1
|
Yao J, Ma F, Shi D, Da M. ZFP1 is a biomarker related to poor prognosis and immunity in gastric cancer. Sci Rep 2024; 14:21233. [PMID: 39261568 PMCID: PMC11390720 DOI: 10.1038/s41598-024-72387-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Accepted: 09/06/2024] [Indexed: 09/13/2024] Open
Abstract
We aimed to determine the prognostic significance of ZFP1 in gastric cancer (GC), its role in the immune microenvironment, and its potential as a therapeutic target using data from The Cancer Genome Atlas (TCGA) database. ZFP1 overexpression was closely associated with tumour T stage and histological grade. Patients with GC and high ZFP1 expression had poor outcomes. Lower ZFP1 expression was associated with longer symptom-free intervals and disease-specific survival. Subgroup analyses of T3 and T4, N0, N1, and M0 patients showed that overall survival (OS), disease-specific survival, and progression-free interval (PFI) were worse in those with high ZFP1 expression. ZFP1 expression in GC was moderately to strongly positively correlated with the infiltration levels of effector central memory T cells and T helper cells and negatively correlated with Th17 cells and NK CD56bright cells. The lncRNA-miRNA-ZFP1 axis was predicted using a public database. CCK8, colony formation, and wound healing assays were conducted to investigate whether ZFP1 promoted the proliferation and migration of GC cells. Our study suggests that ZFP1 plays a key role in the prognosis, immune response, and progression of GC and is a significant factor in the diagnosis and treatment of this disease.
Collapse
Affiliation(s)
- Jibin Yao
- The First School of Clinical Medicine, Lanzhou University, No.204 Donggang West Road, Lanzhou, 730000, People's Republic of China
- Department of Surgical Oncology, Gansu Province Hospital, Lanzhou, People's Republic of China
| | - Fubin Ma
- Department of Surgery, Ningxia Medical University, Yinchuan, People's Republic of China
| | - Donghai Shi
- The First School of Clinical Medicine, Gansu University of Chinese Medicine, Lanzhou, People's Republic of China
| | - Mingxu Da
- The First School of Clinical Medicine, Lanzhou University, No.204 Donggang West Road, Lanzhou, 730000, People's Republic of China.
- Department of Surgical Oncology, Gansu Province Hospital, Lanzhou, People's Republic of China.
| |
Collapse
|
2
|
Wang L, Zheng Z, Zheng J, Zhang G, Wang Z. The Potential Significance of the EMILIN3 Gene in Augmenting the Aggressiveness of Low-Grade Gliomas is Noteworthy. Cancer Manag Res 2024; 16:711-730. [PMID: 38952353 PMCID: PMC11215280 DOI: 10.2147/cmar.s463694] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2024] [Accepted: 06/20/2024] [Indexed: 07/03/2024] Open
Abstract
Purpose Low-grade gliomas (LGG) are common brain tumors with high mortality rates. Cancer cell invasion is a significant factor in tumor metastasis. Novel biomarkers are urgently needed to predict LGG prognosis effectively. Methods The data for LGG were obtained from the Bioinformatics database. A consensus clustering analysis was performed to identify molecular subtypes linked with invasion in LGG. Differential expression analysis was performed to identify differentially expressed genes (DEGs) between the identified clusters. Enrichment analyses were then conducted to explore the function for DEGs. Prognostic signatures were placed, and their predictive power was assessed. Furthermore, the invasion-related prognostic signature was validated using the CGGA dataset. Subsequently, clinical specimens were procured in order to validate the expression levels of the distinct genes examined in this research, and to further explore the impact of these genes on the glioma cell line LN229 and HS-683. Results Two invasion-related molecular subtypes of LGG were identified, and we sifted 163 DEGs between them. The enrichment analyses indicated that DEGs are mainly related to pattern specification process. Subsequently, 10 signature genes (IGF2BP2, SRY, CHI3L1, IGF2BP3, MEOX2, ABCC3, HOXC4, OTP, METTL7B, and EMILIN3) were sifted out to construct a risk model. Besides, the survival (OS) in the high-risk group was lower. The performance of the risk model was verified. Furthermore, a highly reliable nomogram was generated. Cellular experiments revealed the ability to promote cell viability, value-addedness, migratory ability, invasive ability, and colony-forming ability of the glioma cell line LN229 and HS-683. The qRT-PCR analysis of clinical glioma samples showed that these 10 genes were expressed at higher levels in high-grade gliomas than in low-grade gliomas, suggesting that these genes are associated with poor prognosis of gliomas. Conclusion Our study sifted out ten invasion-related biomarkers of LGG, providing a reference for treatments and prognostic prediction in LGG.
Collapse
Affiliation(s)
- Li`ao Wang
- Clinical College of Neurology, Neurosurgery and Neurorehabilitation, Tianjin Medical University, Tianjin, 300203, People’s Republic of China
| | - Zhiming Zheng
- Department of Neurosurgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, People’s Republic of China
| | - Jia Zheng
- Department of Neurosurgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, People’s Republic of China
| | - Guifeng Zhang
- Department of Neurology, Liaocheng People’s Hospital, Liaocheng, 252004, People’s Republic of China
| | - Zheng Wang
- Department of Neurosurgery, Liaocheng Traditional Chinese Medicine Hospital, Liaocheng, 252000, People’s Republic of China
| |
Collapse
|
3
|
Shao H, Zhu J, Zhu Y, Liu L, Zhao S, Kang Q, Liu Y, Zou H. Identification of characteristic genes and construction of regulatory network in gallbladder carcinoma. BMC Med Genomics 2023; 16:240. [PMID: 37821907 PMCID: PMC10566037 DOI: 10.1186/s12920-023-01663-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Accepted: 09/17/2023] [Indexed: 10/13/2023] Open
Abstract
BACKGROUND Gallbladder carcinoma (GBC) is a highly malignant tumor with a poor overall prognosis. This study aimed to identify the characteristic microRNAs (miRNAs) of GBC and the competing endogenous RNA (ceRNA) regulatory mechanisms. METHODS The microarray data of GBC tissue samples and normal gallbladder (NGB) tissue samples from the Gene Expression Omnibus (GEO) database was downloaded. GBC-related differentially expressed miRNAs (DE-miRNAs) were identified by inter-group differential expression analysis and weighted gene co-expression network analysis (WGCNA). Machine learning algorithms were used to screen the characteristic miRNA based on the intersect between least absolute shrinkage and selection operator (LASSO) and Support vector machine-recursive feature elimination (SVM-RFE). Based on the differential expression analysis of GEO database, the ceRNA network of characteristic miRNA was predicted and constructed. The biological functions of the ceRNA network were revealed by carrying out the gene enrichment analysis was implemented. We further screened the key genes of ceRNA network and constructed a protein-protein interaction (PPI) network, and predicted and generated the transcription factors (TFs) network of signature miRNAs. The expression of characteristic miRNA in clinical samples was verified by quantitative real-time polymerase chain reaction (qRT-PCR). RESULTS A total of 131 GBC-related DE-miRNAs were obtained. The hsa-miR-4770 was defined as characteristic miRNA for GBC. The ceRNA network containing 211 mRNAs, one miRNA, two lncRNAs, and 48 circRNAs was created. Gene enrichment analysis suggested that the downstream genes were mainly involved in actin filament organization, cell-substrate adhesion, cell-matrix adhesion, reactive oxygen species metabolic process, glutamine metabolic process and extracellular matrix (ECM)-receptor interaction pathway. 10 key genes in the network were found to be most correlated with disease, and involved in cell cycle-related processes, p53, and extrinsic apoptotic signaling pathways. The qRT-PCR result demonstrated that hsa-miR-4770 is down-regulated in GBC, and the expression trend is consistent with the public database. CONCLUSIONS We identified hsa-miR-4770 as the characteristic miRNA for GBC. The ceRNA network of hsa-miR-4770 may play key roles in GBC. This study provided some basis for potential pathogenesis of GBC.
Collapse
Affiliation(s)
- Hanrui Shao
- Department of Hepatopancreatobiliary Surgery, The Second Affiliated Hospital of Kunming Medical University, 374 Dianmian Avenue, Wu Hua District, Kunming, 650106, Yunnan, P.R. China
| | - Jiahai Zhu
- Department of Hepatopancreatobiliary Surgery, The Second Affiliated Hospital of Kunming Medical University, 374 Dianmian Avenue, Wu Hua District, Kunming, 650106, Yunnan, P.R. China
| | - Ya Zhu
- Department of Hepatopancreatobiliary Surgery, The Second Affiliated Hospital of Kunming Medical University, 374 Dianmian Avenue, Wu Hua District, Kunming, 650106, Yunnan, P.R. China
| | - Lixin Liu
- Department of Hepatopancreatobiliary Surgery, The Second Affiliated Hospital of Kunming Medical University, 374 Dianmian Avenue, Wu Hua District, Kunming, 650106, Yunnan, P.R. China
| | - Songling Zhao
- Department of Hepatopancreatobiliary Surgery, The Second Affiliated Hospital of Kunming Medical University, 374 Dianmian Avenue, Wu Hua District, Kunming, 650106, Yunnan, P.R. China
| | - Qiang Kang
- Department of Hepatopancreatobiliary Surgery, The Second Affiliated Hospital of Kunming Medical University, 374 Dianmian Avenue, Wu Hua District, Kunming, 650106, Yunnan, P.R. China
| | - Yunxia Liu
- Experiment Teaching Center, Basic Medical School, Kunming Medical University, 1168 West Chunrong Road, Kunming, 650500, P.R. China.
| | - Hao Zou
- Department of Hepatopancreatobiliary Surgery, The Second Affiliated Hospital of Kunming Medical University, 374 Dianmian Avenue, Wu Hua District, Kunming, 650106, Yunnan, P.R. China.
| |
Collapse
|
4
|
Ayoufu A, Paierhati P, Qiao L, Zhang N, Abudukeremu M. RUSC1-AS1 promotes the malignant progression of breast cancer depending on the regulation of the miR-326/XRCC5 pathway. Thorac Cancer 2023; 14:2504-2514. [PMID: 37429610 PMCID: PMC10447167 DOI: 10.1111/1759-7714.15035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Revised: 06/24/2023] [Accepted: 06/26/2023] [Indexed: 07/12/2023] Open
Abstract
BACKGROUND Many long noncoding RNAs (lncRNAs) are the key regulators for cancer progression, including breast cancer (BC). RUSC1 antisense 1 (RUSC1-AS1) has been found to be highly expressed in BC, but its role and potential molecular mechanism in BC remain to be further elucidated. METHODS Quantitative reverse transcription-polymerase chain reaction (RT-PCR) was utilized to measure RUSC1-AS1, microRNA (miR)-326 and X-ray repair cross-complementing group 5 (XRCC5) expression. Cell proliferation, metastasis, cell cycle, apoptosis and angiogenesis were determined by cell counting kit-8, colony formation, transwell, flow cytometry and tube formation assays. Protein expression was detected by western blot analysis. The targeted relationship between miR-326 and RUSC1-AS1 or XRCC5 was validated using dual-luciferase reporter assay and RIP assay. Xenograft models were constructed to uncover the effect of RUSC1-AS1 on BC tumorigenesis. RESULTS RUSC1-AS1 was upregulated in BC, and its downregulation suppressed BC proliferation, metastasis, cell cycle, angiogenesis, and tumor growth. MiR-326 was confirmed to be sponged by RUSC1-AS1, and its inhibitor reversed the regulation of RUSC1-AS1 silencing on BC progression. XRCC5 could be targeted by miR-326. Overexpression of XRCC5 reversed the inhibitory impacts of miR-326 on BC progression. CONCLUSION RUSC1-AS1 could serve as a sponge of miR-326 to promote BC progression by targeting XRCC5, suggesting that RUSC1-AS1 might be a target for BC treatment.
Collapse
Affiliation(s)
- Aisikeer Ayoufu
- Department of Breast Surgery Ward TwoAffiliated Cancer Hospital of Xinjiang Medical UniversityUrumqiChina
| | - Puerkaiti Paierhati
- Department of Breast and Thyroid SurgeryAffiliated Cancer Hospital of Xinjiang Medical UniversityUrumqiChina
| | - Lei Qiao
- Department of Breast and Thyroid SurgeryAffiliated Cancer Hospital of Xinjiang Medical UniversityUrumqiChina
| | - Nan Zhang
- Department of Breast and Thyroid SurgeryAffiliated Cancer Hospital of Xinjiang Medical UniversityUrumqiChina
| | - Muzhapaer Abudukeremu
- Department of Breast and Thyroid SurgeryAffiliated Cancer Hospital of Xinjiang Medical UniversityUrumqiChina
| |
Collapse
|
5
|
Sabbir Ahmed CM, Canchola A, Paul B, Alam MRN, Lin YH. Altered long non-coding RNAs expression in normal and diseased primary human airway epithelial cells exposed to diesel exhaust particles. Inhal Toxicol 2023; 35:157-168. [PMID: 36877189 PMCID: PMC10424575 DOI: 10.1080/08958378.2023.2185703] [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: 06/24/2022] [Accepted: 02/24/2023] [Indexed: 03/07/2023]
Abstract
BACKGROUND Exposure to diesel exhaust particles (DEP) has been linked to a variety of adverse health effects, including increased morbidity and mortality from cardiovascular diseases, chronic obstructive pulmonary disease (COPD), metabolic syndrome, and lung cancer. The epigenetic changes caused by air pollution have been associated with increased health risks. However, the exact molecular mechanisms underlying the lncRNA-mediated pathogenesis induced by DEP exposure have not been revealed. METHODS Through RNA-sequencing and integrative analysis of both mRNA and lncRNA profiles, this study investigated the role of lncRNAs in altered gene expression in healthy and diseased human primary epithelial cells (NHBE and DHBE-COPD) exposed to DEP at a dose of 30 μg/cm2. RESULTS We identified 503 and 563 differentially expressed (DE) mRNAs and a total of 10 and 14 DE lncRNAs in NHBE and DHBE-COPD cells exposed to DEP, respectively. In both NHBE and DHBE-COPD cells, enriched cancer-related pathways were identified at mRNA level, and 3 common lncRNAs OLMALINC, AC069234.2, and LINC00665 were found to be associated with cancer initiation and progression. In addition, we identified two cis-acting (TMEM51-AS1 and TTN-AS1) and several trans-acting lncRNAs (e.g. LINC01278, SNHG29, AC006064.4, TMEM51-AS1) only differentially expressed in COPD cells, which could potentially play a role in carcinogenesis and determine their susceptibility to DEP exposure. CONCLUSIONS Overall, our work highlights the potential importance of lncRNAs in regulating DEP-induced gene expression changes associated with carcinogenesis, and individuals suffering from COPD are likely to be more vulnerable to these environmental triggers.
Collapse
Affiliation(s)
- C. M. Sabbir Ahmed
- Environmental Toxicology Graduate Program, University of California, Riverside, United States
| | - Alexa Canchola
- Environmental Toxicology Graduate Program, University of California, Riverside, United States
| | - Biplab Paul
- Gastrointestinal Unit, Massachusetts General Hospital, Boston, Massachusetts 02114, United States
- Harvard Medical School, Boston, Massachusetts 02115, United States
| | - Md Rubaiat Nurul Alam
- Environmental Toxicology Graduate Program, University of California, Riverside, United States
| | - Ying-Hsuan Lin
- Environmental Toxicology Graduate Program, University of California, Riverside, United States
- Department of Environmental Sciences, University of California, Riverside, United States
| |
Collapse
|
6
|
Ranjbar M, Heydarzadeh S, Shekari Khaniani M, Foruzandeh Z, Seif F, Pornour M, Rahmanpour D, Tarhriz V, Alivand M. Mutual interaction of lncRNAs and epigenetics: focusing on cancer. EGYPTIAN JOURNAL OF MEDICAL HUMAN GENETICS 2023. [DOI: 10.1186/s43042-023-00404-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/08/2023] Open
Abstract
AbstractLong noncoding RNAs are characterized as noncoding transcripts longer than 200 nucleotides in response to a variety of functions within the cells. They are involved in almost all cellular mechanisms so as epigenetics. Given that epigenetics is an important phenomenon, which participates in the biology of complex diseases, many valuable studies have been performed to demonstrate the control status of lncRNAs and epigenetics. DNA methylation and histone modifications as epigenetic mechanisms can regulate the expression of lncRNAs by affecting their coding genes. Reciprocally, the three-dimensional structure of lncRNAs could mechanistically control the activity of epigenetic-related enzymes. Dysregulation in the mutual interaction between epigenetics and lncRNAs is one of the hallmarks of cancer. These mechanisms are either directly or indirectly involved in various cancer properties such as proliferation, apoptosis, invasion, and metastasis. For instance, lncRNA HOTAIR plays a role in regulating the expression of many genes by interacting with epigenetic factors such as DNA methyltransferases and EZH2, and thus plays a role in the initiation and progression of various cancers. Conversely, the expression of this lncRNA is also controlled by epigenetic factors. Therefore, focusing on this reciprocated interaction can apply to cancer management and the identification of prognostic, diagnostic, and druggable targets. In the current review, we discuss the reciprocal relationship between lncRNAs and epigenetic mechanisms to promote or prevent cancer progression and find new potent biomarkers and targets for cancer diagnosis and therapy.
Collapse
|
7
|
JHDM1D-AS1-driven inhibition of miR-940 releases ARTN expression to induce breast carcinogenesis. Clin Transl Oncol 2023:10.1007/s12094-023-03102-y. [PMID: 36862282 DOI: 10.1007/s12094-023-03102-y] [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: 11/19/2022] [Accepted: 01/23/2023] [Indexed: 03/03/2023]
Abstract
INTRODUCTION As ceRNA network of long non-coding RNA (lncRNA)-microRNA (miR)-messenger RNAs (mRNA) can be predicted on the basis of bioinformatics tools, we are now one step closer to deeper understanding carcinogenic mechanisms. In this study, we clarified the mechanistic understanding of JHDM1D-AS1-miR-940-ARTN ceRNA network in the development of breast cancer (BC). MATERIALS AND METHODS The lncRNA-miRNA-mRNA interaction of interest was predicted by in silico analysis and identified by conducting RNA immunoprecipitation, RNA pull-down and luciferase assays. The expression patterns of JHDM1D-AS1, miR-940 and ARTN in BC cells were altered by lentivirus infection and plasmid transfection for functional assays on the biological properties of BC cells. Finally, the tumorigenic and metastatic abilities of BC cells were assessed in vivo. RESULTS JHDM1D-AS1 was highly expressed, while miR-940 was poorly expressed in BC tissues and cells. JHDM1D-AS1 could competitively bind to miR-940, whereby promoting the malignant behaviors of BC cells. Furthermore, ARTN was identified as a target gene of miR-940. Through targeting ARTN, miR-940 exerted a tumor-suppressive role. In vivo experiments further confirmed that JHDM1D-AS1 enhanced the tumorigenesis and metastasis through up-regulation of ARTN. CONCLUSIONS Taken together, our study demonstrated the involvement of ceRNA network JHDM1D-AS1-miR-940-ARTN in the progression of BC, which highlighted promising therapeutic targets for BC treatment.
Collapse
|
8
|
Lv Y, Wang Y, Zhang Z. Potentials of lncRNA-miRNA-mRNA networks as biomarkers for laryngeal squamous cell carcinoma. Hum Cell 2023; 36:76-97. [PMID: 36181662 DOI: 10.1007/s13577-022-00799-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2022] [Accepted: 09/26/2022] [Indexed: 01/11/2023]
Abstract
Chemoresistance, radioresistance, and facile spreading of laryngeal squamous cell carcinoma (LSCC) make the practically clinical treatment invalid. Such dismal outcome mainly originates from the lack of effective biomarkers which are highly desirable to understand the pathogenesis of LSCC, and strives to find promising novel biomarkers to improve early screening, effective treatment, and prognosis evaluation in LSCC. Recently, long non-coding RNAs (lncRNAs), a kind of non-coding RNAs longer than 200 nucleotides, can participate in the process of tumorigenesis and progression through many regulatory modalities, such as epigenetic transcriptional regulation and post-transcriptional regulation. Meanwhile, microRNAs (miRNAs, miRs), essentially involved in the post-transcriptional regulation of gene expression, are aberrantly expressed in cancer-related genomic regions or susceptible sites. An increasing number of studies have shown that lncRNAs are important regulators of miRNAs expression in LSCC, and that miRNAs can also target to regulate the expression of lncRNAs, and they can target to regulate downstream messenger RNAs (mRNAs) transcriptionally or post-transcriptionally, thereby affecting various physiopathological processes of LSCC. Complex cross-regulatory networks existing among lncRNAs, miRNAs, and mRNAs can regulate the tumorigenesis and development of LSCC. Such networks may become promising biomarkers and potential therapeutic targets in the research field of LSCC. In this review, we mainly summarize the latest research progress on the regulatory relationships among lncRNAs, miRNAs, and downstream mRNAs, and highlight the potential applications of lncRNA-miRNA-mRNA regulatory networks as biomarkers for the early diagnosis, epithelial-mesenchymal transition (EMT) process, chemoresistance, radioresistance, and prognosis of LSCC, aiming to provide important clues for understanding the pathogenesis of LSCC and developing new diagnostic and therapeutic strategies.
Collapse
Affiliation(s)
- Yan Lv
- The Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, China Three Gorges University, Yichang, 443002, China
| | - Yanhua Wang
- The Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, China Three Gorges University, Yichang, 443002, China. .,Department of Morphology, Medical College of China Three Gorges University, Life Science Building, No.8 Daxue Road, Yichang, 443002, China.
| | - Zhikai Zhang
- The Third-Grade Pharmacological Laboratory on Chinese Medicine Approved by State Administration of Traditional Chinese Medicine, China Three Gorges University, Yichang, 443002, China
| |
Collapse
|
9
|
Wan L, Gu D, Jin X. LncRNA NCK1-AS1 Promotes Malignant Cellular Phenotypes of Laryngeal Squamous Cell Carcinoma via miR-137/NCK1 Axis. Mol Biotechnol 2022; 64:888-901. [PMID: 35218517 DOI: 10.1007/s12033-022-00469-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Accepted: 02/12/2022] [Indexed: 01/20/2023]
Abstract
Increasing evidence demonstrates that many long noncoding RNAs (lncRNAs) are implicated with the development of laryngeal squamous cell carcinoma (LSCC). As shown by bioinformatics analysis, lncRNA non-catalytic region of tyrosine kinase adaptor protein 1-antisense 1 (NCK1-AS1) is upregulated in tissues of head and neck squamous cell carcinoma. The study aimed to explore the role and mechanism of NCK1-AS1 in LSCC. NCK1-AS1 expression in LSCC cells was evaluated by reverse transcription qPCR. The viability, proliferation, invasion, migration, and apoptosis of LSCC cells with indicated transfection were evaluated by CCK-8 assays, Ethynyl deoxyuridine incorporation assays, Transwell assays, wound healing assays, and TUNEL assays, respectively. Subcellular fractionation assays were performed to evaluate the cellular distribution of NCK1-AS1 and NCK1. NCK1 protein level in LSCC cells with indicated transfection was quantified by western blotting. The binding relation between miR-137 and NCK1-AS1 (or NCK1) were determined using RNA immunoprecipitation assays and luciferase reporter assays. NCK1-AS1 was highly expressed in LSCC cell lines. NCK1-AS1 depletion suppressed LSCC cell viability, proliferation, invasion, and migration while enhancing cell apoptosis. NCK1, an adjacent gene of NCK1-AS1, is also highly expressed in LSCC cells and was positively regulated by NCK1-AS1. Moreover, NCK1-AS1 interact with miR-137 to upregulate NCK1 expression. NCK1 was the downstream target of miR-137 and was negatively correlated to miR-137. In addition, overexpressed NCK1 reversed the suppressive impact of NCK1-AS1 depletion on malignant behaviors of LSCC cells. NCK1-AS1 contributes to LSCC cellular behaviors by upregulating NCK1 via interaction with miR-137.
Collapse
Affiliation(s)
- Lanlan Wan
- Department of Otolaryngology, The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, No. 6, Beijing West Road, Huaian, 223300, Jiangsu, China
| | - Dongsheng Gu
- Department of Otolaryngology, The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, No. 6, Beijing West Road, Huaian, 223300, Jiangsu, China
| | - Xin Jin
- Department of Otolaryngology, The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, No. 6, Beijing West Road, Huaian, 223300, Jiangsu, China.
| |
Collapse
|
10
|
Hu YZ, Hu ZL, Liao TY, Li Y, Pan YL. LncRNA SND1-IT1 facilitates TGF-β1-induced epithelial-to-mesenchymal transition via miR-124/COL4A1 axis in gastric cancer. Cell Death Dis 2022; 8:73. [PMID: 35184134 PMCID: PMC8858320 DOI: 10.1038/s41420-021-00793-6] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Revised: 11/16/2021] [Accepted: 12/01/2021] [Indexed: 01/16/2023]
Abstract
AbstractThe transformation of tumor cells from an epithelial to a mesenchymal-like phenotype, designated as epithelial-to-mesenchymal transition (EMT), represents a key hallmark of human cancer metastasis, including gastric cancer (GC). However, a large set of non-coding RNAs have been studied for their functions that initiate or inhibit this phenotypic switch in GC cells by regulating oncogenes or tumor suppressors. In this paper, we aimed to identify lncRNA SND1-IT1, miR-124, and COL4A1 gene in the context of GC with a specific focus on their effects on transforming growth factor β1 (TGF-β1)-induced EMT. The study included 52 paired samples of lesion tissues and adjacent lesion-free tissues surgically resected from patients diagnosed with GC. HGC-27 cells were stimulated with exogenous TGF-β1 (2 ng/mL). Expression of lncRNA SND1-IT1, miR-124, and COL4A1 was determined by RT-qPCR. CCK-8 assays, Transwell assays, immunoblotting analysis of EMT-specific markers, and tumor invasion markers were performed to evaluate cell viability, migration, and invasion of cultured HGC-27 cells. Luciferase activity assay was employed to examine miR-124 binding with lncRNA SND1-IT1 and COL4A1, respectively. LncRNA SND1-IT1 was upregulated in GC tissues and cells. TGF-β1-stimulated EMT and regulated lncRNA SND1-IT1, miR-124, and COL4A1 expressions in HGC-27 cells. LncRNA SND1-IT1 knockdown tempered HGC-27 cell viability, migration and invasion. LncRNA SND1-IT1 participated in TGF-β1-stimulated EMT in GC by sponging miR-124. MiR-124 attenuated TGF-β1-stimulated EMT in GC by targeting COL4A1. These results primarily demonstrated TGF-β1 can regulate cancer cell migration, invasion and stimulate EMT through the SND1-IT1/miR-124/COL4A1 axis in GC.
Collapse
|
11
|
Jiang W, Ou ZL, Zhu Q, Yao YB, Zai HY. LncRNA OIP5-AS1 aggravates the stemness of hepatoblastoma through recruiting PTBP1 to increase the stability of β-catenin. Pathol Res Pract 2022; 232:153829. [DOI: 10.1016/j.prp.2022.153829] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Revised: 02/16/2022] [Accepted: 02/24/2022] [Indexed: 12/12/2022]
|
12
|
Yin DF, Zhou XJ, Li N, Liu HJ, Yuan H. Long non-coding RNA SND1-IT1 accelerates cell proliferation, invasion and migration via regulating miR-132-3p/SMAD2 axis in retinoblastoma. Bioengineered 2022; 12:1189-1201. [PMID: 34969359 PMCID: PMC8806201 DOI: 10.1080/21655979.2021.1909962] [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] [Indexed: 01/02/2023] Open
Abstract
Long noncoding RNAs (lncRNAs) have been identified as prognostic biomarkers and functional regulators in human tumors. In our study, we aim to investigate the roles of lncRNA SND1-IT1 (SND1-IT1) in retinoblastoma (RB). We observed that SND1-IT1 was highly expressed in both RB specimens and cells, and associated with poorer prognosis of RB patients. Functional investigation revealed that downregulation of SND1-IT1 suppressed RB cell proliferation, migration and invasion in vitro and restrained RB tumorigenesis in vivo. MiR-132-3p was predicted to interact with SND1-IT1. RT-qPCR and dual-luciferase reporter assays verified the regulation of miR-132-3p by SND1-IT1 in RB cells. In addition, SND1-IT1 enhanced the expression of SMAD2 by sponging miR-132-3p. Rescue experiments revealed that knockdown of miR-132-3p reversed the inhibiting effects of miR-132-3p knockdown on RB cells. Overall, SND1-IT1 can promote the progression of RB cells through miR-132-3p/SMAD2 axis, suggesting that l SND1-IT1 might be a novel biomarker and potential target for RB.
Collapse
Affiliation(s)
- Dong-Fang Yin
- Medical Department of Ophthalmology, Chinese PLA General Hospital, Beijing, China
| | - Xue-Jun Zhou
- Medical Department of Otolaryngology, Head and Neck Surgery, Chinese PLA General Hospital, Beijing, China
| | - Na Li
- Medical Department of Ophthalmology, Chinese PLA General Hospital, Beijing, China
| | - Hui-Jie Liu
- Medical Department of Ophthalmology, Chinese PLA General Hospital, Beijing, China
| | - Hu Yuan
- Medical Department of Otolaryngology, Head and Neck Surgery, Chinese PLA General Hospital, Beijing, China
| |
Collapse
|
13
|
MiR-106b-5p Promotes Malignant Behaviors of Cervical Squamous Cell Carcinoma Cells by Targeting TIMP2. Reprod Sci 2021; 29:203-211. [PMID: 34767243 DOI: 10.1007/s43032-021-00788-9] [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: 01/06/2021] [Accepted: 10/30/2021] [Indexed: 10/19/2022]
Abstract
The objective of this study was to investigate modulatory mechanism of miR-106b-5p and tissue inhibitor of metalloproteinases 2 (TIMP2) on cervical squamous cell carcinoma cells. Differentially expressed genes in CSCC were analyzed via bioinformatics analysis. The targeting impact of miR-106b-5p on TIMP2 was validated through dual-luciferase assay and RNA immunoprecipitation assay. MiR-106b-5p level and TIMP2 mRNA level were assessed via qRT-PCR. TIMP2 protein level was measured via western blot. Malignant behaviors of CSCC cells were evaluated by functional experiments. The EMT and apoptosis-related proteins were determined via western blot. MiR-106b-5p was noticeably elevated in CSCC cells. Its downstream target was TIMP2. MiR-106b-5p and TIMP2 levels were inversely correlated. MiR-106b-5p overexpression fostered malignant phenotypes of CSCC cells, and vice versus. TIMP2 overexpression weakened the promotive impact of forced expression of miR-106b-5p on CSCC cell growth. EMT was facilitated by forced expression of miR-106b-5p. MiR-106b-5p regulates the progression of CSCC cells via targeting TIMP2, which may provide novel value for development of therapeutic targets for CSCC.
Collapse
|
14
|
Li R, Pang XF, Huang ZG, Yang LH, Peng ZG, Ma J, He RQ. Overexpression of UBE2C in esophageal squamous cell carcinoma tissues and molecular analysis. BMC Cancer 2021; 21:996. [PMID: 34488675 PMCID: PMC8422647 DOI: 10.1186/s12885-021-08634-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Accepted: 07/26/2021] [Indexed: 01/16/2023] Open
Abstract
BACKGROUND Esophageal cancer is a common malignant tumor and its 5-year survival rate is much lower than 30% due to its invasiveness and pronounced metastasis ability, as well as the difficulty in early diagnosis. This study aimed to elucidate the molecular mechanism of ubiquitin conjugating enzyme E2 C (UBE2C) in esophageal squamous cell carcinoma (ESCC). METHODS In this study, we conducted a comprehensive evaluation of the UBE2C expression in ESCC by collecting the protein and mRNA expression data (including in-house RNA-seq, in-hosue immunohistochemistry, TCGA-GTEx RNA-seq and tissue microarray) to calculate a combined standardized mean difference (SMD) and summary receiver operating characteristic curve (sROC). Kaplan-Meier (K-M) method was used for survival analysis. We also explored the mechanism of UBE2C in ESCC by combing the differentially expressed genes (DEGs) of ESCC, related-genes of UBE2C in ESCC and the putative miRNAs and lncRNAs which may regulate UBE2C. RESULTS UBE2C protein and mRNA were highly expressed in ESCC tissues (including 772 ESCC tissue samples and 1837 non-cancerous tissue control samples). The pooled SMD of UBE2C expression values was 1.98 (95% CI: 1.51-2.45, p < 0.001), and the the area under the curve (AUC) of the sROC was 0.93 (95% CI: 0.90-0.95). The results of survival analysis suggested that UBE2C is likely to play different roles in different stages of the ESCC. Pathway anaylsis showed that UBE2C mainly influenced the biological function of esophageal cancer by synergistic effects with CDK1, PTTG1 and SKP2. We also constructed a potential UBE2C-related ceRNA network for ESCC (HCP5/has-miR-139-5p/UBE2C). CONCLUSION UBE2C mRNA and protein level were highly expressed in ESCC and UBE2C was likely to play different roles in different stages of the ESCC.
Collapse
Affiliation(s)
- Rong Li
- Department of Medical Oncology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Xing-Feng Pang
- Department of Medical Oncology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Zhi-Guang Huang
- Department of Pathology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Li-Hua Yang
- Department of Medical Oncology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Zhi-Gang Peng
- Department of Medical Oncology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Jie Ma
- Department of Medical Oncology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Rong-Quan He
- Department of Medical Oncology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China.
| |
Collapse
|
15
|
Bi O, Anene CA, Nsengimana J, Shelton M, Roberts W, Newton-Bishop J, Boyne JR. SFPQ promotes an oncogenic transcriptomic state in melanoma. Oncogene 2021; 40:5192-5203. [PMID: 34218270 PMCID: PMC8376646 DOI: 10.1038/s41388-021-01912-4] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Accepted: 06/17/2021] [Indexed: 02/06/2023]
Abstract
The multifunctional protein, splicing factor, proline- and glutamine-rich (SFPQ) has been implicated in numerous cancers often due to interaction with coding and non-coding RNAs, however, its role in melanoma remains unclear. We report that knockdown of SFPQ expression in melanoma cells decelerates several cancer-associated cell phenotypes, including cell growth, migration, epithelial to mesenchymal transition, apoptosis, and glycolysis. RIP-seq analysis revealed that the SFPQ-RNA interactome is reprogrammed in melanoma cells and specifically enriched with key melanoma-associated coding and long non-coding transcripts, including SOX10, AMIGO2 and LINC00511 and in most cases SFPQ is required for the efficient expression of these genes. Functional analysis of two SFPQ-enriched lncRNA, LINC00511 and LINC01234, demonstrated that these genes independently contribute to the melanoma phenotype and a more detailed analysis of LINC00511 indicated that this occurs in part via modulation of the miR-625-5p/PKM2 axis. Importantly, analysis of a large clinical cohort revealed that elevated expression of SFPQ in primary melanoma tumours may have utility as a prognostic biomarker. Together, these data suggest that SFPQ is an important driver of melanoma, likely due to SFPQ-RNA interactions promoting the expression of numerous oncogenic transcripts.
Collapse
Affiliation(s)
- O Bi
- School of Applied Sciences, University of Huddersfield, Huddersfield, UK
| | - C A Anene
- Centre for Cancer Genomics and Computational Biology, Barts Cancer Institute, Queen Mary University of London, London, UK
| | - J Nsengimana
- Population Health Sciences Institute, Faculty of Medical Sciences, Newcastle University, Newcastle, UK
| | - M Shelton
- School of Applied Sciences, University of Huddersfield, Huddersfield, UK
| | - W Roberts
- School of Clinical and Applied Science, Leeds Beckett University, Leeds, UK
| | | | - J R Boyne
- School of Applied Sciences, University of Huddersfield, Huddersfield, UK.
| |
Collapse
|
16
|
Rothzerg E, Ho XD, Xu J, Wood D, Märtson A, Kõks S. Upregulation of 15 Antisense Long Non-Coding RNAs in Osteosarcoma. Genes (Basel) 2021; 12:genes12081132. [PMID: 34440306 PMCID: PMC8394133 DOI: 10.3390/genes12081132] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Revised: 07/20/2021] [Accepted: 07/22/2021] [Indexed: 12/14/2022] Open
Abstract
The human genome encodes thousands of natural antisense long noncoding RNAs (lncRNAs); they play the essential role in regulation of gene expression at multiple levels, including replication, transcription and translation. Dysregulation of antisense lncRNAs plays indispensable roles in numerous biological progress, such as tumour progression, metastasis and resistance to therapeutic agents. To date, there have been several studies analysing antisense lncRNAs expression profiles in cancer, but not enough to highlight the complexity of the disease. In this study, we investigated the expression patterns of antisense lncRNAs from osteosarcoma and healthy bone samples (24 tumour-16 bone samples) using RNA sequencing. We identified 15 antisense lncRNAs (RUSC1-AS1, TBX2-AS1, PTOV1-AS1, UBE2D3-AS1, ERCC8-AS1, ZMIZ1-AS1, RNF144A-AS1, RDH10-AS1, TRG-AS1, GSN-AS1, HMGA2-AS1, ZNF528-AS1, OTUD6B-AS1, COX10-AS1 and SLC16A1-AS1) that were upregulated in tumour samples compared to bone sample controls. Further, we performed real-time polymerase chain reaction (RT-qPCR) to validate the expressions of the antisense lncRNAs in 8 different osteosarcoma cell lines (SaOS-2, G-292, HOS, U2-OS, 143B, SJSA-1, MG-63, and MNNG/HOS) compared to hFOB (human osteoblast cell line). These differentially expressed IncRNAs can be considered biomarkers and potential therapeutic targets for osteosarcoma.
Collapse
Affiliation(s)
- Emel Rothzerg
- School of Biomedical Sciences, The University of Western Australia, Perth, WA 6009, Australia; (E.R.); (J.X.); (D.W.)
- Perron Institute for Neurological and Translational Science, QEII Medical Centre, Nedlands, WA 6009, Australia
| | - Xuan Dung Ho
- Department of Oncology, College of Medicine and Pharmacy, Hue University, Hue 53000, Vietnam;
| | - Jiake Xu
- School of Biomedical Sciences, The University of Western Australia, Perth, WA 6009, Australia; (E.R.); (J.X.); (D.W.)
| | - David Wood
- School of Biomedical Sciences, The University of Western Australia, Perth, WA 6009, Australia; (E.R.); (J.X.); (D.W.)
| | - Aare Märtson
- Department of Traumatology and Orthopaedics, University of Tartu, Tartu University Hospital, 50411 Tartu, Estonia;
| | - Sulev Kõks
- Perron Institute for Neurological and Translational Science, QEII Medical Centre, Nedlands, WA 6009, Australia
- Centre for Molecular Medicine and Innovative Therapeutics, Murdoch University, Murdoch, WA 6150, Australia
- Correspondence: ; Tel.: +61-(0)-8-6457-0313
| |
Collapse
|
17
|
Jiang R, Zhang Z, Zhong Z, Zhang C. Long-non-coding RNA RUSC1-AS1 accelerates osteosarcoma development by miR-101-3p-mediated Notch1 signalling pathway. J Bone Oncol 2021; 30:100382. [PMID: 34367901 PMCID: PMC8326430 DOI: 10.1016/j.jbo.2021.100382] [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: 12/30/2020] [Revised: 06/07/2021] [Accepted: 07/06/2021] [Indexed: 01/22/2023] Open
Abstract
Background Long non-coding RNA (lncRNA) RUSC1-AS1 has been found to modulate several cancers development. In this study, we explored the role of RUSC1-AS1 on osteosarcoma (OS) progression. Methods Quantitative Real-time PCR (qRT-PCR) was conducted to test the relative expression of RUSC1-AS1, Notch1 mRNA and miR-101-3p in OS tissues and adjacent normal tissues. Gain- or loss- of functional assays were carried out to determine the roles of RUSC1-AS1 and miR-101-3p in OS progression both in vitro and in vivo. The expression of E-cadherin, N-cadherin, Vimentin, Snail, Notch1, Ras and ERK was determined by Western blot. Furthermore, the relationships between RUSC1-AS1 and miR-101-3p, Notch1 and miR-101-3p were confirmed through RNA immunoprecipitation (RIP) and dual luciferase reporter gene assay. Results RUSC1-AS1 and Notch1 were up-regulated in OS cells and tissues. Down-regulating RUSC1-AS1 significantly attenuated the proliferative, epithelial-mesenchymal transition (EMT), growth, lung metastasis, migrative and invasive abilities of MG-63 and Saos-2 cells, and aggravated apoptosis, accompanied with down-regulated Notch1-Ras-ERK1/2 in those cells both in vitro and in vivo, while overexpression of RUSC1-AS1 exerted opposite effects. Overexpressing miR-101-3p in OS cells had similar effects as RUSC1-AS1 inhibition. In addition, RUSC1-AS1 functioned as a competing endogenous RNA (ceRNA) to competitively sponge miR-101-3p, thus upregulating Notch1 expression and mediating the malignant behaviors of OS cells. Conclusion RUSC1-AS1 is a novel oncogenic lncRNA in OS through the miR-101-3p-Notch1-Ras-ERK pathway, which might be a potential therapeutic target for OS.
Collapse
Affiliation(s)
- Rui Jiang
- Department of Orthopedics, China-Japan Union Hospital of Jilin University, Changchun 130033, Jilin, China
| | - Ziyan Zhang
- Department of Orthopedics, The Second Hospital of Jilin University, Changchun 130033, Jilin, China
| | - Zhiwei Zhong
- Department of Pain Medicine, China-Japan Union Hospital of Jilin University, Changchun 130033, Jilin, China
| | - Chao Zhang
- Department of Ophthalmology, The Second Hospital of Jilin University, ChangchCun 130033, Jilin, China
| |
Collapse
|
18
|
Huang Y, Gu M, Tang Y, Sun Z, Luo J, Li Z. Systematic review and meta-analysis of prognostic microRNA biomarkers for survival outcome in laryngeal squamous cell cancer. Cancer Cell Int 2021; 21:316. [PMID: 34158050 PMCID: PMC8220842 DOI: 10.1186/s12935-021-02021-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Accepted: 06/11/2021] [Indexed: 12/29/2022] Open
Abstract
Background Laryngeal carcinoma is a primary malignant tumor originating from the laryngeal mucosa, and its pathogenesis is not fully understood. It is a rare type of cancer that shows a downward trend in the 5-year survival rate. In clinical practice, dysregulated microRNAs are often observed in patients with laryngeal cancer. In recent years, an increasing number of studies have confirmed that the strong biomarker potential of microRNAs. We conducted a systematic review and meta-analysis to identify and highlight multiple microRNAs as biomarkers for disease prognosis in patients with laryngeal cancer. Methods We actively searched the systematic reviews in PubMed, Embase, Web of Science and The Cochrane Library to select the studies that met the proposed guidelines. A total of 5307 patients with laryngeal cancer were included in this study to evaluate the association between microRNAs expression levels and patient outcomes. For overall survival in the clinical stage, a hazard ratio (HR) and corresponding 95% confidence interval (CI) are calculated to assess the effect of survival. Results A total of 36 studies on microRNAs and laryngeal cancer recovery were included in this meta-analysis. The selected endpoints for these studies included overall survival (OS) and disease-free survival (DFS).The comorbidities of overexpression and underexpression of microRNAs were 1.13 (95% CI 1.06–1.20, P < 0.05) and 1.10 (95% CI 1.00–1.20, P < 0.05), respectively. Conclusion MiRNA-100, miRNA-155, miRNA-21, miRNA-34a, miRNA-195 and miR-let-7 are expected to be potential noninvasive and simple markers for laryngeal cancer.
Collapse
Affiliation(s)
- Yan Huang
- Department of Radiotherapy, The Affiliated Changzhou No.2 People's Hospital of Nanjing Medical University, Changzhou, China.,Department of Head and Neck Surgery, Graduate School of Dalian Medical University, Dalian, China
| | - Min Gu
- Department of Stomatology, Affiliated Third Hospital of Soochow University, The First People's Hospital of Changzhou City, Changzhou, China
| | - Yiting Tang
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Soochow University, Suzhou, China
| | - Zhiqiang Sun
- Department of Radiotherapy, The Affiliated Changzhou No.2 People's Hospital of Nanjing Medical University, Changzhou, China
| | - Judong Luo
- Department of Radiotherapy, The Affiliated Changzhou No.2 People's Hospital of Nanjing Medical University, Changzhou, China.
| | - Zhe Li
- Department of Breast Surgery, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China.
| |
Collapse
|
19
|
Tong CJ, Deng QC, Ou DJ, Long X, Liu H, Huang K. LncRNA RUSC1-AS1 promotes osteosarcoma progression through regulating the miR-340-5p and PI3K/AKT pathway. Aging (Albany NY) 2021; 13:20116-20130. [PMID: 34048366 PMCID: PMC8436931 DOI: 10.18632/aging.203047] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Accepted: 04/02/2021] [Indexed: 11/25/2022]
Abstract
Dysregulation of long noncoding RNA (lncRNA) is frequently involved in the progression and development of osteosarcoma. LncRNA RUSC1-AS1 is reported to be upregulated and acts as an oncogene in hepatocellular carcinoma, cervical cancer and breast cancer. However, its role in osteosarcoma has not been studied yet. In the present study, we investigated the role of RUSC1-AS1 in osteosarcoma both in vitro and in vivo. The results showed that the expression of RUSC1-AS1 was significantly upregulated in osteosarcoma cell line U2OS and HOS compared to that in human osteoblast cell line hFOB1.19. Similar results were found in human samples. Silencing RUSC1-AS1 by siRNA significantly inhibited U2OS and HOS cell proliferation and invasion, measured by CCK-8 and transwell assay. Besides, knockdown of RUSC1-AS1 increased cell apoptosis in osteosarcoma cell lines. In addition, RUSC1-AS1 promoted the epithelial-mesenchymal transition (EMT) process of osteosarcoma cells. In vivo experiments confirmed that RUSC1-AS1 knockdown had an inhibitory effect on osteosarcoma tumor growth. Mechanically, we showed that RUSC1-AS1 directly binds to and inhibits miR-340-5p and activates the PI3K/AKT signaling pathway. In conclusion, our study demonstrated that RUSC1-AS1 promoted osteosarcoma development both in vitro and in vivo through sponging to miR-340-5p and activating the PI3K/AKT signaling pathway. Therefore, RUSC1-AS1 becomes a potential therapeutic target for osteosarcoma.
Collapse
Affiliation(s)
- Chang-Jun Tong
- Department of Orthopedics, Huazhong University of Science and Technology Union Shenzhen Hospital, Shenzhen 518000, China
| | - Qing-Chun Deng
- Department of Gynecology, The Second Affiliated Hospital of Hainan Medical University, Haikou 570102, China
| | - Di-Jun Ou
- Department of Orthopedics, Huazhong University of Science and Technology Union Shenzhen Hospital, Shenzhen 518000, China
| | - Xia Long
- Department of Operating Room, Huazhong University of Science and Technology Union Shenzhen Hospital, Shenzhen 518000, China
| | - He Liu
- Department of Orthopedics, Huazhong University of Science and Technology Union Shenzhen Hospital, Shenzhen 518000, China
| | - Kang Huang
- Department of Orthopedics, Huazhong University of Science and Technology Union Shenzhen Hospital, Shenzhen 518000, China
| |
Collapse
|
20
|
Liu C, Tang L, Xu M, Lin Y, Shen J, Zhou L, Ho L, Lu J, Ai X. LncRNA RUSC1-AS1 contributes to the progression of hepatocellular carcinoma cells by modulating miR-340-5p/CREB1 axis. Am J Transl Res 2021; 13:1022-1036. [PMID: 33841637 PMCID: PMC8014390] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Accepted: 12/01/2020] [Indexed: 06/12/2023]
Abstract
BACKGROUND Recent studies have proven that there is a relationship between long non-coding RNAs (lncRNAs) and malignant tumor hepatocellular carcinoma (HCC). However, the function of RUSC1-AS1 and its relative regulators in HCC remains unknown. METHODS In vitro studies, CCK-8 assays, colony formation assays, transwell assays, and wound healing tests were carried out to evaluate the proliferation, migration, and invasion of HCC cells. The correlation between RUSC1-AS1 expression with tumor size or weight was studied in nude mice. Bioinformatics analysis, dual luciferase, quantitative Real-Time PCR (qRT-PCR), and Western blot analysis aimed to discover the relevance between miR-340-5p and RUSC1-AS1 or cAMP responsive element binding protein 1 (CREB1). RESULTS When compared with normal groups, RUSC1-AS1 expression in HCC tissues and HCC cell lines was higher. We also found that knockdown of RUSC1-AS1 inhibited HCC cell progression, including proliferation, migration, and invasion, and suppressed tumorigenesis in vivo. Further studies demonstrated that the expression of RUSC1-AS1 negatively correlated with miR-340-5p expression in HCC cells. In addition, miR-340-5p was identified as a direct target of RUSC1-AS1 and tightly associated with the prevention of tumor progression. Moreover, miR-340-5p bound directly to CREB1. CREB1 overexpression reversed the impact of miR-340-5p on HCC cells. Together, lncRNA RUSC1-AS1 plays a regulatory role in the PI3K/AKT signaling pathway in HCC cells. CONCLUSION We demonstrated that lncRNA RUSC1-AS1 influenced HCC cell progression by modulating its downstream target miR-340-5p/CREB1 axis via the PI3K/AKT signaling pathway, which may be a potential prognostic and therapeutic target for treating HCC.
Collapse
Affiliation(s)
- Chunjiang Liu
- Department of General Surgery, Shaoxing People’s Hospital (Shaoxing Hospital of Zhejiang University)Shaoxing, Zhejiang, China
| | - Liming Tang
- Department of General Surgery, Shaoxing People’s Hospital (Shaoxing Hospital of Zhejiang University)Shaoxing, Zhejiang, China
| | - Miaojun Xu
- Department of General Surgery, Shaoxing People’s Hospital (Shaoxing Hospital of Zhejiang University)Shaoxing, Zhejiang, China
| | - Yuting Lin
- Department of General Surgery, BenQ Medical Center, The Affiliated BenQ Hospital of Nanjing Medical UniversityNanjing, Jiangsu, China
| | - Jingfu Shen
- Department of General Surgery, BenQ Medical Center, The Affiliated BenQ Hospital of Nanjing Medical UniversityNanjing, Jiangsu, China
| | - Liang Zhou
- Department of General Surgery, BenQ Medical Center, The Affiliated BenQ Hospital of Nanjing Medical UniversityNanjing, Jiangsu, China
| | - Lichen Ho
- Department of General Surgery, BenQ Medical Center, The Affiliated BenQ Hospital of Nanjing Medical UniversityNanjing, Jiangsu, China
| | - Jinjing Lu
- Department of General Surgery, BenQ Medical Center, The Affiliated BenQ Hospital of Nanjing Medical UniversityNanjing, Jiangsu, China
| | - Xiaoming Ai
- Department of General Surgery, BenQ Medical Center, The Affiliated BenQ Hospital of Nanjing Medical UniversityNanjing, Jiangsu, China
| |
Collapse
|
21
|
Zou Y, Chen B. Long non-coding RNA HCP5 in cancer. Clin Chim Acta 2020; 512:33-39. [PMID: 33245911 DOI: 10.1016/j.cca.2020.11.015] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2020] [Revised: 11/09/2020] [Accepted: 11/11/2020] [Indexed: 12/13/2022]
Abstract
Cancer remains a major threat to human health worldwide. Long non-coding RNA (lncRNA) comprises a group of single-stranded RNA with lengths longer than 200 bp. LncRNAs are aberrantly expressed and play a variety of roles involving multiple cellular processes in cancer. Histocompatibility leukocyte antigen complex P5 (HCP5), initially reported in 1993, is an important lncRNA located between the MICA and MICB genes in MHC I region. HCP5 is involved many autoimmune diseases as well as malignancies. Abnormal HCP5 expression occurs in many types of cancer and its dysregulation appears closely associated with tumor progression. HCP5 is also involved in anti-tumor drug resistance as well. As such, HCP5 represents a promising biomarker and therapeutic target in cancer. In this review, we summarize recent researches and provide an overview of the role and mechanism of HCP5 in human cancer.
Collapse
Affiliation(s)
- Yuanzhang Zou
- Department of Urology, Affiliated Hospital of Jiangsu University, Jiangsu University, Zhenjiang, Jiangsu, China
| | - Binghai Chen
- Department of Urology, Affiliated Hospital of Jiangsu University, Jiangsu University, Zhenjiang, Jiangsu, China.
| |
Collapse
|
22
|
Yang T, Li S, Liu J, Yin D, Yang X, Tang Q, Wang S. Long non-coding RNA KRT16P2/miR-1294/EGFR axis regulates laryngeal squamous cell carcinoma cell aggressiveness. Am J Transl Res 2020; 12:2939-2955. [PMID: 32655821 PMCID: PMC7344088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Accepted: 05/18/2020] [Indexed: 06/11/2023]
Abstract
Laryngeal squamous cell carcinoma (LSCC) is one of the most commonly seen head and neck malignancies. Identifying potent markers and/or targets for early diagnosis and individualized therapies for LSCC remains a considerable challenge. The present study analyzed online data and identified lncRNA KRT16P2 as a significantly upregulated long non-coding RNA (lncRNA) in LSCC. KRT16P2 knockdown in LSCC cells inhibited cancer cell proliferation, invasion, and migration. Similar to KRT16P2, EGFR expression was also significantly upregulated in LSCC. KRT16P2 and EGFR were positively correlated in LSCC tissue samples. EGFR knockdown also dramatically inhibited LSCC cell proliferation and aggressiveness (invasion and migration). Through online data and online tools, miR-1294 was predicted to target KRT16P2 and EGFR 3'UTR simultaneously. KRT16P2 inhibited miR-1294 expression, and miR-1294 inhibited EGFR expression through direct binding. miR-1294 overexpression repressed LSCC cell proliferation and aggressiveness. The effects of KRT16P2 silence on the expression of EGFR, LSCC cell proliferation, invasion, and migration, the protein levels of ki-67, PCNA, and cleaved-Caspase 3, as well as the phosphorylation of AKT, were all significantly reversed by miR-1294 inhibition. In conclusion, we demonstrated a lncRNA KRT16P2/miR-1294/EGFR axis that regulates LSCC cell proliferation, invasion, and migration. The clinical application of this axis needs further in vivo and clinical investigation.
Collapse
Affiliation(s)
- Tao Yang
- Department of Otolaryngology Head and Neck Surgery, The Second Xiangya Hospital, Central South University Changsha 410011, Hunan, PR China
| | - Shisheng Li
- Department of Otolaryngology Head and Neck Surgery, The Second Xiangya Hospital, Central South University Changsha 410011, Hunan, PR China
| | - Jiajia Liu
- Department of Otolaryngology Head and Neck Surgery, The Second Xiangya Hospital, Central South University Changsha 410011, Hunan, PR China
| | - Danhui Yin
- Department of Otolaryngology Head and Neck Surgery, The Second Xiangya Hospital, Central South University Changsha 410011, Hunan, PR China
| | - Xinming Yang
- Department of Otolaryngology Head and Neck Surgery, The Second Xiangya Hospital, Central South University Changsha 410011, Hunan, PR China
| | - Qinglai Tang
- Department of Otolaryngology Head and Neck Surgery, The Second Xiangya Hospital, Central South University Changsha 410011, Hunan, PR China
| | - Shuhui Wang
- Department of Otolaryngology Head and Neck Surgery, The Second Xiangya Hospital, Central South University Changsha 410011, Hunan, PR China
| |
Collapse
|
23
|
Wang X, Liu L, Zhao W, Li Q, Wang G, Li H. LncRNA SNHG16 Promotes the Progression of Laryngeal Squamous Cell Carcinoma by Mediating miR-877-5p/FOXP4 Axis. Onco Targets Ther 2020; 13:4569-4579. [PMID: 32547087 PMCID: PMC7251222 DOI: 10.2147/ott.s250752] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Accepted: 04/26/2020] [Indexed: 12/24/2022] Open
Abstract
Objective Laryngeal cancer is a common malignant tumor in the ENT, of which laryngeal squamous cell carcinoma (LSCC) accounts for more than 90% of laryngeal cancer. The purpose of this study is to investigate the regulatory mechanism of lncRNA SNHG16 in LSCC. Materials and Methods Real-time quantitative reverse transcription polymerase chain reaction (RT-qPCR) was used to measure mRNA expression. Cell Counting Kit (CCK-8), Transwell and luciferase reporter assays, flow cytometric analysis and Western blot analysis were used to investigate the function of lncRNA SNHG16 in LSCC. Results SNHG16 expression was increased in LSCC tissues and cells. The abnormal expression of SNHG16 was associated with clinical stage and lymph node metastasis in LSCC patients. In addition, knockdown of SNHG16 restrained cell proliferation, migration and invasion in LSCC. More importantly, SNHG16 acted as a competitive endogenous RNA in LSCC and regulated FOXP4 expression by making miR-877-5p sponge. Further, SNHG16 promoted LSCC progression by interacting with miR-877-5p and FOXP4. Conclusion LncRNA SNHG16 promotes the progression of LSCC by sponging miR-877-5p and upregulating FOXP4.
Collapse
Affiliation(s)
- Xiaoli Wang
- Department of Clinical Laboratory, Jinan City People's Hospital, Jinan People's Hospital Affiliated to Shandong First Medical University, Jinan 271199, People's Republic of China
| | - Liming Liu
- Department of Otorhinolaryngology, Juye County Hospital of TCM, Heze 274900, People's Republic of China
| | - Wenfei Zhao
- Department of Comprehensive Oncology Therapy, Qingdao Central Hospital, Qingdao University, Qingdao 266043, People's Republic of China
| | - Qingyan Li
- Department of Clinical Laboratory, The People's Hospital of Zhangqiu Area, Jinan 250200, People's Republic of China
| | - Guangsheng Wang
- Department of Neurology, The People's Hospital of Zhangqiu Area, Jinan 250200, People's Republic of China
| | - Huahui Li
- Department of Clinical Laboratory, Qingdao Municipal Hospital, Qingdao 266071, People's Republic of China
| |
Collapse
|
24
|
Guo Q, Zhang Q, Lu L, Xu Y. Long noncoding RNA RUSC1-AS1 promotes tumorigenesis in cervical cancer by acting as a competing endogenous RNA of microRNA-744 and consequently increasing Bcl-2 expression. Cell Cycle 2020; 19:1222-1235. [PMID: 32264732 DOI: 10.1080/15384101.2020.1749468] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
The expression of a long noncoding RNA termed RUSC1-AS1 is dysregulated in breast cancer and laryngeal squamous cell carcinoma, and this dysregulation affects various tumor-associated biological processes. To our knowledge, the expression status and detailed roles of RUSC1-AS1 in cervical cancer as well as its regulatory mechanisms of action remain unknown. Therefore, the objectives of this study were to measure RUSC1-AS1 expression in cervical cancer, investigate the effects of RUSC1-AS1 on cervical cancer cells, and identify the mechanism underlying these effects. Herein, RUSC1-AS1 was found to be highly expressed in cervical cancer tissues and cell lines. High RUSC1-AS1 expression significantly correlated with the International Federation of Gynecology and Obstetrics (FIGO) stage, lymph node metastasis, and shorter overall survival among the patients with cervical cancer. Functional assays revealed that interference with RUSC1-AS1 expression suppressed cervical cancer cell proliferation, migration, and invasion in vitro; induced apoptosis in vitro; and impeded tumor growth in vivo. In addition, RUSC1-AS1 was demonstrated to act as a competing endogenous RNA of microRNA-744 (miR-744) and consequently increase B-cell lymphoma 2 (Bcl-2 or BCL2) expression levels in cervical cancer cells. Furthermore, either inhibition of miR-744 or restoration of Bcl-2 expression neutralized the effects of the RUSC1-AS1 silencing on the malignant characteristics of cervical cancer cells. Thus, RUSC1-AS1 promotes the aggressiveness of cervical cancer in vitro and in vivo by upregulating miR-744-Bcl-2 axis output. The RUSC1-AS1-miR-744-Bcl-2 pathway may be involved in cervical cancer pathogenesis and could serve as a novel target for anticancer therapies.
Collapse
Affiliation(s)
- Qizhen Guo
- Department of Gynaecology and Obstetrics, Gaomi People's Hospital, Gaomi, Shandong, P.R. China
| | - Qin Zhang
- Department of Neurosurgery, Gaomi People's Hospital, Gaomi, Shandong, P.R. China
| | - Lianwei Lu
- Department of Radiology, Weifang People's Hospital, Weifang, Shandong, P.R. China
| | - Yanping Xu
- Department of General Surgery, Gaomi People's Hospital, Gaomi, Shandong, P.R. China
| |
Collapse
|