• 제목/요약/키워드: zVAD

검색결과 71건 처리시간 0.02초

Effect of Embelin on TRAIL Receptor 2 mAb-induced Apoptosis of TRAIL-resistant A549 Non-small Cell Lung Cancer Cells

  • Jiang, Lei;Hao, Jin-Li;Jin, Mu-Lan;Zhang, Yun-Gang;Wei, Ping
    • Asian Pacific Journal of Cancer Prevention
    • /
    • 제14권10호
    • /
    • pp.6115-6120
    • /
    • 2013
  • Introduction: Some non-small cell lung cancer (NSCLC) tumor cells are insensitive to tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) -based therapy. This study was conducted to examine the effect of embelin on the sensitivity of the A549 NSCLC cell line to TRAIL receptor2 (TRAILR2) monoclonal antibodies and to investigate the potential mechanisms. Materials and Methods: A549 cells were treated with embelin, TRAILR2 mAb or a combination of both. Cell viability was measured using ATPlite assay and apoptosis rates were determined by flow cytometry with AnnexinV-FITC and propidium iodide staining, with the expression levels of proteins analyzed by Western blotting. Results: The cell survival rate of separate treatments with 100 ng/ml TRAILR2 antibody or 25 uM embelin were $81.5{\pm}1.57%$ and $61.7{\pm}2.84%$, respectively. Their combined use markedly decreased cell viability in A549 cells to $28.1{\pm}1.97%$ (P<0.05). The general caspase inhibitor Z-VAD-FMK could inhibit the embelin-enhanced sensitivity of A549 cells to TRAILR2 mAb ($75.97{\pm}3.17%$)(P<0.05). Both flow cytometry and cell morphological analysis showed that embelin was able to increase TRAIL-induced apoptosis in A549 cells. Combined treatment with embelin and TRAILR2 mAb augmented the activation of initiator caspases and effector caspase. In addition, A549 cells showed increasing levels of TRAILR2 protein and decreasing levels of Bcl-2, survivin and c-FLIP following the treatment with embelin+TRAILR2 mAb. Conclusions: Embelin could enhance TRAIL-induced apoptosis in A549 cells. The synergistic effect of the combination treatment might be due to modulation of multiple components in the TRAIL receptor-mediated apoptotic signaling pathway, including TRAILR2, XIAP, survivin, Bcl-2 and c-FLIP.

Beta-asarone Induces LoVo Colon Cancer Cell Apoptosis by Up-regulation of Caspases through a Mitochondrial Pathway in vitro and in vivo

  • Zou, Xi;Liu, Shen-Lin;Zhou, Jin-Yong;Wu, Jian;Ling, Bo-Fan;Wang, Rui-Ping
    • Asian Pacific Journal of Cancer Prevention
    • /
    • 제13권10호
    • /
    • pp.5291-5298
    • /
    • 2012
  • Beta-asarone is one of the main bioactive constituents in traditional Chinese medicine Acorus calamu. Previous studies have shown that it has antifungal and anthelmintic activities. However, little is known about its anticancer effects. This study aimed to determine inhibitory effects on LoVo colon cancer cell proliferation and to clarify the underlying mechanisms in vitro and in vivo. Dose-response and time-course anti-proliferation effects were examined by MTT assay. Our results demonstrated that LoVo cell viability showed dose- and time-dependence on ${\beta}$-asarone. We further assessed anti-proliferation effects as ${\beta}$-asarone-induced apoptosis by annexin V-fluorescein isothiocyanate/propidium iodide assay usinga flow cytometer and observed characteristic nuclear fragmentation and chromatin condensation of apoptosis by microscopy. Moreover, we found the apoptosis to be induced through the mitochondrial/caspase pathway by decreasing mitochondrial membrane potential (MMP) and reducing the Bcl-2-to-Bax ratio, in addition to activating the caspase-9 and caspase-3 cascades. Additionally, the apoptosis could be inhibited by a pan-caspase inhibitor, carbobenzoxy-valyl-alanyl-aspartyl-[O-methyl]-fluoromethylketone (Z-VAD-FMK). When nude mice bearing LoVo tumor xenografts were treated with ${\beta}$-asarone, tumor volumes were reduced and terminal deoxynucleotide transferase-mediated dUTP nick end labeling (TUNEL) assays of excised tissue also demonstrated apoptotic changes. Taken together, these findings for the first time provide evidence that ${\beta}$-asarone can suppress the growth of colon cancer and the induced apoptosis is possibly mediated through mitochondria/caspase pathways.

Down-Regulation of Survivin by Nemadipine-A Sensitizes Cancer Cells to TRAIL-Induced Apoptosis

  • Park, Seong Ho;Park, So Jung;Kim, Joo-Oh;Shin, Ji Hyun;Kim, Eun Sung;Jo, Yoon Kyung;Kim, Jae-Sung;Park, So Jung;Jin, Dong-Hoon;Hwang, Jung Jin;Lee, Seung Jin;Jeong, Seong-Yun;Lee, Chaeyoung;Kim, InKi;Cho, Dong-Hyung
    • Biomolecules & Therapeutics
    • /
    • 제21권1호
    • /
    • pp.29-34
    • /
    • 2013
  • The tumor necrosis factor (TNF)-related apoptosis-inducing ligand (TRAIL) is a member of the tumor necrosis factor family of cytokines. TRAIL selectively induces apoptotic cell death in various tumors and cancer cells, but it has little or no toxicity in normal cells. Agonism of TRAIL receptors has been considered to be a valuable cancer-therapeutic strategy. However, more than 85% of primary tumors are resistant to TRAIL, emphasizing the importance of investigating how to overcome TRAIL resistance. In this report, we have found that nemadipine-A, a cell-permeable L-type calcium channel inhibitor, sensitizes TRAIL-resistant cancer cells to this ligand. Combination treatments using TRAIL with nemadipine-A synergistically induced both the caspase cascade and apoptotic cell death, which were blocked by a pan caspase inhibitor (zVAD) but not by autophagy or a necrosis inhibitor. We further found that nemadipine-A, either alone or in combination with TRAIL, notably reduced the expression of survivin, an inhibitor of the apoptosis protein (IAP) family of proteins. Depletion of survivin by small RNA interference (siRNA) resulted in increased cell death and caspase activation by TRAIL treatment. These results suggest that nemadipine-A potentiates TRAIL-induced apoptosis by down-regulation of survivin expression in TRAIL resistant cells. Thus, combination of TRAIL with nemadipine-A may serve a new therapeutic scheme for the treatment of TRAIL resistant cancer cells, suggesting that a detailed study of this combination would be useful.

β-lapachone-Induced Apoptosis of Human Gastric Carcinoma AGS Cells Is Caspase-Dependent and Regulated by the PI3K/Akt Pathway

  • Yu, Hai Yang;Kim, Sung Ok;Jin, Cheng-Yun;Kim, Gi-Young;Kim, Wun-Jae;Yoo, Young Hyun;Choi, Yung Hyun
    • Biomolecules & Therapeutics
    • /
    • 제22권3호
    • /
    • pp.184-192
    • /
    • 2014
  • ${\beta}$-lapachone is a naturally occurring quinone that selectively induces apoptotic cell death in a variety of human cancer cells in vitro and in vivo; however, its mechanism of action needs to be further elaborated. In this study, we investigated the effects of ${\beta}$-lapachone on the induction of apoptosis in human gastric carcinoma AGS cells. ${\beta}$-lapachone significantly inhibited cellular proliferation, and some typical apoptotic characteristics such as chromatin condensation and an increase in the population of sub-G1 hypodiploid cells were observed in ${\beta}$-lapachone-treated AGS cells. Treatment with ${\beta}$-lapachone caused mitochondrial transmembrane potential dissipation, stimulated the mitochondria-mediated intrinsic apoptotic pathway, as indicated by caspase-9 activation, cytochrome c release, Bcl-2 downregulation and Bax upregulation, as well as death receptor-mediated extrinsic apoptotic pathway, as indicated by activation of caspase-8 and truncation of Bid. This process was accompanied by activation of caspase-3 and concomitant with cleavage of poly(ADP-ribose) polymerase. The general caspase inhibitor, z-VAD-fmk, significantly abolished ${\beta}$-lapachone-induced cell death and inhibited growth. Further analysis demonstrated that the induction of apoptosis by ${\beta}$-lapachone was accompanied by inactivation of the phosphatidylinositol 3-kinase (PI3K)/Akt signaling pathway. The PI3K inhibitor LY29004 significantly increased ${\beta}$-lapachone-induced apoptosis and growth inhibition. Taken together, these findings indicate that the apoptotic activity of ${\beta}$-lapachone is probably regulated by a caspase-dependent cascade through activation of both intrinsic and extrinsic signaling pathways, and that inhibition of the PI3K/Akt signaling may contribute to ${\beta}$-lapachone-mediated AGS cell growth inhibition and apoptosis induction.

Quinacrin Induces Cytochrome c-dependent Apoptotic Signaling in Human Cervical Carcinoma Cells

  • Fasanmade, Adedigbo A.;Owuor, Edward D.;Ee, Rachel P.L.;Qato, Dima;Heller, Mark;Kong, Ah Ng Tony
    • Archives of Pharmacal Research
    • /
    • 제24권2호
    • /
    • pp.126-135
    • /
    • 2001
  • Quinacrine (QU), a phospholipase-A2 (PLA-2) inhibitor has been used clinically as a chemotherapeutic adjuvant. To understand the mechanisms leading to its chemotherapeutic effect, we have investigated QU-induced apoptotic signaling pathways in human cervical squamous carcinoma HeLa cells. In this study, we found that QU induced cytochrome c-dependent apoptotic signaling. The release of pro-apoptotic cytochrome c was QU concentration- and time-dependent, and preceded activation of caspase-9 and -3. Flow cytometric FACScan analysis using fluorescence intensities of $DiOC_6$/ demonstrated that QU-induced cytochrome c release was independent of mitochondrial permeability transition (MPT), since the concentrations of QU that induced cytochrome c release did not alter mitochondrial membrane potential (${\blacktriangle}{\Psi}_m$). Moreover, kinetic analysis of caspase activities showed that cytochrome c release led to the activation of caspase-9 and downstream death effector caspase-3, Caspase-3 inhibitor (Ac-DEVD-CHO) partially blocked QU-induced apoptosis, suggesting the importance of caspase-3 in this apoptotic signaling mechanism. Supplementation with arachidonic acid (AA) sustained caspase-3 activation induced by QU. Using inhibitors against cellular arachidonate metabolism of lipooxygenase (Nordihydroxyguaiaretic Acid, NDGA) and cyclooxygenase (5,8,11,14-Eicosatetraynoic Acid, ETYA) demonstrated that QU-induced apoptotic signaling may be dependent on its role as a PLA-2 inhibitor. Interestingly, NDCA attenuated QU-induced cytochrome c release, caspase activity as well as apoptotic cell death. The blockade of cytochrome c release by NDCA was much more effective than that attained with cyclosporin A (CsA), a MPT inhibitor. ETYA was not effective in blocking cytochrome c release, except under very high concentrations. Caspase inhibitor z-VAD blocked the release of cytochrome c suggesting that this signaling event is caspase dependent, and caspase-8 activation may be upstream of the mitochondrial events. In summary, we report that QU induced cytochrome c-dependent apoptotic signaling cascade, which may be dependent on its role as a PLA-2 inhibitor. This apoptotic mechanism induced by QU may contribute to its known chemotherapeutic effects.

  • PDF

Serine pretease 억제제인 4-(2-aminoethyl) benzensulfonylfluoride (AEBSF)에 의한 호중구의 자연 세포사멸의 지연과 수지상 세포로의 전이분화 연구 (Effects of AEBSF on the Delay of Spontaneous Apoptosis and the Trans-Differentiation of Human Neutrophils into Dendritic Cells)

  • 박해영;곽종영
    • 생명과학회지
    • /
    • 제17권7호통권87호
    • /
    • pp.948-955
    • /
    • 2007
  • 생체 면역반응에 중요한 역할을 하는 호중구의 세포사멸은 자연적으로 일어나거나 여러 외부자극에 의한 신호의 전달에 의해 증가하거나 지연된다. 또한 사이토카인과 같은 분화제에 의해 세포사멸이 지연되고 항원 제시 기능을 가진 수지상 세포로 분화되기도 한다. 본 연구에서는 세포사멸 억제제와 사이토카인을 이용한 시스템에서 호중구가 수지상 세포로 분화되는가를 조사하였다. Pancaspase와 serine protease의 억제제인 zVAD-fmk와 AEBSF를 처리하였을 때 호중구의 세포사멸은 현저히 감소되며 AEBSF는 caspase-3와 serine protease활성을 모두 억제하였다. 호중구의 세포사멸을 효과적으로 억제하는 AEBSF와 함께 분화제로 널리 쓰이는 CM-CSF를 같이 처리하여 3일 동안 배양하면 수지상 세포에서 높이 발현되는 CD8O, CD83 및 MHC class ll의 세포표면 마커의 발현이 증가하였다. AEBSF와 CM-CSF를 처리한 호중구를 T-세포와 함께 배양하였을 때 SEB가 존재할 경우 T-세포가 증식되었으며 SEB가 없이도 $IFN{\gamma}$는 생성되었다. 이들 결과들로 부터 serine protease 억제제인 AEBSF를 호중구에 처리하여 세포사멸을 효과적으로 억제하는 것과 수지상 세포로의 분화를 촉진하는 사이토카인인 CM-CSF의작용을 나타내게 하는 조건과는 서로 상호적으로 연관되어 작용할 수 있다는 것을 제시하고 있다.

인체폐암 A549 세포에서 Baicalein에 의한 세포사멸 유도: Apoptosis와 Autophagy 경로의 역할 (Baicalein induces cell death in Human Lung Carcinoma A549 Cells: Role of Apoptosis and Autophagy pathway)

  • 김철환;황병수;정용태;김민진;신수영;오영택;엄정혜;이승영;최경민;조표연;정진우
    • 한국자원식물학회:학술대회논문집
    • /
    • 한국자원식물학회 2019년도 춘계학술대회
    • /
    • pp.112-112
    • /
    • 2019
  • Baicalein is one of the main flavonoids derived from roots of Scutellaria baicalensis Georgi, a traditional Oriental medicine. Although baicalein has high antitumor effect on several human carcinomas, the mechanism responsible for this property is not unclear. In this study, the data revealed that baicale-ininduced growth inhibition was associated with the induction of apoptosis connecting with cytochrome c release, down-regulation of anti-apoptotic Bcl-xl and increased the percentage of cells with a loss of mitochondria membrane permeabilization. Baicalein also induced the proteolytic activation of caspases and cleavage of PARP; however, blockage of caspases activation by z-VAD-fmk inhibited baicalein-induced apoptosis. In addition, baicalein enhanced the formation of autophagosomes and up-regulated LC3-II/LC3-I ratio. Interestingly, the pretreatment of bafilomycin A1 recovered baicalein-induced cell death suggesting that autophagy by baicalein roles as protective autophagy. Taken together, our results indicated that this flavonoid induces apoptosis and cell protective autophagy. These data means combination treatment with baicalein and autophagy inhibitor might be a promising anticancer drug.

  • PDF

YM155 처리에 의한 두경부 암 AMC-HN4 세포 세포자멸사 유도 효과 (YM155 Induces Apoptosis through Downregulation of Anti-apoptotic Proteins in Head and Neck AMC-HN4 Cells)

  • 장호준;권택규;김동은
    • 생명과학회지
    • /
    • 제29권3호
    • /
    • pp.318-324
    • /
    • 2019
  • 두경부암은 전세계에서 발병률이 여섯 번째로 높은 암으로 그동안 수술적 치료를 선호하였으나 광범위한 절제에 따른 기능적 장애로 인해 항암치료에 대한 관심이 높아지고 있다. 두경부암에서 cisplatin이 가장 많이 사용되는 항암제이나 cisplatin 내성이 문제가 되고 있다. 따라서 부작용은 줄이고, 약제내성 기전에 대해 이해하여 암세포의 사멸은 증대시키는 새로운 항암제의 개발이 필요하다. Survivin은 inhibitor of apoptosis proteins (IAPs) family 중 하나로 두경부암에서 과발현되어 있다. YM155는 survivin을 억제하는 분자로 본 연구를 통해 YM155의 처리 후 두경부 암세포의 세포자멸사가 유도되며, 뇌암 세포와 신장암 세포에서도 세포자멸사가 유도됨을 확인할 수 있었다. 반면에 정상세포인 mesangial cells에는 YM155가 세포자멸사에 영향을 주지 않았다. YM155는 caspase의 활성화를 통해 세포자멸사를 촉진하며, anti-apoptotic protein인 c-FLIP, Mcl-1, survivin의 발현을 저해하는 것으로 확인되었다. YM155는 두경부 뿐만 아니라 다른 장기의 악성종양 치료법의 개발에 활용 될 수 있을 것으로 생각된다.

MHY2251, a New SIRT1 Inhibitor, Induces Apoptosis via JNK/p53 Pathway in HCT116 Human Colorectal Cancer Cells

  • Yong Jung Kang;Young Hoon Kwon;Jung Yoon Jang;Jun Ho Lee;Sanggwon Lee;Yujin Park;Hyung Ryong Moon;Hae Young Chung;Nam Deuk Kim
    • Biomolecules & Therapeutics
    • /
    • 제31권1호
    • /
    • pp.73-81
    • /
    • 2023
  • Sirtuins (SIRTs) belong to the nicotinamide adenine dinucleotide (NAD+)-dependent class III histone deacetylase family. They are key regulators of cellular and physiological processes, such as cell survival, senescence, differentiation, DNA damage and stress response, cellular metabolism, and aging. SIRTs also influence carcinogenesis, making them potential targets for anticancer therapeutic strategies. In this study, we investigated the anticancer properties and underlying molecular mechanisms of a novel SIRT1 inhibitor, MHY2251, in human colorectal cancer (CRC) cells. MHY2251 reduced the viability of various human CRC cell lines, especially those with wild-type TP53. MHY2251 inhibited SIRT1 activity and SIRT1/2 protein expression, while promoting p53 acetylation, which is a target of SIRT1 in HCT116 cells. MHY2251 treatment triggered apoptosis in HCT116 cells. It increased the percentage of late apoptotic cells and the sub-G1 fraction (as detected by flow cytometric analysis) and induced DNA fragmentation. In addition, MHY2251 upregulated the expression of FasL and Fas, altered the ratio of Bax/Bcl-2, downregulated the levels of pro-caspase-8, -9, and -3 proteins, and induced subsequent poly(ADP-ribose) polymerase cleavage. The induction of apoptosis by MHY2251 was related to the activation of the caspase cascade, which was significantly attenuated by pre-treatment with Z-VAD-FMK, a pan-caspase inhibitor. Furthermore, MHY2251 stimulated the phosphorylation of c-Jun N-terminal kinase (JNK), and MHY2251-triggered apoptosis was blocked by pre-treatment with SP600125, a JNK inhibitor. This finding indicated the specific involvement of JNK in MHY2251-induced apoptosis. MHY2251 shows considerable potential as a therapeutic agent for targeting human CRC via the inhibition of SIRT1 and activation of JNK/p53 pathway.

3-Deoxysappanchalcone Inhibits Cell Growth of Gefitinib-Resistant Lung Cancer Cells by Simultaneous Targeting of EGFR and MET Kinases

  • Jin-Young Lee;Seung-On Lee;Ah-Won Kwak;Seon-Bin Chae;Seung-Sik Cho;Goo Yoon;Ki-Taek Kim;Yung Hyun Choi;Mee-Hyun Lee;Sang Hoon Joo;Jin Woo Park;Jung-Hyun Shim
    • Biomolecules & Therapeutics
    • /
    • 제31권4호
    • /
    • pp.446-455
    • /
    • 2023
  • The mechanistic functions of 3-deoxysappanchalcone (3-DSC), a chalcone compound known to have many pharmacological effects on lung cancer, have not yet been elucidated. In this study, we identified the comprehensive anti-cancer mechanism of 3-DSC, which targets EGFR and MET kinase in drug-resistant lung cancer cells. 3-DSC directly targets both EGFR and MET, thereby inhibiting the growth of drug-resistant lung cancer cells. Mechanistically, 3-DSC induced cell cycle arrest by modulating cell cycle regulatory proteins, including cyclin B1, cdc2, and p27. In addition, concomitant EGFR downstream signaling proteins such as MET, AKT, and ERK were affected by 3-DSC and contributed to the inhibition of cancer cell growth. Furthermore, our results show that 3-DSC increased redox homeostasis disruption, ER stress, mitochondrial depolarization, and caspase activation in gefitinib-resistant lung cancer cells, thereby abrogating cancer cell growth. 3-DSC induced apoptotic cell death which is regulated by Mcl-1, Bax, Apaf-1, and PARP in gefitinib-resistant lung cancer cells. 3-DSC also initiated the activation of caspases, and the pan-caspase inhibitor, Z-VAD-FMK, abrogated 3-DSC induced-apoptosis in lung cancer cells. These data imply that 3-DSC mainly increased mitochondria-associated intrinsic apoptosis in lung cancer cells to reduce lung cancer cell growth. Overall, 3-DSC inhibited the growth of drug-resistant lung cancer cells by simultaneously targeting EGFR and MET, which exerted anti-cancer effects through cell cycle arrest, mitochondrial homeostasis collapse, and increased ROS generation, eventually triggering anti-cancer mechanisms. 3-DSC could potentially be used as an effective anti-cancer strategy to overcome EGFR and MET target drug-resistant lung cancer.