• Title/Summary/Keyword: Cell proliferation and growth

Search Result 1,578, Processing Time 0.027 seconds

Snail Promotes Cancer Cell Proliferation via Its Interaction with the BIRC3

  • Rho, Seung Bae;Byun, Hyun-Jung;Kim, Boh-Ram;Lee, Chang Hoon
    • Biomolecules & Therapeutics
    • /
    • v.30 no.4
    • /
    • pp.380-388
    • /
    • 2022
  • Snail is implicated in tumour growth and metastasis and is up-regulated in various human tumours. Although the role of Snails in epithelial-mesenchymal transition, which is particularly important in cancer metastasis, is well known, how they regulate tumour growth is poorly described. In this study, the possible molecular mechanisms of Snail in tumour growth were explored. Baculoviral inhibitor of apoptosis protein (IAP) repeat-containing protein 3 (BIRC3), a co-activator of cell proliferation during tumourigenesis, was identified as a Snail-binding protein via a yeast two-hybrid system. Since BIRC3 is important for cell survival, the effect of BIRC3 binding partner Snail on cell survival was investigated in ovarian cancer cell lines. Results revealed that Bax expression was activated, while the expression levels of anti-apoptotic proteins were markedly decreased by small interfering RNA (siRNA) specific for Snail (siSnail). siSnail, the binding partner of siBIRC3, activated the tumour suppressor function of p53 by promoting p53 protein stability. Conversely, BIRC3 could interact with Snail, for this reason, the possibility of BIRC3 involvement in EMT was investigated. BIRC3 overexpression resulted in a decreased expression of the epithelial marker and an increased expression of the mesenchymal markers. siSnail or siBIRC3 reduced the mRNA levels of matrix metalloproteinase (MMP)-2 and MMP-9. These results provide evidence that Snail promotes cell proliferation by interacting with BIRC3 and that BIRC3 might be involved in EMT via binding to Snail in ovarian cancer cells. Therefore, our results suggested the novel relevance of BIRC3, the binding partner of Snail, in ovarian cancer development.

Silencing of the COPS3 Gene by siRNA Reduces Proliferation of Lung Cancer Cells Most Likely via induction of Cell Cycle Arrest and Apoptosis

  • Wang, Xue-Mei;Cui, Jiu-Wei;Li, Wei;Cai, Lu;Song, Wei;Wang, Guan-Jun
    • Asian Pacific Journal of Cancer Prevention
    • /
    • v.13 no.3
    • /
    • pp.1043-1048
    • /
    • 2012
  • The COPS3 gene has stimulating effect on cell proliferation and progression of osteosarcomas and related cells. However, the features of COPS3 and its potential application as a therapeutic target in other cancers has not yet been studied. In this study, therefore, the effect of COPS3 silencing via COPS3 siRNA on lung cancer cell proliferation was examined. Expression levels of COPS3 gene in COPS3 siRNA infected cells and control siRNA infected cells were compared with real time PCR and Western blot analysis. Cell proliferation levels were comprehensively analyzed by MTT, BrdU incorporationy, and colony formation assays. For mechanistic assessment the effects of COPS3 silencing on cell cycle and apoptosis were analyzed using flow cytometry. Results showed that successful silencing of the COPS3 gene at both translational and transcriptional levels significantly reduced the proliferation and colony formation by lung cancer cells (p<0.01). Flow cytometry showed cell cycle arrest in the G0/G1 phase after COPS3 silencing, and more importantly, apoptosis was induced as a result of COPS3 knockdown, which negatively affected cell survival. Therefore, these results provide another piece of important evidence that the COPS3 gene expressed in lung cancer cells may play a critical role in stimulating proliferation. Down-regulation of COPS3 could significantly inhibit lung cancer cell growth, which was most likely mediated via induction of cell cycle arrest in G0/G1 phase and apoptosis.

YAC tripeptide of epidermal growth factor promotes the proliferation of HaCaT keratinocytes through activation of EGFR

  • Yoo, Yeon Ho;Kim, Yu Ri;Kim, Min Seo;Lee, Kyoung-Jin;Park, Kyeong Han;Hahn, Jang-Hee
    • BMB Reports
    • /
    • v.47 no.10
    • /
    • pp.581-586
    • /
    • 2014
  • Epidermal growth factor (EGF) is known to play key roles in skin regeneration and wound-healing. Here, we demonstrate that Pep2-YAC, a tripeptide covering residues 29-31 in the B loop of EGF, promotes the proliferation of HaCaT keratinocytes with activity comparable to EGF. The treatment of HaCaT cells with Pep2-YAC induced phosphorylation, internalization, and degradation of EGFR and organization of signaling complexes, which consist of Grb2, Gab1, SHP2, and PI3K. In addition, it stimulated the phosphorylation of ERK1/2 at Thr 202/Tyr 204 and of Akt1 at Ser 473 and the nuclear translocation of EGFR, STAT3, c-Jun, and c-Fos. These results suggest that Pep2-YAC may be useful as a therapeutic agent for skin regeneration and wound-healing as an EGFR agonist.

Growth inhibition in head and neck cancer cell lines by gefitinib, an epidermal growth factor receptor tyrosine kinase inhibitor (두경부암 세포주에서 상피성장인자수용체 타이로신 카이네이즈 억제제인 gefitinib의 성장억제에 관한 연구)

  • Song, Seung-Il;Kim, Myung-Jin
    • Journal of the Korean Association of Oral and Maxillofacial Surgeons
    • /
    • v.35 no.5
    • /
    • pp.287-293
    • /
    • 2009
  • Cell survival is the result of a balance between programmed cell death and cellular proliferation. Cell membrane receptors and their associated signal transducing proteins control these processes. Of the numerous receptors and signaling proteins, epidermal growth factor receptor (EGFR) is one of the most important receptors involved in signaling pathways implicated in the proliferation and survival of cancer cells. EGFR is often highly expressed in human tumors including oral squamous cell carcinomas, and there is increasing evidence that high expression of EGFR is correlated with poor clinical outcome of common human cancers. Therefore, we examined the antiproliferative activity of gefitinib, epidermal growth factor receptor tyrosine kinase inhibitor (EGFR TKI), in head and neck cancer cell lines. SCC-9, KB cells were cultured and growth inhibition activity of gefitinib was measured with MTT assay. To study influence of gefitinib in cell cycle, we performed cell cycle analysis with flow cytometry. Western blot was done to elucidate the expression of EGFR in cell lines and phosphorylation of EGFR and downstream kinase protein, Erk and Akt. Significant growth inhibition was observed in SCC-9 cells in contrast with KB cells. Also, flow cytometric analysis showed G1 phase arrest only in SCC-9 cells. In Western blot analysis for investigation of EGFR expression and downstream molecule phosphorylation, gefitinib suppressed phosphorylation of EGFR and downstream protein kinase Erk, Akt in SCC-9. However, in EGFR positive KB cells, weak expression of active form of Erk and Akt and no inhibitory activity of phosphorylation in Erk and Akt was observed. The antiproliferative activity of gefitinib was not correlated with EGFR expression and some possibility of phosphorylation of Erk and Akt as a predictive factor of gefitinib response was emerged. Further investigations on more reliable predictive factor indicating gefitinib response are awaited to be useful gefitinib treatment in head and neck cancer patients.

Caffeic Acid Phenethyl Ester Inhibits Cell Proliferation and Induces Apoptosis in Human Ovarian Cancer Cells

  • Park, Hyung-Joo;Yang, Seung-Joo;Mo, Jin-Young;Ryu, Geun-Chang;Lee, Kyung-Jin
    • Korean Journal of Environmental Biology
    • /
    • v.28 no.4
    • /
    • pp.196-201
    • /
    • 2010
  • The phenethyl ester of caffeic acid (CAPE), an active component of honeybee propolis extract, is shown to inhibit cancer growth previously. However, studies on human ovarian cancer are largely obscure. This study evaluated the effects of CAPE as a potential anti-proliferative and pro-apoptotic agent in the human ovarian cancer line, OVCAR-3. CAPE treated OVCAR-3 cells showed inhibition of cell viability and proliferation in a dose-dependent manner by WST-1 assay, LDH assay and bromodeoxyuridine (BrdU) incorporation assay. Furthermore, CAPE-mediated OVCAR-3 cell growth inhibition was associated with apoptotic changes as evident by cell cycle arrest and accumulation of cells in the apoptotic phase and DNA fragmentation. Taken together, CAPE inhibits cell proliferation via DNA synthesis reduction and induces apoptotic cell death via DNA damage, thus elucidating a novel, plausible mechanism of CAPE anti-tumorigenic property in OVCAR-3 cells.

Expression of Stat5a Gene in Bovine Mammary Gland and its Effect on Proliferation of Mammary Epithelial Cells

  • Jeon, D.H.;Choi, Y.J.;Baik, M.G.
    • Asian-Australasian Journal of Animal Sciences
    • /
    • v.15 no.8
    • /
    • pp.1198-1203
    • /
    • 2002
  • To understand molecular mechanisms involved in bovine mammary gland growth, expression of stat5a gene was examined in bovine mammary tissues. We found that stat5a gene was highly induced at pregnant 7 and 8 months compared to virgin mammary tissues. To examine function of bovine stat5a in mammary epithelial cell proliferation, stat5a expression vector was transfected into mammary epithelial HC11 cells. Cell proliferation rate in stat5a gene-transfected cells was 26%, 95% and 85% higher at 24 h, 48 h and 72 h after seeding, respectively, compared to control vector-transfected cells. Results demonstrate that bovine stat5a enhances proliferation of mammary epithelial cells.

Short-Hairpin RNA-Mediated MTA2 Silencing Inhibits Human Breast Cancer Cell Line MDA-MB231 Proliferation and Metastasis

  • Lu, Jun;Jin, Mu-Lan
    • Asian Pacific Journal of Cancer Prevention
    • /
    • v.15 no.14
    • /
    • pp.5577-5582
    • /
    • 2014
  • Objective: To observe the effects of metastasis-associated tumor gene family 2 (MTA2) depletion on human breast cancer cell proliferation and metastasis. Methods: A short-hairpin RNA targeting MTA2 was chemically synthesized and transfected into a lentivirus to construct Lv-shMTA2 for infection into the MDA-MB231 human breast cancer cell line. At 48 hours after infection cells were harvested and mRNA and protein levels of MTA2 were determined by quantitative real-time polymerase chain reaction (qRT-PCR) and Western blotting, respectively. Cell viability and metastasis were assessed by CCK-8, wound-healing assay and Transwell assay, respectively. In addition, a xenograft model of human breast cancer was constructed to investigate cancerous cell growth and capacity for metastasis. Results: After infection with Lv-shMTA2, mRNA and protein levels of MTA2 was significantly reduced (p<0.05) and MDA-MB231 cell proliferation and metastasis were inhibited (p<0.05). In addition, mean tumor size was smaller than that in control group nude mice (p<0.05) and numbers of metastatic deposits in lung were lower than in control group mice (p<0.05). Depletion of MTA2 affected MMP-2 and apoptosis-related protein expression. Conclusions: For the first time to our knowledge we showed that MTA2 depletion could significantly inhibit human breast cancer cell growth and metastasis, implying that MTA2 might be involved in the progression of breast cancer. The role of MTA2 in breast cancer growth and metastasis might be linked with regulation of matrix metalloproteinase and apoptosis.

The Role of Receptor Activator of NF-κ Ligand in Smooth Muscle Cell Proliferation (Smooth muscle cell 증식에 있어 NF-κ ligand의 receptor activator의 역할)

  • Kim, Hyun-Ju
    • Journal of Life Science
    • /
    • v.16 no.6
    • /
    • pp.1066-1070
    • /
    • 2006
  • Smooth muscle cell (SMC) proliferation is important in the pathogenesis of vascular proliferative disorders. Understanding of the molecular mechanism underlying SMC growth after arterial injury would have therapeutic implications. Here we report that receptor activator of $NF-{\kappa}B$ ligand (RANKL), a member of tumor necrosis factor (TNF) family, promotes the proliferation of SMC, leading to decreased expression of p21 and enhancement of SMC growth. ERK and p38 phosphorylation was enhanced after RANKL treatment in SMC. Inhibition of ERK/p38 MAPK activity by PD98059/SB203580 completely abolished RANKL-induced proliferation of SMC, indicating ERK and p38 MAPK are essential for RANKL-induced SMC proliferation. Taken together, our findings demonstrate that RANK-RANKL-ERK/p38 pathway is important for proliferation of SMC and that these molecules may be the new therapeutic targets for the prevention of vascular diseases.

The Study of Aati-cancer Effects of Bee Venom for Aqua-acupuncure (약침용(藥鍼用) 봉독성분(蜂毒成分) 중(中) Apamin, Melittin의 항암작용(抗癌作用))

  • Kwon, Do-Hee;Lee, Jae-dong;Choi, Do-Yong
    • Journal of Acupuncture Research
    • /
    • v.18 no.1
    • /
    • pp.129-145
    • /
    • 2001
  • Objectives : To characterize the antitumorigenic potential of three representative bee venom components, Melittin, Apamin, and Phospholipase A2, their effects on cell proliferation and apotosis of the human melanoma cell line SK-MEL-2 were analyzed using molecular biological approaches. Methodes & Results : To determine the doses of the drugs that do not induce cytotoxic damage to this cell line, cell viability was examined by MTT assay. While SK-MEL-2 cells treated with 0.5 - 2.0㎍/㎖ of each drug showed no recognizable cytotoxic effect, marked reductions of cell viability were detected at concentrations over 5.0㎍/㎖. [3H]thymidine incorporation assay for cell proliferation demonstrated that DNA replication of SK-MEL-2 cells is inhibited by Apamin and Phospholipase A2 in a dose-dependent manner. Consistent with this result, the cells were accumulated at the G1 phase of the cell cycle after treatment with Apamin and Phospholipase A2, whereas no detectable change in cell proliferation was identified by Melittin treatment. In addition, tryphan blue exclusion and flow cytometric analyses showed that all of these drugs can trigger apoptotic cell death of SK-MEL-2, suggesting that Melittin, Apamin, and Phospholipase A2 have antitumorigenic potential through the suppression of cell growth and/or induction of apoptosis. Qantitative RT-PCR analysis revealed that Apamin and Phospholipase A2 inhibit expression of growth-promoting genes such as c-Jun, c-Fos, and Cyciin D1. Furthermore, Phospholipase A2 induced tumor suppressors p53 and p21/Wafl. In addition, all three drugs were found to activate expression of a representative apoptosis-inducing gene Bax while expression of apoptosis-suppressing Bcl-2 and Bcl-XL genes was not changed. Taken together, this study strongly suggests that Metittin, Apamin, and Phosphalipase A2 may have antitumorigenic activities, which are associated with its growth-inhibiting and/or apoptosis-inducing potentials.

  • PDF

Effects of Ginsenosides $Rg_3$ and $Rh_2$ OH the Proliferation of Prostate Cancer Cells

  • Kim Hyun-Sook;Lee Eun-Hee;Ko Sung-Ryong;Choi Kang-Ju;Park Jong-Hee;Im Dong-Soon
    • Archives of Pharmacal Research
    • /
    • v.27 no.4
    • /
    • pp.429-435
    • /
    • 2004
  • Ginseng has an anti-cancer effect in several cancer models. This study was to characterize active constituents of ginseng and their effects on proliferation of prostate cancer cell lines, LNCaP and PC3. Cell proliferation was measured by $[^3H]$thymidine incorporation, the intracellular calcium concentration by a dual-wavelength spectrophotometer system, effects on mite-gen-activated protein (MAP) kinases by Western blotting, and cell attachment and morphologic changes were observed under a microscope. Among 11 ginsenosides tested, ginsenosides $Rg_3\;and\;Rh_2$ inhibited the proliferation of prostate cancer cells. $EC_{50}s\;of\;Rg_3\;and\;Rh_2$ on PC3 cells were $8.4{\mu}M\;and\;5.5{\mu}M$, respectively, and $14.1{\mu}M\;and\;4.4{\mu}M$ on LNCaP cells, respectively. Both ginsenosides induced cell detachment and modulated three modules of MAP kinases activities differently in LNCaP and PC3 cells. These results suggest that ginsenosides $Rg_3\;and\;Rh_2$-induced cell detachment and inhibition of the proliferation of prostate cancer cells may be associated with modulation of three modules of MAP kinases.