1
|
Qian J, Ge L, Lu C, Han X, Li M, Bian Z. LINC00665 aggravates the malignant phenotypes in chondrosarcoma cells through miR-665/FGF9 pathway. Int J Biol Macromol 2024; 280:135727. [PMID: 39293617 DOI: 10.1016/j.ijbiomac.2024.135727] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Revised: 09/10/2024] [Accepted: 09/14/2024] [Indexed: 09/20/2024]
Abstract
Long non-coding RNAs (lncRNAs) have been demonstrated to participate in a variety of physiological and pathological processes, including tumor initiation and development. Nevertheless, few of them have been investigated in chondrosarcoma. Here, we were intended to unveil the role of long intergenic non-protein coding RNA 665 (LINC00665) in chondrosarcoma. RT-qPCR was adopted for gene expression detection. The biological processes in chondrosarcoma cells were detected by CCK-8, EdU, TUNEL, Transwell and wound healing assays. The relationships between genes in chondrosarcoma cells were evaluated by a series of mechanism experiments including RIP, luciferase reporter assays and so on.LINC00665 expressed at a high level in chondrosarcoma cell lines. LINC00665 interference suppressed cell proliferation, migration and invasion in chondrosarcoma. Besides, LINC00665 interacted with microRNA-665 (miR-665), which was then verified to be down-regulated in chondrosarcoma cells. Additionally, LINC00665 and miR-665 were mutually inhibited by each other in chondrosarcoma cells. Importantly, LINC00665 stimulated fibroblast growth factor 9 (FGF9) expression in chondrosarcoma cells via sponging miR-665. Furthermore, FGF9 participated in the regulation of LINC00665-promoted chondrosarcoma development. CONCLUSION: LINC00665 facilitates chondrosarcoma progression via miR-665/FGF9 axis, which might indicate a new path for the treatment of chondrosarcoma.
Collapse
Affiliation(s)
- Jin Qian
- Department of Orthopedics, Affiliated Hangzhou First People's Hospital, WestLake University School of Medicine, Hangzhou 310006, Zhejiang Province, China
| | - Lujie Ge
- Department of Orthopedics, Affiliated Hangzhou First People's Hospital, WestLake University School of Medicine, Hangzhou 310006, Zhejiang Province, China
| | - Congcong Lu
- Department of Orthopedics, Affiliated Hangzhou First People's Hospital, WestLake University School of Medicine, Hangzhou 310006, Zhejiang Province, China
| | - Xiao Han
- Department of Orthopedics, Affiliated Hangzhou First People's Hospital, WestLake University School of Medicine, Hangzhou 310006, Zhejiang Province, China
| | - Maoqiang Li
- Department of Orthopedics, Affiliated Hangzhou First People's Hospital, WestLake University School of Medicine, Hangzhou 310006, Zhejiang Province, China.
| | - Zhenyu Bian
- Department of Orthopedics, Affiliated Hangzhou First People's Hospital, WestLake University School of Medicine, Hangzhou 310006, Zhejiang Province, China.
| |
Collapse
|
2
|
Yuan M, Gu Y, Chen J, Jiang Y, Qian J, Cao S. LINC00665: A Promising Biomarker in Gastrointestinal Tumors. Curr Mol Med 2024; 24:51-59. [PMID: 36464865 DOI: 10.2174/1566524023666221201141443] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2022] [Revised: 10/19/2022] [Accepted: 10/26/2022] [Indexed: 12/12/2022]
Abstract
An increasing volume of studies has reported that long non-codingRNAs (lncRNAs) are involved in the carcinogenesis of many different cancers. Especially in gastrointestinal tumors, lncRNAs are found to participate in various physiological and pathological processes. LncRNAs can regulate gene expression at multiple levels, including transcriptional, post-transcription, translational, and post-translational levels. Long intergenic non-protein coding RNA 665(LINC00665), a novel cancer-related lncRNA, is frequently dysregulated in multiple gastrointestinal tumors, including gastric and colorectal cancers, hepatocellular carcinoma, and so on. In this review, we analyzed the expression and prognostic value of LINC00665 in human gastrointestinal tumors, systematically summarized the current literature about the clinical significance of this lncRNA, and explored the regulatory mechanisms of LINC00665 as a competing endogenous RNA (ceRNA) in tumor progression. Consequently, we concluded that LINC00665 might act as a prognostic biomarker and a potential target for gastrointestinal tumor diagnosis and treatment.
Collapse
Affiliation(s)
- Mengping Yuan
- Department of Gastroenterology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325000, PR China
| | - Yuyang Gu
- Department of Oncology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325000, PR China
| | - Jiawen Chen
- School of Medicine, Huzhou University, Huzhou, 313000, PR China
| | - Yibin Jiang
- School of Medicine, Huzhou University, Huzhou, 313000, PR China
| | - Jing Qian
- School of Medicine, Huzhou University, Huzhou, 313000, PR China
- Key Laboratory of Vector Biology and Pathogen Control of Zhejiang Province, Huzhou University, Huzhou, 313000, PR China
| | - Shuguang Cao
- Department of Gastroenterology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325000, PR China
| |
Collapse
|
3
|
Jin Z, Meng YJ, Xu YS, Wang MM, Chen D, Jiang X, Xiong ZF. Prognostic and clinicopathological values of LINC00665 in cancers: a systematic review and China population-based meta-analysis. Clin Exp Med 2023; 23:1475-1487. [PMID: 36219365 DOI: 10.1007/s10238-022-00912-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2022] [Accepted: 10/01/2022] [Indexed: 12/24/2022]
Abstract
BACKGROUND Recent studies have uncovered that the aberrant expression of LINC00665 contributes to the malignant pathological process of various cancers and is closely related to the unfavorable prognosis of patients with cancer. However, a systematic analysis of the prognostic and clinicopathologic values of LINC00665 in cancers has not been conducted. OBJECTIVE We aim to clarify the association of LINC00665 expression with patient survival and clinicopathologic phenotypes in cancers. METHODS An electronic search of PubMed, Embase and Web of Science was performed to select eligible literature. Pooled hazard ratio (HR) and odds ratio (OR) were calculated to assess the clinical importance of LINC00665. The fixed-effects model was used to analyze the combined HR values and 95% CI when the studies had no significant heterogeneity (P > 0.1 for the Chi-square test or I2 < 50%). Begg's test and sensitivity analysis were also conducted. This study was registered in The International Prospective Register of Systematic Reviews (PROSPERO registration number: CRD42021290123). RESULTS A total of 710 patients from 10 eligible studies were enrolled in this meta-analysis, which was based on China population. The pooled results of this analysis revealed that high-level expression of LINC00665 was notably correlated with poor overall survival (HR = 2.08, 95% CI = 1.57-2.75) and recurrence-free survival (HR = 2.49, 95% CI = 1.63-3.80) in human cancers. Elevated LINC00665 expression was also correlated with more advanced clinical stage, earlier lymph node metastasis, lower tumor differentiation, earlier distant metastasis and larger tumor size. CONCLUSION LINC00665 expression was critically related to the cancer prognosis, which has important prognostic implications for clinical prediction.
Collapse
Affiliation(s)
- Ze Jin
- Division of Gastroenterology, Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ya-Jun Meng
- Division of Gastroenterology, Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yu-Shuang Xu
- Division of Gastroenterology, Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Meng-Meng Wang
- Division of Gastroenterology, Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Di Chen
- Division of Gastroenterology, Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xin Jiang
- Division of Gastroenterology, Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhi-Fan Xiong
- Division of Gastroenterology, Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
4
|
A Concise Review on Dysregulation of LINC00665 in Cancers. Cells 2022; 11:cells11223575. [PMID: 36429005 PMCID: PMC9688310 DOI: 10.3390/cells11223575] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Revised: 11/05/2022] [Accepted: 11/10/2022] [Indexed: 11/16/2022] Open
Abstract
Long Intergenic Non-Protein Coding RNA 665 (LINC00665) is an RNA gene located on the minus strand of chromosome 19. This lncRNA acts as a competing endogenous RNA for miR-4458, miR-379-5p, miR-551b-5p, miR-3619-5p, miR-424-5p, miR-9-5p, miR-214-3p, miR-126-5p, miR-149-3p, miR-379-5p, miR-665, miR-34a-5p, miR-186-5p, miR-138-5p, miR-181c-5p, miR-98, miR-195-5p, miR-224-5p, miR-3619, miR-708, miR-101, miR-1224-5p, miR-34a-5p, and miR-142-5p. Via influencing expression of these miRNAs, it can enhance expression of a number of oncogenes. Moreover, LINC00665 can influence activity of Wnt/β-Catenin, TGF-β, MAPK1, NF-κB, ERK, and PI3K/AKT signaling. Function of this lncRNA has been assessed through gain-of-function tests and/or loss-of-function studies. Furthermore, diverse research groups have evaluated its expression levels in tissue samples using microarray and RT-qPCR techniques. In this manuscript, we have summarized the results of these studies and categorized them in three sections, i.e., cell line studies, animal studies, and investigations in clinical samples.
Collapse
|
5
|
Hepatitis B Virus-Encoded HBsAg Contributes to Hepatocarcinogenesis by Inducing the Oncogenic Long Noncoding RNA LINC00665 through the NF-κB Pathway. Microbiol Spectr 2022; 10:e0273121. [PMID: 35993712 PMCID: PMC9603668 DOI: 10.1128/spectrum.02731-21] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
Clinical and in vivo studies have demonstrated a role for hepatitis B virus (HBV)-encoded HBsAg (hepatitis B surface antigen) in HBV-related hepatocellular carcinoma (HCC); however, the underlying mechanisms remain largely unknown. Here, we investigated the role of HBsAg in regulating long noncoding RNAs (lncRNAs) involved in HCC progression. Our analysis of microarray data sets identified LINC00665 as an HBsAg-regulated lncRNA. Furthermore, LINC00665 is upregulated in liver samples from HBV-infected patients as well as in HCC, specifically in HBV-related HCC liver samples. These findings were supported by our in vitro data demonstrating that HBsAg, as well as HBV, positively regulates LINC00665 in multiple HBV cell culture models. Next, we evaluated the oncogenic potential of LINC00665 by its overexpression and CRISPR interference (CRISPRi)-based knockdown in various cell-based assays. LINC00665 promoted cell proliferation, migration, and colony formation but inhibited cell apoptosis in vitro. We then identified the underlying mechanism of HBsAg-mediated regulation of LINC00665. We used immunofluorescence assays to show that HBsAg enhanced the nuclear translocation of NF-κB factors in HepG2 cells, confirming that HBsAg activates NF-κB. Inhibition of NF-κB signaling nullified HBsAg-mediated LINC00665 upregulation, suggesting that HBsAg acts through NF-κB to regulate LINC00665. Furthermore, the LINC00665 promoter contains NF-κB binding sites, and their disruption abrogated HBsAg-induced LINC00665 upregulation. Finally, HBsAg facilitated the enrichment of the NF-κB factors NF-κB1, RelA, and c-Rel in the LINC00665 promoter. Taken together, this work shows that HBsAg can drive hepatocarcinogenesis by upregulating oncogenic LINC000665 through the NF-κB pathway, thereby identifying a novel mechanism in HBV-related HCC. IMPORTANCE Hepatitis B virus (HBV) is a major risk factor for hepatocellular carcinoma (HCC). Numerous reports indicate an oncogenic role for HBV-encoded HBsAg; however, the underlying mechanisms are not well understood. Here, we studied the role of HBsAg in regulating lncRNAs involved in hepatocarcinogenesis. We demonstrate that HBsAg, as well as HBV, positively regulates oncogenic lncRNA LINC00665. The clinical significance of this lncRNA is highlighted by our observation that LINC00665 is upregulated in liver samples during HBV infection and HBV-related HCC. Furthermore, we show LINC00665 can drive hepatocarcinogenesis by promoting cell proliferation, colony formation, and cell migration and inhibiting apoptosis. Taken together, this work identified LINC00665 as a novel gene through which HBsAg can drive hepatocarcinogenesis. Finally, we show that HBsAg enhances LINC00665 levels in hepatocytes by activating the NF-κB pathway, thereby identifying a novel mechanism by which HBV may contribute to HCC.
Collapse
|
6
|
Zhao S, Sun W, Chen SY, Li Y, Wang J, Lai S, Jia X. The exploration of miRNAs and mRNA profiles revealed the molecular mechanisms of cattle-yak male infertility. Front Vet Sci 2022; 9:974703. [PMID: 36277066 PMCID: PMC9581192 DOI: 10.3389/fvets.2022.974703] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Accepted: 08/24/2022] [Indexed: 11/04/2022] Open
Abstract
Cattle-yak, the first-generation offspring of cattle and yak, inherited many excellent characteristics from their parents. However, F1 male hybrid infertility restricts the utilization of heterosis greatly. In this study, we first compared the testicular tissue histological characteristics of three cattle, three yaks, and three cattle-yak. Then we explored the miRNA profiles and the target functions of nine samples with RNA-seq technology. We further analyzed the function of DE gene sets of mRNA profiles identified previously with GSEA. Testicular histology indicated that the seminiferous tubules became vacuolated and few active germ cells can be seen. RNA-seq results showed 47 up-regulated and 34 down-regulated, 16 up-regulated and 21 down-regulated miRNAs in cattle and yaks compared with cattle-yak, respectively. From the intersection of DE miRNAs, we identified that bta-miR-7 in cattle-yak is down-regulated. Target prediction indicated that the filtered genes especially MYRFL, FANCA, INSL3, USP9X, and SHF of bta-miR-7 may play crucial roles in the reproductive process. With further network analysis and GSEA, we screened such hub genes and function terms, we also found some DE gene sets that enriched in ATP binding, DNA binding, and reproduction processes. We concluded that bta-miR-7 may play an important role in influencing fecundity. Our study provides new insights for explaining the molecular mechanism of cattle-yak infertility.
Collapse
|
7
|
Wang S, Wang Y, Lu J, Wang J. LncRNA LINC00665 Promotes Ovarian Cancer Cell Proliferation and Inhibits Apoptosis via Targeting miR-181a-5p/FHDC. Appl Biochem Biotechnol 2022; 194:3819-3832. [PMID: 35524876 DOI: 10.1007/s12010-022-03943-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/20/2022] [Indexed: 11/02/2022]
Abstract
Previous reports indicate that long intergenic non-coding RNA LINC00665 naturally occurred vital effects in various cancers. Herein, the role of LINC00665 in ovarian cancer progress was explored. We found that LINC00665 was upregulated in ovarian cancer cell lines. Besides, a series of assays including flow cytometry, wound-healing, transwell, cell counting Kit-8 (CCK-8), and EdU assay confirmed that the knockdown of LINC00665 could reduce the viability, proliferation, and migration of SKOV-3 and OVCAR-3 cells. Accumulating evidence indicates that many lncRNAs can function as endogenous miRNA sponges by competitively binding common miRNAs. In this study, the bioinformatics analysis suggests that LNC00665 specifically binds to miR-181a-5p. LINC00665 downregulated the miR-181a-5p in SKOV-3 and OVCAR-3 cells. The knockdown of miR-181a-5p evidently reverses the inhibitory effect of sh-LINC00662. Besides, FH2 domain containing 1 (FHDC1) has been proved to deed as an effective target of miR-181a-5p. The results reveal the knockdown of LINC00665 facilitates ovarian cancer via development by sponging miR-181a-5p and up-regulating FHDC1 expression. These may contribute to ovarian cancer therapy.
Collapse
Affiliation(s)
- Suli Wang
- Department of Women's Health, Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing, 210004, Jiangsu Province, China
- Department of Gynecological Oncology Surgery, Jiangsu Cancer Hospital (Jiangsu Institute of Cancer Research, Nanjing Medical University Affiliated Cancer Hospital), 42 Baiziting Road, Nanjing, 210009, Jiangsu Province, China
| | - Yingchun Wang
- Department of Gynecology, Nanjing Maternity and Child Health Care Hospital, Nanjing, 210004, Jiangsu Province, China
| | - Jin Lu
- Department of Gynecological Oncology Surgery, Jiangsu Cancer Hospital (Jiangsu Institute of Cancer Research, Nanjing Medical University Affiliated Cancer Hospital), 42 Baiziting Road, Nanjing, 210009, Jiangsu Province, China
| | - Jinhua Wang
- Department of Gynecological Oncology Surgery, Jiangsu Cancer Hospital (Jiangsu Institute of Cancer Research, Nanjing Medical University Affiliated Cancer Hospital), 42 Baiziting Road, Nanjing, 210009, Jiangsu Province, China.
| |
Collapse
|
8
|
Zhang C, Xu SN, Li K, Chen JH, Li Q, Liu Y. The Biological and Molecular Function of LINC00665 in Human Cancers. Front Oncol 2022; 12:886034. [PMID: 35664776 PMCID: PMC9161781 DOI: 10.3389/fonc.2022.886034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Accepted: 04/25/2022] [Indexed: 12/24/2022] Open
Abstract
Long non-coding RNAs (lncRNAs) are more than 200 nucleotides in length and are implicated in the development of human cancers, without protein-coding function. Mounting evidence indicates that cancer initiation and progression are triggered by lncRNA dysregulation. Recently, a growing number of studies have found that LINC00665, a long intergenic non-protein coding RNA, may be associated with various cancers, including gastrointestinal tumors, gynecological tumors, and respiratory neoplasms. LINC00665 was reported to be significantly dysregulated in cancers and has an important clinical association. It participates in cell proliferation, migration, invasion, and apoptosis through different biological pathways. In this review, we summarize the current findings on LINC00665, including its biological roles and molecular mechanisms in various cancers. LINC00665 may be a potential prognostic biomarker and novel therapeutic target for cancers.
Collapse
Affiliation(s)
- Cheng Zhang
- Department of Oncology, The Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Zhengzhou, China
| | - Shu-Ning Xu
- Department of Oncology, The Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Zhengzhou, China
| | - Ke Li
- Department of Oncology, The Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Zhengzhou, China
| | - Jing-Hong Chen
- Department of Oncology, The Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Zhengzhou, China
| | - Qun Li
- Department of Oncology, The Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Zhengzhou, China
| | - Ying Liu
- Department of Oncology, The Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Zhengzhou, China
| |
Collapse
|
9
|
Chen W, Hong L, Hou C, Zong G, Zhang J. Up-regulation of LINC00665 contributes to the progression of glioma and correlates with its MRI characteristics. Am J Transl Res 2022; 14:2988-3002. [PMID: 35702084 PMCID: PMC9185054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Accepted: 03/22/2022] [Indexed: 06/15/2023]
Abstract
BACKGROUND LncRNA LINC00665 partakes in controlling the malignant phenotype of cancer cells, but its role in glioma and the related regulatory mechanism remain uncertain. METHODS RT-PCR was exploited to examine LINC00665 expression. The relationships among the LINC00665 expression, the clinicopathologic values and magnetic resonance imaging (MRI) characteristics of glioma were analyzed. The multiplication, movement, and aggressiveness of glioma cell lines were evaluated by CCK-8, EdU, and Transwell experiments after constructing LINC00665 overexpression and LINC00665 knockdown cell models. A dual-luciferase reporter gene experiment and RIP experiment were executed to validate the interactions between LINC00665 and miR-129-5p, and between miR-129-5p and HMGB1. Western blot and RT-PCR were conducted to find the impact of LINC00665 and miR-129-5p on HMGB1 expression. Nude mouse model was also constructed to examine the impact of LINC00665 on multiplication and aggressiveness of glioma cells in vivo. RESULTS LINC00665 expression was markedly increased in glioma. High LINC00665 expression in glioma was closely linked to larger tumor diameter, higher pathologic grade, heterogeneous MRI signal of the tumor, increased peritumoral edema, and stronger MRI enhancement characteristics. LINC00665 overexpression facilitated the malignant behavior of glioma cells, while LINC00665 knockdown played the reverse role. Mechanistically, LINC00665 could decoy miR-129-5p, and indirectly increased HMGB1 expression. CONCLUSION LINC00665 functions as an oncogenic lncRNA in glioma, to accelerate glioma progression by modulating miR-129-5p and increasing HMGB1 expression.
Collapse
Affiliation(s)
- Wangsheng Chen
- Department of Radiology, Hainan General Hospital, Hainan Hospital of Hainan Medical UniversityHaikou 570311, China
| | - Lan Hong
- Department of Gynecology, Hainan General Hospital/Hainan Hospital of Hainan Medical UniversityHaikou 570311, China
| | - Changlong Hou
- Department of Radiology, Shanghai East Hospital, Tongji University School of MedicineShanghai 200120, China
| | - Genlin Zong
- Department of Radiology, Shanghai East Hospital, Tongji University School of MedicineShanghai 200120, China
| | - Jianhua Zhang
- Department of Radiology, Shanghai East Hospital, Tongji University School of MedicineShanghai 200120, China
| |
Collapse
|
10
|
LINC00665: An Emerging Biomarker for Cancer Diagnostics and Therapeutics. Cells 2022; 11:cells11091540. [PMID: 35563845 PMCID: PMC9102468 DOI: 10.3390/cells11091540] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Revised: 04/25/2022] [Accepted: 04/29/2022] [Indexed: 02/01/2023] Open
Abstract
Long intergenic noncoding RNA 00665 (LINC00665) is located on human chromosome 19q13.12. LINC00665 was upregulated in eighteen cancers and downregulated in two cancers. LINC00665 not only inhibits 25 miRNAs but also directly affects the stability of ten protein-coding genes. Notably, LINC00665 also encodes a micro-peptide CIP2A-BP that promotes triple-negative breast cancer progression. LINC00665 can participate in five signaling pathways to regulate cancer progression, including the Wnt/β-catenin signaling pathway, TGF-β signaling pathway, NF-κB signaling pathway, PI3K/AKT signaling pathway, and MAPK signaling pathway. Aberrant expression of LINC00665 in breast cancer, gastric cancer, and hepatocellular carcinoma can be used for disease diagnosis. In addition, aberrant expression of LINC00665 is closely associated with clinicopathological features and poor prognosis of various cancers. LINC00665 is closely associated with the effects of anticancer drugs, including gefitinib and cisplatin in non-small cell lung cancer, gemcitabine in cholangiocarcinoma, and cisplatin-paclitaxel in breast cancer. This work systematically summarizes the diagnostic and prognostic values of LINC00665 in various tumors, and comprehensively analyzes the molecular regulatory mechanism related to LINC00665, which is expected to provide clear guidance for future research.
Collapse
|
11
|
Zhu J, Zhang Y, Chen X, Bian Y, Li J, Wang K. The Emerging Roles of LINC00665 in Human Cancers. Front Cell Dev Biol 2022; 10:839177. [PMID: 35356290 PMCID: PMC8959703 DOI: 10.3389/fcell.2022.839177] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2021] [Accepted: 02/03/2022] [Indexed: 12/14/2022] Open
Abstract
Long non-coding RNAs (lncRNAs) are non-coding RNAs that have more than 200 nucleotides and can participate in the regulation of gene expression in various ways. An increasing number of studies have shown that the dysregulated expression of lncRNAs is related to the occurrence and progression of human cancers. LINC00665 is a novel lncRNA, which is abnormally expressed in various human cancers, such as lung cancer, breast cancer, prostate cancer, and glioma. LINC00665 functions in many biological processes of tumor cells, such as cell proliferation, migration, invasion, angiogenesis, and metabolism, and is related to the clinicopathological characteristics of cancer patients. LINC00665 can play biological functions as a ceRNA, directly binding and interacting with proteins, and as an upstream molecule regulating multiple signaling pathways. In this review, we comprehensively summarize the expression level, function, and molecular mechanisms of LINC00665 in different human cancers and emphasize that LINC00665 is a promising new diagnostic, prognostic biomarker, and therapeutic target.
Collapse
Affiliation(s)
| | | | | | | | - Juan Li
- *Correspondence: Keming Wang, ; Juan Li,
| | | |
Collapse
|
12
|
Identification of prognostic signature with seven LncRNAs for papillary thyroid carcinoma. Adv Med Sci 2022; 67:103-113. [PMID: 35121283 DOI: 10.1016/j.advms.2021.11.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Revised: 10/15/2021] [Accepted: 11/01/2021] [Indexed: 12/24/2022]
Abstract
PURPOSE With the increasing incidence of thyroid cancer (TC), the prognostic risk assessment of thyroid cancer has been becoming more and more important. The aim of this study was to screen TC-related biomarkers and identify key multi-long non coding RNA (lncRNA) signature for prognostic risk assessment of papillary TC. MATERIAL AND METHODS The lncRNAs differentially expressed between TC tissue and adjacent normal tissue was identified by R language. Bioinformatics analysis was applied to screen the lncRNAs significantly associated with prognosis in TC patients and build the multi-lncRNA signature. The lncRNAs were annotated by co-expression and enrichment analysis to demonstrate the underlying mechanism of their effect on prognosis. RESULTS 285 up-regulated and 174 down-regulated differently expressed lncRNAs were identified. Based on seven signature lncRNAs (AL591846.2, AC253536.3, AC004112.1, LINC00900, AC008555.1, TNRC6C-AS1, LINC01736) a prognostic risk assessment model was built. The model can segregate the patients into the high-risk and low-risk groups (P value <0.0001, CI: 0.02∼0.14). ROC analysis revealed that the area under the curve reached 0.86, indicating that this model had an excellent sensitivity and specificity. Also, the model could act as an independent prognostic indication (HR = 2.90, P value = 0.0094 with multivariate analysis). Annotation results further supported and enriched our understanding of the seven signature lncRNAs. Importantly, expression levels of three of the seven lncRNAs were confirmed in Gene Expression Omnibus (GEO) data. CONCLUSIONS This study has provided a promising method for the prognostic risk assessment in patients with TC.
Collapse
|
13
|
Cao W, Liu X, Su W, Liang H, Tang H, Zhang W, Huang S, Dang N, Qiao A. LINC00665 sponges miR-641 to promote the progression of breast cancer by targeting the SNF2-related CREBBP activator protein (SRCAP). Bioengineered 2022; 13:4573-4586. [PMID: 35152838 PMCID: PMC8974044 DOI: 10.1080/21655979.2022.2031402] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
The regulatory network of competing endogenous RNAs (ceRNA) exists widely in tumors and affects the expression of cancer-related genes, thus playing an important role in the development and prognosis of human tumors. In this research, we explored the role and mechanism of LINC00665 as a ceRNA in breast cancer. We analyzed the expression and targets of LINC00665 in breast cancer using bioinformatics, and detected their effects on breast cancer cells by CCK8, transwell, colony formation and flow cytometry assays. From our results, LINC00665 knockdown suppressed the proliferation, migration and invasion and induced the apoptosis through inactivating the AKT/mTOR signaling pathway in MCF7 and MDA-MB-231 cells. LINC00665 had five potential downstream target miRNAs (miR-542-3p, miR-624-5p, miR-641, miR-425-5p, and miR-30-3p). In dual-luciferase report gene assay, the fluorescence activity of cells transfected with miR-641 mimics decreased, and the expression of miR-641 decreased significantly after knocking down LINC00665. miR-641 mimics significantly inhibited cell proliferation and invasion in MCF7 and MDA-MB-231 cells. We detected five potential direct targets of miR-641 using qPCR (SRCAP, SIKE1, NADK, KHDC4, and HSPG2). SRCAP expression decreased significantly in miR-641 overexpression cells and the binding of SRCAP’s 3ʹUTR and miR-641 was further confirmed by dual-luciferase report gene assay. SRCAP blocked the proliferation and invasion inhibition induced by miR-641 or si-LINC00665 in MCF7 and MDA-MB-231 cells. In conclusion, LINC00665 could promote the survival and metastasis of breast cancer cells through sponging miR-641 and targeting SRCAP. This research provided new potential targets for targeted therapy in human breast cancer.
Collapse
Affiliation(s)
- Wen Cao
- Health College, Yantai Nanshan University, Yantai, Shandong, China
| | - Xiaojing Liu
- Department of Clinical Laboratory Medicine, Shandong University Qilu Hospital, Jinan, China
| | - Weijia Su
- Institute of Basic Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Hao Liang
- Institute of Basic Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Huiru Tang
- Institute of Basic Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Weiliang Zhang
- Institute of Basic Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Shuhong Huang
- Institute of Basic Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Ningning Dang
- Department of Dermatology, Shandong Provincial Hospital Affiliated to Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Aiguo Qiao
- Health College, Yantai Nanshan University, Yantai, Shandong, China
| |
Collapse
|
14
|
Lei M, Du X, Li X, Wang F, Gu L, Guo F. LINC00665 regulates hepatocellular carcinoma by modulating mRNA via the m6A enzyme. Open Life Sci 2022; 17:71-80. [PMID: 35233461 PMCID: PMC8847717 DOI: 10.1515/biol-2022-0003] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Revised: 11/05/2021] [Accepted: 12/12/2021] [Indexed: 12/13/2022] Open
Abstract
This study aimed to reveal the mechanism by which long noncoding RNAs (lncRNAs) modulate hepatocellular carcinoma (HCC) by regulating mRNA via the N6-methyladenosine (m6A) enzyme. The expression and clinical data of 365 HCC patients and 50 healthy control samples were downloaded from the the Cancer Genome Atlas (TCGA) database. Differentially expressed lncRNAs (DElncRNAs) and differentially expressed mRNAs (DEmRNAs) screened using limma packages from the R. m6A2Target database were used to predict the relationship between m6A enzyme-lncRNA and m6A enzyme-mRNA. The mRNAs in the lncRNA-m6A enzyme-mRNA network were subjected to enrichment analysis. Cox regression analysis was used to screen for RNAs significantly related to HCC prognosis. The expression of differentially expressed RNAs (DERs) was verified using the TCGA dataset and GSE55092. Eighty-five DElncRNAs and 747 DEmRNAs were identified. The mRNAs in the lncRNA-m6A enzyme-mRNA network were primarily involved in mitotic cell division, the p53 signaling pathway, and the cell cycle. Three lncRNAs and 14 mRNAs were significantly associated with HCC prognosis. Furthermore, the expression of 12 DERs differed significantly between patients in the early and advanced stages. LINC00665 was predicted to regulate 11 mRNAs by modulating IGF2BP1, IGF2BP2, and YTHDF1. Thus, this study provides new insights into the roles of lncRNA and m6A enzymes in HCC.
Collapse
Affiliation(s)
- Ming Lei
- Nursing Health Sciences College, Yunnan Open University , Kunming , Yunnan, 650500 , China
| | - Xinghua Du
- Laboratory Medicine Department, The Integrated Traditional Chinese and Western Medicine Hospital of Yunnan Province , Kunming , Yunnan, 650224 , China
| | - Xiaokai Li
- Hepatobiliary Surgery Department, The First Affiliated Hospital of Kunming Medical University , Kunming , Yunnan, 650032 , China
| | - Fuke Wang
- Sport Medicine Department, The First Affiliated Hospital of Kunming Medical University , Kunming , Yunnan, 650032 , China
| | - Ling Gu
- Pain Department, The First Affiliated Hospital of Kunming Medical University , Kunming , Yunnan, 650032 , China
| | - Feng Guo
- The Clinical Skills Training Center, Kunming Medical University, No. 1168 Chunrongxi Road Chenggong District , Kunming , Yunnan, 650500 , China
| |
Collapse
|
15
|
Rejali L, Seyedna SY, Asadzadeh Aghdaei H, Nazemalhosseini Mojarad E, Hashemi M. Expression and Clinical Significance of Novel Long Noncoding RNA Fibroblast Growth Factor 10AS and FGF10 in Colorectal Cancer. CELL JOURNAL 2021; 23:665-673. [PMID: 34939760 PMCID: PMC8665978 DOI: 10.22074/cellj.2021.7487] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Accepted: 05/30/2020] [Indexed: 11/09/2022]
Abstract
OBJECTIVE Colorectal cancer (CRC) imposes great health burdens worldwide. Growth factors contribute to cell growth, differentiation, angiogenesis and, most importantly, tumour formation in many types of cancers. Natural antisense transcripts (NATs) are inclusively predicted to play a major role in cancer progression. The present study aims to evaluate the relationship of fibroblast growth factor 10 (FGF10) and novel long noncoding RNA (lncRNA) antisense FGF10 (FGF10AS) expression with clinicopathologic features in CRC progression to designate a biomarker for CRC early detection. MATERIALS AND METHODS This cross-sectional study was conducted on 100 CRC tumour and parallel adjacent normal tissues. We added 30 normal cases to enhance accuracy of the test. The expression levels of FGF10 and FGF10AS were evaluated by real-time polymerase chain reaction (PCR). The findings were validated by measuring expression levels in the HT29 and SW480 cell lines. Immunohistochemistry analysis was performed systematically to evaluate FGF10 protein expression. The Mann-Whitney U test with Cox regression analysis were applied. P<0.05 were designated as significant. RESULTS A significant increase in expression was observed in FGF10 (P<0.001) along with a significant decrease in FGF10AS (P<0.02) in the tumour tissues in comparison with the adjacent normal tissues. Upregulation of FGF10 and downregulation of FGF10AS expression were strongly correlated with the Tumour, Node, Metastasis (TNM) stage (P<0.007 and P<0.004), vascular invasion (P<0.03 and P<0.01), lymph invasion (P<0.02 and P<0.04), and differentiation (P<0.01 and P<0.02), respectively. Moreover, the area under the receiver operating characteristic (ROC) curve for the prognostic value of FGF10 was about 0.84 (95% confidence interval [CI]: 0.771-0.912). Linear regression analysis confirmed a negative correlation between FGF10 expression and its antisense transcript (r=-0.02). CONCLUSION The relationship between the expression levels of FGF10 and FGF10AS in tumour tissues and adjacent normal tissues indicated that sense and antisense FGF RNAs could be remarkable prognostic biomarkers for achieving effective and primitive treatment.
Collapse
Affiliation(s)
- Leili Rejali
- Department of Biology, Faculty of Biological Sciences, Islamic Azad University, North Tehran Branch, Tehran, Iran,Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Centre, Research Institute for Gastroenterology and Liver
Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Seyed Yoosef Seyedna
- Department of Biology, Faculty of Biological Sciences, Islamic Azad University, North Tehran Branch, Tehran, Iran
| | - Hamid Asadzadeh Aghdaei
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Centre, Research Institute for Gastroenterology and Liver
Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Ehsan Nazemalhosseini Mojarad
- Gastroenterology and Liver Diseases Research Centre, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti
University of Medical Sciences, Tehran, Iran
| | - Mehrdad Hashemi
- Department of Genetics, Faculty of Advanced Science and Technology, Islamic Azad University, Tehran Medical Sciences, Tehran, Iran,Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital, Islamic Azad University, Tehran Medical
Sciences, Tehran, Iran,P.O.Box: 1916893813Department of GeneticsFaculty of Advanced Science and TechnologyIslamic Azad UniversityTehran Medical SciencesTehranIran
| |
Collapse
|
16
|
Wei W, Zhao X, Liu J, Zhang Z. Downregulation of LINC00665 suppresses the progression of lung adenocarcinoma via regulating miR-181c-5p/ZIC2 axis. Aging (Albany NY) 2021; 13:17499-17515. [PMID: 34232917 PMCID: PMC8312465 DOI: 10.18632/aging.203240] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Accepted: 05/31/2021] [Indexed: 12/24/2022]
Abstract
Long non-coding RNA (lncRNA) LINC00665 was demonstrated to be upregulated in lung adenocarcinoma (LUAD) and target miR-181c-5p. ZIC2, which is upregulated in LUAD, serves as a putative target of miR-181c-5p. In this study, we aimed to reveal whether LINC00665 regulates miR-181c-5p/ZIC2 axis to promote LUAD progression. The results showed that LINC00665, HOXA1, ZIC2, and HOXA11 levels were increased in LUAD tissues, while miR-181c-5p level was decreased when compared to the adjacent normal tissues. High expression levels of LINC00665, ZIC2, HOXA1 and HOXA11, and low expression of miR-181c-5p were closely linked to poor prognosis of LUAD patients. Knockdown of LINC00665 induced obvious inhibitions in cell viability, clone formation, invasion and tumorigenesis in LUAD cells, whereas miR-181c-5p downregulation significantly neutralized these effects. In addition, downregulation of ZIC2 obviously reversed the enhancements of cell viability, clone formation, invasion and tumorigenesis induced by miR-181c-5p knockdown. In summary, the present study reveals that silencing of LINC00665 suppresses LUAD progression through targeting miR-181c-5p/ZIC2 axis.
Collapse
Affiliation(s)
- Wei Wei
- Department of Lung Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin’s Clinical Research Center for Cancer, Tianjin Lung Cancer Center, Tianjin 300060, China
| | - Xiaoliang Zhao
- Department of Lung Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin’s Clinical Research Center for Cancer, Tianjin Lung Cancer Center, Tianjin 300060, China
| | - Jiang Liu
- Department of Molecule Imaging and Nuclear Medicine in Diagnosis and Treatment, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin’s Clinical Research Center for Cancer, Tianjin Lung Cancer Center, Tianjin 300060, China
| | - Zhenfa Zhang
- Department of Lung Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin’s Clinical Research Center for Cancer, Tianjin Lung Cancer Center, Tianjin 300060, China
| |
Collapse
|
17
|
Wang A, Zhang T, Wei W, Wang H, Zhang Z, Yang W, Xia W, Mao Q, Xu L, Jiang F, Dong G. The Long Noncoding RNA LINC00665 Facilitates c-Myc Transcriptional Activity via the miR-195-5p MYCBP Axis to Promote Progression of Lung Adenocarcinoma. Front Oncol 2021; 11:666551. [PMID: 34277412 PMCID: PMC8281894 DOI: 10.3389/fonc.2021.666551] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Accepted: 06/15/2021] [Indexed: 12/21/2022] Open
Abstract
Long noncoding RNAs (lncRNAs) have recently received growing substantial attention in cancer research due to their important roles in various cancer types. However, the underlying mechanisms and functions of lncRNAs, especially in lung adenocarcinoma (LUAD), remain elusive. Based on pan-cancer screening analyses, we identified that the noncoding RNA LINC00665 was up-regulated in lung adenocarcinoma, which was subsequently confirmed in clinical samples and cell lines. Higher expression of LINC00665 was positively associated with poor prognosis and advanced T stage. Next, using gain- and loss- of function approaches, we revealed that LINC00665 promotes cell proliferation, cell migration, invasion, and suppresses cell apoptosis in LUAD through in vitro and in vivo experiments. Additionally, our findings showed that LINC00665 was predominately localized in the cytoplasm so as to interact with Ago2 protein, which could function as miRNA sponges. The results of bioinformatics prediction and RNA pull-down assay indicated that LINC00665 directly interacted with miR-195-5p. This was also confirmed by fluorescence colocalization. Furthermore, luciferase reporter assay demonstrated that Myc binding protein (MYCBP, also called AMY-1), which enhanced c-Myc transcriptional activity, was the target gene of LINC00665 dependent on miR-195-5p. Finally, rescue functional assay results uncovered that the oncogenic capability of LINC00665 was dependent on miR-195-5p and c-Myc transcriptional activity. In summary, this work elucidates that LINC00665 accelerates LUAD progression via the miR-195-5p/MYCBP axis by acting as a competing endogenous RNA (ceRNA), suggesting that LINC00665 may represent a potential therapeutic target for clinical intervention of LUAD.
Collapse
Affiliation(s)
- Anpeng Wang
- Department of Thoracic Surgery, Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research & The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, China.,Department of Geriatric Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China.,Jiangsu Key Laboratory of Molecular and Translational Cancer Research, Cancer Institute of Jiangsu Province, Nanjing, China.,The Fourth Clinical College of Nanjing Medical University, Nanjing, China
| | - Te Zhang
- Department of Thoracic Surgery, Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research & The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, China.,Jiangsu Key Laboratory of Molecular and Translational Cancer Research, Cancer Institute of Jiangsu Province, Nanjing, China.,The Fourth Clinical College of Nanjing Medical University, Nanjing, China
| | - Wei Wei
- Department of Thoracic Surgery, Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research & The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, China.,Jiangsu Key Laboratory of Molecular and Translational Cancer Research, Cancer Institute of Jiangsu Province, Nanjing, China
| | - Hui Wang
- Department of Thoracic Surgery, Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research & The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, China.,Jiangsu Key Laboratory of Molecular and Translational Cancer Research, Cancer Institute of Jiangsu Province, Nanjing, China.,The Fourth Clinical College of Nanjing Medical University, Nanjing, China
| | - Zeyu Zhang
- Department of Thoracic Surgery, Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research & The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, China.,Jiangsu Key Laboratory of Molecular and Translational Cancer Research, Cancer Institute of Jiangsu Province, Nanjing, China.,The Fourth Clinical College of Nanjing Medical University, Nanjing, China
| | - Wenming Yang
- Department of Thoracic Surgery, Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research & The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, China.,Jiangsu Key Laboratory of Molecular and Translational Cancer Research, Cancer Institute of Jiangsu Province, Nanjing, China.,The Fourth Clinical College of Nanjing Medical University, Nanjing, China
| | - Wenjie Xia
- Department of Thoracic Surgery, Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research & The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, China.,Jiangsu Key Laboratory of Molecular and Translational Cancer Research, Cancer Institute of Jiangsu Province, Nanjing, China
| | - Qixing Mao
- Department of Thoracic Surgery, Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research & The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, China.,Jiangsu Key Laboratory of Molecular and Translational Cancer Research, Cancer Institute of Jiangsu Province, Nanjing, China
| | - Lin Xu
- Department of Thoracic Surgery, Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research & The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, China.,Jiangsu Key Laboratory of Molecular and Translational Cancer Research, Cancer Institute of Jiangsu Province, Nanjing, China
| | - Feng Jiang
- Department of Thoracic Surgery, Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research & The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, China.,Jiangsu Key Laboratory of Molecular and Translational Cancer Research, Cancer Institute of Jiangsu Province, Nanjing, China.,The Fourth Clinical College of Nanjing Medical University, Nanjing, China
| | - Gaochao Dong
- Department of Thoracic Surgery, Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research & The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, China.,Jiangsu Key Laboratory of Molecular and Translational Cancer Research, Cancer Institute of Jiangsu Province, Nanjing, China.,The Fourth Clinical College of Nanjing Medical University, Nanjing, China
| |
Collapse
|
18
|
Gui T, Yao C, Jia B, Shen K. Identification and analysis of genes associated with epithelial ovarian cancer by integrated bioinformatics methods. PLoS One 2021; 16:e0253136. [PMID: 34143800 PMCID: PMC8213194 DOI: 10.1371/journal.pone.0253136] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Accepted: 05/31/2021] [Indexed: 12/24/2022] Open
Abstract
Background Though considerable efforts have been made to improve the treatment of epithelial ovarian cancer (EOC), the prognosis of patients has remained poor. Identifying differentially expressed genes (DEGs) involved in EOC progression and exploiting them as novel biomarkers or therapeutic targets is of great value. Methods Overlapping DEGs were screened out from three independent gene expression omnibus (GEO) datasets and were subjected to Gene ontology (GO) and Kyoto encyclopedia of genes and genomes (KEGG) pathway enrichment analyses. The protein-protein interactions (PPI) network of DEGs was constructed based on the STRING database. The expression of hub genes was validated in GEPIA and GEO. The relationship of hub genes expression with tumor stage and overall survival and progression-free survival of EOC patients was investigated using the cancer genome atlas data. Results A total of 306 DEGs were identified, including 265 up-regulated and 41 down-regulated. Through PPI network analysis, the top 20 genes were screened out, among which 4 hub genes, which were not researched in depth so far, were selected after literature retrieval, including CDC45, CDCA5, KIF4A, ESPL1. The four genes were up-regulated in EOC tissues compared with normal tissues, but their expression decreased gradually with the continuous progression of EOC. Survival curves illustrated that patients with a lower level of CDCA5 and ESPL1 had better overall survival and progression-free survival statistically. Conclusion Two hub genes, CDCA5 and ESPL1, identified as probably playing tumor-promotive roles, have great potential to be utilized as novel therapeutic targets for EOC treatment.
Collapse
Affiliation(s)
- Ting Gui
- Department of Obstetrics and Gynecology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Chenhe Yao
- Department of R&D Technology Center, Beijing Zhicheng Biomedical Technology Co, Ltd, Beijing, China
| | - Binghan Jia
- Department of R&D Technology Center, Beijing Zhicheng Biomedical Technology Co, Ltd, Beijing, China
| | - Keng Shen
- Department of Obstetrics and Gynecology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
- * E-mail:
| |
Collapse
|
19
|
Yan J, Li P, Gao R, Li Y, Chen L. Identifying Critical States of Complex Diseases by Single-Sample Jensen-Shannon Divergence. Front Oncol 2021; 11:684781. [PMID: 34150649 PMCID: PMC8212786 DOI: 10.3389/fonc.2021.684781] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Accepted: 04/29/2021] [Indexed: 12/23/2022] Open
Abstract
MOTIVATION The evolution of complex diseases can be modeled as a time-dependent nonlinear dynamic system, and its progression can be divided into three states, i.e., the normal state, the pre-disease state and the disease state. The sudden deterioration of the disease can be regarded as the state transition of the dynamic system at the critical state or pre-disease state. How to detect the critical state of an individual before the disease state based on single-sample data has attracted many researchers' attention. METHODS In this study, we proposed a novel approach, i.e., single-sample-based Jensen-Shannon Divergence (sJSD) method to detect the early-warning signals of complex diseases before critical transitions based on individual single-sample data. The method aims to construct score index based on sJSD, namely, inconsistency index (ICI). RESULTS This method is applied to five real datasets, including prostate cancer, bladder urothelial carcinoma, influenza virus infection, cervical squamous cell carcinoma and endocervical adenocarcinoma and pancreatic adenocarcinoma. The critical states of 5 datasets with their corresponding sJSD signal biomarkers are successfully identified to diagnose and predict each individual sample, and some "dark genes" that without differential expressions but are sensitive to ICI score were revealed. This method is a data-driven and model-free method, which can be applied to not only disease prediction on individuals but also targeted drug design of each disease. At the same time, the identification of sJSD signal biomarkers is also of great significance for studying the molecular mechanism of disease progression from a dynamic perspective.
Collapse
Affiliation(s)
- Jinling Yan
- School of Mathematics and Statistics, Henan University of Science and Technology, Luoyang, China
| | - Peiluan Li
- School of Mathematics and Statistics, Henan University of Science and Technology, Luoyang, China
| | - Rong Gao
- School of Mathematics and Statistics, Henan University of Science and Technology, Luoyang, China
| | - Ying Li
- School of Mathematics and Statistics, Henan University of Science and Technology, Luoyang, China
| | - Luonan Chen
- State Key Laboratory of Cell Biology, Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai, China
- Center for Excellence in Animal Evolution and Genetics, Chinese Academy of Sciences, Kunming, China
- Key Laboratory of Systems Health Science of Zhejiang Province, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, China
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| |
Collapse
|
20
|
Zhang X, Wu J. LINC00665 promotes cell proliferation, invasion, and metastasis by activating the TGF-β pathway in gastric cancer. Pathol Res Pract 2021; 224:153492. [PMID: 34091388 DOI: 10.1016/j.prp.2021.153492] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Revised: 05/16/2021] [Accepted: 05/20/2021] [Indexed: 12/20/2022]
Abstract
BACKGROUND AND AIMS Accumulating studies have demonstrated that long noncoding RNA plays a vital role in cancer progression. A previous study reported that LINC00665 was overexpressed and acted as a key tumor promoter in lung cancer, but the role of LINC00665 in gastric cancer (GC) remained uncharacterized. Thus, this study aimed to explore the mechanism of LINC00665 in GC. METHODS LINC00665 expression was explored using the Cancer Genome Atlas (TCGA), and a meta-analysis was conducted to assess the expression and prognostic value of LINC00665 in GC from Gene Expression Omnibus databases and the TCGA dataset. Real-time polymerase chain reaction (RT-PCR) was then conducted to verify the LINC00665 expression in GC tissues and cell lines. The effects of LINC00665 on cell proliferation, invasion, metastasis, and cell cycle in GC were evaluated using the CCK-8, wound healing, Transwell, and flow cytometry assays. In vitro validation was also performed. RESULTS LINC00665 overexpression was found in GC, and LINC00665 upregulation was significantly related to poor overall survival and disease-free survival. LINC00665 expression was associated with tumor depth, lymph node metastasis, and TNM stage. Univariate and multivariate analyses proved that LINC00665 could be an independent prognostic biomarker in GC. LINC00665 knockdown subsequently inhibited cell proliferation, invasion, and metastasis in GC cell lines; promoted cell apoptosis; and arrested GC cell lines in the G0/G1 phase. Western blot analysis indicated that LINC00665 silencing inhibited epithelial-mesenchymal transition and decreased the expression levels of TGF-β1, Smad2, and α-SMA. CONCLUSION LINC00665 can be a potential diagnostic and prognostic biomarker for GC patients, and LINC00665 promotes GC cell proliferation, invasion, and metastasis by activating the TGF-β signal pathway.
Collapse
Affiliation(s)
- Xiaohu Zhang
- Department of General Surgery, Beijing Tongren Hospital, Capital Medical University, Beijing, 100730, China
| | - Jixiang Wu
- Department of General Surgery, Beijing Tongren Hospital, Capital Medical University, Beijing, 100730, China.
| |
Collapse
|
21
|
LINC00665 activates Wnt/β-catenin signaling pathway to facilitate tumor progression of colorectal cancer via upregulating CTNNB1. Exp Mol Pathol 2021; 120:104639. [PMID: 33865827 DOI: 10.1016/j.yexmp.2021.104639] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Revised: 03/29/2021] [Accepted: 04/13/2021] [Indexed: 01/03/2023]
Abstract
Background LINC00665 is a newly identified oncogene, which has been reported to be oncogene in various cancers. Nevertheless, its role in the progression of colorectal cancer (CRC) remains obscure to the extent. This study aimed at exploring the role and mechanism of LINC00665 in CRC progression. Materials and methods RNA and protein expression were detected via qRT-PCR and western blot. Functional assays were conducted to investigate the role of LINC00665 in the CRC cellular processes. TOP/FOP assay was performed to detect the activity of Wnt/β-catenin signaling pathway. Mechanism investigations were carried out to explore the regulatory relationship among genes. Results LINC00665 was overtly expressed in CRC cell lines at high levels. Functionally, silencing of LINC00665 could curb in vitro CRC cell growth, migration and invasion, while stimulating cell apoptosis. Mechanically, LINC00665 sponged miR-214-3p to up-regulate CTNNB1 expression, consequently activating Wnt/β-catenin signaling pathway. Furthermore, LINC00665 could bind to U2AF2 and enhance the association between U2AF2 and CTNNB1, increasing the stability of CTNNB1. CTNNB1 overexpression could reverse the suppressive effects of LINC00665 downregulation. Conclusion LINC00665 stimulates CRC progression through the activation of Wnt/β-catenin signaling pathway, which hopefully might be a therapeutic target for CRC.
Collapse
|
22
|
Yang X, Wang Y, Pang S, Li X, Wang P, Ma R, Ma Y, Song C. LINC00665 promotes the progression of acute myeloid leukemia by regulating the miR-4458/DOCK1 pathway. Sci Rep 2021; 11:5009. [PMID: 33658535 PMCID: PMC7930206 DOI: 10.1038/s41598-021-82834-9] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Accepted: 01/21/2021] [Indexed: 12/14/2022] Open
Abstract
This study aimed to explore the role of LINC00665, miR-4458 and DOCK1 and their interactions in the development of acute myeloid leukemia (AML). The relative expression of LINC00665, miR-4458 and DOCK1 in AML samples was measured using qRT-PCR, and the protein level of DOCK1 in AML cell lines was examined using western blot. CCK8, BrdU, transwell, cell adhesion, and caspase-3 activity assays were carried out to evaluate the viability, proliferation, migration, adhesion, and apoptosis of AML cells, respectively. Luciferase reporter, RIP, and RNA pull-down assays were also performed to confirm the target relationship among LINC00665, miR-4458 and DOCK1. Findings revealed that LINC00665 and DOCK1 were aberrantly overexpressed in AML tissues and that the expression of miR-4458 was low in AML tissues. Silencing LINC00665 or DOCK1 presented significant restriction to the proliferation, migration and adhesion of AML cells. Apart from that, it was found that inhibiting miR-4458 could enhance the proliferation, migration and adhesion of AML cells but suppress the apoptosis of AML cells. Experimental results also indicated that LINC00665 exerted its positive function on AML cells by sponging miR-4458 and that miR-4458 influenced the progression of AML cells by targeting DOCK1 directly. Overall, this finding not only provided a novel molecular pathway for the diagnosis and treatment of AML but also showed that LINC00665 could enhance the progression of AML by regulating the miR-4458/DOCK1 pathway.
Collapse
MESH Headings
- Adult
- Aged
- Apoptosis/genetics
- Base Pairing
- Case-Control Studies
- Cell Adhesion
- Cell Line, Tumor
- Cell Movement
- Cell Proliferation
- Disease Progression
- Female
- Gene Expression Regulation, Neoplastic
- Gene Regulatory Networks
- HL-60 Cells
- Humans
- Interferon Regulatory Factors/genetics
- Interferon Regulatory Factors/metabolism
- Leukemia, Myeloid, Acute/diagnosis
- Leukemia, Myeloid, Acute/genetics
- Leukemia, Myeloid, Acute/metabolism
- Leukemia, Myeloid, Acute/pathology
- Male
- MicroRNAs/antagonists & inhibitors
- MicroRNAs/genetics
- MicroRNAs/metabolism
- Middle Aged
- RNA, Long Noncoding/genetics
- RNA, Long Noncoding/metabolism
- RNA, Small Interfering/genetics
- RNA, Small Interfering/metabolism
- Signal Transduction
- rac GTP-Binding Proteins/antagonists & inhibitors
- rac GTP-Binding Proteins/genetics
- rac GTP-Binding Proteins/metabolism
Collapse
Affiliation(s)
- Xiaoyu Yang
- Department of Hematology, The Fifth Affiliated Hospital of Zhengzhou University, No.3 Kangfu Front Road, ZhengzhouHenan, 450052, China.
| | - Yan Wang
- Department of Hematology, The Fifth Affiliated Hospital of Zhengzhou University, No.3 Kangfu Front Road, ZhengzhouHenan, 450052, China
| | - Sulei Pang
- Department of Hematology, The Fifth Affiliated Hospital of Zhengzhou University, No.3 Kangfu Front Road, ZhengzhouHenan, 450052, China
| | - Xiaojie Li
- Department of Hematology, The Fifth Affiliated Hospital of Zhengzhou University, No.3 Kangfu Front Road, ZhengzhouHenan, 450052, China
| | - Panpan Wang
- Department of Hematology, The Fifth Affiliated Hospital of Zhengzhou University, No.3 Kangfu Front Road, ZhengzhouHenan, 450052, China
| | - Ruojin Ma
- Department of Hematology, The Fifth Affiliated Hospital of Zhengzhou University, No.3 Kangfu Front Road, ZhengzhouHenan, 450052, China
| | - Yunyun Ma
- Department of Hematology, The Fifth Affiliated Hospital of Zhengzhou University, No.3 Kangfu Front Road, ZhengzhouHenan, 450052, China
| | - Chunge Song
- Department of Hematology, The Fifth Affiliated Hospital of Zhengzhou University, No.3 Kangfu Front Road, ZhengzhouHenan, 450052, China
| |
Collapse
|
23
|
Sharma A, Colonna G. System-Wide Pollution of Biomedical Data: Consequence of the Search for Hub Genes of Hepatocellular Carcinoma Without Spatiotemporal Consideration. Mol Diagn Ther 2021; 25:9-27. [PMID: 33475988 PMCID: PMC7847983 DOI: 10.1007/s40291-020-00505-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/01/2020] [Indexed: 12/17/2022]
Abstract
Biomedical institutions rely on data evaluation and are turning into data factories. Big-data storage centers, supercomputing systems, and increased algorithmic efficiency allow us to analyze the ever-increasing amount of data generated every day in biomedical research centers. In network science, the principal intrinsic problem is how to integrate the data and information from different experiments on genes or proteins. Data curation is an essential process in annotating new functional data to known genes or proteins, undertaken by a biobank curator, which is then reflected in the calculated networks. We provide an example of how protein-protein networks today have space-time limits. The next step is the integration of data and information from different biobanks. Omics data and networks are essential parts of this step but also have flawed protocols and errors. Consider data from patients with cancer: from biopsy procedures to experimental tests, to archiving methods and computational algorithms, these are continuously handled so require critical and continuous "updates" to obtain reproducible, reliable, and correct results. We show, as a second example, how all this distorts studies in cellular hepatocellular carcinoma. It is not unlikely that these flawed data have been polluting biobanks for some time before stringent conditions for the veracity of data were implemented in Big data. Therefore, all this could contribute to errors in future medical decisions.
Collapse
Affiliation(s)
- Ankush Sharma
- Department of Biosciences, University of Oslo, Oslo, Norway.
- Department of Informatics, University of Oslo, Oslo, Norway.
- Institute of Cancer Research, Institute of Clinical medicine, University of Oslo, Oslo, Norway.
| | - Giovanni Colonna
- Medical Informatics, AOU-Vanvitelli, Università della Campania, Naples, Italy
| |
Collapse
|
24
|
Wang X, Wang Y, Lin F, Xu M, Zhao X. Long non-coding RNA LINC00665 promotes melanoma cell growth and migration via regulating the miR-224-5p/VMA21 axis. Exp Dermatol 2020; 31:64-73. [PMID: 33247967 DOI: 10.1111/exd.14246] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Revised: 10/15/2020] [Accepted: 11/11/2020] [Indexed: 12/12/2022]
Abstract
Melanoma is an aggressive malignant skin tumor endangering the health of patients. Long non-coding RNAs (lncRNAs) and microRNAs (miRNAs) have been increasingly reported to be implicated in the carcinogenesis of melanoma. Long intergenic non-coding RNA 00665 (LINC00665) has been found to exert important regulatory roles in some cancers, yet its function in melanoma remains to be investigated. QRT-PCR analysis was conducted to evaluate the relative expression of RNAs. Functional experiments in vitro including colony formation, EdU, wound-healing and transwell assays, as well as in vivo xenograft assays, were utilized to study the role of LINC00665 in melanoma. Mechanical experiments were implemented to probe into the molecular linkage of LINC00665, miR-224-5p and VMA21. LINC00665 was abnormally highly expressed in melanoma cells. Silencing LINC00665 could inhibit the proliferation and migration of melanoma cells. LINC00665 sponged miR-224-5p to upregulate VMA21. VMA21 knockdown exerted similarly interfering effects on above biological processes in melanoma cells. However, VMA21 overexpression abolished the in vitro and in vivo outcomes of LINC00665 silencing. LINC00665 promotes proliferative and migrating abilities of melanoma cells via targeting miR-224-5p/VMA21 axis.
Collapse
Affiliation(s)
- Xiaonan Wang
- Orthopaedic Medical Center, The Second Hospital of Jilin University, Changchun, China
| | - Yanbing Wang
- Orthopaedic Medical Center, The Second Hospital of Jilin University, Changchun, China
| | - Feifei Lin
- Orthopaedic Medical Center, The Second Hospital of Jilin University, Changchun, China
| | - Meng Xu
- Orthopaedic Medical Center, The Second Hospital of Jilin University, Changchun, China
| | - Xin Zhao
- Orthopaedic Medical Center, The Second Hospital of Jilin University, Changchun, China
| |
Collapse
|
25
|
Peng X, Wang J, Li D, Chen X, Liu K, Zhang C, Lai Y. Identification of grade-related genes and construction of a robust genomic-clinicopathologic nomogram for predicting recurrence of bladder cancer. Medicine (Baltimore) 2020; 99:e23179. [PMID: 33217824 PMCID: PMC7676566 DOI: 10.1097/md.0000000000023179] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND Bladder cancer (BC) is a common tumor in the urinary system with a high recurrence rate. The individualized treatment and follow-up after surgery is the key to a successful outcome. Currently, the surveillance strategies are mainly depending on tumor stage and grade. Previous evidence has proved that tumor grade was a significant and independent risk factor of BC recurrence. Exploring the grade-related genes may provide us a new approach to predict prognosis and guide the post-operative treatment in BC patients. METHODS In this study, the weighted gene co-expression network analysis was applied to identify the hub gene module correlated with BC grade using GSE71576. After constructing a protein-protein interaction (PPI) network with the hub genes inside the hub gene module, we identified some potential core genes. TCGA and another independent dataset were used for further validation. RESULTS The results revealed that the expression of AURKA, CCNA2, CCNB1, KIF11, TTK, BUB1B, BUB1, and CDK1 were significantly higher in high-grade BC, showing a strong ability to distinguish BC grade. The expression levels of the 8 genes in normal, paracancerous, tumorous, and recurrent bladder tissues were progressively increased. By conducting survival analysis, we proved their prognostic value in predicting the recurrence of BC. Eventually, we constructed a prognostic nomogram by combining the 8-core-gene panel with clinicopathologic features, which had shown great performance in predicting the recurrence of BC. CONCLUSION We identified 8 core genes that revealed a significant correlation with the tumor grade as well as the recurrence of BC. Finally, we proved the value of a novel prognostic nomogram for predicting the relapse-free survival of BC patients after surgery, which could guide their treatment and follow-up.
Collapse
Affiliation(s)
- Xiqi Peng
- Guangdong and Shenzhen Key Laboratory of Male Reproductive Medicine and Genetics, Peking University Shenzhen Hospital, Shenzhen
- Shantou University Medical College, Shantou, Guangdong
| | - Jingyao Wang
- Guangdong and Shenzhen Key Laboratory of Male Reproductive Medicine and Genetics, Peking University Shenzhen Hospital, Shenzhen
| | - Dongna Li
- Shantou University Medical College, Shantou, Guangdong
| | - Xuan Chen
- Guangdong and Shenzhen Key Laboratory of Male Reproductive Medicine and Genetics, Peking University Shenzhen Hospital, Shenzhen
- Shantou University Medical College, Shantou, Guangdong
| | - Kaihao Liu
- Guangdong and Shenzhen Key Laboratory of Male Reproductive Medicine and Genetics, Peking University Shenzhen Hospital, Shenzhen
- Anhui Medical University, Hefei, Anhui, China
| | - Chunduo Zhang
- Guangdong and Shenzhen Key Laboratory of Male Reproductive Medicine and Genetics, Peking University Shenzhen Hospital, Shenzhen
| | - Yongqing Lai
- Guangdong and Shenzhen Key Laboratory of Male Reproductive Medicine and Genetics, Peking University Shenzhen Hospital, Shenzhen
| |
Collapse
|
26
|
Ding J, Zhao J, Huan L, Liu Y, Qiao Y, Wang Z, Chen Z, Huang S, Zhao Y, He X. Inflammation-Induced Long Intergenic Noncoding RNA (LINC00665) Increases Malignancy Through Activating the Double-Stranded RNA-Activated Protein Kinase/Nuclear Factor Kappa B Pathway in Hepatocellular Carcinoma. Hepatology 2020; 72:1666-1681. [PMID: 32083756 DOI: 10.1002/hep.31195] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2019] [Revised: 01/09/2020] [Accepted: 01/26/2020] [Indexed: 12/20/2022]
Abstract
BACKGROUND AND AIMS The nuclear factor kappa B (NF-κB) signaling pathway is important for linking inflammation and tumorigenesis. Here, we characterized an NF-κB signaling activation-induced long intergenic noncoding (LINC) RNA in hepatocellular carcinoma (HCC), LINC00665, that contributes to the enhanced cell proliferation of HCC cells both in vitro and in vivo. APPROACH AND RESULTS LINC00665 physically interacts with the double-stranded RNA (dsRNA)-activated protein kinase (PKR), enhances its activation, and maintains its protein stability by blocking ubiquitin/proteasome-dependent degradation, resulting in a positive feedback regulation of NF-κB signaling in HCC cells. Notably, patients with HCC and higher LINC00665 have poorer outcomes in the clinic. CONCLUSIONS Our findings indicate that LINC00665 is involved in the NF-κB signaling activation in HCC cells and that the inflammatory LINC00665/PKR/NF-κB loop plays important oncogenic roles in hepatic cancer progression and may be a potential therapeutic target.
Collapse
Affiliation(s)
- Jie Ding
- Fudan University Shanghai Cancer Center and Institutes of Biomedical Sciences, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Jingjing Zhao
- Fudan University Shanghai Cancer Center and Institutes of Biomedical Sciences, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Lin Huan
- Fudan University Shanghai Cancer Center and Institutes of Biomedical Sciences, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yizhe Liu
- Fudan University Shanghai Cancer Center and Institutes of Biomedical Sciences, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yejun Qiao
- Fudan University Shanghai Cancer Center and Institutes of Biomedical Sciences, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Zhen Wang
- Fudan University Shanghai Cancer Center and Institutes of Biomedical Sciences, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Zhiao Chen
- Fudan University Shanghai Cancer Center and Institutes of Biomedical Sciences, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Shenglin Huang
- Fudan University Shanghai Cancer Center and Institutes of Biomedical Sciences, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.,Key Laboratory of Breast Cancer in Shanghai, Fudan University Shanghai Cancer Center, Fudan University, Shanghai, China
| | - Yingjun Zhao
- Fudan University Shanghai Cancer Center and Institutes of Biomedical Sciences, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.,Key Laboratory of Breast Cancer in Shanghai, Fudan University Shanghai Cancer Center, Fudan University, Shanghai, China
| | - Xianghuo He
- Fudan University Shanghai Cancer Center and Institutes of Biomedical Sciences, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.,Key Laboratory of Breast Cancer in Shanghai, Fudan University Shanghai Cancer Center, Fudan University, Shanghai, China
| |
Collapse
|
27
|
Lv M, Mao Q, Li J, Qiao J, Chen X, Luo S. Knockdown of LINC00665 inhibits proliferation and invasion of breast cancer via competitive binding of miR-3619-5p and inhibition of catenin beta 1. Cell Mol Biol Lett 2020; 25:43. [PMID: 32983239 PMCID: PMC7513511 DOI: 10.1186/s11658-020-00235-8] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Accepted: 09/09/2020] [Indexed: 12/12/2022] Open
Abstract
Background Long intergenic non-protein coding RNA00665 (LINC00665) plays a crucial tumorigenic role in many cancers, such as gastric cancer and lung adenocarcinoma. However, its role and mechanism of action in the progression of breast cancer (BC) are unknown. Methods LINC00665 expression levels were determined using quantitative polymerase chain reaction analysis with BC tissues and cell lines. BC cell proliferation was tested by performing 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assays, whereas BC cell migration and invasion capabilities were analyzed by performing transwell migration assays. Percentages of apoptotic cells were measured by flow cytometry. Interactions between LINC00665 and miR-3169-5p were examined by performing luciferase reporter assays, and the expression levels of proteins, such as β-catenin, were examined by western blot analysis. Results LINC00665 was expressed at high levels in BC tissues and cells. Upregulated LINC00665 expression correlated with tumor size and tumor, node, and metastasis stages, but not with the age of patients. LINC00665 knockdown inhibited BC cell proliferation, migration, and invasion, whereas it promoted apoptosis. Moreover, bioinformatics analysis and the luciferase reporter assay revealed that LINC00665 bound the microRNA (miR) miR-3619-5p. miR-3619-5p expression correlated negatively with LINC00665 expression in BC tissues. miR-3619-5p overexpression inhibited BC cell proliferation, migration, and invasion, but promoted apoptosis. Simultaneous knockdown of LINC00665 and miR-3619-5p led to increased cell proliferation, migration, and invasion, and inhibited apoptosis. Additionally, catenin beta 1, which encodes the β-catenin protein, was the target gene of miR-3619-5p. β-catenin expression clearly decreased after LINC00665 knockdown and miR-3619-5p overexpression, but increased after simultaneous knockdown of LINC00665 and miR-3619-5p. Conclusion LINC00665 knockdown inhibited BC cell proliferation and invasion by binding miR-3619-5p and inhibiting β-catenin expression.
Collapse
Affiliation(s)
- Minhao Lv
- Department of Breast Surgery, The Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, Henan P.R. China
| | - Qixin Mao
- Department of Breast Surgery, The Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, Henan P.R. China
| | - Juntao Li
- Department of Breast Surgery, The Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, Henan P.R. China
| | - Jianghua Qiao
- Department of Breast Surgery, The Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, Henan P.R. China
| | - Xiuchun Chen
- Department of Breast Surgery, The Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, Henan P.R. China
| | - Suxia Luo
- Department of Medical Oncology, The Affiliated Cancer Hospital of Zhengzhou University, No. 127, Dongming Road, Jinshui District, Zhengzhou, 450008 Henan P.R. China
| |
Collapse
|
28
|
Wang C, Li M, Wang S, Jiang Z, Liu Y. LINC00665 Promotes the Progression of Multiple Myeloma by Adsorbing miR-214-3p and Positively Regulating the Expression of PSMD10 and ASF1B. Onco Targets Ther 2020; 13:6511-6522. [PMID: 32764956 PMCID: PMC7368456 DOI: 10.2147/ott.s241627] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Accepted: 06/01/2020] [Indexed: 12/11/2022] Open
Abstract
Background Although assertion that long non-coding RNA (lncRNA) exerts crucial functions in the progression of multiple myeloma (MM) is well documented, few studies investigate function and underlying mechanism of long intergenic non-protein coding RNA 665 (LINC00665) in MM. Patients and Methods A total of 25 MM patient samples and 15 healthy volunteer samples were collected, and quantitative real-time polymerase chain reaction (qRT-PCR) was employed to detect the expressions of LINC00665. PSMD10 and ASF1B expressions were determined by qRT-PCR and Western blot assays. U266 cell and H929 cell were used in functional experiments. Besides, CCK-8 assay and flow cytometry analysis were utilized to determine cell proliferation and apoptosis. Bioinformatics analysis and dual-luciferase reporter assays were used to predict and verify the targeting relationships between LINC00665 and miR-214-3p, PSMD10 and miR-214-3p, as well as ASF1B and miR-214-3p. Moreover, the regulatory function of LINC00665 on the expression of PSMD10 and ASF1B was detected by Western blot. Results The expression of LINC00665 was up-regulated in MM samples and cell lines. In vitro functional assays indicated that LINC00665 enhanced MM cell proliferation and inhibited its apoptosis. PSMD10 and ASF1B were identified as target genes of miR-214-3p. Additionally, LINC00665 negatively regulated miR-214-3p expression through sponging miR-214-3p and positively regulated PSMD10 and ASF1B. Conclusion LINC00665 can promote the expression of PSMD10 and ASF1B by inhibiting the expression of miR-214-3p, thus facilitating the proliferation and inhibiting apoptosis of MM cells.
Collapse
Affiliation(s)
- Chong Wang
- Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan Province, People's Republic of China
| | - Mengya Li
- Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan Province, People's Republic of China
| | - Shujuan Wang
- Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan Province, People's Republic of China
| | - Zhongxing Jiang
- Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan Province, People's Republic of China
| | - Yanfang Liu
- Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan Province, People's Republic of China
| |
Collapse
|
29
|
Zhang Y, Tao Y, Ji H, Li W, Guo X, Ng DM, Haleem M, Xi Y, Dong C, Zhao J, Zhang L, Zhang X, Xie Y, Dai X, Liao Q. Genome-wide identification of the essential protein-coding genes and long non-coding RNAs for human pan-cancer. Bioinformatics 2020; 35:4344-4349. [PMID: 30923830 DOI: 10.1093/bioinformatics/btz230] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2018] [Revised: 01/17/2019] [Accepted: 03/26/2019] [Indexed: 01/11/2023] Open
Abstract
MOTIVATION Genome-scale CRISPR/Cas9 system has been a democratized gene editing technique and widely used to investigate gene functions in some biological processes and diseases especially cancers. Aiming to characterize gene aberrations and assess their effects on cancer, we designed a pipeline to identify the essential genes for pan-cancer. METHODS CRISPR screening data were used to identify the essential genes that were collected from published data and integrated by Robust Rank Aggregation algorithm. Then, hypergeometrics test and random walks with restart (RWR) were used to predict additional essential genes on broader scale. Finally, the expression status and potential roles of these genes were explored based on TCGA portal and regulatory network analysis. RESULTS We collected 926 samples from 10 CRISPR-based screening studies involving 33 different types of cancer to identify cancer-essential genes, which consists of 799 protein-coding genes (PCGs) and 97 long non-coding RNAs (lncRNAs). Then, we constructed a 'bi-colored' network with both PCGs and lncRNAs and applied it to predict additional essential genes including 495 PCGs and 280 lncRNAs on a broader scale using hypergeometrics test and RWR. After obtaining all essential genes, we further investigated their potential roles in cancer and found that essential genes have higher and more stable expression levels, and are associated with multiple cancer-associated biological processes and survival time. The regulatory network analysis detected two intriguing modules of essential genes participating in the regulation of cell cycle and ribosome biogenesis in cancer. AVAILABILITY AND IMPLEMENTATION . SUPPLEMENTARY INFORMATION Supplementary data are available at Bioinformatics online.
Collapse
Affiliation(s)
- Yuwei Zhang
- Department of Preventative Medicine, Zhejiang Provincial Key Laboratory of Pathological and Physiological Technology, Medicine School of Ningbo University, Ningbo, Zhejiang, China
| | - Yang Tao
- Department of Preventative Medicine, Zhejiang Provincial Key Laboratory of Pathological and Physiological Technology, Medicine School of Ningbo University, Ningbo, Zhejiang, China
| | - Huihui Ji
- Zhejiang Provincial Key Laboratory of Pathophysiology, School of Medicine, Ningbo University, Zhejiang, Ningbo, China
| | - Wei Li
- Center for Genetic Medicine Research, Children's National Medical Center, Department of Genomics and Precision Medicine, George Washington University, Washington, DC, USA
| | - Xingli Guo
- School of Computer Science and Technology, Xidian University, Xi'an, Shaanxi Province 710071, China
| | - Derry Minyao Ng
- Department of Preventative Medicine, Zhejiang Provincial Key Laboratory of Pathological and Physiological Technology, Medicine School of Ningbo University, Ningbo, Zhejiang, China
| | - Maria Haleem
- Department of Preventative Medicine, Zhejiang Provincial Key Laboratory of Pathological and Physiological Technology, Medicine School of Ningbo University, Ningbo, Zhejiang, China
| | - Yang Xi
- Zhejiang Provincial Key Laboratory of Pathophysiology, School of Medicine, Ningbo University, Zhejiang, Ningbo, China
| | - Changzheng Dong
- Department of Preventative Medicine, Zhejiang Provincial Key Laboratory of Pathological and Physiological Technology, Medicine School of Ningbo University, Ningbo, Zhejiang, China
| | - Jinshun Zhao
- Department of Preventative Medicine, Zhejiang Provincial Key Laboratory of Pathological and Physiological Technology, Medicine School of Ningbo University, Ningbo, Zhejiang, China
| | - Lina Zhang
- Department of Preventative Medicine, Zhejiang Provincial Key Laboratory of Pathological and Physiological Technology, Medicine School of Ningbo University, Ningbo, Zhejiang, China
| | - Xiaohong Zhang
- Department of Preventative Medicine, Zhejiang Provincial Key Laboratory of Pathological and Physiological Technology, Medicine School of Ningbo University, Ningbo, Zhejiang, China
| | - Yangyang Xie
- Anorectal Surgery, Ningbo Second Hospital, Ningbo, China
| | - Xiaoyu Dai
- Anorectal Surgery, Ningbo Second Hospital, Ningbo, China
| | - Qi Liao
- Department of Preventative Medicine, Zhejiang Provincial Key Laboratory of Pathological and Physiological Technology, Medicine School of Ningbo University, Ningbo, Zhejiang, China
| |
Collapse
|
30
|
Wang H, Wang L, Zhang S, Xu Z, Zhang G. Downregulation of LINC00665 confers decreased cell proliferation and invasion via the miR-138-5p/E2F3 signaling pathway in NSCLC. Biomed Pharmacother 2020; 127:110214. [PMID: 32403047 DOI: 10.1016/j.biopha.2020.110214] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Revised: 04/25/2020] [Accepted: 04/28/2020] [Indexed: 11/19/2022] Open
Abstract
Non-small cell lung cancer (NSCLC) is a type of malignant tumor which threatens human health and life. Recently, some researches on long non-coding RNAs (lncRNAs) in NSCLC has elucidated critical regulatory roles in cell proliferation, migration, and invasion, the relative clinical significance and mechanisms of action are still unclear. This study focuses on the important role of a novel lncRNA LINC00665 in the development of NSCLC. Long intergenic non-protein coding RNA 665 gene (LINC00665) was found through microarray analysis and was measured by real-time quantitative PCR (RT-qPCR). The interactions between LINC00665 and miR-138-5p as well as the interactions between miR-138-5p and E2F3 (E2F transcription factor 3) were explored by bioinformatics analysis and dual-luciferase assays. CCK-8, transwell and mouse xenograft assays were performed to investigate the effects of LINC00665 and miR-138-5p on NSCLC proliferation and invasion. As a result, LINC00665 expression was upregulated in NSCLC lung tissues and cells. Downregulated LINC00665 could arrest A549 and H1299 cell proliferation and invasion in vitro, and this finding was recapitulated in vivo. LINC00665 directly regulated the expression of miR-138-5p. Additionally, E2F3 was one of the targets of miR-138-5p; E2F3 without 3'UTR could reverse the inhibitory effects of downregulated LINC00665 on proliferation and invasion in A549 and H1299 cells. In conclusion, dysregulation of LINC00665 plays a vital role in NSCLC progression, indicating that its downregulation may confer decreased cell proliferation and invasion via the miR-138-5p/E2F3 signaling pathway.
Collapse
Affiliation(s)
- Huaqi Wang
- Department of Respiratory Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Lei Wang
- Department of Emergency, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China.
| | - Shijie Zhang
- Department of Clinical Laboratory, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Zhexuan Xu
- Department of Respiratory Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Guojun Zhang
- Department of Respiratory Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China.
| |
Collapse
|
31
|
Ruan X, Zheng J, Liu X, Liu Y, Liu L, Ma J, He Q, Yang C, Wang D, Cai H, Li Z, Liu J, Xue Y. lncRNA LINC00665 Stabilized by TAF15 Impeded the Malignant Biological Behaviors of Glioma Cells via STAU1-Mediated mRNA Degradation. MOLECULAR THERAPY-NUCLEIC ACIDS 2020; 20:823-840. [PMID: 32464546 PMCID: PMC7256440 DOI: 10.1016/j.omtn.2020.05.003] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Revised: 04/23/2020] [Accepted: 05/01/2020] [Indexed: 12/11/2022]
Abstract
Glioma is a brain cancer characterized by strong invasiveness with limited treatment options and poor prognosis. Recently, dysregulation of long non-coding RNAs (lncRNAs) has emerged as an important component in cellular processes and tumorigenesis. In this study, we demonstrated that TATA-box binding protein associated factor 15 (TAF15) and long intergenic non-protein coding RNA 665 (LINC00665) were both downregulated in glioma tissues and cells. TAF15 overexpression enhanced the stability of LINC00665, inhibiting malignant biological behaviors of glioma cells. Both metal regulatory transcription factor 1 (MTF1) and YY2 transcription factor (YY2) showed high expression levels in glioma tissues and cells, and their knockdown inhibited malignant progression. Mechanistically, overexpression of LINC00665 was confirmed to destabilize MTF1 and YY2 mRNA by interacting with STAU1, and knockdown of STAU1 could rescue the MTF1 and YY2 mRNA degradation caused by LINC00665 overexpression. G2 and S-phase expressed 1 (GTSE1) was identified as an oncogene in glioma, and knockdown of MTF1 or YY2 decreased the mRNA and protein expression levels of GTSE1 through direct binding to the GTSE1 promoter region. Our study highlights a key role of the TAF15/LINC00665/MTF1(YY2)/GTSE1 axis in modulating the malignant biological behaviors of glioma cells, suggesting novel mechanisms by which lncRNAs affect STAU1-mediated mRNA stability, which can inform new molecular therapies for glioma.
Collapse
Affiliation(s)
- Xuelei Ruan
- Department of Neurobiology, School of Life Sciences, China Medical University, Shenyang 110122, China; Key Laboratory of Cell Biology, Ministry of Public Health of China, China Medical University, Shenyang 110122, China; Key Laboratory of Medical Cell Biology, Ministry of Education of China, China Medical University, Shenyang 110122, China
| | - Jian Zheng
- Department of Neurosurgery, Shengjing Hospital of China Medical University, Shenyang 110004, China; Liaoning Clinical Medical Research Center in Nervous System Disease, Shenyang 110004, China; Key Laboratory of Neuro-oncology in Liaoning Province, Shenyang 110004, China
| | - Xiaobai Liu
- Department of Neurosurgery, Shengjing Hospital of China Medical University, Shenyang 110004, China; Liaoning Clinical Medical Research Center in Nervous System Disease, Shenyang 110004, China; Key Laboratory of Neuro-oncology in Liaoning Province, Shenyang 110004, China
| | - Yunhui Liu
- Department of Neurosurgery, Shengjing Hospital of China Medical University, Shenyang 110004, China; Liaoning Clinical Medical Research Center in Nervous System Disease, Shenyang 110004, China; Key Laboratory of Neuro-oncology in Liaoning Province, Shenyang 110004, China
| | - Libo Liu
- Department of Neurobiology, School of Life Sciences, China Medical University, Shenyang 110122, China; Key Laboratory of Cell Biology, Ministry of Public Health of China, China Medical University, Shenyang 110122, China; Key Laboratory of Medical Cell Biology, Ministry of Education of China, China Medical University, Shenyang 110122, China
| | - Jun Ma
- Department of Neurobiology, School of Life Sciences, China Medical University, Shenyang 110122, China; Key Laboratory of Cell Biology, Ministry of Public Health of China, China Medical University, Shenyang 110122, China; Key Laboratory of Medical Cell Biology, Ministry of Education of China, China Medical University, Shenyang 110122, China
| | - Qianru He
- Department of Neurobiology, School of Life Sciences, China Medical University, Shenyang 110122, China; Key Laboratory of Cell Biology, Ministry of Public Health of China, China Medical University, Shenyang 110122, China; Key Laboratory of Medical Cell Biology, Ministry of Education of China, China Medical University, Shenyang 110122, China
| | - Chunqing Yang
- Department of Neurosurgery, Shengjing Hospital of China Medical University, Shenyang 110004, China; Liaoning Clinical Medical Research Center in Nervous System Disease, Shenyang 110004, China; Key Laboratory of Neuro-oncology in Liaoning Province, Shenyang 110004, China
| | - Di Wang
- Department of Neurosurgery, Shengjing Hospital of China Medical University, Shenyang 110004, China; Liaoning Clinical Medical Research Center in Nervous System Disease, Shenyang 110004, China; Key Laboratory of Neuro-oncology in Liaoning Province, Shenyang 110004, China
| | - Heng Cai
- Department of Neurosurgery, Shengjing Hospital of China Medical University, Shenyang 110004, China; Liaoning Clinical Medical Research Center in Nervous System Disease, Shenyang 110004, China; Key Laboratory of Neuro-oncology in Liaoning Province, Shenyang 110004, China
| | - Zhen Li
- Department of Neurosurgery, Shengjing Hospital of China Medical University, Shenyang 110004, China; Liaoning Clinical Medical Research Center in Nervous System Disease, Shenyang 110004, China; Key Laboratory of Neuro-oncology in Liaoning Province, Shenyang 110004, China
| | - Jing Liu
- Department of Neurosurgery, Shengjing Hospital of China Medical University, Shenyang 110004, China; Liaoning Clinical Medical Research Center in Nervous System Disease, Shenyang 110004, China; Key Laboratory of Neuro-oncology in Liaoning Province, Shenyang 110004, China
| | - Yixue Xue
- Department of Neurobiology, School of Life Sciences, China Medical University, Shenyang 110122, China; Key Laboratory of Cell Biology, Ministry of Public Health of China, China Medical University, Shenyang 110122, China; Key Laboratory of Medical Cell Biology, Ministry of Education of China, China Medical University, Shenyang 110122, China.
| |
Collapse
|
32
|
Wu M, Shang X, Sun Y, Wu J, Liu G. Integrated analysis of lymphocyte infiltration-associated lncRNA for ovarian cancer via TCGA, GTEx and GEO datasets. PeerJ 2020; 8:e8961. [PMID: 32419983 PMCID: PMC7211406 DOI: 10.7717/peerj.8961] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2019] [Accepted: 03/23/2020] [Indexed: 12/18/2022] Open
Abstract
Background Abnormal expression of long non-coding RNAs (lncRNA) play a significant role in the incidence and progression of high-grade serous ovarian cancer (HGSOC), which is a leading cause of mortality among gynecologic malignant tumor patients. In this study, our aim is to identify lncRNA-associated competing endogenous RNA (ceRNA ) axes that could define more reliable prognostic parameters of HGSOC, and to investigate the lncRNAs’ potential mechanism of in lymphocyte infiltration. Methods The RNA-seq and miRNA expression profiles were downloaded from The Cancer Genome Atlas (TCGA) and the Genotype-Tissue Expression (GTEx) database; while for obtaining the differentially expressed lncRNAs (DELs), miRNAs (DEMs), and genes (DEGs), we used edgeR, limma and DESeq2. After validating the RNA, miRNA and gene expressions, using integrated three RNA expression profiles (GSE18520, GSE27651, GSE54388) and miRNA profile (GSE47841) from the Gene Expression Omnibus (GEO) database, we performed Gene Ontology (GO) and Kyoto Encyclopedia of Gene and Genome (KEGG) pathway analyses through ClusterProfiler. The prognostic value of these genes was determined with Kaplan–Meier survival analysis and Cox regression analysis. The ceRNA network was constructed using Cytoscape. The correlation between lncRNAs in ceRNA network and immune infiltrating cells was analyzed by using Tumor IMmune Estimation Resource (TIMER), and gene markers of tumor-infiltrating immune cells were identified using Spearman’s correlation after removing the influence of tumor purity. Results A total of 33 DELs (25 upregulated and eight downregulated), 134 DEMs (76 upregulated and 58 downregulated), and 1,612 DEGs (949 upregulated and 663 downregulated) were detected that could be positively correlated with overall survival (OS) of HGSOC. With the 1,612 analyzed genes, we constructed a ceRNA network, which indicated a pre-dominant involvement of the immune-related pathways. Furthermore, our data revealed that LINC00665 influenced the infiltration level of macrophages and dendritic cells (DCs). On the other hand, FTX and LINC00665, which may play their possible roles through the ceRNA axis, demonstrated a potential to inhibit Tregs and prevent T-cell exhaustion. Conclusion We defined several prognostic biomarkers for the incidence and progression of HGSOC and constructed a network for ceRNA axes; among which three were indicated to have a positive correlation with lymphocyte infiltration, namely: FTX-hsa-miR-150-5p-STK11, LINC00665-hsa-miR449b-5p-VAV3 and LINC00665-hsa-miR449b-5p-RRAGD.
Collapse
Affiliation(s)
- Meijing Wu
- Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, Tianjin, China
| | - Xiaobin Shang
- Department of Esophageal Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Yue Sun
- Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, Tianjin, China
| | - Jing Wu
- Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, Tianjin, China
| | - Guoyan Liu
- Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, Tianjin, China
| |
Collapse
|
33
|
Zhang L, Makamure J, Zhao D, Liu Y, Guo X, Zheng C, Liang B. Bioinformatics analysis reveals meaningful markers and outcome predictors in HBV-associated hepatocellular carcinoma. Exp Ther Med 2020; 20:427-435. [PMID: 32537007 PMCID: PMC7281962 DOI: 10.3892/etm.2020.8722] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2019] [Accepted: 12/05/2019] [Indexed: 12/18/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is the most common type of malignant neoplasm of the liver with high morbidity and mortality. Extensive research into the pathology of HCC has been performed; however, the molecular mechanisms underlying the development of hepatitis B virus-associated HCC have remained elusive. Thus, the present study aimed to identify critical genes and pathways associated with the development and progression of HCC. The expression profiles of the GSE121248 dataset were downloaded from the Gene Expression Omnibus database and the differentially expressed genes (DEGs) were identified. Gene Ontology (GO) and Kyoto Encyclopedia of Gene and Genome (KEGG) analyses were performed by using the Database for Annotation, Visualization and Integrated Discovery. Subsequently, protein-protein interaction (PPI) networks were constructed for detecting hub genes. In the present study, 1,153 DEGs (777 upregulated and 376 downregulated genes) were identified and the PPI network yielded 15 hub genes. GO analysis revealed that the DEGs were primarily enriched in ‘protein binding’, ‘cytoplasm’ and ‘extracellular exosome’. KEGG analysis indicated that DEGs were accumulated in ‘metabolic pathways’, ‘chemical carcinogenesis’ and ‘fatty acid degradation’. After constructing the PPI network, cyclin-dependent kinase 1, cyclin B1, cyclin A2, mitotic arrest deficient 2 like 1, cyclin B2, DNA topoisomerase IIα, budding uninhibited by benzimidazoles (BUB)1, TTK protein kinase, non-SMC condensin I complex subunit G, NDC80 kinetochore complex component, aurora kinase A, kinesin family member 11, cell division cycle 20, BUB1B and abnormal spindle microtubule assembly were identified as hub genes based on the high degree of connectivity by using Cytoscape software. In addition, overall survival (OS) and disease-free survival (DFS) analyses were performed using the Gene Expression Profiling Interactive Analysis online database, which revealed that the increased expression of all hub genes were associated with poorer OS and DFS outcomes. Receiver operating characteristic curves were constructed using GraphPad prism 7.0 software. The results confirmed that 15 hub genes were able to distinguish HCC form normal tissues. Furthermore, the expression levels of three key genes were analyzed in tumor and normal samples of the Human Protein Atlas database. The present results may provide further insight into the underlying mechanisms of HCC and potential therapeutic targets for the treatment of this disease.
Collapse
Affiliation(s)
- Lijie Zhang
- Department of Radiology, Hubei Key Laboratory of Molecular Imaging, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Joyman Makamure
- Department of Radiology, Hubei Key Laboratory of Molecular Imaging, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Dan Zhao
- Department of Radiology, Hubei Key Laboratory of Molecular Imaging, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Yiming Liu
- Department of Radiology, Hubei Key Laboratory of Molecular Imaging, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Xiaopeng Guo
- Department of Radiology, Hubei Key Laboratory of Molecular Imaging, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Chuansheng Zheng
- Department of Radiology, Hubei Key Laboratory of Molecular Imaging, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Bin Liang
- Department of Radiology, Hubei Key Laboratory of Molecular Imaging, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| |
Collapse
|
34
|
Zhu LJ, Pan Y, Chen XY, Hou PF. BUB1 promotes proliferation of liver cancer cells by activating SMAD2 phosphorylation. Oncol Lett 2020; 19:3506-3512. [PMID: 32269624 PMCID: PMC7114935 DOI: 10.3892/ol.2020.11445] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Accepted: 01/15/2020] [Indexed: 12/26/2022] Open
Abstract
Budding uninhibited by benzimidazoles 1 (BUB1) is a mitotic checkpoint serine/threonine kinase that has been reported as an oncogene or tumor suppressor gene in various types of cancer, including breast cancer, pancreatic ductal adenocarcinoma, prostate and gastric cancers. However, its role in liver cancer remains unclear. The present study aimed to explore the biological function of BUB1 in liver cancer. The present study demonstrated that BUB1 mRNA expression levels and the intensity of immunohistochemical staining were significantly increased in liver cancer tissues compared with normal tissues. The role of BUB1 in cell proliferation was also determined. Overexpression of BUB1 significantly promoted cell proliferation, whereas knockdown of BUB1 expression inhibited the proliferation of liver cancer cell lines. In experiments investigating the underlying mechanism, overexpression of BUB1 increased the levels of SMAD2 phosphorylation, whereas knockdown of BUB1 reduced the levels of SMAD2 phosphorylation. Therefore, BUB1 may promote proliferation of liver cancer cells by activating phosphorylation of SMAD2, and BUB1 may serve as a potential target in the diagnosis and/or treatment of liver cancer.
Collapse
Affiliation(s)
- Li-Jing Zhu
- Department of Radiation Oncology, Lianshui County People's Hospital, Huaian, Jiangsu 223400, P.R. China
| | - Yan Pan
- Department of Clinical Laboratory, Lianshui County People's Hospital, Huaian, Jiangsu 223400, P.R. China
| | - Xiao-Ying Chen
- Clinical Laboratory, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, P.R. China
| | - Pan-Fei Hou
- Department of Clinical Laboratory, Lianshui County People's Hospital, Huaian, Jiangsu 223400, P.R. China
| |
Collapse
|
35
|
LINC00665 promotes breast cancer progression through regulation of the miR-379-5p/LIN28B axis. Cell Death Dis 2020; 11:16. [PMID: 31907362 PMCID: PMC6944690 DOI: 10.1038/s41419-019-2213-x] [Citation(s) in RCA: 63] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2019] [Revised: 12/10/2019] [Accepted: 12/11/2019] [Indexed: 12/11/2022]
Abstract
Breast cancer is the most common malignant tumor among women worldwide. Although increasing evidence indicates that long noncoding RNAs (lncRNAs) play critical roles during breast tumorigenesis and progression, the involvement of most lncRNAs in breast cancer remains largely unknown. In the current study, we demonstrated that LINC00665 promotes breast cancer cell proliferation, migration, and invasion. Accumulating evidence indicates that many lncRNAs can function as endogenous miRNA sponges by competitively binding common miRNAs. In this study, we demonstrated that LINC00665 functions as a sponge for miR-379-5p, reducing the ability of miR-379-5p to repress LIN28B. LINC00665 promoted breast cancer progression and induced an epithelial-mesenchymal transition-like phenotype via the upregulation of LIN28B expression. Clinically, LINC00665 expression was increased but miR-379-5p expression was decreased in breast cancer tissues compared with that in normal breast tissues in the TCGA database. Furthermore, the expression of LINC00665 was negatively related with miR-379-5p expression. Collectively, our results reveal the LINC00665-miR-379-5p-LIN28B axis and shed light on breast cancer therapy.
Collapse
|
36
|
Gao L, Xiong DD, He RQ, Yang X, Lai ZF, Liu LM, Huang ZG, Wu HY, Yang LH, Ma J, Li SH, Lin P, Yang H, Luo DZ, Dang YW, Chen G. MIR22HG As A Tumor Suppressive lncRNA In HCC: A Comprehensive Analysis Integrating RT-qPCR, mRNA-Seq, And Microarrays. Onco Targets Ther 2019; 12:9827-9848. [PMID: 31819482 PMCID: PMC6875507 DOI: 10.2147/ott.s227541] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2019] [Accepted: 10/14/2019] [Indexed: 12/12/2022] Open
Abstract
Introduction MIR22HG has a reported involvement in the tumorigenesis of a variety of cancers, including hepatocellular carcinoma (HCC). However, the exact molecular mechanism of MIR22HG in HCC has not been clarified. Methods In the present study, we integrated data from in-house RT-qPCR, RNA-sequencing, microarray, and literature studies to conduct a comprehensive evaluation of the clinico-pathological and prognostic significance of MIR22HG in an extremely large group of HCC samples. We also explored the potential mechanism of MIR22HG in HCC by analyzing the alteration profiles of MIR22HG in HCC to predict transcription factors (TFs) that may interact with MIR22HG and to annotate the biological functions of genes co-expressed with MIR22HG. MIR22HG expression was also compared in HCC nude mice xenografts before and after a treatment with nitidine chloride. Results We found that MIR22HG was downregulated in HCC and that this downregulation correlated with the malignant phenotype of HCC. Comprehensive analysis of the prognostic impact of MIR22HG in HCC revealed a beneficial effect of MIR22HG on the survival outcome of HCC patients. Seven cases of MIR22HG deep deletion occurred in 360 of the cancer genome atlas (TCGA) provisional HCC samples. A total of 22 MIR22HG-TF-mRNA triplets in HCC were predicted by the lncRNAmap. Co-expressed genes of MIR22HG, identified by weighted correlation network analysis (WGCNA), mainly participated in the pathways involving osteoclast differentiation, chemokine signaling pathways, and hematopoietic cell lineage. In vivo experiments demonstrated that nitidine chloride could stimulate MIR22HG expression in HCC xenografts. Conclusion In summary, MIR22HG may play a tumor-suppressive role in HCC by coordinating with predicted TFs and co-expressed genes, such as NLRP3, CSF1R, SIGLEC10, and ZEB2, or by being controlled by nitidine chloride.
Collapse
Affiliation(s)
- Li Gao
- Department of Pathology, First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, Zhuang Autonomous Region 530021, People's Republic of China
| | - Dan-Dan Xiong
- Department of Pathology, First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, Zhuang Autonomous Region 530021, People's Republic of China
| | - Rong-Quan He
- Department of Medical Oncology, First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, Zhuang Autonomous Region 530021, People's Republic of China
| | - Xia Yang
- Department of Pathology, First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, Zhuang Autonomous Region 530021, People's Republic of China
| | - Ze-Feng Lai
- School of Pharmacy, Guangxi Medical University, Nanning, Guangxi, Zhuang Autonomous Region 530021, People's Republic of China
| | - Li-Min Liu
- School of Pharmacy, Guangxi Medical University, Nanning, Guangxi, Zhuang Autonomous Region 530021, People's Republic of China
| | - Zhi-Guang Huang
- Department of Pathology, First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, Zhuang Autonomous Region 530021, People's Republic of China
| | - Hua-Yu Wu
- Department of Cell Biology and Genetics, School of Pre-Clinical Medicine, Guangxi Medical University, Nanning, Guangxi, Zhuang Autonomous Region 530021, People's Republic of China
| | - Li-Hua Yang
- Department of Medical Oncology, First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, Zhuang Autonomous Region 530021, People's Republic of China
| | - Jie Ma
- Department of Medical Oncology, First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, Zhuang Autonomous Region 530021, People's Republic of China
| | - Sheng-Hua Li
- Department of Urology Surgery, First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, Zhuang Autonomous Region 530021, People's Republic of China
| | - Peng Lin
- Department of Ultrasound, First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, Zhuang Autonomous Region 530021, People's Republic of China
| | - Hong Yang
- Department of Ultrasound, First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, Zhuang Autonomous Region 530021, People's Republic of China
| | - Dian-Zhong Luo
- Department of Pathology, First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, Zhuang Autonomous Region 530021, People's Republic of China
| | - Yi-Wu Dang
- Department of Pathology, First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, Zhuang Autonomous Region 530021, People's Republic of China
| | - Gang Chen
- Department of Pathology, First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, Zhuang Autonomous Region 530021, People's Republic of China
| |
Collapse
|
37
|
Yang B, Bai Q, Chen H, Su K, Gao C. LINC00665 induces gastric cancer progression through activating Wnt signaling pathway. J Cell Biochem 2019; 121:2268-2276. [PMID: 31736127 DOI: 10.1002/jcb.29449] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2019] [Accepted: 10/10/2019] [Indexed: 12/14/2022]
Affiliation(s)
- Bo Yang
- Department of Oncology Affiliated Hospital of Xuzhou Medical University Xuzhou Jiangsu China
| | - Qingqing Bai
- Department of Oncology Suqian Hospital of Traditional Chinese Medicine Suqian Jiangsu China
| | - Huidong Chen
- Department of Oncology Suqian Hospital of Traditional Chinese Medicine Suqian Jiangsu China
| | - Kun Su
- Department of Oncology Suqian Hospital of Traditional Chinese Medicine Suqian Jiangsu China
| | - Chao Gao
- Department of Oncology Affiliated Hospital of Xuzhou Medical University Xuzhou Jiangsu China
| |
Collapse
|
38
|
Shan Y, Li P. Long Intergenic Non-Protein Coding RNA 665 Regulates Viability, Apoptosis, and Autophagy via the MiR-186-5p/MAP4K3 Axis in Hepatocellular Carcinoma. Yonsei Med J 2019; 60:842-853. [PMID: 31433582 PMCID: PMC6704017 DOI: 10.3349/ymj.2019.60.9.842] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/22/2018] [Revised: 03/23/2019] [Accepted: 03/29/2019] [Indexed: 02/07/2023] Open
Abstract
PURPOSE Long intergenic non-protein coding RNA 665 (LINC00665) plays a vital role in the development of cancer. Its function in hepatocellular carcinoma (HCC), however, remains largely unknown. MATERIALS AND METHODS The expressions of LINC00665, miR-186-5p, and MAP4K3 were determined by qRT-PCR. Cell viability and apoptosis were evaluated by MTT and flow cytometry, respectively. Autophagic puncta formation was observed by fluorescence microscopy. Bioinformatics analysis, luciferase reporter assay, RNA immunoprecipitation, and RNA pulldown were performed to identify associations among LINC00665, miR-186-5p, and MAP4K3. Western blot was utilized to examine the expressions of MAP4K3, Beclin-1, and LC3. Tumor growth was evaluated in a xenograft model. RESULTS Elevations in LINC00665 were observed in HCC tissues and cells. The overall survival of HCC patients with high levels of LINC00665 was shorter than those with low levels. In vitro, LINC00665 depletion inhibited viability and induced apoptosis and autophagy. miR-186-5p interacted with LINC00665 and was downregulated in HCC tissues and cells. Upregulation of miR-186-5p inhibited viability and induced apoptosis and autophagy, which were attenuated by upregulation of LINC00665. MAP4K3 was found to possess binding sites with miR-186-5p and was upregulated in HCC tissues and cells. MAP4K3 depletion inhibited viability and induced apoptosis and autophagy, which were attenuated by miR-186-5p inhibitor. In vivo, miR-186-5p expression was negatively correlated with LINC00665 or MAP4K3 in HCC tissues, while LINC00665 was positively correlated with MAP4K3. LINC00665 knockdown suppressed tumor growth. CONCLUSION LINC00665 was involved in cell viability, apoptosis, and autophagy in HCC via miR-186-5p/MAP4K3 axis, which may provide a new approach for HCC treatment.
Collapse
Affiliation(s)
- Yong Shan
- Department of General Surgery, Jinchang Central Hospital, Jinchang, Gansu, China.
| | - Ping Li
- Department of Ultrasound, Shenzhen Hospital of Southern Medical University, Shenzhen, China
| |
Collapse
|
39
|
Qi H, Xiao Z, Wang Y. Long non-coding RNA LINC00665 gastric cancer tumorigenesis by regulation miR-149-3p/RNF2 axis. Onco Targets Ther 2019; 12:6981-6990. [PMID: 31695413 PMCID: PMC6717843 DOI: 10.2147/ott.s214588] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2019] [Accepted: 07/30/2019] [Indexed: 12/16/2022] Open
Abstract
Background Recently, LINC00665 has been reported to be a pivotal regulator in kinds of malignancy, such as lung cancer and liver cancer. However, the functions and underlying mechanisms of LINC00665 in gastric cancer (GC) remain unclear. Materials and methods We recruited 49 paired GC tissue to explore LINC00665 expression by qRT-PCR. In vitro function assays were used to explore the roles of LINC00665 in GC progression. Moreover, the interaction among LINC00665, miR-149-3p and RNF2 was explored by bioinformatics analysis and luciferase reporter assay. Results In the present study, we found that LINC00665 expression was significantly elevated in GC tissues and cell lines. High LINC00665 expression was associated with TNM stage, histological grade, and poor prognosis of GC patients. Function assays showed that LINC00665 suppression significantly reduced GC cells viability and invasion ability in vitro. Mechanistic analysis showed that LINC00665 might serve as a ceRNA for miR-149-3p to regulate the expression of RNF2. Conclusion Our current study revealed the LINC00665/miR-149-3p/RNF2 axis was involved in GC progression, providing novel insights into the treatment for GC.
Collapse
Affiliation(s)
- Hongyang Qi
- Department of Gastroenterology, The Central Hospital of Xinxiang, Xinxiang, Henan 453000, People's Republic of China
| | - Zhanyu Xiao
- Department of Gastroenterology, The Central Hospital of Xinxiang, Xinxiang, Henan 453000, People's Republic of China
| | - Yunxi Wang
- Department of Gastroenterology, The Central Hospital of Xinxiang, Xinxiang, Henan 453000, People's Republic of China
| |
Collapse
|
40
|
Liu L, Lin J, He H. Identification of Potential Crucial Genes Associated With the Pathogenesis and Prognosis of Endometrial Cancer. Front Genet 2019; 10:373. [PMID: 31105744 PMCID: PMC6499025 DOI: 10.3389/fgene.2019.00373] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2019] [Accepted: 04/09/2019] [Indexed: 12/13/2022] Open
Abstract
Background and Objective Endometrial cancer (EC) is a common gynecological malignancy worldwide. Despite advances in the development of strategies for treating EC, prognosis of the disease remains unsatisfactory, especially for advanced EC. The aim of this study was to identify novel genes that can be used as potential biomarkers for identifying the prognosis of EC and to construct a novel risk stratification using these genes. Methods and Results An mRNA sequencing dataset, corresponding survival data and expression profiling of an array of EC patients were obtained from The Cancer Genome Atlas and Gene Expression Omnibus, respectively. Common differentially expressed genes (DEGs) were identified based on sequencing and expression as given in the profiling dataset. Pathway enrichment analysis of the DEGs was performed using the Database for Annotation, Visualization, and Integrated Discovery. The protein-protein interaction network was established using the string online database in order to identify hub genes. Univariate and multivariable Cox regression analyses were used to screen prognostic DEGs and to construct a prognostic signature. Survival analysis based on the prognostic signature was performed on TCGA EC dataset. A total of 255 common DEGs were found and 11 hub genes (TOP2A, CDK1, CCNB1, CCNB2, AURKA, PCNA, CCNA2, BIRC5, NDC80, CDC20, and BUB1BA) that may be closely related to the pathogenesis of EC were identified. A panel of 7 DEG signatures consisting of PHLDA2, GGH, ESPL1, FAM184A, KIAA1644, ESPL1, and TRPM4 were constructed. The signature performed well for prognosis prediction (p < 0.001) and time-dependent receiver-operating characteristic (ROC) analysis displayed an area under the curve (AUC) of 0.797, 0.734, 0.729, and 0.647 for 1, 3, 5, and 10-year overall survival (OS) prediction, respectively. Conclusion This study identified potential genes that may be involved in the pathophysiology of EC and constructed a novel gene expression signature for EC risk stratification and prognosis prediction.
Collapse
Affiliation(s)
- Li Liu
- Department of Obstetrics and Gynecology, Liuzhou Worker's Hospital, Fourth Affiliated Hospital of Guangxi Medical University, Liuzhou, China
| | - Jiajing Lin
- Department of Obstetrics and Gynecology, Liuzhou Worker's Hospital, Fourth Affiliated Hospital of Guangxi Medical University, Liuzhou, China
| | - Hongying He
- Department of Obstetrics and Gynecology, Liuzhou Worker's Hospital, Fourth Affiliated Hospital of Guangxi Medical University, Liuzhou, China
| |
Collapse
|
41
|
Xi X, Chu Y, Liu N, Wang Q, Yin Z, Lu Y, Chen Y. Joint bioinformatics analysis of underlying potential functions of hsa-let-7b-5p and core genes in human glioma. J Transl Med 2019; 17:129. [PMID: 30995921 PMCID: PMC6471881 DOI: 10.1186/s12967-019-1882-7] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2018] [Accepted: 04/11/2019] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Glioma accounts for a large proportion of cancer, and an effective treatment for this disease is still lacking because of the absence of specific driver molecules. Current challenges in the treatment of glioma are the accurate and timely diagnosis of brain glioma and targeted treatment plans. To investigate the diagnostic biomarkers and prospective role of miRNAs in the tumorigenesis and progression of glioma, we analyzed the expression of miRNAs and key genes in glioma based on The Cancer Genome Atlas database. METHODS Of the 701 cases that were downloaded, five were normal and 696 were glioma. Then, 1626 differentially expressed genes were identified, and 173 aberrantly expressed miRNAs were calculated by edgeR. GO and KEGG pathway enrichment analyses were performed using Cytoscape software. A coexpression network was built by weighted correlation network analysis (WGCNA). A cell scratch test and transwell, cell apoptosis and cell cycle assays were performed to validate the function of hsa-let-7b-5p. RESULTS Based on crosstalk genes in the KEGG, PPI network, and WGCNA analyses, PLK1, CCNA2, cyclin B2 (CCNB2), and AURKA were screened as candidate diagnostic marker genes. The survival analysis revealed that high mRNA expression of PLK1, CCNA2, and AURKA was significantly associated with poor overall survival. Furthermore, hsa-let-7b-5p was identified as a core miRNA in the regulation of candidate genes involved in glioma development. We confirmed that hsa-let-7b-5p could inhibit the migration, invasion, and cell cycle of glioma cells. CONCLUSIONS This study provides four potential biomarkers for the diagnosis of glioma, offers a potential explanation of its pathogenesis, and proposes hsa-let-7b-5p as a therapeutic target.
Collapse
Affiliation(s)
- Xiaonan Xi
- College of Pharmacy, Nankai University, Tianjin, 300350 People’s Republic of China
- State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin, 300350 People’s Republic of China
| | - Yahui Chu
- College of Pharmacy, Nankai University, Tianjin, 300350 People’s Republic of China
- State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin, 300350 People’s Republic of China
| | - Ning Liu
- State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin, 300350 People’s Republic of China
| | - Qianqian Wang
- State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin, 300350 People’s Republic of China
| | - Zheng Yin
- College of Pharmacy, Nankai University, Tianjin, 300350 People’s Republic of China
| | - Yaxin Lu
- College of Pharmacy, Nankai University, Tianjin, 300350 People’s Republic of China
- State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin, 300350 People’s Republic of China
| | - Yue Chen
- College of Pharmacy, Nankai University, Tianjin, 300350 People’s Republic of China
- State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin, 300350 People’s Republic of China
| |
Collapse
|
42
|
Cong Z, Diao Y, Xu Y, Li X, Jiang Z, Shao C, Ji S, Shen Y, De W, Qiang Y. Long non-coding RNA linc00665 promotes lung adenocarcinoma progression and functions as ceRNA to regulate AKR1B10-ERK signaling by sponging miR-98. Cell Death Dis 2019; 10:84. [PMID: 30692511 PMCID: PMC6349882 DOI: 10.1038/s41419-019-1361-3] [Citation(s) in RCA: 102] [Impact Index Per Article: 20.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2018] [Revised: 01/03/2019] [Accepted: 01/08/2019] [Indexed: 12/23/2022]
Abstract
Long non-coding RNAs (lncRNAs) are frequently dysregulated in multiple malignancies, demonstrating their potential oncogenic or tumor-suppressive roles in tumorigenesis. Herein, we reported the identification of a novel lncRNA, linc00665 (ENST00000590622), which was markedly upregulated in lung adenocarcinoma (LUAD) tissues and might serve as an independent predictor for poor prognosis. Functional assays indicated that linc00665 reinforced LUAD cell proliferation and metastasis in vitro and in vivo. Mechanistically, transcription factor SP1 induced the transcription of linc00665 in LUAD cells, which exerted its oncogenic role by functioning as competing endogenous RNA (ceRNA) for miR-98 and subsequently activating downstream AKR1B10-ERK signaling pathway. Together, our study elucidates oncogenic roles of linc00665-miR98-AKR1B10 axis in LUAD tumorigenesis, which may serve as potential diagnostic biomarkers and therapeutic targets.
Collapse
Affiliation(s)
- Zhuangzhuang Cong
- Department of Cardiothoracic Surgery, Jinling Hospital, Medical School of Nanjing University, 210000, Nanjing, China
| | - Yifei Diao
- Department of Cardiothoracic Surgery, Jinling Hospital, Southeast University, 210000, Nanjing, China
| | - Yang Xu
- Department of Cardiothoracic Surgery, Jinling Hospital, Medical School of Nanjing University, 210000, Nanjing, China
- Department of Cardiothoracic Surgery, Jinling Hospital, Nanjing Medical University, 210000, Nanjing, China
| | - Xiaokun Li
- Department of Cardiothoracic Surgery, Jinling Hospital, Southeast University, 210000, Nanjing, China
| | - Zhisheng Jiang
- Department of Cardiothoracic Surgery, Jinling Hospital, Bengbu Medical College, 233030, Anhui, China
| | - Chenye Shao
- Department of Cardiothoracic Surgery, Jinling Hospital, Nanjing Medical University, 210000, Nanjing, China
| | - Saiguang Ji
- Department of Cardiothoracic Surgery, Jinling Hospital, Bengbu Medical College, 233030, Anhui, China
| | - Yi Shen
- Department of Cardiothoracic Surgery, Jinling Hospital, Medical School of Nanjing University, 210000, Nanjing, China.
- Department of Cardiothoracic Surgery, Jinling Hospital, Southeast University, 210000, Nanjing, China.
- Department of Cardiothoracic Surgery, Jinling Hospital, Nanjing Medical University, 210000, Nanjing, China.
- Department of Cardiothoracic Surgery, Jinling Hospital, Bengbu Medical College, 233030, Anhui, China.
| | - Wei De
- Department of Biochemistry and Molecular Biology, Nanjing Medical University, 210000, Nanjing, China
| | - Yong Qiang
- Department of Cardiothoracic Surgery, Jinling Hospital, Medical School of Nanjing University, 210000, Nanjing, China.
| |
Collapse
|
43
|
Guo YN, Dong H, Ma FC, Huang JJ, Liang KZ, Peng JL, Chen G, Wei KL. The clinicopathological significance of decreased miR-125b-5p in hepatocellular carcinoma: evidence based on RT-qPCR, microRNA-microarray, and microRNA-sequencing. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2019; 12:21-39. [PMID: 31933718 PMCID: PMC6944034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 11/01/2018] [Accepted: 11/26/2018] [Indexed: 06/10/2023]
Abstract
The aim of the study was to comprehensively evaluate the clinical value of miR-125b-5p in hepatocellular carcinoma (HCC) and its potential molecular mechanisms. MiR-125b-5p expression was remarkably lower as examined by real-time reverse transcription quantitative polymerase chain reaction (RT-qPCR) in 95 paired HCC and nonmalignant liver tissues in house (P<0.001), which was in accord with the results from miRNA-sequencing data with 371 cases of HCC. miRNA-chips from Gene Expression Omnibus (GEO) and ArrayExpress were screened. Among the seven included miRNA-chips, the relative expression of miR-125b-5p expression levels showed decreasing trends in HCC tissue samples compared with non-cancerous liver tissue samples. Altogether, A total of 655 cases of HCC tissues and 334 non-HCC liver tissues were included in the final meta-analysis. We observed that the expression of miR-125b-5p indeed decreased markedly in HCC tissues compared with the non-HCC tissues (SMD: -1.414, 95% CI: -1.894 to -0.935, P<0.001). The area under the SROC curve of lower expression of miR-125b-5p was 0.91 (95% CI: 0.89 to 0.94). A Kaplan-Meier survival analysis indicated that the lower expression or the absence of miR-125b-5p may be a risk factor for the poor outcome of HCC patients. Furthermore, the potential target genes of miR-125b-5p from 11 miRNA target prediction databases were intersected with 1,486 differentially expressed genes (DEGs) as calculated by RNA-sequencing data. Finally, a total of 330 GEGs were collected and enriched in the pathways of lysosome, focal adhesion, and pathways in cancer. In conclusion, this study utilizes a variety of research methods to confirm the lower level of miR-125b-5p in HCC tissues. This lower expression level of miR-125b-5p is closely related to increased disease progression in HCC patients.
Collapse
Affiliation(s)
- Yi-Nan Guo
- Department of Pathology, First Affiliated Hospital of Guangxi Medical UniversityNanning 530021, Guangxi, Zhuang Autonomous Region, People’s Republic of China
| | - Hao Dong
- Department of Oncology, First Affiliated Hospital of Guangxi Medical UniversityNanning 530021, Guangxi, Zhuang Autonomous Region, People’s Republic of China
| | - Fu-Chao Ma
- Department of Oncology, First Affiliated Hospital of Guangxi Medical UniversityNanning 530021, Guangxi, Zhuang Autonomous Region, People’s Republic of China
| | - Jing-Jv Huang
- Department of Oncology, First Affiliated Hospital of Guangxi Medical UniversityNanning 530021, Guangxi, Zhuang Autonomous Region, People’s Republic of China
| | - Kai-Zhi Liang
- Department of Oncology, First Affiliated Hospital of Guangxi Medical UniversityNanning 530021, Guangxi, Zhuang Autonomous Region, People’s Republic of China
| | - Jia-Li Peng
- Department of Oncology, First Affiliated Hospital of Guangxi Medical UniversityNanning 530021, Guangxi, Zhuang Autonomous Region, People’s Republic of China
| | - Gang Chen
- Department of Pathology, First Affiliated Hospital of Guangxi Medical UniversityNanning 530021, Guangxi, Zhuang Autonomous Region, People’s Republic of China
| | - Kang-Lai Wei
- Department of Pathology, First Affiliated Hospital of Guangxi Medical UniversityNanning 530021, Guangxi, Zhuang Autonomous Region, People’s Republic of China
| |
Collapse
|
44
|
Li R, Jiang X, Zhang Y, Wang S, Chen X, Yu X, Ma J, Huang X. Cyclin B2 Overexpression in Human Hepatocellular Carcinoma is Associated with Poor Prognosis. Arch Med Res 2019; 50:10-17. [PMID: 31101236 DOI: 10.1016/j.arcmed.2019.03.003] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2018] [Revised: 02/25/2019] [Accepted: 03/11/2019] [Indexed: 12/14/2022]
Abstract
BACKGROUND AND AIMS Cyclin B2 (CCNB2) has been reported to be highly expressed in a few malignancies. However, the biological function of CCNB2 in hepatocellular carcinoma (HCC) is largely unknown. We aimed to investigate the effect of CCNB2 in HCC. METHODS The expression of CCNB2 in HCC and normal liver tissues and connection of its expression with prognosis and clinical parameters were studied. The effect of knocking down CCNB2 on cell proliferation, migration, cell cycle distribution, and apoptosis were estimated in BEL-7404 cells. RESULTS Compared to normal liver tissues, the level of CCNB2 was higher in HCC tissues from the Gene Expression Profiling Interactive Analysis (GEPIA). The 5 year overall survival and disease-free survival of HCC patients with high CCNB2 levels were shorter than that of those with low CCNB2 levels. Immunohistochemistry analysis also discovered the expression differences of CCNB2 in HCC and normal liver tissues and showed that CCNB2 expression was significantly associated with tumor number, tumor size, tumor thrombus, and alanine aminotransferase level. CCNB2 expression was higher in HCC cell lines (BEL-7404, Hep3B, BEL-7402, and SMMC-7721) than that in the normal hepatic cell line (HL-7702). Knockdown of CCNB2 inhibited cell proliferation and migration, promoted cell apoptosis, and caused S phase arrest in BEL-7404 cells. Finally, CCNB2 was associated with Polo Like Kinase 1 (PLK1) in the GEPIA database and BEL-7404 cells. CONCLUSIONS CCNB2 may serve as a prognostic factor and participated in the development and progression and promote cell proliferation and migration through CCNB2/PLK1 pathway in HCC.
Collapse
Affiliation(s)
- Rong Li
- Department of gastroenterology, Central South University Xiangya School of Medicine Affiliated Haikou Hospital, Haikou, China
| | - Xuemei Jiang
- Department of gastroenterology, Central South University Xiangya School of Medicine Affiliated Haikou Hospital, Haikou, China
| | - Yingai Zhang
- Center Laboratory, Central South University Xiangya School of Medicine Affiliated Haikou Hospital, Haikou, China
| | - Shunlan Wang
- Center Laboratory, Central South University Xiangya School of Medicine Affiliated Haikou Hospital, Haikou, China
| | - Xijie Chen
- Department of Gastroenterology, The First People's Hospital of Chenzhou, Chenzhou, China
| | - Xiangnan Yu
- Department of Gastroenterology, Zhongshan Hospital of Fudan University, Shanghai, China
| | - Jiamei Ma
- Department of gastroenterology, Central South University Xiangya School of Medicine Affiliated Haikou Hospital, Haikou, China
| | - Xiaoxi Huang
- Department of gastroenterology, Central South University Xiangya School of Medicine Affiliated Haikou Hospital, Haikou, China.
| |
Collapse
|