scholarly journals MicroRNA-224 Promotes Pancreatic Cancer Cell Proliferation and Migration by Targeting the TXNIP-Mediated HIF1α Pathway

2018 ◽  
Vol 48 (4) ◽  
pp. 1735-1746 ◽  
Author(s):  
Guanghui Zhu ◽  
Lianming Zhou ◽  
Haijun Liu ◽  
Yuanzhou Shan ◽  
Xueli Zhang

Background/Aims: MicroRNAs (miRNAs) have been shown to participate in the development of pancreatic ductal adenocarcinoma (PDAC) by modulating multiple cellular processes. Increased miR-224 expression enhances proliferation and metastasis in human cancers. This study aimed to investigate the role of miR-224 and its underlying mechanism of action in PDAC. Methods: BrdU, MTT, and cell migration assays were performed to determine cell proliferation, viability, and migration, respectively. The binding sites of miR-224 were identified using a luciferase reporter system, whereas protein expression of target genes was determined by immunoblotting and immunofluorescence analyses. A BALB/c nude mouse xenograft model was used to evaluate the role of miR-224 in vivo. Results: We demonstrated that miR-224 expression was enhanced in PDAC cells and tissues, and was related to migration and proliferation. Noticeably, miR-224 overexpression promoted the proliferation, migration, and metastasis of Panc1 cells, while miR-224 inhibition had the reverse effect on PDAC cells. Moreover, we found that thioredoxin-interacting protein (TXNIP) is a target of miR-224. The results also indicated that miR-224 inversely regulated TXNIP by binding directly to its 3′-untranslated region, which resulted in the activation of hypoxia-inducible factor 1α (HIF1α). Further, either TXNIP re-expression or HIF1α depletion abolished the effects of miR-224 on the proliferation and migration of PDAC cells in vitro and in vivo. Regarding the relationship of TXNIP and HIF1α, we found that TXNIP mediated the nuclear export of HIF1α and its degradation by forming a complex with HIF1α. Conclusion: The miR-224-TXNIP-HIF1α axis may be useful in developing novel therapies for PDAC.

2020 ◽  
Author(s):  
Yongbin Chi ◽  
Yan Wang ◽  
Dawei Zhou ◽  
Wanchao Liu ◽  
Ruodong Han ◽  
...  

Abstract Background: Circular RNAs (circRNAs) are a new family of abundant regulatory RNAs with roles in various types of cancer. While the hsa_circ_0046701 (circ-YES1) function in non-small cell lung cancer (NSCLC) is unclear. Methods: Circ-YES1 expression in normal pulmonary epithelial and NSCLC cells was examined. The small interfering RNA for circ-YES1 was prepared, cell proliferation and migration were assessed. Tumorigenesis in nude mice was assayed to validate the role of circ-YES1. Bioinformatics analyses and luciferase reporter assays were utilized to identify downstream targets of circ-YES1. Results: Compared to normal pulmonary epithelial cells, the circ-YES1 expression increased in NSCLC cells, and cell proliferation and migration were suppressed after circ-YES1 knockdown. Both high mobility group protein B1 (HMGB1) and miR-142-3p were found to be downstream targets of circ-YES1, and miR-142-3p inhibition and HMGB1 overexpression reversed the effects of circ-YES1 knockdown on cell proliferation and migration. Similarly, HMGB1 overexpression reversed the miR-142-3p overexpression effects on these two processes. The imaging experiment results revealed that circ-YES1 knockdown impeded tumor development and metastasis in a nude mouse xenograft model. Conclusion: Taken together, our results show that circ-YES1 promotes tumor development through the miR-142-3p–HMGB1 axis and support the development of circ-YES1 as a new therapeutic NSCLC target.


2022 ◽  
Vol 19 (1) ◽  
Author(s):  
Min Chu ◽  
Yingchao Fan ◽  
Liting Wu ◽  
Xiaoyan Ma ◽  
Jinfeng Sao ◽  
...  

Abstract Purpose This study aimed to explore the role of long non-coding RNA (lncRNA) BDNF-AS in the progression of multiple myeloma (MM). Methods The expression of BDNF-AS, miR-125a-5p, and miR-125b-5p in MM serum and cell lines were detected by quantitative reverse transcriptase PCR (qRT-PCR). The binding relationships between miR-125a/b-5p and BDNF-AS or Bcl-2 were predicted by Starbase and verified by luciferase reporter assay and RNA immunoprecipitation (RIP) assay. Cell proliferation was evaluated by Cell Counting Kit-8 (CCK-8) assay and 5-ethynyl-2′-deoxyuridine (EdU) staining assay. Cell migration was evaluated by wound healing assay. The expression levels of apoptosis-related proteins were evaluated by Western blot analysis. The role of BDNF-AS was also investigated in a xenograft tumor model in vivo. Results BDNF-AS was significantly upregulated, while miR-125a-5p and miR-125b-5p were downregulated in MM serum and corresponding cancer cell lines. Knockdown of BDNF-AS effectively inhibited the proliferation and migration of MM.1S and U266 cells, and co-transfection of miR-125a-5p or miR-125b-5p inhibitor and sh-BDNF-AS enhanced cell proliferation and migration compared with that in sh-BDNF-AS group. Knockdown of miR-125a-5p or miR-125b-5p significantly enhanced the proliferation and migration of MM.1S and U266 cells, and co-transfection of sh-Bcl-2 and miR-125a/b-5p inhibitor inhibited cell proliferation compared with that in miR-125a/b-5p inhibitor group. Moreover, knockdown of BDNF-AS increased the expression levels of apoptosis-related proteins (cleaved caspase 3 and cleaved PARP), while knockdown of miR-125a-5p or miR-125b-5p reduced the expression levels of these apoptosis-related proteins compared with knockdown of BDNF-AS. Furthermore, knockdown of BDNF-AS effectively suppressed MM tumor growth in vivo. Conclusion Our findings revealed that knockdown of BDNF-AS inhibited the progression of MM by targeting the miR-125a/b-5p-Bcl-2 axis, indicating that BDNF-AS might serve as a novel drug target for MM.


2021 ◽  
Vol 21 (1) ◽  
Author(s):  
Quan Lin ◽  
Yan Jia ◽  
Duo Zhang ◽  
Hongjuan Jin

Abstract Background Long non-coding RNAs (lncRNAs) are vital regulators of gene expression and cellular processes in multiple cancers, including melanoma. Nevertheless, the function of lncRNA NCK1-antisense 1 (NCK1-AS1) in melanoma remains unknown. Methods RT-qPCR was used to analyze the expression of NCK1-AS1, microRNA-526b-5p (miR-526b-5p) and ADAM metallopeptidase domain 15 (ADAM15). Cell proliferation was determined by CCK-8, colony formation and EdU assays. Cell migration was assessed by transwell migration and wound healing assays. Mechanism experiments including luciferase reporter, RIP and RNA pull down assays were conducted to demonstrate the interactions between RNAs. Xenograft model was established to verify the function of NCK1-AS1 and miR-526b-5p in melanoma in vivo. Results NCK1-AS1 was overexpressed in melanoma cell lines and NCK1-AS1 knockdown hampers the proliferation and migration of melanoma cells. Besides, miR-526b-5p binds to NCK1-AS1 in melanoma and ADAM15 was validated as its downstream target. Further, the inhibitory effects of NCK1-AS1 knockdown on cell proliferation and migration in melanoma were reversed by the depletion of miR-526b-5p and further counteracted by ADAM15 knockdown. The growth of melanoma tumors was hindered by the down-regulation of NCK1-AS1 or up-regulation of miR-526b-5p. Conclusion NCK1-AS1 facilitates cell proliferation and migration in melanoma via targeting miR-526b-5p/ADAM15 axis.


2020 ◽  
Author(s):  
Guan-Bin Qi ◽  
Lei Li

Abstract Background: LINC00958, a newly identified lncRNA, has been reported to be closely linked to tumorigenesis in multiple cancers. However, its specific role in non-small cell lung cancer (NSCLC) remains unclear.Methods: The expression of LINC00958 was determined by RT-qPCR analysis. Cell proliferation and migration were evaluated by CCK-8 and transwell assays, respectively. Xenograft tumor models were established to examine the effect of LINC00958 on tumor growth in vivo. Luciferase reporter assays were performed to determine the interaction between LINC00958 and miR-204-3p and the interaction between miR-204-3p and KIF2A.Results: We found that LINC00958 was up-regulated in NSCLC tissues and cell lines. Down-regulation of LINC00958 inhibited cell proliferation and migration in vitro and suppressed tumor growth in vivo. Mechanically, we revealed that LINC00958 influenced NSCLC progression partly by sponging miR-204-3p and regulating KIF2A expression.Conclusions: Our study provided new insights into the role of LINC00958 as a promising prognostic biomarker and a therapeutic target for NSCLC.


2020 ◽  
Author(s):  
He Chen ◽  
Pengcheng Liu ◽  
Lei Wang ◽  
Yanxia Yu ◽  
Yan Zhao ◽  
...  

Abstract BACKGROUND: Gastric cancer (GC) is one of the most common malignant cancers, with high morbidity and mortality rates worldwide. The present study was to explore whether miR-200b is a tumor suppressor in GC and to unveil the potential mechanisms. METHODS: Levels of c-Myc, Cyclin D1, MMP-3 and MMP-9 expression were detected respectively by qRT-PCR and Western blot assay. BrdU proliferation assay, Cell cycle analysis, Wound-healing and Transwell assays were used to study the role of miR-200b with inhibitor, mimics or ZEB1-RNAi in TGF-β1-treated SGC-7901/DDP cells. The xenograft model with nude mice was established to unveil the role of miR-200b in vivo.RESULTS: Compared with the paracancerous tissues, miR-200b was decreased in GC patients and SGC-7901/DDP cells. Lower level of miR-200b induced by its inhibitor promoted TGF-β1-treated SGC-7901/DDP cells proliferation and migration, and increased the levels of c-Myc, Cyclin D1, MMP-3, MMP-9, β-catenin and APC. Interestingly, miR-200b mimics and ZEB1-RNAi were able to reduce the proliferation and migration of TGF-β1-induced SGC-7901/DDP cells as well as their levels of c-Myc, Cyclin D1, MMP-3 MMP-9, β-catenin and APC. In addition, ZEB1 was indeed the potential target of miR-200b identified by dual luciferase reporter gene assay. Xenograft model also suggested that over-expression of miR-200b suppressed the growth of tumor in vivo. CONCLUSION: Taken together, our findings suggest that miR-200b be likely to play an important role in activating TGF-β1-induced SGC-7901/DDP cells and perform as a tumor suppressor by targeting ZEB1 in GC. What’s more, miR-200b may modulate Wnt/β-catenin signaling pathway in TGF-β1-induced SGC-7901/DDP cells.


2020 ◽  
Author(s):  
He Chen ◽  
Pengcheng Liu ◽  
Lei Wang ◽  
Yanxia Yu ◽  
Yan Zhao ◽  
...  

Abstract BACKGROUND: Gastric cancer (GC) is one of the most common malignant cancers, with high morbidity and mortality rates worldwide. The present study was to explore whether miR-200b is a tumor suppressor in GC and to unveil the potential mechanisms. METHODS: Levels of c-Myc, Cyclin D1, MMP-3 and MMP-9 expression were detected respectively by qRT-PCR and Western blot assay. BrdU proliferation assay, Cell cycle analysis, Wound-healing and Transwell assays were used to study the role of miR-200b with inhibitor, mimics or ZEB1-RNAi in TGF-β1-treated SGC-7901/DDP cells. The xenograft model with nude mice was established to unveil the role of miR-200b in vivo.RESULTS: Compared with the paracancerous tissues, miR-200b was decreased in GC patients and SGC-7901/DDP cells. Lower level of miR-200b induced by its inhibitor promoted TGF-β1-treated SGC-7901/DDP cells proliferation and migration, and increased the levels of c-Myc, Cyclin D1, MMP-3, MMP-9, β-catenin and APC. Interestingly, miR-200b mimics and ZEB1-RNAi were able to reduce the proliferation and migration of TGF-β1-induced SGC-7901/DDP cells as well as their levels of c-Myc, Cyclin D1, MMP-3 MMP-9, β-catenin and APC. In addition, ZEB1 was indeed the potential target of miR-200b identified by dual luciferase reporter gene assay. Xenograft model also suggested that over-expression of miR-200b suppressed the growth of tumor in vivo. CONCLUSION: Taken together, our findings suggest that miR-200b be likely to play an important role in activating TGF-β1-induced SGC-7901/DDP cells and perform as a tumor suppressor by targeting ZEB1 in GC. What’s more, miR-200b may modulate Wnt/β-catenin signaling pathway in TGF-β1-induced SGC-7901/DDP cells.


2020 ◽  
Author(s):  
He Chen ◽  
Pengcheng Liu ◽  
Lei Wang ◽  
Yanxia Yu ◽  
Yan Zhao ◽  
...  

Abstract BACKGROUND : Gastric cancer (GC) is one of the most common malignant cancers, with high morbidity and mortality rates worldwide. The present study was to explore whether miR-200b is a tumor suppressor in GC and to unveil the potential mechanisms. METHODS: Levels of c-Myc, Cyclin D1, MMP-3 and MMP-9 expression were detected respectively by qRT-PCR and Western blot assay. BrdU proliferation assay, Cell cycle analysis, Wound-healing and Transwell assays were used to study the role of miR-200b with inhibitor, mimics or ZEB1-RNAi in TGF-β1-treated SGC-7901/DDP cells. The xenograft model with nude mice was established to unveil the role of miR-200b in vivo . RESULTS : Compared with the paracancerous tissues, miR-200b was decreased in GC patients and SGC-7901/DDP cells. Lower level of miR-200b induced by its inhibitor promoted TGF-β1-treated SGC-7901/DDP cells proliferation and migration, and increased the levels of c-Myc, Cyclin D1, MMP-3, MMP-9, β-catenin and APC. Interestingly, miR-200b mimics and ZEB1-RNAi were able to reduce the proliferation and migration of TGF-β1-induced SGC-7901/DDP cells as well as their levels of c-Myc, Cyclin D1, MMP-3 MMP-9, β-catenin and APC. In addition, ZEB1 was indeed the potential target of miR-200b identified by dual luciferase reporter gene assay. Xenograft model also suggested that over-expression of miR-200b suppressed the growth of tumor in vivo. CONCLUSION : Taken together, our findings suggest that miR-200b be likely to play an important role in activating TGF-β1-induced SGC-7901/DDP cells and perform as a tumor suppressor by targeting ZEB1 in GC. What’s more, miR-200b may modulate Wnt/β-catenin signaling pathway in TGF-β1-induced SGC-7901/DDP cells.


Author(s):  
Zizhen Si ◽  
Lei Yu ◽  
Haoyu Jing ◽  
Lun Wu ◽  
Xidi Wang

Abstract Background Long non-coding RNAs (lncRNA) are reported to influence colorectal cancer (CRC) progression. Currently, the functions of the lncRNA ZNF561 antisense RNA 1 (ZNF561-AS1) in CRC are unknown. Methods ZNF561-AS1 and SRSF6 expression in CRC patient samples and CRC cell lines was evaluated through TCGA database analysis, western blot along with real-time PCR. SRSF6 expression in CRC cells was also examined upon ZNF561-AS1 depletion or overexpression. Interaction between miR-26a-3p, miR-128-5p, ZNF561-AS1, and SRSF6 was examined by dual luciferase reporter assay, as well as RNA binding protein immunoprecipitation (RIP) assay. Small interfering RNA (siRNA) mediated knockdown experiments were performed to assess the role of ZNF561-AS1 and SRSF6 in the proliferative actives and apoptosis rate of CRC cells. A mouse xenograft model was employed to assess tumor growth upon ZNF561-AS1 knockdown and SRSF6 rescue. Results We find that ZNF561-AS1 and SRSF6 were upregulated in CRC patient tissues. ZNF561-AS1 expression was reduced in tissues from treated CRC patients but upregulated in CRC tissues from relapsed patients. SRSF6 expression was suppressed and enhanced by ZNF561-AS1 depletion and overexpression, respectively. Mechanistically, ZNF561-AS1 regulated SRSF6 expression by sponging miR-26a-3p and miR-128-5p. ZNF561-AS1-miR-26a-3p/miR-128-5p-SRSF6 axis was required for CRC proliferation and survival. ZNF561-AS1 knockdown suppressed CRC cell proliferation and triggered apoptosis. ZNF561-AS1 depletion suppressed the growth of tumors in a model of a nude mouse xenograft. Similar observations were made upon SRSF6 depletion. SRSF6 overexpression reversed the inhibitory activities of ZNF561-AS1 in vivo, as well as in vitro. Conclusion In summary, we find that ZNF561-AS1 promotes CRC progression via the miR-26a-3p/miR-128-5p-SRSF6 axis. This study reveals new perspectives into the role of ZNF561-AS1 in CRC.


2021 ◽  
Vol 12 (4) ◽  
Author(s):  
Xiaoguang Gu ◽  
Jianan Zhang ◽  
Yajuan Ran ◽  
Hena Pan ◽  
JinHong Jia ◽  
...  

AbstractCircular RNAs have been reported to play significant roles in regulating pathophysiological processes while also guiding clinical diagnosis and treatment of hepatocellular carcinoma (HCC). However, only a few circRNAs have been identified thus far. Herein, we investigated the role of a specific closed-loop structure of hsa_circ_101555 that was generated by back-splicing of the host gene casein kinase 1 gamma 1 (CSNK1G1) in the development and proliferation of HCC. We investigated the expression of Hsa_circ_101555 in HCC and normal tissues using bioinformatics. The expression level of hsa_circ_101555 was further detected by fluorescence in situ hybridization and qRT-PCR in ten HCC patients. Transwell, migration, WST-1 assays, and colony formation assays were used to evaluate the role of hsa_circ_101555 in HCC development and proliferation. The regulatory mechanisms of hsa_circ_101555 in miR-145-5p and CDCA3 were determined by dual luciferase reporter assay. A mouse xenograft model was also used to determine the effect of hsa_circ_101555 on HCC growth in vivo. hsa_circ_101555 showed greater stability than the linear RNA; while in vitro and in vivo results demonstrated that hsa_circ_101555 silencing significantly suppressed cell proliferation, migration, and invasion of HCC cells. Rescue experiments further demonstrated that suppression of miR-145-5p significantly attenuated the biological effects of hsa_circ_101555 knockdown in HCC cells. We also identified a putative oncogene CDCA3 as a potential miR-145-5p target. Thus, our results demonstrated that hsa_circ_101555 might function as a competing endogenous RNA of miR-145-5p to upregulate CDCA3 expression in HCC. These findings suggest that hsa_circ_101555 may be a potential therapeutic target for patients with HCC.


2021 ◽  
Vol 21 (1) ◽  
Author(s):  
Jiong Ma ◽  
Chunxia Zhou ◽  
Xuejun Chen

Abstract Background Hedgehog (Hh) signaling pathway, which is essential for cell proliferation and differentiation, is noted to be aberrantly activated in tumor from increasing studies in recent years. MicroRNAs (miRNAs) as an important non-coding RNA in cells have been proven to possess a regulatory role specific to the Hh signaling pathway. Here, in vitro and in vivo cellular/molecular experiments were adopted to clarify the regulatory mechanism linking miR-636 to the Hh signaling pathway in ovarian cancer (OVC). Methods Protein–protein interaction analysis was performed to identify the hub gene in the Hh pathway. TargetScan database was used to predict the potential upstream regulators for Gli2. qRT-PCR was performed to test the expression of miR-636, while Western blot was conducted to detect the expression of proteins related to the Hh pathway and epithelial-mesenchymal transition (EMT). For cell functional experiments, HO-8910PM OVC cell line was used. MTT assay and wound healing assay were used to measure the effect of miR-636 on cell proliferation and migration. Flow cytometry was carried out to examine the effect of miR-636 on cell cycle, and Western blot was used to identify the change in expression of Hh and EMT-related proteins. Dual-luciferase reporter gene assay was implemented to detect the targeting relationship between miR-636 and Gli2. Xenotransplantation models were established for in vivo examination. Results Gli2 was identified as the hub gene of the Hh pathway and it was validated to be regulated by miR-636 based on the data from TargetScan and GEO databases. In vitro experiments discovered that miR-636 was significantly lowly expressed in OVC cell lines, and overexpressing miR-636 significantly inhibited HO-8910PM cell proliferation, migration and induced cell cycle arrest in G0/G1 phase, while the inhibition of miR-636 caused opposite results. Dual-luciferase reporter gene assay revealed that Gli2 was the target gene of miR-636 in OVC. Besides, overexpressed miR-636 decreased protein expression of Gli2, and affected the expression of proteins related to the Hh signaling pathway and EMT. Rescue experiments verified that overexpression of Gli2 reversed the inhibitory effect of miR-636 on HO-8910PM cell proliferation and migration, and attenuated the blocking effect of miR-636 on cell cycle. The xenotransplantation experiment suggested that miR-636 inhibited cell growth of OVC by decreasing Gli2 expression. Besides, overexpressing Gli2 potentiated the EMT process of OVC cells via decreasing E-cadherin protein expression and increasing Vimentin protein expression, and it reversed the inhibitory effect of miR-636 on OVC cell proliferation in vivo. Conclusion miR-636 mediates the activation of the Hh pathway via binding to Gli2, thus inhibiting EMT, suppressing cell proliferation and migration of OVC. Trial registration: The experimental protocol was established, according to the ethical guidelines of the Helsinki Declaration and was approved by the Human Ethics Committee of The Second Affiliated hospital of Zhejiang University School of Medicine (IR2019001235). Written informed consent was obtained from individual or guardian participants.


Sign in / Sign up

Export Citation Format

Share Document