scholarly journals Effect of UBE2Q on BECN and CEA Protein in Colorectal Cancer Cells

Author(s):  
Niloofar Boroumand

Background: Expression of UBE2Q1, UBE2Q2, and members of the ubiquitinconjugating enzyme family (E2) are affected in colorectal cancer (CRC). The BECN gene plays a key role in CRC cells. In gastrointestinal carcinoma therapy, tumorassociated antigens such as CEA are typically used.To investigate the association between UBE2Q1 and Beclin1 autophagy marker and CEA protein expression in LS180 CRC cell line. Materials and Methods: In this study, changes in the expression of BECN marker in LS180 cell lines with the vector containing UBE2Q1 were investigated using real-time PCR. The expression of CEA protein was also evaluated by western blotting. Statistical analyses were performed with Graph Pad Prism software. Results: The results indicated reduced expression of BECN autophagy marker (P=0). Therefore, in the presence of UBE2Q1, cancer cells have less ability to induce autophagy. However, CEA protein levels in LS180 transfected cells with a UBE2Q1- ORF-containing plasmid decreased when compared to non-transfected cells. Conclusion: The use of pharmacologic inhibitors related to the autophagy mechanism can be a novel approach in cancer therapy.

2021 ◽  
Author(s):  
Bin Chen ◽  
Haijuan Xiao ◽  
Linguangjin Wu ◽  
Ting Wang ◽  
Shuyun Wang ◽  
...  

Abstract Background This study was intended to investigate the function of Quercetin in chemoresistant colorectal cancer (CRC) cells. In addition, this research aimed to explore the mechanism by which Quercetin regulates the malignant behavior of CRC cells. Methods To induce THP-1 cells into M2 tumor-associated macrophages (M2-TAMs), THP-1 cells were stimulated by PMA and IL-4. MDC staining was used to investigate the autophagy in M2-TAMs. Meanwhile, cell proliferation was tested by colony formation assay. In addition, wound healing and transwell assay were performed to detect the cell migration and invasion, respectively. Dual luciferase assay was used to investigate the correlation between hsa_circ_0006990 and miR-132-3p/miR-532-3p. Furthermore, mRNA and protein levels were detected by RT-qPCR and western blot, respectively. Results Quercetin suppressed autophagy of M2-TAMs. In addition, M2-TAMs significantly inhibited the apoptosis and promoted the proliferation of CRC cells, while this phenomenon was reversed by Quercetin. Meanwhile, the expression of hsa_circ_0006990 in CRC cells was decreased by M2-TAMs, while Quercetin reversed this phenomenon. Furthermore, overexpression of hsa_circ_0006990 significantly reversed the anti-tumor effect of Quercetin on CRC. Conclusion Quercetin inhibited the tumorigenesis of colorectal cancer cells through downregulation of hsa_circ_0006990. Thus, our study might shed new lights on exploring the new strategies against CRC.


2018 ◽  
Vol 46 (2) ◽  
pp. 492-504 ◽  
Author(s):  
Xiangyuan Chen ◽  
Qichao Wu ◽  
Pengfei Sun ◽  
Yanjun Zhao ◽  
Minmin Zhu ◽  
...  

Background/Aims: To investigate the effect of propofol on glucose metabolism in colorectal cancer cells and in an in vivo xenograft model. Methods: Glucose metabolism was assessed by measuring the extracellular acidification rate in HT29 and SW480 colorectal cancer cells. Quantitative real-time PCR and western blot analyses were used to detect mRNA and protein levels, respectively. Intracellular calcium was assessed by using a Fluo-3 AM fluorescence kit. Micro-positron emission tomography/computed tomography (microPET/CT) imaging was used to analyze glucose metabolism in the tumors of the xenograft model. Results: Propofol exposure induced a dose-dependent decrease of aerobic glycolysis in HT29 and SW480 colorectal cancer cells. MicroPET/CT indicated that propofol also inhibited 18F-FDG uptake in the xenograft model. In addition, hypoxia-inducible factor 1α (HIF1α) was also reduced by propofol dose-dependently. Propofol repressed the NMDAR-CAMKII-ERK pathway to inactivate HIF1α and therefore reduced glycolysis. Conclusion: Propofol inhibited aerobic glycolysis in colorectal cancer cells through the inactivation of the NMDAR-CAMKII-ERK pathway, which may facilitate a better understanding of the use of propofol in the clinical setting.


2018 ◽  
Vol 47 (2) ◽  
pp. 680-693 ◽  
Author(s):  
Shu Gong ◽  
Dongsheng Xu ◽  
Jialin Zhu ◽  
Fangdong Zou ◽  
Rui Peng

Background/Aims: Mutations in the Ras/Raf/MEK/ERK pathway are detected in 50% of colorectal cancer cases and play a crucial role in cancer development and progression. Cobimetinib is a MEK inhibitor approved for the treatment of advanced melanoma and inhibits the cell viability of other types of cancer cells. Methods: HCT116 colorectal cancer cells were treated with cobimetinib, and MTT assay, colony formation assay, and flow cytometry were used to evaluate cell viability, cell cycle, and apoptosis, respectively. The expression of genes associated with the cell cycle and apoptosis were evaluated by quantitative real-time PCR and western blotting. To explore use of cobimetinib in colorectal cancer treatment and further understand its mechanisms, RNA-seq technology was used to identify differentially expressed genes (DEGs) between cobimetinib-treated and untreated HCT116 cells. Furthermore, we compared these DEGs with Gene Expression Omnibus data from colorectal cancer tissues and normal colonic epithelial tissues. Results: We found that cobimetinib not only inhibited cell proliferation but also induced G1 phase arrest and apoptosis in HCT116 colorectal cancer cells, suggesting that cobimetinib may useful in colorectal cancer therapy. After cobimetinib treatment, 3,495 DEGs were obtained, including 2,089 upregulated genes and 1,406 downregulated genes, and most of these DEGs were enriched in the cell cycle, DNA replication, and DNA damage repair pathways. Our results revealed that some genes with high expression in colorectal cancer tissues were downregulated by cobimetinib in HCT116 cells, including CCND1, E2F1, CDC25C, CCNE2, MYC, and PCNA. These genes have vital roles in DNA replication and the cell cycle. Furthermore, genes with low expression in colorectal cancer tissues were upregulated by cobimetinib, including PRKCA, PI3K, RTK, and PKC. Based on our results, the PKC and PI3K pathways were activated after cobimetinib treatment, and inhibition of these two pathways can increase the cytotoxicity of cobimetinib in HCT116 cells. Notably, cobimetinib appeared to enhance the efficacy of 5-fluorouracil (5-FU) by decreasing TYMS expression, high expression of which is responsible for 5-FU resistance in colorectal cancer. Conclusions: Our results suggest the potential use of cobimetinib in colorectal cancer therapy.


Molecules ◽  
2018 ◽  
Vol 23 (11) ◽  
pp. 2968 ◽  
Author(s):  
Yi Yang ◽  
Suresh Bhosle ◽  
Young Yu ◽  
So-Yeon Park ◽  
Rui Zhou ◽  
...  

Lichens produce various unique chemicals that are used in the pharmaceutical industry. To screen for novel lichen secondary metabolites that inhibit the stemness potential of colorectal cancer cells, we tested acetone extracts of 11 lichen samples collected in Chile. Tumidulin, isolated from Niebla sp., reduced spheroid formation in CSC221, DLD1, and HT29 cells. In addition, mRNA expressions and protein levels of cancer stem markers aldehyde dehydrogenase-1 (ALDH1), cluster of differentiation 133 (CD133), CD44, Lgr5, and Musashi-1 were reduced after tumidulin treatment. Tumidulin decreased the transcriptional activity of the glioma-associated oncogene homolog zinc finger protein (Gli) promoter in reporter assays, and western blotting confirmed decreased Gli1, Gli2, and Smoothened (SMO) protein levels. Moreover, the tumidulin activity was not observed in the presence of Gli and SMO inhibitors. Together, these results demonstrate for the first time that tumidulin is a potent inhibitor of colorectal cancer cell stemness.


2020 ◽  
Vol 11 (11) ◽  
Author(s):  
Yong Xia ◽  
Shuzhi Liu ◽  
Changlin Li ◽  
Zhiying Ai ◽  
Wenzhi Shen ◽  
...  

AbstractFerropotsis is among the most important mechanisms of cancer suppression, which could be harnessed for cancer therapy. However, no natural small-molecule compounds with cancer inhibitory activity have been identified to date. In the present study, we reported the discovery of a novel ferroptosis inducer, talaroconvolutin A (TalaA), and the underlying molecular mechanism. We discovered that TalaA killed colorectal cancer cells in dose-dependent and time-dependent manners. Interestingly, TalaA did not induce apoptosis, but strongly triggered ferroptosis. Notably, TalaA was significantly more effective than erastin (a well-known ferroptosis inducer) in suppressing colorectal cancer cells via ferroptosis. We revealed a dual mechanism of TalaA’ action against cancer. On the one hand, TalaA considerably increased reactive oxygen species levels to a certain threshold, the exceeding of which induced ferroptosis. On the other hand, this compound downregulated the expression of the channel protein solute carrier family 7 member 11 (SLC7A11) but upregulated arachidonate lipoxygenase 3 (ALOXE3), promoting ferroptosis. Furthermore, in vivo experiments in mice evidenced that TalaA effectively suppressed the growth of xenografted colorectal cancer cells without obvious liver and kidney toxicities. The findings of this study indicated that TalaA could be a new potential powerful drug candidate for colorectal cancer therapy due to its outstanding ability to kill colorectal cancer cells via ferroptosis induction.


2020 ◽  
Author(s):  
Cong Tian ◽  
Tingyuan Lang ◽  
Jiangfeng Qiu ◽  
Kun Han ◽  
Lei Zhou ◽  
...  

Abstract Background: Cancer stem cells (CSCs) have been recognized as an important drug target, however, the underlying mechanisms have not been fully understood. SKP1 is a traditional drug target for cancer therapy, while, whether SKP1 promotes colorectal cancer (CRC) stem cells (CRC-SCs) and the underlying mechanisms have remained elusive.Methods: Human CRC cell lines HCT-116 and HT-29 and primary human colorectal cancer cells were used in this study. Gene manipulation was performed by lentivirus system. The mRNA and protein levels were examined by qRT-PCR and western blot, respectively. Sphere formation and transwell assay were employed for examination of sphere-forming and migration capacities. The self-renewal capacity was determined by limiting dilution assay. The tumorigenicity was examined by xenograft model. The transcriptional activities of the promoters were examined by luciferase reporter assay. Co-immunoprecipitation assay was used to test protein-protein interaction. The transcription and protein-DNA interaction were examined by nuclear run-on and ChIP-PCR assay. The relationship between gene expression and survival was analyzed by Kaplan-meier analysis. The correlation between two genes was analyzed by Spearman analysis. Data are represented as mean ± s.d. and the significance was determined by Student’s t-test.Results: SKP1 is upregulated in colorectal cancer stem cells and predicts poor prognosis of colon cancer patients. Overexpression of SKP1 promotes the sphere-forming and migration capacities as well as self-renewal of CRC cells, and upregulates the expression of CSCs markers. In contrast, SKP1 depletion produces the opposite effects. SKP1 strengthens YAP activity and knockdown of YAP abolished the effect of SKP1 on the stemness of colorectal cancer cells. SKP1 suppresses RASSF1 at both mRNA and protein levels and overexpression of RASSF1 abolished the effect of SKP1.Conclusion: Our results demonstrated that SKP1 suppresses RASSF1 at both mRNA and protein level, attenuates Hippo signaling, activates YAP, and thereby promoting the stemness of CRC cells. Our works thus revealed a novel underlying mechanism of CRC-SCs maintenance and suggested a novel drug target for eradicating CRC-SCs.


2021 ◽  
Vol 20 (11) ◽  
pp. 2333-2338
Author(s):  
Li Peng ◽  
Lin Yu ◽  
Chen Yong-Le

Purpose: To determine the effect of polysaccharides from Gleditsia on apoptosis of colorectal cancer cells, and the mechanism involved.Methods: Polysaccharides were extracted from Lycium barbarum, and their concentration was more than 85 %. Then, DLD-1 cells were cultured in medium with the polysaccharides at concentrations of 75 and 150 μg/mL. Cell proliferation was determined with MTT (3-(4,5)-dimethylthiahiazo (-z-y1)-3,5- diphenytetrazoliumromide) assay and colony formation assay, while apoptosis was determined with flow cytometry. Changes in MMP were measured flow cytometrically. The protein levels of PARP, Bcl-2, Bax, and caspases 3 and 9 were determined with Western blot assay.Results: Cell viability decreased time-dependently. Compared with control without polysaccharide exposure, cell viability, colony forming cells, % apoptosis, red: green fluorescence ratio, and bcl-2 expression were significantly and concentration-dependently decreased, while the expression levels of PARP, Bax, caspase-3 and caspase-8 were significantly increased (p < 0.05).Conclusion: These results indicate that the polysaccharides suppressed apoptosis of colorectal cancer cells by inhibiting the mitochondrial and caspase-dependent apoptosis pathways. Gleditsia polysaccharide may be used as an adjuvant therapy for colorectal cancer.


Oncogene ◽  
2021 ◽  
Author(s):  
Xiaoyan Fan ◽  
Yitao Wang ◽  
Tinghui Jiang ◽  
Tao Liu ◽  
Yuelei Jin ◽  
...  

AbstractB-Myb is an important transcription factor that plays a critical role in gene expression regulation and tumorigenesis. However, its functional implication in colorectal cancer remains elusive. In this study, we found that B-Myb was significantly upregulated at both mRNA and protein levels in colorectal cancer samples compared to non-tumor counterparts. B-Myb overexpression accelerated cell proliferation, cell cycle progression and cell motility in colorectal cancer cells, and promoted tumor growth in orthotopic nude mouse models in vivo. In contrast, B-Myb depletion inhibited these malignant phenotypes. Mechanistic investigations revealed that E2F2 was a novel transcriptional target of B-Myb and is essential to B-Myb-induced malignant phenotypes. Notably, B-Myb and E2F2 exhibited positive expression correlation, and interacted with each other in colorectal cancer cells. In addition to their autoregulatory mechanisms, B-Myb and E2F2 can also directly transactivate each other, thus constituting consolidated reciprocal feed-forward transactivation loops. Moreover, both B-Myb and E2F2 are required for the activation of ERK and AKT signaling pathways in colorectal cancer cells. Taken together, our data clarified a critical role for B-Myb in colorectal cancer and unraveled an exquisite mutual collaboration and reciprocal cross regulation between B-Myb and E2F2 that contribute to the malignant progression of human colorectal cancer.


Sign in / Sign up

Export Citation Format

Share Document