scholarly journals Long non-coding RNA LINC01116 is activated by EGR1 and facilitates lung adenocarcinoma oncogenicity via targeting miR-744-5p/CDCA4 axis

2021 ◽  
Vol 21 (1) ◽  
Author(s):  
Ping Ren ◽  
Liang Chang ◽  
Xiaodong Hong ◽  
Lei Xing ◽  
Hong Zhang

Abstract Background Lung adenocarcinoma (LAD) is one of the most frequently diagnosed pathological categories of human lung cancer. Nevertheless, the link between long non-coding RNA (lncRNA) LINC01116 and LAD remains poorly investigated. Methods QRT-PCR and western blot were applied for quantifying the expression of RNAs and proteins. Both functional experiments assays in vitro and xenografts model in vivo were implemented for analyzing LINC01116 function in LAD while molecular relationship among RNAs was investigated via mechanism experiments. Results LINC01116 was expressed at an abnormally high level in LAD, which was induced by transcription activator EGR1. LINC01116 depletion restrained proliferation, migration and invasion, yet facilitated apoptosis of LAD cells. MiR-744-5p could bind to LINC01116. MiR-744-5p inhibitor reversed the inhibitory effects of silencing LINC01116 on LAD malignant behaviors. In addition, cell division cycle-associated protein 4 (CDCA4) shared binding sites with miR-744-5p. Silencing LINC01116 elicited decline in CDCA4 mRNA and protein levels. Moreover, CDCA4 up-regulation could counteract the biological effects of LINC01116 knockdown on LAD cells. Conclusion Our data revealed that LINC01116 promoted malignant behaviors of LAD cells by targeting miR-744-5p/CDCA4 axis, implying the theoretical potential of LINC01116 as a novel target for LAD treatment.

Pathobiology ◽  
2021 ◽  
pp. 1-12
Author(s):  
Ling Zhou ◽  
Xiao-li Xu

<b><i>Background:</i></b> Emerging research has demonstrated that long non-coding RNAs (lncRNAs) attach great importance to the progression of cervical cancer (CC). LncRNA ARAP1-AS1 was involved in the development of several cancers; however, its role in CC is far from being elucidated. <b><i>Methods:</i></b> Real-time PCR (RT-PCR) was employed to detect ARAP1-AS1 and miR-149-3p expression in CC samples. CC cell lines (HeLa and C33A cells) were regarded as the cell models. The biological effect of ARAP1-AS1 on cancer cells was measured using CCK-8 assay, colony formation assay, flow cytometry, Transwell assay and wound healing assay in vitro, and subcutaneous xenotransplanted tumor model and tail vein injection model in vivo. Furthermore, interactions between ARAP1-AS1 and miR-149-3p, miR-149-3p and POU class 2 homeobox 2 (POU2F2) were determined by bioinformatics analysis, qRT-PCR, Western blot, luciferase reporter and RNA immunoprecipitation assay, respectively. <b><i>Results:</i></b> The expression of ARAP1-AS1 was enhanced in CC samples, while miR-149-3p was markedly suppressed. Additionally, ARAP1-AS1 overexpression enhanced the viability, migration, and invasion of CC cells. ARAP1-AS1 downregulated miR-149-3p via sponging it. ARAP1-AS1 and miR-149-3p exhibited a negative correlation in CC samples. On the other hand, ARAP1-AS1 enhanced the expression of POU2F2, which was validated as a target gene of miR-149-3p. <b><i>Conclusion:</i></b> ARAP1-AS1 was abnormally upregulated in CC tissues and indirectly modulated the POU2F2 expression via reducing miR-149-3p expression. Our study identified a novel axis, ARAP1-AS1/miR-149-3p/POU2F2, in CC tumorigenesis.


2020 ◽  
Vol 40 (3) ◽  
Author(s):  
Ke Wu ◽  
Lei Li ◽  
Lin Li ◽  
Dong Wang

Abstract Objective: To investigate the specific function of long non-coding RNA HAL in serous ovarian cancer (SOC) and to further clarify the regulation of HAL on EMT pathway. Materials and methods: The expression of HAL and TWIST1 was detected by qRT-PCR. CCK8 assay, wound healing assay, transwell assay and flow cytometry were used to detect the HAL function on proliferation, migration, invasion and apoptosis in SOC cells. Western blot was used to calculate protein level of Vimentin, N-cadherin and E-cadherin. The effect of HAL on tumorigenesis of SOC was confirmed by xenograft nude mice model. Results: HAL was significantly decreased in SOC tissues and cells. Overexpression of HAL inhibited the proliferation, migration and invasion of SKOV3 cells, but promoted apoptosis. Furthermore, overexpression of HAL decreased the mRNA and protein levels of TWIST1 via a binding between HAL and TWIST1. Forced expression of TWIST1 reversed the inhibitory role of HAL on SOC cells’ migration and invasion. The in vivo tumor growth assay showed that HAL suppressed SOC tumorigenesis with inhibiting EMT pathway. Conclusions: Our research emphasized HAL acting as a tumor-inhibiting gene by regulating EMT signaling pathway, thus providing some novel experimental basis for clinical treatment of SOC.


2021 ◽  
Vol 11 ◽  
Author(s):  
Yuanyong Wang ◽  
Minge Ma ◽  
Chuan Li ◽  
Yuling Yang ◽  
Maolong Wang

GAS6 antisense RNA 1 (GAS6-AS1) is a long non-coding RNA involved in hepatocellular carcinoma and gastric cancer. However, the functional role of GAS6-AS1 in lung adenocarcinoma (LUAD) remains unclear. In the present study, qRT-PCR was used to measure the levels of GAS6-AS1, GIMAP6 and miR-24-3p expression in LUAD samples and cell lines. CCK-8 and colony formation assays were used to determine cell proliferation. Cell migration and invasion were evaluated using wound healing and transwell assays, respectively. The potential interactions between molecules were assessed using RNA immunoprecipitation and luciferase reporter assays. Western blot analysis was used to quantify protein expression. The anti-tumor effect of over-expressed GAS6-AS1 on LUAD was also examined in vivo in xenograft tumor experiments. The expression of GAS6-AS1 was notably downregulated in LUAD samples and cell lines and associated with a poor prognosis. GAS6-AS1 overexpression inhibited the migration and invasion of A549 and H1650 cells. Down-expressed GAS6-AS1 acted as a sponge for miR-24-3p and down-regulated the expression of its target, GTPase IMAP Family Member 6. These findings suggested that GAS6-AS1 might represent a potential diagnostic biomarker for LUAD.


2021 ◽  
Author(s):  
Xin Liu ◽  
Zhenghao Huang ◽  
Honglei Qin ◽  
Jingwen Chen ◽  
Yang Zhao

Abstract BackgroundLong non-coding RNA (LncRNA) has been exhibited to exert significant function among human cancers. AC022306.2, as a newly discovered lncRNA, has an unclear function in ovarian cancer (OC). This study aims to uncover the functional role of AC022306.2 in OC and discover its possible mechanism. MethodsThe expression of AC022306.2 and Galactokinase 2 (GALK2) in OC tissues and adjacent non-tumor tissues was detected via qRT-PCR. The CCK-8 assay, cell clonogenesis assay, scratch healing assay and trans-well assay were used to reveal the function of AC022306.2 and GALK2 in ovarian cancer cell lines. Mice xenografts experiment was performed. Bioinformatics predicted the microRNA (miRNA) that bond with AC022306.2 and GALK2, and dual luciferase reporter system confirmed it. Rescue experiments of miRNA mimics and siGALK2 transfection on the basis of AC022306.2 over-expression were carried out to uncover the mechanism by which AC022306.2 played cancer-promoting roles in ovarian cancer.ResultsIt was found that AC022306.2 was up-regulated in EOC tissues compared with adjacent non-tumor tissues. The elevated expression of AC022306.2 was related to the FIGO stage of OC. Functional experiments showed that AC022306.2 overexpression accelerated proliferation and aggression of OC cells in vitro and accelerated tumor growth in vivo. We also found that GALK2 was up-regulated in OC tissues. The expression of GALK2 mRNA in OC tissue was positively associated with the expression of AC022306.2. After AC022306.2 was knocked down, the expression of GALK2 was down-regulated. In addition, GALK2 depletion restored the proliferation and aggression capabilities of OC cells after AC022306.2 overexpression. Mechanically, AC022306.2 acted as a competitive endogenous RNA (ceRNA) of miR-369-3p to modulate the expression of GALK2. The up-regulating of miR-369-3p or the down-regulating of GALK2 partially reversed the effect of AC022306.2 overexpressed on cell propagation and aggression in OC. ConclusionsAC022306.2 is a new oncogene in the carcinogenesis and development of OC. AC022306.2 improves the development of OC by regulating the miR-369-3p / GALK2 axis, indicating that AC022306.2 may have the potential to become a new molecular target for the treatment of OC.


2020 ◽  
Vol 11 ◽  
Author(s):  
Qifeng Cao ◽  
Weiqin Yang ◽  
Xili Ji ◽  
Wei Wang

Emerging evidence suggests that long non-coding RNA (lncRNA) plays a critical role in human disease progression. Recently, a novel lncRNA ST8SIA6-AS1 was shown as an important driver in various cancer types. Nevertheless, its contribution to lung adenocarcinoma (LUAD) remains undocumented. Herein, we found that ST8SIA6-AS1 was frequently overexpressed in LUAD cell lines, tissues, and plasma. Depletion of ST8SIA6-AS1 significantly inhibited LUAD cell proliferation and invasion in vitro and tumor growth in vivo. In term of mechanism, ST8SIA6-AS1 was transcriptionally repressed by tumor suppressor p53, and ST8SIA6-AS1 was mainly located in the cytoplasm and could abundantly sponge miR-125a-3p to increase nicotinamide N-methyltransferase (NNMT) expression, thereby facilitating LUAD malignant progression. Clinically, high ST8SIA6-AS1 was positively correlated with larger tumor size, lymph node metastasis, and later TNM stage. Moreover, ST8SIA6-AS1 was identified as an excellent indicator for MM diagnosis and prognosis. Collectively, our data demonstrate that ST8SIA6-AS1 is a carcinogenic lncRNA in LUAD, and targeting the axis of ST8SIA6-AS1/miR-125a-3p/NNMT may be a promising treatment for LUAD patients.


Author(s):  
Fenrong Chen ◽  
Yan Wang ◽  
Yan Cheng ◽  
Haitao Shi ◽  
Hong Li ◽  
...  

Hepatocellular carcinoma (HCC) is a considerable threat to human life, and patients with HCC are usually diagnosed in the later stages. Although treatment for HCC has recently advanced rapidly, novel targets for HCC are still desperately needed, especially for precision medicine. Here, we identified an HCC enriched long non-coding RNA, AC006262.5, that promoted the proliferation, migration, and invasion of HCC both in vitro and in vivo. In addition, our results revealed that AC006262.5 bound to and regulated miR-7855-5p, a tumor suppressive miRNA in HCC. Moreover, our data illustrated that AC006262.5 regulated the expression of BPY2C via miR-7855-5p. Finally, we found that AC006262.5 and miR-7855-5p formed a regulatory loop. Upregulation of AC006262.5 resulted in the decreased expression of miR-7855-5p, and downregulation of miR-7855-5p further facilitated the expression of AC006262.5. Our study provides novel targets for HCC diagnosis and treatment and sheds light on the lncRNA-miRNA regulatory nexus that controls the pathology of HCC.


Author(s):  
Fengqiang Yu ◽  
Mingqiang Liang ◽  
Yu Huang ◽  
Weidong Wu ◽  
Bin Zheng ◽  
...  

Abstract Background Exosomes have emerged as critical mediators of intercellular communication. Hypoxia is widely recognized as a key regulator of tumor aggressiveness, and significantly affects exosome release by tumor cells. However, the effects of exosomes derived from hypoxic lung adenocarcinoma (LUAD) cells are poorly understood. Methods Samples of miRNA isolated from hypoxic LUAD cell-derived exosomes (HExo) and normoxic LUAD cell-derived exosomes (NExo) were sequenced to identify miRNAs that might mediate tumor progression. Exosomal miRNA was co-cultured with LUAD cells to assess its biological effects on cell migration and metastasis both in vitro and in vivo. The cellular target of exosomal miRNA was confirmed by dual-luciferase assays. Western blot studies showed that exosomal miRNA regulated the related pathway. The availability of circulating exosomal miRNA derived from plasma was also evaluated. Results We found that HExo could significantly enhance the migration and invasion of normoxic LUAD cells. MiRNA sequencing results suggested that miR-31-5p was largely internalized within HExo and could be taken up by normoxic LUAD cells. Exosomal miR-31-5p was found to directly target Special AT-Rich Sequence-Binding Protein 2 (SATB2)-revered epithelial mesenchymal transition and significantly increase activation of MEK/ERK signaling, thereby contributing to tumor progression both in vitro and in vivo. Furthermore, higher levels of circulating exosomal miR-31-5p were detected in LUAD patients, especially in patients with metastatic disease. Conclusions Our findings demonstrate that exosomal miR-31-5p exerts a crucial role in LUAD progression, and could serve as a diagnostic biomarker for LUAD.


2020 ◽  
Author(s):  
Meichen Pan ◽  
Jingren Shi ◽  
Shangqi Yin ◽  
Huan Meng ◽  
Chaonan He ◽  
...  

Abstract Background: Glioma is the most frequent primary malignant brain tumor, characterized by high morbidity, high mortality and dismal prognosis. Numerous analyses have revealed the abnormal expression of long non-coding RNAs (lncRNAs) in glioma cells. This study aims to explore its role in glioma development and prognosis.Methods: The gene expression in cell lines were measured by qRT-PCR. The role of LINC00663 in glioma was confirmed by CCK8, EdU assay, transwell and western blot as well as by in vivo experiments. Besides, Pearson's correlation analysis and Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis were also performed as needed.Results: Firstly, data from our preliminary work (NSFC NO. 81572474) showed that LINC00663 might be largely implicated in glioma. Meanwhile, LINC00663 upregulation confirmed in glioma predicted poor clinical outcomes. Functionally, LINC00663 knockdown restrained cell proliferation, migration and invasion in vitro. Mechanistic investigations validated that LINC00663 silencing decreased AKT activation in glioma cells.Conclusions: LINC00663 promotes glioma development and progression through regulating AKT pathway, suggesting LINC00663 as a probable target for glioma treatment.


2020 ◽  
Vol 40 (9) ◽  
Author(s):  
Li-li Zhou ◽  
Meng Zhang ◽  
Yan-zhen Zhang ◽  
Mei-fen Sun

Abstract Glioma is the most prevalent solid tumor in the central nervous system (CNS). Recently, it has been indicated that long non-coding RNAs (lncRNAs) substantially adjust the development of a variety of human cancers. In the present study, it was found and verified via microarray analysis that lncRNA PSMA3-AS1 exhibited a high expression in glioma tissues and cell lines. Then CCK-8, 5-Ethynyl-2′-deoxyuridine (EdU) staining, plate clone assay, Transwell assay, Western blotting and nude mouse model were adopted to verify PSMA3-AS1’s effects on glioma. Knockdown of PSMA3-AS1 inhibited the migration, proliferation and invasion of glioma cells in vivo and in vitro. Besides, PSMA3-AS1 bound to miR-302a-3p directly reduced the expression of miR-302a-3p, thus functioning as an endogenous sponge confirmed by luciferase reporter assay and bioinformatics analysis. PSMA3-AS1 knockdown remarkably enhanced the role of miR-302a-3p overexpression in cell behaviors in glioma. Moreover, these assays also confirmed that RAB22A was a target of miR-302a-3p. In this research, therefore, the PSMA3-AS1/miR-302a-3p/RAB22A pathway regulatory axis may be revealed in the pathogenesis of glioma, and PSMA3-AS1 can be used as an underlying target for the treatment and prognosis of glioma.


2021 ◽  
Vol 21 (1) ◽  
Author(s):  
Liting You ◽  
Qian Wu ◽  
Zhaodan Xin ◽  
Huiyu Zhong ◽  
Juan Zhou ◽  
...  

Abstract Background miR-124-3p can inhibit integrin β3 (ITGB3) expression to suppress the migration and invasion of gastric cancer (GC), and in the process lncRNA HOXA11-AS may act as a molecular sponge. Methods Luciferase reporter assay was conducted to verify the binding of miR-124-3p and HOXA11-AS. RT-PCR and western blot were performed to detect the expression of HOXA11-AS, miR-124-3p and ITGB3 in GC tissues and cells. Gene silence and overexpression experiments as well as cell migration and invasion assays on GC cell lines were performed to determine the regulation of molecular pathways, HOXA11-AS/miR-124-3p/ITGB3. Furthermore, the role of HOXA11-AS in GC was confirmed in mice models. Results We found HOXA11-AS is up-regulated in GC tissues and can bind with miR-124-3p. Through overexpression/knockdown experiments and function tests in vitro, we demonstrated HOXA11-AS can promote ITGB3 expression by sponging miR-124-3p, consequently enhance the proliferation, migration, and invasion of GC cells. Meanwhile, we validated that HOXA11-AS promotes migration and invasion of GC cells via down-regulating miR-124-3p and up-regulating ITGB3 in vivo. Conclusions We demonstrated that lncRNA HOXA11-AS can increase ITGB3 expression to promote the migration and invasion of gastric cancer by sponging miR-124-3p. Our results suggested that HOXA11-AS may reasonably serve as a promising diagnostic biomarker and a potential therapeutic target of GC.


Sign in / Sign up

Export Citation Format

Share Document