scholarly journals MiR-122-5p suppresses the proliferation, migration, and invasion of gastric cancer cells by targeting LYN

2019 ◽  
Vol 52 (1) ◽  
pp. 49-57 ◽  
Author(s):  
Lei Meng ◽  
Zhangming Chen ◽  
Zhe Jiang ◽  
Ting Huang ◽  
Jie Hu ◽  
...  

Abstract Gastric cancer (GC) is one of malignant tumors with high mortality and morbidity in the world. MicroRNA-122 (miR-122) acts as a tumor suppressor in a variety of cancers and has been found to be dominant in gastric adenocarcinoma. However, the specific biological function of miR-122-5p in GC is not completely clear. In this study, we found that miR-122-5p was low-expressed in GC tissues and cell lines by using qRT-PCR. Overexpression of miR-122-5p inhibited the proliferation, migration, and invasion of GC cells by using CCK-8 and transwell assays. On the contrary, downregulation of miR-122-5p promoted the proliferation, migration, and invasion of GC cells. In addition, we found that the expression of LYN, an Src family tyrosine kinase, was inversely correlated with miR-122-5p expression in GC tissues by using western blot analysis, immunohistochemistry, and qRT-PCR assays. Meanwhile, luciferase assay results indicated that LYN is a direct target of miR-122-5p in GC cells. Moreover, silencing LYN expression by its siRNA inhibited the proliferation, migration, and invasion of GC cells. Importantly, overexpression of LYN restored miR-122-5p-mediated inhibition of the proliferation, migration, and invasion of GC cells. Taken together, our results indicated miR-122-5p inhibited the proliferation, migration, and invasion by targeting LYN in GC.

2019 ◽  
Author(s):  
Yue Pan ◽  
Weixing Chen ◽  
Xin Yuan ◽  
Hongpeng Lu ◽  
Lei Xu ◽  
...  

Abstract Background: Recent studies have shown that microRNA-99a(miR-99a)plays a key role in the development of virious malignancies; however, its relationship with gastric cancer remains unclear. In this study, we investigated the functions and potential mechanisms of miR-99a in gastric cancer. Methods: Real-time qRT-PCR was used to assess the expression levels of miR-99a in gastric cancer tissue samples and cell lines compared to their matched adjacent normal tissues and a normal gastric mucosa epithelial cell line, respectively. SGC-7901 cells were transfected with miR-99a mimics and negative controls to determine the effects of miR-99a overexpression on cell proliferation, cell cycle transition, migration and invasion of gastric cancer cells in vitro . The role of miR-99a in endogenous c-Src expression in gastric cancer cells was also investigated by qRT-PCR and Western blotting. Results: Our results showed a significant increase in miR-99a expression in both gastric cancer tissues and cells compared to normal tissues and cells. Overexpression of miR-99a significantly promoted the cell proliferation, migration and invasion of gastric cancer cells compared to normal cells, with a concurrent increase in the S+G2 phases of the cell cycle. Further investigations found that miR-99a overexpression led to significant upregulation of endogenous c-Src. Conclusion: Taken together, our findings suggest that miR-99a may act as a tumour promoter in the pathogenesis of gastric cancer by indirectly modulating c-Src expression.


2020 ◽  
Vol 11 (9) ◽  
Author(s):  
Jie Zhang ◽  
Wei-qing Qiu ◽  
Hongyi Zhu ◽  
Hua Liu ◽  
Jian-hua Sun ◽  
...  

Abstract Gastric cancer (GC) is one of the most leading malignancies. Long noncoding RNA is related to GC. In this study, 11 miRNAs in the exosomes and six lncRNAs in the tissues was examined by qRT-PCR. Correlation analysis was used to analyze the relationship between miRNAs in exosome and lncRNAs in the tissues. Four miRNAs level in GC tissues were examined by qRT-PCR. MTT was used to determine cell viability. Flow cytometry was used to quantify the apoptotic cells. Transwell assay was used to examine the migration and invasion capacity. Dual-luciferase assay was used to examine the interaction between HOTAIR and miR-30a or -b. Capillary formation was used to determine the capillary formation capacity. Weak negative correlations were found between HOTAIR and miR-30a or -b in GC tissue samples. Interestingly, strong negative correlations were identified between the HOTAIR level in GC tissue samples and the miR-30a or -b levels in plasma exosomes. HOTAIR knockdown GC cells exhibited decreased migration, invasion, proliferation, and upregulated apoptosis, which released more miR-30a and -b into the exosomes. KRAS was upregulated when co-cultured with exosomes from HOTAIR overexpressed cells, and promoted GC cells proliferation, migration, and invasion. Meanwhile, HUVEC cells expressed increased VEGF-A and formatted more capillaries. Subsequently, we identified a 10mer target site of miR-30a or -b in HOTAIR sequence, and the overexpression of HOTAIR induced the degradation of miR-30a or -b, indicating a ceRNA role of HOTAIR. We report the negative correlation between the plasma miRNAs level and GC tissue HOTAIR expression for the first time and unveiled the ceRNA role of HOTAIR in GC. HOTAIR functions as an onco-lncRNA regulating the level of miR-30a and -b in both GC cells and exosomes. These findings may give insight into understanding the mechanism of GC pathogenesis and provide new biomarkers for clinical diagnosis.


2019 ◽  
Vol 47 (2) ◽  
pp. 926-935 ◽  
Author(s):  
Fang Wang

Objective MicroRNA-384 (miR-384) has been reported to function as a tumor suppressor in multiple cancers; however, its role in gastric cancer (GC) remains unclear. Methods We measured expression levels of miR-384 in GC cell lines and in a normal gastric cell line (GES-1). The association between miR-384 and the metadherin gene ( MTDH) was assessed by luciferase reporter assay and western blot. The effects of the miR-384/MTDH axis on GC cell behaviors were measured by CCK-8, wound-healing, and transwell invasion assays. Results miR-384 was significantly downregulated in GC cell lines compared with normal gastric cells. MTDH was identified as a direct target of miR-384 by bioinformatics analysis, luciferase assay, and western blot. Functional assays demonstrated that miR-384 inhibited GC cell proliferation, migration, and invasion through targeting MTDH. Conclusion These results reveal that miR-384 acts as a tumor suppressor in GC and suggest that the miR-384/MTDH axis may be a potential therapeutic target for GC.


2019 ◽  
Vol 60 (5-6) ◽  
pp. 208-218 ◽  
Author(s):  
Tao Xiao ◽  
Zhigang Jie

Background: Gastric cancer (GC) is one of the most common malignant tumors. It is likely to occur in lymph nodes and is prone to distant metastasis in its early stages, which portends a poor prognosis. Previous studies have shown that miRNA-21 was abnormally highly expressed and associated with early metastasis in GC, but the mechanism by which it regulates the invasion and metastasis of GC has not been elucidated. Methods: Epithelial-mesenchymal transition (EMT) is an important pathologic basis of tumor invasion and metastasis, and in this study, the relationship between miRNA-21 and EMT in GC invasion and metastasis was investigated using RT-qPCR, Western blot, and wound scratch and transwell assays. Results: We found that miRNA-21 expression in GC cell lines was higher than in a gastric mucosal epithelial cell line. After transfection with an miRNA-21 mimic, the upregulation of EMT was found to promote migration and invasion of MGC-803 cells. However, the downregulation of EMT was found to accompany the inhibition of invasion and migration of GC cells after downregulation of miRNA-21 expression due to the transfection of an miRNA-21 inhibitor. Conclusions: These findings suggest that miRNA-21 might promote the invasion and metastasis of GC by upregulating EMT.


2014 ◽  
Vol 32 (3_suppl) ◽  
pp. 56-56
Author(s):  
Xu Yanjun ◽  
Zhou Tianhua ◽  
Si Jianmin ◽  
Zhuo Wei

56 Background: Migration and invasion of cancer cells are essential process during cancer metastatic procession. In gastric cancer, cells invasion into the surrounding tissue is a crucial early step. However, the mechanisms have not been fully understood. MicroRNAs, which are a class of small single-stranded non-coding RNA, participate in the malignant progressions of cancer, including metastasis. We study the association between specific dysregulated miRNA and specific metastasis step of gastric cancer, which will provide insights into the potential mechanisms of gastric cancer cells migration, invasion and metastasis. Methods: The expression of miR-375 was assayed using the quantitative real-time PCR analysis. Scratch-wound healing assay, Transwell migration and invasion assay were conducted to study the migration and invasion abilities of cells. Animal experiment was also conducted to examine the effect on liver and lung metastases by overexpression of miR-375. Luciferase assay was conducted to study the association between Snail and miR-375. Results: MiR-375 is downregulated in gastric cancer cells with greater migration and invasion abilities. The expression level of miR-375 is decreased in gastric cancer tissues from metastasis-positive patients compared with that from metastasis-free patients. Overexpression of miR-375 inhibits the migration and invasion abilities of gastric cancer cells. JAK2, which may be a target gene of miR-375, could reverses miR-375 induced inhibition of gastric cancer cells migration and invasion. Liver metastasis was not detected in mice injected with miR-375 overexpressed cells but was apparent in mice injected with cells which were transfected with control vector. The transcription factor Snail, which binds directly to the putative promoter of miR-375, could reduce the expression level of miR-375 significantly. A distinct inverse correlation was found between miR-375 expression and Snail mRNA level. Conclusions: These findings demonstrate that tumor suppressor miR-375, whose expression is directly regulated by the transcription factor Snail, inhibits gastric cancer cells migration, invasion and metastasis by targeting an important protein JAK2.


2021 ◽  
Vol 11 ◽  
Author(s):  
Dejun Yang ◽  
Zunqi Hu ◽  
Yu Zhang ◽  
Xin Zhang ◽  
Jiapeng Xu ◽  
...  

Circular RNA is a kind of RNA with a covalently closed loop, which has a complex ability to modulate genes in the process of tumorigenesis and metastasis. Nevertheless, how circular RNA functions in gastric cancer (GC) remains unclear. The effect of circHIPK3 in vitro was studied here. Quantitative real-time PCR (qRT-PCR) was employed to found that circHIPK3 markedly increased in GC tissues and cell lines. And low expression of circHIPK3 suppressed the GC cells growing and metabolizing. Then the bioinformatics tool predicted the downstream target of circHIPK3, and it was proved by the dual-luciferase report experiment. According to the results of bioinformatics analysis and experimental data, it was clarified that circHIPK3 acted as a sponge of miR-637, releasing its direct target AKT1. The dual-luciferase assay revealed that mir-637 could bind circHIPK3 and AKT1. qRT-PCR data indicated that overexpression circHIPK3 led to the low level of miR-637 and overexpressed miR-637 would reduce AKT1 level. Finally, we demonstrated that the low expression of miR-637 or overexpression of AKT1 could attenuate the anti-proliferative effects of si-circHIPK3. These results suggest that the circHIPK3/miR-637/AKT1 regulatory pathway may be associated with the oncogene and growth of gastric cancer. In short, a new circular RNA circHIPK3 and its function are identified, and the regulatory pathway of circHIPK3/miR-637/AKT1 in the tumorigenesis and development of gastric cancer is discovered.


2021 ◽  
Vol 2021 ◽  
pp. 1-11
Author(s):  
Haibo Yao ◽  
Qinshu Shao ◽  
Yanfei Shao

Objective. To explore the relationship between CTCFL and DPPA2 and validate the positive role of CTCFL/DPPA2 in cell malignant behaviors in gastric cancer. Methods. We predicted gastric cancer-related transcription factors and corresponding target mRNAs through bioinformatics. Levels of CTCFL and DPPA2 were assessed via qRT-PCR and western blot. In vitro experiments were utilized to assay the cell biological behaviors. CHIP was utilized for the assessment of the targeted relationship between CTCFL and DPPA2. Results. CTCFL and DPPA2 were both highly expressed in gastric cancer cells, and high CTCFLL and DPPA2 could promote cell malignant behaviors. CHIP validated that DPPA2 was a target of CTCFL. In addition, high DPPA2 rescued the repressive impact of CTCFL silencing on the cell proliferation, migration, and invasion in gastric cancer. Conclusion. The transcription factor CTCFL fosters cell proliferative, migratory, and invasive properties via activating DPPA2 in gastric cancer.


2021 ◽  
Author(s):  
Yi-Chen Wang ◽  
Song Lu ◽  
Xiao-Jiang Zhou ◽  
Li Yang ◽  
ping liu ◽  
...  

Abstract Purpose: microRNAs (miRNAs), which may function as oncogenes or tumor suppressors, have been verified in the development of breast carcinoma, melanoma, and some other tumors. The dysregulated miR-1273h-5p in tissue samples of gastric cancer (GC) may be involved in the progression of GC. The aim of this study was to verify the biological function of miR-1273h-5p in GC progression.Method: The differential expression of microRNAs between GC and tumor-adjacent normal tissues was detected by microarrays, and polymerase chain reaction (PCR) analysis was used for miR-1273h-5p and chemokine (C-X-C motif) ligand 12 (CXCL12) mRNA expressions. The effect of miR-1273h-5p on cell proliferation and apoptosis was evaluated by CCK-8 assay and flow cytometry; cell migration and invasion were observed by using the transwell method. In addition, protein levels were determined by Western blot. SGC-7901 cell transfected gene sequences were injected into BALB/c-nu mice to establish a xenograft model in order to validate the biological function of miR-1273h-5p in vivo.Results: Compared to tumor-adjacent normal tissue and GES-1 cells, miR-1273h-5p was significantly down-regulated in tissues and cells of GC. The overexpression of miR-1273h-5p could inhibit cell proliferation, migration, invasion, and promote cell apoptosis; in contrast, inhibition of miR-1273h-5p expression could reverse this process. Moreover, a significant up-regulation of CXCL12 was observed when the miR-1273h-5p was down-regulated in GC cells. Additionally, tumor tissues were collected from mice after 21 days of feeding, revealing that miR-1273h-5p significantly reduces tumor volume and tumor weight. Conclusions: miR-1273h-5p regulates cell proliferation, migration, invasion, and apoptosis during GC progression by directly binding to CXCL12 mRNA 3'-UTR, thus can be used as a potential diagnostic and a novel therapeutic target for GC in clinical practice.


2021 ◽  
Author(s):  
Yan Ding ◽  
Sujie Gao ◽  
Jiabin Zheng ◽  
Xuebo Chen

Abstract Gastric cancer (GC) is one of the most human malignancies. The 5-fluorouracil (5-Fu) is a first-line anti-gastric cancer chemotherapeutic agent. However, a large number of GC patients developed 5-Fu resistance. Currently, the biological roles and molecular mechanisms of lncRNA-SNHG16 in 5-Fu resistant gastric cancer still remain elusive. Here we report SNHG16 as well as PTBP1 are positively associated with 5-Fu resistance of gastric cancer. SNHG16 and PTBP1 are significantly upregulated in gastric tumors and cell lines. Silencing SNHG16 or PTBP1 effectively sensitized GC cells to 5-Fu. Furthermore, the glucose metabolism was remarkedly elevated in 5-Fu resistant GC cells. Under low glucose supply, 5-Fu resistant cells displayed higher vulnerability than parental GC cells. Bioinformatical analysis and luciferase assay demonstrated that SNHG16 downregulated miR-506-3p by sponging it. We identified PTBP1 was a direct target of miR-506-3p in GC cells. RNA-seq results indicated PTBP1 positively regulated multiple glycolysis enzymes expressions, including GLUT1, HK2 and LDHA. Bioinformatics analysis illustrated the 3’UTRs of glycolysis enzymes contained multiple PTBP1 binding sites, which were further verified by RNA-pull down and RNA immunoprecipitation assays. Consequently, we demonstrated PTBP1 upregulated the mRNAs of glycolysis enzymes via promoting the mRNAs stabilities. Finally, in vivo xenograft experiments validated that blocking the SNHG16-mediated miR-506-3p-PTBP1 axis effectively sensitized xenograft tumors to 5-Fu treatments. Summarily, our study reports biological roles and molecular mechanisms underlying the lncRNA SNHG16-mediated 5-Fu resistance in GC through modulating the miR-506-3p-PTBP1-glycolysis axis, presenting a promising prospect in improvement of 5-Fu therapy for chemoresistant GC patients.


Author(s):  
Lei Zheng ◽  
Junli Cao ◽  
Lijie Liu ◽  
Hongmei Xu ◽  
Lanlan Chen ◽  
...  

Upregulating the expression of long non-coding RNA LINC00982 controlled cell proliferation in gastric cancer, but the regulatory molecular mechanisms are yet to be expounded. We here aimed to elaborate how LINC00982 regulated the malignancy of gastric cancer cells. RT-qPCR and Western blot analysis were used to detect the expression of LINC00982 and CTSF in gastric cancer tissues and cells. Modulatory effect of LINC00982 on gastric cancer cells was assessed by CCK-8, colony formation, Transwell migration and invasion assays. The relationship between LINC00982, HEY1 and CTSF was examined by RIP, luciferase assay, and ChIP, and their interaction in the regulation of gastric cancer cellular functions was analyzed by performing gain-of-function and rescue assays. The nude mouse model of tumor formation was developed to examine the effects of LINC00982 on tumorigenesis. LINC00982 was lowly expressed in gastric cancer tissues, while its overexpression impaired the proliferative, migratory and invasive properties of gastric cancer cells. Furthermore, LINC00982 could bind to transcription factor HEY1 and inhibited its expression. Through blocking the binding of HEY1 to CTSF promoter. LINC00982 promoted the expression of CTSF. Overexpression of HEY1 or inhibition of CTSF could reverse the anti-tumor effects of LINC00982 on gastric cancer, which were further demonstrated in vivo. Taken together, LINC00982 acted as a tumor suppressor in gastric cancer, which is therefore suggested to be a potential anti-tumor target for gastric cancer.


Sign in / Sign up

Export Citation Format

Share Document