1
|
Chen N, Li Y, Li X. Dynamic role of long noncoding RNA in liver diseases: pathogenesis and diagnostic aspects. Clin Exp Med 2025; 25:160. [PMID: 40369230 PMCID: PMC12078412 DOI: 10.1007/s10238-025-01678-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2025] [Accepted: 04/09/2025] [Indexed: 05/16/2025]
Abstract
Liver disease (LD) is complex pathological condition that has emerged as a major threat to human health and the quality of life. Nonetheless, the molecular mechanisms underlying the pathogenesis of LD have not yet been fully elucidated. Recently, a large amount of evidence has shown that long noncoding RNAs (lncRNAs) play important roles in diverse biological processes in the liver. The dysregulation of lncRNAs in the liver, for example, can affect tumor proliferation, migration, and invasion, contribute to hepatic metabolism disorder of lipid and glucose, and shape of hepatic tumoral microenvironment. Thus, a comprehensive understanding of the functional roles of lncRNAs in LD pathogenesis may provide new perspectives for the development of novel diagnostic and therapeutic tools. In the present review, we summarize the current findings on the relationship between lncRNAs and LD, including the modes of action of lncRNAs, the biological significance of lncRNAs in the pathogenesis of LD, especially in hepatocellular carcinoma (HCC) as well as in some non-neoplastic disorders, and the potential use of lncRNAs as diagnostic biomarkers and therapeutic targets for LD.
Collapse
Affiliation(s)
- Ningning Chen
- Department of Neonatology, Children's Hospital Affiliated to Shandong University (Jinan Children's Hospital), No. 23976, Jingshi Road, Jinan, 250022, Shandong, China
- School of Basic Medical Sciences, Shandong University, Jinan, China
| | - Yunxia Li
- Department of Neonatology, Children's Hospital Affiliated to Shandong University (Jinan Children's Hospital), No. 23976, Jingshi Road, Jinan, 250022, Shandong, China
| | - Xiaoying Li
- Department of Neonatology, Children's Hospital Affiliated to Shandong University (Jinan Children's Hospital), No. 23976, Jingshi Road, Jinan, 250022, Shandong, China.
| |
Collapse
|
2
|
Alimohammadi M, Fooladi AAI, Mafi A, Alavioun SM, Cho WC, Reiter RJ, Khormizi FZ, Yousefi T, Farahani N, Khoshnazar SM, Hushmandi K. Long noncoding RNAs and HPV-related cervical cancer: Uncovering molecular mechanisms and clinical applications. Transl Oncol 2025; 55:102363. [PMID: 40121995 PMCID: PMC11982485 DOI: 10.1016/j.tranon.2025.102363] [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: 10/30/2024] [Revised: 03/08/2025] [Accepted: 03/16/2025] [Indexed: 03/25/2025] Open
Abstract
Cervical cancer (CC) is the primary cause of cancer-related mortality among women in developing countries and is the most prevalent disease linked to human papillomavirus (HPV). Over 70 % of CC cases result from persistent infections with high-risk HPV types. The virus typically targets the mucocutaneous epithelium, generating viral particles in mature epithelial cells, which leads to disruptions in normal cell-cycle regulation and promotes uncontrolled cellular proliferation. This unchecked cell division results in the accumulation of genetic damage, contributing to the pathogenesis of CC. While HPV infection is a key etiological factor, the disease's progression also necessitates the involvement of genetic and epigenetic influences. One of the epigenetic regulators, long noncoding RNAs (lncRNAs), are characterized by transcripts exceeding 200 nucleotides. These molecules play crucial roles in various cellular processes, including transcription regulation, RNA metaboli35 per 100,000sm, and apoptosis. Investigating the specific roles of lncRNAs in modulating gene expression related to the oncogenic mechanisms of CC, particularly in the context of high-risk HPV infections, may provide valuable insights for diagnostic and therapeutic advancements. Herein, we first review key molecular mechanisms by which lncRNAs interfere with CC-related HPV development. Then, diagnostic, prognostic, and therapeutic potentials of these lncRNA molecules will be highlighted in depth. The focus of this article is on the role of lncRNAs associated with HPV-related CC, emphasizing the investigation of signaling pathways and their underlying molecular mechanisms. Furthermore, we explore the therapeutic potential and diagnostic relevance of the most significant lncRNAs in the context of CC, thereby highlighting their importance in advancing treatment strategies and improving patient outcomes.
Collapse
Affiliation(s)
- Mina Alimohammadi
- Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Abbas Ali Imani Fooladi
- Applied Microbiology Research Center, Biomedicine Technologies Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Alireza Mafi
- Nutrition and Food Security Research Center, Isfahan University of Medical Sciences, Isfahan, Iran; Department of Clinical Biochemistry, School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Seyedeh Mana Alavioun
- Department of Basic sciences, Faculty of Veterinary Medicine, Urmia university, Urmia, Iran
| | - William C Cho
- Department of Clinical Oncology, Queen Elizabeth Hospital, Kowloon, Hong Kong
| | - Russel J Reiter
- Department of Cell Systems and Anatomy, UT Health San Antonio, Long School of Medicine, San Antonio, TX, USA
| | | | - Tooba Yousefi
- Department of Clinical Biochemistry, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Najma Farahani
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
| | - Seyedeh Mahdieh Khoshnazar
- Gastroenterology and Hepatology Research Center, Institute of Basic and Clinical Physiology Sciences, Kerman University of Medical Sciences, Kerman, Iran.
| | - Kiavash Hushmandi
- Nephrology and Urology Research Center, Clinical Sciences Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
3
|
Cui Y. Analysis of Long Noncoding RNA in Fatty Acid Metabolism to Identify Prognostic Markers and Predict Immunotherapy Response in Low-Grade Glioma. World Neurosurg 2025; 196:123723. [PMID: 39952400 DOI: 10.1016/j.wneu.2025.123723] [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] [Received: 01/15/2025] [Accepted: 01/20/2025] [Indexed: 02/17/2025]
Abstract
BACKGROUND Low-grade gliomas (LGGs) are notorious for their difficult early-stage diagnosis, limited treatment options, and poor prognosis, making them a focal point in cancer research. Long noncoding RNAs (lncRNAs) have been identified as regulators of metabolic reprogramming in tumor cells, offering new directions for LGG treatment. METHODS This study employed data from The Cancer Genome Atlas, focusing on key fatty acid metabolism-related lncRNA. A risk scoring model was developed using univariate/multifactorial and least absolute shrinkage and selection operator Cox regression. Additionally, the study evaluated the role of these prognostic lncRNAs in LGG progression by assessing associations between LGG immune markers and tumor drug resistance. Finally, functional enrichment analysis highlighted the molecular roles of these lncRNAs. RESULTS In this study, a total of 14 prognostic lncRNAs were obtained. The risk model demonstrated excellent validity and reliability, making it a superior predictor of prognosis among patients with varying LGG risks. Among the identified lncRNAs, GHET-1 was notably associated with LGG sensitivity to current chemotherapy options and might be a crucial lncRNA affecting LGG progression. High-risk patients exhibited T-helper cell-mediated immunosuppression, potentially paving new paths for future LGG immunotherapy. CONCLUSIONS Focusing on lncRNA regulation and fatty acid metabolism reprogramming, this study established an innovative prognostic prediction model for LGGs, showing outstanding validity and reliability. The findings offer new molecular and cellular targets for the future development of LGG treatments.
Collapse
Affiliation(s)
- Yang Cui
- Department of Neurosurgery, Hebei Yanda Hospital, Langfang, He Bei, China.
| |
Collapse
|
4
|
Lu Z, Dong H, Tu Z, Liu H. Expression, molecular mechanisms and therapeutic potentials of ATF1 in cancers. Life Sci 2025; 360:123256. [PMID: 39580140 DOI: 10.1016/j.lfs.2024.123256] [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] [Received: 09/17/2024] [Revised: 11/06/2024] [Accepted: 11/14/2024] [Indexed: 11/25/2024]
Abstract
Activating transcription factor 1 (ATF1) is a crucial cellular regulator, with its misregulation implicated in numerous cancers. As a key player in the ATF/CREB family, ATF1 modulates gene expression in response to extracellular signals, significantly impacting cancer progression. This review examines ATF1's structural features, its role in tumorigenesis, and its potential therapeutic applications. Data from various databases consistently show ATF1 overexpression in diverse cancers, associated with poor prognosis and aggressive phenotypes. The review explores ATF1's complex regulatory mechanisms, influencing cell proliferation, apoptosis, migration, invasion, and therapeutic resistance, and its interactions with regulatory networks. Emerging strategies targeting ATF1, such as engineered antibodies, natural compounds, and small molecule inhibitors, show efficacy in preclinical models. ATF1 may also act as a biomarker for personalized therapeutic response and resistance. Future research should focus on ATF1's role in the tumor microenvironment and its interaction with the immune system, potentially leading to new immunotherapeutic strategies. A deeper understanding of ATF1 could enhance cancer treatment and patient outcomes.
Collapse
Affiliation(s)
- Ziwen Lu
- School of Life Sciences, Jiangsu University, Zhenjiang, Jiangsu 212013, China
| | - Hangyu Dong
- School of Pharmacy, Jiangsu University, Zhenjiang, Jiangsu 212013, China
| | - Zhigang Tu
- School of Life Sciences, Jiangsu University, Zhenjiang, Jiangsu 212013, China.
| | - Hanqing Liu
- School of Pharmacy, Jiangsu University, Zhenjiang, Jiangsu 212013, China.
| |
Collapse
|
5
|
Zhai X, Zhou J, Huang X, Weng J, Lin H, Sun S, Chi J, Meng L. LncRNA GHET1 from bone mesenchymal stem cell-derived exosomes improves doxorubicin-induced pyroptosis of cardiomyocytes by mediating NLRP3. Sci Rep 2024; 14:19078. [PMID: 39154102 PMCID: PMC11330485 DOI: 10.1038/s41598-024-70151-w] [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: 03/04/2023] [Accepted: 08/13/2024] [Indexed: 08/19/2024] Open
Abstract
Doxorubicin (DOX) is an important chemotherapeutic agent for the treatment of hematologic tumors and breast carcinoma. However, its clinical application is limited owing to severe cardiotoxicity. Pyroptosis is a form of programmed cell death linked to DOX-induced cardiotoxicity. Bone mesenchymal stem cell-derived exosomes (BMSC-Exos) and endothelial progenitor cells-derived exosomes (EPC-Exos) have a protective role in the myocardium. Here we found that BMSC-Exos could improve DOX-induced cardiotoxicity by inhibiting pyroptosis, but EPC-Exos couldn't. Compared with EPCs-Exo, BMSC-Exo-overexpressing lncRNA GHET1 more effectively suppressed pyroptosis, protecting against DOX-induced cardiotoxicity. Further studies showed that lncRNA GHET1 effectively decreased the expression of Nod-like receptor protein 3 (NLRP3), which plays a vital role in pyroptosis by binding to IGF2 mRNA-binding protein 1 (IGF2BP1), a non-catalytic posttranscriptional enhancer of NLRP3 mRNA. In summary, lncRNA GHET1 released by BMSC-Exo ameliorated DOX-induced pyroptosis by targeting IGF2BP1 to reduce posttranscriptional stabilization of NLRP3.
Collapse
Affiliation(s)
- Xiaoya Zhai
- Department of Cardiology, Shaoxing People's Hospital, Shaoxing, China
| | - Jiedong Zhou
- Department of Clinical Medicine, School of Medicine, Shaoxing University, Shaoxing, Zhejiang, China
| | - Xingxiao Huang
- Department of Cardiology, Shaoxing People's Hospital, Shaoxing, China
| | - Jingfan Weng
- Department of Cardiac Rehabilitation, Zhejiang Hospital, Hangzhou, China
| | - Hui Lin
- Department of Cardiology, Shaoxing People's Hospital, Shaoxing, China
| | - Shimin Sun
- Department of Cardiology, Shaoxing People's Hospital, Shaoxing, China
| | - Jufang Chi
- Department of Cardiology, Shaoxing People's Hospital, Shaoxing, China
| | - Liping Meng
- Department of Cardiology, Shaoxing People's Hospital, Shaoxing, China.
| |
Collapse
|
6
|
Liu Q, Zhou H, Wang Y, Gui J, Yang D, Sun J, Ge D, Wu S, Liu Q, Zhu L, Mi Y. H3K27 acetylation activated-PDLIM7 promotes castration-resistant prostate cancer progression by inducing O-Glycosylation of YAP1 protein. Transl Oncol 2024; 40:101830. [PMID: 38056280 PMCID: PMC10714362 DOI: 10.1016/j.tranon.2023.101830] [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: 07/19/2023] [Revised: 10/17/2023] [Accepted: 11/11/2023] [Indexed: 12/08/2023] Open
Abstract
Castration-resistant prostate cancer (CRPC) is a fatal disease that evolves from prostate cancer due to drug resistance after long-term androgen deprivation therapy. In this study, we aimed to find novel molecular targets for treating CRPC. Through peptidome, we screened out polypeptides dysregulated in the serum of CRPC patients. According to RT-qPCR analysis and cell viability detection, we chose PDZ and LIM Domain 7 (PDLIM7) as the research object. As demonstrated by loss-of-function assays, silencing of PDLIM7 could suppress CRPC cell proliferation, migration, and angiogenesis. Moreover, PDLIM7 knockdown enhanced the sensitivity of CRPC cells to docetaxel treatment. Subsequently, we found that CBP/p300 increases the H3K27ac level in the PDLIM7 promoter to activate PDLIM7. Mechanism experiments such as IP and western blot revealed that PDLIM7 interacted with YAP1 to induce O-Glycosylation of YAP1 and thus stabilize YAP1 protein. Rescue assays demonstrated that PDLIM7 promoted the malignant processes of CRPC cells through YAP1. Finally, an animal study validated that PDLIM7 aggravated tumor growth. In conclusion, our findings highlighted the oncogenic role of PDLIM7 upregulated by CBP/p300-induced H3K27ac enhancement in CRPC by stabilizing YAP1.
Collapse
Affiliation(s)
- Qing Liu
- Affiliated Hospital of Jiangnan University, No. 1000 Hefeng Road, Wuxi, Jiangsu 214122, China; Wuxi Medical College, Jiangnan University, Wuxi 214122, China; Department of Health and Wellness, Huadong Sanatorium, Wuxi, China
| | - Hangsheng Zhou
- Department of Urology, Affiliated Hospital of Jiangnan University, No. 1000 Hefeng Road, Wuxi, Jiangsu 214122, China; Wuxi Medical College, Jiangnan University, Wuxi 214122, China
| | - Yanjuan Wang
- Department of Nuclear Medicine, Affiliated Hospital of Jiangnan University, No. 1000 Hefeng Road, Wuxi, Jiangsu 214122, China
| | - Jiandong Gui
- Department of Urology, Affiliated Hospital of Jiangnan University, No. 1000 Hefeng Road, Wuxi, Jiangsu 214122, China; Wuxi Medical College, Jiangnan University, Wuxi 214122, China
| | - Dongjie Yang
- Department of Pathology, Affiliated Hospital of Jiangnan University, No. 1000 Hefeng Road, Wuxi, Jiangsu 214122, China
| | - Jian Sun
- Department of Urology, Affiliated Hospital of Jiangnan University, No. 1000 Hefeng Road, Wuxi, Jiangsu 214122, China; Wuxi Medical College, Jiangnan University, Wuxi 214122, China
| | - Dongsheng Ge
- Department of Urology, Affiliated Hospital of Jiangnan University, No. 1000 Hefeng Road, Wuxi, Jiangsu 214122, China; Wuxi Medical College, Jiangnan University, Wuxi 214122, China
| | - Sheng Wu
- Department of Urology, Affiliated Hospital of Jiangnan University, No. 1000 Hefeng Road, Wuxi, Jiangsu 214122, China; Wuxi Medical College, Jiangnan University, Wuxi 214122, China
| | - Qin Liu
- Department of Health and Wellness, Huadong Sanatorium, Wuxi, China
| | - Lijie Zhu
- Department of Urology, Affiliated Hospital of Jiangnan University, No. 1000 Hefeng Road, Wuxi, Jiangsu 214122, China; Wuxi Medical College, Jiangnan University, Wuxi 214122, China
| | - Yuanyuan Mi
- Department of Urology, Affiliated Hospital of Jiangnan University, No. 1000 Hefeng Road, Wuxi, Jiangsu 214122, China; Wuxi Medical College, Jiangnan University, Wuxi 214122, China.
| |
Collapse
|
7
|
Lin H, Hu S, Li Y, Li S, Teng D, Yang Y, Liu B, Du X. H3K27ac-activated LncRNA NUTM2A-AS1 Facilitated the Progression of Colorectal Cancer Cells via MicroRNA-126-5p/FAM3C Axis. Curr Cancer Drug Targets 2024; 24:1222-1234. [PMID: 38347779 DOI: 10.2174/0115680096277956240119065938] [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] [Received: 10/18/2023] [Revised: 01/04/2024] [Accepted: 01/10/2024] [Indexed: 09/25/2024]
Abstract
OBJECTIVE Long non-coding RNAs (lncRNAs) are of great importance in the process of colorectal cancer (CRC) tumorigenesis and progression. However, the functions and underlying molecular mechanisms of the majority of lncRNAs in CRC still lack clarity. METHODS A Quantitative real-time polymerase chain reaction (qRT-PCR) was employed to detect lncRNA NUTM2A-AS1 expression in CRC cell lines. Cell counting kit 8 (CCK-8) assay and flow cytometry were used to examine the biological functions of lncRNA NUTM2A-AS1 in the proliferation and apoptosis of CRC cells. RT-qPCR and western blot were implemented for the detection of cell proliferation-, apoptosis-related proteins, and FAM3C. Bioinformatics analysis and dual- luciferase reporter assays were utilized to identify the mutual regulatory mechanism of ceRNAs. RESULTS lncRNA NUTM2A-AS1 notably elevated in CRC cell lines and the silenced of NUTM2A- AS1 declined proliferation and facilitated apoptosis. Mechanistically, NUTM2A-AS1 was transcriptionally activated by histone H3 on lysine 27 acetylation (H3K27ac) enriched at its promoter region, and NUTM2A-AS1 acted as a sponge for miR-126-5p, leading to the upregulation of FAM3C expression in CRC cell lines. CONCLUSION Our research proposed NUTM2A-AS1 as an oncogenic lncRNA that facilitates CRC malignancy by upregulating FAM3C expression, which might provide new insight and a promising therapeutic target for the diagnosis and treatment of CRC.
Collapse
Affiliation(s)
- Haiguan Lin
- Department of General Surgery, the First Medical Centre, Chinese PLA General Hospital, Beijing, China
- Department of General Surgery, PLA Strategic Support Force Characteristic Medical Center, Beijing, China
| | - Shidong Hu
- Department of General Surgery, the First Medical Centre, Chinese PLA General Hospital, Beijing, China
| | - Yuxuan Li
- Department of General Surgery, the First Medical Centre, Chinese PLA General Hospital, Beijing, China
| | - Songyan Li
- Department of General Surgery, the First Medical Centre, Chinese PLA General Hospital, Beijing, China
| | - Da Teng
- Department of General Surgery, the First Medical Centre, Chinese PLA General Hospital, Beijing, China
| | - Yan Yang
- Department of General Surgery, the First Medical Centre, Chinese PLA General Hospital, Beijing, China
| | - Boyan Liu
- Department of General Surgery, the First Medical Centre, Chinese PLA General Hospital, Beijing, China
| | - Xiaohui Du
- Department of General Surgery, the First Medical Centre, Chinese PLA General Hospital, Beijing, China
| |
Collapse
|
8
|
Deng L, Bao W, Zhang B, Zhang S, Chen Z, Zhu X, He B, Wu L, Chen X, Deng T, Chen B, Yu Z, Wang Y, Chen G. AZGP1 activation by lenvatinib suppresses intrahepatic cholangiocarcinoma epithelial-mesenchymal transition through the TGF-β1/Smad3 pathway. Cell Death Dis 2023; 14:590. [PMID: 37669935 PMCID: PMC10480466 DOI: 10.1038/s41419-023-06092-5] [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: 08/25/2022] [Revised: 08/08/2023] [Accepted: 08/21/2023] [Indexed: 09/07/2023]
Abstract
Intrahepatic cholangiocarcinoma (ICC) is a primary liver malignancy and is characterized by highly aggressive and malignant biological behavior. Currently, effective treatment strategies are limited. The effect of lenvatinib on ICC is unknown. In this study, we found that AZGP1 was the key target of lenvatinib in ICC, and its low expression in ICC cancer tissues was associated with a poor prognosis in patients. Lenvatinib is a novel AZGP1 agonist candidate for ICC that inhibits ICC-EMT by regulating the TGF-β1/Smad3 signaling pathway in an AZGP1-dependent manner. Furthermore, we found that lenvatinib could increase AZGP1 expression by increasing the acetylation level of H3K27Ac in the promoter region of the AZGP1 gene, thereby inhibiting EMT in ICC cells. In conclusion, lenvatinib activates AZGP1 by increasing the acetylation level of H3K27Ac on the AZGP1 promoter region and regulates the TGF-β1/Smad3 signaling pathway in an AZGP1-dependent manner to inhibit ICC-EMT. This study offers new insight into the mechanism of lenvatinib in the treatment of ICC and provides a theoretical basis for new treatment methods.
Collapse
Affiliation(s)
- Liming Deng
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325035, China
- Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325035, China
- The Second Affiliated Hospital, Department of General Surgery, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Wenming Bao
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325035, China
- Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325035, China
| | - Baofu Zhang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325035, China
- Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325035, China
| | - Sina Zhang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325035, China
- Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325035, China
| | - Ziyan Chen
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325035, China
- Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325035, China
| | - Xuewen Zhu
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325035, China
- Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325035, China
| | - Bangjie He
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325035, China
- Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325035, China
| | - Lijun Wu
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325035, China
- Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325035, China
| | - Xiaohu Chen
- Department of Pathology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325035, China
| | - Tuo Deng
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325035, China
- Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325035, China
| | - Bo Chen
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325035, China
- Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325035, China
| | - Zhengping Yu
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325035, China
- Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325035, China
| | - Yi Wang
- Department of Epidemiology and Biostatistics, School of Public Health and Management, Wenzhou Medical University, Wenzhou, 325035, China.
| | - Gang Chen
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325035, China.
- Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325035, China.
- Hepatobiliary Pancreatic Tumor Bioengineering Cross International Joint Laboratory of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325035, China.
| |
Collapse
|
9
|
Ghafouri-Fard S, Ahmadi Teshnizi S, Hussen BM, Taheri M, Zali H. A review on the role of GHET1 in different cancers. Pathol Res Pract 2023; 247:154545. [PMID: 37244053 DOI: 10.1016/j.prp.2023.154545] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Revised: 05/13/2023] [Accepted: 05/16/2023] [Indexed: 05/29/2023]
Abstract
Gastric cancer High Expressed Transcript 1 (GHET1) is an RNA gene located on chromosome 7q36.1. This non-coding RNA is involved in the pathology of different cancers. It can regulate cell proliferation, apoptosis and cell cycle transition. Moreover, it induces epithelial-mesenchymal transition. Up-regulation of GHET1 has been correlated with poor prognosis of patients with different malignancies. Besides, its up-regulation has been mostly detected in later stages and advanced grades of cancers. This review summarizes recent studies on the expression of GHET1, its in vitro functions, and its impact on the beginning and progression of cancer based on xenograft models of cancer.
Collapse
Affiliation(s)
- Soudeh Ghafouri-Fard
- Department of Medical Genetics, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Sara Ahmadi Teshnizi
- Phytochemistry Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Bashdar Mahmud Hussen
- Department of Clinical Analysis, College of Pharmacy, Hawler Medical University, Kurdistan Region, Erbil, Iraq
| | - Mohammad Taheri
- Institute of Human Genetics, Jena University Hospital, Jena, Germany; Urology and Nephrology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Hakimeh Zali
- Proteomics Research Center, Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
10
|
Konani M, Pourhoseini M, Hashemi M, Entezari M, Khaleghi S. LncRNA GHET1 and LncRNA ZXF2 as New Biomarkers in Oral Squamous Cell Carcinoma in Relation to Clinicopathological Variables. INTERNATIONAL JOURNAL OF CANCER MANAGEMENT 2023. [DOI: 10.5812/ijcm-121372] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/09/2023]
Abstract
Background: The utilization of molecular techniques in detecting cancer has resulted in an improved prediction of outcomes. Oral squamous cell carcinoma (OSCC) is a prevalent illness that is frequently detected in its late stages. Therefore, finding molecular biomarkers that lead to the early detection of OSCC is of great importance. Objectives: This study aimed at evaluating the expression levels of long non-coding RNA (LncRNA) gastric carcinoma highly expressed transcript 1 (GHET1) and lncRNA ZXF2 in OSCC patients and their relationship with clinical pathology variables due to biomarker discovery and early diagnosis of OSCC. Methods: Tissue sampling was performed after selecting 30 OSCC patients and 30 healthy individuals from Emam-Khomeini Hospital, Tehran, Iran. Then, RNA extraction and cDNA synthesis were performed from these samples, using the relevant kits and their quantity and quality were measured, using nanodrop and agarose gel electrophoresis, respectively. For molecular biomarker identification and validation, real-time PCR (RT-PCR) was utilized to assess the expression of lncRNA GHET1 and lncRNA ZXF2. Data analysis was done, using GraphPad prism V.8 software. Results: The results showed that the expressions of both lncRNA GHET1 and lncRNA ZXF2 in OSCC tumor tissue increased compared to normal tissue (P < 0.0001). Receiver operating characteristic (ROC) analysis indicated that lncRNA GHET1 and lncRNA ZXF2 have the capability to be employed as biomarkers for OSCC detection. However, no significant relationship was observed between lncRNA GHET1 and lncRNA ZXF2 expressions with clinicopathological variables such as tumor stage and grade as well as patients' age. Conclusions: LncRNA GHET1 and lncRNA ZXF2 have the potential to be used as biomarkers in the early detection of OSCC and evaluating their expression in clinical settings are recommended. The use of these biomarkers in the early detection of OSCC can prevent the high mortality rate of OSCC patients. In the current study, the important role of the studied lncRNAs in OSCC diagnosis was shown. However, further studies are needed to confirm this.
Collapse
|
11
|
Fu Y, Si A, Wei X, Lin X, Ma Y, Qiu H, Guo Z, Pan Y, Zhang Y, Kong X, Li S, Shi Y, Wu H. Combining a machine-learning derived 4-lncRNA signature with AFP and TNM stages in predicting early recurrence of hepatocellular carcinoma. BMC Genomics 2023; 24:89. [PMID: 36849926 PMCID: PMC9972730 DOI: 10.1186/s12864-023-09194-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Accepted: 02/17/2023] [Indexed: 03/01/2023] Open
Abstract
BACKGROUND Near 70% of hepatocellular carcinoma (HCC) recurrence is early recurrence within 2-year post surgery. Long non-coding RNAs (lncRNAs) are intensively involved in HCC progression and serve as biomarkers for HCC prognosis. The aim of this study is to construct a lncRNA-based signature for predicting HCC early recurrence. METHODS Data of RNA expression and associated clinical information were accessed from The Cancer Genome Atlas Liver Hepatocellular Carcinoma (TCGA-LIHC) database. Recurrence associated differentially expressed lncRNAs (DELncs) were determined by three DEG methods and two survival analyses methods. DELncs involved in the signature were selected by three machine learning methods and multivariate Cox analysis. Additionally, the signature was validated in a cohort of HCC patients from an external source. In order to gain insight into the biological functions of this signature, gene sets enrichment analyses, immune infiltration analyses, as well as immune and drug therapy prediction analyses were conducted. RESULTS A 4-lncRNA signature consisting of AC108463.1, AF131217.1, CMB9-22P13.1, TMCC1-AS1 was constructed. Patients in the high-risk group showed significantly higher early recurrence rate compared to those in the low-risk group. Combination of the signature, AFP and TNM further improved the early HCC recurrence predictive performance. Several molecular pathways and gene sets associated with HCC pathogenesis are enriched in the high-risk group. Antitumor immune cells, such as activated B cell, type 1 T helper cell, natural killer cell and effective memory CD8 T cell are enriched in patients with low-risk HCCs. HCC patients in the low- and high-risk group had differential sensitivities to various antitumor drugs. Finally, predictive performance of this signature was validated in an external cohort of patients with HCC. CONCLUSION Combined with TNM and AFP, the 4-lncRNA signature presents excellent predictability of HCC early recurrence.
Collapse
Affiliation(s)
- Yi Fu
- grid.507037.60000 0004 1764 1277Shanghai Key Laboratory of Molecular Imaging, Zhoupu Hospital, Shanghai University of Medicine and Health Sciences, Shanghai, China ,grid.507037.60000 0004 1764 1277Collaborative Innovation Center for Biomedicines, Shanghai University of Medicine and Health Sciences, Shanghai, China ,grid.507037.60000 0004 1764 1277School of Medical Instruments, Shanghai University of Medicine and Health Sciences, Shanghai, China
| | - Anfeng Si
- grid.41156.370000 0001 2314 964XDepartment of Surgical Oncology, Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Xindong Wei
- grid.412585.f0000 0004 0604 8558Central Laboratory, Department of Liver Diseases, Shuguang Hospital, Shanghai University of Chinese Traditional Medicine, Shanghai, China
| | - Xinjie Lin
- grid.507037.60000 0004 1764 1277Shanghai Key Laboratory of Molecular Imaging, Zhoupu Hospital, Shanghai University of Medicine and Health Sciences, Shanghai, China ,grid.507037.60000 0004 1764 1277Collaborative Innovation Center for Biomedicines, Shanghai University of Medicine and Health Sciences, Shanghai, China
| | - Yujie Ma
- grid.507037.60000 0004 1764 1277Shanghai Key Laboratory of Molecular Imaging, Zhoupu Hospital, Shanghai University of Medicine and Health Sciences, Shanghai, China ,grid.507037.60000 0004 1764 1277Collaborative Innovation Center for Biomedicines, Shanghai University of Medicine and Health Sciences, Shanghai, China
| | - Huimin Qiu
- grid.507037.60000 0004 1764 1277Collaborative Innovation Center for Biomedicines, Shanghai University of Medicine and Health Sciences, Shanghai, China ,grid.267139.80000 0000 9188 055XSchool of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai, China
| | - Zhinan Guo
- grid.507037.60000 0004 1764 1277Collaborative Innovation Center for Biomedicines, Shanghai University of Medicine and Health Sciences, Shanghai, China ,grid.412543.50000 0001 0033 4148School of Kinesiology, Shanghai University of Sport, Shanghai, China
| | - Yong Pan
- grid.268099.c0000 0001 0348 3990Department of Infectious Disease, Zhoushan Hospital, Wenzhou Medical University, Zhoushan, China
| | - Yiru Zhang
- grid.268099.c0000 0001 0348 3990Department of Infectious Disease, Zhoushan Hospital, Wenzhou Medical University, Zhoushan, China
| | - Xiaoni Kong
- grid.412585.f0000 0004 0604 8558Central Laboratory, Department of Liver Diseases, Shuguang Hospital, Shanghai University of Chinese Traditional Medicine, Shanghai, China
| | - Shibo Li
- Department of Infectious Disease, Zhoushan Hospital, Wenzhou Medical University, Zhoushan, China.
| | - Yanjun Shi
- Abdominal Transplantation Center, General Surgery, School of Medicine, Ruijin Hospital, Shanghai Jiao Tong University, Shanghai, China.
| | - Hailong Wu
- Shanghai Key Laboratory of Molecular Imaging, Zhoupu Hospital, Shanghai University of Medicine and Health Sciences, Shanghai, China. .,Collaborative Innovation Center for Biomedicines, Shanghai University of Medicine and Health Sciences, Shanghai, China. .,School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai, China. .,School of Kinesiology, Shanghai University of Sport, Shanghai, China.
| |
Collapse
|
12
|
Shen H, Gu X, Li H, Tang M, Li X, Zhang Y, Su F, Wang Z. Exploring Prognosis, Tumor Microenvironment and Tumor Immune Infiltration in Hepatocellular Carcinoma Based on ATF/CREB Transcription Factor Family Gene-Related Model. J Hepatocell Carcinoma 2023; 10:327-345. [PMID: 36874250 PMCID: PMC9983578 DOI: 10.2147/jhc.s398713] [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: 12/02/2022] [Accepted: 02/20/2023] [Indexed: 03/02/2023] Open
Abstract
Introduction Hepatocellular carcinoma (HCC) is the most common form of primary liver cancer. It is the fourth leading cause of cancer-related death worldwide. Deregulation of the ATF/CREB family is associated with the progression of metabolic homeostasis and cancer. Because the liver plays a central role in metabolic homeostasis, it is critical to assess the predictive value of the ATF/CREB family in the diagnosis and prognosis of HCC. Methods Using data from The Cancer Genome Atlas (TCGA), this research evaluated the expression, copy number variations, and frequency of somatic mutations of 21 genes in the ATF/CREB family in HCC. A prognostic model based on the ATF/CREB gene family was developed via Lasso and Cox regression analyses, with the TCGA cohort serving as the training dataset and the International Cancer Genome Consortium (ICGC) cohort serving as the validation set. Kaplan-Meier and receiver operating characteristic analyses verified the accuracy of the prognostic model. Furthermore, the association among the prognostic model, immune checkpoints, and immune cells was examined. Results High-risk patients exhibited an unfavorable outcome as opposed to those in the low-risk category. Multivariate Cox analysis revealed that the risk score calculated based on the prognostic model was an independent prognostic factor for HCC. Analysis of immune mechanisms revealed that the risk score had a positive link to the expression of immune checkpoints, particularly CD274, PDCD1, LAG3, and CTLA4. Differences in immune cells and immune-associated roles were found between the high- and low-risk patients, as determined by single-sample gene set enrichment analysis. The core genes ATF1, CREB1, and CREB3 in the prognostic model were shown to be upregulated in HCC tissues as opposed to adjoining normal tissues, and the 10-year overall survival (OS) rate was worse among patients with elevated expression levels of ATF1, CREB1, and CREB3. Elevated expression levels of ATF1, CREB1, and CREB3 in HCC tissues were confirmed by qRT-PCR and immunohistochemistry studies. Conclusion According to the results of our training set and test set, the risk model based on the six ATF/CREB gene signatures predicting prognosis has certain predictive accuracy in predicting the survival of HCC patients. This study provides novel insights into the individualized treatment of patients with HCC.
Collapse
Affiliation(s)
- Honghong Shen
- Department of Medical Oncology, First Affiliated Hospital of Bengbu Medical College, Bengbu, People's Republic of China
| | - Xianhua Gu
- Department of Gynecology Oncology, First Affiliated Hospital of Bengbu Medical College, Bengbu, People's Republic of China
| | - Huiyuan Li
- Department of Medical Oncology, First Affiliated Hospital of Bengbu Medical College, Bengbu, People's Republic of China
| | - Mingyue Tang
- Department of Medical Oncology, First Affiliated Hospital of Bengbu Medical College, Bengbu, People's Republic of China
| | - Xinwei Li
- Department of Medical Oncology, First Affiliated Hospital of Bengbu Medical College, Bengbu, People's Republic of China
| | - Yue Zhang
- Department of Medical Oncology, First Affiliated Hospital of Bengbu Medical College, Bengbu, People's Republic of China
| | - Fang Su
- Department of Medical Oncology, First Affiliated Hospital of Bengbu Medical College, Bengbu, People's Republic of China
| | - Zishu Wang
- Department of Medical Oncology, First Affiliated Hospital of Bengbu Medical College, Bengbu, People's Republic of China
| |
Collapse
|
13
|
Xia JK, Qin XQ, Zhang L, Liu SJ, Shi XL, Ren HZ. Roles and regulation of histone acetylation in hepatocellular carcinoma. Front Genet 2022; 13:982222. [PMID: 36092874 PMCID: PMC9452893 DOI: 10.3389/fgene.2022.982222] [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: 06/30/2022] [Accepted: 08/03/2022] [Indexed: 11/13/2022] Open
Abstract
Hepatocellular Carcinoma (HCC) is the most frequent malignant tumor of the liver, but its prognosis is poor. Histone acetylation is an important epigenetic regulatory mode that modulates chromatin structure and transcriptional status to control gene expression in eukaryotic cells. Generally, histone acetylation and deacetylation processes are controlled by the opposing activities of histone acetyltransferases (HATs) and histone deacetylases (HDACs). Dysregulation of histone modification is reported to drive aberrant transcriptional programmes that facilitate liver cancer onset and progression. Emerging studies have demonstrated that several HDAC inhibitors exert tumor-suppressive properties via activation of various cell death molecular pathways in HCC. However, the complexity involved in the epigenetic transcription modifications and non-epigenetic cellular signaling processes limit their potential clinical applications. This review brings an in-depth view of the oncogenic mechanisms reported to be related to aberrant HCC-associated histone acetylation, which might provide new insights into the effective therapeutic strategies to prevent and treat HCC.
Collapse
Affiliation(s)
- Jin-kun Xia
- Department of Hepatobiliary Surgery, Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China
- Hepatobiliary Institute Nanjing University, Nanjing, China
| | - Xue-qian Qin
- Department of Hepatobiliary Surgery, Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China
| | - Lu Zhang
- Department of Hepatobiliary Surgery, Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China
| | - Shu-jun Liu
- Department of Hepatobiliary Surgery, Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China
| | - Xiao-lei Shi
- Department of Hepatobiliary Surgery, Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China
- Hepatobiliary Institute Nanjing University, Nanjing, China
| | - Hao-zhen Ren
- Department of Hepatobiliary Surgery, Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China
- Hepatobiliary Institute Nanjing University, Nanjing, China
| |
Collapse
|
14
|
Chen M, Liu Y, Yang Y, Qiu Y, Wang Z, Li X, Zhang W. Emerging roles of activating transcription factor (ATF) family members in tumourigenesis and immunity: Implications in cancer immunotherapy. Genes Dis 2022; 9:981-999. [PMID: 35685455 PMCID: PMC9170601 DOI: 10.1016/j.gendis.2021.04.008] [Citation(s) in RCA: 52] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2020] [Revised: 04/20/2021] [Accepted: 04/26/2021] [Indexed: 12/12/2022] Open
Abstract
Activating transcription factors, ATFs, are a group of bZIP transcription factors that act as homodimers or heterodimers with a range of other bZIP factors. In general, ATFs respond to extracellular signals, indicating their important roles in maintaining homeostasis. The ATF family includes ATF1, ATF2, ATF3, ATF4, ATF5, ATF6, and ATF7. Consistent with the diversity of cellular processes reported to be regulated by ATFs, the functions of ATFs are also diverse. ATFs play an important role in cell proliferation, apoptosis, differentiation and inflammation-related pathological processes. The expression and phosphorylation status of ATFs are also related to neurodegenerative diseases and polycystic kidney disease. Various miRNAs target ATFs to regulate cancer proliferation, apoptosis, autophagy, sensitivity and resistance to radiotherapy and chemotherapy. Moreover, ATFs are necessary to maintain cell redox homeostasis. Therefore, deepening our understanding of the regulation and function of ATFs will provide insights into the basic regulatory mechanisms that influence how cells integrate extracellular and intracellular signals into genomic responses through transcription factors. Under pathological conditions, especially in cancer biology and response to treatment, the characterization of ATF dysfunction is important for understanding how to therapeutically utilize ATF2 or other pathways controlled by transcription factors. In this review, we will demonstrate how ATF1, ATF2, ATF3, ATF4, ATF5, ATF6, and ATF7 function in promoting or suppressing cancer development and identify their roles in tumour immunotherapy.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Wenling Zhang
- Corresponding author. Department of Medical Laboratory Science, the Third Xiangya Hospital, Central South University, Tongzipo Road 172, Yuelu District, Changsha, Hunan 410013, PR China.
| |
Collapse
|
15
|
Kong F, Ma L, Wang X, You H, Zheng K, Tang R. Regulation of epithelial-mesenchymal transition by protein lysine acetylation. Cell Commun Signal 2022; 20:57. [PMID: 35484625 PMCID: PMC9052664 DOI: 10.1186/s12964-022-00870-y] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Accepted: 03/20/2022] [Indexed: 01/01/2023] Open
Abstract
The epithelial-mesenchymal transition (EMT) is a vital driver of tumor progression. It is a well-known and complex trans-differentiation process in which epithelial cells undergo morphogenetic changes with loss of apical-basal polarity, but acquire spindle-shaped mesenchymal phenotypes. Lysine acetylation is a type of protein modification that favors reversibly altering the structure and function of target molecules via the modulation of lysine acetyltransferases (KATs), as well as lysine deacetylases (KDACs). To date, research has found that histones and non-histone proteins can be acetylated to facilitate EMT. Interestingly, histone acetylation is a type of epigenetic regulation that is capable of modulating the acetylation levels of distinct histones at the promoters of EMT-related markers, EMT-inducing transcription factors (EMT-TFs), and EMT-related long non-coding RNAs to control EMT. However, non-histone acetylation is a post-translational modification, and its effect on EMT mainly relies on modulating the acetylation of EMT marker proteins, EMT-TFs, and EMT-related signal transduction molecules. In addition, several inhibitors against KATs and KDACs have been developed, some of which can suppress the development of different cancers by targeting EMT. In this review, we discuss the complex biological roles and molecular mechanisms underlying histone acetylation and non-histone protein acetylation in the control of EMT, highlighting lysine acetylation as potential strategy for the treatment of cancer through the regulation of EMT. Video Abstract
Collapse
Affiliation(s)
- Fanyun Kong
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogenic Biology and Immunology, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Lihong Ma
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogenic Biology and Immunology, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Xing Wang
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogenic Biology and Immunology, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Hongjuan You
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogenic Biology and Immunology, Xuzhou Medical University, Xuzhou, Jiangsu, China.
| | - Kuiyang Zheng
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogenic Biology and Immunology, Xuzhou Medical University, Xuzhou, Jiangsu, China.,National Demonstration Center for Experimental Basic Medical Sciences Education, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Renxian Tang
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogenic Biology and Immunology, Xuzhou Medical University, Xuzhou, Jiangsu, China. .,National Demonstration Center for Experimental Basic Medical Sciences Education, Xuzhou Medical University, Xuzhou, Jiangsu, China.
| |
Collapse
|
16
|
Tang K, Lv D, Miao L, Mao Y, Yu X. LncRNA TUG1 functions as a ceRNA for miR-1-3p to promote cell proliferation in hepatic carcinogenesis. J Clin Lab Anal 2022; 36:e24415. [PMID: 35421276 PMCID: PMC9102766 DOI: 10.1002/jcla.24415] [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: 11/05/2021] [Revised: 03/27/2022] [Accepted: 03/28/2022] [Indexed: 12/19/2022] Open
Abstract
Background Hepatocellular carcinoma (HCC) is characterised by high malignancy, metastasis and recurrence, but the specific mechanism that drives these outcomes is unclear. Recent studies have shown that long noncoding RNAs (lncRNAs) can regulate the proliferation and apoptosis of hepatic cells. Methods We searched for lncRNAs and microRNAs (miRNAs), which can regulate IGF1 expression, through a bioinformatics website, and predicted that lncRNA taurine‐upregulated gene 1 (TUG1) would have multiple targets for miR‐1‐3p binding, meaning that lncRNA TUG1 played an adsorption role. A double luciferase assay was used to verify the targeting relationship between lncRNA TUG1 and miR‐1‐3p. Western blotting and qPCR were used to verify the targeting relationship between miR‐1‐3p and IGF1, and qPCR was used to verify the regulatory relationship between the lncRNA TUG1‐miR‐1‐3p‐IGF1 axis. CCK‐8 was used to detect the growth activity of miRNA‐transfected L‐O2 cells, and flow cytometry was used to detect cell cycle changes and apoptosis. Result The proliferation cycle of L‐O2 cells transfected with miR‐1‐3p mimics was significantly slowed. Flow cytometry showed that the proliferation of L‐O2 cells was slowed, and the apoptosis rate was increased. In contrast, when L‐O2 cells were transfected with miR‐1‐3p inhibitor, the expression of IGF1 was significantly upregulated, and the cell proliferation cycle was significantly accelerated. Flow cytometry showed that the cell proliferation rate was accelerated, and the apoptosis rate was reduced. Conclusion LncRNA TUG1 can adsorb miR‐1‐3p as a competitive endogenous RNA (ceRNA) to promote the expression of IGF1 and promote cell proliferation in hepatic carcinogenesis.
Collapse
Affiliation(s)
- Keke Tang
- The Affiliated Hospital of the Medical School of Ningbo University, Ningbo, China
| | - Di Lv
- The Affiliated Hospital of the Medical School of Ningbo University, Ningbo, China
| | - Lingling Miao
- The Affiliated Hospital of the Medical School of Ningbo University, Ningbo, China
| | - Yushan Mao
- The Affiliated Hospital of the Medical School of Ningbo University, Ningbo, China
| | - Xiaoyan Yu
- The Affiliated Hospital of the Medical School of Ningbo University, Ningbo, China
| |
Collapse
|
17
|
Chen F, Wang J, Wu Y, Gao Q, Zhang S. Potential Biomarkers for Liver Cancer Diagnosis Based on Multi-Omics Strategy. Front Oncol 2022; 12:822449. [PMID: 35186756 PMCID: PMC8851237 DOI: 10.3389/fonc.2022.822449] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Accepted: 01/17/2022] [Indexed: 12/11/2022] Open
Abstract
Liver cancer is the fourth leading cause of cancer-related death worldwide. Hepatocellular carcinoma (HCC) accounts for about 85%-90% of all primary liver malignancies. However, only 20-30% of HCC patients are eligible for curative therapy mainly due to the lack of early-detection strategies, highlighting the significance of reliable and accurate biomarkers. The integration of multi-omics became an important tool for biomarker screening and unique alterations in tumor-associated genes, transcripts, proteins, post-translational modifications and metabolites have been observed. We here summarized the novel biomarkers for HCC diagnosis based on multi-omics technology as well as the clinical significance of these potential biomarkers in the early detection of HCC.
Collapse
Affiliation(s)
- Fanghua Chen
- Center for Tumor Diagnosis & Therapy, Jinshan Hospital, Fudan University, Shanghai, China
- Liver Cancer Institute, Zhongshan Hospital, and Key Laboratory of Carcinogenesis and Cancer Invasion (Ministry of Education), Fudan University, Shanghai, China
| | - Junming Wang
- Liver Cancer Institute, Zhongshan Hospital, and Key Laboratory of Carcinogenesis and Cancer Invasion (Ministry of Education), Fudan University, Shanghai, China
| | - Yingcheng Wu
- Liver Cancer Institute, Zhongshan Hospital, and Key Laboratory of Carcinogenesis and Cancer Invasion (Ministry of Education), Fudan University, Shanghai, China
| | - Qiang Gao
- Center for Tumor Diagnosis & Therapy, Jinshan Hospital, Fudan University, Shanghai, China
- Liver Cancer Institute, Zhongshan Hospital, and Key Laboratory of Carcinogenesis and Cancer Invasion (Ministry of Education), Fudan University, Shanghai, China
| | - Shu Zhang
- Center for Tumor Diagnosis & Therapy, Jinshan Hospital, Fudan University, Shanghai, China
- Liver Cancer Institute, Zhongshan Hospital, and Key Laboratory of Carcinogenesis and Cancer Invasion (Ministry of Education), Fudan University, Shanghai, China
- *Correspondence: Shu Zhang,
| |
Collapse
|
18
|
Li J, Wang H. H3K27ac-activated EGFR-AS1 promotes cell growth in cervical cancer through ACTN4-mediated WNT pathway. Biol Direct 2022; 17:3. [PMID: 34998421 PMCID: PMC8742952 DOI: 10.1186/s13062-021-00315-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2021] [Accepted: 11/29/2021] [Indexed: 01/09/2023] Open
Abstract
Background Recently, extensive studies unveiled that lncRNAs exert critical function in the development and progression of cervical cancer (CC). EGFR-AS1 is a novel lncRNA which has not been well-explored in CC. Aims Our study aimed to research the function and molecular mechanism of EGFR-AS1 in CC cells. qRT-PCR analysis was performed to detect gene expression. Colony formation, EdU, flow cytometry, TUNEL, western blot and transwell assays were performed to assess the effect of EGFR-AS1 on CC cell growth. The regulatory mechanism of EGFR-AS1 was dug out through mechanism experiments. Results EGFR-AS1 was notably overexpressed in CC cell lines. Loss-of-functional experiments revealed that EGFR-AS1 promoted CC cell proliferation, migration and invasion, and suppressed cell apoptosis. Mechanistically, up-regulation of EGFR-AS1 was attributed to the activation of H3K27 acetylation (H3K27ac). Further, EGFR-AS1 was revealed to function as miR-2355-5p sponge. Additionally, miR-2355-5p was down-regulated in CC cells and ACTN4 was identified as a target gene of miR-2355-5p. Ultimately, overexpressed ACTN4 could reserve the suppressive role of EGFR-AS1 silencing in CC cell growth. Last but not least, EGFR-AS1 facilitated CC cell growth via ACTN4-mediated WNT pathway. Conclusions H3K27ac-activated EGFR-AS1 sponged miR-2355-5p and promoted CC cell growth through ACTN4-mediated WNT pathway. Supplementary Information The online version contains supplementary material available at 10.1186/s13062-021-00315-5.
Collapse
Affiliation(s)
- Jingyan Li
- Zibo Maternal and Child Health Hospital of Shandong Province, Zibo, 255000, Shandong, China
| | - Hongbing Wang
- Department of Gynecology and Oncology, Hubei Cancer Hospital Affiliated to Tongji Medical College of Huazhong University of Science and Technology, Hongshan District, No. 116 Zhuodaoquan South Road, Wuhan, 430079, Hubei, China.
| |
Collapse
|
19
|
Yue M, Liu T, Yan G, Luo X, Wang L. LINC01605, regulated by the EP300-SMYD2 complex, potentiates the binding between METTL3 and SPTBN2 in colorectal cancer. Cancer Cell Int 2021; 21:504. [PMID: 34544413 PMCID: PMC8451128 DOI: 10.1186/s12935-021-02180-8] [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: 08/09/2021] [Accepted: 08/25/2021] [Indexed: 12/28/2022] Open
Abstract
Background Colorectal cancer (CC) is one of the major contributors to tumor-related death worldwide, and its main cause of death is distant metastasis. Dysregulation of long non-coding RNA (lncRNA) LINC01605 has been implicated in CC. However, its role in metastasis of CC remains elusive. The goal of the study is to uncover the biological function and molecular mechanism of LINC01605 in CC. Methods The differentially expressed lncRNAs were first screened from GSE97300, GSE84983, GSE110715, GSE70880, and GSE75970 microarrays. The correlation between the expression of LINC01605 and the clinical phenotypes of enrolled CC patients (n = 134) was subsequently analyzed. The upstream and downstream regulatory mechanisms of LINC01605 in CC were identified through bioinformatics and RNA-seq analyses. Finally, the effects of related factors on CC cell growth and metastasis were confirmed through functional validation experiments. Results LINC01605, significantly highly expressed in CC, was a prognostic factor for patients with CC. Functional experiments revealed that LINC01605 knockdown inhibited the proliferatory and metastatic potential of CC cells in vitro and in vivo. Moreover, LINC01605 was regulated by SMYD2-EP300-mediated modifications of histone H3K4me3 as well as H3K27ac. LINC01605 was found to bind to METTL3 and promote the m6A modification of SPTBN2 mRNA, thereby facilitating the translation of SPTBN2. Conclusions Overexpression of LINC01605, regulated by SMYD2-EP300-mediated H3K27ac and H3K4me3 modifications, bound to METTL3 protein to promote m6A modification of SPTBN2 mRNA, leading to the development of CC. Supplementary Information The online version contains supplementary material available at 10.1186/s12935-021-02180-8.
Collapse
Affiliation(s)
- Meng Yue
- Department of Colorecal and Anal Surgery, The First Hospital of Jilin University, No. 71, Xinmin Street, Chaoyang District, Changchun, 130021, Jilin, People's Republic of China
| | - Tao Liu
- Department of Colorecal and Anal Surgery, The First Hospital of Jilin University, No. 71, Xinmin Street, Chaoyang District, Changchun, 130021, Jilin, People's Republic of China
| | - Guoqiang Yan
- Department of Colorecal and Anal Surgery, The First Hospital of Jilin University, No. 71, Xinmin Street, Chaoyang District, Changchun, 130021, Jilin, People's Republic of China
| | - Xiaofan Luo
- Department of Colorecal and Anal Surgery, The First Hospital of Jilin University, No. 71, Xinmin Street, Chaoyang District, Changchun, 130021, Jilin, People's Republic of China
| | - Lei Wang
- Department of Colorecal and Anal Surgery, The First Hospital of Jilin University, No. 71, Xinmin Street, Chaoyang District, Changchun, 130021, Jilin, People's Republic of China.
| |
Collapse
|
20
|
Liu Y, Shi P, Fang H, Zhao Z, Yang F, Zhang J, Jing S, Geng C. Long non-coding RNA GHET1 promotes thyroid cancer cell proliferation and invasion. Transl Cancer Res 2021; 10:4148-4157. [PMID: 35116711 PMCID: PMC8797618 DOI: 10.21037/tcr-21-1615] [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: 07/16/2021] [Accepted: 09/07/2021] [Indexed: 12/13/2022]
Abstract
BACKGROUND We aimed to evaluate the role of long non-coding RNA (LncRNA) gastric carcinoma proliferation-enhancing transcript 1 (GHET1) on thyroid cancer (TC) behavior in vitro. METHODS TC tissues and paired adjacent normal tissues were obtained after surgical excision from 43 patients with TC. The expression of LncRNA GHET1 was analyzed by real-time (RT) PCR. Human papillary thyroid cancer cell lines (TPC-1, BCPAP) were used to examine the role of LncRNA GHET1 in vitro. Cell proliferation was determined by CCK8 and cell colony formation assays. Transwell and wound-healing assays were used to detect the invasion and migration of thyroid cancer cells. RESULTS Our results showed that LncRNA GHET1 was significantly more upregulated in TC tissues than in adjacent normal tissues. LncRNA GHET1 was also increased in thyroid cancer cell lines compared to normal thyroid cell lines. The upregulation of LncRNA GHET1 was significantly associated with tumor invasion, gender, and lymph node metastasis in patients with thyroid cancer. The in vitro studies showed that silencing LncRNA GHET1 in BCPAP cells inhibited cell proliferation, cell invasion, and migration. Silencing of LncRNA GHETI also promoted the cell apoptotic rate, caused an increase in the cell population at the G0/G1 phase, and decreased the cell population at the S phase. In contrast, the overexpression of LncRNA GHET1 promoted cell proliferation, invasion, and migration, inhibited cell apoptosis, and increased cell population at the S phase in TPC cells. CONCLUSIONS LncRNA GHET1 dysregulation might be involved in the carcinogenesis of thyroid cancer. LncRNA GHET1 could be used as a potential molecular marker and molecular target for TC.
Collapse
Affiliation(s)
- Yan Liu
- Department of Otorhinolaryngology, Hebei Medical University Fourth Affiliated Hospital and Hebei Provincial Tumor Hospital, Shijiazhuang, China
| | - Ping Shi
- Department of Otorhinolaryngology, Hebei Medical University Fourth Affiliated Hospital and Hebei Provincial Tumor Hospital, Shijiazhuang, China
| | - Hao Fang
- Hepatobiliary Surgery Department, Hebei Medical University Fourth Affiliated Hospital and Hebei Provincial Tumor Hospital, Shijiazhuang, China
| | - Zhen Zhao
- Department of Otorhinolaryngology, Hebei Medical University Fourth Affiliated Hospital and Hebei Provincial Tumor Hospital, Shijiazhuang, China
| | - Fei Yang
- Department of Otorhinolaryngology, Hebei Medical University Fourth Affiliated Hospital and Hebei Provincial Tumor Hospital, Shijiazhuang, China
| | - Jie Zhang
- Department of Otorhinolaryngology, Hebei Medical University Fourth Affiliated Hospital and Hebei Provincial Tumor Hospital, Shijiazhuang, China
| | - Shanghua Jing
- Department of Otorhinolaryngology, Hebei Medical University Fourth Affiliated Hospital and Hebei Provincial Tumor Hospital, Shijiazhuang, China
| | - Cuizhi Geng
- Breast Oncology Department, Hebei Medical University Fourth Affiliated Hospital and Hebei Provincial Tumor Hospital, Shijiazhuang, China
| |
Collapse
|
21
|
He Y, Gong P, Wang S, Xu Q, Chen J. The significance of homeodomain transcription factor 2 in colon cancer cells. Biomed Eng Online 2021; 20:81. [PMID: 34372865 PMCID: PMC8351361 DOI: 10.1186/s12938-021-00912-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Accepted: 07/22/2021] [Indexed: 12/01/2022] Open
Abstract
Background Colon cancer is a serious malignant tumor. It has been reported that paired-like homeodomain transcription factor 2 (PITX2) can promote the progression of several types of cancer via regulating the Wnt/β-catenin pathway. It has also been demonstrated that high levels of long non-coding RNA (lncRNA) gastric carcinoma high expressed transcript 1 (GHET1) can also promote the development of cervical cancer via activating the Wnt/β-catenin pathway. However, whether PITX2 can affect the development of colon cancer via regulating the expression of lncRNA GHET1 remains unclear. Results The results demonstrated that PITX2 knockdown attenuated the proliferation, migration and invasion abilities of colon cancer cells. Additionally, PITX2 promoted the expression of lncRNA GHET1 via binding to its promoter. Overexpression of lncRNA GHET1 induced the expression of Wnt/β-catenin signaling-related proteins, cyclin D1, c-Myc and MMP-7. Furthermore, lncRNA GHET1 overexpression abrogated the PITX2 silencing-mediated decreased proliferation, migration and invasion abilities of colon cancer cells. Conclusion The findings of the present study suggested that PITX2 could enhance the proliferation, migration and invasion abilities of colon cancer cells via upregulating lncRNA GHET1 and activating the Wnt/β-catenin pathway.
Collapse
Affiliation(s)
- Yang He
- Department of Oncology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, 301 Yanchang Road Middle, Shanghai, 200072, People's Republic of China.,Department of Interventional Oncology, Shanghai DaHua Hospital, Shanghai, 200072, People's Republic of China
| | - Peng Gong
- Department of Oncology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, 301 Yanchang Road Middle, Shanghai, 200072, People's Republic of China
| | - Sitong Wang
- Department of Oncology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, 301 Yanchang Road Middle, Shanghai, 200072, People's Republic of China
| | - Qing Xu
- Department of Oncology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, 301 Yanchang Road Middle, Shanghai, 200072, People's Republic of China.
| | - Jianhua Chen
- Department of Oncology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, 301 Yanchang Road Middle, Shanghai, 200072, People's Republic of China.
| |
Collapse
|
22
|
Chen C, Chen Q, Wu J, Zou H. H3K27ac-induced FOXC2-AS1 accelerates tongue squamous cell carcinoma by upregulating E2F3. J Oral Pathol Med 2021; 50:1018-1030. [PMID: 34358374 DOI: 10.1111/jop.13232] [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: 05/25/2021] [Revised: 07/09/2021] [Accepted: 07/13/2021] [Indexed: 12/23/2022]
Abstract
BACKGROUND The important roles of lncRNAs have been reported in cancers, including tongue squamous cell carcinoma (TSCC). Here, we investigated the functional role and molecular mechanisms of lncRNA FOXC2-AS1 in TSCC. METHODS The expression level of FOXC2-AS1 in TSCC was determined by RT-qPCR. Its biological role was evaluated through colony formation assay, flow cytometry, wound healing, transwell, and Western blot analyses. The interactions among gene were tested by mechanistic investigations. RESULTS FOXC2-AS1 expression was high in TSCC tissues and cells. Functional assays in vitro showed that silencing FOXC2-AS1 restrained cell proliferation, cell cycle, migration, invasion, and EMT. In the mechanism, it was verified that H3K27 acetylation (H3K27ac) triggered an increase in FOXC2-AS1 expression. Furthermore, FOXC2-AS1 was identified as a cytoplasmic lncRNA and served as a ceRNA to upregulate E2F3 expression via sponging miR-6868-5p. CONCLUSION H3K27ac-induced FOXC2-AS1 exhibits carcinogenic property in TSCC by the miR-6868-5p/E2F3 axis.
Collapse
Affiliation(s)
- Cuiting Chen
- Department of Stomatology, The Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Qiongyu Chen
- Department of Stomatology, The Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Jian Wu
- Department of Stomatology, The Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Han Zou
- Department of Stomatology, The Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| |
Collapse
|
23
|
Ma Y, Zheng W. H3K27ac-induced lncRNA PAXIP1-AS1 promotes cell proliferation, migration, EMT and apoptosis in ovarian cancer by targeting miR-6744-5p/PCBP2 axis. J Ovarian Res 2021; 14:76. [PMID: 34108034 PMCID: PMC8191132 DOI: 10.1186/s13048-021-00822-z] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Accepted: 05/07/2021] [Indexed: 02/02/2023] Open
Abstract
We aimed to explore role of lncRNA PAX-interacting protein 1-antisense RNA1 (PAXIP1-AS1) in ovarian cancer (OC). RT-qPCR analysis identified upregulation of PAXIP1-AS1 in OC cell lines. Functionally, PAXIP1-AS1 knockdown inhibited cell proliferation, accelerated cell apoptosis, and suppressed cell migration and epithelial-mesenchymal transition (EMT) process. Upregulation of PAXIP1-AS1 was induced by CBP-mediated H3K27 acetylation (H3K27ac) via bioinformatic analysis and ChIP assay. Furthermore, PAXIP1-AS1 served as a competing endogenous RNA (ceRNA) to regulate PCBP2 expression by sponging microRNA-6744-5p (miR-6744-5p). Restoration experiments showed that overexpressed PCBP2 rescued effects of silenced PAXIP1-AS1 on cell proliferation, apoptosis, migration and EMT. Overall, lncRNA PAXIP1-AS1 activated by H3K27ac functioned as a tumor promoter in OC via mediating miR-6744-5p/PCBP2 axis, which provided promising insight into exploration on OC therapy.
Collapse
Affiliation(s)
- Yimin Ma
- Department of Gynecology, Ningbo Medical Center Lihuili Hospital, Ningbo, 315040, Zhejiang, China.
| | - Wei Zheng
- Department of Gynecology, Xi'an Military Industry Hospital, Xi'an, 710065, Shaanxi, China
| |
Collapse
|
24
|
Liu Y, Huang X, Guo L, Luo N. LINC00649 Facilitates the Cellular Process of Bladder Cancer Cells via Signaling Axis miR-16-5p/JARID2. Urol Int 2021; 106:304-312. [PMID: 33789312 DOI: 10.1159/000506239] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Accepted: 01/27/2020] [Indexed: 11/19/2022]
Abstract
Bladder cancer (BC), as one of the most common cancers around the world, begins in the inner side of the bladder and is inclined to spread to the remaining parts of the body. Extensive documents have shown that long noncoding RNAs function as stimuli in various cancer types. With regard to LINC00649, there is limited investigation on its role previously. In our research, we discovered that LINC00649 was considerably highly expressed in BC cells and the lack of LINC00649 would cause inactivity in cellular proliferation, migration, and invasion. miR-16-5p turned out to be competitively incorporated by LINC00649 in the upstream or JARID2 downstream. In BC cells, LINC00649 was found to bind with miR-16-5p to increase the expression of JARID2. Overly expressed JARID2 was found to reverse the LINC00649 shortage-mediated suppressive impacts on the cellular process of BC cells. Concisely, it was the first study on the molecular mechanism of LINC00649 in BC. This work detected that LINC00649 enhanced cell proliferation, migration, and invasion of BC cells by acting as a sponge of miR-16-5p and upregulating JARID2, providing novel insight into understating BC.
Collapse
Affiliation(s)
- Yongsong Liu
- Department of Urology, The Second People's Hospital of Yibin, Yibin, China
| | - Xiande Huang
- Department of Urology, Gansu Provincial Hospital, Lanzhou, China
| | - Lijun Guo
- Department of Urology, Gansu Provincial Hospital, Lanzhou, China
| | - Nengqin Luo
- Department of Urology, Gansu Provincial Hospital, Lanzhou, China
| |
Collapse
|
25
|
Yue M, Yun Z, Li S, Yan G, Kang Z. NEDD4 triggers FOXA1 ubiquitination and promotes colon cancer progression under microRNA-340-5p suppression and ATF1 upregulation. RNA Biol 2021; 18:1981-1995. [PMID: 33530829 DOI: 10.1080/15476286.2021.1885232] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
NEDD4 is an E3 ubiquitin ligase that recognizes substrates through protein-protein interactions and is involved in cancer development. This study aimed to elucidate the function of NEDD4 in colon cancer (CC) progression and its mechanism of action. NEDD4 was abundantly expressed in CC tissues and cells, and the overexpression of NEDD4 promoted the growth and metastasis of xenograft tumours as well as the tumorigenesis rate of primary CC in mouse models. In in vitro experiments, the silencing (or upregulation) of NEDD4 inhibited (or increased) the viability, invasion, and epithelial-to-mesenchymal transition of CC cells. The binding relationships between NEDD4 and FOXA1, FOXA1 and microRNA (miRNA)-340-5p, and miR-340-5p and ATF1 were validated by Co-immunoprecipitation, chromatin immunoprecipitation and luciferase assays, and NEDD4 was demonstrated to trigger FOXA1 ubiquitination and degradation. FOXA1 transcriptionally activated miR-340-5p, which subsequently bound to ATF1 mRNA. The upregulation of FOXA1 or miR-340-5p or the downregulation of ATF1 blocked certain functions of NEDD4 in CC cells. Altogether, NEDD4 was demonstrated to trigger FOXA1 ubiquitination and promote CC progression under the involvement of microRNA-340-5p suppression and ATF1 upregulation.
Collapse
Affiliation(s)
- Meng Yue
- Department of Colorecal & Anal Surgery, The First Hospital of Jilin University, Changchun, P.R. China
| | - Zhennan Yun
- Department of Colorecal & Anal Surgery, The First Hospital of Jilin University, Changchun, P.R. China
| | - Shiquan Li
- Department of Colorecal & Anal Surgery, The First Hospital of Jilin University, Changchun, P.R. China
| | - Guoqiang Yan
- Department of Colorecal & Anal Surgery, The First Hospital of Jilin University, Changchun, P.R. China
| | - Zhenhua Kang
- Department of Colorecal & Anal Surgery, The First Hospital of Jilin University, Changchun, P.R. China
| |
Collapse
|
26
|
Khajehdehi M, Khalaj-Kondori M, Ghasemi T, Jahanghiri B, Damaghi M. Long Noncoding RNAs in Gastrointestinal Cancer: Tumor Suppression Versus Tumor Promotion. Dig Dis Sci 2021; 66:381-397. [PMID: 32185664 DOI: 10.1007/s10620-020-06200-x] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2019] [Accepted: 03/07/2020] [Indexed: 01/17/2023]
Abstract
Approximately 80% of the human genome harbors biochemical marks of active transcription that its majority transcribes to noncoding RNAs, namely long noncoding RNAs (lncRNAs). LncRNAs are heterogeneous RNA transcripts that regulate critical biological processes such as cell survival and death. They involve in the progression of different cancers by affecting transcriptional and post-transcriptional modifications as well as epigenetic control of numerous tumor suppressors and oncogenes. Recent findings show that aberrant expression of lncRNAs is associated with tumor initiation, progression, invasion, and overall survival of patients with gastrointestinal (GI) cancers. Some lncRNAs play as tumor suppressors in all GI cancers, but others play as tumor promoters. However, some other lncRNAs might function as a tumor suppressor in one GI cancer, but as a tumor promoter in another GI cancer type. This fact highlights possible context dependency of the expression patterns and roles of at least some lncRNAs in GI cancer development and progression. Here, we review the functional relation of lncRNAs involved in the development and progression of GI cancer by focusing on their roles as tumor suppressor and tumor promoter genes.
Collapse
Affiliation(s)
- Mina Khajehdehi
- Department of Animal Biology, Faculty of Natural Science, University of Tabriz, Tabriz, Iran
| | - Mohammad Khalaj-Kondori
- Department of Animal Biology, Faculty of Natural Science, University of Tabriz, Tabriz, Iran.
| | - Tayyebeh Ghasemi
- Department of Animal Biology, Faculty of Natural Science, University of Tabriz, Tabriz, Iran
| | - Babak Jahanghiri
- Department of Molecular Medicine, Institute of Medical Biotechnology, National Institute of Genetic Engineering and Biotechnology, Tehran, Iran
| | - Mehdi Damaghi
- Department of Oncologic Sciences, Morsani College of Medicine, University of South Florida, Tampa, 33612, FL, USA
| |
Collapse
|
27
|
Ganguly N, Chakrabarti S. Role of long non‑coding RNAs and related epigenetic mechanisms in liver fibrosis (Review). Int J Mol Med 2021; 47:23. [PMID: 33495817 PMCID: PMC7846421 DOI: 10.3892/ijmm.2021.4856] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2020] [Accepted: 10/29/2020] [Indexed: 02/07/2023] Open
Abstract
Liver fibrosis is one of the major liver pathologies affecting patients worldwide. It results from an improper tissue repair process following liver injury or inflammation. If left untreated, it ultimately leads to liver cirrhosis and liver failure. Long non‑coding RNAs (lncRNAs) have been implicated in a wide variety of diseases. They can regulate gene expression and modulate signaling. Some of the lncRNAs promote, while others inhibit liver fibrosis. Similarly, other epigenetic processes, such as methylation and acetylation regulate gene transcription and can modulate gene expression. Notably, there are several regulatory associations of lncRNAs with other epigenetic processes. A major mechanism of action of long non‑coding RNAs is to competitively bind to their target microRNAs (miRNAs or miRs), which in turn affects miRNA availability and bioactivity. In the present review, the role of lncRNAs and related epigenetic processes contributing to liver fibrosis is discussed. Finally, various potential therapeutic approaches targeting lncRNAs and related epigenetic processes, which are being considered as possible future treatment targets for liver fibrosis are identified.
Collapse
Affiliation(s)
- Niladri Ganguly
- Department of Pathology and Laboratory Medicine, University of Western Ontario, London, ON N6A 5C1, Canada
| | - Subrata Chakrabarti
- Department of Pathology and Laboratory Medicine, University of Western Ontario, London, ON N6A 5C1, Canada
| |
Collapse
|
28
|
Yuan D, Chen Y, Li X, Li J, Zhao Y, Shen J, Du F, Kaboli PJ, Li M, Wu X, Ji H, Cho CH, Wen Q, Li W, Xiao Z, Chen B. Long Non-Coding RNAs: Potential Biomarkers and Targets for Hepatocellular Carcinoma Therapy and Diagnosis. Int J Biol Sci 2021; 17:220-235. [PMID: 33390845 PMCID: PMC7757045 DOI: 10.7150/ijbs.50730] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Accepted: 11/01/2020] [Indexed: 12/24/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is one of the leading causes of cancer-related death worldwide. Increasing studies showed that long non-coding RNAs (lncRNAs), a novel class of RNAs that are greater than 200 nucleotides in length but lack the ability to encode proteins, exert crucial roles in the occurrence and progression of HCC. LncRNAs promote the proliferation, migration, invasion, autophagy, and apoptosis of tumor cells by regulating downstream target gene expression and cancer-related signaling pathways. Meanwhile, lncRNA can be used as biomarkers to predict the efficacy of HCC treatment strategies, such as surgery, radiotherapy, chemotherapy, and immunotherapy, and as a potential individualized tool for HCC diagnosis and treatment. In this review, we overview up-to-date findings on lncRNAs as potential biomarkers for HCC surgery, radiotherapy, chemotherapy resistance, target therapy, and immunotherapy, and discuss the potential clinical application of lncRNA as tools for HCC diagnosis and treatment.
Collapse
Affiliation(s)
- Donghong Yuan
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China.,South Sichuan Institute of Translational Medicine, Luzhou, Sichuan, China
| | - Yu Chen
- Department of Oncology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Xiaobing Li
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China
| | - Jing Li
- Department of Oncology and Hematology, Hospital (T.C.M) Affiliated to Southwest Medical University, Luzhou, Sichuan, China
| | - Yueshui Zhao
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China.,South Sichuan Institute of Translational Medicine, Luzhou, Sichuan, China
| | - Jing Shen
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China.,South Sichuan Institute of Translational Medicine, Luzhou, Sichuan, China
| | - Fukuan Du
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China.,South Sichuan Institute of Translational Medicine, Luzhou, Sichuan, China
| | - Parham Jabbarzadeh Kaboli
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China.,South Sichuan Institute of Translational Medicine, Luzhou, Sichuan, China
| | - Mingxing Li
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China.,South Sichuan Institute of Translational Medicine, Luzhou, Sichuan, China
| | - Xu Wu
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China.,South Sichuan Institute of Translational Medicine, Luzhou, Sichuan, China
| | - Huijiao Ji
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China.,South Sichuan Institute of Translational Medicine, Luzhou, Sichuan, China
| | - Chi Hin Cho
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China.,South Sichuan Institute of Translational Medicine, Luzhou, Sichuan, China
| | - Qinglian Wen
- South Sichuan Institute of Translational Medicine, Luzhou, Sichuan, China.,Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Wanping Li
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China
| | - Zhangang Xiao
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China.,South Sichuan Institute of Translational Medicine, Luzhou, Sichuan, China
| | - Bo Chen
- Science and Technology Achievement Incubation Center, Kunming Medical University, Kunming, Yunnan, China
| |
Collapse
|
29
|
Poursheikhani A, Nokhandani N, Yousefi H, Rad DM, Sahebkar A. Clinicopathological Significance of Long Non-Coding RNA GHET1 in Human Cancers: A Meta-Analysis. Curr Pharm Biotechnol 2020; 21:1422-1432. [DOI: 10.2174/1389201021999200727163238] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Revised: 03/30/2020] [Accepted: 06/14/2020] [Indexed: 02/06/2023]
Abstract
Introduction:
Cancer is considered as the main public health problem and the second leading
cause of morbidity and mortality worldwide. Numerous environmental-lifestyle related risk factors
account for around one-third of cancer deaths. Recently, the key role of lncRNAs has been widely investigated
in a variety of disorders, including cancer. The lncRNA GHET1 has been considered as an
essential oncogenic lncRNA in many types of human cancers. Clinical investigations indicated that expression
of lncRNA GHET1 is correlated with clinicopathological characteristics in cancer. This metaanalysis
investigated the correlation between the lncRNA GHET1 expression and clinicopathological
features in different types of cancers.
Materials and Methods:
Comprehensive literature searches in PubMed, Scopus, and Web of
Knowledge were conducted up to April 11, 2019. Sixteen studies were included in this meta-analysis.
All statistical analyses were conducted using Stata software, version 12.0.
Results:
The pooled OR and 95%CIs of the sixteen relevant studies showed that over expression of
lncRNA GHET1 was associated with tumor-size ≥5 cm (OR= 2.51, 95% CI: 1.89-3.33, p=0.00,
I2=38.30%), positive lymph node metastasis (OR= 2.83, 95% CI: 1.78-4.52, p=0.00, I2=45.60%), advanced
tumor stage (OR= 3.92, 95% CI: 2.97-5.19, p=0.00, I2=0.00%), positive distant metastasis
(OR= 5.74, 95% CI: 2.58-12.77, p=0.00, I2=0.00%), advanced tumor status (OR= 2.97, 95% CI: 1.40-
6.29, p=0.01, I2=34.70%), and positive vascular invasion (OR= 2.69, 95% CI: 1.61-4.50, p=0.00,
I2=29.20%).
Conclusion:
Taken together, the current study demonstrated that overexpression of lncRNA GHET1 is
significantly associated with clinicopathological features in human cancers. Our results suggested that
lncRNA GHET1 can be utilized as a prognostic biomarker in human cancer.
Collapse
Affiliation(s)
- Arash Poursheikhani
- Medical Genetics Research Centre, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Negin Nokhandani
- Department of Medical Immunology, Faculty of Medicine, Golestan University of Medical Sciences, Golestan, Iran
| | - Hassan Yousefi
- Louisiana State University, School of Medicine, New Orleans, LA, United States
| | - Dorsa M. Rad
- Medical Genetics Research Centre, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | | |
Collapse
|
30
|
Shin TJ, Lee KH, Cho JY. Epigenetic Mechanisms of LncRNAs Binding to Protein in Carcinogenesis. Cancers (Basel) 2020; 12:E2925. [PMID: 33050646 PMCID: PMC7599656 DOI: 10.3390/cancers12102925] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Revised: 10/09/2020] [Accepted: 10/09/2020] [Indexed: 02/06/2023] Open
Abstract
Epigenetic dysregulation is an important feature for cancer initiation and progression. Long non-coding RNAs (lncRNAs) are transcripts that stably present as RNA forms with no translated protein and have lengths larger than 200 nucleotides. LncRNA can epigenetically regulate either oncogenes or tumor suppressor genes. Nowadays, the combined research of lncRNA plus protein analysis is gaining more attention. LncRNA controls gene expression directly by binding to transcription factors of target genes and indirectly by complexing with other proteins to bind to target proteins and cause protein degradation, reduced protein stability, or interference with the binding of other proteins. Various studies have indicated that lncRNA contributes to cancer development by modulating genes epigenetically and studies have been done to determine which proteins are combined with lncRNA and contribute to cancer development. In this review, we look in depth at the epigenetic regulatory function of lncRNAs that are capable of complexing with other proteins in cancer development.
Collapse
Affiliation(s)
| | | | - Je-Yoel Cho
- Department of Biochemistry, BK21 Plus and Research Institute for Veterinary Science, School of Veterinary Medicine, Seoul National University, Seoul 08826, Korea; (T.-J.S.); (K.-H.L.)
| |
Collapse
|
31
|
Prognostic significance of LncRNA GHET1 expression in various cancers: a systematic review and meta-analysis. Biosci Rep 2020; 39:BSR20190608. [PMID: 31227613 PMCID: PMC6822487 DOI: 10.1042/bsr20190608] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2019] [Revised: 06/04/2019] [Accepted: 06/14/2019] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Dysregulated expression of long non-coding RNA gastric carcinoma high expressed transcript 1 (lncRNA GHET1) has been observed in several cancers, however, definite conclusion on the prognostic value of lncRNA GHET1 expression in human cancers has not been determined. The aim of this meta-analysis was to evaluate the prognostic significance of lncRNA GHET1 expression in cancers. METHODS PubMed, Web of Science and Embase were comprehensively searched for relevant studies. Meta-analyses of overall survival (OS) and clinicopathological features were conducted. RESULTS Ten studies were finally analyzed in the present study. High lncRNA GHET1 expression was associated with shorter OS than low lncRNA GHET1 expression in cancers (hazard ratio (HR) = 2.59, 95% CI = 1.93-3.47, P<0.01). Online cross-validation using The Cancer Genome Atlas (TCGA) data observed similar results (HR = 1.10, P<0.05). When compared with low lncRNA GHET1 expression, high lncRNA GHET1 expression was related to larger tumor size (P<0.01), worse differentiation (P<0.01), earlier distant metastasis (P=0.02), earlier lymph node metastasis (P<0.01) and more advanced clinical stage (P<0.01). CONCLUSION High lncRNA GHET1 expression is associated with worse cancer prognosis and can serve as a promising prognostic factor of human cancers.
Collapse
|
32
|
Zhao Q, Zhang Z, Li J, Xu F, Zhang B, Liu M, Liu Y, Chen H, Yang J, Zhang J. Lysine Acetylome Study of Human Hepatocellular Carcinoma Tissues for Biomarkers and Therapeutic Targets Discovery. Front Genet 2020; 11:572663. [PMID: 33093847 PMCID: PMC7527632 DOI: 10.3389/fgene.2020.572663] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Accepted: 08/26/2020] [Indexed: 12/16/2022] Open
Abstract
Lysine acetylation is a vital post-translational modification (PTM) of proteins, which plays an important role in cancer development. In healthy human liver tissues, multiple non-histone proteins were identified with acetylation modification, however, the role of acetylated proteins in hepatocellular carcinoma (HCC) development remains largely unknown. Here we performed a quantitative acetylome study of tumor and normal liver tissues from HCC patients. Overall, 598 lysine acetylation sites in 325 proteins were quantified, and almost 59% of their acetylation levels were significantly changed. The differentially acetylated proteins mainly consisted of non-histone proteins located in mitochondria and cytoplasm, which accounted for 42% and 24%, respectively. Bioinformatics analysis showed that differentially acetylated proteins were enriched in metabolism, oxidative stress, and signal transduction processes. In tumor tissues, 278 lysine sites in 189 proteins showed decreased acetylation levels, which occupied 98% of differentially acetylated proteins. Moreover, we collected twenty pairs of tumor and normal liver tissues from HCC male patients, and found that expression levels of SIRT1 (p = 0.002), SIRT2 (p = 0.01), and SIRT4 (p = 0.045) were significantly up-regulated in tumor tissues. Over-expression of possibly accounted for the widespread deacetylation of non-histone proteins identified in HCC tumor tissues, which could serve as promising predictors of HCC. Taken together, our work illustrates abundant differentially acetylated proteins in HCC tumor tissues, and offered insights into the role of lysine acetylation in HCC development. It provided potential biomarker and drug target candidates for clinical HCC diagnosis and treatment.
Collapse
Affiliation(s)
- Qianwei Zhao
- BGI College and Henan Institute of Medical and Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, China.,Henan Key Laboratory for Pharmacology of Liver Diseases, Zhengzhou University, Zhengzhou, China
| | - Zhendong Zhang
- BGI College and Henan Institute of Medical and Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, China
| | - Jinxia Li
- School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Fang Xu
- BGI College and Henan Institute of Medical and Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, China
| | - Bingxia Zhang
- School of Life Sciences, Zhengzhou University, Zhengzhou, China
| | - Mengduan Liu
- BGI College and Henan Institute of Medical and Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, China
| | - Yixian Liu
- BGI College and Henan Institute of Medical and Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, China
| | - Huiping Chen
- BGI College and Henan Institute of Medical and Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, China
| | - Junxia Yang
- BGI College and Henan Institute of Medical and Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, China
| | - Jintao Zhang
- BGI College and Henan Institute of Medical and Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, China
| |
Collapse
|
33
|
Clinicopathological and Prognostic Value of Gastric Carcinoma Highly Expressed Transcript 1 in Cancer: A Meta-Analysis. JOURNAL OF ONCOLOGY 2020; 2020:6341093. [PMID: 32908508 PMCID: PMC7471811 DOI: 10.1155/2020/6341093] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Revised: 07/20/2020] [Accepted: 07/24/2020] [Indexed: 11/25/2022]
Abstract
Background Long noncoding RNA gastric cancer highly expressed transcript 1 (lncRNA GHET1) is often reported to be abnormally expressed in multiple cancers, but the situation is different in different cancers. Therefore, a meta-analysis is necessary to clarify the value of lncRNA GHET1 as a prognostic indicator in cancer. Methods Relevant research studies on lncRNA GHET1 and cancer were retrieved from three electronic literature databases of Web of Science, PubMed, and OVID. Meanwhile, hazard ratios (HRs) and 95% confidence intervals (CIs) were calculated to explore the relationship between lncRNA GHET1 expression and survival of cancer patients. The odds ratios (ORs) and 95% CIs were calculated to assess the association of lncRNA GHET1 expression with pathological parameters of cancer patients. Results The meta-analysis included a total of 11 studies involving 714 cancer patients. The pooled HR suggests that high lncRNA GHET1 expression is associated with poor overall survival. In addition, high expression of lncRNA GHET1 was found to be associated with larger tumor size, poor histological grade, high tumor stage, lymph node metastasis, and distant metastasis. Conclusions High lncRNA GHET1 expression can predict poor survival and pathological parameters. And lncRNA GHET1 could serve as a new indicator in multiple cancers.
Collapse
|
34
|
Wang H, Meng F, Dong S. Aberrant Expression of Gastric Carcinoma High Expressed Transcript 1 is a Diagnostic and Prognostic Biomarker in Human Cancer. J Natl Med Assoc 2020; 112:602-612. [PMID: 32653201 DOI: 10.1016/j.jnma.2020.05.017] [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] [Received: 05/11/2020] [Revised: 05/25/2020] [Accepted: 05/25/2020] [Indexed: 11/19/2022]
Abstract
BACKGROUND Gastric carcinoma high expressed transcript 1 (GHET1), a long noncoding RNA (LncRNA), has been reported to be involved in tumor genesis and cancer progression. High GHET1 expression is correlated with a poor prognosis in cancer. In this meta-analysis, we investigated the association between GHET1 and lymph node metastasis, differentiation, vascular invasion and so on in human cancer. METHODS We performed systematic search in PubMed, EMBASE, Wiley Online Library, Wiley Online Library, and Medline from January 1, 1996 to September 25, 2017. A total of 8 studies were selected for further research. RESULTS The result showed that GHET1 expression was significantly associated with OS (hazard ratio [HR] = 2.23; 95% CI, 1.86-2.67; P < 0.0001), including digestive system tumor (HR = 2.17; 95% CI, 1.63-2.89; P < 0.0001). Moreover, high GHET1 expression was related to tumor size (odds ratio [OR] = 2.15, 95% CI: 1.57-2.94; P < 0.00001), TNM stage (Ⅲ+Ⅳ vs.Ⅰ+Ⅱ; OR = 4.02, 95% CI:3.06-5.28; P < 0.00001), lymph node metastasis (Yes vs. No; OR = 3.55, 95% CI:2.54-4.95; P < 0.00001), differentiation (poor vs. well or moderate; OR = 1.72, 95% CI:1.22-2.42; P = 0.002), vascular invasion (Yes vs. No; OR = 3.03, 95% CI:1.80-5.12; P = 0.00001). Also, we found that high expression of GHET1 has a significant relationship with analysis method and sample size. It almost higher in different human cancers. CONCLUSIONS GHET1 may serve as a potential clinical biomarker and poor survival and high-risk recurrence in cancers.
Collapse
Affiliation(s)
- Hua Wang
- School of Medicine, Jiangsu University, 301 Xuefu Road, Zhenjiang, Jiangsu, 212013, PR China.
| | - Fanyu Meng
- Department of Cardiology, Jilin Provincial People's Hospital, 1183 Gongnong Road, Changchun, Jilin, 130033, PR China
| | - Surong Dong
- Department of Laboratory Medicine, Taizhou People's Hospital, 366 Taihu Road, Taizhou, Jiangsu, 225300, PR China.
| |
Collapse
|
35
|
Cui X, Chen J, Zheng Y, Shen H. Circ_0000745 Promotes the Progression of Cervical Cancer by Regulating miR-409-3p/ATF1 Axis. Cancer Biother Radiopharm 2020; 37:766-778. [PMID: 32644859 DOI: 10.1089/cbr.2019.3392] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Background: Cervical cancer (CC) is a common gynecological malignancy with a high risk of recurrence and death. Circular RNAs play a crucial role in the occurrence and development of tumors. This study aimed to investigate the function and mechanism of circ_0000745 in CC. Methods: The levels of circ_0000745, miR-409-3p, and activating transcription factor 1 (ATF1) were determined by quantitative real-time polymerase chain reaction or western blot assay. Cell proliferation was assessed by colony formation assay. Cell migration and invasion were evaluated by transwell assay. Glycolysis was analyzed by measuring extracellular acidification rate, glucose uptake, and lactate production. Also, the protein levels of glucose transporter 1 and lactate dehydrogenase A were detected using western blot. The relationship among circ_0000745, miR-409-3p, and ATF1 were confirmed by dual-luciferase reporter assay. Moreover, xenograft assay was performed to analyze tumor growth in vivo. Results: Circ_0000745 and ATF1 were upregulated, whereas miR-409-3p was downregulated in CC tissues and cells. Knockdown of circ_0000745 repressed proliferation, migration, invasion, and glycolysis of CC cells. Circ_0000745 regulated CC progression by targeting miR-409-3p. Circ_0000745 modulated ATF1 expression through sponging miR-409-3p. MiR-409-3p hindered CC progression by targeting ATF1. Furthermore, depletion of circ_0000745 impeded tumor growth in vivo. Conclusion: Circ_0000745 promoted the progression of CC through modulating miR-409-3p/ATF1 axis, indicating a promising biomarker for CC therapy.
Collapse
Affiliation(s)
- Xia Cui
- Department of Gynecology, Changzhou No. 2 People's Hospital Affiliated to Nanjing Medical University, Changzhou, China
| | - Jiming Chen
- Department of Gynecology, Changzhou No. 2 People's Hospital Affiliated to Nanjing Medical University, Changzhou, China
| | - Yafeng Zheng
- Department of Gynecology, Changzhou No. 2 People's Hospital Affiliated to Nanjing Medical University, Changzhou, China
| | - Huaji Shen
- Department of Gynecology, Changzhou No. 2 People's Hospital Affiliated to Nanjing Medical University, Changzhou, China
| |
Collapse
|
36
|
LincRNA-ROR is activated by H3K27 acetylation and induces EMT in retinoblastoma by acting as a sponge of miR-32 to activate the Notch signaling pathway. Cancer Gene Ther 2020; 28:42-54. [PMID: 32439866 DOI: 10.1038/s41417-020-0181-z] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2020] [Revised: 05/06/2020] [Accepted: 05/07/2020] [Indexed: 12/19/2022]
Abstract
Recent studies have suggested that lincRNA-ROR is involved in the tumorigenesis of different types of cancers. However, the role of lincRNA-ROR in retinoblastoma has not been determined. We investigated lincRNA-ROR levels in 58 retinoblastoma and adjacent non-tumor tissues by quantitative reverse transcription PCR. Recurrence-free survival was analyzed using Cox regression analyses. Cell migration and invasion abilities were detected by wound-healing, Transwell invasion, and bioluminescence imaging assays. Western blotting was performed to detect epithelial-mesenchymal transition markers. Interactions between lincRNA-ROR, miR-32-5p, and Notch1 were confirmed by Luciferase, RNA pull-down, and RIP assays. Histone acetylation was detected by chromatin immunoprecipitation assays. We showed that lincRNA-ROR was significantly upregulated in retinoblastoma tissues, and overexpression of lincRNA-ROR was significantly correlated with optic nerve invasion, nodal or distant metastasis, and recurrence. We also showed that lincRNA-ROR is a critical promoter of retinoblastoma cell metastasis, both in vivo and in vitro. Further, we demonstrated that lincRNA-ROR activates the Notch signaling pathway by acting as a sponge of miR-32-5p. Upregulation of lincRNA-ROR was attributed to the CBP-mediated H3K27 acetylation at the promoter region. Our results reveal a potential competing endogenous RNA regulatory pathway, in which lincRNA-ROR modulates the epithelial-mesenchymal transition program by competitively binding to endogenous miR-32-5p and regulating Notch signaling pathway activity in retinoblastoma cells, which may provide new insights into novel molecular therapeutic targets for retinoblastoma.
Collapse
|
37
|
Ye P, Lv X, Aizemaiti R, Cheng J, Xia P, Di M. H3K27ac-activated LINC00519 promotes lung squamous cell carcinoma progression by targeting miR-450b-5p/miR-515-5p/YAP1 axis. Cell Prolif 2020; 53:e12797. [PMID: 32297697 PMCID: PMC7260072 DOI: 10.1111/cpr.12797] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2019] [Revised: 02/24/2020] [Accepted: 03/02/2020] [Indexed: 12/17/2022] Open
Abstract
Objectives Long non‐coding RNAs (lncRNAs) are extensively reported as participants in the biological process of diverse malignancies, including lung squamous cell carcinoma (LUSC). Long intergenic non‐protein coding RNA 519 (LINC00519) is identified as a novel lncRNA which has not yet been studied in cancers. Materials and Methods LINC00519 expression was detected by qRT‐PCR. The effect of LINC00519 on LUSC cellular activities was determined by in vitro and in vivo assays. Subcellular fractionation and FISH assays were conducted to identify the localization of LINC00519. The interaction between miR‐450b‐5p/miR‐515‐5p and LINC00519/YAP1 was verified by RIP, RNA pull‐down and luciferase reporter assays. Results Elevated level of LINC00519 was identified in LUSC tissues and cell lines. High LINC00519 level predicted unsatisfactory prognosis. Then, loss‐of‐function assays suggested the inhibitive role of silenced LINC00519 in cell proliferation, migration, invasion and tumour growth and promoting effect on cell apoptosis in LUSC. Mechanically, LINC00519 was activated by H3K27 acetylation (H3K27ac). Moreover, LINC00519 sponged miR‐450b‐5p and miR‐515‐5p to up‐regulate Yes1 associated transcriptional regulator (YAP1). Additionally, miR‐450b‐5p and miR‐515‐5p elicited anti‐carcinogenic effects in LUSC. Finally, rescue assays validated the effect of LINC00519‐miR‐450b‐5p‐miR‐515‐5p‐YAP1 axis in LUSC. Conclusions H3K27ac‐activated LINC00519 acts as a competing endogenous RNA (ceRNA) to promote LUSC progression by targeting miR‐450b‐5p/miR‐515‐5p/YAP1 axis.
Collapse
Affiliation(s)
- Peng Ye
- Department of Thoracic Surgery, The First Affiliated Hospital of Zhejiang University, Hangzhou, China
| | - Xiayi Lv
- Department of Thoracic Surgery, The First Affiliated Hospital of Zhejiang University, Hangzhou, China
| | - Rusidanmu Aizemaiti
- Department of Thoracic Surgery, The First Affiliated Hospital of Zhejiang University, Hangzhou, China
| | - Jun Cheng
- Department of Thoracic Surgery, The First Affiliated Hospital of Zhejiang University, Hangzhou, China
| | - Pinghui Xia
- Department of Thoracic Surgery, The First Affiliated Hospital of Zhejiang University, Hangzhou, China
| | - Meng Di
- Department of Thoracic Surgery, The First Affiliated Hospital of Zhejiang University, Hangzhou, China
| |
Collapse
|
38
|
Wang D, Zhang H, Fang X, Zhang X, Liu H. Prognostic value of long non-coding RNA GHET1 in cancers: a systematic review and meta-analysis. Cancer Cell Int 2020; 20:109. [PMID: 32280301 PMCID: PMC7137500 DOI: 10.1186/s12935-020-01189-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2019] [Accepted: 03/27/2020] [Indexed: 12/24/2022] Open
Abstract
Background A number of studies have demonstrated the critical role of long non-coding RNA gastric cancer high expressed transcript 1 (GHET1) in many cancers. This meta-analysis provides an evidence-based evaluation of the prognostic role of GHET1 in cancer. Materials and methods Literature searches were conducted in several databases including Medline, Cochrane, EMBASE, CNKI, and Wanfang. The pooled odds ratio (OR) and hazard ratio (HR) with 95% confidence interval (CI) were used to evaluate the role of GHET1 in cancer. The study protocol was registered at PROSPERO (ID: CRD42018111252). Results Sixteen studies, containing 1315 patients, were analyzed in this meta-analysis. The pooled results indicated that GHET1 overexpression was significantly associated with poor overall survival (OS) and disease-free survival (DFS) in cancer. Moreover, up-regulation of GHET1 expression predicted larger tumor size, positive lymph node metastasis, positive distant metastasis, and advanced TNM (tumor-node-metastases) stage in human cancers. Conclusion There is a significant correlation between up-regulation of GHET1 and both poor prognosis and advanced clinicopathological cancer characteristics. GHET1 may be a potential prognostic predictor for human cancers.
Collapse
Affiliation(s)
- Dingding Wang
- 1Department of Pathology, Beijing Tongren Hospital, Capital Medical University, Beijing, 100730 China
| | - Hong Zhang
- 1Department of Pathology, Beijing Tongren Hospital, Capital Medical University, Beijing, 100730 China
| | - Xiaolian Fang
- 2Department of Otolaryngology, Head and Neck Surgery, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, No.56 Nanlishi Rd, Beijing, 100045 China
| | - Xue Zhang
- 1Department of Pathology, Beijing Tongren Hospital, Capital Medical University, Beijing, 100730 China
| | - Honggang Liu
- 1Department of Pathology, Beijing Tongren Hospital, Capital Medical University, Beijing, 100730 China
| |
Collapse
|
39
|
Xia X, Ruan Y, Li B, Yu Y, Kong X, Zhuang P, Wu H. The Long Non-coding RNA lnc-DMP1 Regulates Dmp1 Expression Through H3K27Ac Modification. Front Genet 2020; 11:233. [PMID: 32256524 PMCID: PMC7093497 DOI: 10.3389/fgene.2020.00233] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Accepted: 02/27/2020] [Indexed: 12/19/2022] Open
Abstract
Several long non-coding RNAs (lncRNAs) have been reported regulate the expression of neighbor protein-coding genes at post-transcriptional, transcriptional and epigenetic levels. Dmp1 (Dentin matrix protein 1), encoding a non-collagenous extracellular matrix protein, plays an important role in dentin and bone mineralization. However, the transcriptional regulation of lncRNA on Dmp1 has not been reported. In this study, we identified a novel lncRNA named lnc-DMP1, which is near the Dmp1 gene region and undergoes remarkable changes during mandible development. lnc-DMP1 is co-localized and significantly expressed correlation with Dmp1 in embryonic and postnatal mouse mandibles. In MC3T3-E1 cells, lnc-DMP1 positively regulates DMP1 expression and skeletal mineralization. Furthermore, lnc-DMP1 induces the promoter activity of Dmp1 by modulating H3K27Ac enrichment in the Dmp1 promoter. In conclusion, our results indicate that lnc-DMP1 is a novel lncRNA near the Dmp1 gene region and regulates Dmp1 expression by modulating the H3K27 acetylation level of Dmp1 promoter.
Collapse
Affiliation(s)
- Xin Xia
- Department of Stomatology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yi Ruan
- Department of Stomatology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Boya Li
- Department of Stomatology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yansong Yu
- Department of Stomatology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Xiangbo Kong
- Department of Stomatology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Peilin Zhuang
- Department of Stomatology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Hong Wu
- Department of Stomatology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
40
|
Talebi A, Masoodi M, Mirzaei A, Mehrad-Majd H, Azizpour M, Akbari A. Biological and clinical relevance of metastasis-associated long noncoding RNAs in esophageal squamous cell carcinoma: A systematic review. J Cell Physiol 2020; 235:848-868. [PMID: 31310341 DOI: 10.1002/jcp.29083] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2019] [Accepted: 06/21/2019] [Indexed: 12/12/2022]
Abstract
Esophageal squamous cell carcinoma (ESCC) is a foremost cancer-related death worldwide owing to rapid metastasis and poor prognosis. Metastasis, as the most important reason for death, is biologically a multifaceted process involving a range of cell signaling pathways. Long noncoding RNAs (lncRNAs), as transcriptional regulators, can regulate numerous genomic processes and cellular processes such as cell proliferation, migration, and invasion. LncRNAs have also been shown to involve in/regulate the cancer metastasis-related signaling pathways. Hence, they have increasingly been brought to international attention in molecular oncology research. A number of researchers have attempted to reveal the biological and clinical relevance of lncRNAs in ESCC tumourigenesis and metastasis. The aberrant expression of these molecules in ESCC has regularly been reported to involve in various cellular processes and clinical features, including diagnosis, prognosis, and therapeutic responses. Here, we especially consider the pathways in which lncRNAs act as metastasis-mediated effectors, mainly by interacting with epithelial-mesenchymal transition-associated factors. We review the biological roles of lncRNAs through involving in ESCC metastasis as well as the clinical significance of the metastasis-related lncRNAs in cancer diagnosis and prognosis.
Collapse
Affiliation(s)
- Atefeh Talebi
- Colorectal Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Mohsen Masoodi
- Colorectal Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Alireza Mirzaei
- Bone and Joint Reconstruction Research Center, Shafa Orthopedic Hospital, Iran University of Medical Sciences, Tehran, Iran
| | - Hassan Mehrad-Majd
- Cancer Molecular Pathology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mazaher Azizpour
- Department of Orthopedic Surgery, Aarhus University Hospital, Aarhus, Denmark
| | - Abolfazl Akbari
- Colorectal Research Center, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
41
|
The Value of lncRNA GHET1 as a Prognostic Factor for Survival of Chinese Cancer Outcome: A Meta-Analysis. DISEASE MARKERS 2019; 2019:5824190. [PMID: 31885739 PMCID: PMC6914916 DOI: 10.1155/2019/5824190] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Revised: 09/06/2019] [Accepted: 10/30/2019] [Indexed: 12/30/2022]
Abstract
Aim There is increasing evidence that high expression levels of the gastric carcinoma highly expressed transcript 1 (GHET1), a long noncoding RNA (lncRNA), are associated with cancer prognosis and may be used as a valuable biomarker for cancer patients. The purpose of this meta-analysis was to analyze existing data to reveal potential clinical applications of GHET1 for cancer prognosis and tumor progression. All of these studies included in this meta-analysis were collected through a variety of retrieval strategies; and the enrolled articles were qualified via the meta-analysis of enrolled studies in epidemiology (MOOSE) and the preferred reporting items for systematic reviews and meta-analyses (PRISMA) checklists. Materials and Methods The literature collection was performed by a comprehensive search through electronic databases for studies published on or before March 10, 2019. These included the Cochrane library, PubMed, Embase, Web of Science, Springer, Science Direct, and three Chinese databases: CNKI, Weipu, and Wanfang. Seven studies that met the specified criteria were analyzed in the present research. Results The combined results indicate that an elevated GHET1 expression level is significantly associated with poor overall survival (OS) (HR = 2.40, 95% CI: 1.87–3.08, p < 0.001) and tumor progression (III/IV vs. I/II: HR = 1.80, 95% CI: 1.48–2.18, p < 0.001) in multiple cancers. The elevated GHET1 expression was also associated with lymph node metastasis (LNM) (HR = 2.44, 95% CI: 1.86–3.20, p < 0.001) in Chinese cancer patients. Conclusions. The present findings indicate that an increased GHET1 expression level is associated with poor OS, tumor progression, and LNM in patients with multiple tumors and may serve as a useful prognostic biomarker in Chinese cancer patients.
Collapse
|
42
|
Liu D, Zhang H, Cong J, Cui M, Ma M, Zhang F, Sun H, Chen C. H3K27 acetylation‐induced lncRNA EIF3J‐AS1 improved proliferation and impeded apoptosis of colorectal cancer through miR‐3163/YAP1 axis. J Cell Biochem 2019; 121:1923-1933. [PMID: 31709617 DOI: 10.1002/jcb.29427] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2019] [Accepted: 10/08/2019] [Indexed: 01/03/2023]
Affiliation(s)
- Dingsheng Liu
- Department of Colorectal Surgery, Shengjing Hospital China Medical University Shenyang Liaoning China
| | - Hong Zhang
- Department of Colorectal Surgery, Shengjing Hospital China Medical University Shenyang Liaoning China
| | - Jinchun Cong
- Department of Colorectal Surgery, Shengjing Hospital China Medical University Shenyang Liaoning China
| | - Mingming Cui
- Department of Colorectal Surgery, Shengjing Hospital China Medical University Shenyang Liaoning China
| | - Mingxing Ma
- Department of Colorectal Surgery, Shengjing Hospital China Medical University Shenyang Liaoning China
| | - Fangyuan Zhang
- Department of Colorectal Surgery, Shengjing Hospital China Medical University Shenyang Liaoning China
| | - Huayi Sun
- Department of Colorectal Surgery, Shengjing Hospital China Medical University Shenyang Liaoning China
| | - Chunsheng Chen
- Department of Colorectal Surgery, Shengjing Hospital China Medical University Shenyang Liaoning China
| |
Collapse
|
43
|
Jiang Y, Zhang H, Ke J, Shen H, Ou H, Liu Y. Overexpression of LncRNA GHET1 predicts an unfavourable survival and clinical parameters of patients in various cancers. J Cell Mol Med 2019; 23:4891-4899. [PMID: 31251476 PMCID: PMC6653440 DOI: 10.1111/jcmm.14486] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Revised: 04/15/2019] [Accepted: 04/29/2019] [Indexed: 12/12/2022] Open
Abstract
Recently, increasing studies have reported that long non-coding RNA (lncRNA) gastric carcinoma highly expressed transcript 1 (GHET1) is highly expressed in variety of cancers and relevant to poor prognosis of cancer patients. Nevertheless, the results were inconsistent and the systematic analysis of lncRNA GHET1 in cancers has not been inspected. Thus, we aim to evaluate the relationship between lncRNA GHET1 expression and clinical outcomes in human cancers. We searched keywords in PubMed, Web of Science, Embase, Cochrane Library and ClinicalTrial.gov. Stata SE12.0 software was used in the quantitative meta-analysis. Pooled hazard ratio (HR) and odds ratio with 95% confidence interval (95% Cl) were calculated to evaluate the clinical significance of lncRNA GHET1. Twelve studies totalling 761 patients with cancers were included for analysis. The pooled results of this study indicated that high lncRNA GHET1 expression level was significantly associated with poor overall survival (OS, HR = 2.30, 95% CI: 1.75-3.02) in human cancers. The statistical significance was also detected in subgroup analysis stratified by analysis method, cancer type, sample size and follow-up time respectively. In addition, the elevated lncRNA GHET1 expression was also significantly related to more advanced clinical stage, earlier lymph node metastasis, earlier distant metastasis and bigger tumour size. LncRNA GHET1 may serve as a promising biomarker for prognosis in Asians with cancers.
Collapse
Affiliation(s)
- Yao‐Fei Jiang
- Hubei Cancer Clinical Study Centre & Hubei Key Laboratory of Tumor Biological BehaviorsZhongnan Hospital, Wuhan UniversityWuhanChina
- Department of Radiation and Medical OncologyZhongnan Hospital, Wuhan UniversityWuhanChina
| | - Hong‐Yan Zhang
- Hubei Cancer Clinical Study Centre & Hubei Key Laboratory of Tumor Biological BehaviorsZhongnan Hospital, Wuhan UniversityWuhanChina
- Department of Radiation and Medical OncologyZhongnan Hospital, Wuhan UniversityWuhanChina
| | - Jin Ke
- Department of Oral and Maxillofacial Surgery, School and Hospital of StomatologyWuhan UniversityWuhanChina
| | - Hui Shen
- Hubei Cancer Clinical Study Centre & Hubei Key Laboratory of Tumor Biological BehaviorsZhongnan Hospital, Wuhan UniversityWuhanChina
- Department of Radiation and Medical OncologyZhongnan Hospital, Wuhan UniversityWuhanChina
| | - Hai‐Bin Ou
- Hubei Cancer Clinical Study Centre & Hubei Key Laboratory of Tumor Biological BehaviorsZhongnan Hospital, Wuhan UniversityWuhanChina
- Department of Radiation and Medical OncologyZhongnan Hospital, Wuhan UniversityWuhanChina
| | - Yu Liu
- Hubei Cancer Clinical Study Centre & Hubei Key Laboratory of Tumor Biological BehaviorsZhongnan Hospital, Wuhan UniversityWuhanChina
- Department of Radiation and Medical OncologyZhongnan Hospital, Wuhan UniversityWuhanChina
| |
Collapse
|
44
|
GHET1 acts as a prognostic indicator and functions as an oncogenic lncRNA in cervical cancer. Biosci Rep 2019; 39:BSR20182506. [PMID: 30948501 PMCID: PMC6488859 DOI: 10.1042/bsr20182506] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2018] [Revised: 03/28/2019] [Accepted: 03/30/2019] [Indexed: 12/29/2022] Open
Abstract
Gastric carcinoma proliferation enhancing transcript 1 (GHET1) has been suggested to serve as a promising oncogenic lncRNA in various types of human cancer. However, the role of GHET1 remained unknown in cervical cancer. In our study, we found GHET1 expression was markedly elevated in cervical cancer tissue specimens and cell lines compared with adjacent normal cervical tissue specimens and human normal cervical cell line, respectively. Then, we found high expression of GHET1 is a useful biomarker to discriminate cervical cancer tissues from non-tumorous tissues, and associated with advanced clinical stage, lymph node metastasis, distant metastasis and poor histological grade in cervical cancer patients. The survival analysis showed high GHET1 expression was an independent unfavorable prognostic factor in cervical cancer patients. Knockdown of GHET1 expression markedly inhibits cervical cancer cell proliferation, migration, and invasion. The loss-of-function study indicated knockdown of GHET1 expression markedly inhibits cervical cancer cell proliferation, migration, and invasion. In conclusion, GHET1 acts as an oncogenic lncRNA in cervical cancer.
Collapse
|
45
|
Yang W, Shan Z, Zhou X, Peng L, Zhi C, Chai J, Liu H, Yang J, Zhang Z. Knockdown of lncRNA GHET1 inhibits osteosarcoma cells proliferation, invasion, migration and EMT in vitro and in vivo. Cancer Biomark 2019; 23:589-601. [PMID: 30475755 DOI: 10.3233/cbm-181863] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
OBJECTIVE Osteosarcoma is the most common primary malignant skeleton tumor that derives from mesenchymal cells. Emerging evidences have identified the vital role of long non-coding RNAs (lncRNAs) in the development of osteosarcoma. In this study, we aimed to investigate the role of lncRNA gastric carcinoma highly expressed transcript 1 (GHET1) in osteosarcoma progression. METHODS The expression levels of relevant genes in clinical samples and cell lines were determined by quantitative real-time PCR. Cell proliferation was determined by CCK8 and cell colony formation assays. Transwell assay was used to detect the invasion and migration of osteosarcoma cells. Cell apoptosis and cell cycle were detected by flow cytometry. Protein levels were detected by western blot. In vivo tumor growth was investigated in the xenograft nude mice model. To determine whether growth inhibition and apoptosis are responsible for antitumor activity of silencing GHET1, immunohistochemistry for proliferation and TUNEL assay was performed in xenograft tissues. In vivo lung metastasis was performed to detect the effect of GHET1 on cell metastasis ability. RESULTS Our results revealed that GHET1 was up-regulated in osteosarcoma tissues compared to normal tissues. GHET1 was also increased in osteosarcoma cell lines compared to normal osteoplastic cell line. The up-regulation of GHET1 was significantly associated with TNM stage, distant metastasis and lymph node metastasis in patients with osteosarcoma. In vitro studies showed that silencing GHET1 in MG-63 and U2OS cells inhibited cell proliferation, cell invasion and migration and epithelial-to-mesenchymal transition (EMT), promoted cell apoptotic rate, and also caused an increase in cell population at G0/G1 phase with a decrease in cell population at S phase. Overexpression of GHET1 promoted the proliferation, invasion and migration of osteosarcoma cells. Importantly, silencing GHET1 inhibited tumor growth and tumor metastasis in mice MG-63-xenograft model in association with changes of EMT-related genes, reduced expression of Ki-67 and promotion of apoptosis. CONCLUSION GHET1 was up-regulated in osteosarcoma tissues and cell lines, inhibited cell apoptosis, promoted cell proliferation, invasion and migration by affecting EMT in vitro, and was correlated with the tumor growth and metastasis in vivo. GHET1 may be a potential therapeutic target of osteosarcoma treatment.
Collapse
Affiliation(s)
- Wei Yang
- Department of General Surgery, Affiliated Tumor Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, Henan, China
| | - Zhiming Shan
- Clinical Laboratory, The Affiliated Children Hospital of Zhengzhou University, The Children's Hospital of Henan Province, Zhengzhou Children's Hospital, Zhengzhou, Henan, China
| | - Xinfang Zhou
- Department of General Surgery, Affiliated Tumor Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, Henan, China
| | - Liangqun Peng
- Department of General Surgery, Affiliated Tumor Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, Henan, China
| | - Chongyang Zhi
- Department of General Surgery, Affiliated Tumor Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, Henan, China
| | - Junhui Chai
- Department of General Surgery, Affiliated Tumor Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, Henan, China
| | - Hongxing Liu
- Department of General Surgery, Affiliated Tumor Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, Henan, China
| | - Junmei Yang
- Clinical Laboratory, The Affiliated Children Hospital of Zhengzhou University, The Children's Hospital of Henan Province, Zhengzhou Children's Hospital, Zhengzhou, Henan, China
| | - Zhandong Zhang
- Department of General Surgery, Affiliated Tumor Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, Henan, China
| |
Collapse
|
46
|
Chen F, Qi S, Zhang X, Wu J, yang X, Wang R. lncRNA PLAC2 activated by H3K27 acetylation promotes cell proliferation and invasion via the activation of Wnt/β‑catenin pathway in oral squamous cell carcinoma. Int J Oncol 2019; 54:1183-1194. [PMID: 30720068 PMCID: PMC6411352 DOI: 10.3892/ijo.2019.4707] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2018] [Accepted: 12/21/2018] [Indexed: 12/22/2022] Open
Abstract
As a new group of important effector molecules involved in multiple cancer types, including breast cancer, lung cancer and oral squamous cell carcinoma, long noncoding RNAs (lncRNAs) have attracted considerable attention recently. However, the underlying cause that induces the dysregulated lncRNAs in cancer remains poorly understood. In the present study, the regulatory model of the lncRNA placenta‑specific protein 2 (PLAC2) upregulation in oral squamous cell carcinoma (OSCC) was investigated and its biological functions in OSCC malignant progression was identified. A reverse transcription‑quantitative polymerase chain reaction assay identified that PLAC2 is upregulated in OSCC cell lines and primary tissue samples. Furthermore, bioinformatic analysis followed by chromatin immunoprecipitation verified an enriched histone H3 on lysine 27 (H3K27) acetylation (H3K27ac) at the promoter region of the PLAC2 gene. Knockdown of cAMP‑response element binding protein‑binding protein (CBP) significantly reduced the enrichment level of H3K27ac, and thereby induced a decreased expression of PLAC2. Functionally, overexpression of PLAC2 promotes OSCC cell proliferation, migration and invasion, whereas knockdown of PLAC2 exerted an opposite effect. Furthermore, the Wnt/β‑catenin signaling pathway was activated by PLAC2 and mediated the PLAC2‑induced malignant progress of OSCC. In conclusion, the present results indicated that lncRNA PLAC2 is transcriptionally activated by H3K27ac modification at the promoter region in OSCC, and promotes cell growth and metastasis via activating Wnt/β‑catenin signaling pathway. Therefore, PLAC2 may serve as a promising biomarker for OSCC prognosis and therapy.
Collapse
Affiliation(s)
- Fubo Chen
- Department of Stomatology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072
| | - Shengcai Qi
- Department of Stomatology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072
| | - Xu Zhang
- Department of Stomatology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072
| | - Jinjin Wu
- Department of Stomatology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072
| | - Xi yang
- Department of Oral and Maxillofacial Head and Neck Oncology, Ninth People's Hospital, Shanghai JiaoTong University School of Medicine
- Shanghai Key Laboratory of Stomatology and Shanghai Research Institute of Stomatology
- National Clinical Research Center of Stomatology, Shanghai 200011, P.R. China
| | - Raorao Wang
- Department of Stomatology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072
| |
Collapse
|
47
|
Li S, Liu J, Kong F, Wang Y, Li N, Zou Y. lncRNA GHET1 has effects in development of pre-eclampsia. J Cell Biochem 2019; 120:12647-12652. [PMID: 30861585 DOI: 10.1002/jcb.28531] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2019] [Revised: 02/01/2019] [Accepted: 02/01/2019] [Indexed: 02/01/2023]
Abstract
To explain long noncoding RNA (lncRNA) gastric carcinoma high expressed transcript 1 (GHET1) affects the mechanism in development of pre-eclampsia. The pathological changes of normal, mild, and severe pre-eclampsia were evaluated by hematoxylin and eosin staining and measured the lncRNA GHET1 expression in different tissues by reverse-transcription polymerase chain reaction. In the cell experiment, the BeWo cells were randomly divided into three groups: normal control (NC) group, model group, and lncRNA group. The JEG3 cells of the model and lncRNA groups were cultured in the hypoxia condition. The JEG3 cells invasion and migration abilities were measured by Tanswell and wound-healing assays. The relative protein expressions of different groups were evaluated by Western blot (WB) assay. Compared with normal puerperal, the lncRNA GHET1 gene expression of pre-eclampsia was significantly downregulated (P < 0.05, respectively). In the cell experiment, the invasion cell number and wound-healing rate of the model group were significantly suppressed compared with the NC group (P < 0.05, respectively). However, the invasion cell number and wound-healing rate of lncRNA group were enhanced by lncRNA GHET1 overexpression (P < 0.05, respectively). In WB assay, the E-cadherin, fibronectin, and vimentin proteins expression showed significant differences between the model and lncRNA groups (P < 0.05, respectively). lncRNA GHET1 overexpression had restored cell invasion and migration abilities reduced by hypoxia in pre-eclampsia.
Collapse
Affiliation(s)
- Shuhong Li
- Departments of Obstetrics and Gynecology, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, People's Republic of China
| | - Jie Liu
- Departments of Obstetrics and Gynecology, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, People's Republic of China
| | - Fanqiang Kong
- Departments of Emergency, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, People's Republic of China
| | - Yuanli Wang
- Departments of Obstetrics and Gynecology, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, People's Republic of China
| | - Ning Li
- Departments of Central Laboratory, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, People's Republic of China
| | - Yanfen Zou
- Departments of Obstetrics and Gynecology, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, People's Republic of China
| |
Collapse
|
48
|
Li J, Jiang X, Li Z, Huang L, Zhou Y, Liu Y, Cui Y. Long noncoding RNA GHET1 in human cancer. Clin Chim Acta 2018; 488:111-115. [PMID: 30399371 DOI: 10.1016/j.cca.2018.11.007] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2018] [Revised: 11/01/2018] [Accepted: 11/02/2018] [Indexed: 02/06/2023]
Abstract
LncRNAs are a group of noncoding RNAs that are >200 nucleotides in length. These RNAs have no significant protein-coding potential due to the lack of obvious open reading frames. To date, accumulating evidence has demonstrated that dysregulation of lncRNAs exhibits indispensable roles in the pathological processes of human cancers. These RNAs function as either oncogenes or tumor suppressor genes to regulate proliferation, migration and invasion of cancer cells. GHET1, a prominent oncogenic lncRNA, is highly expressed in diverse malignancies. Furthermore, GHET1 performs key functions in carcinogenesis and progression, suggesting that GHET1 is expected to be a prospective biomarker or therapeutic target for cancers. In this review, we provide a summary of the current evidence concerning the biological functions, underlying mechanisms and clinical significance of GHET1 during tumor development.
Collapse
Affiliation(s)
- Jinglin Li
- Department of Hepatopancreatobiliary Surgery, The Second Affiliated Hospital of Harbin Medical University, No.246 Xuefu Avenue, Harbin 150086, China
| | - Xingming Jiang
- Department of Hepatopancreatobiliary Surgery, The Second Affiliated Hospital of Harbin Medical University, No.246 Xuefu Avenue, Harbin 150086, China
| | - Zhenglong Li
- Department of Hepatopancreatobiliary Surgery, The Second Affiliated Hospital of Harbin Medical University, No.246 Xuefu Avenue, Harbin 150086, China
| | - Lining Huang
- Department of Hepatopancreatobiliary Surgery, The Second Affiliated Hospital of Harbin Medical University, No.246 Xuefu Avenue, Harbin 150086, China
| | - Yuanshi Zhou
- Department of Hepatopancreatobiliary Surgery, The Second Affiliated Hospital of Harbin Medical University, No.246 Xuefu Avenue, Harbin 150086, China
| | - Yueping Liu
- Department of Hepatopancreatobiliary Surgery, The Second Affiliated Hospital of Harbin Medical University, No.246 Xuefu Avenue, Harbin 150086, China
| | - Yunfu Cui
- Department of Hepatopancreatobiliary Surgery, The Second Affiliated Hospital of Harbin Medical University, No.246 Xuefu Avenue, Harbin 150086, China.
| |
Collapse
|
49
|
Yu X, Yuan Z, Yang Z, Chen D, Kim T, Cui Y, Luo Q, Liu Z, Yang Z, Fan X, Chen D, Wang L. The novel long noncoding RNA u50535 promotes colorectal cancer growth and metastasis by regulating CCL20. Cell Death Dis 2018; 9:751. [PMID: 29970882 PMCID: PMC6030363 DOI: 10.1038/s41419-018-0771-y] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2018] [Revised: 05/30/2018] [Accepted: 06/06/2018] [Indexed: 01/15/2023]
Abstract
Long noncoding RNAs (lncRNAs) have been emerging as master regulators of tumor growth and metastasis, but the functions and underlying mechanisms of lncRNAs in colorectal cancer (CRC) still need to be clarified. Here, we found a novel lncRNA u50535, which was greatly overexpressed in CRC tissues and was associated with poor prognosis in CRC patients. Function studies showed that u50535 was an oncogene in CRC both in vitro and in vivo. In mechanism, through RNA sequencing and rescue assay, we found that u50535 activates CCL20 signaling to promote cell proliferation and migration in CRC. Taken together, these findings suggest that u50535 can promote CRC growth and metastasis and may serve as a potential biomarker in CRC.
Collapse
Affiliation(s)
- Xihu Yu
- Guangdong Institute of Gastroenterology, Guangzhou, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Zixu Yuan
- Department of Gastrointestinal Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Zuli Yang
- Department of Gastrointestinal Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Daici Chen
- Guangdong Institute of Gastroenterology, Guangzhou, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Taewan Kim
- The Ohio State University Comprehensive Cancer Center, Columbus, OH, USA
| | - Yanmei Cui
- Guangdong Institute of Gastroenterology, Guangzhou, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Qianxin Luo
- Guangdong Institute of Gastroenterology, Guangzhou, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Zhihang Liu
- Guangdong Institute of Gastroenterology, Guangzhou, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Zihuan Yang
- Guangdong Institute of Gastroenterology, Guangzhou, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Xinjuan Fan
- Department of Pathology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - DianKe Chen
- Department of Medical Oncology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.
| | - Lei Wang
- Guangdong Institute of Gastroenterology, Guangzhou, China.
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.
- Department of Gastrointestinal Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.
| |
Collapse
|
50
|
Sheng N, Li Y, Qian R, Li Y. The clinical significance and biological function of lncRNA RGMB-AS1 in hepatocellular carcinoma. Biomed Pharmacother 2018; 98:577-584. [DOI: 10.1016/j.biopha.2017.12.067] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2017] [Revised: 12/02/2017] [Accepted: 12/15/2017] [Indexed: 02/09/2023] Open
|