• Title/Summary/Keyword: Clonogenic assay

Search Result 60, Processing Time 0.029 seconds

Mechanism Underlying a Proteasome Inhibitor, Lactacystin-Induced Apoptosis on SCC25 Human Tongue Squamous Cell Carcinoma Cells (사람혀편평상피세포암종세포에서 proteasome 억제제인 lactacystin에 의해 유도된 세포자멸사의 기전에 대한 연구)

  • Baek, Chul-Jung;Kim, Gyoo-Cheon;Kim, In-Ryoung;Lee, Seung-Eun;Kwak, Hyun-Ho;Park, Bong-Soo;Tae, Il-Ho;Ko, Myung-Yun;Ahn, Yong-Woo
    • Journal of Oral Medicine and Pain
    • /
    • v.34 no.3
    • /
    • pp.261-276
    • /
    • 2009
  • Lactacystin, a microbial natural product synthesized by Streptomyces, has been commonly used as a selective proteasome inhibitor in many studies. Proteasome inhibitors is known to be preventing the proliferation of cancer cells in vivo as well as in vitro. Furthermore, proteasome inhibitors, as single or combined with other anticancer agents, are suggested as a new class of potential anticancer agents. This study was undertaken to examine in vitro effects of cytotoxicity and growth inhibition, and the molecular mechanism underlying induction of apoptosis in SCC25 human tongue sqaumous cell carcinoma cell line treated with lactacystin. The viability of SCC25 cells, human normal keratinocytes (HaCaT cells) and human gingiva fibroblasts (HGF-1 cells), and the growth inhibition of SCC25 cells were assessed by MTT assay and clonogenic assay respectively. The hoechst staining, hemacolor staining and TUNEL staining were conducted to observe SCC25 cells undergoing apoptosis. SCC25 cells were treated with lactacystin, and Western blotting, immunocytochemistry, confocal microscopy, FAScan flow cytometry, MMP activity, and proteasome activity were performed. Lactacystin treatment of SCC25 cells resulted in a time- and does-dependent decrease of cell viability and a does-dependent inhibition of cell growth, and induced apoptotic cell death. Interestingly, lactacytin remarkably revealed cytotoxicity in SCC25 cells but not normal cells. And tested SCC25 cells showed several lines of apoptotic manifestation such as nuclear condensation, DNA fragmentation, the reduction of MMP and proteasome activity, the decrease of DNA contents, the release of cytochrome c into cytosol, the translocation of AIF and DFF40 (CAD) onto nuclei, the up-regulation of Bax, and the activation of caspase-7, caspase-3, PARP, lamin A/C and DFF45 (ICAD). Flow cytometric analysis revealed that lactacystin resulted in G1 arrest in cell cycle progression which was associated with up-regulation in the protein expression of CDK inhibitors, $p21^{WAF1/CIP1}$ and $p27^{KIP1}$. We presented data indicating that lactacystin induces G1 cell cycle arrest and apoptois via proteasome, mitochondria and caspase pathway in SCC25 cells. Therefore our data provide the possibility that lactacystin could be as a novel therapeutic strategy for human tongue squamous cell carcinoma.

Plumbagin from Plumbago Zeylanica L Induces Apoptosis in Human Non-small Cell Lung Cancer Cell Lines through NF-κB Inactivation

  • Xu, Tong-Peng;Shen, Hua;Liu, Ling-Xiang;Shu, Yong-Qian
    • Asian Pacific Journal of Cancer Prevention
    • /
    • v.14 no.4
    • /
    • pp.2325-2331
    • /
    • 2013
  • Objective: To detect effects of plumbagin on proliferation and apoptosis in non-small cell lung cancer cell lines, and investigate the underlying mechanisms. Materials and Methods: Human non-small cell lung cancer cell lines A549, H292 and H460 were treated with various concentrations of plumbagin. Cell proliferation rates was determined using both cell counting kit-8 (CCK-8) and clonogenic assays. Apoptosis was detected by annexin V/propidium iodide double-labeled flow cytometry and TUNEL assay. The levels of reactive oxygen species (ROS) were detected by flow cytometry. Activity of NF-${\kappa}B$ was examined by electrophoretic mobility shift assay (EMSA) and luciferase reporter assay. Western blotting was used to assess the expression of both NF-${\kappa}B$ regulated apoptotic-related gene and activation of p65 and $I{\kappa}B{\kappa}$. Results: Plumbagin dose-dependently inhibited proliferation of the lung cancer cells. The IC50 values of plumbagin in A549, H292, and H460 cells were 10.3 ${\mu}mol/L$, 7.3 ${\mu}mol/L$, and 6.1 ${\mu}mol/L$ for 12 hours, respectively. The compound concentration-dependently induced apoptosis of the three cell lines. Treatment with plumbagin increased the intracellular level of ROS, and inhibited the activation of NK-${\kappa}B$. In addition to inhibition of NF-${\kappa}B$/p65 nuclear translocation, the compound also suppressed the degradation of $I{\kappa}B{\kappa}$. ROS scavenger NAC highly reversed the effect of plumbagin on apoptosis and inactivation of NK-${\kappa}B$ in H460 cell line. Treatment with plumbagin also increased the activity of caspase-9 and caspase-3, downregulated the expression of Bcl-2, upregulated the expression of Bax, Bak, and CytC. Conclusions: Plumbagin inhibits cell growth and induces apoptosis in human lung cancer cells through an NF-${\kappa}B$-regulated mitochondrial-mediated pathway, involving activation of ROS.

Dexamethasone Interferes with Autophagy and Affects Cell Survival in Irradiated Malignant Glioma Cells

  • Komakech, Alfred;Im, Ji-Hye;Gwak, Ho-Shin;Lee, Kyue-Yim;Kim, Jong Heon;Yoo, Byong Chul;Cheong, Heesun;Park, Jong Bae;Kwon, Ji Woong;Shin, Sang Hoon;Yoo, Heon
    • Journal of Korean Neurosurgical Society
    • /
    • v.63 no.5
    • /
    • pp.566-578
    • /
    • 2020
  • Objective : Radiation is known to induce autophagy in malignant glioma cells whether it is cytocidal or cytoprotective. Dexamethasone is frequently used to reduce tumor-associated brain edema, especially during radiation therapy. The purpose of the study was to determine whether and how dexamethasone affects autophagy in irradiated malignant glioma cells and to identify possible intervening molecular pathways. Methods : We prepared p53 mutant U373 and LN229 glioma cell lines, which varied by phosphatase and tensin homolog (PTEN) mutational status and were used to make U373 stable transfected cells expressing GFP-LC3 protein. After performing cell survival assay after irradiation, the IC50 radiation dose was determined. Dexamethasone dose (10 μM) was determined from the literature and added to the glioma cells 24 hours before the irradiation. The effect of adding dexamethasone was evaluated by cell survival assay or clonogenic assay and cell cycle analysis. Measurement of autophagy was visualized by western blot of LC3-I/LC3-II and quantified by the GFP-LC3 punctuated pattern under fluorescence microscopy and acridine orange staining for acidic vesicle organelles by flow cytometry. Results : Dexamethasone increased cell survival in both U373 and LN229 cells after irradiation. It interfered with autophagy after irradiation differently depending on the PTEN mutational status : the autophagy decreased in U373 (PTEN-mutated) cells but increased in LN229 (PTEN wild-type) cells. Inhibition of protein kinase B (AKT) phosphorylation after irradiation by LY294002 reversed the dexamethasone-induced decrease of autophagy and cell death in U373 cells but provoked no effect on both autophagy and cell survival in LN229 cells. After ATG5 knockdown, radiation-induced autophagy decreased and the effect of dexamethasone also diminished in both cell lines. The diminished autophagy resulted in a partial reversal of dexamethasone protection from cell death after irradiation in U373 cells; however, no significant change was observed in surviving fraction LN229 cells. Conclusion : Dexamethasone increased cell survival in p53 mutated malignant glioma cells and increased autophagy in PTEN-mutant malignant glioma cell but not in PTEN-wildtype cell. The difference of autophagy response could be mediated though the phosphatidylinositol 3-kinase/AKT/mammalian target of rapamycin signaling pathway.

Reduction of Radiation-induced Chromosome Aberration and Apoptosis by Dithiothreitol

  • Kim, Jeong Hee;Lee, Eun Ju;Hyun, Jin Won;Kim, Sung Ho;Mar, Woongchon;Kim, Jin Kyu
    • Archives of Pharmacal Research
    • /
    • v.21 no.6
    • /
    • pp.683-687
    • /
    • 1998
  • We have examined in vitro and in vivo radioprotective effects of a well-known thiol-containing compound, dithiothreitol (DTT). The treatment of both 0.5 and 1mM of DTT significantly increased clonogenic survival of ${\gamma}$-ray irradiated Chinese hamster (V79-4) cells. In order to investigate the possible radioprotective mechanism of DTT, we measured gamma-ray induced chromosome aberration by micronucleus assay. In the presence of 0.5mM or 1mM DTT, the frequencies of micronuclei were greatly reduced in all dose range examined (1.5-8 GY). Slightly higher reduction in micronucleus formation was observed in 1mM DTT-treated cells than in 0.5mM DTT-treated cells. In addition, incubation with both 0.5 and 1mM of DTT prior to gamma-ray irradiation reduced nucleosomal DNA fragmentation at about same extent, this result suggests that treatment of DTT at concentrations of 0.5 and 1mM reduced radiation-induced apoptosis. In vivo experiments, we also observed that DTT treatment reduced the incidence of apoptotic cells in mouse small intestine crypts. In irradiated control group 4.4${\pm}$0.5 apoptotic cells per crypt were observed. In DTT-administered and irradiated mice, only 2.1${\pm}$0.4 apoptotic cells per crypt was observed. In vitro and in vivo data obtained in this study showed that DTT reduced radiation-induced damages and it seems that the possible radioprotective mechanisms of action of DTT are prevention of chromosome aberration.

  • PDF

Differential Protein Expression Profile Between CD20 Positive and Negative Cells of the NCI-H929 Cell Line

  • Geng, Chuan-Ying;Liu, Nian;Yang, Guang-Zhong;Liu, Ai-Jun;Leng, Yun;Wang, Hui-Juan;Li, Li-Hong;Wu, Yin;Li, Yan-Chen;Chen, Wen-Ming
    • Asian Pacific Journal of Cancer Prevention
    • /
    • v.13 no.11
    • /
    • pp.5409-5413
    • /
    • 2012
  • At present, multiple myeloma (MM) remains an incurable disease and cologenic cells may be responsible for disease relapse. It has been proposed that CD20+/CD138- NCI-H929 cells could be hallmarks of MM clonogenic cells. Here, the immunology phenotype of NCI-H929 cells is described. Only a small population of CD20+/CD138- cells (<1%) was found in the NCI-H929 cell line, but CD20+/CD138- cells were not detected. We found that CD20+/CD138+ cells were able to exhibit cologenic capacity by colony formation assay and continuous passage culture. Proteins were analyzed by 1D-SDS-PAGE and TMT based quantitative differential liquid chromatography tandem mass spectrometry (LC-MS/MS). 1,082 non-redundant proteins were identified, 658 of which were differentially expressed with at least a 1.5-fold difference. 205 proteins in CD20+ cells were expressed at higher levels and 453 proteins were at lower levels compared with CD20- cells. Most proteins had catalytic and binding activity and mainly participated in metabolic processes, cell communication and molecular transport. These results proved that there are different biological features and protein expression profile between CD20+ and CD20- cells in the NCI-H929 cell line.

IGF-1 from Adipose-Derived Mesenchymal Stem Cells Promotes Radioresistance of Breast Cancer Cells

  • Yang, Hui-Ying;Qu, Rong-Mei;Lin, Xiao-Shan;Liu, Tong-Xin;Sun, Quan-Quan;Yang, Chun;Li, Xiao-Hong;Lu, Wei;Hu, Xiao-Fang;Dai, Jing-Xing;Yuan, Lin
    • Asian Pacific Journal of Cancer Prevention
    • /
    • v.15 no.23
    • /
    • pp.10115-10119
    • /
    • 2015
  • Purpose: The aim of this study was to investigate effects of adipose-derived mesenchymal stem cells (AMSCs) on radioresistance of breast cancer cells. Materials and Methods: MTT assays were used to detect any influence of AMSC supernatants on proliferation of breast cancer cells; cell migration assays were used to determine the effect of breast cancer cells on the recruitment of AMSCs; the cell survival fraction post-irradiation was assessed by clonogenic survival assay; ${\gamma}$-H2AX foci number post-irradiation was determined via fluorescence microscopy; and expression of IGF-1R was detected by Western blotting. Results: AMSC supernatants promoted proliferation and radioresistance of breast cancer cells. Breast cancer cells could recruit AMSCs, especially after irradiation. IGF-1 derived from AMSCs might be responsible for the radioresistance of breast cancer cells. Conclusions: Our results suggest that AMSCs in the tumor microenvironment may affect the outcome of radiotherapy for breast cancer in vitro.

Enhancing effect of Canavalia gladiata DC semen on the hematopoietic expansion and function of stem cells (도두(刀豆)의 조혈증진 작용에 관한 연구)

  • Kim, Seung-Hyung;Kim, Kun-Hoae;Chi, Gyeong-Yup;Cho, In-Sik;Kim, Han-Young;Lee, Young-Cheol
    • The Korea Journal of Herbology
    • /
    • v.27 no.4
    • /
    • pp.9-16
    • /
    • 2012
  • Objective : Canavalia gladiata DC semen (CGS) have been used to improve hematopoietic activity. In the current study, we investigated whether CGS regulate hemato-potentiating function using hematopoietic stem cells (HSCs) as a testing system. Methods : HSCs isolated from femur in mice with leukopenia and thrombocytopenia induced induced by CTX. Then, Real-time PCR was performed to measure the mRNA expression and hematopoietic related gene (EPO, IL-3, SCF, c-kit, GM-CSF), the phoaphorylation of GATA-1 and STAT-5a/b were observed by ELISA method, and the number of granulocyte erythrocyte monocyte macrophage colony-forming units (CFU-GEMM) and erythroid burst forming units (BFU-E), semisolid clonogenic assay was performed. Result : When HSCs were treated with CGS, the expression of hematopoietic related genes (EPO, IL-3, SCF, c-kit, and GM-CSF) were significantly increased at the levels of mRNA as well as production in HSCs. Additionally, CGS enhanced phosphorylation of STAT-1 and signal transducer and activator of transcription-5a/b (STAT-5a/b) in HSCs. Furthermore, CGS significantly enhanced the growth rate of granulocyte erythrocyte monocyte macrophage colony-forming units (CFU-GEMM) and erythroid burst forming units (BFU-E) in vitro. Conclusion : These result suggest that CGS has hematopoietic enhancement via hematopoietic cytokine-mediated GATA-1/STAT-5a/b pathway.

Mechanism Underlying Shikonin-induced Apoptosis and Cell Cycle Arrest on SCC25 Human Tongue Squamous Cell Carcinoma Cell Line

  • Oh, Sang-Hun;Park, Sung-Jin;Yu, Su-Bin;Kim, Yong-Ho;Kim, In-Ryoung;Park, Bong-Soo
    • International Journal of Oral Biology
    • /
    • v.40 no.1
    • /
    • pp.51-61
    • /
    • 2015
  • Shikonin, a major ingredient in the traditional Chinese herb Lithospermumerythrorhizon, exhibits multiple biological functions including antimicrobial, anti-inflammatory, and antitumor effects. It has recently been reported that shikonin displays antitumor properties in many cancers. This study was aimed to investigate whether shikonin could inhibit oral squamous carcinoma cell (OSCC) growth via mechanisms of apoptosis and cell cycle arrest. The effects of shikonin on the viability and growth of OSCC cell line, SCC25 cells were assessed by MTT assay and clonogenic assays, respectively. Hoechst staining and DNA electrophoresis indicated that the shikonin-treated SCC25 cells were undergoing apoptosis. Western blotting, immunocytochemistry, confocal microscopy, flow cytometry, MMP activity, and proteasome activity also supported the finding that shikonin induces apoptosis. Shikonin treatment of SCC25 cells resulted in a time- and dose-dependent decrease in cell viability, inhibition of cell growth, and increase in apoptotic cell death. The treated SCC25 cells showed several lines of apoptotic manifestation as follows: nuclear condensation; DNA fragmentation; reduced MMP and proteasome activity; decrease in DNA contents; release of cytochrome c into cytosol; translocation of AIF and DFF40 (CAD) onto the nuclei; a significant shift in Bax/Bcl-2 ratio; and activation of caspase-9, -7, -6, and -3, as well as PARP, lamin A/C, and DFF45 (ICAD). Shikonin treatment also resulted in down-regulation of the G1 cell cycle-related proteins and up-regulation of $p27^{KIP1}$. Taken together, our present findings demonstrate that shikonin strongly inhibits cell proliferation by modulating the expression of the G1 cell cycle-related proteins, and that it induces apoptosis via the proteasome, mitochondria, and caspase cascades in SCC25 cells.

Effects of Mitosene Analogues on Growth Inhibition of Human Cervical Cancer Cell Lines (Mitosene유사체의 자궁암세포주 성장억제 효과)

  • Dong-Soo Cha;Soo-Kie Kim;Chan-Mug Ahn;Sun-Ju Choi;Yoon-Sun Park;Sang-Won Han
    • Biomedical Science Letters
    • /
    • v.3 no.2
    • /
    • pp.71-76
    • /
    • 1997
  • To develop a promising alkylating agents for anti-cervical cancer chemotherapy, five mitosene analogues were synthesized. Despite the potentiality of better cytotoxicity on solid tumor cells as opposed to that on rapidly-doubled leukemic cells, there have been no reports on the inhibition of the cervical cancer cell line by mitosene analogues. The present experiment was designed to investigate whether mitosene analogues can effectively inhibit the cellular proliferation of cervical cancer cells by using an in vitro chemosensitivty system. The mitosene analogues displayed a potent cytotoxic effect on the tested cervical cancer cell lines. Among the analogues, (22) compound gave the best inhibitory effect on SiHa tumor colonies formation. These data indicate that mitosene analogues can effectively inhibit the growth of cervical cancer cells in vitro.

  • PDF

Effects of Korean Traditional Medicine on Murine Hematopiesis (Regulation of Hematopoietic Cytokine & $CD34^{+}$ cell Expression) (수 종의 한약제제가 조혈작용에 미치는 영향)

  • 전재현;김영철;이장훈;우흥정
    • The Journal of Korean Medicine
    • /
    • v.22 no.3
    • /
    • pp.156-168
    • /
    • 2001
  • Objectives : To evaluate the diverse actions of stimulation on the hematopoietic system, 4 formulas (KH I, KH 2, KH 3, KH 4) were studied. Method and Result : RT-PCR was performed to measure the gene expression of hematopoietic cytokines (TPO, GM-CSF, SCF, IL-3). When bone marrow cells were treated with KH 1, 2, 3, 4, the gene expressions of TPO, SCF, IL-3, and GM-CSF were increased. Flow cytometric analysis was performed to measure the expression of CD34+ cell activity. After 72 hrs culture supplemented with KH 1, 2, 3, 4, the percent of CD34+ cell of KH 2, 3, 4 were increased. To measure the expression of colony forming units - granulocyte erythrocytes, macrophages, megakaryocytes (CFU-GEMM) and burst forming unit-erythroid (BFU-E), semisolid clonogenic assay was performed. After 14 days of culture the number of CFU-GEMM and BFU-E of KH I, 2, 3, 4 were significantly increased compared to those of EPO groups (KH 1 P<0.0l, KH 2 P<0.05, KH 3 P<0.001, KH 4 P<0.0l). To determine the intracelluar TPO expression by KH 3, KH 4 in bone marrow cells, intracelluar staining and flow cytometric analysis were performed. After 24 hrs cultures, the TPO expression of the KH 3 and KH 4 treated groups were increased over those of the controlled groups (control : 50%, KH 3 : 87%, KH 4 : 78%). Conclusion : These results suggest that KH I, KH 2, KH 3, KH 4 have hematopoietic effects through increasing the production of hematopoietic cytokines and stimulating the activity of $CD34^{+}$ cells. This study also shows that KH 3 has a more effective hematopoietic effect than KH 1, 2, 4. These results suggest that the formulas (KH I, 2, 3, 4) can be applied to the patients with inappropriate hematopoietic system, and that KH 3 can be the most effective formula among these 4 in treating bone marrow disease in clinics.

  • PDF