• 제목/요약/키워드: human gastric cancer cell

검색결과 284건 처리시간 0.03초

Ginkgo biloba Leaf Extract Regulates Cell Proliferation and Gastric Cancer Cell Death

  • Kim, Da Hyun;Yang, Eun Ju;Lee, JinAh;Chang, Jeong Hyun
    • 대한의생명과학회지
    • /
    • 제28권2호
    • /
    • pp.92-100
    • /
    • 2022
  • Ginkgo biloba Leaf Extract (GBE) is an extract from leaves of the Ginkgo biloba tree, widely used as a health supplement. GBE can inhibit the proliferation of several types of tumor cell. Although it is known to have anti-cancer effects in breast cancer and skin cancer, research related to gastric cancer is still insufficient. Based on results showing anti-cancer effects on solid cancer, we aimed to determine whether GBE has similar effects on gastric cancer. In this study, the anti-cancer effect of GBE in gastric adenocarcinoma was investigated by confirming the cell proliferation inhibitory effect of AGS cells. We also evaluated whether GBE regulates expression of the tumor suppressor protein p53 and Rb. GBE has apoptotic effects on AGS cells that were confirmed by changes in anti-apoptosis protein Bcl-2, Bcl-xl and pro-apoptosis protein Bax levels. Wound healing and cell migration were also decreased by treatment with GBE. Furthermore, we verified the effects of GBE on mitogenic signaling by investigating AKT target gene expression levels and revealed downregulated Sod2 and Bcl6 expression. We also confirmed that expression of inflammation-related genes decreased in a time-dependent manner. These results indicate that GBE has an anti-cancer effect on human gastric cancer cell lines. Further research on the mechanism of the anti-cancer effect will serve as basic data for possible anti-cancer drug development.

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
    • /
    • 제50권8호
    • /
    • 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.

Hesperidin Induces Apoptosis in SNU-668, Human Gastric Cancer Cells

  • Park, Hae-Jeong;Ra, Je-Hyun;Han, Mi-Young;Chung, Joo-Ho
    • Molecular & Cellular Toxicology
    • /
    • 제3권1호
    • /
    • pp.31-35
    • /
    • 2007
  • Hesperidin, known as a flavonoid constituent of citrus, has been known to reduce the proliferation of several cancer cells. We investigated whether hesperidin-induced cell death on SNU-668, human gastric cancer cells. The cytotoxicity of hesperidin on SNU-668 cells was determined by 3-(4, 5-dimethylthiazol-2-yl)-2, 5-diphenyltetrazolium bromide (MTT) assay at the concentration of 1, 10, 50, and 100 ${\mu}M$. Cell viability by hesperidin was 53.18$\pm$2.85% of control value at 100 ${\mu}M$. The cell death by hesperidin showed apoptotic features, which were confirmed using a combination of 4, 6-diamidino-2-phenylindole (DAPI) staining and terminal deoxynucleotidyl transferase (TdT)-mediated dUTP nick end labeling (TUNEL) assay. In the apoptosis-regulating genes, treatment of hesperidin decreased mRNA expression of B-cell CLL/lymphoma 2 (BCL2), whereas expression of BCL2-associated X protein (BAX) was increased. The mRNA expression and the activity of caspase3 (CASP3), a major apoptotic factor, was significantly increased by hesperidin treatment. These results suggest that hesperidin could induce apoptosis through CASP3 activation on SNU-668, human gastric cancer cells.

위암에서 새로운 종양원인 유전자 Nemo-like Kinase의 발현 증가 (The Overexpression of Oncogenic Nemo-like Kinase in Gastric Cancer)

  • 김민규;정광화;남석우
    • 약학회지
    • /
    • 제56권6호
    • /
    • pp.358-363
    • /
    • 2012
  • Nemo-like kinase (NLK), an evolutionarily conserved serine/threonine protein kinase, plays an important role in wide variety of developmental events. NLK phosphorylates T-cell factor/lymphoid enhancer factor (TCF/LEF) transcriptional complex and suppresses wnt signaling pathway through inhibition of ${\beta}$-catenin/TCF complex interaction. However, the function of NLK in gastric carcinogenesis has not been investigated. In the present study, we have examined whether the NLK gene is involved in the development and/or progression of gastric cancers. NLK expression was analyzed by immunohistochemical staining in 153 advanced gastric cancer specimens. Immunhistochemical analysis showed increased expression of NLK in 91 (59.5%) out of 153 gastric cancer specimens. Statistically, there was no significant relationship between altered expression of NLK protein and clinicopathological parameters, including tumor differentiation, location, lymph node metastasis. We identified that mRNA and protein expression of NLK was significantly up-regulated in human gastric cancer tissues compare to corresponding normal gastric tissues. In addition, we found that human gastric cancer cell lines exhibited relatively high expression of NLK, as compared with normal gastric cells. The results of this study suggest that aberrant regulation of NLK may contribute to the development or progression of gastric cancers and serve as a potential biomarker for advanced gastric cancer patients.

MicroRNA-146a Enhances Helicobacter pylori Induced Cell Apoptosis in Human Gastric Cancer Epithelial Cells

  • Wu, Kai;Yang, Liu;Li, Cong;Zhu, Chao-Hui;Wang, Xin;Yao, Yi;Jia, Yu-Jie
    • Asian Pacific Journal of Cancer Prevention
    • /
    • 제15권14호
    • /
    • pp.5583-5586
    • /
    • 2014
  • Helicobacter pylori (H. pylori) infection induces apoptosis in gastric epithelial cells, and this occurrence may link to gastric carcinogenesis. However, the regulatory mechanism of H. pylori-induced apoptosis is not clear. MicroRNA-146a has been implicated as a key regulator of the immune system. This report describes our discovery of molecular mechanisms of microRNA-146a regulation of apoptosis in human gastric cancer cells. We found that overexpression of microRNA-146a by transfecting microRNA-146a mimics could significantly enhance apoptosis, and this upregulation was triggered by COX-2 inhibition. Furthermore, we found that microRNA-146a density was positively correlated with apoptosis rates in H. pylori-positive gastric cancer tissues and intratumoral microRNA-146a density was negatively correlated with lymph node metastasis among H. pylori-positive gastric cancer patients. Understanding the important roles of microRNA-146a in regulating cell apoptosis in H. pylori infected human gastric cancer cells will contribute to the development of microRNA targeted therapy in the future.

Knockdown of a Proliferation-inducing Ligand (PRIL) Suppresses the Proliferation of Gastric Cancer Cells

  • Cui, Jiu-Wei;Li, Yan;Wang, Chang;Yao, Cheng;Li, Wei
    • Asian Pacific Journal of Cancer Prevention
    • /
    • 제13권2호
    • /
    • pp.633-636
    • /
    • 2012
  • Purpose: PRIL (proliferation-inducing ligand) is a newly identified member of the tumor necrosis factor (TNF) family and modulates death ligand-induced apoptosis. Here, we investigated the effect of PRIL on cellular characteristics relating to tumor progression in human gastric cancer. Method: Recombinant lentivirus containing PRIL siRNA was constructed and then infected MGC803 and SGC7901 gastric cancer cells. MTT [3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide] colony formation and cell cycle analysis were used to study the effect of PRIL knockdown on gastric cancer cell proliferation. Results: PRIL expression in lentivirus infected cells was significantly reduced as evidenced by quantitative real-time PCR. Cell viability and colony formation of MGC803 and SGC7901 cells were significantly hampered in PRIL knock-down cells. Moreover, the cell cycle was arrested at G2/M phase, elucidating the mechanism underlying the inhibitory effect of siRNA on cell proliferation. Conclusions: Our study indicated that PRIL functions in promoting cell growth, and lentivirus-mediated PRIL gene knockdown might be a promising strategy in the treatment of gastric cancer.

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
    • /
    • 제20권5호
    • /
    • 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

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
    • /
    • 제13권4호
    • /
    • 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-${\kappa}B$.

한국인 위암에서 Survivin과 HIAP-1 유전자 발현 (Expression of Survivin and HIAP-1 in Korean Gastric Cancers)

  • 박찬진;류승완;김인호;백원기;서성일;서민호;손수상
    • Journal of Gastric Cancer
    • /
    • 제3권1호
    • /
    • pp.19-25
    • /
    • 2003
  • Purpose: Dysregulation of apoptosis may attribute to development of cancer by abnormally prolonging cell viability with accumulation of transforming mutations. Survivin and HIAP (Human Inhibitors of Apoptosis)-1 were recently described as apoptosis inhibitors. Their pathogenic roles in gastric cancer are largely unknown. In the present study, we examined the expression of survivin and HIAP-1 in gastric cancer tissues and cell lines in order to elucidate the roles of survivin and HIAP-1 in the process of gastric carcinogenesis. Materials and Methods: Eight gastric cancer cell lines and five gastric cancer tissues were studied. The expression of survivin and HIAP-1 were evaluated by reverse transcription -polymerase chain reaction (RT-PCR), immunohistochemistry, and Western blot. Results: Western blot and RT-PCR analysis revealed survivin and HIAP-1 expression in all gastric cancer cell lines. Increased expression of survivin and HIAP-1 were found in all cases of gastric cancer tissues compared to normal tissues by Western blot analysis. In immunohistochemical analysis tumor cells were stained with anti-survivin and anti-HIAP-1 antibodies. Cell cycle dependence of survivin expression was preserved in gastric cancer cell lines. Conclusion: The results indicate that increased expression of survivin and HIAP-1 genes may play an important role in gastric cancer.

  • PDF

G protein-coupled estrogen receptor-1 agonist induces chemotherapeutic effect via ER stress signaling in gastric cancer

  • Lee, Seon-Jin;Kim, Tae Woo;Park, Gyeong Lim;Hwang, Yo Sep;Cho, Hee Jun;Kim, Jong-Tae;Lee, Hee Gu
    • BMB Reports
    • /
    • 제52권11호
    • /
    • pp.647-652
    • /
    • 2019
  • G protein-coupled estrogen receptor (GPER) is known to play an important role in hormone-associated cancers. G-1, a novel synthetic GPER agonist, has been reported to exhibit anti-carcinogenic properties. However, the chemotherapeutic mechanism of GPER is yet unclear. Here, we evaluated GPER expression in human gastric cancer tissues and cells. We found that G-1 treatment attenuates GPER expression in gastric cancer. GPER expression increased G-1-induced antitumor effects in mouse xenograft model. We analyzed the effects of knockdown/overexpression of GPER on G-1-induced cell death in cancer cells. Increased GPER expression in human gastric cancer cells increased G-1-induced cell death via increased levels of cleaved caspase-3, -9, and cleaved poly ADP-ribose polymerase. Interestingly, during G-1-induced cell death, GPER mRNA and protein expression was attenuated and associated with ER stress-induced expression of PERK, ATF-4, GRP-78, and CHOP. Furthermore, PERK-dependent induction of ER stress activation increased G-1-induced cell death, whereas PERK silencing decreased cell death and increased drug sensitivity. Taken together, the data suggest that the induction of ER stress via GPER expression may increase G-1-induced cell death in gastric cancer cells. These results may contribute to a new paradigm shift in gastric cancer therapy.