• Title/Summary/Keyword: Gastric cancer cells

Search Result 496, Processing Time 0.13 seconds

The Growth Inhibitiory Effect of New Pyrrolo[1,2-${\alpha}$]benzimidazole Derivatives on Human Gastric Cancer Cells

  • Kim, Soo-Kie;Ahn, Chan-Mug;Choi, Sun-Ju;Park, Yoon-Sun;Cho, Hyung-Chul;Koh, Choon-Myung
    • Archives of Pharmacal Research
    • /
    • v.20 no.5
    • /
    • pp.410-413
    • /
    • 1997
  • In the course of screening synthetic compounds to inhibit tumor cell growth, pyrrolo[1,2-.alpha.] benzimidazole (PBI), an intermediate of azamitosene, was found to inhibit a proliferation of gastric cancer cell lines. Despite a potential cytotoxic activity against solid tumor cells as opposed to that against rapidly-doubled leukemic cells, there has been no report on the inhibition of gastric cancer cell line by PBI and its' derivatives. The present experiment was designed to determine if PBI derivatives can effectively inhibit the cellular proliferation of gastric cancer cells by using in vitro as well as in vivo chemosensitivity system (MTT assay, clonogenic assay and human tumor xenografted assay). Of the tested PBI derivatives, PBI (18) and PBI (20), displayed the effective growth inhibition of cultured gastric cancer cells or even in the xenografted nude mouse model.

  • PDF

The Growth Inhibition against Gastric Cancer Cell in Germanium or Soybean Sprouts Cultured with Germanium (게르마늄 및 게르마늄 분말 용해수로 재배한 콩나물의 위암세포 성장억제 작용)

  • 김은정;이경임;박건영
    • Korean journal of food and cookery science
    • /
    • v.20 no.3
    • /
    • pp.287-291
    • /
    • 2004
  • The growth inhibitory effect of germanium, or soybean sprouts cultured with germanium, on cancer cells was determined in the cultured gastric cancer cell line, AGS. The growth of AGS was significantly inhibited by the addition of 0.01-1% organic germanium (Ge-132) and germanium stone powder in MTT cytotoxicity assays. The juice from germanium treated soybean sprouts (GTS) inhibited the growth of AGS gastric cancer cells by 78-88% at concentrations of 2.5 or 5${\mu}\ell$. The juice from Seomoktae GTS revealed an especially higher growth inhibitory effect than that from the control soybean sprouts (germanium non-treated soybean sprouts, GNTS) in AGS. The results suggest that soybean sprouts cultured with germanium may exert an anticancer effect against gastric cancer cells.

Hypermethylation and Clinicopathological Significance of RASAL1 Gene in Gastric Cancer

  • Chen, Hong;Pan, Ying;Cheng, Zheng-Yuan;Wang, Zhi;Liu, Yang;Zhao, Zhu-Jiang;Fan, Hong
    • Asian Pacific Journal of Cancer Prevention
    • /
    • v.14 no.11
    • /
    • pp.6261-6265
    • /
    • 2013
  • Background: Recent studies have suggested that expression of the RAS protein activator like-1 gene (RASAL1) is decreased in gastric carcinoma tissues and cell lines, indicated a role in tumorigenesis and development of gastric cancer. Reduced expression of RASAL1 could result in aberrant increase of activity of RAS signaling pathways in cancer cells. However, the exact mechanism which induces down-regulation of the RASAL1 gene remains unclear. This study aimed to determine the methylation status and regulation of RASAL1 in gastric cancer. Materials and Methods: Using the methylation-specific polymerase chain reaction (MSP), the methylation status of CpG islands in the RASAL1 promoter in gastric cancers and paired adjacent non-cancerous tissues from 40 patients was assessed and its clinicopathological significance was analyzed. The methylation status of RASAL1 in gastric cancer lines MKN-28, SGC-790l, BGC-823, as well as in normal gastric epithelial cell line GES-l was also determined after treatment with a DNA methyltransferase inhibitor, 5-aza-2'-doexycytidine (5-Aza-CdR). RAS activity (GAS-GTP) was assessed through a pull-down method, while protein levels of ERK1/2, a downstream molecule of RAS signaling pathways, were determined by Western blotting. Results: The frequencies of RASAL1 promoter methylation in gastric cancer and paired adjacent non-cancerous tissues were 70% (28/40) and 30% (12/40) respectively (P<0.05). There were significantly correlations between RASAL1 promoter methylation with tumor differentiation, tumor size, invasive depth and lymph node metastasis in patients with gastric cancer (all P<0.05), but no correlation was found for age or gender. Promoter hypermethylation of the RASAL1 gene was detected in MKN-28, SGC-790l and BGC-823 cancer cells, but not in the normal gastric epithelial cell line GES-1. Elevated expression of the RASAL1 protein, a decreased RAS-GTP and p-ERK1/2 protein were detected in three gastric cancer cell lines after treatment with 5-Aza-CdR. Conclusions: Aberrant hypermethylation of the RASAL1 gene promoter frequently occurs in gastric cancer tissues and cells. In addition, the demethylating agent 5-Aza-CdR can reverse the hypermethylation of RASAL1 gene and up-regulate the expression of RASAL1 significantly in gastric cancer cells in vivo. Our study suggests that RASAL1 promoter methylation may have a certain relationship with the reduced RASAL1 expression in gastric cancer.

Anti-proliferation Effects of Interferon-gamma on Gastric Cancer Cells

  • Zhao, Ying-Hui;Wang, Tao;Yu, Guang-Fu;Zhuang, Dong-Ming;Zhang, Zhong;Zhang, Hong-Xin;Zhao, Da-Peng;Yu, Ai-Lian
    • Asian Pacific Journal of Cancer Prevention
    • /
    • v.14 no.9
    • /
    • pp.5513-5518
    • /
    • 2013
  • IFN-${\gamma}$ plays an indirect anti-cancer role through the immune system but may have direct negative effects on cancer cells. It regulates the viability of gastric cancer cells, so we examined whether it affects their proliferation and how that might be brought about. We exposed AGS, HGC-27 and GES-1 gastric cancer cell lines to IFN-${\gamma}$ and found significantly reduced colony formation ability. Flow cytometry revealed no effect of IFN-${\gamma}$ on apoptosis of cell lines and no effect on cell aging as assessed by ${\beta}$-gal staining. Microarray assay revealed that IFN-${\gamma}$ changed the mRNA expression of genes related to the cell cycle and cell proliferation and migration, as well as chemokines and chemokine receptors, and immunity-related genes. Finally, flow cytometry revealed that IFN-${\gamma}$ arrested the cells in the G1/S phase. IFN-${\gamma}$ may slow proliferation of some gastric cancer cells by affecting the cell cycle to play a negative role in the development of gastric cancer.

CD44 and CD133 as Cancer Stem Cell Markers for Gastric Cancer

  • Lee, Hyun-Joo;Choi, Young-Sil;Kim, Sung-Joo;Moon, Hyoun-Jong
    • Journal of Gastric Cancer
    • /
    • v.10 no.3
    • /
    • pp.99-105
    • /
    • 2010
  • Purpose: Currently, the two most influential gastric stem cell marker candidates are CD44 and CD133. The aim of this study was to make a comparison and determine the appropriate marker for use in gastric cancer stem cell research. Materials and Methods: We analyzed the expressions of CD44, CD133, and CD24 from the gastric cancer cell lines MKN45, MKN74, KATO-III, NCI-N87, SNU-1, SNU-216, SNU-601, SNU-638, and SNU-688 using flow cytometry. In addition, we measured the change in viability after applying 5 fluorouracil (5-FU) to the MKN45, MKN74, KATO-III, and NCI-N87 cell lines using a Cell Counting Kit 8. Results: CD133 expression was above moderate in the KATO-III, SNU-216, SNU-601 cell lines, whereas it was below 1% in the remaining cell lines. CD44 was expressed at levels above 5% in all gastric cancer cell lines. The effect of 5-FU on viability and CD133 or CD44 expression in the cell lines were not related. Conclusions: Expression of CD133 positive cells was insufficient in the gastric cancer cell lines. Therefore, of the cell lines tested, CD44 was the most appropriate tumor maker for research on gastric cancer stem cells.

Exogenous Morphine Inhibits Human Gastric Cancer MGC-803 Cell Growth by Cell Cycle Arrest and Apoptosis Induction

  • Qin, Yi;Chen, Jing;Li, Li;Liao, Chun-Jie;Liang, Yu-Bing;Guan, En-Jian;Xie, Yu-Bo
    • Asian Pacific Journal of Cancer Prevention
    • /
    • v.13 no.4
    • /
    • pp.1377-1382
    • /
    • 2012
  • Morphine is not only an analgesic treating pain for patients with cancer but also a potential anticancer drug inhibiting tumor growth and proliferation. To gain better insight into the involvement of morphine in the biological characteristics of gastric cancer, we investigated effects on progression of gastric carcinoma cells and the expression of some apoptosis-related genes including caspase-9, caspase-3, survivin and NF-${\kappa}B$ using the MGC-803 human gastric cancer cell line. The viability of cells was assessed by MTT assay, proliferation by colony formation assay, cell cycle progression and apoptosis by flow cytometry and ultrastructural alteration by transmission electron microscopy. The influences of morphine on caspase-9, caspase-3, survivin and NF-${\kappa}B$ were evaluated by semi-quantitative RT-PCR and Western blot. Our data showed that morphine could significantly inhibit cell growth and proliferation and cause cell cycle arrest in the G2/M phase. MGC-803 cells which were incubated with morphine also had a higher apoptotic rate than control cells. Morphine also led to morphological changes of gastric cancer cells. The mechanism of morphine inhibiting gastric cancer progression in vitro might be associated with activation of caspase-9 and caspase-3 and inhibition of survivin and NF-${\kappa}B$.

Ursolic Acid Promotes Apoptosis of SGC-7901 Gastric Cancer Cells through ROCK/PTEN Mediated Mitochondrial Translocation of Cofilin-1

  • Li, Rui;Wang, Xia;Zhang, Xiao-Hong;Chen, Hong-Hai;Liu, Yan-Dong
    • Asian Pacific Journal of Cancer Prevention
    • /
    • v.15 no.22
    • /
    • pp.9593-9597
    • /
    • 2014
  • Ursolic acid, extracted from the traditional Chinese medicine bearberry, can induce apoptosis of gastric cancer cells. However, its pro-apoptotic mechanism still needs further investigation. More and more evidence demonstrates that mitochondrial translocation of cofilin-1 appears necessary for the regulation of apoptosis. Here, we report that ursolic acid (UA) potently induces the apoptosis of gastric cancer SGC-7901 cells. Further mechanistic studies revealed that the ROCK1/PTEN signaling pathway plays a critical role in UA-mediated mitochondrial translocation of cofilin-1 and apoptosis. These findings imply that induction of apoptosis by ursolic acid stems primarily from the activation of ROCK1 and PTEN, resulting in the translocation of cofilin-1 from cytoplasm to mitochondria, release of cytochrome c, activation of caspase-3 and caspase-9, and finally inducing apoptosis of gastric cancer SGC-7901 cells.

Involvement of melastatin type transient receptor potential 7 channels in ginsenoside Rd-induced apoptosis in gastric and breast cancer cells

  • Kim, Byung Joo
    • Journal of Ginseng Research
    • /
    • v.37 no.2
    • /
    • pp.201-209
    • /
    • 2013
  • Ginsenoside, one of the active ingredients of Panax ginseng, has a variety of physiologic and pharmacologic effects. The purpose of this study was to explore the effects of ginsenoside Rd (G-Rd) on melastatin type transient receptor potential 7 (TRPM7) channels with respect to the proliferation and survival of AGS and MCF-7 cells (a gastric and a breast cancer cell line, respectively). AGS and MCF-7 cells were treated with different concentrations of G-Rd, and caspase-3 activities, mitochondrial depolarizations, and sub-G1 fractions were analyzed to determine if cell death occurred by apoptosis. In addition, human embryonic kidney (HEK) 293 cells overexpressing TRPM7 channels were used to confirm the role of TRPM7 channels. G-Rd inhibited the proliferation and survival of AGS and MCF-7 cells and enhanced caspase-3 activity, mitochondrial depolarization, and sub-G1 populations. In addition, G-Rd inhibited TRPM7-like currents in AGS and MCF-7 cells and in TRPM7 channel overexpressing HEK 293 cells, as determined by whole cell voltage-clamp recordings. Furthermore, TRPM7 overexpression in HEK 293 cells promoted G-Rd induced cell death. These findings suggest that G-Rd inhibits the proliferation and survival of gastric and breast cancer cells by inhibiting TRPM7 channel activity.

Eupatilin Inhibits Gastric Cancer Cell Growth by Blocking STAT3-Mediated VEGF Expression

  • Cheong, Jae-Ho;Hong, Sung-Yi;Zheng, Yanjun;Noh, Sung-Hoon
    • Journal of Gastric Cancer
    • /
    • v.11 no.1
    • /
    • pp.16-22
    • /
    • 2011
  • Purpose: Eupatilin is an antioxidative flavone and a phytopharmaceutical derived from Artemisia asiatica. It has been reported to possess anti-tumor activity in some types of cancer including gastric cancer. Eupatilin may modulate the angiogenesis pathway which is part of anti-inflammatory effect demonstrated in gastric mucosal injury models. Here we investigated the anti-tumor effects of eupatilin on gastric cancer cells and elucidated the potential underlying mechanism whereby eupatilin suppresses angiogenesis and tumor growth. Materials and Methods: The impact of eupatilin on the expression of angiogenesis pathway proteins was assessed using western blots in MKN45 cells. Using a chromatin immunoprecipitation assay, we tested whether eupatilin affects the recruitment of signal transducer and activator of transcription 3 (STAT3), aryl hydrocarbon receptor nuclear translocator (ARNT) and hypoxia-inducible factor-$1{\alpha}$ (HIF-$1{\alpha}$) to the human VEGF promoter. To investigate the effect of eupatilin on vasculogenesis, tube formation assays were conducted using human umbilical vein endothelial cells (HUVECs). The effect of eupatilin on tumor suppression in mouse xenografts was assessed. Results: Eupatilin significantly reduced VEGF, ARNT and STAT3 expression prominently under hypoxic conditions. The recruitment of STAT3, ARNT and HIF-$1{\alpha}$ to the VEGF promoter was inhibited by eupatilin treatment. HUVECs produced much foreshortened and severely broken tubes with eupatilin treatment. In addition, eupatilin effectively reduced tumor growth in a mouse xenograft model. Conclusions: Our results indicate that eupatilin inhibits angiogenesis in gastric cancer cells by blocking STAT3 and VEGF expression, suggesting its therapeutic potential in the treatment of gastric cancer.

ER membrane protein complex subunit 6 (EMC6) is a novel tumor suppressor in gastric cancer

  • Wang, Xiaokun;Xia, Yan;Xu, Chentong;Lin, Xin;Xue, Peng;Zhu, Shijie;Bai, Yun;Chen, Yingyu
    • BMB Reports
    • /
    • v.50 no.8
    • /
    • pp.411-416
    • /
    • 2017
  • The endoplasmic reticulum (ER) membrane protein complex subunit 6 (EMC6) is a novel human autophagy-related molecule. Here, using tissue microarray and immunohistochemistry, we report that EMC6 protein is lost or reduced in glandular cells of patients with gastric adenocarcinoma, compared to normal stomach mucosa. Overexpression of EMC6 in gastric cancer cells inhibited cell growth, migration, invasion, and induced apoptosis and cell cycle arrest at S-phase. Further investigation suggested that EMC6 overexpression in BGC823 human adenocarcinoma gastric cancer cells reduced tumorigenicity in a xenograft model, demonstrating that EMC6 has the characteristics of a tumor suppressor. This is the first study to show that EMC6 induces cell death in gastric cancer cells. The molecular mechanism of how EMC6 functions as a tumor suppressor needs to be further explored.