• Title/Summary/Keyword: DNA damage checkpoint

Search Result 33, Processing Time 0.027 seconds

Identification of Biomarkers for Radiation Response Using cDNA Microarray

  • Park, Woong-Yang
    • Proceedings of the Korean Society for Bioinformatics Conference
    • /
    • 2001.10a
    • /
    • pp.29-44
    • /
    • 2001
  • DNA damage by physical insult including UV and g-radiation might provoke genetic alterations in cells, which is followed by either acute cell death or tumorigenesis. The responsiveness to g-radiation depends on cellular context of target cells. To understand the mechanisms of checkpoint control, repair and cell death following genotoxic stimu]i, cDNA microarray can provide the gene expression profile. To make a profile of gene expression in irradiated Jurkat T cells, we hybridized the cDNA microarray using cDNA from g-irradiated Jurkat T cells. Jurkat T cells were exposed to 4Gy to 16Gy, and total RNA were extracted at 4 to 24 hrs after irradiation. The hybridization of the microarray to fluorescence-labeled cDNA from treated and untreated cells was analyzed by bioinformatic analysis to address relative changes in expression levels of the genes present in the array. Responses varied widely in different time points, suggesting acute stress response and chronic restoration or cell death. From these results we could select 384 genes related to radiation response in Tcells, and radiation response might be different in various types of cells. Using Radchip, we could separate "the exposed" from control PBMCs. We propose that Radchip might be useful to check the radiation research as well as radiation carcinogenesis.

  • PDF

The emerging role of myeloid-derived suppressor cells in radiotherapy

  • Kang, Changhee;Jeong, Seong-Yun;Song, Si Yeol;Choi, Eun Kyung
    • Radiation Oncology Journal
    • /
    • v.38 no.1
    • /
    • pp.1-10
    • /
    • 2020
  • Radiotherapy (RT) has been used for decades as one of the main treatment modalities for cancer patients. The therapeutic effect of RT has been primarily ascribed to DNA damage leading to tumor cell death. Besides direct tumoricidal effect, RT affects antitumor responses through immune-mediated mechanism, which provides a rationale for combining RT and immunotherapy for cancer treatment. Thus far, for the combined treatment with RT, numerous studies have focused on the immune checkpoint inhibitors and have shown promising results. However, treatment resistance is still common, and one of the main resistance mechanisms is thought to be due to the immunosuppressive tumor microenvironment where myeloid-derived suppressor cells (MDSCs) play a crucial role. MDSCs are immature myeloid cells with a strong immunosuppressive activity. MDSC frequency is correlated with tumor progression, recurrence, negative clinical outcome, and reduced efficacy of immunotherapy. Therefore, increasing efforts to target MDSCs have been made to overcome the resistance in cancer treatments. In this review, we focus on the role of MDSCs in RT and highlight growing evidence for targeting MDSCs in combination with RT to improve cancer treatment.

Kaempferol Activates G2-Checkpoint of the Cell Cycle Resulting in G2-Arrest and Mitochondria-Dependent Apoptosis in Human Acute Leukemia Jurkat T Cells

  • Kim, Ki Yun;Jang, Won Young;Lee, Ji Young;Jun, Do Youn;Ko, Jee Youn;Yun, Young Ho;Kim, Young Ho
    • Journal of Microbiology and Biotechnology
    • /
    • v.26 no.2
    • /
    • pp.287-294
    • /
    • 2016
  • The effect of kaempferol (3,5,7,4-tetrahydroxyflavone), a flavonoid compound that was identified in barnyard millet (Echinochloa crus-galli var. frumentacea) grains, on G2-checkpoint and apoptotic pathways was investigated in human acute leukemia Jurkat T cell clones stably transfected with an empty vector (J/Neo) or a Bcl-xL expression vector (J/Bcl-xL). Exposure of J/Neo cells to kaempeferol caused cytotoxicity and activation of the ATM/ATR-Chk1/Chk2 pathway, activating the phosphorylation of p53 (Ser-15), inhibitory phosphorylation of Cdc25C (Ser-216), and inactivation of cyclin-dependent kinase 1 (Cdk1), with resultant G2-arrest of the cell cycle. Under these conditions, apoptotic events, including upregulation of Bak and PUMA levels, Bak activation, mitochondrial membrane potential (Δψm) loss, activation of caspase-9, -8, and -3, anti-poly (ADP-ribose) polymerase (PARP) cleavage, and accumulation of apoptotic sub-G1 cells, were induced without accompanying necrosis. However, these apoptotic events, except for upregulation of Bak and PUMA levels, were completely abrogated in J/Bcl-xL cells overexpressing Bcl-xL, suggesting that the G2-arrest and the Bcl-xL-sensitive mitochondrial apoptotic events were induced, in parallel, as downstream events of the DNA-damage-mediated G2-checkpoint activation. Together these results demonstrate that kaempferol-mediated antitumor activity toward Jurkat T cells was attributable to G2-checkpoint activation, which caused not only G2-arrest of the cell cycle but also activating phosphorylation of p53 (Ser-15) and subsequent induction of mitochondria-dependent apoptotic events, including Bak and PUMA upregulation, Bak activation, Δψm loss, and caspase cascade activation.

Antitumor effects of valdecoxib on hypopharyngeal squamous carcinoma cells

  • Trang, Nguyen Thi Kieu;Yoo, Hoon
    • The Korean Journal of Physiology and Pharmacology
    • /
    • v.26 no.6
    • /
    • pp.439-446
    • /
    • 2022
  • The antitumoral effects of valdecoxib (Val), an United States Food and Drug Administration-approved anti-inflammatory drug that was withdrawn due to the side effects of increased risk of cardiovascular adverse events, were investigated in hypopharyngeal squamous cell carcinoma cells by performing a cell viability assay, transwell assay, immunofluorescence imaging, and Western blotting. Val markedly inhibited cell viability with an IC50 of 67.3 µM after 48 h of treatment, and also downregulated cell cycle proteins such as Cdks and their regulatory cyclin units. Cell migration and invasion were severely suppressed by inhibiting integrin α4/FAK expression. In addition, Val activated the cell cycle checkpoint CHK2 in response to excessive DNA damage, which led to the activation of caspase-3/9 and induced caspase-dependent apoptosis. Furthermore, the signaling cascades of the PI3K/AKT/mTOR and mitogen-activated protein kinase pathways were significantly inhibited by Val treatment. Taken together, our results indicate that Val can be used for the treatment of hypopharyngeal squamous cell carcinoma.

Knockdown of UHRF1 by Lentivirus-mediated shRNA Inhibits Ovarian Cancer Cell Growth

  • Yan, Feng;Shao, Li-Jia;Hu, Xiao-Ya
    • Asian Pacific Journal of Cancer Prevention
    • /
    • v.16 no.4
    • /
    • pp.1343-1348
    • /
    • 2015
  • Human UHRF1 (ubiquitin-like PHD and RING finger domain-containing 1) has been reported to be over-expressed in many cancers, but its role in ovarian cancer remains elusive. Here, we determined whether knockdown of UHRF1 by lentivirus-mediated shRNA could inhibit ovarian cancer cell growth. Lentivirus-mediated short hairpin RNAs (lv-shRNAs-UHRF1) were designed to trigger the gene silencing RNA interference (RNAi) pathway. The efficiency of lentivirus-mediated shRNA infection into HO-8910 and HO-8910 PM cells was determined using fluorescence microscopy to observe lentivirus-mediated GFP expression and was confirmed to be over 80 percent. UHRF1 expression in infected HO-8910 and HO-8910 PM was evaluated by real-time PCR and Western blot analysis. The Cell Counting Kit-8 (CCK-8) assay was used to measure cell viability; flow cytometry and Hoechst 33342 assay was applied to measure cell cycle arrest and apoptosis. Cell invasion was assessed using transwell chambers. Our results demonstrated that the loss of UHRF1 promoted HO-8910 and HO-8910 PM cell apoptosis, while inhibiting cell proliferation. In addition, UHRF1 knockdown significantly inhibited the invasion of human ovarian cancer cells. In the present study, we also showed that depleting HO-8910 cells of UHRF1 caused activation of the DNA damage response pathway, with the cell cycle arrested in G2/M-phase. The DNA damage response in cells depleted of UHRF1 was illustrated by phosphorylation of CHK (checkpoint kinase) 2 on Thr68, phosphorylation of CDC25 (cell division control 25) on Ser 216 and phosphorylation of CDK1 (cyclin-dependent kinase 1) on Tyr 15.

CELL MORPHOLOGY CHANGE BY THE ULTRAVIOLET RAY IRRADIATION

  • Park, Myoung-Joo;Matuo, Yoichirou;Akiyama, Yoko;Izumi, Yoshinobu;Nishijima, Shigehiro
    • Journal of Radiation Protection and Research
    • /
    • v.34 no.1
    • /
    • pp.15-24
    • /
    • 2009
  • The effect of low doses of ultraviolet (UV) irradiation on morphology changes of cell has been studied based on the observation of the cell length. It was shown that UV-irradiated cell has different behavior in comparison with non-irradiated cell. From the histogram of cell-length distribution, it was confirmed that cell cycle of non irradiated cell was 28 hours, and that cell cycle of irradiated cell with dose of $20\;Jm^{-2}$ was delayed (39 hours), while irradiated cell with $40\;Jm^{-2}$ and $60\;Jm^{-2}$ did not divide and kept growing continuously. It was supposed that in case of $20\;Jm^{-2}$ of irradiation dose, the cell cycle was delayed because the checkpoint worked in order to repair DNA damage induced by generation of pyrimidine dimer, reactive oxygen species and so on. It was also supposed that in case of $40\;Jm^{-2}$ and $60\;Jm^{-2}$ of irradiation dose, overgrowth was induced because the checkpoint was not worked well. The morphology of overgrown cell was similar to that of normally senescent cell. Therefore, it was considered that cell senescence was accelerated by UV irradiation with irradiation doses of $40\;Jm^{-2}$ and $60\;Jm^{-2}$.

Mutation of the Chk1 Gene in Gastric Cancers with Microsatellite Instability (현미부수체 불안정성을 동반한 위암에서 Chk1 유전자의 돌연변이)

  • Lee, Jong-Heun;Cho, Young-Gu;Song, Jae-Whie;Park, Cho-Hyun;Kim, Su-Yeong;Nam, Suk-Woo;Lee, Sug-Hyung;Yoo, Nam-Jin;Lee, Jung-Young;Park, Won-Sang
    • Journal of Gastric Cancer
    • /
    • v.5 no.4 s.20
    • /
    • pp.260-265
    • /
    • 2005
  • Purpose: The protein kinase Chk1 is required for cell cycle arrest in response to DNA damage and is shown to play an important role in the G2/M checkpoint. The aim of this study was to investigate the relationship between microsatellite instability and frameshift mutation of the Chk1 gene in gastric cancers. Materials and Methods: The microsatellite instability was analyzed in 95 primary gastric carcinomas by using microdissection and 6 microsatellite markers. We also peformed single strand conformational polymorphism and sequencing to detect frameshift mutation of the Chk1 gene. Results: We found positive microsatellite instability in 19 (20%) of the 95 gastric cancers, 13 high- and 6 low-frequency microsatellite instability cases. The frameshift mutation of Chk1, which resulted in a truncated Chk1 protein, was detected in two high-frequency microsatellite instability cases. Conclusion: These data suggest that the microsatellite instability may contribute to the development of gastric carcinomas through inactivation of Chk1.

  • PDF

The Effects of Injinchunggantang-derivative on Cell Viability, Cell Cycle Progression and Apoptosis of Hepatocytes (인진청간탕가미방(茵陳淸肝湯加味方)이 간세포활성(肝細胞活性), 세포주기(細胞週期) 및 APOPTOSIS에 미치는 영향(影響))

  • Hong, Sang-Hoon;Lee, Jang-Hoon;Woo, Hong-Jung
    • The Journal of Korean Medicine
    • /
    • v.19 no.2
    • /
    • pp.337-372
    • /
    • 1998
  • To evaluate the effects of Injinchunggantang-derivative on cell viability, cell cycle progression, and apoptosis, MTT assay, cell cycle analysis, Cpp32 protease assay, DNA fragnemtation assay, quantitative RT-PCR, and Western blotting were performed. The results were as followes. In MTT assay, etoposide+Injinchunggantang-derivative-treated cells as well as Injinchunggantang-derivative-treated cells showed higher viability than etoposide-treated cells with no time-concentration-dependence, which implied that Injinchunggantang-derivative has hepato-protective effect Cell cycle analysis showed that Injinchunggantang-derivative has no significant effect on the cell cycle. Cpp32 protease assav and DNA fragmentation assay Injinchunggantang-derivative carry inhibitory effects on apoptosis induction. It was suggested that Injinchunggantang-delivative might regulate the cell cycle, in particular $G_1$ checkpoint by blocking p53 and Watl pathway. Injinchunggantang-derivative inhibited the mRNA expressions of Cpp32, Fas, and Bcl-2, which could result in inhibition of apoptosis. These results imply that Injinchunggantang-derivative increases hepatocyte viability, and protects hepatocyte from damage by regulating the expression of genes associated with cell cycle and apoptosis, which explains the mechanism of the clinical effect of Injinchunggantang-derivative on liver diseases.

  • PDF

Effects of an Anti-cancer Drug, Tubastatin A, on the Growth and Development of Immature Oocytes in Mice (항암제 tubastatin A에 의한 생쥐 미성숙 난모세포의 성장과 발달에 미치는 효과)

  • Choi, Yun-Jung;Min, Gyesik
    • Journal of Life Science
    • /
    • v.29 no.1
    • /
    • pp.105-111
    • /
    • 2019
  • In recent years, progress has been made in the search for the development of new anti-cancer agents by employing specific inhibitors of histone deacetylase (HDAC)-6 to block signal transduction pathways in cancer cells. This study examined the effects of tubastatin A (TubA), an HDAC-6 inhibitor, on the growth and development of immature oocytes in murine ovaries using RNA sequencing analysis. The results from a gene set enrichment analysis (GSEA) indicated that the expression of most of the gene sets involved in the cell cycle and control and progression of meiosis decreased in the TubA-treated group as compared with that in germinal vesicle (GV) stage oocytes. In addition, an ingenuity pathway analysis (IPA) suggested that TubA not only caused increased expression of p53 and pRB and decreased expression of CDK4/6 and cyclin D but also caused elevated expression of genes involved in the control of the DNA check point in G2/M stage oocytes. These results suggest that TubA may induce cell cycle arrest and apoptosis through the induction of changes in the expression of genes involved in signal transduction pathways associated with DNA damage and the cell cycle of immature oocytes in the ovary.