• Title/Summary/Keyword: Cell cycle inhibitor

Search Result 320, Processing Time 0.03 seconds

Effect of Cell Cycle of Donor Nucleus on In Vitro Development in Nuclear Transplant Rabbit Embryos (토끼 핵이식 수정란의 체외 발달에 미치는 공핵란 세포주기의 효과)

  • 박충생;전병균;윤희준;이효종;최상용
    • Korean Journal of Animal Reproduction
    • /
    • v.20 no.2
    • /
    • pp.143-153
    • /
    • 1996
  • To improve the efficiency of nuclear transplantation in the rabbit, this study were evaluated the influence of celly cycle of donor nuclei on the in vitro developmental potential in the nuclear transplant embryos. The embryos of 16-cell stage were collected from the mated does at 48h post-hCG injection and they were synchronized to G1 phase of 32-cell stage. Synchronization of the cell cylce of blastomeres were induced, first, using an microtubules polymerization inhibitor, 0.5$\mu\textrm{g}$/ml colcemid for 10h to arrest blastomeres in metaphase, and secondly, using a DNA synthesis inhibitor, 0.1$\mu\textrm{g}$/ml aphidicolin for 1.5 to 2h to cleave to 32-cell stage and arrest them in G1 phase. The separated G1 phase blastomeres of 32-cell stage were injectied into enucleated recipient cytoplasms by micromanipulation. After culture until 20h post-hCG injection, the nuclear transplant oocytes were electrofused and activated by electrical stimulation. The nuclear transplant embryos were co-cultured for 120h. In vitro cultured embryos were monitored every 24h to assess for development rate. After in vitro cultue for 120h, the nuclear transplant embryos developed to blastocyst stage were stained with Hoechst 33342 dye for counting the number of blastomeres under a fluorescence microscopy. The cleavage rate of blastomeres from 16-cell stage stage rabbit embryos treated with colcemid for 10h or aphidicolin for 6h following colcemid for 10h were not significantly different. The electrofusion rate was similar by high in S and G1 phase donor nuclei as 80.6 and 79.1%, respectively. However, the nuclear transplant embryos using G1 phase donor nuclei were developed to blastocyst at high rate(60.3%) than those using S phase donor nuclei(26.0%). Moreover, the mean blastocyst stage were increased significantly(P<0.05) with the G1 phase donor nuclei(176.6 cells and 1.50 cycles), as compared with the S phase donor nuclei(136.6 cells and 1.42 cycles). These results show that the blastomeres of G1 phase were more successful as donor nuclei in the nuclear transplant procedure, compared with S phase.

  • PDF

G1 Arrest of the Cell Cycle by Onchungeum in Human Hepatocarcinoma Cells (온청음(溫淸飮)이 인체 간암세포의 세포주기 G1 Arrest에 미치는 영향)

  • Goo, In-Moo;Shin, Heung-Mook
    • Journal of Physiology & Pathology in Korean Medicine
    • /
    • v.22 no.4
    • /
    • pp.821-828
    • /
    • 2008
  • Onchungeum, a herbal formula, which has been used for treatment of anemia due to bleeding, discharging blood and skin disease. In the present study, it was examined the effects of extract of Onchungeum (OCE) on the growth of human hepatocarcinoma cell lines Hep3B (p53 null type) and HepG2 (p53 wild type) in order to investigate the anti-proliferative mechanism by OCE. Treatment of Hep3B and HepG2 cells to OCE resulted in the growth inhibition in a dose-dependent manner, however Hep3B cell line exhibited a relatively strong anti-proliferative activity to OEC. Flow cytometric analysis revealed that OCE treatment in Hep3B cells caused G1 phase arrest of the cell cycle, which was associated with various morphological changes in a dose-dependent fashion. RT-PCR and immunoblotting data revealed that treatment of OCE caused the down-regulation of cyclin D1 expression, however the levels of cyclin E expression were not changed by OCE. The G1 arrest of the cell cycle was also associated with the induction of Cdk inhibitor p27 by OCE. Because the p53 gene is null in Hep3B cells, it is most likely that the induction of p21 is mediated through a p53-independent pathway. Moreover, p27 detected in anti-Cdk4 and anti-Cdk2 immunoprecipitates from the OCE-treated cells, suggesting that OCE-induced p27 protein blocks Cdk kinase activities by directing binding to the cyclin/Cdk complexes. Furthermore, OCE treatment potently suppresses the phosphorylation of retinoblastoma proteins and the levels of the transcription factor E2F-1 expression. Taken together, these results indicated that the growth inhibitory effect of OCE in Hep3B hepatoma cells was associated with the induction of G1 arrest of the cell cycle through regulation of several major growth regulatory gene products.

G0/G1 Cell Cycle Arrest and Activation of Caspases in Honokiol-mediated Growth Inhibition of Human Gastric Cancer Cells

  • Kang, You-Jin;Chung, Hwa-Jin;Min, Hye-Young;Song, Ja-Young;Park, Hyen-Joo;Youn, Ui-Joung;Bae, Ki-Hwan;Kim, Yeong-Shik;Lee, Sang-Kook
    • Natural Product Sciences
    • /
    • v.18 no.1
    • /
    • pp.16-21
    • /
    • 2012
  • Honokiol, a naturally occurring neolignan mainly found in Magnolia species, has been shown to have the anti-angiogenic, anti-invasive and cancer chemopreventive activities, but the molecular mechanism of actions has not been fully elucidated yet. In the present study, we investigated the effect of honokiol on the growth inhibitory activity in cultured SNU-638 human gastric cancer cells. We found that honokiol exerted potent antiproliferative activity against SNU-638 cells. Honokiol also arrested the cell cycle progression at the G0/G1 phase and induced the apoptotic cell death in a concentration-dependent manner. The cell cycle arrest was well correlated with the downregulation of Rb, cyclin D1, cyclin A, cyclin E, and CDK4 expression, and the induction of cyclin-dependent kinase inhibitor p27. The increase of sub-G1 peak by honokiol was closely related to the induction of apoptosis, which was evidenced by the induction of DNA fragmentation, the cleavage of poly(ADPribose) polymerase, and the sequential activation of caspase cascade. These findings suggest the cell cycle arrest and induction of apoptosis might be one possible mechanism of actions for the anti-proliferative activity of honokiol in human gastric cancer cell.

Picropodophyllotoxin Induces G1 Cell Cycle Arrest and Apoptosis in Human Colorectal Cancer Cells via ROS Generation and Activation of p38 MAPK Signaling Pathway

  • Lee, Seung-On;Kwak, Ah-Won;Lee, Mee-Hyun;Seo, Ji-Hye;Cho, Seung-Sik;Yoon, Goo;Chae, Jung-Il;Joo, Sang Hoon;Shim, Jung-Hyun
    • Journal of Microbiology and Biotechnology
    • /
    • v.31 no.12
    • /
    • pp.1615-1623
    • /
    • 2021
  • Picropodophyllotoxin (PPT), an epimer of podophyllotoxin, is derived from the roots of Podophyllum hexandrum and exerts various biological effects, including anti-proliferation activity. However, the effect of PPT on colorectal cancer cells and the associated cellular mechanisms have not been studied. In the present study, we explored the anticancer activity of PPT and its underlying mechanisms in HCT116 cells. The 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay was used to monitor cell viability. Flow cytometry was used to evaluate cell cycle distribution, the induction of apoptosis, the level of reactive oxygen species (ROS), assess the mitochondrial membrane potential (Δψm), and multi-caspase activity. Western blot assays were performed to detect the expression of cell cycle regulatory proteins, apoptosis-related proteins, and p38 MAPK (mitogen-activated protein kinase). We found that PPT induced apoptosis, cell cycle arrest at the G1 phase, and ROS in the HCT116 cell line. In addition, PPT enhanced the phosphorylation of p38 MAPK, which regulates apoptosis and PPT-induced apoptosis. The phosphorylation of p38 MAPK was inhibited by an antioxidant agent (N-acetyl-L-cysteine, NAC) and a p38 inhibitor (SB203580). PPT induced depolarization of the mitochondrial inner membrane and caspase-dependent apoptosis, which was attenuated by exposure to Z-VAD-FMK. Overall, these data indicate that PPT induced G1 arrest and apoptosis via ROS generation and activation of the p38 MAPK signaling pathway.

Helicobacter pylori-Induced Progranulin Promotes the Progression of the Gastric Epithelial Cell Cycle by Regulating CDK4

  • Ren, Zongjiao;Li, Jiayi;Du, Xianhong;Shi, Wenjing;Guan, Fulai;Wang, Xiaochen;Wang, Linjing;Wang, Hongyan
    • Journal of Microbiology and Biotechnology
    • /
    • v.32 no.7
    • /
    • pp.844-854
    • /
    • 2022
  • Helicobacter pylori, a group 1 carcinogen, colonizes the stomach and affects the development of stomach diseases. Progranulin (PGRN) is an autocrine growth factor that regulates multiple cellular processes and plays a tumorigenic role in many tissues. Nevertheless, the mechanism of action of PGRN in gastric cancer caused by H. pylori infection remains unclear. Here, we investigated the role of PGRN in cell cycle progression and the cell proliferation induced by H. pylori infection. We found that the increased PGRN was positively associated with CDK4 expression in gastric cancer tissue. PGRN was upregulated by H. pylori infection, thereby promoting cell proliferation, and that enhanced level of proliferation was reduced by PGRN inhibitor. CDK4, a target gene of PGRN, is a cyclin-dependent kinase that binds to cyclin D to promote cell cycle progression, which was upregulated by H. pylori infection. We also showed that knockdown of CDK4 reduced the higher cell cycle progression caused by upregulated PGRN. Moreover, when the PI3K/Akt signaling pathway (which is promoted by PGRN) was blocked, the upregulation of CDK4 mediated by PGRN was reduced. These results reveal the potential mechanism by which PGRN plays a major role through CDK4 in the pathological mechanism of H. pylori infection.

The Effect of Carex dispalata Extract on the Activation of Anagen Pathway (삿갓사초 추출물의 모발 성장 기전 활성화 효과)

  • Kang, Jung-Il;Seo, Min Jeong;Choi, Youn Kyung;Shin, Su Young;Kim, Sun Yu;Yoo, Eun-Sook;Kim, Sang-Cheol;Kang, Hee-Kyoung
    • Korean Journal of Pharmacognosy
    • /
    • v.52 no.4
    • /
    • pp.234-241
    • /
    • 2021
  • Dermal papilla cells (DPCs) are present throughout the hair cycle and play an essential role in hair cycle and hair growth. In this study, we investigated the effect of Carex dispalata on the activation of anagen pathway in DPCs. C. dispalata extract increased the proliferation of DPCs and induced changes in the levels of cell cycle-related proteins. To elucidate the mechanism by which C. dispalata extract stimulates the anagen pathway related to the proliferation of DPCs, we evaluated the effect of C. dispalata extract on the activation of Akt signaling. The increase in the level of phospho-Akt by C. dispalata extract was inhibited by PI3K inhibitor (wortmannin). Wortmannin reduced the effects of C. dispalata extract on the levels of cell cycle-related proteins and proliferation of DPCs. C. dispalata extract increased the levels of Wnt/β-catenin proteins. Wnt/β-catenin inhibitor (XAV939) inhibited changes in cell cycle, cell cycle-related proteins, Wnt/β-catenin proteins, and proliferation induced by C. dispalata extract. C. dispalata extract increased the level of autophagy protein (LC3I/II), and this change was inhibited by XAV939. These results suggest that C. dispalata extract can activate PI3K/Akt, Wnt/β-catenin, and autophagy pathways in DPCs to induce cell proliferation, and thereby promote hair growth phase.

Induction of cytoprotective autophagy by morusin via AMP-activated protein kinase activation in human non-small cell lung cancer cells

  • Park, Hyun-Ji;Park, Shin-Hyung
    • Nutrition Research and Practice
    • /
    • v.14 no.5
    • /
    • pp.478-489
    • /
    • 2020
  • BACKGROUND/OBJECTIVES: Morusin, a marker component of Morus alba L., possesses anti-cancer activity. The objective of this study was to determine autophagy-inducing effect of morusin in non-small cell lung cancer (NSCLC) cells and investigate the underlying mechanism. SUBJECTS/METHODS: Autophagy induction and the expression of autophagy-related proteins were analyzed by LC3 immunofluorescence and western blot, respectively. The role of autophagy and AMP-activated protein kinase (AMPK) was determined by treating NSCLC cells with bafilomycin A1, an autophagy inhibitor, and compound C, an AMPK inhibitor. Cytotoxicity and apoptosis induction were determined by MTT assay, trypan blue exclusion assay, annexin V-propidium iodide (PI) double staining assay, and cell cycle analysis. RESULTS: Morusin increased the formation of LC3 puncta in the cytoplasm and upregulated the expression of autophagy-related 5 (Atg5), Atg12, beclin-1, and LC3II in NSCLC cells, demonstrating that morusin could induce autophagy. Treatment with bafilomycin A1 markedly reduced cell viability but increased proportions of sub-G1 phase cells and annexin V-positive cells in H460 cells. These results indicate that morusin can trigger autophagy in NSCLC cells as a defense mechanism against morusin-induced apoptosis. Furthermore, we found that AMPK and its downstream acetyl-CoA carboxylase (ACC) were phosphorylated, while mammalian target of rapamycin (mTOR) and its downstream p70S6 kinase (p70S6K) were dephosphorylated by morusin. Morusin-induced apoptosis was significantly increased by treatment with compound C in H460 cells. These results suggest that morusin-induced AMPK activation could protect NSCLC cells from apoptosis probably by inducing autophagy. CONCLUSIONS: Our findings suggest that combination treatment with morusin and autophagy inhibitor or AMPK inhibitor might enhance the clinical efficacy of morusin for NSCLC.

Identification of Protein Phosphatase 4 Inhibitory Protein That Plays an Indispensable Role in DNA Damage Response

  • Park, Jaehong;Lee, Jihye;Lee, Dong-Hyun
    • Molecules and Cells
    • /
    • v.42 no.7
    • /
    • pp.546-556
    • /
    • 2019
  • Protein phosphatase 4 (PP4) is a crucial protein complex that plays an important role in DNA damage response (DDR), including DNA repair, cell cycle arrest and apoptosis. Despite the significance of PP4, the mechanism by which PP4 is regulated remains to be elucidated. Here, we identified a novel PP4 inhibitor, protein phosphatase 4 inhibitory protein (PP4IP) and elucidated its cellular functions. PP4IP-knockout cells were generated using the CRISPR/Cas9 system, and the phosphorylation status of PP4 substrates (H2AX, KAP1, and RPA2) was analyzed. Then we investigated that how PP4IP affects the cellular functions of PP4 by immunoprecipitation, immunofluorescence, and DNA double-strand break (DSB) repair assays. PP4IP interacts with PP4 complex, which is affected by DNA damage and cell cycle progression and decreases the dephosphorylational activity of PP4. Both overexpression and depletion of PP4IP impairs DSB repairs and sensitizes cells to genotoxic stress, suggesting timely inhibition of PP4 to be indispensable for cells in responding to DNA damage. Our results identify a novel inhibitor of PP4 that inhibits PP4-mediated cellular functions and establish the physiological importance of this regulation. In addition, PP4IP might be developed as potential therapeutic reagents for targeting tumors particularly with high level of PP4C expression.

Study on the Pro-apoptotic Effect of Artemisiae Capillaris Herba Extracted with Ethanol on Biliary Tract Cancer Cell Line, SNU-1196 (인진(茵蔯)의 에탄올 추출물이 담도암 세포주 SNU-1196의 apoptosis에 미치는 효과에 관한 연구)

  • Lee, Kyung-Wook;Woo, Hong-Jung
    • The Journal of Internal Korean Medicine
    • /
    • v.33 no.4
    • /
    • pp.587-598
    • /
    • 2012
  • Objectives : This study was performed to elucidate the pro-apoptotic effect of Artemisiae capillaris herba extracted with ethanol on biliary tract cancer cells. Materials and Methods : The biliary tract cancer cell line SNU-1196 was used in this study. Cells were treated with different concentrations of Artemisiae capillaris herba for 24, 48 and 72 hours. After the treatment, cell viability, apoptosis, caspase activities and the mRNA expressions of the Bcl-2, Bax, P53, and P21 were measured by using MTT assay, cell cycle analysis, apoptosis assay, and RT-PCR. The cell cycle analysis was done by flow cytometry and apoptosis assay by cell death detection ELISA kit. Results : Artemisiae capillaris herba inhibited proliferation of SNU-1196 in long-time culture group with dose-dependent manner. All cells treated with Artemisiae capillaris herba showed increased apoptosis with dose- and time-dependent manner. Exposure of SNU-1196 to Artemisiae capillaris herba induced caspase-3 activation. However, apeoptosis was blocked when SNU-1196 was treated together with the pan-caspase inhibitor Z-VAD-FMK and the caspase-3 inhibitor Z-DEVE-FMK. After the treatment of Artemisiae capillaris herba, the mRNA expressions of caspase -3, -8, -9, p53, and p21 was increased in all cells. Artemisiae capillaris herba resulted in a significant decrease in Bcl-2 and an increase in Bax mRNA levels. Conclusions : These results suggest that Artemisiae capillaris herba would be beneficial in the treatment of biliary tract cancer.

Combination Therapy with a PI3K/mTOR Dual Inhibitor and Chloroquine Enhances Synergistic Apoptotic Cell Death in Epstein-Barr Virus-Infected Gastric Cancer Cells

  • Kim, Mi-Young;Kruger, Annie J.;Jeong, Ju-Yeon;Kim, Jaehee;Shin, Phil kyung;Kim, Sun Young;Cho, Joo Young;Hahm, Ki Baik;Hong, Sung Pyo
    • Molecules and Cells
    • /
    • v.42 no.6
    • /
    • pp.448-459
    • /
    • 2019
  • The phosphoinositide 3-kinase/protein kinase B/mammalian target of rapamycin (PI3K/AKT/mTOR) signaling pathway is a promising target for gastric cancer (GC) treatment; however the efficacy of PI3K/mTOR dual inhibitors in GC has not yet been maximized. Additionally, the effect of autophagy regulation by PI3K/mTOR dual inhibitors has not been clearly elucidated in GC treatment. We aimed to show that our newly developed PI3K/mTOR dual inhibitor, CMG002, when combined with an autophagy inhibitor, chloroquine (CQ), potently induces effective cancer cell death in Epstein-Barr virus (EBV)-associated gastric cancer (EBVaGC) cells, where both the PI3K/AKT/mTOR and autophagy pathways play important roles in disease pathogenesis. EBV- and mock-infected AGS and NUGC3 GC cell lines were treated with CMG002 +/- CQ. PI3K/AKT/mTOR signaling pathway mediators, cellular apoptosis and autophagy markers were confirmed by Western blot assay. Cell viability was assessed using the Cell Counting Kit-8 (CCK-8) assay. CMG002 effectively blocked the PI3K/AKT/mTOR pathway by markedly decreasing phosphorylation of AKT and its downstream mediator S6. CMG002 induced G0/G1 cell cycle arrest and enhanced apoptotic cell death in AGS and NUGC3 cells, particularly EBV-infected cells compared with mock-infected cells, as confirmed by flow cytometric analyses and TUNEL (terminal deoxynucleotidyl transferase dUTP nick end labeling) assays. The combination of CMG002 plus CQ synergistically increased apoptotic cell death in EBV-infected GC cell lines when compared with CMG002 alone (P < 0.05). Our results suggest that the new PI3K/mTOR dual inhibitor, CMG002, when used in combination with the autophagy inhibitor, CQ, provides enhanced therapeutic efficacy against EBVaGC.