• 제목/요약/키워드: colorectal cancer cell

검색결과 359건 처리시간 0.021초

홍화가 인체 대장암세포에 미치는 효과 (Effects of Carthami Flos on Human Colorectal Adenocarcinoma Cells)

  • 한송이;김정아;송호준;채한;권영규;김병주
    • 한국한의학연구원논문집
    • /
    • 제17권2호
    • /
    • pp.129-134
    • /
    • 2011
  • Objective : The purpose of this study was to investigate the anti-cancer effects of Carthami Flos in some kinds of human colorectal adenocarcinoma cells. Method : We used two kinds of human colorectal adenocarcinoma cell lines, such as HT-29 and WiDr cells. We examined cell death by MTT assay and observed the morphological changes with Carthami Flos. Result : We showed that the combination of sub-optimal doses of Carthami Flos and cisplatin noticeably suppresses in HT-29 cells and doxorubicin in WiDr cells. Furthermore, we studied the caspase 3 activity to identify the apoptosis. Conclusion : Our findings provide insight into unraveling the effects of Carthami Flos in human colorectal adenocarcinoma cells and developing therapeutic agents against colorectal cancer.

Role of Innate Immunity in Colorectal Cancer

  • Bora Keum
    • Journal of Digestive Cancer Research
    • /
    • 제6권1호
    • /
    • pp.11-15
    • /
    • 2018
  • Chemotherapy and surgical resection are the mainstay of cancer treatment. Particularly for chemotherapy, although it is effective method to care, sometimes cure various cancers, there are many different status of cancer not being controlled by chemotherapy such as recurrence and resistance to chemotherapy. In order to overcome those difficulties during cancer therapy, immunotherapy targeting immune cells and immune associated factors to enhance cancer immunity has been highlighted. Innate immunity plays important roles on initial stage of cancer immunity that are detecting, killing cancer cells and initiating adaptive immunity for cancer. So many basic and clinical studies to manage innate immunity for cancer therapy have been going on, and most of them were to stimulate innate immune cells including dendritic cell, macrophage, monocyte, and natural killer cell in various ways. They showed promising results but still there are many things to be resolved before clinical application. Herein, I review the role of innate immune cells and therapeutic trials for colorectal cancer.

  • PDF

SOCS1 counteracts ROS-mediated survival signals and promotes apoptosis by modulating cell cycle to increase radiosensitivity of colorectal cancer cells

  • Ryu, Ji-Yoon;Oh, Jiyoung;Kim, Su-Min;Kim, Won-Gi;Jeong, Hana;Ahn, Shin-Ae;Kim, Seol-Hee;Jang, Ji-Young;Yoo, Byong Chul;Kim, Chul Woo;Lee, Choong-Eun
    • BMB Reports
    • /
    • 제55권4호
    • /
    • pp.198-203
    • /
    • 2022
  • As negative regulators of cytokine signaling pathways, suppressors of cytokine signaling (SOCS) proteins have been reported to possess both pro-tumor and anti-tumor functions. Our recent studies have demonstrated suppressive effects of SOCS1 on epithelial to mesenchymal signaling in colorectal cancer cells in response to fractionated ionizing radiation or oxidative stress. The objective of the present study was to determine the radiosensitizing action of SOCS1 as an anti-tumor mechanism in colorectal cancer cell model. In HCT116 cells exposed to ionizing radiation, SOCS1 over-expression shifted cell cycle arrest from G2/M to G1 and promoted radiation-induced apoptosis in a p53-dependent manner with down-regulation of cyclin B and up-regulation of p21. On the other hand, SOCS1 knock-down resulted in a reduced apoptosis with a decrease in G1 arrest. The regulatory action of SOCS1 on the radiation response was mediated by inhibition of radiation-induced Jak3/STAT3 and Erk activities, thereby blocking G1 to S transition. Radiation-induced early ROS signal was responsible for the activation of Jak3/Erk/STAT3 that led to cell survival response. Our data collectively indicate that SOCS1 can promote radiosensitivity of colorectal cancer cells by counteracting ROS-mediated survival signal, thereby blocking cell cycle progression from G1 to S. The resulting increase in G1 arrest with p53 activation then contributes to the promotion of apoptotic response upon radiation. Thus, induction of SOCS1 expression may increase therapeutic efficacy of radiation in tumors with low SOCS1 levels.

RNA polymerase I subunit D activated by Yin Yang 1 transcription promote cell proliferation and angiogenesis of colorectal cancer cells

  • Jianfeng Shan;Yuanxiao Liang;Zhili Yang;Wenshan Chen;Yun Chen;Ke Sun
    • The Korean Journal of Physiology and Pharmacology
    • /
    • 제28권3호
    • /
    • pp.265-273
    • /
    • 2024
  • This study aims to explore possible effect of RNA polymerase I subunit D (POLR1D) on proliferation and angiogenesis ability of colorectal cancer (CRC) cells and mechanism herein. The correlation of POLR1D and Yin Yang 1 (YY1) expressions with prognosis of CRC patients in TCGA database was analyzed. Quantitative realtime polymerase chain reaction (qRT-PCR) and Western blot were applied to detect expression levels of POLR1D and YY1 in CRC cell lines and CRC tissues. SW480 and HT-29 cells were transfected with si-POLR1D or pcDNA3.1-POLR1D to achieve POLR1D suppression or overexpression before cell migration, angiogenesis of human umbilical vein endothelial cells were assessed. Western blot was used to detect expressions of p38 MAPK signal pathway related proteins and interaction of YY1 with POLR1D was confirmed by dual luciferase reporter gene assay and chromatin immunoprecipitation (ChIP). TCGA data showed that both POLR1D and YY1 expressions were up-regulated in CRC patients. High expression of POLR1D was associated with poor prognosis of CRC patients. The results showed that POLR1D and YY1 were highly expressed in CRC cell lines. Inhibition or overexpression of POLR1D can respectively suppress or enhance proliferation and angiogenesis of CRC cells. YY1 inhibition can suppress CRC progression and deactivate p38 MAPK signal pathway, which can be counteracted by POLR1D overexpression. JASPAR predicted YY1 can bind with POLR1D promoter, which was confirmed by dual luciferase reporter gene assay and ChIP. YY1 transcription can up-regulate POLR1D expression to activate p38 MAPK signal pathway, thus promoting proliferation and angiogenesis ability of CRC cells.

Fentanyl Increases Colorectal Carcinoma Cell Apoptosis by Inhibition of NF-κB in a Sirt1-dependent Manner

  • Zhang, Xiu-Lai;Chen, Min-Li;Zhou, Sheng-Li
    • Asian Pacific Journal of Cancer Prevention
    • /
    • 제15권22호
    • /
    • pp.10015-10020
    • /
    • 2014
  • Background: Fentanyl is used as an analgesic to treat pain in a variety of patients with cancer and recently it has become considered to also act as an antitumor agent. The study present was designed to investigate the effects of fentanyl on colorectal cancer cell growth and plausible mechanisms. Materials and Methods: The human colorectal carcinoma cell line HCT116 was subcutaneously injected into nude mice. The viability of HCT116 was tested by MTT assay, and apoptosis by flow cytometry and caspase-3 activity. The expression of Sirt1 and NF-${\kappa}B$ were evaluated by Western blotting and the levels of Sirt1 and NF-${\kappa}B$ by fluorescence method. SiRNA was used to silence and Ad-Sirt1 to overexpress Sirt1. Results: Our data showed that fentanyl could inhibit tumor growth, with increased expression of Sirt1 and down-regulation of Ac-p65 in tumors. Compared with control cells without treatment, HCT116 cells that were incubated with fentanyl had a higher apoptotic rate. Moreover, fentanyl could increase expression and activity of Sirt1 and inhibitor expression and activity of NF-${\kappa}B$, which might be mechanisms of fentanyl action. Conclusions: Fentanyl increased colorectal carcinoma cell apoptosis by inhibition of NF-${\kappa}B$ activation in a Sirt1-dependent manner.

Anti-Proliferative Activity of Ethanol Extracts from Taxilli Ramulus (Taxillus chinensis (DC.) Danser) Through Cyclin D1 Proteasomal Degradation in Human Colorectal Cancer Cells

  • Park, Gwang Hun;Song, Hun Min;Park, Su Bin;Park, Ji Hye;Shin, Myeong Su;Son, Ho-Jun;Um, Yurry;Jeong, Jin Boo
    • 한국자원식물학회지
    • /
    • 제30권6호
    • /
    • pp.640-646
    • /
    • 2017
  • In this study, we elucidated anti-cancer activity and potential molecular mechanism of 70% ethanol extracts from Taxilli Ramulus (Taxillus chinensis (DC.) Danser) (TR-E70) against human colorectal cancer cells. Anti-cell proliferative effect of TR-E70 was evaluated by MTT assay. The effect of TR-E70 on the expression of cyclin D1 in the protein and mRNA level was evaluated by Western blot and RT-PCR, respectively. TR-E70 suppressed the proliferation of human colorectal cancer cell lines, HCT116 and SW480. Although TR-E70 decreased cyclin D1 expression in protein and mRNA level, decreased level of cyclin D1 protein by TR-E70 more dramatically occurred than that of cyclin D1 mRNA. Cyclin D1 downregulation by TR-E70 was attenuated in presence of MG132. In addition, TR-E70 phosphorylated threonine-286 (T286) of cyclin D1. TR-E70-mediated cyclin D1 degradation was blocked in presence of LiCl as an inhibitor $GSK3{\beta}$ but not PD98059 as an ERK1/2 inhibitor and SB203580 as a p38 inhibitor. Our results suggest that TR-E70 may downregulate cyclin D1 as one of the potential anti-cancer targets through $GSK3{\beta}$-dependent cyclin D1 degradation. From these findings, TR-E70 has potential to be a candidate for the development of chemoprevention or therapeutic agents for human colorectal cancer.

함유황 다당체 Fucoidan의 인체 대장암세포(HT-29) 사멸과 Apoptosis에 미치는 영향 (Effects of Fucoidan, a Sulfur-Containing Polysaccharide, on Cytotoxicity and Apoptosis in HT-29 Human Colorectal Cancer Cells)

  • 김민지;정하숙
    • 한국식품영양학회지
    • /
    • 제35권3호
    • /
    • pp.204-212
    • /
    • 2022
  • The purpose of this study was to investigate the biological activity of fucoidan, a sulfur-containing polysaccharide, on cytotoxicity and apoptosis in the human HT-29 colorectal cancer cell line using cell viability, Flow cytometry, Western blot, and RT-PCR analyses. Fucoidan inhibited the proliferation of HT-29 cells by 39.6% at a concentration of 100 ㎍/mL for 72 h. The inhibition was dose-dependent and accompanied by apoptosis. Flow cytometric analysis showed that fucoidan increased early apoptosis and late apoptosis by 65.84% and 72.09% at concentrations of 25 and 100 ㎍/mL, respectively. Analysis of the mechanism of these events indicated that fucoidan-treated cells exhibited increases in the activation of caspase-3, caspase-8, and PARP in a dose-dependent manner. These results suggest that fucoidan may inhibit the growth of human colorectal cancer cells by various apoptosis-promoting effects, as well as by apoptosis itself.

Preliminary Proteomic Analysis of Indomethacin's Effect on Tumor Transplanted with Colorectal Cancer Cell in Nude Mice

  • Wang, Yu-Jie;Zhang, Gui-Ying;Xiao, Zhi-Qiang;Wang, Hong-Mei;Chen, Zhu-Chu
    • BMB Reports
    • /
    • 제39권2호
    • /
    • pp.171-177
    • /
    • 2006
  • Nonsteroidal anti-inflammatory drugs such as indomethacin (IN) can exert anti-colorectal cancer (CRC) activity through cyclooxygenase independent mechanism, but the exactly biological mechanism is not completely known. Here we use proteomic tools to investigate the molecular mechanism of this action. First, nude mice bearing tumors derived from subcutaneous injection with human CRC cell line HCT116 were randomly allocated to groups treated with or without indomethacin. Later, tumor lumps were incised and then total proteins extracted. After separated with two-dimensional electrophoresis, thirty-one differently expressed spots were found between IN-treated and non-IN-treated groups, of which 25 spots decreased and 6 spots increased in abundance in IN-treated group. Through matrix-assisted laser desorption ionization time of flight mass spectrometry and then NCBInr and SWISS-PROT databases searching, 12 protein spots were finally identified including galectin-1, annexin A1, annexin IV, trancription factor BTF3A, calreticulin. Most of the identified proteins are correlated with tumor's biological prosperities of proliferation, invasion, apoptosis and immunity, or take part in cell's signal transduction. From above we thought that indomethacin can exert its effect on colorectal cancer through regulating several proteins' expression directly or indirectly. Further study of these proteins may be helpful in founding new targets of drugs for cancer chemotherapy.

Anti-Proliferative Effect of Naringenin through p38-Dependent Downregulation of Cyclin D1 in Human Colorectal Cancer Cells

  • Song, Hun Min;Park, Gwang Hun;Eo, Hyun Ji;Lee, Jin Wook;Kim, Mi Kyoung;Lee, Jeong Rak;Lee, Man Hyo;Koo, Jin Suk;Jeong, Jin Boo
    • Biomolecules & Therapeutics
    • /
    • 제23권4호
    • /
    • pp.339-344
    • /
    • 2015
  • Naringenin (NAR) as one of the flavonoids observed in grapefruit has been reported to exhibit an anti-cancer activity. However, more detailed mechanism by which NAR exerts anti-cancer properties still remains unanswered. Thus, in this study, we have shown that NAR down-regulates the level of cyclin D1 in human colorectal cancer cell lines, HCT116 and SW480. NAR inhibited the cell proliferation in HCT116 and SW480 cells and decreased the level of cyclin D1 protein. Inhibition of proteasomal degradation by MG132 blocked NAR-mediated cyclin D1 downregulation and the half-life of cyclin D1 was decreased in the cells treated with NAR. In addition, NAR increased the phosphorylation of cyclin D1 at threonine-286 and a point mutation of threonine-286 to alanine blocked cyclin D1 downregulation by NAR. p38 inactivation attenuated cyclin D1 downregulation by NAR. From these results, we suggest that NAR-mediated cyclin D1 downregulation may result from proteasomal degradation through p38 activation. The current study provides new mechanistic link between NAR, cyclin D1 downregulation and cell growth in human colorectal cancer cells.

MiR-454 Prompts Cell Proliferation of Human Colorectal Cancer Cells by Repressing CYLD Expression

  • Liang, Hong-Liang;Hu, Ai-Ping;Li, Sen-Lin;Xie, Jia-Ping;Ma, Qing-Zhu;Liu, Ji-Yong
    • Asian Pacific Journal of Cancer Prevention
    • /
    • 제16권6호
    • /
    • pp.2397-2402
    • /
    • 2015
  • Previous studies have shown that miR-454 plays an important role in a variety of biological processes in various human cancer cells. However, the underlying mechanisms of this microRNA in colorectal cancer (CRC) cells remain largely unknown. In the present study, we investigated the miR-454 role in CRC cell proliferation. We found that miR-454 expression is markedly upregulated in CRC tissues and CRC cells compared with the matched tumor adjacent tissues and the FHC normal colonic cell line. Ectopic expression of miR-454 promoted the proliferation and anchorage-independent growth of CRC cells, whereas inhibition of miR-454 reduced this effect. Bioinformatics analysis further revealed cylindromatosis (CYLD), a putative tumor suppressor as a potential target of miR-454. Data from luciferase reporter assays showed that miR-454 directly binds to the 3'-untranslated region (3'-UTR) of CYLD mRNA and repressed expression at both transcriptional and translational levels. In functional assays, CYLD-silenced in miR-454-in-transfected SW480 cells have positive effect to promote cell proliferation, suggesting that direct CYLD downregulation is required for miR-454-induced CRC cell proliferation. In sum, our data provide compelling evidence that miR-454 functions as an onco-miRNA, playing a crucial role in the promoting cell proliferation in CRC, and its oncogenic effect is mediated chiefly through direct suppression of CYLD expression.