• Title/Summary/Keyword: anti-proliferative effect

Search Result 247, Processing Time 0.024 seconds

A Tuber Lectin from Arisaema jacquemontii Blume with Anti-insect and Anti-proliferative Properties

  • Kaur, Manpreet;Singh, Kuljinder;Rup, Pushpinder Jai;Kamboj, Sukhdev Singh;Saxena, Ajit Kumar;Sharma, Madhunika;Bhagat, Madhulika;Sood, Sarvesh Kumar;Singh, Jatinder
    • BMB Reports
    • /
    • v.39 no.4
    • /
    • pp.432-440
    • /
    • 2006
  • A tuber lectin from Arisaema jacquemontii Blume belonging to family Araceae was purified by employing a single step affinity chromatography using column of asialofetuin-linked amino activated silica beads and the bound lectin was eluted with 100 mM glycine-HCl buffer pH 2.5. The purified A. jacquemontii lectin (AJL) showed a single protein band with an apparent molecular mass of 13.4 kDa when submitted to SDS-polyacrylamide gel electrophoresis under reducing as well as non-reducing conditions. The native molecular mass of AJL determined by gel filtration on a Biogel P-200 column was 52 kDa and its carbohydrate content was estimated to be 3.40%. Thus AJL is a tetrameric glycoprotein. The purified lectin agglutinated erythrocytes from rabbit but not from human. Its activity was not inhibited by any of the mono- and disaccharides tested except N-acetyl-D-lactosamine having minimal inhibitory sugar concentration (MIC) 25 mM. Among the glycoproteins tested only asialofetuin was found to be inhibitory (MIC $125\;{\mu}g/mL$). A single band was obtained in native PAGE at pH 4.5 while PAGE at pH 8.3 showed two bands. Isoelectric focusing of AJL gave multiple bands in the pI range of 4.6-5.5. When incorporated in artificial diet AJL significantly affected the development of Bactrocera cucurbitae (Coquillett) larvae indicating the possibility of using this lectin in a biotechnological strategy for insect management of cucurbits. Larvae fed on artificial diet containing sub-lethal dose of AJL showed a significant decrease in acid phosphatase and alkaline phosphatase activity while esterase activity markedly increased as compared to larvae fed on diet without lectin. Out of various human cancer cell lines employed in sulphorhodamine B (SRB) assay, this lectin was found to have appreciable inhibitory effect on the in vitro proliferation of HCT-15, HOP-62, SW-620, HT-29, IMR-32, SKOV-3, Colo-205, PC-3, HEP-2 and A-549 cancer cell lines by 82, 77, 73, 70, 41, 41, 37, 29, 21 and 21% respectively.

Effect of Arresting MCF-7 Human Breast Carcinoma Cell at G2/M Phase of Trichosanthes Kirilowii (천화분이 MCF-7 유방암 세포주의 G2/M 세포주기 억제에 미치는 영향)

  • Jeong, Seung-Min;Jeong, Mi-Kyung;Ko, Seong-Gyu;Choi, You-Kyung;Park, Jong-Hyeong;Jun, Chan-Yong
    • Journal of Physiology & Pathology in Korean Medicine
    • /
    • v.25 no.5
    • /
    • pp.857-862
    • /
    • 2011
  • The purpose of this study is to investigate the anti-proliferative mechanism by Trichosanthes kirilowii (TCK) in MCF-7 human breast carcinoma cell. In this study, we used human breast cancer cell line, Michigan cancer foundation-7 cells (MCF-7 cells). They were co-incubated with 30~200 ${\mu}g$/ml TCK for 48 hours, and cell viability was measured by Water-soluble tetrazolium salt-1 (WST-1) assay. After MCF-7 cells were exposed to 60 ${\mu}g$/ml of TCK for 0, 3, 6, 12, 24, 48 hours, We performed flow analysis cytometry sorting(FACS) and western blot analysis. We investigated the effect of dose-dependent cell growth inhibition by TCK, which could be proved by WST-1 assay. Also, flow cytometry analysis showed that TCK increased percentage of subG1 phase and G2/M phase cell cycle. In addition, TCK induced apoptosis through the expression of caspase-9, -3 and poly(ADP-ribose) polymerase(PARP) activation. Moreover, we showed that ATM-dependent G2/M phase arrest by DNA damage and phosphorylation of chk2, cdc25C, cdc2(Tyr15). Taken together, these results suggest that by G2/M phase arrest through DNA damage and inducing of apoptosis through intrinsic pathway, TCK may have potential tumor suppressor in breast cancer.

The Effects of Yunpyesan on Cell Proloferation, Apoptosis and Cell Cycle Progression of Human Lung Cancer A549 Cells (윤폐산에 의한 폐암세포 증식억제기전에 관한 연구)

  • Kang Yun-Keong;Park Dong Il;Lee Jun Hyuk;Choi Yung Hyun
    • Journal of Physiology & Pathology in Korean Medicine
    • /
    • v.16 no.4
    • /
    • pp.745-755
    • /
    • 2002
  • To examine the effects of Yunpyesan on the cell proliferation of A549 human lung carcinoma cell line, we performed various experiments such as dose-dependent effect of Yunpyesan on cell proliferation and viability, morphological changes, quantification of apoptotic cell death and alterations of apoptosis/cell cycle-regulatory gene products. Yunpyesan declined cell viability and proliferation in both a dose- and a time-dependent manner. The anti-proliferative effect by Yunpyesan treatment in A459 cells was associated with morphological changes such as membrane shrinking and cell rounding up. Yunpyesan Induced apoptotic cell death in a time-dependent manner, which was associated with degradation of poly-(ADP-ribose) polymerase (PARP), an apoptotic target protein, without alterations of the balance between Bcl-2 and Bax expressions. DNA flow cytometric histograms showed that population of G1 phase of the cell cycle was increased by Yunpyesan treatment in a dose-dependent manner. Western blot analysis revealed that cyclin D1 and A were reduced by Yunpyesan treatment, whereas cyclin dependent kinase (Cdk) inhibitor p27 was markedly increased in a time-dependent fashion. The level of tumor suppressor p53 proteins was also increased by Yunpyesan treatment and its increase might be linked to increase of Cdk inhibitor p27. In addition, Mdm2, negative regulator of p53, was down-regulated by Yunpyesan treatment. Since the expression of retinoblastome protein (pRB), a key regulator of G1/S progression, was reduced by Yunpyesan treatment, we supposed that phosphorylation of pRB might be also blocked. The present results indicated that Yunpyesan-induced inhibition of lung cancer cell proliferation is associated with the induction of apoptosis and the blockage of G1/S progression.

Effect of First Line Gastric Cancer Chemotherapy Regime on the AGS Cell Line - MTT Assay Results

  • Alizadeh-Navaei, Reza;Rafiei, Alireza;Abedian-Kenari, Saeid;Asgarian-Omran, Hossein;Valadan, Reza;Hedayatizadeh-Omran, Akbar
    • Asian Pacific Journal of Cancer Prevention
    • /
    • v.17 no.1
    • /
    • pp.131-133
    • /
    • 2016
  • Background: Combination chemotherapy regimes are common treatments for cancer. The aim of this study was to evaluation the effect of individual chemotherapeutic agents in comparison with a first line chemotherapy regime treatment in the AGS gastric cancer cell line by MTT assay. Materials and Methods: In this experimental study, AGS cells were grown in RPMI-1640 supplemented with 10% fetal calf serum and 100 IU/ml penicillin, and $10{\mu}g/ml$ streptomycinin, under a humidified condition at $37^{\circ}C$ with 5% CO2. All cells were washed with PBS and detached with trypsin, centrifuged and 8000 cells re-plated on to 96- well plates. LD50 doses of Epirubicin, Cisplatin and 5-fluorouracil were added to each well in mono or triple therapy. Anti-proliferative activities were determined by MTT assay after 24, 48 or 72 h. Results: Results of MTT assays showed that there were no significant differences among 3 drugs in monotherapy (p=0.088), but there was significant difference between combination therapy with epirubicin (P=0.031) and 5FU (p=0.013) on cell survival at 24 h. After 48 and 72 hours, cell viability showed significant differences between the 3 drugs (p=0.048 and P=0.000 for 48 and 72 h, respectively) and there was significant difference between combination therapy with epirubicin (P=0.035 and P=0.002 for 48 and 72 h, respectively). Conclusions: The results showed no significant differences between these chemotherapy drugs each given alone, but combination therapy with 3 drugs had significant effects on cell viability in comparison with epirubicin alone.

Anti-proliferative Effect of Paclitaxel in Multicellular Layers of Human Cancer Cells (다층 배양된 암세포에서 파크리탁셀의 항증식효과 분석)

  • Kang, Choon-Mo;Lee, Joo-Ho;Cha, Jung-Ho;Kuh, Hyo-Jeong
    • Journal of Pharmaceutical Investigation
    • /
    • v.36 no.1
    • /
    • pp.1-9
    • /
    • 2006
  • Human solid tumors exhibit a multicellular resistance (MCR) resulting from limited drug penetration and decreased sensitivity of tumor cells when interacting with their microenvironments. Multicellular cultures represent solid tumor condition in vivo and provide clinically relevant data. There is little data on antitumor effect of paclitaxel (PTX) in multicellular cultures although its MCR has been demonstrated. In the present study, we evaluated antiproliferative effects of PTX in multicellular layers (MCL) of DLD-1 human colorectal carcinoma cells. BrdU labeling index (LI), thickness of MCL, cell cycle distribution and cellular uptake of calcein were measured before and after exposure to PTX at 0.1 to 50 ${\mu}M$ for 24, 48 and 72 hrs. BrdU LI and thickness of MCL showed a concentration- and time-dependent decrease and the changes in both parameters were similar, i.e., 34.2% and 40.6% decrease in BrdU LI and thickness, respectively, when exposed to $50\;{\mu}M$ for 72 hr. The DLD-1 cells grown in MCL showed increase in $%G_{0}/G_{1}$ and resistance to cell cycle arrest and apoptosis compared to monolayers. Calcein uptake in MCL did not change upon PTX exposure, indicating technical problems in multicellular system. Overall, these data indicate that antitumor activity of PTX may be limited in human solid tumors (a multicellular system) and MCL may be an appropriate model to study further pharmacodynamics of PTX.

A Study on the Effects of Anticareinogenie Activity of Chondria Crassicaulis (서실 분획물의 암예방효과)

  • Jeon Kwang-Hye;Shin Mi-Ok;Bae Song-Ja
    • Journal of Nutrition and Health
    • /
    • v.38 no.7
    • /
    • pp.503-511
    • /
    • 2005
  • In this study we investigated the biological activity of Chondria Crassicaulis (CC) on the human cancer cells. CC was extracted with methanol and further fractionated into four different types: hexane (CCMH), methanol (CCMM), butanol (CCMB), and aqueous (CCMA) partition layers. We determined the cytotoxic effect of these layers on human cancer cells by MTT assay. Among various partition layers of CC, the CCMM and CCMB showed the strong cytotoxic effects at 150 ${\mu}g$/ml which resulted $98.91\%$, $92.96\%$ on HeLa cell lines and $95.47\%$, $77.05\%$ on MCF-7 cell lines. And, the anti-proliferative effect of CC was accompanied by a marked inhibition of cyclooxygenase (COX-2), Caspase-3 and IAP (cIAP-1, cIAP-2 and XIAP) protein and concomitant induction of p53, p21 and Survivin protein. However, CC did not affect the level of Bax, Bcl-2 and Bcl-XL- protein. Also, we observed quinone reductase (QR) induced effects in all fraction layers of CC on HepG2 cells. The QR induced effects of the CCMH and CCMM on HePG2 cells at 120 ${\mu}g$/mL concentration indicated 3.73 and 2.45 with the control value of 1.0. Although further studies are needed, the present work suggests that CC may be a chemopreventive agent for the treatment of human cancer cells.

Apoptotic activity of demethoxycurcumin in MG-63 human osteosarcoma cells

  • Kang, Kyeong-Rok;Kim, Jae-Sung;Kim, Tae-Hyeon;Seo, Jeong-Yeon;Park, Jong-Hyun;Chun, Hong Sung;Yu, Sun-Kyoung;Kim, Heung-Joong;Kim, Chun Sung;Kim, Do Kyung
    • International Journal of Oral Biology
    • /
    • v.46 no.1
    • /
    • pp.23-29
    • /
    • 2021
  • Demethoxycurcumin (DMC), which is a curcuminoid found in turmeric, has anti-proliferative effects on cancer cells. However, the effect of DMC on osteosarcoma has not been established. The aim of this study was to examine the effects of DMC on cell growth and apoptosis induction in MG-63 human osteosarcoma cells. This study was investigated using 3-[4, 5-dimethylthiazol-2-yl]-2, 5 diphenyl tetrazolium bromid assay, Live/Dead cell assay, 4', 6-diamidino-2-phenylindole staining, and immunoblotting in MG-63 cells. DMC induced MG-63 cell death in a dose-dependent manner, with an estimated IC50 value of 54.4 µM. DMC treatment resulted in nuclear condensation in MG-63 cells. DMC-induced apoptosis in MG-63 cells was mediated by the expression of Fas and activation of caspase-8, caspase-3, and poly (ADP-ribose) polymerase. Immunoblotting results showed that Bcl-2 and Bcl-xL were downregulated, while Bax and Bad were upregulated by DMC in MG-63 cells. These results indicated that DMC inhibits cell proliferation and induces apoptotic cell death in MG-63 human osteosarcoma cells via the death receptor-mediated extrinsic apoptotic pathway and mitochondria-mediated intrinsic apoptotic pathway.

8-Methoxypsoralen Induces Apoptosis by Upregulating p53 and Inhibits Metastasis by Downregulating MMP-2 and MMP-9 in Human Gastric Cancer Cells

  • Eun Kyoung, Choi;Hae Dong, Kim;Eun Jung, Park;Seuk Young, Song;Tien Thuy, Phan;Miyoung, Nam;Minjung, Kim;Dong-Uk, Kim;Kwang-Lae, Hoe
    • Biomolecules & Therapeutics
    • /
    • v.31 no.2
    • /
    • pp.219-226
    • /
    • 2023
  • Furanocoumarin 8-methoxypsoralen (8-MOP) is the parent compound that naturally occurs in traditional medicinal plants used historically. 8-MOP has been employed as a photochemotherapeutic component of Psoralen + Ultraviolet A (PUVA) therapy for the treatment of vitiligo and psoriasis. Although the role of 8-MOP in PUVA therapy has been studied, little is known about the effects of 8-MOP alone on human gastric cancer cells. In this study, we observed anti-proliferative effect of 8-MOP in several human cancer cell lines. Among these, the human gastric cancer cell line SNU1 is the most sensitive to 8-MOP. 8-MOP treated SNU1 cells showed G1-arrest by upregulating p53 and apoptosis by activating caspase-3 in a dose-dependent manner, which was confirmed by loss-of-function analysis through the knockdown of p53-siRNA and inhibition of apoptosis by Z-VAD-FMK. Moreover, 8-MOP-induced apoptosis is not associated with autophagy or necrosis. The signaling pathway responsible for the effect of 8-MOP on SNU1 cells was confirmed to be related to phosphorylated PI3K, ERK2, and STAT3. In contrast, 8-MOP treatment decreased the expression of the typical metastasis-related proteins MMP-2, MMP-9, and Snail in a p53-independent manner. In accordance with the serendipitous findings, treatment with 8-MOP decreased the wound healing, migration, and invasion ability of cells in a dose-dependent manner. In addition, combination treatment with 8-MOP and gemcitabine was effective at the lowest concentrations. Overall, our findings indicate that oral 8-MOP has the potential to treat early human gastric cancer, with fewer side effects.

Modulacon of Cell Cycle Control by Histone Deacetylase Inhibitor Trichostatin A in A549 Human Non-small Cell Lung Cancer Cells (인체폐암세포 A549의 세포주기 조절인자에 미치는 histone deacetylase inhibitor trichostatin A의 영향)

  • Hwang Ji Won;Kim Young Min;Hong Su Hyun;Choi Byung Tae;Lee Won Ho;Choi Yung Hyun
    • Journal of Life Science
    • /
    • v.15 no.5 s.72
    • /
    • pp.726-733
    • /
    • 2005
  • Histone deacetylase (HDAC) inhibitors target key steps of tumor development. They inhibit proliferation, induce differentiation and/or apoptotic cell death, and exhibit potent antimetastatic and antiangiogenic properties in cancer cells in vitro and in vivo. Although they are emerging as a promising new treatment strategy in malignancy, how they exert their effect on human non-small cell lung cancer cells is as yet unclear. The present study was undertaken to investiate the underlying mechanism of a HDAC inhibitor trichostatin A (TSA)-induced growth arrest and its effect on the cell cycle control gene products in a human lung carcinoma cell line A549. TSA treaoent induced the growth inhibition and morphological changes in a concentration-dependent manner. Treatment of A549 cells with TSA resulted in a concentration-dependent increased G1 (under 100 ng/ml) and/or G2/M (200 ng/ml) cell population of the cell cycle as determined by flow cytometry Moreover, 200 ng/ml TSA treatment significantly induced the population of sub-G1 cells (23.0 fold of control). This anti-proliferative effect of TSA was accompanied by a marked inhibition of cyclins, positive regulators of cell cycle progression, and cyclin-dependent kinases (Cdks) expression and concomitant induction of tumor suppressor p53 and Cdk inhibitors such as p21 and p27 Although further studies are needed, these findings provide important insights into the possible molecular mechanisms of the anti-cancer activity of TSA in human lung carcinoma cells.

Induction of G1 Arrest by Methanol Extract of Lycopus lucidus in Human Lung Adenocarcinoma A549 Cells (택란 메탄올 추출물에 의한 인체 폐암 세포주 A549의 G1 arrest 유발)

  • Park, Hyun-Jin;Jin, Soojung;Oh, You Na;Yun, Seung-Geun;Lee, Ji-Young;Kwon, Hyun Ju;Kim, Byung Woo
    • Journal of Life Science
    • /
    • v.23 no.9
    • /
    • pp.1109-1117
    • /
    • 2013
  • Induction of G1 Arrest by Methanol Extract of Lycopus lucidus in Human Lung Adenocarcinoma A549 Cells Lycopus lucidus, a herbaceous perennial, is used as a traditional remedy in East Asia, including China and Korea. It has been reported that L. lucidus has anti-allergic effects, inhibitory effects on cholesterol acyltransferase in high glucose-induced vascular inflammation, and anti-proliferative effects in human breast cancer cells. However, the molecular mechanisms of the anti-cancer effects of L. lucidus have not yet been fully determined. In this study, we evaluated the anti-cancer effect and the mechanism of action of L. lucidus in human lung adenocarcinoma A549 cells using methanol extracts of L. lucidus (MELL). MELL treatment showed cytotoxic activity in a dose-dependent manner and induced G1 arrest in A549 cells. The induction of G1 arrest by MELL was associated with the up-regulation of phospho-CHK2 and the down-regulation of Cdc25A phosphatase. In addition, MELL treatment induced decreased expression of G1/S transition-related proteins, including CDK2, CDK4, CDK6, cyclin D1 and cyclin E. MELL also regulated the mRNA expression of CDK2 and cyclin E. On the other hand, the expression of p53 and the cyclin-dependent kinase inhibitor p21 was not induced by MELL. Collectively, these results suggest that MELL may exert an anti-cancer effect by cell cycle arrest at G1 phase through the ATM/CHK2/Cdc25A/CDK2 pathway in A549 cells.