and thus promoted the aggressiveness of nasopharyngeal carcinoma cells through AKT signalling pathway and induced epithelial‐mesenchymal transition (EMT).
Collapse
Affiliation(s)
- Xintang Lan
- Department of Otolaryngology Head and Neck Surgery, Weihai Municipal Hospital, Weihai, China
| | - Xiuling Liu
- Department of Otolaryngology Head and Neck Surgery, Weihai Municipal Hospital, Weihai, China
| |
Collapse
|
81
|
Aird J, Baird AM, Lim MC, McDermott R, Finn SP, Gray SG. Carcinogenesis in prostate cancer: The role of long non-coding RNAs. Noncoding RNA Res 2018; 3:29-38. [PMID: 30159437 PMCID: PMC6084828 DOI: 10.1016/j.ncrna.2018.01.001] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2017] [Accepted: 01/16/2018] [Indexed: 12/28/2022] Open
Abstract
LncRNAs appear to play a considerable role in tumourigenesis through regulating key processes in cancer cells such as proliferative signalling, replicative immortality, invasion and metastasis, evasion of growth suppressors, induction of angiogenesis and resistance to apoptosis. LncRNAs have been reported to play a role in prostate cancer, particularly in regulating the androgen receptor signalling pathway. In this review article, we summarise the role of 34 lncRNAs in prostate cancer with a particular focus on their role in the androgen receptor signalling pathway and the epithelial to mesenchymal transition pathway.
Collapse
Affiliation(s)
- John Aird
- Department of Histopathology and Morbid Anatomy, School of Medicine, Trinity College Dublin, Ireland
| | - Anne-Marie Baird
- Department of Histopathology and Morbid Anatomy, School of Medicine, Trinity College Dublin, Ireland
- Thoracic Oncology Research Group, Trinity Translational Medical Institute, St. James's Hospital, Dublin, Ireland
- Department of Clinical Medicine, Trinity College Dublin, Dublin, Ireland
- Cancer and Ageing Research Program, Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, QLD, Australia
| | - Marvin C.J. Lim
- Department of Histopathology and Morbid Anatomy, School of Medicine, Trinity College Dublin, Ireland
- Department of Medical Oncology, St. Vincent's University Hospital, Dublin, Ireland
- Department of Medical Oncology, Tallaght Hospital, Dublin, Ireland
| | - Ray McDermott
- Department of Medical Oncology, St. Vincent's University Hospital, Dublin, Ireland
- Department of Medical Oncology, Tallaght Hospital, Dublin, Ireland
| | - Stephen P. Finn
- Department of Histopathology and Morbid Anatomy, School of Medicine, Trinity College Dublin, Ireland
- Department of Clinical Medicine, Trinity College Dublin, Dublin, Ireland
| | - Steven G. Gray
- Department of Clinical Medicine, Trinity College Dublin, Dublin, Ireland
- HOPE Directorate, St. James's Hospital, Dublin, Ireland
- Labmed Directorate, St. James's Hospital, Dublin, Ireland
- School of Biological Sciences, Dublin Institute of Technology, Dublin, Ireland
| |
Collapse
|
82
|
Lu Q, Shan S, Li Y, Zhu D, Jin W, Ren T. Long noncoding RNA SNHG1 promotes non-small cell lung cancer progression by up-regulating MTDH via sponging miR-145-5p. FASEB J 2018; 32:3957-3967. [PMID: 29466052 DOI: 10.1096/fj.201701237rr] [Citation(s) in RCA: 118] [Impact Index Per Article: 16.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Long noncoding RNAs participate in the progression and initiation of non-small cell lung cancer (NSCLC), although the mechanism remains unknown. The lncRNA identified as small nucleolar RNA host gene 1 ( SNHG1) is a novel lncRNA that is increased in multiple human cancers; however, the regulatory mechanism requires further investigation. In this study, we discovered that SNHG1 was markedly up-regulated in NSCLC tissues and cells and that SNHG1 silencing decreased tumor volumes. Moreover, we explored its regulatory mechanism and found that SNHG1 directly bound to microRNA (miRNA)-145-5p, isolating miR-145-5p from its target gene MTDH. Inhibition of SNHG1 suppressed NSCLC cell viability, proliferation, migration, and invasion in vitro, but its effect was rescued by miR-145-5p inhibition. These results demonstrate that SNHG1 contributes to NSCLC progression by modulating the miR-145-5p/ MTDH axis, and it could potentially be a therapeutic target as well as a diagnostic marker.-Lu, Q., Shan, S., Li, Y., Zhu, D., Jin, W., Ren, T. Long noncoding RNA SNHG1 promotes non-small cell lung cancer progression by up-regulating MTDH via sponging miR-145-5p.
Collapse
Affiliation(s)
- Qingchun Lu
- Department of Respiratory Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Shan Shan
- Department of Respiratory Medicine, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Yanyan Li
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Dongyi Zhu
- Department of Respiratory Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Wenjing Jin
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China; and.,Department of Intensive Care Unit, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, China
| | - Tao Ren
- Department of Respiratory Medicine, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| |
Collapse
|
83
|
Zhao J, Zhang L, Zheng L, Hong Y, Zhao L. LncRNATCF7 promotes the growth and self-renewal of glioma cells via suppressing the miR-200c-EpCAM axis. Biomed Pharmacother 2018; 97:203-208. [DOI: 10.1016/j.biopha.2017.10.039] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2017] [Revised: 10/08/2017] [Accepted: 10/09/2017] [Indexed: 12/11/2022] Open
|
84
|
SNHG1 promotes cell proliferation by acting as a sponge of miR-145 in colorectal cancer. Oncotarget 2017; 9:2128-2139. [PMID: 29416759 PMCID: PMC5788627 DOI: 10.18632/oncotarget.23255] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2017] [Accepted: 12/05/2017] [Indexed: 02/07/2023] Open
Abstract
ncRNAs are important regulatory molecules and involve in many physiological cellular processes. Small nucleolar RNA host gene 1 (SNHG1) is a host to 8 snoRNAs and is located in 11q12.3 region of the chromosome. It has been reported to be involved in several cancers. However, the role of SNHG1 in the tumorigenesis of colorectal cancer is still unknown. In this study, SNHG1 was upregulated in colorectal cancers, and SNHG1 expression was correlated with advanced colorectal cancer stage and tumor recurrence. We found that SNHG1 promoted cell proliferation by acting as a sponge of miR-145, a well known tumor suppressor of colorectal cancer. Furthermore, the survival analysis indicated that colorectal cancer patients with higher expression of SNHG1 had a worse prognosis. These findings suggested that SNHG1 may act as a potential therapeutic target for the treatment of colorectal cancer.
Collapse
|
85
|
Chen Y, Lian YJ, Ma YQ, Wu CJ, Zheng YK, Xie NC. LncRNA SNHG1 promotes α-synuclein aggregation and toxicity by targeting miR-15b-5p to activate SIAH1 in human neuroblastoma SH-SY5Y cells. Neurotoxicology 2017; 68:212-221. [PMID: 29217406 DOI: 10.1016/j.neuro.2017.12.001] [Citation(s) in RCA: 81] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2017] [Revised: 11/23/2017] [Accepted: 12/03/2017] [Indexed: 02/07/2023]
Abstract
Numerous long non-coding RNAs (lncRNAs) have been identified as aberrantly expressed in Parkinson's disease (PD). However, limited knowledge is available concerning the roles of dysregulated lncRNAs and the underlying molecular regulatory mechanism in the pathological process of PD. In this study, we found that lncRNA small nucleolar RNA host gene 1 (SNHG1) and seven in absentia homolog 1 (SIAH1) were upregulated, but microRNA-15b-5p (miR-15b-5p) was downregulated in SH-SY5Y cells pretreated with MPP+, as well as in MPTP-induced mouse model of PD. Overexpression of SIAH1 enhanced cellular toxicity of α-synuclein in SH-SY5Y cells, as indicated by the reduction of cell viability and elevation of LDH release. The percentage of α-synuclein aggregate-positive cells and the number of α-synuclein aggregates per cell were increased in SH-SY5Y cells transfected with pcDNA-SIAH1, while decreased after transfection with short interfering RNA specific for SIAH1 (si-SIAH1). Bioinformatics and luciferase reporter assay revealed that SIAH1 was a direct target of miR-15b-5p. We also found that SNHG1 could directly bind to miR-15-5p and repress miR-15-5p expression. Upregulation of miR-15b-5p alleviated α-synuclein aggregation and apoptosis by targeting SIAH1 in SH-SY5Y cells overexpressing α-synuclein. Overexpression of SNHG1 enhanced, whereas SNHG1 knockdown inhibited α-synuclein aggregation and α-synuclein-induced apoptosis. Moreover, the neuroprotective effect of si-SNHG1 was abrogated by downregulation of miR-15b-5p. In summary, our data suggest that SNHG1, as a pathogenic factor, promotes α-synuclein aggregation and toxicity by targeting the miR-15b-5p/SIAH1 axis, contributing to a better understanding of the mechanisms of Lewy body formation and loss of dopaminergic neurons in PD.
Collapse
Affiliation(s)
- Yuan Chen
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan Province, PR China
| | - Ya-Jun Lian
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan Province, PR China.
| | - Yun-Qing Ma
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan Province, PR China
| | - Chuan-Jie Wu
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan Province, PR China
| | - Ya-Ke Zheng
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan Province, PR China
| | - Nan-Chang Xie
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan Province, PR China
| |
Collapse
|
86
|
Zhu Y, Li B, Liu Z, Jiang L, Wang G, Lv M, Li D. Up-regulation of lncRNA SNHG1 indicates poor prognosis and promotes cell proliferation and metastasis of colorectal cancer by activation of the Wnt/β-catenin signaling pathway. Oncotarget 2017; 8:111715-111727. [PMID: 29340086 PMCID: PMC5762354 DOI: 10.18632/oncotarget.22903] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2017] [Accepted: 11/16/2017] [Indexed: 01/05/2023] Open
Abstract
Recently, the lncRNA small nucleolar RNA host gene (SNHG1) has been exhibited to be upregulated, which plays a crucial role in the development and prognosis of several cancers. However, the role of the biology and clinical significance of SNHG1 in the tumorigenesis of colorectal cancer (CRC) has rarely been reported. In this work, we firstly found that SNHG1 expression levels were upregulated aberrantly in colorectal cancer tissues and colorectal cancer cell lines. By Kaplan-Meier survival analysis, patients with high SNHG1 expression level had poorer overall survival (OS) and progression-free survival (PFS) than those with low SNHG1 expression. In multivariate analysis, increased SNHG1 expression was proved to be an independent unfavorable prognostic indicator for CRC. In vitro experiments revealed that SNHG1 silencing inhibited the growth and metastasis and induced apoptosis of CRC cell lines. Finally, we found that SNHG1 may induce the activation of the WNT/β-catenin pathway through regulating β-catenin expression and transcription factor-4 (TCF-4), cyclin D1 and MMP-9. Altogether, our findings demonstrated that lncRNA SNHG1, was high expressed in colorectal cancer tissues and may serve as a tumor oncogene through regulating WNT/β-catenin signal pathway, which provided a candidate diagnostic biomarker and a promising therapeutic target for patients with CRC.
Collapse
Affiliation(s)
- Yuping Zhu
- Department of Colorectal Surgery, Zhejiang Cancer Hospital, Hangzhou 310022, Zhejiang Province, China
| | - Bo Li
- Department of Colorectal Surgery, Zhejiang Cancer Hospital, Hangzhou 310022, Zhejiang Province, China
| | - Zhuo Liu
- Department of Colorectal Surgery, Zhejiang Cancer Hospital, Hangzhou 310022, Zhejiang Province, China
| | - Lai Jiang
- Department of Colorectal Surgery, Zhejiang Cancer Hospital, Hangzhou 310022, Zhejiang Province, China
| | - Gang Wang
- Department of Colorectal Surgery, Zhejiang Cancer Hospital, Hangzhou 310022, Zhejiang Province, China
| | - Min Lv
- Department of Ultrasonic, Zhejiang Cancer Hospital, Hangzhou 310022, Zhejiang Province, China
| | - Dechuan Li
- Department of Colorectal Surgery, Zhejiang Cancer Hospital, Hangzhou 310022, Zhejiang Province, China
| |
Collapse
|
87
|
Wang J, Cao L, Wu J, Wang Q. Long non-coding RNA SNHG1 regulates NOB1 expression by sponging miR-326 and promotes tumorigenesis in osteosarcoma. Int J Oncol 2017; 52:77-88. [PMID: 29115574 PMCID: PMC5743365 DOI: 10.3892/ijo.2017.4187] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2017] [Accepted: 10/08/2017] [Indexed: 01/17/2023] Open
Abstract
The long non-coding RNA (lncRNA) small nucleolar RNA host gene 1 (SNHG1) has been demonstrated to participate in the deterioration of many types of cancer. However, the underlying mechanisms of SNHG1-mediating functions in osteosarcoma (OS) have yet to be elucidated. In the present study, our results showed that SNHG1 was upregulated in OS tissues and cell lines, and high SNHG1 expression predicts poor overall survival of OS patients. Knockdown of SNHG1 inhibited cell growth and metastasis of OS in vitro and in vivo. Furthermore, our data demonstrated that there was reciprocal repression between SNHG1 and miR-326 which act as a tumor suppressor in OS cells, and exhibiting a strong negative relationship between SNHG1 and miR-326 expression in OS tissues. Additionally, we identified that SNHG1 increased human nin one binding protein (NOB1), an oncogene, through sponging miR-326 as competing endogenous RNA (ceRNA), finally prompting cell growth, migration and invasion in OS. Collectively, these findings not only uncovered that the SNHG1/miR-326/NOB1 signaling axis has a key role in OS progression but also suggested the potential application of SNHG1 and miR-326 as biomarkers in the OS diagnosis and treatment.
Collapse
Affiliation(s)
- Jiandong Wang
- Department of Trauma and Orthopedics, Trauma Emergency Center, Shanghai General Hospital, Shanghai Jiao Tong University, Shanghai 200080, P.R. China
| | - Lei Cao
- Department of Trauma and Orthopedics, Trauma Emergency Center, Shanghai General Hospital, Shanghai Jiao Tong University, Shanghai 200080, P.R. China
| | - Jianhong Wu
- Department of Trauma and Orthopedics, Trauma Emergency Center, Shanghai General Hospital, Shanghai Jiao Tong University, Shanghai 200080, P.R. China
| | - Qiugen Wang
- Department of Trauma and Orthopedics, Trauma Emergency Center, Shanghai General Hospital, Shanghai Jiao Tong University, Shanghai 200080, P.R. China
| |
Collapse
|
88
|
Zhang Y, Huang JC, Cai KT, Yu XB, Chen YR, Pan WY, He ZL, Lv J, Feng ZB, Chen G. Long non‑coding RNA HOTTIP promotes hepatocellular carcinoma tumorigenesis and development: A comprehensive investigation based on bioinformatics, qRT‑PCR and meta‑analysis of 393 cases. Int J Oncol 2017; 51:1705-1721. [PMID: 29039502 PMCID: PMC5673011 DOI: 10.3892/ijo.2017.4164] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2017] [Accepted: 10/06/2017] [Indexed: 12/20/2022] Open
Abstract
HOTTIP functions as an independent biomarker in multiple cancers. However, the role of HOTTIP in hepatocellular carcinoma (HCC) remains unclear. In this study, we sought to investigate the HOTTIP expression in HCC and normal liver. We combined quantitative reverse transcription-polymerase chain reactions (qRT-PCR), Gene Expression Omnibus (GEO) and The Cancer Genome Atlas (TCGA), Multi Experiment Matrix (MEM) and Oncomine database to assess the clinical role and the potential molecular mechanism of HOTTIP in HCC. Furthermore, a meta-analysis was performed to evaluate the relationship between HOTTIP and HCC tumorigenesis and development. Additionally, bioinformatics analysis, which contained Gene Ontology (GO), Kyoto Encyclopedia of Genes and Genomes (KEGG) and network analysis, were applied to investigate the underlying functions, pathways and networks of the potential genes. HOTTIP was obviously upregulated in HCC. A statistically significant higher expression of HOTTIP was found in TNM (III +IV), age (≥60), sex (male), race (white) and cirrhosis (no) compared to the control groups (P<0.05). Furthermore, the meta-analysis of 393 cases from multiple centers indicated that HOTTIP had high diagnostic value in HCC. Additionally, according to GO and KEGG analyses, we found that the most strongly enriched functional terms were gland development, transcription factor activity and extrinsic to membrane. Also, the HOTTIP co-expressed genes were significantly related to PPAR signaling pathway. We speculate that HOTTIP might play a vital part in HCC via regulating various pathways, especially PPAR signaling pathway. However, the detailed mechanism should be confirmed by functional experiments.
Collapse
Affiliation(s)
- Yu Zhang
- Department of Pathology, First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region 530021, P.R. China
| | - Jia-Cheng Huang
- Department of Pathology, First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region 530021, P.R. China
| | - Kai-Teng Cai
- Department of Pathology, First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region 530021, P.R. China
| | - Xi-Bing Yu
- Department of Hepatobiliary Surgery, First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region 530021, P.R. China
| | - You-Rong Chen
- Department of Pathology, First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region 530021, P.R. China
| | - Wen-Ya Pan
- Department of Pathology, First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region 530021, P.R. China
| | - Ze-Liang He
- Department of Hepatobiliary Surgery, First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region 530021, P.R. China
| | - Jun Lv
- Department of Hepatobiliary Surgery, First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region 530021, P.R. China
| | - Zhen-Bo Feng
- Department of Pathology, First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region 530021, P.R. China
| | - Gang Chen
- Department of Pathology, First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region 530021, P.R. China
| |
Collapse
|
89
|
Hu Y, Ma Z, He Y, Liu W, Su Y, Tang Z. LncRNA-SNHG1 contributes to gastric cancer cell proliferation by regulating DNMT1. Biochem Biophys Res Commun 2017; 491:926-931. [PMID: 28754593 DOI: 10.1016/j.bbrc.2017.07.137] [Citation(s) in RCA: 84] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2017] [Accepted: 07/24/2017] [Indexed: 12/24/2022]
Abstract
OBJECTIVE The objective of this study was to determine the expression of long-chain non-coding RNA SNHG1 (lncRNA-SNHG1) in gastric carcinoma and explore its function on cancer cell proliferation. METHODS The expression of lncRNA-SNHG1 in tumor tissues and corresponding adjacent tissues from 50 patients with gastric cancer was detected with realtime-PCR. The relationships between the expression of lncRNA-SNHG1 and clinicopathological features of gastric cancer patients were analyzed. Survival analysis was performed to study the correlation between lncRNA SNHG1 expression and patient prognosis. To assess the effect of LncRNA SNHG1 on proliferation in cancer cells, cell viability and colony formation assays were conducted when lncRNA SNHG1 was upregulated or downregulated by Lentivirus or plasmid in gastric cancer cells. Furthermore, in vivo tumor assay was performed to confirm the impact of lncRNA SNHG1 on proliferation of gastric cancer. RESULTS The expression of lncRNA SNHG1 in gastric cancer tissues was significantly higher than that in adjacent tissues and was correlated with TNM stage, T stage, and lymph node metastasis. The survival time of patients with higher expression level of lncRNA-SNHG1 was significantly lower than that of the lower expression level. LncRNA-SNHG1 accelerated the proliferation of gastric cancer cells obviously and increased the expression of DNMT1. CONCLUSION LncRNA SNHG1 promotes DNMT1 expression, which facilitates the gastric cancer proliferation.
Collapse
Affiliation(s)
- Yongbo Hu
- Department of General Surgery, Xiantao First People's Hospital Affiliated to Yangtze University, Xiantao, Hubei, China
| | - Zhen Ma
- Department of General Surgery, Xiantao First People's Hospital Affiliated to Yangtze University, Xiantao, Hubei, China
| | - Yiming He
- Department of General Surgery, Xiantao First People's Hospital Affiliated to Yangtze University, Xiantao, Hubei, China
| | - Wei Liu
- Department of General Surgery, Xiantao First People's Hospital Affiliated to Yangtze University, Xiantao, Hubei, China
| | - Yu Su
- Department of General Surgery, Xiantao First People's Hospital Affiliated to Yangtze University, Xiantao, Hubei, China
| | - Zongbin Tang
- Department of General Surgery, Xiantao First People's Hospital Affiliated to Yangtze University, Xiantao, Hubei, China.
| |
Collapse
|
90
|
Cao Z, Huang S, Li J, Bai Y, Dou C, Liu C, Kang F, Gong X, Ding H, Hou T, Dong S. Long noncoding RNA expression profiles in chondrogenic and hypertrophic differentiation of mouse mesenchymal stem cells. Funct Integr Genomics 2017; 17:739-749. [PMID: 28735352 DOI: 10.1007/s10142-017-0569-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2017] [Revised: 07/09/2017] [Accepted: 07/13/2017] [Indexed: 02/07/2023]
Abstract
Long noncoding RNAs (lncRNAs) are important regulators for a variety of biological processes. Chondrogenic differentiation of mesenchymal stem cells (MSCs) is a crucial stage in chondrogenesis while chondrocyte hypertrophy is related to endochondral ossification and osteoarthritis. However, the effects of lncRNAs on chondrogenic and hypertrophic differentiation of mouse MSCs are unclear. To explore the potential mechanisms of lncRNAs during chondrogenesis and chondrocyte hypertrophy, microarray was performed to investigate the expression profiles of lncRNA and mRNA in MSCs, pre-chondrocytes, and hypertrophic chondrocytes. Then, we validated microarray data by RT-PCR and screened three lncRNAs from upregulating groups during chondrogenesis and chondrocyte hypertrophy respectively. After downregulating any of the above lncRNAs, we found that the expression of chondrogenesis-related genes such as Sox9 and Col2a1 and hypertrophy-related genes including Runx2 and Col10a1 was inhibited, respectively. Furthermore, the target genes of above lncRNAs were predicted by bioinformatics approaches. Gene ontology and Kyoto encyclopedia of genes and genome biological pathway analysis were also made to speculate the functions of above lncRNAs. In conclusion, the study first revealed the expression profile of lncRNAs in chondrogenic and hypertrophic differentiations of mouse MSCs and presented a new prospect for the underlying mechanisms of chondrogenesis and endochondral ossification.
Collapse
Affiliation(s)
- Zhen Cao
- Department of Biomedical Materials Science, School of Biomedical Engineering, Third Military Medical University, Gaotanyan Street No. 30, Chongqing, 400038, China.,Department of Anatomy, Third Military Medical University, Chongqing, 400038, China
| | - Song Huang
- School of Pathology and Laboratory Medicine, The University of Western Australia, Nedlands, WA, 6009, Australia
| | - Jianmei Li
- Department of Biomedical Materials Science, School of Biomedical Engineering, Third Military Medical University, Gaotanyan Street No. 30, Chongqing, 400038, China
| | - Yun Bai
- Department of Biomedical Materials Science, School of Biomedical Engineering, Third Military Medical University, Gaotanyan Street No. 30, Chongqing, 400038, China
| | - Ce Dou
- Department of Biomedical Materials Science, School of Biomedical Engineering, Third Military Medical University, Gaotanyan Street No. 30, Chongqing, 400038, China.,National & Regional United Engineering Laboratory of Tissue Engineering, Department of Orthopedics, Southwest Hospital, Third Military Medical University, Chongqing, 400038, China
| | - Chuan Liu
- Department of Biomedical Materials Science, School of Biomedical Engineering, Third Military Medical University, Gaotanyan Street No. 30, Chongqing, 400038, China
| | - Fei Kang
- Department of Biomedical Materials Science, School of Biomedical Engineering, Third Military Medical University, Gaotanyan Street No. 30, Chongqing, 400038, China
| | - Xiaoshan Gong
- Department of Biomedical Materials Science, School of Biomedical Engineering, Third Military Medical University, Gaotanyan Street No. 30, Chongqing, 400038, China
| | - Haibin Ding
- Department of Biomedical Materials Science, School of Biomedical Engineering, Third Military Medical University, Gaotanyan Street No. 30, Chongqing, 400038, China
| | - Tianyong Hou
- National & Regional United Engineering Laboratory of Tissue Engineering, Department of Orthopedics, Southwest Hospital, Third Military Medical University, Chongqing, 400038, China
| | - Shiwu Dong
- Department of Biomedical Materials Science, School of Biomedical Engineering, Third Military Medical University, Gaotanyan Street No. 30, Chongqing, 400038, China.
| |
Collapse
|