scholarly journals miR-339-5p Inhibits Autophagy to Reduce the Resistance of Laryngeal Carcinoma on Cisplatin via Targeting TAK1

2021 ◽  
Vol 2021 ◽  
pp. 1-9
Author(s):  
Guang Li ◽  
Zexing Cheng

Laryngeal carcinoma is a malignant disease with high morbidity and mortality. Several studies have indicated that miRNA dysfunction involves in the development of laryngeal carcinoma. In this study, the connection of miR-339-5p and laryngeal carcinoma was investigated, and qRT-PCR, CCK-8, and flow cytometry assay were used to observe the function of miR-339-5p on laryngeal carcinoma. Besides, the target database, dual-luciferase reporter assay, and western blot were used to explore the regulation mechanism of miR-339-5p on the progression of laryngeal carcinoma. The results showed that miR-339-5p was significantly downregulated in cisplatin-resistant cells of laryngeal carcinoma, and miR-339-5p upregulation could weaken the resistance of laryngeal carcinoma cells on cisplatin. Moreover, miR-339-5p could directly react with 3 ′ -UTR of TAK1, and TAK1 could reverse the effects of miR-339-5p on the progression of autophagy. In conclusion, this study suggests that miR-339-5p can inhibit the autophagy to decrease the cisplatin resistance of laryngeal carcinoma via targeting TAK1.

2021 ◽  
Vol 2021 ◽  
pp. 1-8
Author(s):  
Qilong Wang ◽  
Xiaomin Hao ◽  
Gang Xu ◽  
Tiesheng Lv

Colon cancer is a common malignant disease with high morbidity and mortality, and miRNA dysfunction has been confirmed as an important reason for cancer development. Several studies have verified miR-605-3p as a tumor inhibitor while its roles in colon cancer remain uncertain. In this study, the specimen of the patients and the cell lines of colon cancer were used to observe the expression of miR-605-3p, and the CCK-8, Transwell assay, and flow cytometry assay were used to observe the functions of miR-605-3p in colon cancer cells. The downstream factors of miR-605-3p were predicted by TargetScan and then were verified by dual-luciferase reporter assay. Moreover, western blot was used to investigate the effect of miR-605-3p on Wnt/β-catenin signal pathway. The result showed that miR-605-3p was extremely downregulated in the pathological tissues and tumor cells, and miR-605-3p could effectively induce the apoptosis and impede the proliferation and invasion of the tumor cells. It was found that KIF3B was a target of KIF3B; decreased KIF3B could reverse the effects of miR-605-3p on colon cancer. Besides, the inactivated Wnt/β-catenin pathway was also observed in colon cells when miR-605-3p was upregulated, and the phenomenon could be rescued by KIF3B upregulation. In conclusion, miR-605-3p could inactivate the Wnt/β-catenin pathway induced via promoting KIF3B expression.


2021 ◽  
Vol 2021 ◽  
pp. 1-9
Author(s):  
Jianwei Li ◽  
Su Yan

Hepatocellular carcinoma (HCC) has threatened the health of humans, and some evidence has indicated that miR-466 involves the progressions of some cancers. This study focused on the role of miR-466 in the formation and development of HCC. The expression levels of miR-466 in the tissues of patients and HCC cell lines were measured by qRT-PCR, and CCK-8, transwell assay, and flow cytometry assay were used to observe the functions of miR-466 on the HCC cells. Moreover, the miRNA databases, dual-luciferase reporter assay, and Western blot were used for the investigation of the regulation mechanism of miR-466 on HCC cells. The results showed that miR-466 was significantly downregulated in HCC tissues and cell lines, and inhibited proliferation, invasion, and high apoptosis were found in HCC cells when miR-466 was overexpressed. The results confirmed that FMNL2 was a target of miR-466, and increased FMNL2 could reverse the effects of miR-466 on the phenotype of HCC cells. Besides, it was also found that miR-466 was involved in the regulation of NF-κB and Wnt/β-catenin pathways in HCC cells via targeting FMNL2. In conclusion, the results of this study suggest that miR-466 regulates the activities of NF-κB and Wnt/β-catenin pathways to inhibit the progression of HCC cells via targeting FMNL2.


2022 ◽  
Vol 8 (1) ◽  
Author(s):  
Changyu Zhu ◽  
Xiaolei Jiang ◽  
Hua Xiao ◽  
Jianmei Guan

AbstractRadioresistance prevails as one of the largest obstacles in the clinical treatment of nasopharyngeal carcinoma (NPC). Meanwhile, tumor-derived extracellular vesicles (TEVs) possess the ability to manipulate radioresistance in NPC. However, its mechanism remains to be further explored. Therefore, the current study set out to explore the mechanism of microRNA (miR)-142-5p delivered by TEVs in regard to the radiosensitivity of NPC. Firstly, peripheral blood samples were collected from patients with radioresistance and radiosensitivity, followed by RT-qPCR detection of miR-142-5p expression. A dual-luciferase reporter assay was carried out to elucidate the targeting relationship of miR-142-5p with HGF and EGF. In addition, radiotherapy-resistant NPC cell models were established by screening NPC cells with gradient increasing radiation exposure, and co-incubated with EVs isolated from miR-142-5p mimic-transfected NPC cells, followed by overexpression of HGF and EGF. Moreover, cell viability was detected by means of MTS, cell proliferation with a colony formation assay, cell apoptosis with flow cytometry, and expression patterns of related genes with the help of Western blot analysis. NPC xenotransplantation models in nude mice were also established by subcutaneous injection of 5-8FR cells to determine apoptosis, tumorigenicity, and radiosensitivity in nude mice. It was found that miR-142-5p was poorly expressed in peripheral blood from NPC patients with radioresistance. Mechanistic experimentation illustrated that miR-142-5p inversely targeted HGF and EGF to inactivate the HGF/c-Met and EGF/EGFR pathways, respectively. NPC cell apoptosis was observed to be augmented, while their radioresistance and proliferation were restricted by EVs-miR-142-5p or HGF silencing, or EGF silencing. Furthermore, EVs-miR-142-5p inhibited growth and radioresistance and accelerated the apoptosis of radiotherapy-resistant NPC cells in nude mice by inhibiting the HGF/c-Met and EGF/EGFR pathways. Collectively, our findings indicated that TEVs might inhibit the HGF/c-Met and EGF/EGFR pathways by delivering miR-142-5p into radiotherapy-resistant NPC cells to enhance radiosensitivity in NPC.


2020 ◽  
Author(s):  
Dan Wang ◽  
Jingbo Yang

Abstract Objective To probe into the regulatory mechanism of miR-375 in hepatocellular carcinoma (HCC) cells under sorafenib treatment. Methods Western blotting and qRT-PCR were applied to measure the expressions of miR-375 and SIRT5 in parental HCC cells (HepG2 and Huh7) and sorafenib-resistant HCC cells (HepG2/so and Huh7/so). HepG2/so cells were accordingly transfected with miR-375 mimic, miR-375 inhibitor, sh-SIRT5, pcDNA3.1-SIRT5 or negative control. Western blotting measured the expressions of p62, LC3I and LC3II in HCC cells. CCK-8 and flow cytometry assessed the survivability and apoptosis of HCC cells, respectively. Bioinformatics techniques and dual-luciferase reporter assay predicted and verified the targeting relationship between miR-375 and SIRT5. Results MiR-375 was under-expressed and SIRT5 was over-expressed in HCC cells. Autophagy inhibitor impaired the survival of HepG2/so cells transfected with miR-375 inhibitor. Autophagy activator enhanced the drug resistance of HepG2/so cells transfected with miR-375 mimic. MiR-375 suppressed the drug resistance of HepG2/so cells by inhibiting autophagy. SIRT5 enhanced the drug resistance of HepG2/so cells by promoting autophagy and it could be targeted by miR-375. Conclusion MiR-375 suppresses autophagy to attenuate the drug resistance of HCC cells by regulating SIRT5. The findings of this study may provide new therapeutic targets for treating hepatocellular carcinoma.


2021 ◽  
Vol 65 (4) ◽  
Author(s):  
Yang Liu ◽  
Weichun Guo ◽  
Shuo Fang ◽  
Bin He ◽  
Xiaohai Li ◽  
...  

Osteosarcoma (OS), characterized by high morbidity and mortality, is the most common bone malignancy worldwide. MicroRNAs (miRNAs) play a crucial role in the initiation and development of OS. The purpose of this study was to investigate the roles of miR-1270 in OS. RT-qPCR and Western blot were applied to detect the mRNA and protein level, respectively. CCK-8, colony formation, and TUNEL assays were conducted to determine the cell viability, proliferation, and apoptosis of OS cells. Wound healing and transwell assay were performed to detect the migration and invasion ability of OS cells. Bioinformatics analysis and dual-luciferase reporter assay were used to predict the target genes of miR-1270. Tumor xenograft in vivo assay was carried out to determine miR-1270 effect on the tumor size, volume, and weight. In this study, miR-1270 was overexpressed in OS tissues and cells. However, miR-1270 knockdown inhibited the proliferation, migration and invasion, and promoted the OS cells’ apoptosis. Mechanistically, cingulin (CGN) was predicted and proved to be a target of miR-1270 and partially alleviated the effects of miR-1270 on the proliferation, migration and invasion ability of OS cells. Taken together, knockdown of miR-1270 may inhibit the development of OS via targeting CGN. This finding may provide a novel therapeutic strategy for OS.


2021 ◽  
Author(s):  
Chun Duan ◽  
Bin Quan ◽  
Ni Wang ◽  
Jianghua Yang ◽  
Yan-Lin Yu

Abstract Background: Hepatocellular carcinoma (HCC) is a common malignancy with high morbidity. The current study aimed to explore the molecular mechannism of lncRNA SLC16A1-AS1 in the tumorigenesis of HCC.Material and Methods: The expression of SLC16A1-AS1 and miR-411 were examined in clinical HCC tissues. HCC cell lines Hep3B and Huh-7 were employed and transfected with si-SLC16A1-AS1. The correlation between SLC16A1-AS1 and miR-411 was verified by luciferase reporter assay. Cell viability was detected by CCK-8 assay. Cell migration and invasion capacity were examined by transwell assay. The protein level of MITD1 was analyzed by western blotting.Results: The expression of SLC16A1-AS1 markedly increased in HCC tissues and cell lines. Subsequent studies identified SLC16A1-AS1 as a downstream target of miR-411. In addition, SLC16A1-AS1 knockdown and miR-411 overexpression significantly stagnated progression of HCC cells. SLC16A1-AS1 knockdown also downregulated MITD1 levels. Conclusion: Our findings showed that SLC16A1-AS1 was overexpressed in HCC cells and tissues. SLC16A1-AS1 promoted the malignant characteristics of HCC cells and acted as an oncogene. Its regulatory effect may be associated with miR-411/MITD1 axis. Therefore, SLC16A1-AS1 has a potential be used as a biomarker or therapeutic target for the treatment of HCC.


Open Medicine ◽  
2020 ◽  
Vol 15 (1) ◽  
pp. 1193-1201
Author(s):  
Zhang E ◽  
Chunli Li ◽  
Yuandi Xiang

AbstractThis research aimed to illustrate the biological function and associated regulatory mechanism of lncRNA FOXD3-AS1 (FOXD3-AS1) in nasopharyngeal carcinoma (NPC). This research initially found that FOXD3-AS1 was obviously upregulated in NPC cell lines by quantitative reverse transcription polymerase chain reaction (qRT-PCR) detection. Next, the direct target of FOXD3-AS1 was predicted by bioinformatics and further verified by dual-luciferase reporter assay. MiroRNA-135a-5p (miR-135a-5p) was identified as the target gene of FOXD3-AS1 and down-expressed in C666-1 cells compared to NP69. In addition, function assays were conducted in C666-1 cells, including methyl tetrazolium assay, flow cytometry, Caspase3 activity detection, and western blot assay. Our results suggested that miR-135a-5p upregulation inhibited NPC cell growth, enhanced cell apoptosis, promoted Caspase3 activity, increased cleaved-Caspase3, and reduced pro-Caspase3 level. Moreover, we found that FOXD3-AS1 knockdown notably inhibited C666-1 cell proliferation, increased cell apoptosis, enhanced Caspase3 activity, enhanced cleaved-Caspase3 expression, and suppressed pro-Caspase3 level in C666-1 cells. However, these findings were reversed in C666-1 cells by miR-135a-5p mimic co-transfection. To sum up, our data showed that FOXD3-AS1 knockdown regulated cell growth and apoptosis in NCP cells via altering miR-135a-5p expression, suggesting that FOXD3-AS1 might be a therapeutic target for NPC diagnosis and treatment.


2021 ◽  
Vol 2021 ◽  
pp. 1-12
Author(s):  
Xu Niu ◽  
Haitao Sun ◽  
Feng Qiu ◽  
Jing Liu ◽  
Tianchi Yang ◽  
...  

Objective. To analyze the function of miR-10b-5p in suppressing the invasion and proliferation of primary hepatic carcinoma cells by downregulating erythropoietin-producing hepatocellular receptor A2 (EphA2). Material and Methods. Eighty-six hepatic carcinoma (HCC) tissue specimens and 86 corresponding adjacent tissue specimens were collected, and the mRNA expression of miR-10b-5p and Ephrin type-A receptor 2 (EphA2) in the specimens was determined using a reverse transcription-polymerase chain reaction (RT-PCR) assay. Western blot was employed to quantify EphA2, B-cell chronic lymphocytic leukemia/lymphoma-2 (Bcl-2), Bcl-2-associated X protein (Bax), and Caspase-3 in the cells, and CCK8, Transwell assay, and flow cytometry were applied to evaluate the proliferation, invasion, and apoptosis of cells, respectively. Moreover, the dual luciferase reporter assay was utilized for correlation analysis between miR-10b-5p and EphA2. Results. miR-10b-5p was lowly expressed in HCC, while EphA2 was highly expressed. Cell experiments revealed that miR-10b-5p overexpression or EphA2 knockdown could reduce cell proliferation, accelerate apoptosis, strongly upregulate Bax and Caspase-3, and downregulate Bcl-2. In contrast, miR-10b-5p knockdown or EphA2 overexpression gave rise to reverse biological phenotypes. Furthermore, dual luciferase reporter assay verified that miR-10b-5p was a target of EphA2, and the rescue experiment implied that transfection of pCMV-EphA2 or Si-EphA2 could reverse EphA2 expression and cell biological functions caused by miR-10b-5p overexpression or knockdown. Conclusions. miR-10b-5p reduced HCC cell proliferation but accelerate apoptosis by regulating EphA2, suggesting it has the potential to be a clinical target for HCC.


2021 ◽  
Vol 18 (2) ◽  
pp. 97-106
Author(s):  
Jianxing Zheng ◽  
Daming Cheng ◽  
Dongyang Wu ◽  
Libing Wang ◽  
Fengzhi Qu ◽  
...  

Objective: This study explored the potential function of miR-452-5p in hepatocellular carcinoma (HCC) and clarified the mechanism underlying HCC progression. Materials & methods: Real-time quantitative PCR was used to detect miR-452-5p and COLEC10 mRNA expression in HCC, western blot was performed to test COLEC10 protein expression. The regulatory mechanism of miR-452-5p/COLEC10 in HCC cells was explored using CCK-8, wound healing assay, Transwell and dual-luciferase reporter assay. Results: MiR-452-5p was greatly upregulated in HCC cells, and it served as an oncogene playing an active role in HCC cell proliferation, migration and invasion. COLEC10 was identified as the target of miR-452-5p in HCC attenuating the promoting effect of miR-452-5p on HCC cells upon overexpression. Conclusion: MiR-452-5p can promote the progression of HCC via targeting COLEC10.


2020 ◽  
Author(s):  
Shengming Fan ◽  
Pei Chen ◽  
Shugang Li

Abstract Background: The study aimed to investigate the regulatory relationship between miR-145-5p and ABRACL, and tried to clarify the mechanisms underlying the proliferation, migration and invasion of esophageal cancer (EC) cells.Methods: Gene expression data related to EC were accessed from TCGA database and the “edgeR” package was used to screen the differentially expressed miRNA (DEmiRNAs) and genes (DEGs). TargetScan, miRDB and miRTarBase databases were used to predict potential targets for the target miRNA miR-145-5p. qRT-PCR and Western blot were performed to assess the expression of miR-145-5p and ABRACL in EC cells. Dual-luciferase reporter assay was performed for verification of the targeting relationship between miR-145-5p and ABRACL. Functional experiments including colony formation assay, Transwell migration and invasion assays were used to detect the proliferation, migration and invasion abilities of EC cells. Results: The expression of miR-145-5p was significantly decreased in EC, while ABRACL was remarkably increased. In addition, there was a negative correlation identified between miR-145-5p and ABRACL expression levels. Overexpressing miR-145-5p was able to suppress cell proliferation, migration and invasion, whereas silencing miR-145-5p posed an opposite effect. In the meantime, ABRACL was identified as a direct target of miR-145-5p by dual-luciferase reporter assay. Furthermore, miR-145-5p could inhibit the expression of ABRACL, in turn inhibiting the proliferation, migration and invasion of EC cells. Conclusion: miR-145-5p functions on the proliferation, migration and invasion of EC cells via targeting ABRACL, and it may be a novel therapeutic target for EC treatment.


Sign in / Sign up

Export Citation Format

Share Document