1
|
Wang Q, Zhao C, Du Q, Cao Z, Pan J. Non-coding RNA in infantile hemangioma. Pediatr Res 2024; 96:1594-1602. [PMID: 38750296 DOI: 10.1038/s41390-024-03250-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Revised: 04/18/2024] [Accepted: 04/27/2024] [Indexed: 01/29/2025]
Abstract
Infantile hemangioma (IH) is the most common benign vascular tumor of infancy, but its pathogenesis has not been fully discovered. From the cellular perspective, CD133+ stem cells orchestrate the proliferation and development of IH. Regarding molecular mechanisms, hypoxia inducible factor-1α, renin-angiotensin system, and vascular endothelial growth factor are current study hotspots, while non-coding RNAs (ncRNAs) might be essential factors participating in this network. Therefore, this article reviewed published studies concerning the roles of ncRNAs in IH and listed noted miRNAs, lncRNAs, and circRNAs. Other ncRNAs, such as snRNAs, snoRNAs, and tsRNAs, though have not been examined in IH, are mentioned as well to discuss their potential functions. Due to the continuous development of sequencing technologies and computational pipelines for ncRNAs annotation, relevant studies will provide evidence to gradually enhance acknowledgments of ncRNAs' role in IH. The pathogenesis of IH might be revealed and the treatment protocol would be optimized in the future. IMPACT: Non-coding RNAs (ncRNAs) play critical roles in infantile hemangioma. This article thoroughly reviewed all ncRNAs (miRNAs, lncRNAs, and circRNAs) mentioned in previous studies regarding the pathogenesis of infantile hemangioma. Other ncRNAs are promising subjects for further investigation. This review introduced the emerging ncRNAs that need to be explored in IH.
Collapse
Affiliation(s)
- Qizhang Wang
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Chengzhi Zhao
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Qianxin Du
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Zhiwei Cao
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Jian Pan
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, China.
| |
Collapse
|
2
|
Gao X, Chen L, Zuo H, Li Q. A novel hsa_circ_0006903 circular RNA promotes tumor development and dendritic cells activated expression in infantile hemangioma. Heliyon 2024; 10:e34186. [PMID: 39082028 PMCID: PMC11284419 DOI: 10.1016/j.heliyon.2024.e34186] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2023] [Revised: 07/04/2024] [Accepted: 07/04/2024] [Indexed: 08/02/2024] Open
Abstract
Background Increasing reports revealed that circular RNAs (circRNAs) and immune cells infiltration were related with tumor development. However, its role in infantile hemangioma (IH) is unknown. We will explore a novel hsa_circ_0006903-based ceRNA network and investigate the landscape of dendritic cells activated expression in IH. Material and methods Differentially expressed circRNAs (DECs) were identified from Gene Expression Omnibus (GEO) database. Regulatory networks and functional enrichment analysis were constructed. CIBERSORT was used to characterize immune cells composition. qRT-PCR was performed to detect the expression of hsa_circ_0006903 in cell lines. Then, the role of hsa_circ_0006903 in IH were validated in vitro using transwell assay. Immunofluorescence was applied to the colocalization of CD11b for dendritic cells activated as a biomarker in IH tissues. Results Using GEO database, a total of 67 DECs were screened out in IH. Hsa_circ_0006903 was the most significant DECs. Then, a novel hsa_circ_0006903 circular RNA-ceRNA network was constructed. Mechanistically, functional enrichment analysis showed that the p53 signaling pathway played the most important roles, and hsa_circ_0006903/miR-6721-5p/CACNA2D2 and hsa_circ_0006903/miR-4786-3p/ATP13A4 axis were identified. CACNA2D2, ATP13A4, and P53 were significantly downregulated in IH cell lines. We validated that dendritic cell activated was significantly overexpressed. Moreover, CD11b as a biomarker of dendritic cells activated were tested in IH tissues. Finally, hsa_circ_0006903 was significantly overexpressed, and hsa_circ_0006903 promoted infantile hemangioma cell proliferation, invasion, and migration in vitro. Conclusion Overall, our study revealed that a novel hsa_circ_0006903 promoted tumor progression, and indicated a potential biomarker CD11b of dendritic cells activated in IH.
Collapse
Affiliation(s)
- Xibo Gao
- Department of Dermatology, Tianjin Children’s Hospital, Tianjin, 300134, China
| | - Lixin Chen
- Department of Dermatology, Tianjin Children’s Hospital, Tianjin, 300134, China
| | - Hailiang Zuo
- Department of Orthopaedics, Tianjin Children’s Hospital, Tianjin, 300134, China
| | - Qinfeng Li
- Department of Dermatology, Tianjin Children’s Hospital, Tianjin, 300134, China
| |
Collapse
|
3
|
Zhao B, Li B, Zhang J, Qi Y, Chen B, Chen L. Construction of miRNA-mRNA Regulatory Network to Identify Potential Biomarkers in Infantile Hemangioma by Integrated Bioinformatics Analysis. Crit Rev Eukaryot Gene Expr 2023; 33:61-71. [PMID: 37199314 DOI: 10.1615/critreveukaryotgeneexpr.v33.i5.60] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/19/2023]
Abstract
Infantile hemangioma (IH) is the most common vascular tumor among infants and children. However, the understanding of pathogenesis about IH has not been fully elucidated, and the potential diagnostic maker remains further explored. In this study, we aimed to find miRNAs as potential biomarkers of IH through bioinformatic analysis. The microarray datasets GSE69136, GSE100682 were downloaded from the GEO database. The co-expressed differential miRNAs were identified by analyzing these two datasets. The downstream common target genes were predicted by the ENCORI, Mirgene, miRWalk, and Targetscan databases. GO annotation and KEGG pathway enrichment analysis for target genes were performed. The STRING database and Cytoscape software were used to construct the protein-protein interaction network and screen hub genes. Then potential diagnostic markers for IH were further screened and identified by using Receiver operating characteristic curve analysis. A total of thirteen co-expressed up-regulated miRNAs were screened out in the above two datasets, and 778 down-regulated target genes were then predicted. GO annotation and KEGG pathway enrichment analysis indicated that the common target genes strongly correlated with IH. Through the DEM-hub gene network construction, six miRNAs associated with the hub genes were identified. Finally, has-miR-522-3p, has-miR-512-3p, has-miR-520a-5p with high diagnostic values were screened out by receiver operating characteristic analysis. In the study, the potential miRNA-mRNA regulatory network was firstly constructed in IH. And, the three miRNAs might be used as potential biomarkers for IH, which also provided novel strategies for the therapeutic intervention of IH.
Collapse
Affiliation(s)
- Boming Zhao
- Division of Joint Surgery and Sports Medicine, Department of Orthopedic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, Hubei 430071, China
| | - Bin Li
- Division of Joint Surgery and Sports Medicine, Department of Orthopedic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, Hubei 430071, China
| | - Jun Zhang
- Division of Joint Surgery and Sports Medicine, Department of Orthopedic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, Hubei 430071, China
| | - Yongjian Qi
- Department of Spine Surgery and Musculoskeletal Tumor, Department of Orthopedic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, Hubei 430071, China
| | - Biao Chen
- Division of Joint Surgery and Sports Medicine, Department of Orthopedic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, Hubei 430071, China
| | - Liaobin Chen
- Division of Joint Surgery and Sports Medicine, Department of Orthopedic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, Hubei 430071, China
| |
Collapse
|
4
|
Li T, Tong H, Zhu J, Qin Z, Yin S, Sun Y, Liu X, He W. Identification of a Three-Glycolysis-Related lncRNA Signature Correlated With Prognosis and Metastasis in Clear Cell Renal Cell Carcinoma. Front Med (Lausanne) 2022; 8:777507. [PMID: 35083240 PMCID: PMC8785401 DOI: 10.3389/fmed.2021.777507] [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: 09/15/2021] [Accepted: 12/06/2021] [Indexed: 12/20/2022] Open
Abstract
The clear cell renal cell carcinoma (ccRCC) is not only a malignant disease but also an energy metabolic disease, we aimed to identify a novel prognostic model based on glycolysis-related long non-coding RNA (lncRNAs) and explore its mechanisms. With the use of Pearson correlation analysis between the glycolysis-related differentially expressed genes and lncRNAs from The Cancer Genome Atlas (TCGA) dataset, we identified three glycolysis-related lncRNAs and successfully constructed a prognostic model based on their expression. The diagnostic efficacy and the clinically predictive capacity of the signature were evaluated by univariate and multivariate Cox analyses, Kaplan–Meier survival analysis, and principal component analysis (PCA). The glycolysis-related lncRNA signature was constructed based on the expressions of AC009084.1, AC156455.1, and LINC00342. Patients were grouped into high- or low-risk groups according to risk score demonstrated significant differences in overall survival (OS) period, which were validated by patients with ccRCC from the International Cancer Genome Consortium (ICGC) database. Univariate Cox analyses, multivariate Cox analyses, and constructed nomogram-confirmed risk score based on our signature were independent prognosis predictors. The CIBERSORT algorithms demonstrated significant correlations between three-glycolysis-related lncRNAs and the tumor microenvironment (TME) components. Functional enrichment analysis demonstrated potential pathways and processes correlated with the risk model. Clinical samples validated expression levels of three-glycolysis-related lncRNAs, and LINC00342 demonstrated the most significant aberrant expression. in vitro, the general overexpression of LINC00342 was detected in ccRCC cells. After silencing LINC00342, the aberrant glycolytic levels and migration abilities in 786-O cells were decreased significantly, which might be explained by suppressed Wnt/β-catenin signaling pathway and reversed Epithelial mesenchymal transformation (EMT) process. Collectively, our research identified a novel three-glycolysis-related lncRNA signature as a promising model for generating accurate prognoses for patients with ccRCC, and silencing lncRNA LINC00342 from the signature could partly inhibit the glycolysis level and migration of ccRCC cells.
Collapse
Affiliation(s)
- Tinghao Li
- Department of Urology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Hang Tong
- Department of Urology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Junlong Zhu
- Department of Urology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Zijia Qin
- Department of Urology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Siwen Yin
- Department of Urology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yan Sun
- Department of Urology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xudong Liu
- Department of Urology, Bishan Hospital of Traditional Chinese Medicine, Chongqing, China
| | - Weiyang He
- Department of Urology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
5
|
Liu R, Yang X. LncRNA LINC00342 promotes gastric cancer progression by targeting the miR-545-5p/CNPY2 axis. BMC Cancer 2021; 21:1163. [PMID: 34715819 PMCID: PMC8556989 DOI: 10.1186/s12885-021-08829-x] [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: 06/23/2021] [Accepted: 09/30/2021] [Indexed: 12/29/2022] Open
Abstract
Background This study aimed to explore the role and underlying molecular mechanisms of long non-coding RNA (lncRNA) LINC00342 in gastric cancer (GC). Methods The expression of LINC00342 in GC tissues was evaluated by Quantitative reverse transcription polymerase chain reaction (qRT-PCR). Silencing of LINC00342 was conducted to investigate the effect of LINC00342 in vitro and in vivo. The underlying molecular mechanisms of LINC00342 were determined by dual luciferase reporter assay, Western blotting analysis and rescue experiments. Biological functions of LINC00342 were evaluated by cell counting kit-8 (CCK-8) assay, colony formation assay, wound healing assay and Transwell assays. In addition, a tumor model was used to verify the effect of LINC00342 in tumorigenesis in vivo. Results LINC00342 was significantly upregulated in GC tissues and cell lines. Silencing of LINC00342 efficiently inhibited proliferation, migration and invasion of AGS cells in vitro, and also suppressed the tumorigenesis of GC in vivo. Functional experiments showed that LINC00342 regulated the expression of canopy fibroblast growth factor signaling regulator 2 (CNPY2) by competitively sponging miR-545-5p. Rescue experiments showed that inhibition of miR-545-5p and overexpression of CNPY2 significantly reversed cell phenotypes caused by silencing of LINC00342. Conclusion LINC00342 plays a potential oncogenic role in GC by targeting the miR545-5p/CNPY2 axis, and might act as a novel therapeutic target for GC.
Collapse
Affiliation(s)
- Run Liu
- Department of Gastroenterology, The Shijiazhuang People's Hospital, 365 Jianhuanan street, Yuhua District, Shijiazhuang, 050000, Hebei, China
| | - Xianwu Yang
- Department of Gastroenterology, The Shijiazhuang People's Hospital, 365 Jianhuanan street, Yuhua District, Shijiazhuang, 050000, Hebei, China.
| |
Collapse
|
6
|
Mei H, Xian H, Ke J. LncRNA-MCM3AP-AS1 Promotes the Progression of Infantile Hemangiomas by Increasing miR-138-5p/HIF-1α Axis-Regulated Glycolysis. Front Mol Biosci 2021; 8:753218. [PMID: 34660700 PMCID: PMC8511435 DOI: 10.3389/fmolb.2021.753218] [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: 08/04/2021] [Accepted: 09/13/2021] [Indexed: 01/02/2023] Open
Abstract
Infantile hemangioma (IH) is a common benign tumor of endothelial cells in infants. Most hemangiomas are self-limited, but a few may develop and lead to serious complications that affect the normal life of children. Therefore, finding an effective treatment strategy for IH is a pressing need. Recent studies have demonstrated that non-coding RNAs affect the progression of multiple tumors. This study aims to investigate the mechanism by which LncRNA-MCM3AP-AS1 promotes glycolysis in the pathogenesis of IH. We first documented that the expression of LncRNA MCM3AP-AS1 was significantly upregulated in IH. Furthermore, we demonstrated that MCM3AP-AS1 bound to miR-106b-3p which promotes glycolysis in IH. In addition, we found that inhibition of HIF-1α contributed to the transformation of glycolysis to normal aerobic oxidation, partially reversed the promoting effect on glycolysis by the up-regulation of LncRNA MCM3AP-AS1 in IH disease. More importantly, we demonstrated this phenomenon existed in IH patients. Taken together, we demonstrate that LncRNA-MCM3AP-AS1 promotes the progression of infantile hemangiomas by increasing the glycolysis via regulating miR-138-5p/HIF-1α axis.
Collapse
Affiliation(s)
- Haijun Mei
- Department of General Surgery, Affiliated Hospital of Nantong University, Nantong, China
| | - Hua Xian
- Department of General Surgery, Affiliated Hospital of Nantong University, Nantong, China
| | - Jing Ke
- Department of General Surgery, Affiliated Hospital of Nantong University, Nantong, China
| |
Collapse
|
7
|
Lu Y, Yuan R, Huang H, Chen Z, Qin X, Chen J, Jiang J, Zhuo Y. A Six Autophagy-Related LncRNA Signature Associated with Clinical Prognosis in Prostate Cancer. Genet Test Mol Biomarkers 2021. [DOI: 10.1089/gtmb.2021.0024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Affiliation(s)
- Yangbai Lu
- Department of Urology, The First Affiliated Hospital of Jinan University, Guangzhou, China
- Department of Urology, Zhongshan City People's Hospital Affiliated to Sun Yat‑sen University, Zhongshan, China
| | - Runqiang Yuan
- Department of Urology, Zhongshan City People's Hospital Affiliated to Sun Yat‑sen University, Zhongshan, China
| | - Hongxing Huang
- Department of Urology, Zhongshan City People's Hospital Affiliated to Sun Yat‑sen University, Zhongshan, China
| | - Zheng Chen
- Department of Urology, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Xiaoping Qin
- Department of Urology, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Jianfan Chen
- Department of Urology, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Junbin Jiang
- Department of Urology, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Yumin Zhuo
- Department of Urology, The First Affiliated Hospital of Jinan University, Guangzhou, China
| |
Collapse
|
8
|
Lv D, Tan L, Ma H, Zhang Y, Lin J, Yu Y, Chen Z, Zhang Q, Ding Q, Deng Z. WITHDRAWN: LINC00342 promotes thyroid carcinoma progression by targeting miR-384/CHMP5 pathway. Pathol Res Pract 2021; 223:153272. [PMID: 34044217 DOI: 10.1016/j.prp.2020.153272] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Revised: 10/22/2020] [Accepted: 10/25/2020] [Indexed: 11/25/2022]
Affiliation(s)
- Dan Lv
- Department of Pulmonology, the Affiliated Hospital of Medical School of Ningbo University, Ningbo, 315020, China
| | - Lin Tan
- Department of Gastrointestinal Surgery, Ningbo First Hospital, Ningbo, 315010, China
| | - Hongying Ma
- Department of Pulmonology, the Affiliated Hospital of Medical School of Ningbo University, Ningbo, 315020, China
| | - Yun Zhang
- Department of Pulmonology, the Affiliated Hospital of Medical School of Ningbo University, Ningbo, 315020, China
| | - Jun Lin
- Department of Neurology, Xiangshan County Red Cross Taiyou Hospital General Hospital of Medical and Health Group, Ningbo, 315731, China
| | - Yiming Yu
- Department of Pulmonology, the Affiliated Hospital of Medical School of Ningbo University, Ningbo, 315020, China
| | - Zhongbo Chen
- Department of Pulmonology, the Affiliated Hospital of Medical School of Ningbo University, Ningbo, 315020, China
| | - Qiaoli Zhang
- Department of Pulmonology, the Affiliated Hospital of Medical School of Ningbo University, Ningbo, 315020, China
| | - Qunli Ding
- Department of Pulmonology, the Affiliated Hospital of Medical School of Ningbo University, Ningbo, 315020, China.
| | - Zaichun Deng
- Department of Pulmonology, the Affiliated Hospital of Medical School of Ningbo University, Ningbo, 315020, China.
| |
Collapse
|
9
|
Wu M, Chen Y, Feng L, Dai H, Fang S, Xu J. MiR-206 promotes extracellular matrix accumulation and relieves infantile hemangioma through targeted inhibition of DNMT3A. Cell Cycle 2021; 20:978-992. [PMID: 33945391 PMCID: PMC8172163 DOI: 10.1080/15384101.2021.1919820] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Revised: 12/02/2020] [Accepted: 12/07/2020] [Indexed: 10/21/2022] Open
Abstract
MiR-206 is abnormally expressed in infant hemangioma endothelial cells (HemECs), but the mechanism is not clear. We explored the intervention of miR-206 in HemECs in relation to extracellular matrix (ECM) metabolism. We selected 48 cases of infantile hemangioma (IH) from volunteer organizations. After the isolated and extracted HemECs were interfered with overexpressed or silenced miR-206, the effects of miR-206 on the proliferation, migration and invasion of HemECs were examined through basic cell function experiments. The expression differences of miR-206, DNA Methyltransferase 3A (DNMT3A) and ECM-related genes were analyzed as needed by qRT-PCR or Western blot. TargetScan and dual-luciferase experiments were applied to predict and confirm the binding relationship between miR-206 and DNMT3A. The correlation between miR-206 and DNMT3A was analyzed in IH tissues by Pearson correlation coefficient, and further confirmed in HemECs by conducting rescue experiments. A nude mouse model of xenograft tumor was constructed to verify the results of in vitro experiments. MiR-206, which was downregulated in proliferative hemangioma, suppressed the malignant development of HemECs by regulating ECM-related genes. As the target gene of miR-206, DNMT3A was high-expressed in IH tissues and was negatively correlated with miR-206. Overexpressed DNMT3A counteracted the inhibitory effect of miR-206 mimic on HemECs and its regulatory effect on ECM. The results of in vivo experiments were consistent with those from cell experiments. Thus, miR-206 could promote ECM accumulation through targeted inhibition of DNMT3A, further inhibiting the malignant development of HemECs and relieving IH.
Collapse
Affiliation(s)
- Minliang Wu
- Department of Plastic Surgery, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Yong Chen
- Department of Plastic Surgery, School of Medicine, Jinling Hospital, Nanjing University, Nanjing, Jiangsu, China
| | - Ling Feng
- Department of Pharmacy, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Haiying Dai
- Department of Plastic Surgery, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Shuo Fang
- Department of Plastic Surgery, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Jianguo Xu
- Department of Plastic Surgery, Changhai Hospital, Naval Medical University, Shanghai, China
| |
Collapse
|
10
|
lncRNA HEIH accelerates cell proliferation and inhibits cell senescence by targeting miR-3619-5p/CTTNBP2 axis in ovarian cancer. ACTA ACUST UNITED AC 2021; 27:1302-1314. [PMID: 33110047 DOI: 10.1097/gme.0000000000001655] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
OBJECTIVE Epithelial ovarian cancer is the most lethal malignancy in gynecology. Numerous studies have confirmed that long noncoding RNAs (lncRNAs) are abnormally expressed in ovarian cancer and are closely associated with the cell proliferation and senescence in cancers. However, the role and underlying molecular mechanism of long noncoding RNA high expression in hepatocellular carcinoma (HEIH) in ovarian cancer remain unknown. METHODS Experiments including Real-time quantitative polymerase chain reaction, RNA immunoprecipitation, luciferase reporter, Fluorescence in situ hybridization, western blot, colony formation assays, β-galactosidase senescence assay, cell apoptosis, proliferation, invasion, and migration assays were applied to investigate the role of HEIH in ovarian cancer. The data were expressed as the mean ± standard deviation. Student t test was used to compare the data between two groups. The one-way analysis of variance was applied to compare the data among multiple groups with Tukey post hoc test. All experiments were repeated three times. P < 0.05 was considered statistically significant. RESULTS Herein, HEIH expression was found to be up-regulated in ovarian cancer tissues (n = 25; twofold higher than normal tissues, P < 0.05) and cell lines (sixfold higher than normal ovarian epithelial cell line on average, P < 0.05), and high HEIH expression predicted poor prognosis (survival rate is about 25% after 40 mo; P < 0.05). Moreover, we found that HEIH accelerated proliferation, migration, and invasion, whereas inhibited cell senescence in ovarian cancer (P < 0.05). In mechanism, HEIH was confirmed to serve as a sponge for miR-3619-5p, and miR-3619-5p counteracted HEIH-mediated regulation of ovarian cancer (P < 0.05). Besides, cortactin-binding protein 2 (CTTNBP2) was found to be the downstream target of miR-3619-5p. Rescue assays validated that CTTNBP2 up-regulation significantly reversed the inhibitory effects of HEIH knockdown on ovarian cancer progression (P < 0.05). Furthermore, we found that HEIH facilitated tumor growth in vivo by regulating CTTNBP2 expression (P < 0.05). CONCLUSIONS In conclusion, our research revealed that HEIH accelerated cell proliferation, migration and invasion, whereas inhibited cell senescence in ovarian cancer via targeting the miR-3619-5p/CTTNBP2 axis. These findings may be valuable for finding new therapeutic targets to improve ovarian cancer treatment.
Collapse
|
11
|
Shen P, Qu L, Wang J, Ding Q, Zhou C, Xie R, Wang H, Ji G. LncRNA LINC00342 contributes to the growth and metastasis of colorectal cancer via targeting miR-19a-3p/NPEPL1 axis. Cancer Cell Int 2021; 21:105. [PMID: 33588834 PMCID: PMC7885559 DOI: 10.1186/s12935-020-01705-x] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Accepted: 12/07/2020] [Indexed: 12/19/2022] Open
Abstract
Background Long intergenic non-protein coding RNA 00342 (LINC00342) has been identified as a novel oncogene. However, the functional role of LINC00342 in colorectal cancer (CRC) remains unclear. Methods The expression of LINC00342 is detected by real-time PCR (RT-PCR) analysis. Cell proliferation, migration and invasion and xenograft model are examined to analyze the biological functions of LINC00342 in vitro and in vivo using colony formation, would healing and transwell analyses. Dual-luciferase reporter and RNA immunoprecipitation (RIP) assays are used to identify the target interactions between LINC00342, miR-19a-3p and aminopeptidase like 1 (NPEPL1). Results LINC00342 was highly expressed in CRC. Down-regulation of LINC00342 inhibited cell proliferation and metastasis of CRC cells. Moreover, knocking down LINC00342 inhibited the tumor growth in vivo. Mechanistic investigation revealed that LINC00342 might sponge miR-19a-3p to regulate NPEPL1 expression. Further investigation indicated that the ontogenesis facilitated by LINC00342 was inhibited due to the depletion of NPEPL1. Conclusion LINC00342 promotes CRC progression by competitively binding miR-19a-3p with NPEPL1.
Collapse
Affiliation(s)
- Peng Shen
- The Second Affiliated Hospital of Nanjing Medical University, Nanjing, 210000, China
| | - Lili Qu
- The Second Affiliated Hospital of Nanjing Medical University, Nanjing, 210000, China
| | - Jingjing Wang
- The Second Affiliated Hospital of Nanjing Medical University, Nanjing, 210000, China
| | - Quchen Ding
- The Second Affiliated Hospital of Nanjing Medical University, Nanjing, 210000, China
| | - Chuanwen Zhou
- The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, Huai'an, 223300, China
| | - Rui Xie
- The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, Huai'an, 223300, China
| | - Honggang Wang
- The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, Huai'an, 223300, China
| | - Guozhong Ji
- The Second Affiliated Hospital of Nanjing Medical University, Nanjing, 210000, China.
| |
Collapse
|
12
|
Yang P, Li J, Peng C, Tan Y, Chen R, Peng W, Gu Q, Zhou J, Wang L, Tang J, Feng Y, Sun Y. TCONS_00012883 promotes proliferation and metastasis via DDX3/YY1/MMP1/PI3K-AKT axis in colorectal cancer. Clin Transl Med 2020; 10:e211. [PMID: 33135346 PMCID: PMC7568852 DOI: 10.1002/ctm2.211] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Revised: 10/02/2020] [Accepted: 10/04/2020] [Indexed: 01/07/2023] Open
Abstract
Background Long noncoding RNAs (lncRNAs) have emerged as key regulators in multiple cancers, including colorectal cancer (CRC). However, the biological functions and molecular mechanisms underlying most lncRNAs in CRC remain largely unknown. Methods A novel lncRNA (TCONS_00012883) was identified using RNA sequencing. The level of TCONS_00012883 expression in CRC was analyzed by qRT‐PCR. The biological functions of TCONS_00012883 in CRC were investigated by a series of in vitro and in vivo experiments: CCK8, colony formation, EdU, flow cytometric assays, transwell assays, and mouse xenograft. The molecular mechanisms of TCONS_00012883 were demonstrated by RNA pulldown, mass spectrometry analysis, RIP, coimmunoprecipitation, RNA sequencing, chromatin immunoprecipitation, and rescue experiments. Results Elevated expression of TCONS_00012883 was confirmed in CRC and positively associated with a poor prognosis. Functionally, gain‐ and loss‐of‐function assays indicated that TCONS_00012883 promoted proliferation and metastasis of CRC cell lines in vitro and in vivo. Mechanistically, RNA pulldown and mass spectrometry analysis showed that DEAD‐box helicase 3 (DDX3) was the protein partner of TCONS_00012883. Furthermore, RNA sequencing assay revealed that matrix metallopeptidase 1 (MMP1) was the downstream of TCONS_00012883. Intriguingly, we found that transcription factor (YY1) could serve as a bridge between TCONS_00012883, DDX3, and MMP1. Conclusions TCONS_00012883 significantly promoted CRC progression via the DDX3/YY1/MMP1 axis, and thus, may act as a major role in diagnosis and therapy of CRC.
Collapse
Affiliation(s)
- Peng Yang
- The First School of Clinical Medicine, Nanjing Medical University, Nanjing, China.,Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Jie Li
- The First School of Clinical Medicine, Nanjing Medical University, Nanjing, China.,Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Chaofan Peng
- The First School of Clinical Medicine, Nanjing Medical University, Nanjing, China.,Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Yuqian Tan
- The First School of Clinical Medicine, Nanjing Medical University, Nanjing, China.,Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Ranran Chen
- The First School of Clinical Medicine, Nanjing Medical University, Nanjing, China.,Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Wen Peng
- The First School of Clinical Medicine, Nanjing Medical University, Nanjing, China.,Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Qiou Gu
- The First School of Clinical Medicine, Nanjing Medical University, Nanjing, China.,Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Jiahui Zhou
- The First School of Clinical Medicine, Nanjing Medical University, Nanjing, China.,Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Lu Wang
- The First School of Clinical Medicine, Nanjing Medical University, Nanjing, China.,Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Junwei Tang
- The First School of Clinical Medicine, Nanjing Medical University, Nanjing, China.,Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Yifei Feng
- The First School of Clinical Medicine, Nanjing Medical University, Nanjing, China.,Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Yueming Sun
- The First School of Clinical Medicine, Nanjing Medical University, Nanjing, China.,Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| |
Collapse
|
13
|
Bai Z, Li H, Li C, Sheng C, Zhao X. Integrated analysis identifies a long non-coding RNAs-messenger RNAs signature for prediction of prognosis in hepatitis B virus-hepatocellular carcinoma patients. Medicine (Baltimore) 2020; 99:e21503. [PMID: 33019382 PMCID: PMC7535691 DOI: 10.1097/md.0000000000021503] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Hepatitis B virus (HBV) infection is a leading cause of hepatocellular carcinoma (HCC), but HBV-HCC related prognosis signature remains rarely investigated. This study was to identify an integrated long non-coding RNAs-messenger RNAs (lncRNA-mRNA) signature for prediction of overall survival (OS) and explore their underlying functions.One RNA-sequencing dataset (training set, n = 95) and one microarray dataset E-TABM-36 (validation set, n = 44) were collected. Least absolute shrinkage and selection operator analysis was performed to identify an lncRNA-mRNA prognosis signature. The OS difference of patients in the high-risk and low-risk risk groups was evaluated by Kaplan-Meier curve. Area under the receiver operating characteristic curve (AUC), Harrell concordance index (C-index) calculation, and multivariate analyses with clinical characteristics were used to determine the prognostic ability. Furthermore, a coexpression network was constructed to interpret the functions.Nine signature genes (3 lncRNAs and 6 mRNAs) were selected to generate the risk score model. Patients belonging to the high-risk group showed a significantly shorter survival than those of the low-risk group. The prediction accuracy of the risk score for 5-year OS was 0.936 and 0.905 for the training set and validation set, respectively. Also, this risk score was independent of various clinical variables for the prognosis prediction. Incorporation of the risk score remarkably increased the predictive power of the routine clinical prognostic factors (vascular invasion status, tumor recurrence status) (AUC = 0.942 vs 0.628; C-index = 0.7997 vs 0.6908). Furthermore, LncRNA insulin-like growth factor 2 antisense RNA (IGF2-AS) and long intergenic non-protein coding RNA 342 (LINC00342) were predicted to exert tumor suppression effects by regulating homeobox D1 (HOXD1) and secreted frizzled related protein 5 (SFRP5), respectively; while lncRNA rhophilin Rho GTPase binding protein 1 antisense RNA 1 (RHPN1-AS1) may possess carcinogenic potential by promoting the transcription of chromobox 2 (CBX2), cell division cycle 20 (CDC20), matrix metallopeptidase 12 (MMP12), stratifin (SFN), tripartite motif containing 16 (TRIM16), and uroplakin 3A (UPK3A). These mRNAs may be associated with cell proliferation or apoptosis related pathways.This study may provide a novel, effective prognostic biomarker, and some therapeutic targets for HBV-HCC patients.
Collapse
|
14
|
Li Z, Jiang M, Zhang T, Liu S. GAS6-AS2 Promotes Hepatocellular Carcinoma via miR-3619-5p/ARL2 Axis Under Insufficient Radiofrequency Ablation Condition. Cancer Biother Radiopharm 2020; 36:879-887. [PMID: 32799553 DOI: 10.1089/cbr.2019.3541] [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: 12/30/2022] Open
Abstract
Background: Hepatocellular carcinoma (HCC) is a common malignancy worldwide. Radiofrequency ablation (RFA) is applied for treating HCC; however, insufficient RFA promotes HCC development and accelerates HCC recurrence. Therefore, the molecular functions underlying this process have gradually attracted attention. Aim of the study: We sought to examine whether GAS6-AS2 (also known as GAS6-DT: growth arrest specific 6 divergent transcript) played a role in the development of HCC after insufficient RFA. Methods: The in vitro model was established by heating Huh7 and MHCC97 cells in water bath at 47°C, named as Huh7-H and MHCC97-H. Colony formation, transwell and western blot assays were conducted for functional analysis. Results: GAS6-AS2 was upregulated in Huh7-H and MHCC97-H cells relative to Huh7 and MHCC97 cells. GAS6-AS2 deficiency hampered cell proliferation, migration, invasion, epithelial-mesenchymal transition, and stemness in Huh7-H and MHCC97-H cells. Moreover, microRNA-3619-5p (miR-3619-5p) combined with GAS6-AS2 and ARL2 (ADP ribosylation factor-like GTPase 2) was the target gene of miR-3619-5p. GAS6-AS2 served as the competing endogenous RNA (ceRNA) of ARL2 via absorbing miR-3619-5p. Conclusion: On the whole, present study uncovered a novel ceRNA mechanism of GAS6-AS2/miR-3619-5p/ARL2 in HCC after insufficient RFA, which might shed a new insight into treatment of HCC after insufficient RFA.
Collapse
Affiliation(s)
- Zilin Li
- Invasive Technology Department, Hubei Cancer Hospital, Wuhan, China
| | - Manhong Jiang
- Oncology Department, Hubei Provincial Hospital of Traditional Chinese Medicine, Wuhan, China
| | - Ting Zhang
- Chest Radiotherapy Department 1, Hubei Cancer Hospital, Wuhan, China
| | - Suhua Liu
- Anesthesiology Department, Hubei Rongjun Hospital, Wuhan, China
| |
Collapse
|
15
|
Yu L, Shu H, Xing L, Lv MX, Li L, Xie YC, Zhang Z, Zhang L, Xie YY. Silencing long non‑coding RNA NEAT1 suppresses the tumorigenesis of infantile hemangioma by competitively binding miR‑33a‑5p to stimulate HIF1α/NF‑κB pathway. Mol Med Rep 2020; 22:3358-3366. [PMID: 32945470 PMCID: PMC7453642 DOI: 10.3892/mmr.2020.11409] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Accepted: 05/20/2020] [Indexed: 02/07/2023] Open
Abstract
Infantile hemangioma (IH) is one of the most common vascular tumors that occurs during childhood, but its pathogenesis is currently not completely understood. Even though lncRNA nuclear paraspeckle assembly transcript 1 (NEAT1) plays vital roles in tumorigenesis of malignant tumors, its roles in IH remain unclear. Therefore, we evaluate the function of lncRNA NEAT1 in IH. Reverse transcription-quantitative PCR indicated that IH tissues exhibited high expression levels of NEAT1 and hypoxia-inducible factor 1α (HIF1α), and low expression levels of the microRNA (miR)-33a-5p. Small interfering RNA-mediated depletion of NEAT1 suppressed hemangioma endothelial cell (HemEC) proliferation, migration and invasion. The data suggested that NEAT1 positively regulated HIF1α expression by sponging miR-33a-5p in HemECs. miR-33a-5p overexpression or HIF1α silencing also acted to suppress HemEC proliferation, migration and invasion. Furthermore, the results indicated that the NEAT1/miR-33a-5p/HIF1α axis regulated the NF-κB signaling pathway. Collectively, the results revealed that depletion of lncRNA NEAT1 suppressed the tumorigenesis of IH by competitively binding miR-33a-5p and thereby stimulating the HIF1α/NF-κB signaling pathway.
Collapse
Affiliation(s)
- Li Yu
- Department of Dermatology, Kunming Children's Hospital, Kunming, Yunnan 650228, P.R. China
| | - Hong Shu
- Department of Dermatology, Kunming Children's Hospital, Kunming, Yunnan 650228, P.R. China
| | - Lu Xing
- Department of Dermatology, Kunming Children's Hospital, Kunming, Yunnan 650228, P.R. China
| | - Meng-Xing Lv
- Department of Pathology, Kunming Children's Hospital, Kunming, Yunnan 650228, P.R. China
| | - Li Li
- Department of Institute Pediatrics, Kunming Children's Hospital, Kunming, Yunnan 650228, P.R. China
| | - Yu-Cheng Xie
- Department of Pathology, Kunming Children's Hospital, Kunming, Yunnan 650228, P.R. China
| | - Zhao Zhang
- Department of Dermatology, Kunming Children's Hospital, Kunming, Yunnan 650228, P.R. China
| | - Li Zhang
- Department of Dermatology, Kunming Children's Hospital, Kunming, Yunnan 650228, P.R. China
| | - Yu-Yan Xie
- Department of Dermatology, Kunming Children's Hospital, Kunming, Yunnan 650228, P.R. China
| |
Collapse
|
16
|
Zhou Q, Guo J, Huang W, Yu X, Xu C, Long X. Linc-ROR promotes the progression of breast cancer and decreases the sensitivity to rapamycin through miR-194-3p targeting MECP2. Mol Oncol 2020; 14:2231-2250. [PMID: 32335998 PMCID: PMC7463371 DOI: 10.1002/1878-0261.12700] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2020] [Revised: 03/16/2020] [Accepted: 04/22/2020] [Indexed: 12/13/2022] Open
Abstract
linc‐ROR is reported to be a potential biomarker of breast cancer, but the detailed mechanism of linc‐ROR‐mediated breast cancer regulation has not been fully studied. We aimed to explore how linc‐ROR affects proliferation, metastasis, and drug sensitivity in breast cancer. Cell lines in which linc‐ROR was overexpressed or knocked down were constructed, and the cell proliferation, colony formation, cell migration, and invasion abilities of these lines were explored. A CCK‐8 assay was performed to determine the sensitivity of the breast cancer cells to rapamycin. Next‐generation sequencing was conducted to explore the detailed regulatory mechanism of linc‐ROR; differentially expressed RNAs in the linc‐ROR‐overexpressing cell line compared with the negative control were screened out, and their target genes were chosen to perform Gene Ontology analysis, Kyoto Encyclopedia of Genes and Genomes analysis, protein–protein interaction network analysis, and competing endogenous RNA (ceRNA) network analysis. The ceRNA mechanism of linc‐ROR for miR‐194‐3p, which targets MECP2, was determined through dual‐luciferase reporter assay, RT–qPCR, western blot, and rescue experiments. Finally, we found that linc‐ROR was upregulated in breast tumor tissues. linc‐ROR promoted the cell proliferation, colony formation, cell migration, and invasion of breast cancer and decreased the sensitivity of breast cancer cells to rapamycin. The overexpression of linc‐ROR triggered changes in the whole transcriptome of breast cancer cells, and a total of 85 lncRNAs, 414 microRNAs, 490 mRNAs, and 92 circRNAs were differentially expressed in the linc‐ROR‐overexpressing cell line compared with the negative control. Through a series of bioinformatic analyses, the ‘linc‐ROR/miR‐194‐3p/MECP2’ ceRNA regulatory axis was confirmed to be involved in the linc‐ROR‐mediated progression and drug sensitivity of breast cancer. In conclusion, linc‐ROR serves as an onco‐lncRNA in breast cancer and promotes the survival of breast cancer cells during rapamycin treatment by functioning as a ceRNA sponge for miR‐194‐3p, which targets MECP2.
Collapse
Affiliation(s)
- Qian Zhou
- Department of Laboratory Medicine, Zhongnan Hospital of Wuhan University, China
| | - Juan Guo
- Department of Laboratory Medicine, Zhongnan Hospital of Wuhan University, China
| | - Wenjie Huang
- Department of Laboratory Medicine, Zhongnan Hospital of Wuhan University, China
| | - Xiaosi Yu
- Department of Laboratory Medicine, Zhongnan Hospital of Wuhan University, China
| | - Chen Xu
- Department of Laboratory Medicine, Zhongnan Hospital of Wuhan University, China
| | - Xinghua Long
- Department of Laboratory Medicine, Zhongnan Hospital of Wuhan University, China
| |
Collapse
|
17
|
Liu Y, Li J, Wang S, Song H, Yu T. STAT4-mediated down-regulation of miR-3619-5p facilitates stomach adenocarcinoma by modulating TBC1D10B. Cancer Biol Ther 2020; 21:656-664. [PMID: 32397798 DOI: 10.1080/15384047.2020.1754690] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
BACKGROUND MicroRNAs (miRNAs) as the subtype of non-coding RNAs are revealed to be crucial players in cellular activities. It has been reported that miR-3619-5p functions as a tumor inhibitor in several cancers. However, the connection between miR-3619-5p and stomach adenocarcinoma (STAD) remains to be discovered. AIM OF THE STUDY The purpose of the study is to figure out the role and molecular regulation mechanism of miR-3619-5p in STAD. METHODS The expression of miR-3619-5p was evaluated via qRT-PCR analysis. Gain-of-function experiments demonstrated the effects of miR-3619-5p on cellular functions. The upper-stream transcription factor STAT4 and downstream target gene TBC1D10B of miR-3619-5p were identified by bioinformatic analysis. The binding and interaction between the indicated molecules were verified by RNA pull-down and luciferase reporter assays. RESULTS The expression of miR-3619-5p was prominently down-regulated in STAD cells and tissues. MiR-3619-5p suppresses cell proliferation, migration, invasion and tumor growth in STAD. Further, STAT4 bound with miR-3619-5p promoter and inhibited its transcription. MiR-3619-5p was also recognized to modulate STAD progression through the regulation of downstream target gene TBC1D10B. CONCLUSION STAT4-mediated miR-3619-5p controls STAD carcinogenesis and progression through modulating TBC1D10B expression, which may provide a novel insight for researching the STAD-related molecular mechanism.
Collapse
Affiliation(s)
- Yinhua Liu
- Department of Pathology, Wannan Medical College First Affiliated Hospital, Yijishan Hospital , Wuhu, Anhui Province, China
| | - Jiaping Li
- Department of Cardiothoracic Surgery, Wannan Medical College First Affiliated Hospital, Yijishan Hospital , Wuhu, Anhui Province, China
| | - Sufeng Wang
- Department of Pathology, Wannan Medical College First Affiliated Hospital, Yijishan Hospital , Wuhu, Anhui Province, China
| | - Hong Song
- Department of Pathology, Wannan Medical College First Affiliated Hospital, Yijishan Hospital , Wuhu, Anhui Province, China
| | - Tao Yu
- Department of Neurosurgical Intensive Care Unit, Wannan Medical College First Affiliated Hospital, Yijishan Hospital , Wuhu, Anhui Province, China.,Research Center for Functional Maintenance and Reconstruction of Viscera, Wannan Medical College First Affiliated Hospital, Yijishan Hospital , Wuhu, Anhui Province, China
| |
Collapse
|
18
|
Miao Z, Liu S, Xiao X, Li D. LINC00342 regulates cell proliferation, apoptosis, migration and invasion in colon adenocarcinoma via miR-545-5p/MDM2 axis. Gene 2020; 743:144604. [PMID: 32213297 DOI: 10.1016/j.gene.2020.144604] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Revised: 03/11/2020] [Accepted: 03/16/2020] [Indexed: 01/28/2023]
Abstract
AIM Colon adenocarcinoma (COAD) is the third most common cancer in the world. We aimed to explore the functional mechanism of LINC00342 in COAD. METHODS The LINC00342 expressions in COAD tissues were detected via qRT-PCR and in situ hybridization analysis. Statistical analysis was performed to analyze the relationship between LINC00342 expression and COAD clinical features. Small interfering LINC00342 (siLINC00342)/siCtrl were synthesized and then transfected into COAD cells. Cell apoptosis and proliferation were respectively assessed by flow cytometry and cell counting kit-8 assay. Cell migration and invasion were both measured using transwell assay. The target miRNA of LINC00342 were predicted and verified by online bioinformatics tools and luciferase reporter assay and RNA pull-down assay. Mice COAD models were constructed to explore the effects of LINC00342 on COAD in vivo. RESULTS LINC00342 was over-expressed in COAD tissues and LINC00342 overexpression was related to the poor prognosis of COAD patients. LINC00342 knockdown inhibited cell proliferation, migration and invasion and promoted apoptosis of COAD cells. LINC00342 targeted to miR-545-5p/murine double minute 2 (MDM2) in COAD. In COAD tissues and cell lines, LINC00342 expression was positively correlated to MDM2 expression, while it was negatively correlated to miR-545-5p expression. Both miR-545-5p-mimic and siMDM2 transfection could recover the changes of cell biological activities which were induced by LINC00342 overexpression. LINC00342 knockdown suppressed COAD tumor growth in vivo. CONCLUSION LINC00342 affected cell biological activities of COAD in vivo and in vitro via regulating miR-545-5p/MDM2. It might a novel target in COAD therapy.
Collapse
Affiliation(s)
- Zuohua Miao
- Department of Pathology, The First Affiliated Hospital of Gannan Medical University, Ganzhou 341000, China
| | - Suyun Liu
- Department of Critical Care Medicine, The First Affiliated Hospital of Nanchang University, Nanchang 330000, China
| | - Xuewen Xiao
- Department of Pathology, The First Affiliated Hospital of Gannan Medical University, Ganzhou 341000, China
| | - Dan Li
- Department of Pathology, The First Affiliated Hospital of Gannan Medical University, Ganzhou 341000, China.
| |
Collapse
|