1
|
Liu X, Jiang D, Liu Y, Xie K, Zhao Y, Liu F. Crispr-Cas9-based long non-coding RNA interference and activation identified that the aberrant expression of Myc-regulated ST8SIA6 antisense RNA 1 promotes tumorigenesis and metastasis in hepatocellular carcinoma. Cytojournal 2024; 21:53. [PMID: 39737136 PMCID: PMC11683396 DOI: 10.25259/cytojournal_109_2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Accepted: 09/26/2024] [Indexed: 01/01/2025] Open
Abstract
Objective Long non-coding RNAs (lncRNAs) participate in the formation, progression, and metastasis of cancer. This study aimed to explore the roles of the lncRNA ST8SIA6 antisense RNA 1 (ST8SIA6-AS1) in tumorigenesis and elucidate the underlying molecular mechanism of its upregulation in hepatocellular carcinoma (HCC). Material and Methods A total of 56 in-house pairs of HCC tissues were examined, and ST8SIA6-AS1 levels were determined through real-time polymerase chain reaction (PCR). The biological behavior of ST8SIA6-AS1 by Crispr-Cas9-based gene repression and activation was determined in vitro and in vivo. The binding sites and biological behavior of Myc proto-oncogene and forkhead box A on chromatin were investigated through luciferase reporter assays, chromatin immunoprecipitation-quantitative PCR, and co-immunoprecipitation (co-IP) assays. The regulatory mechanisms of ST8SIA6-AS1 expression were analyzed with encyclopedia of DNA elements and gene expression profiling interactive analysis. Results The expression of ST8SIA6-AS1 significantly increased in multiple HCC cell lines and the 56 in-house pairs of HCC tissues (P = 0.0018). Functionally, high-efficiency Crispr-Cas9-based knockdown of ST8SIA6-AS1 revealed that ST8SIA6-AS1 knockdown attenuated the proliferation, migration, and infiltration of HCC cells and considerably reduced the growth rate of subcutaneous and orthotopic HCC tumors. Conversely, ST8SIA6-AS1 overexpression considerably improved the oncogenic characteristics of the HCC cells. Furthermore, ST8SIA6-AS1 upregulation was regulated by the direct binding of transcription factor Myc to the -260 bp to+155 bp and +1003 bp to +1312 bp regions of the ST8SIA6-AS1 transcription start site, which is a segment with high level of H3K27 acetylation. Myc knockdown or treatment with the BET bromodomain inhibitor JQ-1 considerably reduced ST8SIA6-AS1 RNA expression in the HCC cells. Conclusion Our study has established the oncogenic role of ST8SIA6-AS1 and elucidated the Myc-dependent upregulation mechanism of ST8SIA6-AS1 in HCC, providing a profound theoretical molecular basis for the carcinogenic function of ST8SIA6-AS1 in HCC.
Collapse
Affiliation(s)
- Xueqian Liu
- Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Dong Jiang
- Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Yang Liu
- Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Kun Xie
- Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Yijun Zhao
- Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Fubao Liu
- Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| |
Collapse
|
2
|
Qiu C, Fan H, Tao S, Deng Z, Luo H, Liu F. ST8SIA6-AS1, a novel lncRNA star in liver cancer. Front Cell Dev Biol 2024; 12:1435664. [PMID: 39211393 PMCID: PMC11358109 DOI: 10.3389/fcell.2024.1435664] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Accepted: 07/26/2024] [Indexed: 09/04/2024] Open
Abstract
Liver cancer is one of the most lethal gastrointestinal malignancies. Emerging evidence has underscored the pivotal role of long non-coding RNAs (lncRNAs) in tumorigenesis, with ST8SIA6-AS1 identified as a novel oncogenic lncRNA contributing to liver cancer progression. ST8SIA6-AS1 is consistently upregulated in hepatic cancer tissues and is strongly associated with unfavorable prognosis. Moreover, it demonstrates high diagnostic efficacy in detecting HCC. ST8SIA6-AS1 is involved in various cellular processes including proliferation, migration, and invasion, primarily through its function as a competing endogenous RNA (ceRNA), thereby facilitating hepatocarcinogenesis and disease advancement. This review provides a detailed examination of the molecular functions and regulatory mechanisms of ST8SIA6-AS1 in hepatocellular carcinoma (HCC) and highlights its potential as a promising biomarker for liver cancer, aiming to propel the development of innovative therapeutic strategies for HCC management.
Collapse
Affiliation(s)
- Cheng Qiu
- Department of General Surgery, Pingxiang People’s Hospital, Pingxiang, Jiangxi, China
| | - Haoran Fan
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Siyu Tao
- Second School of Clinical Medicine, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Ziqing Deng
- Department of General Surgery, Nanchang Third Hospital, Nanchang, Jiangxi, China
| | - Hongliang Luo
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Fangteng Liu
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| |
Collapse
|
3
|
Chen Y, Gao F, He Y, Liu M, Quan Y, Zhang P. DUB3 is a MAGEA3 deubiquitinase and a potential therapeutic target in hepatocellular carcinoma. iScience 2024; 27:109181. [PMID: 38414853 PMCID: PMC10897913 DOI: 10.1016/j.isci.2024.109181] [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: 08/21/2023] [Revised: 12/24/2023] [Accepted: 02/06/2024] [Indexed: 02/29/2024] Open
Abstract
Although melanoma-associated antigen A3 and A6 (MAGEA3/6)-specific tumor vaccines have shown antitumor effects in melanoma and non-small cell lung cancer (NSCLC), many cancers do not respond because MAGEA3 can promote cancer without triggering an immune response. Here, we identified DUB3 as the MAGEA3 deubiquitinase. DUB3 interacts with, deubiquitinates and stabilizes MAGEA3. Depletion of DUB3 in hepatocellular carcinoma (HCC) cells results in MAGEA3 degradation and P53-dependent growth inhibition. Moreover, DUB3 knockout attenuates HCC tumorigenesis in vivo, which can be rescued by restoration of MAGEA3. Intriguingly, pharmacological inhibition of DUB3 by palbociclib promotes degradation of MAGEA3 and inhibits tumor growth in preclinical models implanted with parental HCC cells but not with DUB3 knockout HCC cells. In patients with HCC, DUB3 is highly expressed, and its levels positively correlate with MAGEA3 levels. Taken together, DUB3 is a MAGEA3 deubiquitinase, and abrogating DUB3 enzymatic activity by palbociclib is a promising therapeutic strategy for HCC.
Collapse
Affiliation(s)
- Yuanhong Chen
- National Engineering Research Center for Nanomedicine, Key Laboratory of Molecular Biophysics of Ministry of Education, College of Life Science and Technology, Department of Oncology, Tongji Hospital, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Feng Gao
- National Engineering Research Center for Nanomedicine, Key Laboratory of Molecular Biophysics of Ministry of Education, College of Life Science and Technology, Department of Oncology, Tongji Hospital, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Yan He
- National Engineering Research Center for Nanomedicine, Key Laboratory of Molecular Biophysics of Ministry of Education, College of Life Science and Technology, Department of Oncology, Tongji Hospital, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Meijun Liu
- National Engineering Research Center for Nanomedicine, Key Laboratory of Molecular Biophysics of Ministry of Education, College of Life Science and Technology, Department of Oncology, Tongji Hospital, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Yuan Quan
- Stem Cell Laboratory, the Second Affiliated Hospital, Fujian Medical University, Quanzhou 362000, China
| | - Peijing Zhang
- National Engineering Research Center for Nanomedicine, Key Laboratory of Molecular Biophysics of Ministry of Education, College of Life Science and Technology, Department of Oncology, Tongji Hospital, Huazhong University of Science and Technology, Wuhan 430074, China
| |
Collapse
|
4
|
Xue J, Zhao H, Fu Y, Liu X, Wu X. Integrated analysis of multiple transcriptomic data identifies ST8SIA6‑AS1 and LINC01093 as potential biomarkers in HBV‑associated liver cancer. Oncol Lett 2023; 25:185. [PMID: 37065781 PMCID: PMC10091192 DOI: 10.3892/ol.2023.13771] [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: 07/26/2022] [Accepted: 12/02/2022] [Indexed: 04/18/2023] Open
Abstract
The mechanisms of long-non-coding RNAs (lncRNAs) in hepatitis B virus (HBV) infection-associated liver cancer remain largely unclear. Therefore, the aim of the present study was to investigate the regulatory mechanisms of lncRNAs in this disease. HBV-liver cancer related transcriptome expression profile data (GSE121248 and GSE55092) from the Gene Expression Omnibus database and survival prognosis information from The Cancer Genome Atlas (TCGA) database were obtained for analysis. The limma package was used to identify the overlapped differentially expressed RNAs (DERs), including DElncRNAs and DEmRNAs, in the GSE121248 and GSE55092 datasets. The screened optimized lncRNA signatures were used to develop a nomogram model based on the GSE121248 dataset, which was validated using the GSE55092 and TCGA datasets. A competitive endogenous RNA (ceRNA) network was constructed based on the screened prognosis-associated lncRNA signatures from TCGA dataset. In addition, the levels of specific lncRNAs were evaluated in HBV-infected human liver cancer tissues and cells, and Cell Counting Kit-8, ELISA and Transwell assays were performed to evaluate the effects of the lncRNAs in HBV-expressing liver cancer cells. A total of 535 overlapped DERs, including 30 DElncRNAs and 505 DEmRNAs, were identified in the GSE121248 and GSE55092 datasets. An optimized DElncRNA signature comprising 10 lncRNAs was used to establish a nomogram. ST8SIA6-AS1 and LINC01093 were identified as lncRNAs associated with HBV-liver cancer prognosis in TCGA dataset, and were applied to construct a ceRNA network. Reverse transcription-quantitative PCR analysis showed that ST8SIA6-AS1 was upregulated and LINC01093 was downregulated in HBV-infected human liver cancer tissues and HBV-expressing liver cancer cells compared with non-HBV-infected controls. ST8SIA6-AS1 knockdown and LINC01093 overexpression independently reduced the number of copies of HBV DNA, the levels of hepatitis B surface antigen and hepatitis B e antigen, as well as cell proliferation, migration and invasion. In summary, the present study identified ST8SIA6-AS1 and LINC01093 as two potential biomarkers that may be effective therapeutic targets for HBV-associated liver cancer.
Collapse
Affiliation(s)
- Jianhua Xue
- Department of Infectious Diseases, Hospital for Infectious Diseases of Pudong District, Shanghai 201318, P.R. China
| | - Hui Zhao
- Department of Infectious Diseases, Hospital for Infectious Diseases of Pudong District, Shanghai 201318, P.R. China
| | - Yifei Fu
- Department of Infectious Diseases, Hospital for Infectious Diseases of Pudong District, Shanghai 201318, P.R. China
| | - Xu Liu
- Department of Infectious Diseases, Hospital for Infectious Diseases of Pudong District, Shanghai 201318, P.R. China
| | - Xiangxiang Wu
- Department of Traditional Chinese Medicine, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200437, P.R. China
- Correspondence to: Dr Xiangxiang Wu, Department of Traditional Chinese Medicine, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, 110 Ganhe Road, Hongkou, Shanghai 200437, P.R. China, E-mail:
| |
Collapse
|
5
|
Qiao Y, Wang B, Yan Y, Niu L. Long noncoding RNA ST8SIA6-AS1 promotes cell proliferation and metastasis in triple-negative breast cancer by targeting miR-145-5p/CDCA3 to inactivate the p53/p21 signaling pathway. ENVIRONMENTAL TOXICOLOGY 2022; 37:2398-2411. [PMID: 35730485 DOI: 10.1002/tox.23605] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Revised: 05/08/2022] [Accepted: 06/09/2022] [Indexed: 06/15/2023]
Abstract
BACKGROUND Triple-negative breast cancer (TNBC), the most aggressive subtype of breast cancer, always exhibits a poor prognosis due to high risk of early recurrence and distant metastasis. Long noncoding RNAs (lncRNAs) have been reported as crucial regulators in breast cancer. However, the functions and action mechanisms of lncRNA ST8SIA6-AS1 in TNBC are largely unknown. METHODS Quantitative real-time PCR and western blot assays were used to measure the expression levels of different genes and proteins. Cell proliferation ability was monitored by CCK-8, colony forming and flow cytometry assays. Wound healing and transwell assays were performed to evaluate cell migration and invasion. The regulatory mechanisms of ST8SIA6-AS1 in TNBC were confirmed by dual luciferase reporter and RIP assays. A mouse xenograft model was established to investigate the role of ST8SIA6-AS1 in TNBC tumor growth. RESULTS ST8SIA6-AS1 displayed a higher expression in TNBC cells. Silencing ST8SIA6-AS1 impaired cell proliferation, cell cycle progression, migration, and invasion in vitro, and slowed tumor growth in vivo. Mechanistically, ST8SIA6-AS1 could facilitate the expression of its target CDCA3 (cell division cycle associated protein 3) and inactivate the p53/p21 signaling by inhibiting miR-145-5p. Moreover, miR-145-5p exerted a tumor-suppressive activity by targeting CDCA3. The tumor-suppressive effects induced by ST8SIA6-AS1 knockdown were abated by the down-regulation of miR-145-5p or the up-regulation of CDCA3. CONCLUSION ST8SIA6-AS1 exerts an oncogenic role in TNBC by interacting with miR-145-5p to up-regulate CDCA3 expression and inactivate the p53/p21 signaling, highlighting ST8SIA6-AS1 as a promising molecular target to combat TNBC.
Collapse
Affiliation(s)
- Yan Qiao
- Department of Breast Surgery, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Bin Wang
- Department of Breast Surgery, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Yu Yan
- Department of Breast Surgery, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Ligang Niu
- Department of Breast Surgery, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| |
Collapse
|
6
|
LncRNA ST8SIA6-AS1 facilitates hepatocellular carcinoma progression by governing miR-651-5p/TM4SF4 axis. Anticancer Drugs 2022; 33:741-751. [PMID: 35946523 DOI: 10.1097/cad.0000000000001326] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
The oncogenic role of ST8SIA6-AS1 in different cancers was reported, including hepatocellular carcinoma (HCC). However, the underlying mechanism has not been completely explored. Real time quantitative PCR analysis was conducted to assess the ST8SIA6-AS1, miR-651-5p and TM4SF4 expression in HCC tissues and cells. Cell counting kit-8 and wound-healing migration assays were adopted to evaluate the HCC cell proliferation and migration, respectively. The expression of apoptosis-related proteins (Bax and Bcl-2) in human colorectal cancer cells (HCC) was determined by western blotting. In addition to bioinformatics analysis, RNA immunoprecipitation studies and luciferase reporter assays were undertaken to investigate the direct target relationship among ST8SIA6-AS1 and miR-651-5p or TM4SF4. Highly expressed ST8SIA6-AS1 and TM4SF4 as well as poorly expressed miR-651-5p were detected in HCC tissues and cells. Clinically, miR-651-5p expression in HCC tissues is negatively correlated with ST8SIA6-AS1 or TM4SF4. Cell functional assays demonstrated that ST8SIA6-AS1 silencing resulted in weakened proliferative and migratory capacities in HCC cells in addition to increase Bax expression and reduced Bcl-2 expression. ST8SIA6-AS1 exhibited its oncogenic function by sponging tumor suppressor miR-651-5p, and the anti-oncogenic of miR-651-5p was offset by its TM4SF4. The manipulation of ST8SIA6-AS1/miR-651-5p/TM4SF4 axis-mediated oncogenicity in HCC might shed new light on HCC diagnosis and therapy.
Collapse
|
7
|
HOXA-AS3 Promotes Proliferation and Migration of Hepatocellular Carcinoma Cells via the miR-455-5p/PD-L1 Axis. J Immunol Res 2022; 2021:9289719. [PMID: 34988230 PMCID: PMC8723882 DOI: 10.1155/2021/9289719] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Accepted: 10/20/2021] [Indexed: 12/14/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is the most prevalent type of hepatic carcinoma. Long noncoding RNAs (lncRNAs) are considered crucial regulators of gene expression; however, their functions in HCC are not well understood. Thus, the present study is aimed at elucidating the functions of the lncRNA HOXA-AS3 in HCC. The functions of the HOXA-AS3/miR-455-5p/programmed death-ligand 1 (PD-L1) axis were investigated in vitro via qRT-PCR and dual-luciferase reporter assays. The effect of HOXA-AS3 expression on tumor growth and metastasis was assessed using a mouse xenograft model. High HOXA-AS3 expression was observed in the HCC cell lines. Furthermore, overexpression of HOXA-AS3 in HCC cells enhanced proliferation, migration, and invasion, regulated the cell cycle, and retarded apoptosis. We also identified an miR-455-5p binding site in HOXA-AS3. By sponging miR-455-5p, HOXA-AS3 increased the expression of PD-L1. Additionally, both the inhibition of PD-L1 and overexpression of miR-455-5p reversed the effects on cell proliferation and invasion triggered by the overexpression of HOXA-AS3. In conclusion, HOXA-AS3 modulated the functions of HCC cells through the miR-455-5p/PD-L1 axis. Therefore, HOXA-AS3 may be a novel therapeutic target for HCC.
Collapse
|
8
|
Yan G, Chang Z, Wang C, Gong Z, Xin H, Liu Z. LncRNA ILF3-AS1 promotes cell migration, invasion and EMT process in hepatocellular carcinoma via the miR-628-5p/MEIS2 axis to activate the Notch pathway. Dig Liver Dis 2022; 54:125-135. [PMID: 34053876 DOI: 10.1016/j.dld.2021.04.036] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Revised: 04/08/2021] [Accepted: 04/25/2021] [Indexed: 12/11/2022]
Abstract
BACKGROUND Long non-coding RNAs (lncRNAs) are essential indicators for hepatocellular carcinoma. LncRNAs can exert the same functions as their antisense mRNAs. ILF3 is an oncogene in hepatocellular carcinoma. ILF3 divergent transcript (ILF3-AS1) is the antisense RNA of ILF3, and has been reported as an oncogene in various cancers. AIMS To explore the role of lncRNA ILF3-AS1 in malignant phenotypes of hepatocellular carcinoma cells. METHODS AND RESULTS RT-qPCR analysis revealed that ILF3-AS1 was significantly upregulated in hepatocellular carcinoma cells. The hepatocellular carcinoma cell viability was suppressed by silenced ILF3-AS1. Transwell and wound healing assays showed that ILF3-AS1 downregulation inhibited cell invasion and migration. The levels of proteins associated with epithelial-mesenchymal transition (EMT) process and the Notch pathway were detected by western blot analysis. Luciferase reporter, RNA pull down and RIP assays were used to investigate the relationship between ILF3-AS1 and downstream target genes. ILF3-AS1 competed with meis homeobox 2 (MEIS2) for miR-628-5p in hepatocellular carcinoma cells. ILF3-AS1 elevated the levels of key proteins on the Notch pathway. Rescue assays demonstrated that MEIS2 reversed the antitumor effects of silenced ILF3-AS1 on hepatocellular carcinoma. In vivo assays demonstrated that ILF3-AS1 silencing inhibited the hepatocellular carcinoma tumor growth. CONCLUSIONS ILF3-AS1 promoted hepatocellular carcinoma progression via the Notch pathway and miR-628-5p/MEIS2 axis.
Collapse
Affiliation(s)
- Guangxin Yan
- Department of Radiology, Shengjing Hospital of China Medical University, No. 36, Sanhao Street, Heping District, Shenyang 110000, Liaoning, China
| | - Zhihui Chang
- Department of Radiology, Shengjing Hospital of China Medical University, No. 36, Sanhao Street, Heping District, Shenyang 110000, Liaoning, China
| | - Chuanzhuo Wang
- Department of Radiology, Shengjing Hospital of China Medical University, No. 36, Sanhao Street, Heping District, Shenyang 110000, Liaoning, China
| | - Zheng Gong
- Department of Radiology, Shengjing Hospital of China Medical University, No. 36, Sanhao Street, Heping District, Shenyang 110000, Liaoning, China
| | - He Xin
- Department of Radiology, Shengjing Hospital of China Medical University, No. 36, Sanhao Street, Heping District, Shenyang 110000, Liaoning, China
| | - Zhaoyu Liu
- Department of Radiology, Shengjing Hospital of China Medical University, No. 36, Sanhao Street, Heping District, Shenyang 110000, Liaoning, China.
| |
Collapse
|
9
|
Hugonnet M, Singh P, Haas Q, von Gunten S. The Distinct Roles of Sialyltransferases in Cancer Biology and Onco-Immunology. Front Immunol 2021; 12:799861. [PMID: 34975914 PMCID: PMC8718907 DOI: 10.3389/fimmu.2021.799861] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Accepted: 12/02/2021] [Indexed: 12/24/2022] Open
Abstract
Aberrant glycosylation is a key feature of malignant transformation. Hypersialylation, the enhanced expression of sialic acid-terminated glycoconjugates on the cell surface, has been linked to immune evasion and metastatic spread, eventually by interaction with sialoglycan-binding lectins, including Siglecs and selectins. The biosynthesis of tumor-associated sialoglycans involves sialyltransferases, which are differentially expressed in cancer cells. In this review article, we provide an overview of the twenty human sialyltransferases and their roles in cancer biology and immunity. A better understanding of the individual contribution of select sialyltransferases to the tumor sialome may lead to more personalized strategies for the treatment of cancer.
Collapse
Affiliation(s)
- Marjolaine Hugonnet
- Institute of Pharmacology, University of Bern, Bern, Switzerland
- Bern Center for Precision Medicine (BCPM), University of Bern, Bern, Switzerland
| | - Pushpita Singh
- Institute of Pharmacology, University of Bern, Bern, Switzerland
| | - Quentin Haas
- Institute of Pharmacology, University of Bern, Bern, Switzerland
| | - Stephan von Gunten
- Institute of Pharmacology, University of Bern, Bern, Switzerland
- Bern Center for Precision Medicine (BCPM), University of Bern, Bern, Switzerland
| |
Collapse
|
10
|
Zhang Y, Yang Y, Zhang Y, Liu Z. lncRNA ST8SIA6-AS1 facilitates proliferation and invasion in liver cancer by regulating miR-142-3p. Exp Ther Med 2021; 22:1348. [PMID: 34659494 PMCID: PMC8515546 DOI: 10.3892/etm.2021.10783] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Accepted: 06/09/2021] [Indexed: 12/25/2022] Open
Abstract
Long non-coding RNA ST8 α-N-acetyl-neuraminide α-2,8-sialyltransferase 6 antisense 1 (ST8SIA6-AS1) has been identified as a novel oncogene in breast cancer. However, its involvement in liver cancer has remained elusive. In the present study, the expression of ST8SIA6-AS1 and microRNA (miR)-142-3p in liver cancer tissues and cell lines was detected by reverse transcription-quantitative PCR. Tumor cell proliferation, migration and invasion assays were performed to determine the biological functions of ST8SIA6-AS1. The targeting interaction between ST8SIA6-AS1 and miR-142-3p predicted by bioinformatics was verified by a luciferase reporter assay and a biotin pulldown assay. The results indicated that ST8SIA6-AS1 was highly expressed in liver cancer tissues and cell lines, and the high expression of ST8SIA6-AS1 in liver cancer tissues was associated with poor prognosis. Knockdown of ST8SIA6-AS1 inhibited the proliferation, metastasis and invasion of liver cancer cells. Mechanistic investigation revealed that ST8SIA6-AS1 sequesters miR-142-3p and negatively regulates miR-142-3p expression in liver cancer cells. Further investigation indicated that the tumor-inhibitory effect of ST8SIA6-AS1 silencing was reversed by miR-142-3p depletion. In conclusion, ST8SIA6-AS1 was indicated to exert an oncogenic function in liver cancer by competitively sponging miR-142-3p.
Collapse
Affiliation(s)
- Yang Zhang
- Department of General Surgery, The General Hospital of The Central Military Theater of The People's Liberation Army, Wuhan, Hubei 430030, P.R. China
| | - Yan Yang
- Department of Pediatric Hematology, Maternal and Child Health Hospital of Hubei Province, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430031, P.R. China
| | - Yi Zhang
- Department of General Surgery, The General Hospital of The Central Military Theater of The People's Liberation Army, Wuhan, Hubei 430030, P.R. China
| | - Zhisu Liu
- Department of Hepatobiliary Surgery, Zhongnan Hospital of Wuhan University, Wuhan, Hubei 430071, P.R. China
| |
Collapse
|
11
|
The Role of Long Non-Coding RNA and microRNA Networks in Hepatocellular Carcinoma and Its Tumor Microenvironment. Int J Mol Sci 2021; 22:ijms221910630. [PMID: 34638971 PMCID: PMC8508708 DOI: 10.3390/ijms221910630] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Revised: 09/27/2021] [Accepted: 09/27/2021] [Indexed: 02/07/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is a common liver malignancy with high morbidity and poor prognosis. Long non-coding RNAs (lncRNAs) are involved in crucial biological processes of tumorigenesis and progression, and play four major regulatory roles, namely signal, decoy, guide, and scaffold, to regulate gene expression. Through these processes, lncRNAs can target microRNAs (miRNAs) to form lncRNA and miRNA networks, which regulate cancer cell proliferation, metastasis, drug resistance, and the tumor microenvironment. Here, we summarize the multifaceted functions of lncRNA and miRNA networks in the pathogenesis of HCC, the potential use of diagnostic or prognostic biomarkers, and novel therapeutic targets in HCC. This review also highlights the regulatory effects of lncRNA and miRNA networks in the tumor microenvironment of HCC.
Collapse
|
12
|
Lv C, Wan Q, Shen C, Wu H, Zhou B, Wang W. Long non‑coding RNA ZSCAN16‑AS1 promotes the malignant properties of hepatocellular carcinoma by decoying microRNA‑451a and consequently increasing ATF2 expression. Mol Med Rep 2021; 24:780. [PMID: 34498716 PMCID: PMC8436228 DOI: 10.3892/mmr.2021.12420] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2021] [Accepted: 07/26/2021] [Indexed: 11/05/2022] Open
Abstract
The importance of long noncoding RNAs (lncRNAs) in the oncogenicity of hepatocellular carcinoma (HCC) has been widely studied. However, the detailed functions of ZSCAN16 antisense RNA 1 (ZSCAN16‑AS1) have seldom been explored in HCC until the present study. In the present study, experiments were performed to clarify whether ZSCAN16‑AS1 is implicated in the oncogenesis and progression of HCC and to explore the possible underlying mechanisms. ZSCAN16‑AS1 expression was analyzed using reverse transcription‑quantitative PCR. The effects of ZSCAN16‑AS1 on the biological behavior of HCC cells were demonstrated by functional experiments. The direct binding capacity of ZSCAN16‑AS1 with microRNA‑451a (miR‑451a) was indicated by the luciferase reporter assay and RNA immunoprecipitation. The high expression of ZSCAN16‑AS1 was confirmed in HCC by The Cancer Genome Atlas database and the cohort of the present study. Survival data revealed that patients with a high ZSCAN16‑AS1 level had worse prognosis compared with those with a low ZSCAN16‑AS1 level. Following ZSCAN16‑AS1 knockdown, HCC cell proliferation, migration and invasion were curbed, whereas cell apoptosis was promoted in vitro. The absence of ZSCAN16‑AS1 restricted tumor growth of HCC cells in vivo. Mechanistically, ZSCAN16‑AS1 acted as a competing endogenous RNA by decoying miR‑451a in HCC cells. Furthermore, activating transcription factor 2 (ATF2), a direct target of miR‑451a, was under the regulation of ZSCAN16‑AS1, which was exerted by sequestering miR‑451a. In addition, miR‑451a knockdown or ATF2 resumption reversed the proliferation suppression, apoptosis promotion and migration and invasion inhibition triggered by ZSCAN16‑AS1 silencing. In conclusion, ZSCAN16‑AS1, a pro‑oncogenic lncRNA, aggravated the malignancy of HCC by controlling the miR‑451a/ATF2 axis. An understanding of the competing endogenous RNA network of ZSCAN16‑AS1/miR‑451a/ATF2 in HCC might be instrumental in the development of attractive targets for molecular therapy.
Collapse
Affiliation(s)
- Chaohua Lv
- Department of Hepatobiliary and Pancreatic Surgery, The People's Hospital of Tongliang, Chongqing 402575, P.R. China
| | - Qingsong Wan
- Department of Hepatobiliary and Pancreatic Surgery, The People's Hospital of Tongliang, Chongqing 402575, P.R. China
| | - Chengxiang Shen
- Department of Hepatobiliary and Pancreatic Surgery, The People's Hospital of Tongliang, Chongqing 402575, P.R. China
| | - Hongsheng Wu
- Department of Hepatobiliary and Pancreatic Surgery, The People's Hospital of Tongliang, Chongqing 402575, P.R. China
| | - Bin Zhou
- Department of Hepatobiliary and Pancreatic Surgery, The People's Hospital of Tongliang, Chongqing 402575, P.R. China
| | - Weiwei Wang
- Department of Hepatobiliary and Pancreatic Surgery, The People's Hospital of Tongliang, Chongqing 402575, P.R. China
| |
Collapse
|
13
|
Kuai J, Zheng L, Yi X, Liu Z, Qiu B, Lu Z, Jiang Y. ST8SIA6-AS1 promotes the development of hepatocellular carcinoma cells through miR-338-3p/NONO Axis. Dig Liver Dis 2021; 53:1192-1200. [PMID: 33722502 DOI: 10.1016/j.dld.2021.02.012] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Revised: 01/19/2021] [Accepted: 02/09/2021] [Indexed: 12/11/2022]
Abstract
BACKGROUND Increasing studies have shown a vital fact that long non-coding RNAs (lncRNAs) play a considerable regulatory role in hepatocellular carcinoma (HCC) progression. However, whether ST8 alpha-N-acetyl-neuraminide alpha-2, 8-sialyltransferase 6 antisense RNA 1 (ST8SIA6-AS1) affects the development of HCC is unclear. METHODS The target genes in HCC cell lines were quantified via utilzing quantitative real-time polymerase chain reaction (RT-qPCR) analysis and western blot. Effects of ST8SIA6-AS1 on proliferative, apoptosis and migratory ability of HCC cells were proved by a series of function experiments. The cellular distribution of ST8SIA6-AS1 was examined through fluorescent in situ hybridization (FISH) assay and subcellular fractionation experiments. RNA pulldown assay was implemented to explore the target of ST8SIA6-AS1. RNA Binding Protein Immunoprecipitation (RIP) and luciferase reporter assays were performed to identify the specific relationships between miR-338-3p and ST8SIA6-AS1/ non-POU domain containing octamer binding (NONO). RESULTS The expression of ST8SIA6-AS1 was apparently elevated in HCC cell. Silenced ST8SIA6-AS1 reduced proliferative, migratory and invasive ability of HCC cells. Moreover, ST8SIA6-AS1 targeted miR-338-3p to modulate the expression of NONO in HCC cells. CONCLUSIONS ST8SIA6-AS1 enhances the progression of HCC via miR-338-3p/NONO axis in vitro.
Collapse
Affiliation(s)
- Jinghua Kuai
- Department of Gastroenterology, Qilu Hospital of Shandong University, Qingdao 266000, Shandong, China
| | - Lijie Zheng
- Department of General Surgery, Qilu Hospital of Shandong University, No.758 Hefei Road, Shibei District, Qingdao 266000, Shandong, China.
| | - Xin Yi
- Department of General Surgery, Qilu Hospital of Shandong University, No.758 Hefei Road, Shibei District, Qingdao 266000, Shandong, China
| | - Zengli Liu
- Department of General Surgery, Qilu Hospital of Shandong University, No.758 Hefei Road, Shibei District, Qingdao 266000, Shandong, China
| | - Bo Qiu
- Department of General Surgery, Qilu Hospital of Shandong University, No.758 Hefei Road, Shibei District, Qingdao 266000, Shandong, China
| | - Zhihua Lu
- Department of General Surgery, Qilu Hospital of Shandong University, No.758 Hefei Road, Shibei District, Qingdao 266000, Shandong, China
| | - Yuanhui Jiang
- Department of General Surgery, Qilu Hospital of Shandong University, No.758 Hefei Road, Shibei District, Qingdao 266000, Shandong, China
| |
Collapse
|
14
|
Oncogenic activity and cellular functionality of melanoma associated antigen A3. Biochem Pharmacol 2021; 192:114700. [PMID: 34303709 DOI: 10.1016/j.bcp.2021.114700] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Revised: 07/16/2021] [Accepted: 07/20/2021] [Indexed: 12/27/2022]
Abstract
Cancer testis antigen Melanoma associated antigen A3 (MAGE-A3) has been subject of research for many years. Being expressed in various tumor types and influencing proliferation, metastasis, and tumor pathogenicity, MAGE-A3 is an attractive target for cancer therapy, particularly because in healthy tissues, MAGE-A3 is only expressed in testes and placenta. MAGE-A3 acts as a cellular master regulator by stimulating E3 ubiquitin ligase tripartite motif-containing protein 28 (TRIM28), resulting in regulation of various cellular targets. These include tumor suppressor protein p53 and cellular energy sensor AMP-activated protein kinase (AMPK). The restricted expression of MAGE-A3 in tumor cells makes MAGE-A3 an attractive target for vaccine-based immune therapy. However, although phase I and phase II clinical trials involving MAGE-A3-specific immunotherapeutic interventions were promising, large phase III studies failed. This article gives an overview about the role of MAGE-A3 as a cellular master switch and discusses approaches to improve MAGE-A3-based immunotherapies.
Collapse
|
15
|
Aberrant Sialylation in Cancer: Biomarker and Potential Target for Therapeutic Intervention? Cancers (Basel) 2021; 13:cancers13092014. [PMID: 33921986 PMCID: PMC8122436 DOI: 10.3390/cancers13092014] [Citation(s) in RCA: 60] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Revised: 04/19/2021] [Accepted: 04/20/2021] [Indexed: 12/14/2022] Open
Abstract
Simple Summary Sialylation is a post-translational modification that consists in the addition of sialic acid to growing glycan chains on glycoproteins and glycolipids. Aberrant sialylation is an established hallmark of several types of cancer, including breast, ovarian, pancreatic, prostate, colorectal and lung cancers, melanoma and hepatocellular carcinoma. Hypersialylation can be the effect of increased activity of sialyltransferases and results in an excess of negatively charged sialic acid on the surface of cancer cells. Sialic acid accumulation contributes to tumor progression by several paths, including stimulation of tumor invasion and migration, and enhancing immune evasion and tumor cell survival. In this review we explore the mechanisms by which sialyltransferases promote cancer progression. In addition, we provide insights into the possible use of sialyltransferases as biomarkers for cancer and summarize findings on the development of sialyltransferase inhibitors as potential anti-cancer treatments. Abstract Sialylation is an integral part of cellular function, governing many biological processes including cellular recognition, adhesion, molecular trafficking, signal transduction and endocytosis. Sialylation is controlled by the levels and the activities of sialyltransferases on glycoproteins and lipids. Altered gene expression of these enzymes in cancer yields to cancer-specific alterations of glycoprotein sialylation. Mounting evidence indicate that hypersialylation is closely associated with cancer progression and metastatic spread, and can be of prognostic significance in human cancer. Aberrant sialylation is not only a result of cancer, but also a driver of malignant phenotype, directly impacting key processes such as tumor cell dissociation and invasion, cell-cell and cell-matrix interactions, angiogenesis, resistance to apoptosis, and evasion of immune destruction. In this review we provide insights on the impact of sialylation in tumor progression, and outline the possible application of sialyltransferases as cancer biomarkers. We also summarize the most promising findings on the development of sialyltransferase inhibitors as potential anti-cancer treatments.
Collapse
|