scholarly journals Circulating microRNA-762 upregulates colorectal cancer might through Wnt-1/β-catenin signaling

2020 ◽  
Author(s):  
Peng-Sheng Lai ◽  
Wei-Min Chang ◽  
Ying-Yin Chen ◽  
Yi-Feng Lin ◽  
Hui-Fen Liao ◽  
...  

ABSTRACTBackgroundA number of microRNAs (miRNAs) have been demonstrated to be correlated with the diagnosis, progression and prognosis of colorectal cancer (CRC). However, the key miRNAs and the associated signaling pathways that regulate the growth and metastasis of CRC remain unclear.MethodsThe circulating miRNAs from BALB/c mice with CRC CT26 cell implantation were assayed by microarray. Then, mmu-miR-762 mimic and inhibitor were transfected to CT26 cells for analysis of cell viability, invasion, and epithelial-mesenchymal transition (EMT), cell cycle, and regulatory molecule expression. Human subjects were included for comparison the circulating has-miR-762 levels in CRC patients and control donors, as well as the patients with and without distant metastasis.ResultsThe miRNA levels in mice with CRC cell implantation indicated that plasma mmu-miR-762 was upregulated. Transfection of mmu-miR-762 mimic to CT26 cells increased cell viability, invasion, and EMT, whereas transfection of mmu-miR-762 inhibitor decreased the above abilities. Cells treated with high-concentration mmu-miR-762 inhibitor induced cell cycle arrest at G0/G1 phase. Western blot analysis showed that mmu-miR-762 mimic transfection upregulated the expression of Wnt-1 and β-catenin. Further analysis was performed to demonstrate the correlation of has-miR-762 with CRC patients. The results showed that serum has-miR-762 levels in CRC patients were higher than in control donors. Among the CRC patients, patients with distant metastasis showed higher serum has-miR-762 levels than patients without distant metastasis.ConclusionsThe present study demonstrated that circulating miR-762 might be a biomarker with upregulation of CRC cell growth and invasion through the Wnt/β-catenin signaling.

2019 ◽  
Author(s):  
Peng-Sheng Lai ◽  
Wei-Min Chang ◽  
Ying-Yin Chen ◽  
Yi-Feng Lin ◽  
Hui-Fen Liao ◽  
...  

Abstract Background: A number of microRNAs (miRNAs) have been demonstrated to be associated with the diagnosis, progression and prognosis of colorectal cancer (CRC). However, the function of miRNA-762 (miR-762) in CRC remains unclear, and the molecular mechanisms underlying the effects of miR‑762 in CRC require further investigation. Methods: The circulating miRNAs from BALB/c mice with CRC CT26 cell implantation were assayed by microarray. Then, miR-762 mimic and inhibitor were transfected to CT26 cells for analysis of cell viability, invasion, and epithelial-mesenchymal transition (EMT), cell cycle, and regulatory molecule expression. Human subjects were included for comparison the circulating miR-762 levels in CRC patients and control donors, as well as the patients with and without distant metastasis. Results: The screening for miRNA levels in mice with CRC cell implantation indicated that plasma miR-762 was upregulated. Transfection of miR-762 mimic to CT26 cells increased cell viability, invasion, and EMT, whereas transfection of miR-762 inhibitor decreased the above abilities. Western blot analysis showed that miR-762 mimic transfection upregulated the expression of Wnt-1 and b-catenin, as well as increased the nuclear translocation of b-catenin. Further analysis showed that serum miR-762 levels in CRC patients were higher than in control donors. Among the CRC patients (n = 20), six patients with distant metastasis showed higher serum miR-762 levels than the patients without distant metastasis. Conclusions: Circulating miR-762 could promote CRC disease development and progression through the Wnt/b-catenin signaling. miR-762 might be used as a biomarker for CRC diagnosis and targeted therapy.


2021 ◽  
pp. 1-12
Author(s):  
Peng-Sheng Lai ◽  
Wei-Min Chang ◽  
Ying-Yin Chen ◽  
YiFeng Lin ◽  
Hui-Fen Liao ◽  
...  

Colorectal cancer (CRC) has become the third most common cause of cancer-related deaths. CRC occurs because of abnormal growth of cells that can invade other tissues and cause distant metastases. Researchers have suggested that aberrant microRNA (miRNA) expression is involved in the initiation and progression of cancers. However, the key miRNAs that regulate the growth and metastasis of CRC remain unclear. The circulating miRNAs from BALB/c mice with CRC CT26 cell implantation were assayed by microarray. Then, Mus musculus (house mouse) mmu-miR-762 mimic and inhibitor were transfected to CT26 cells for analysis of cell viability, invasion, and epithelial-mesenchymal transition (EMT), cell cycle, and regulatory molecule expression. Human subjects were included for comparison the circulating Homo sapiens (human) has-miR-762 levels in CRC patients and control donors, as well as the patients with and without distant metastasis. The result for miRNA levels in mice with CRC cell implantation indicated that plasma mmu-miR-762 was upregulated. Transfection of mmu-miR-762 mimic to CT26 cells increased cell viability, invasion, and EMT, whereas transfection of mmu-miR-762 inhibitor decreased the above abilities. Cells treated with high-concentration mmu-miR-762 inhibitor induced cell cycle arrest at G0/G1 phase. However, mmu-miR-762 did not cause apoptosis of cells. Western blot analysis showed that mmu-miR-762 mimic transfection upregulated the expression of Wnt-1 and β-catenin, as well as increased the nuclear translocation of β-catenin. Further analysis was performed to demonstrate the correlation of miR-762 with CRC, and blood samples were collected from CRC patients and control donors. The results showed that serum has-miR-762 levels in CRC patients were higher than in control donors. Among the CRC patients (n= 20), six patients with distant metastasis showed higher serum has-miR-762 levels than patients without distant metastasis. Conclusions, the present study suggests that circulating miR-762 might be a potential biomarker for upregulation of CRC cell growth and invasion, and may be accompanied by the Wnt/β-catenin signaling.


2020 ◽  
Author(s):  
Peng-Sheng Lai ◽  
Wei-Min Chang ◽  
Ying-Yin Chen ◽  
Yi-Feng Lin ◽  
Hui-Fen Liao ◽  
...  

Abstract Background A number of microRNAs (miRNAs) have been demonstrated to be correlated with the diagnosis, progression and prognosis of colorectal cancer (CRC). However, the key miRNAs and the associated signaling pathways that regulate the growth and metastasis of CRC remain unclear. Methods miRNA array was analyzed in CRC CT26 cell-transplanted BALB/c mice. The effects of miR-762 mimics and inhibitors on the growth, invasion, cell cycle, and regulatory pathways of CRC CT26 cells were assessed. Cell cycle and sub-G1 assay were collected from the treated CT26 cells. Finally, blood samples were collected from patients with CRC. Further analysis to compare miR-762 levels in blood samples collected from CRC patients and normal control donors. Patients’ basic data were retrieved from electronic medical records and analyzed for age, gender, tumor staging, and survival. Results The miRNA levels in mice with CRC cell implantation indicated that plasma mmu-miR-762 was upregulated. Transfection of mmu-miR-762 mimic to CT26 cells increased cell viability, invasion, and EMT, whereas transfection of mmu-miR-762 inhibitor decreased the above abilities. Cells treated with high-concentration mmu-miR-762 inhibitor induced cell cycle arrest at G0/G1 phase. However, mmu-miR-762 did not cause apoptosis of cells. Western blot analysis showed that mmu-miR-762 mimic transfection upregulated the expression of Wnt-1 and β-catenin, as well as increased the nuclear translocation of β-catenin. Further analysis was performed to demonstrate the correlation of has-miR-762 with CRC, and blood samples were collected from CRC patients and control donors. The results showed that serum has-miR-762 levels in CRC patients were higher than in control donors. Among the CRC patients, patients with distant metastasis showed higher serum has-miR-762 levels than patients without distant metastasis. Conclusions The present study demonstrated that circulating miR-762 might be a biomarker with upregulation of CRC cell growth and invasion through the Wnt/β-catenin signaling.


2020 ◽  
Author(s):  
Zhenxian Xiang ◽  
Guoquan Huang ◽  
Haitao Wu ◽  
Qiuming He ◽  
Chaogang Yang ◽  
...  

Abstract Background: Circulating tumor cells are important precursor of colorectal cancer metastasis, which attributes to the main cause of cancer-related death. The ability to adopt epithelial-mesenchymal transition (EMT) process facilitates CTCs generation, thereby overcoming metastatic bottlenecks and realizing distant metastasis. However, the potential molecular mechanism of CRC EMT remains largely unknown.Methods: RT-qPCR, immunohistochemical staining, and western blot were used to detect the expression of mRNA and protein in CRC. Loss- and gain-of-function approaches were performed to investigate the effect of SNHG16 on CRC cell phenotypes. Function assays, including wounding healing, transwell assay, and clone formation were used to assess the effect of SNHG16 on tumor biological behavior. Then, RNA immunoprecipitation, Chromatin Immunoprecipitation, Co-Immunoprecipitation, GST-pull down, biotin-labeled miR-195-5p pull down, and dual-luciferase assay were performed to uncover the underlying mechanism for molecular interaction. Finally, CRC nude mice xenograft model experiment was performed to evaluate the influence of SNHG16 on tumor progression in vivo Results: Compared with normal tissue and cell line, SNHG16 was significantly upregulated in CRC. Clinical investigation revealed that SNHG16 high expression was correlated with advanced TNM stage, distant metastasis, and poor prognosis of cancer patients. According to Loss- and gain-of-function experiment, SNHG16 could promote CRC proliferation, migration, invasion, EMT, mesenchymal-type CTCs (MCTCs) generation, and liver metastasis through YAP1 in vitro and in vivo. Mechanistic research indicates that, SNHG16 could act as miRNA sponge to sequester miR-195-5p on Ago2, thereby protecting YAP1 from repression and facilitating CRC liver metastasis and tumor progression. Moreover, YAP1 could combine with TEA Domain Transcription Factor 1 (TEAD1) to form a YAP1/TEAD1 complex, which could in turn bind to the promoter of SNHG16 and regulate its transcription. In addition, both of YAP1 and TEAD1 are indispensable during this process. Finally, we demonstrated that YAP1 significantly promoted the tumor progression, and SNHG16 could rescue the effect of YAP1 on tumor progressionConclusion: Herein, we clarified a hitherto unexplored positive feedback loop between SNHG16 and YAP1/TEAD1. These findings provided new sights in CRC liver metastasis, and it may act as a potential candidate in the treatment of CRC.


2022 ◽  
Author(s):  
Jiayan Wu ◽  
Hongquan Zhu ◽  
Jiandong Yu ◽  
Zhiping Chen ◽  
Zeyu Lin ◽  
...  

Abstract OBJECTIVE: Long non-coding RNA HOXB-AS3 has been implicated in tumor progression in a variety of carcinomas. However, its biological role in gallbladder cancer (GBC) is unknown. The biological function and underlying mechanism of the lncRNA HOXB-AS3 for GBC were investigated in this study.MATERIALS AND METHODS: To investigate the function of lncRNA HOXB-AS3 in GBC, the level of lncRNA HOXB-AS3 in GBC cells was detected by quantitative reverse-transcription polymerase chain reaction. The cell viability was tested by cell counting kit-8 assay and colony formation assay. Flow cytometry was performed to investigate cell apoptosis and cell cycle. In addition, cell migration ability was assessed by wound healing assay and cell invasion ability by transwell invasion assay. RESULTS: It was found that HOXB-AS3 was obviously elevated in GBC tissues and cells. However, inhibition of HOXB-AS3 could depress NOZ and GBC-SD cell viability as well as induce cell apoptosis. Also, the gallbladder cancer cell cycle was blocked in the G1 phase. Meanwhile, NOZ and GBC-SD cell migration, invasion, and epithelial-mesenchymal transition were obviously suppressed by knockdown of HOXB-AS3. What is more, we found that HOXB-AS3 might promote gallbladder progress by activating the MEK/ERK pathway.CONCLUSION: The results show that lncRNA HOXB-AS3 serves as a key regulator in GBC progression, which provides a new treatment strategy for GBC.


2019 ◽  
Vol 2019 ◽  
pp. 1-10 ◽  
Author(s):  
Ling Zhang ◽  
Abid Naeem ◽  
Shaofeng Wei ◽  
Zexie Li ◽  
Zhenzhong Zang ◽  
...  

The current study investigates the inhibitory effects of Pulsatilla pentacyclic triterpenoid saponins extract (PPTS) on epithelial-mesenchymal transition (EMT) triggered by the transforming growth factor-β1 (TGF-β1) in human colorectal cancer SW480 cell line, further illustrates the possible mechanism of PPTS inhibition of growth and invasion from the perspective of EMT, and provides new theoretical support for the treatment of tumor by Chinese medicine. The SW480 cells were treated in groups: blank control, TGF-β1 (10 ng/mL), and varying concentrations of PPTS cotreated with TGF-β1-induced (10 ng/mL) groups. CCK8 was used to detect cell viability; transwell was applied to detect invasion ability, cell migration ability was also determined, ELISA and RT-qPCR were utilized for the determination of CYP3A, CYP2C9, CYP2C19, N-cadherin, and MMP-9 expression. Flow cytometry detection was applied to detect cell cycle and apoptosis. The results obtained have shown that PPTS can significantly inhibit the invasion and migration of tumors in SW480 cells and can also block the S phase in the cell cycle but may produce cytotoxicity in higher doses. The present research work provides substantial evidence that PPTS has a significant inhibitory effect on TGF-β1-induced EMT in SW480 cells and it also promotes apoptosis.


2021 ◽  
Author(s):  
Zhi-Hao Zhong ◽  
Yu-Qing Zhang

Abstract Mulberry tree branches are one of the largest agro-wastes produced in silk industry. How to make full use of this waste has always been one of the most important issues for the silk industry and even the entire biological industry. The paper has first reported that the inhibition of morusin recovered from mulberry branch barks, a prenylated flavonoid, on 20 kinds of tumour cells, of which the IC50 values of the 80% cells reaches about 15 µM. Second, effects on the proliferation, invasion and apoptosis of two cancer cells were investigated in detail. The experimental results showed that the apoptotic ratio of the high concentration was 77.73% in MDA-MB-453 cells. Western blotting displayed that morusin upregulated E-cadherin and downregulated vimentin and N-cadherin in a dose-dependent manner, and thus reversed epithelial-mesenchymal transition. It could upregulate cleaved Caspase-3 and Bax and downregulate Caspase-3 and Bcl-2, which indicate that the cell apoptosis is induced by morusin. These cancer cells, MDA-MB-453, were blocked in G2 phase, and HCT116 were arrested in S phase when treated with morusin, which is possible that the cell cycle is disturbed. Therefore, morusin could inhibit cancer migration and growth and promote cancer cell apoptosis.


Marine Drugs ◽  
2018 ◽  
Vol 16 (8) ◽  
pp. 252 ◽  
Author(s):  
Li-Chun Lin ◽  
Tzu-Ting Kuo ◽  
Hsin-Yi Chang ◽  
Wen-Shan Liu ◽  
Shih-Min Hsia ◽  
...  

Marine sponges are known to produce numerous bioactive secondary metabolites as defense strategies to avoid predation. Manzamine A is a sponge-derived β-carboline-fused pentacyclic alkaloid with various bioactivities, including recently reported anticancer activity on pancreatic cancer. However, its cytotoxicity and mode of action against other tumors remain unclear. In this study, we exhibit that manzamine A reduced cell proliferation in several colorectal cancer (CRC) cell lines. To further investigate the manzamine A triggered molecular regulation, we analyzed the gene expression with microarray and revealed that pathways including cell cycle, DNA repair, mRNA metabolism, and apoptosis were dysregulated. We verified that manzamine A induced cell cycle arrest at G0/G1 phase via inhibition of cyclin-dependent kinases by p53/p21/p27 and triggered a caspase-dependent apoptotic cell death through mitochondrial membrane potential depletion. Additionally, we performed bioinformatics analysis and demonstrated that manzamine A abolished epithelial–mesenchymal transition process. Several mesenchymal transcriptional factors, such as Snail, Slug, and Twist were suppressed and epithelial marker E-cadherin was induced simultaneously in HCT116 cells by manzamine A, leading to the epithelial-like phenotype and suppression of migration. These findings suggest that manzamine A may serve as a starting point for the development of an anticancer drug for the treatment of metastatic CRC.


2016 ◽  
Vol 6 (1) ◽  
Author(s):  
Hugh Colvin ◽  
Naohiro Nishida ◽  
Masamitsu Konno ◽  
Naotsugu Haraguchi ◽  
Hidekazu Takahashi ◽  
...  

2021 ◽  
Vol 21 (1) ◽  
Author(s):  
Jia-Lu Zheng ◽  
Shuang-Shuang Wang ◽  
Ke-Ping Shen ◽  
Lei Chen ◽  
Xiao Peng ◽  
...  

Abstract Background Ursolic acid (UA) is an anti-cancer herbal compound. In the present study, we observed the effects of UA on anchorage-dependent and -independent growth of human colorectal cancer (CRC) RKO cells. Methods RKO cells were cultured in conventional and detached condition and treated with UA. Cell viability was evaluated by CCK-8 assay. Cell cycle was analyzed by flow cytometry. Apoptosis was identified by Hoechst 33258 staining and flow cytometry analysis. Activities of caspases were measured by commercial kits. Reactive oxygen species (ROS) was recognized by DCFH-DA fluorescent staining. Anoikis was identified by EthD-1 fluorescent staining and flow cytometry analysis. Expression and phosphorylation of proteins were analyzed by western blot. Results UA inhibited RKO cell viability in both a dose- and time-dependent manner. UA arrested the cell cycle at the G0/G1 phase, and induced caspase-dependent apoptosis. UA inhibited Bcl-2 expression and increased Bax expression. In addition, UA up-regulated the level of ROS that contributed to UA activated caspase-3, − 8 and − 9, and induced apoptosis. Furthermore, UA inhibited cell growth in a detached condition and induced anoikis in RKO cells that was accompanied by dampened phosphorylation of FAK, PI3K and AKT. UA also inhibited epithelial-mesenchymal transition (EMT) as indicated by the down-regulation of N-Cad expression and up-regulation of E-Cad expression. Conclusions UA induced caspase-dependent apoptosis, and FAK/PI3K/AKT singling and EMT related anoikis in RKO cells. UA was an effective anti-cancer compound against both anchorage-dependent and -independent growth of RKO cells.


Sign in / Sign up

Export Citation Format

Share Document