• Title/Summary/Keyword: Poly (ADP-ribose) polymerase-1 inhibitor

Search Result 82, Processing Time 0.026 seconds

Reversine induces cell cycle arrest and apoptosis via upregulation of the Fas and DR5 signaling pathways in human colorectal cancer cells

  • YOUNG-LAN PARK;SANG-YOON HA;SUN-YOUNG PARK;JUNG-HO CHOI;MIN-WOO JUNG;DAE-SEONG MYUNG;HYUN-SOO KIM;YOUNG-EUN JOO
    • International Journal of Oncology
    • /
    • v.54 no.5
    • /
    • pp.1875-1883
    • /
    • 2019
  • Reversine, a 2,6-diamino-substituted purine analogue, has been reported to be effective in tumor suppression via induction of cell growth arrest and apoptosis of cancer cells. However, it remains unclear whether reversine exerts anticancer effects on human colorectal cancer cells. In the present study, in vitro experiments were conducted to investigate the anticancer properties of reversine in human colorectal cancer cells. The effect of reversine on human colorectal cancer cell lines, SW480 and HCT-116, was examined using a WST-1 cell viability assay, fluorescence microscopy, flow cytometry, DNA fragmentation, small interfering RNA (siRNA) and western blotting. Reversine treatment demonstrated cytotoxic activity in human colorectal cancer cells. It also induced apoptosis by activating poly(ADP-ribose) polymerase, caspase-3, -7 and -8, and increasing the levels of the pro-apoptotic protein second mitochondria-derived activator of caspase/direct inhibitor of apoptosis-binding protein with low pI. The pan-caspase inhibitor Z-VAD-FMK attenuated these reversine-induced apoptotic effects on human colorectal cancer cells. Additionally, reversine treatment induced cell cycle arrest in the subG1 and G2/M phases via increase in levels of p21, p27 and p57, and decrease in cyclin D1 levels. The expression of Fas and death receptor 5 (DR5) signaling proteins in SW480 and HCT116 cells was upregulated by reversine treatment. Reversine-induced apoptosis and cell cycle arrest were suppressed by inhibition of Fas and DR5 expression via siRNA. In conclusion, Reversine treatment suppressed tumor progression by the inhibition of cell proliferation, induction of cell cycle arrest and induction of apoptosis via upregulation of the Fas and DR5 signaling pathways in human colorectal cancer cells. The present study indicated that reversine may be used as a novel anticancer agent in human colorectal cancer.

The Histone Deacetylase Inhibitor Trichostatin A Sensitizes Human Renal Carcinoma Cells to TRAIL-Induced Apoptosis through Down-Regulation of c-FLIPL

  • Han, Min Ho;Park, Cheol;Kwon, Taek Kyu;Kim, Gi-Young;Kim, Wun-Jae;Hong, Sang Hoon;Yoo, Young Hyun;Choi, Yung Hyun
    • Biomolecules & Therapeutics
    • /
    • v.23 no.1
    • /
    • pp.31-38
    • /
    • 2015
  • Histone acetylation plays a critical role in the regulation of transcription by altering the structure of chromatin, and it may influence the resistance of some tumor cells to tumor necrosis factor (TNF)-related apoptosis-inducing ligand (TRAIL) by regulating the gene expression of components of the TRAIL signaling pathway. In this study, we investigated the effects and molecular mechanisms of trichostatin A (TSA), a histone deacetylase inhibitor, in sensitizing TRAIL-induced apoptosis in Caki human renal carcinoma cells. Our results indicate that nontoxic concentrations of TSA substantially enhance TRAIL-induced apoptosis compared with treatment with either agent alone. Cotreatment with TSA and TRAIL effectively induced cleavage of Bid and loss of mitochondrial membrane potential (MMP), which was associated with the activation of caspases (-3, -8, and -9) and degradation of poly (ADP-ribose) polymerase (PARP), contributing toward the sensitization to TRAIL. Combined treatment with TSA and TRAIL significantly reduced the levels of the cellular Fas-associated death domain (FADD)-like interleukin-$1{\beta}$-converting enzyme (FLICE) inhibitory protein (c-FLIP), whereas those of death receptor (DR) 4, DR5, and FADD remained unchanged. The synergistic effect of TAS and TRAIL was perfectly attenuated in c-$FLIP_L$-overexpressing Caki cells. Taken together, the present study demonstrates that down-regulation of c-FLIP contributes to TSA-facilitated TRAIL-induced apoptosis, amplifying the death receptor, as well as mitochondria-mediated apoptotic signaling pathways.

Mechanism of Apoptotic Cell Death by 2,4,3',5'-Tetramethoxystilbene in Human Promyelocytic Leukemic HL-60 Cells

  • Lee, Sang-Kwang;Chae, Ah-Reum;Chun, Young-Jin
    • Biomolecules & Therapeutics
    • /
    • v.15 no.3
    • /
    • pp.145-149
    • /
    • 2007
  • We have previously shown that 2,4,3',5'-tetramethoxystilbene (TMS), a synthetic trans-stilbene analogue acting as a potent inhibitor of human cytochrome P450 1B1, induces apoptotic cell death in human cancer cells. In the present studies, we report the mechanisms of apoptotic cell death by TMS in human promyelocytic leukemic HL-60 cells. We found that treatment of HL-60 cells with TMS suppressed the cell growth in a concentration-dependent manner with $IC_{50}$ value of about 0.8 ${\mu}M$. Immunoblot experiments revealed that DMHS-induced apoptosis was associated with cleavage of poly (ADP-ribose) polymerase. The release of cytochrome c from mitochondria into the cytosol was significantly increased in response to TMS. TMS caused activation of caspase-3 in a concentration-dependent manner and TMS-mediated caspase-3 activation was partially prevented by the caspase inhibitor, zVAD-fmk. Interestingly, we found that the cytotoxic effect of anticancer drugs such as paclitaxel, docetaxel, or etoposide was enhanced in the presence of TMS. Simultaneous treatment with TCDD also significantly increased cytotoxic effects of TMS alone or TMS and anti-cancer agents. Taken together, our present results indicated that TMS leads to apoptotic cell death in HL-60 cells through activation of caspase-3 activity and release of cytochrome c into cytosol. The ability of TMS to increase cytotoxic effect of anticancer drugs may contribute to its usefulness for cancer chemotherapy.

Suppression of EGFR/STAT3 activity by lupeol contributes to the induction of the apoptosis of human non-small cell lung cancer cells

  • TAE‑RIN MIN;HYUN‑JI PARK;KI‑TAE HA;GYOO‑YONG CHI;YUNG‑HYUN CHOI;SHIN‑HYUNG PARK
    • International Journal of Oncology
    • /
    • v.55 no.1
    • /
    • pp.320-330
    • /
    • 2019
  • The aim of this study was to investigate the underlying mechanisms responsible for the anticancer effects of lupeol on human non-small cell lung cancer (NSCLC). MTT assay and Trypan blue exclusion assay were used to evaluate the cell viability. DAPI staining and flow cytometric analysis were used to detect apoptosis. Molecular docking and western blot analysis were performed to determine the target of lupeol. We found that lupeol suppressed the proliferation and colony formation of NSCLC cells in a dose-dependent manner. In addition, lupeol increased chromatin condensation, poly(ADP-ribose) polymerase (PARP) cleavage, sub-G1 cell populations, and the proportion of Annexin V-positive cells, indicating that lupeol triggered the apoptosis of NSCLC cells. Notably, lupeol inhibited the phosphorylation of epithelial growth factor receptor (EGFR). A docking experiment revealed that lupeol directly bound to the tyrosine kinase domain of EGFR. We observed that the signal transducer and activator of transcription 3 (STAT3), a downstream molecule of EGFR, was also dephosphorylated by lupeol. Lupeol suppressed the nuclear translocation and transcriptional activity of STAT3 and downregulated the expression of STAT3 target genes. The constitutive activation of STAT3 by STAT3 Y705D overexpression suppressed lupeol-induced apoptosis, demonstrating that the inhibition of STAT3 activity contributed to the induction of apoptosis. The anticancer effects of lupeol were consistently observed in EGFR tyrosine kinase inhibitor (TKI)-resistant H1975 cells (EGFR L858R/T790M). Taken together, the findings of this study suggest that lupeol may be used, not only for EGFR TKI-naïve NSCLC, but also for advanced NSCLC with acquired resistance to EGFR TKIs.

Induction of Apoptosis and Inhibition of NO Production by Piceatannol in Human Lung Cancer A549 Cells (A549 인체 폐암세포에서 piceatannol에 의한 apoptosis 유발과 NO 생성의 억제)

  • Choi, Yung-Hyun
    • Journal of Life Science
    • /
    • v.22 no.6
    • /
    • pp.815-822
    • /
    • 2012
  • Piceatannol (trans-3,4,3',5'-tetrahydroxystilbene), a natural stilbene, is an analogue of resveratrol. Although recent experimental data have revealed the health benefit potency of piceatannol, the molecular mechanisms underlying the anti-cancer activity have not yet been studied in detail. In the present study, the further possible mechanisms by which piceatannol exerts its pro-apoptotic action in cultured human lung cancer A549 cells were investigated. Exposure of A549 cells to piceatannol resulted in growth inhibition and induction of apoptosis. Apoptosis induction of A549 cells by piceatannol showed correlation with proteolytic activation of caspase-3, -8, and -9, and concomitant degradation of activated caspase-3 target proteins such as poly (ADP-ribose) polymerase, phospholipase C-${\gamma}1$, ${\beta}$-catenin, and Inhibitor caspase-activated DNase. The increase in apoptosis by piceatannol treatment was also associated with an increase of pro-apoptotic Bax expression and decrease of anti-apoptotic Bcl-2 and Bcl-xL expression, and caused down-regulation of the inhibitor of apoptosis protein family members and up-regulation of Fas and Fas legend. In addition, piceatannol treatment markedly inhibited the expression of mRNA and proteins of inducible nitric oxide (NO) synthase, and the levels of NO production were progressively down-regulated by piceatannol treatment in a dose-dependent fashion. The results indicate that piceatannol may have therapeutic potential against human gastric cancer cells.

Herb medicine Bo-du-san induces caspase dependent apoptosis and cell cycle arrest human gastric cancer cells, SNU-1 (보두산(寶豆散)에 의한 SNU-1 세포의 Apoptosis 유도와 Cell cycle arrest)

  • Yun, Hyun-Joung;Seo, Gyo-Soo;Choi, Jae-Woo;Lee, Hyun-Woo;Heo, Sook-Kyoung;Park, Won-Hwan;Park, Sun-Dong
    • The Korea Journal of Herbology
    • /
    • v.22 no.2
    • /
    • pp.35-43
    • /
    • 2007
  • Objectives : The purpose of this study was to investigate the effect of Bo-du-san (BOS) on apoptosis in human gastric cancer cells, SNU-l cells. BOS, a drug preparation consisting of two herbs, that is, Crotonis Fructus (Strychni ignatii Semen, bodu in Korean) and Glycyrrhizae Radix (Glycyrrhizae uralensis FISCH, Gamcho in Korean). Methodss : In this study, methanol extract of BOS was examined for cytotoxic activity on human gastric cancer cells, SNU-1 cells, using XTT assay, with an IC50 value was 0.7 mg/ml and 0.3 mg/ml at 24 hrs and 48 hrs, respectively. Apoptosis induction by BDS in SNU-l cells was verified by the induction of DNA fragmentation, cleavage of poly ADP-ribose polymerase (PARP), and activation of caspase-3, -8 and -9. Inhibitors of caspase-3, -8 and -9 (Ac-DEVD-CHO, Z-IETD-FMK and Z-LEHD-FMK) efficiently blocked BOS-induced cell death of SNU-l. Resultss : BOS-induced cell death was via caspase dependent apoptosis. Moreover, treatment of BOS result in the decrease the G1/S cycle regulation proteins (cyclin D1 and E) expression and increase CDK inhibitor proteins (p21 and p27) expression, and increase apoptotic protein, p53 expression. Thus, BOS induces apoptosis in SNU-1 cells via cell cycle arrested in G1 phase. Conclusions : These results indicated that BOS has some potential for use as an anti-cancer agent.

  • PDF

Apicidin Induces Apoptosis via Cytochrome c-Mediated Intrinsic Pathway in Human Ovarian Cancer Cells

  • Ahn, Mee-Young;Na, Yong-Jin;Lee, Jae-Won;Lee, Byung-Mu;Kim, Hyung-Sik
    • Biomolecules & Therapeutics
    • /
    • v.17 no.1
    • /
    • pp.17-24
    • /
    • 2009
  • Histone deacetylase (HDAC) inhibitors are a promising class of anticancer agents that inhibit cancer cell growth in vitro and in vivo. Previous report has shown that apicidin inhibited SK-OV-3 cells proliferation and down-regulation of cyclin B1 and CDK1, and up-regulation of $p21^{WAF1}$ and p27. However, the mechanism of apicidin-mediated apoptotic cell death is not clearly understood. For this study, we investigated the mechanism of apoptotic pathway induced by apicidin in human ovarian cancer cell. We found that SK-OV-3 cells treated with apicidin caused an increase in the percentage of cells in the G2/M phase, which preceded apoptosis characterized by the appearance of cells with sub-G1 population. To further investigate the mechanism of apoptosis induction by apicidin, we measured TUNEL assay, poly-ADP ribose polymerase (PARP) cleavage, and caspase activity in SK-OV-3 cells treated with apicidin for 48 h. Apicidin significantly enhanced apoptosis as measured by TUNEL positive apoptotic cells, PARP cleavage, and increased Bax/Bcl-2 ratio. Induction of apoptosis was confirmed by the release of cytochrome c to cytosol. Our data suggest that apicidin-induced apoptosis in SK-OV-3 cells was accompanied by caspase-3 activation and the increase in Bax/Bcl-2 ratio. These data suggest that apicidin may be effective in the treatment of ovarian cancer through activation of intrinsic apoptotic pathway.

Effect of Prunellae Spica on Oxidative Stress and Mitochondrial Dysfunction in the Hepatocyte (하고초(夏枯草)의 간세포에서 항산화 및 미토콘드리아 보호효과)

  • Jang, Mihee;Seo, Hye Lim;Kim, Sang Chan;Kim, Young Woo
    • Journal of Physiology & Pathology in Korean Medicine
    • /
    • v.30 no.1
    • /
    • pp.20-26
    • /
    • 2016
  • Prunellae Spica, the herbaceous plant in the genus Prunella, is a traditional herbal medicine and has been reported to have diuretic, anti-bacterial and anti-oxidant effects. However, the mechanism of its action was not clearly identified. In the present study, we investigated the hepatoprotective effect of Prunellae Spica extract (PSE) against the damage of mitochondria and death in hepatocyte induced by oxidative stress. Treatment of arachidonic acid (AA)+iron significantly induced oxidative stress and apoptosis in the hepatocytes. However, PSE protected cells and inhibited apoptosis by altering the protein levels such as poly(ADP-ribose) polymerase and pro-caspase 3. Moreover, AA+iron induced reactive oxygen species production and mitochondrial dysfunction, and Both of them were inhibited by PSE treatment. PSE markedly activated AMP-activated protein kinase (AMPK), an important regulator in cell survival. Furthermore, this activation by PSE was mediated with liver kinase B1, a major upstream kinase that phosphorylates Thr 172 of AMPKα, and this activation was associated with its cell protection, as assessed by an experiment of a chemical inhibitor. In conclusion, this study demonstrate that PSE protects hepatocytes against oxidative stress as mediated with activation of LKB1-dependent AMPK pathway.

Effects of Rhus verniciflua Stokes Extract on Cell Viability, Cell Cycle Progression and Apoptosis of AGS Cell (건칠(乾漆)이 위암세포의 활성, 세포사멸 및 세포주기관련 유전자 발현에 미치는 영향)

  • An, Jin-Yeong;Ko, Seong-Gyu;Ko, Heung
    • Journal of Physiology & Pathology in Korean Medicine
    • /
    • v.20 no.3
    • /
    • pp.701-709
    • /
    • 2006
  • The Rhus verniciflua Stokes (乾漆-RVS) has been used in traditional East Asia medicine for the therapy of gastritis, stomach cancer, although the mechanism for the biological activity is unclear. In the present study aims to investigate RVS extract contributes to growth inhibitory effect and it's the molecular mechanism on the human gastric cancer cells. AGS (gastric cancer cells) and RIEI (normal cells) were treated to different concentrations and periods of RVS extract $(10{\;}{\sim{{\;}100{\;}ug/mil)$. Growth inhibitory effect was analyzed by measuring FACS study and MTS assay. Cell cycle inhibition was confirmed by measuring CDK2 kinase activity by immunoprecipitation and kinase assay. And apoptosis was confirmed by surveying caspase cascades activation using a pan caspase inhibitor Exposure to RVS extract (50 ug/mll) resulted in a synergistic inhibitory effect on cell growth in AGS cells. Growth inhibition was related with the inhibition of proliferation and induction of apoptosis. The extract induces Gl -cell cycle arrest through the regulation of cyclins, the induction of p27kip1, and the decrease CDK2 kinase activity. And upregulated p27kip1 level is caused by protein stability increment by the reduction of S-phase kinase-associated protein 2 (Skp2), a key molecule related with p27kip1 ubiquitination and degradation, and do novo protein synthesis. Besides, 乾漆 extract induces apoptosis through the expression of Bax, poly(ADP-ribose) polymerase (PARP) and activation of caspase-3. RVS extract induces Gl -cell cycle arrest via accumulation of p27kip1 and apoptosis in human gastric cancer cells but not in normal cells, therefore we suggest that the extract can be used as a novel class of anti-cancer drugs.

Antiproliferative Effect and Apoptotic Mechanism of Extract of Corydalis Yanhusuo on Human Hepatocarcinoma Cells (현호색(玄胡索)이 인체간암세포 증식억제 및 apoptosis 유발에 미치는 영향)

  • Oh, Myun- Taek;Eom, Hyun-Sup;Chi, Gyoo-Yong
    • Journal of Physiology & Pathology in Korean Medicine
    • /
    • v.21 no.6
    • /
    • pp.1437-1449
    • /
    • 2007
  • In this study, the effect of extract of Corydalis yanhusuo (ECT) used in Oriental medicine therapy was investigated on the cell growth and apoptosis of HepG2 human hepatoma cells. It was found that ECT could inhibit the cell growth effectively in a dose-dependent manner, which was associated with morphological change and apoptotic cell death such as formation of apoptotic bodies, DNA fragmentation and increased populations of apoptotic-sub G1 phase. And we observed the effects of ECT on loss of mitochondrial membrane potential (MMP), using the JC-1 probe by DNA flow cytometric analysis. Apoptosis of HepG2 cells by ECT was associated with a down-regulation of anti apoptotic Bcl-2 expression, inhibitor of apoptosis proteins (IAPs) expression and proteolytic activation of caspase-3 and caspase-9. However, ECT did not affect the pro-apoptotic Bax expression and activity of caspase-8. ECT treatment also concomitant degradation and /or inhibition of poly (ADP-ribose) polymerase (PARP), phospholipase C-1 ($PLC{\gamma}1$). Furthermore, ECT treatment caused a dose-dependent inhibition of iNOS and cyclooxygenase-2 (Cox-2). Additionally ECT have been implicated in the regulation of telomerase expression. ECT treatment induced the down-regulation of telomerase reverse transcriptase mRNA (hTERT) expression of HepG2 cells. Taken together, these findings suggest that ECT may be a potential chemotherapeutic agent for the control of HepG2 human hepatoma cells.