scholarly journals MicroRNA silencing of CNN1 and CNN2 protein family members regulates biology processes in colorectal cancer by targeting the p53 signal pathway

2020 ◽  
Author(s):  
Fuda Huang ◽  
Mingwei Wei ◽  
Anmin Wang ◽  
Ya Zhang ◽  
Zebang Qin ◽  
...  

Abstract BackgroundCalponin was first defined as a striated muscle troponin T-like protein that binds actin thin filaments to regulate smooth muscle contraction. There are few studies of CNN1 and CNN2 in colorectal cancer, and the roles these two genes play in colorectal cancer cell lines and the mechanisms by which they act are unknown.MethodsWe used immunohistochemistry to identify expression of the two genes in the cancer tissues. RT-PCR was used to measure expression levels of microRNA. W performed western blots to measure changes in signaling pathways in the context of expression interference.Meanwhile, the same method was used to measure binding relationship between the two genes and key pathway proteins. To determine the relationship between microRNA and gene mRNA, we used the reporter gene method. We used the chi-square and t-test methods to analyze the significance and correlations of the data.Results and conclusionsExpression levels of CNN1 were lower in colon cancer tissues than in normal mucosal tissues. After downregulating CNN1, the cell cycle in colon cancer cell lines progressed quickly, and the expression of related pathway proteins also increased. Expression levels of CNN2 were higher in colon cancer tissues, and its downregulation significantly inhibited cell cycle progression in colon cancer cell lines. We confirmed correlations between the expression of microRNA and CNN2 using data analysis.Bars indicate ± standard errors.*p < 0.05; **p < 0.01 compared with the control. The inhibition of the expression of CNN2 mRNA using microRNA was confirmed using western blot. The combination of the two at the mechanism level was also demonstrated using the reporter gene method.

2021 ◽  
Author(s):  
Baiyou Tang ◽  
Lihua Zhang ◽  
Jing Yu ◽  
Mingjing Peng ◽  
Yu Cheng ◽  
...  

Abstract Background: Solute carrier family 7 member 5 (SLC7A5) was identified highly expressed and as a key participant in various tumor development; however, the role it played in colorectal cancer remains unclear. Methods and Results: In the current study, the expression of SLC7A5 were systematically mined in public databases and validated by real-time PCR in colon cancer and normal tissues. And then, the co-expression and pathway analysis got from public database, which indicated the potential influence of SLC7A5 for the etiology of colorectal cancer, were evaluated in the colon cancer cell lines by loss of SLC7A5 function experiment, flow cytometry, western blot, and wound healing assay. The results showed that the mRNA expression of SLC7A5 was significantly higher in colorectal cancer tissues than that in the non-tumor controls for GEO and TCGA datasets as well as 40 pairs of Xiangya clinical samples. The functional enrichment analysis based on public database showed that the pathways enriched most were cell cycle and epithelial-to-mesenchymal transition (EMT), and Cyclin D1 (CCND1) were the only gene that had a significant positive correlation with SLC7A5. Loss of SLC7A5 function in colon cancer cell lines could arrest cell cycle at G1 phase by down-regulating CCND1 and CDK2 protein expression, and may reduce cell migration by reversing EMT though upregulation of E-Cadherin and downregulation of zonula occludens-1. Conclusion: SLC7A5 is likely associated with the progression of colon cancer.


2016 ◽  
Vol 16 (4) ◽  
pp. 585-596 ◽  
Author(s):  
Ji-Ye Kee ◽  
Yo-Han Han ◽  
Jinbong Park ◽  
Dae-Seung Kim ◽  
Jeong-Geon Mun ◽  
...  

Background: β-Lapachone is a quinone-containing compound found in red lapacho ( Tabebuia impetiginosa, syn. T avellanedae) trees. Lapacho has been used in traditional medicine by several South and Central American indigenous people to treat various types of cancer. The purpose of this study was to investigate the antimetastatic properties of β-lapachone and the underlying mechanisms using colon cancer cells. Methods: This research used metastatic murine colon cancer cell lines, colon 26 (CT26) and colon 38 (MC38). A WST assay, annexin V assay, cell cycle analysis, wound healing assay, invasion assay, western blot analysis, and real-time reverse transcription–polymerase chain reaction were performed to examine the effects of β-lapachone on metastatic phenotypes and molecular mechanisms. The effect of β-lapachone on lung metastasis was assessed in a mouse experimental metastasis model. Results: We found that the inhibition of proliferation of the colon cancer cell lines by β-lapachone was due to the induction of apoptosis and cell cycle arrest. β-Lapachone induced the apoptosis of CT26 cells through caspase-3, -8, and -9 activation; poly(ADP-ribose) polymerase cleavage; and downregulation of the Bcl-2 family in a dose- and time-dependent manner. In addition, a low concentration of β-lapachone decreased the cell migration and invasion by decreasing the expression of matrix metalloproteinases-2 and -9, and increased the expression of tissue inhibitors of metalloproteinases-1 and -2. Moreover, β-lapachone treatment regulated the expression of epithelial-mesenchymal transition markers such as E- and N-cadherin, vimentin, β-catenin, and Snail in CT26 cells. In the mouse experimental metastasis model, β-lapachone significantly inhibited the lung metastasis of CT26 cells. Conclusions: Our results demonstrated the inhibitory effect of β-lapachone on colorectal lung metastasis. This compound may be useful for developing therapeutic agents to treat metastatic cancer.


2012 ◽  
Vol 30 (13) ◽  
pp. 1505-1512 ◽  
Author(s):  
Andrew J. Weickhardt ◽  
Tim J. Price ◽  
Geoff Chong ◽  
Val Gebski ◽  
Nick Pavlakis ◽  
...  

Purpose This preclinical and phase II study evaluated the efficacy and safety of the combination of cetuximab and erlotinib in metastatic colorectal cancer (mCRC). Patients and Methods The activity and mechanism of action of the combination of cetuximab plus erlotinib were investigated in vitro in colorectal cancer cell lines. In the clinical study, patients with chemotherapy-refractory mCRC were treated with cetuximab 400 mg/m2 as a loading dose and then weekly cetuximab 250 mg/m2 with erlotinib 100 mg orally daily. The primary end point was response rate (RR), which was evaluated separately in KRAS wild-type (WT) versus KRAS mutant tumors. Secondary end points included toxicity, progression-free survival (PFS), and overall survival. Target accrual was 50 patients, with a one-stage design. Results Preclinical studies demonstrated synergistic activity of cetuximab and erlotinib cotreatment on growth inhibition of colon cancer cell lines both as a result of enhanced inhibition of the epidermal growth factor receptor pathway and differential effects on STAT3. In the clinical study, 50 patients were enrolled, with 48 patients evaluable for response. The overall RR was 31% (95% CI, 26% to 57%), with a median PFS of 4.6 months (95% CI, 2.8 to 5.6 months). RR was 41% (95% CI, 26% to 57%) in KRAS WT tumors, with a median PFS of 5.6 months (95% CI, 2.9 to 5.6 months). There was no response in 11 patients with KRAS mutations. Frequent grade 3 and 4 toxicities were rash (48%), hypomagnesaemia (18%), and fatigue (10%). Conclusion The combination of cetuximab and erlotinib synergistically inhibits growth of colon cancer cell lines, achieves promising efficacy in patients with KRAS WT mCRC, and merits evaluation in further randomized studies.


2012 ◽  
Vol 10 (1) ◽  
pp. 109 ◽  
Author(s):  
Sebastian Gnosa ◽  
Yang-Mei Shen ◽  
Chao-Jie Wang ◽  
Hong Zhang ◽  
Johannes Stratmann ◽  
...  

2020 ◽  
Vol 8 (Suppl 2) ◽  
pp. A50.1-A50
Author(s):  
M Mianowska ◽  
M Zaremba-Czogalla ◽  
A Zygmunt ◽  
J Gubernator

BackgroundColorectal cancer is the third most commonly diagnosed malignant tumor, taking fourth place in terms of cause of cancer deaths worldwide.1 Unfortunately, the ability of the immune system to distinguish its own from foreign cells is often limited. One of the overexpressed receptors is receptor CD47 - widely distributed glycoprotein on the cell surface of various kind of tumors. It plays a role as ‘don’t eat me’ signal by binding with receptor SIRPα, presents on the cell surface of macrophages.2 Calreticulin, protein occurring on the surface of tumor cells and phagocytes, acts as protein with pro-phagocytic properties. Several natural bioactive substances are predicted to induce immunogenic cell death by translocation calreticulin on the surface of cancer cells which significantly increases the efficiency of their phagocytosis. Moreover, one of the well-known TLR-7 receptor agonists - imiquimod, is involved in phosphorylation of Bruton’s tyrosine kinase leading to the appearance of calreticulin on the surface of macrophages, which increases the efficiency of phagocytosis of tumor cells.3 Combination therapy composed of berberine and imiquimod can be highlighted as effective immunotherapy for colon cancer. However, such an approach remains very limited. Liposomes can serve as promising carriers for targeting delivery and controlled release of anti-cancer agents.Material and MethodsLiposomes were prepared by the thin-film hydration method followed by extrusion. Human colon cancer cell line (LS180 I SW620) and human monocytic cell line (THP-1) were used for experiments. Calreticulin was detected by using confocal microscopy.ResultsThe work presented aimed to develop novel liposomal formulations of berberine and imiquimod which were examined for their efficacy in combination against colorectal cancer cell lines. Liposomal formulations of both compounds were successfully prepared using active loading method with different pH generating agents. All loading methods showed desired characteristics in terms of mean liposome size and polydispersity. The encapsulation efficiency was higher than 95% for almost all used formulations. The in vitro study proved cytotoxicity of berberine loaded liposomal formulations on tested colon cancer cell lines. The results of the immunofluorescence staining indicated that the both compounds triggered calreticulin on the cell surface (colon cancer or macrophages).ConclusionsThe combination of both substances in the liposomal form may generate a synergistic effect on phagocytosis of colon cancer cells.ReferencesArnold M, Sierra MS, Laversanne M, et al. Global patterns and trends in colorectal cancer incidence and mortality. Gut 2017;66:683–691.Sick E, Jeanne A, Schneider C, Dedieu S, Takeda K, Martiny L. CD47 update: a multifaceted actor in the tumor microenvironment of potential therapeutic interest, Br J Pharmacol 2012, 167(7):1415–30.M. Feng, et al., Macrophages eat cancer cells using their own calreticulin as a guide: Roles of TLR and Btk. PNAS 2015;112( 7):2145–2150.Disclosure InformationM. Mianowska: B. Research Grant (principal investigator, collaborator or consultant and pending grants as well as grants already received); Significant; National Science Center, Poland. M. Zaremba-Czogalla: None. A. Zygmunt: None. J. Gubernator: None.


2015 ◽  
Vol 112 (8) ◽  
pp. 2467-2472 ◽  
Author(s):  
James R. Bayrer ◽  
Sridevi Mukkamala ◽  
Elena P. Sablin ◽  
Paul Webb ◽  
Robert J. Fletterick

Colorectal cancers (CRCs) account for nearly 10% of all cancer deaths in industrialized countries. Recent evidence points to a central role for the nuclear receptor liver receptor homolog-1 (LRH-1) in intestinal tumorigenesis. Interaction of LRH-1 with the Wnt/β-catenin pathway, highly active in a critical subpopulation of CRC cells, underscores the importance of elucidating LRH-1’s role in this disease. Reduction of LRH-1 diminishes tumor burden in murine models of CRC; however, it is not known whether LRH-1 is required for tumorigenesis, for proliferation, or for both. In this work, we address this question through shRNA-mediated silencing of LRH-1 in established CRC cell lines. LRH-1 mRNA knockdown results in significantly impaired proliferation in a cell line highly expressing the receptor and more modest impairment in a cell line with moderate LRH-1 expression. Cell-cycle analysis shows prolongation of G0/G1 with LRH-1 silencing, consistent with LRH-1 cell-cycle influences in other tissues. Cluster analysis of microarray gene expression demonstrates significant genome wide alterations with major effects in cell-cycle regulation, signal transduction, bile acid and cholesterol metabolism, and control of apoptosis. This study demonstrates a critical proproliferative role for LRH-1 in established colon cancer cell lines. LRH-1 exerts its effects via multiple signaling networks. Our results suggest that selected CRC patients could benefit from LRH-1 inhibitors.


Sign in / Sign up

Export Citation Format

Share Document