• Title/Summary/Keyword: Caspase 1

Search Result 1,285, Processing Time 0.03 seconds

Pro-Apoptotic Effect of Mori Cortex Radicis in A549 Lung Cancer Cells (상백피가 A549 폐암세포주의 세포사에 미치는 영향)

  • Bae Oh-Sung;Yoo Yeong-Min;Lee Seon-Goo
    • Journal of Physiology & Pathology in Korean Medicine
    • /
    • v.19 no.6
    • /
    • pp.1563-1567
    • /
    • 2005
  • Mori Cortex Radicis is distributed in Northwestern China, northern Asia, northern Europe, North America, and Korea. This extracts drops sugar in bloods and inhibits cyclic AMP phophodiesterase. In this study, we investigated whether Mori Cortex Radicis would cause apoptotic death of A549 lung cancer cells. To examine the apoptotic effect of Mori Cortex Radicis, cytotoxicity assay, DNA fragmentation analysis, caspase-3 activity assay, and Western blotting for caspase-3, caspase-9 and poly(ADP-ribose) polymerase (PARP) and cytochrome c were performed. Treatment of cells with Mori Cortex Radicis was shown to induce cell death in a dose-dependent manner. DNA fragmentation was made in response to Mori Cortex Radicis. The active fragments of caspase-3, caspase-9 and PARP were almost completely induced and cytochrome c was released following exposure to Mori Cortex Radicis. To elucidate the apoptotic mechanisms, RT-PCR and Western blot analyses for the expression of Bcl-2, Bu and Cox-2 were carried out. Treatment with Mori Cortex Radicis was expressed the reduction of Bcl-2 and Cox-2 and the induction of Bax. Especially p21 and p53 were increased prior to untreated control, while cyclin E and cyclin D1 decreased in the cytosol. These results suggest that the effect Mori Cortex Radicis is associated with the cell cycle arrest and pro-apoptotic cell death in A549 lung cancer cells.

Potentiation of Apoptin-Induced Apoptosis by Cecropin B-Like Antibacterial Peptide ABPs1 in Human HeLa Cervical Cancer Cell Lines is Associated with Membrane Pore Formation and Caspase-3 Activation

  • Birame, Basse Mame;Wang, Jigui;Yu, Fuxian;Sun, Jiazeng;Li, Zhili;Liu, Weiquan
    • Journal of Microbiology and Biotechnology
    • /
    • v.24 no.6
    • /
    • pp.756-764
    • /
    • 2014
  • Apoptin, a chicken anemia virus-encoded protein, induces apoptosis in chicken or human tumor cells, localizing in their nuclei as opposed to the cytoplasm of non-transformed cells. The present study was undertaken to investigate whether ABPs1 could potentiate apoptin-induced apoptosis in HeLa cells. ABPs1 and the apoptin genes were successfully cloned into pIRES2-EGFP expression vector and expressed in HeLa cells. We report that ABPs1 augments apoptin cell growth inhibition in a concentration- and time-dependent manner. The DAPI staining and scanning electron microscopy observations revealed apoptotic bodies and plasma membrane pores, which were attributed to apoptin and ABPs1, respectively. Further, ABPs1 in combination with apoptin was found to increase the expression of Bax and to decrease the expression of survivin compared with either agent alone or the control. The apoptotic rate of HeLa cells treated with ABPs1 and apoptin in combination for 48 h was 53.95%. The two-gene combination increased the caspase-3 activity of HeLa cells. Taken together, our study suggests that ABPs1 combined with apoptin significantly inhibits HeLa cell proliferation, and induces cell apoptosis through membrane defects, up-regulation of Bax expression, down-regulation of survivin expression, and activation of the caspase-3 pathway. Thus, the combination of ABPs1 and apoptin could serve as a means to develop novel gene therapeutic agents against human cervical cancer.

Ginsenoside Rd protects cerebral endothelial cells from oxygen-glucose deprivation/reoxygenation induced pyroptosis via inhibiting SLC5A1 mediated sodium influx

  • Li, Suping;Yu, Nengwei;Xu, Fei;Yu, Liang;Yu, Qian;Fu, Jing
    • Journal of Ginseng Research
    • /
    • v.46 no.5
    • /
    • pp.700-709
    • /
    • 2022
  • Background: Ginsenoside Rd is a natural compound with promising neuroprotective effects. However, the underlying mechanisms are still not well-understood. In this study, we explored whether ginsenoside Rd exerts protective effects on cerebral endothelial cells after oxygen-glucose deprivation/reoxygenation (OGD/R) treatment and its potential docking proteins related to the underlying regulations. Method: Commercially available primary human brain microvessel endothelial cells (HBMECs) were used for in vitro OGD/R studies. Cell viability, pyroptosis-associated protein expression and tight junction protein degradation were evaluated. Molecular docking proteins were predicted. Subsequent surface plasmon resonance (SPR) technology was utilized for validation. Flow cytometry was performed to quantify caspase-1 positive and PI positive (caspase-1+/PI+) pyroptotic cells. Results: Ginsenoside Rd treatment attenuated OGD/R-induced damage of blood-brain barrier (BBB) integrity in vitro. It suppressed NLRP3 inflammasome activation (increased expression of NLRP3, cleaved caspase-1, IL-1β and GSDMD-N terminal (NT)) and subsequent cellular pyroptosis (caspase-1+/PI + cells). Ginsenoside Rd interacted with SLC5A1 with a high affinity and reduced OGD/R-induced sodium influx and potassium efflux in HBMECs. Inhibiting SLC5A1 using phlorizin suppressed OGD/R-activated NLRP3 inflammasome and pyroptosis in HBMECs. Conclusion: Ginsenoside Rd protects HBMECs from OGD/R-induced injury partially via binding to SLC5A1, reducing OGD/R-induced sodium influx and potassium efflux, thereby alleviating NLRP3 inflammasome activation and pyroptosis.

Effect of Acacetin on the Apoptosis Induction of HeLa Cells (Acacetin의 HeLa 세포 Apoptosis 유도 효과)

  • Han, Su Jung;Kim, An Keun
    • YAKHAK HOEJI
    • /
    • v.59 no.1
    • /
    • pp.17-22
    • /
    • 2015
  • In this study, we examined the effect of acacetin on the apoptosis induction of HeLa human cervical cancer cells. The results showed that acacetin inhibited the cell viability and induced apoptosis, leading to PARP cleavage and activation of caspase-9, -3, and -7. Moreover, acacetin-induced apoptosis was attenuated by a broad caspase inhibitor, z-VAD-fmk. Also, acacetin resulted in a loss of mitochondria membrane potential. Taken together, our results demonstrate that the induction of apoptosis by acacetin in HeLa cells is associated with caspase activation via the mitochondria pathway.

Induction of apoptosis in human promyelocytic leukaemia HL -60 cells by yomogin involves release of cytochrome c and activation of caspase

  • Jeong, Seoung-Hee;Koo, Sung-Ja;Ryu, Shi-Yong;Park, Hee-Jun;Lee, Kyung-Tae
    • Proceedings of the PSK Conference
    • /
    • 2002.10a
    • /
    • pp.319.1-319.1
    • /
    • 2002
  • Yomogin. an eudesmane sesquiterpene isolated from Artemisia princeps, was found to induce apoptosis in human promyelocytic leukaemia, HL -60 cell with characteristic apoptotic features like nuclear condensation, apoptotic body formation, flipping of membrane phosphatidylserine, release of mitochondrial cytochrome c and caspase-8. -9. and -3 activation. Furthermore. early yomogin-induced cytochrome c release was not affected by the caspase inhibitor Z-VAd fmk and preceded loss of mitochondrial membrane potential. The results suggest that induction of apoptosis by yomogin may provide a pivotal mechanism for their cancer chemopreventive function.

  • PDF

Green Tea (-)EGCG Induces the Apoptotic Death of Lung Cancer Cells via Activation of c-Jun N-terminal Kinase 1 and Activating Protein-1 (녹차의 (-)EGCG에 의한 사람 폐암 세포주 A549의 c-Jun N-terminal Kinase 1과 Activating Protein-1활성화를 통한 세포고사)

  • 박지선;신미경;손희숙;박래길;김명선;정원훈
    • Journal of Nutrition and Health
    • /
    • v.35 no.1
    • /
    • pp.53-59
    • /
    • 2002
  • Green tea has been recognized as a favorite beverage for centuries in Easter and Westers cultures. Recently, anti-tumor effects of green tea constituents have received increasing attention. However, the mechanism of catechin-mediated cytotoxicity against tumor cells remains to be elusive. To elucidate the mechanical insights of anti-tumor effects, (-)epigallocatechin-gallate(EGCG) of catechin was applied to human lung cancer A549 cells. (-)EGCG induced the death of A549 cells, which was revealed as apoptosis in DNA fragmentation assay. (-)EGCG induced the activation of caspase family cysteine proteases including capase-3, -8 and -9 proteases in A549 cells. Furthermore, (-)EGCG increased the phosphotransferase activity of c-Jun N-terminal kinase 1JNK 1), which further induced tole transcriptional activation of activating protein-1(AP-1) in A549 cells. We suggest that (-)EGCG-induced apotosis of A549 cells is mediated by signaling pathway involving caspase family cysteine protease, JNK1 and transcription factor, AP-1.

Sodium Salicylate(NaSaL) Induces Apoptosis of NCI-H1299 Lung Carcinoma Cells via Activation Caspase-3 Protease (NCI-H1299 폐암 세포주에서 Caspase-3 Protease 활성을 통한 Sodium Salicylate(NaSaL)의 세포고사)

  • Shim, Hyeok;Yang, Sei-Hoon;Bak, Sang-Myeon;Jeong, Eun-Taik
    • Tuberculosis and Respiratory Diseases
    • /
    • v.53 no.5
    • /
    • pp.485-496
    • /
    • 2002
  • Background : Nonsteroidal anti-inflammatory drugs (NSAIDs) are useful in the chemoprevention of colon cancers. Continuous NSAID use results in a 40% to 50% reduction in the relative risk of colorectal cancer. The precise mechanism by which NSAIDs prevent and/or cause the regression of colorectal tumors is not known. Some investigators have reported that certain NSAIDs induce apoptosis and alter the expression of the cell cycle regulatory genes in some carcinoma cells when administered at a relatively high concentration. However, the possibility of NSAIDs application as chemopreventive agents in lung cancers remains to be elucidated. To address this question, the human lung cancer cell line NCI-H1299 was used to investigate whether or not NSAIDs might induce the apoptotic death of NCI-H1299 cells. Methods : A viability test was carried out using a MTT assay. Apoptosis was measured by flow cytometric analysis and unclear staining(DAPI). The talytic activity of the caspase family was measured by the fluirigenic cleavage of biosubstrates. To define the mechanical basis of apoptosis, western blot was performed to analyze the expression of the death substrates(PARP and ICAD). Results : NaSaL significantly decreased the viability of the NCI-H1299 cells, which was revealed as apoptosis characterized by an increase in the $subG_0/G_1$ population and unclear fragmentation. The catalytic activity of caspase-3 protease began to increase after 24 Hr and reached a peak 30 Hr after treatment with 10 mM NaSaL. In contrast, caspase-6, 8, and 9 proteases did not have a significantly altered enzymatic activity. Consistent with activation of caspase-3 protease, NaSaL induced the cleavage of the protease biosubstrate. Conclusion : NaSaL induces the apoptotic death of NCI-H1299 human lung cancer cells via the activation of caspase-3 protease.

HS 1-Associated Protein X-1 Is Cleaved by Caspase-3 During Apoptosis

  • Lee, Ah Young;Lee, Yoora;Park, Yun Kyung;Bae, Kwang-Hee;Cho, Sayeon;Lee, Do Hee;Park, Byoung Chul;Kang, Sunghyun;Park, Sung Goo
    • Molecules and Cells
    • /
    • v.25 no.1
    • /
    • pp.86-90
    • /
    • 2008
  • Caspase-3 (CASP3) plays a key role in apoptosis. In this study, HAX-1 was identified as a new substrate of CASP3 during apoptosis. HAX-1 was cleaved by CASP3 during etoposide-(ETO) induced apoptosis, and this event was inhibited by a CASP3-specific inhibitor. The cleavage site of HAX-1, at $Asp^{127}$, was located using N-terminal amino acid sequencing of in vitro cleavage products of recombinant HAX-1. Overexpression of HAX-1 inhibited ETO-induced apoptotic cell death. It also inhibited CASP3 activity. Together, these results suggest that HAX-1, a substrate of CASP3, inhibits the apoptotic process by inhibiting CASP3 activity.

Calpains and Apoptosis

  • Tagliarino, Colleen;Pink, John J.;Boothman, David A.
    • Animal cells and systems
    • /
    • v.5 no.4
    • /
    • pp.267-274
    • /
    • 2001
  • Calpains are a family of cysteine proteases existing primarily in two forms designated by the $Ca^{2+}$ concentration needed for activation in vitro, $\mu$-calpain (calpain-I) and m-calpain (calpain-II). The physiologica1 roles of calpains remain unclear. Many groups have proposed a role for calpains In apoptosis, but their patterns of activation are not well characterized. Calpains have been implicated in neutrophil apoptosis, glucocorticoid-induced thymocyte apoptosis, as well as many other apoptotic pathways. Calpain activation in apoptosis is usually linked upstream or downstream to caspase activation, or in a parallel pathway alongside caspase activation. Calpains have been suggested to be involved in DNA fragmentation (via endonuclease activation), but also as effector proteases that cleave cellular proteins involved in DNA repair, membrane associated proteins and other homeostatic regulatory proteins. Recently, our laboratory demonstrated $\mu$-calpain activation in NAD(P)H: quinone oxidoreducatse 1 (NQO1)-expressing cells after exposure to $\beta$-lapachone, a novel quinone and potential chemo- and radio-therapeutic agent. Increased cytosolic $Ca^{2+}$ in NQO1-expressing cells after $\beta$-lapachone exposures were shown to lead to $\mu$-calpain activation. In turn, $\mu$-calpain activation was important for substrate proteolysis and DNA fragmentation associated with apoptosis. Upon activation, $\mu$-calpain translocated to the nucleus where it could proteolytically cleave PARP and p53. We provided evidence that $\beta$-lapachone-induced, $\mu$-calpain stimulated, apoptosis did not involve any of the known caspases; known apoptotic caspases were not activated after $\beta$-lapachone treatment of NQO1-expressing cells, nor did caspase inhibitors have any effect on $\beta$-1apachone-induced cell death. Elucidation of processes by which $\beta$-1apachone-stimulated $\mu$-calpain activation and calpains ability to activate endonucleases and induce apoptosis independent of caspase activity will be needed to further develop/modulate $\beta$-lapachone for treatment of human cancers that over-express NQO1.

  • PDF

Effects of Melatonin and Fluid Shear Stress on 3T3-L1 Preadipocytes (3T3-L1 지방전구세포에서 멜라토닌과 유체전단응력의 영향)

  • Lee, Jeongkun;Lee, Yeong Hun;Park, Chae Lim;Kim, Chi Hyun
    • Journal of Biomedical Engineering Research
    • /
    • v.39 no.3
    • /
    • pp.109-115
    • /
    • 2018
  • Obesity is a worldwide disease caused by the excessive proliferation of adipocytes. Multiple factors, including melatonin and physical loading, are involved in the control of obesity. Melatonin has been shown to induce apoptosis on preadipocytes while physical loading such as fluid shear stress (FSS) affects the proliferation and differentiation of adipocytes. Here, we studied the combined effects of melatonin and FSS on 3T3-L1 preadipocytes. For physical loading, preadipocytes were stimulated with a maximum dynamic fluid shear stress of 1 Pa at 1 Hz for 2 hours with/without melatonin. The experiment conditions were divided into four groups: (1) control, (2) 1 mM melatonin treatment, (3) FSS, and (4) combined 1 mM melatonin and FSS. All groups had a fixed duration time of 2 hours. ERK, p-ERK, COX-2, $C/EBP{\beta}$, $PPAR{\gamma}$, osteopontin, Bax, caspase-3 and caspase-8 proteins were assessed by Western blot analysis. GAPDH was used as a control. Results showed that combined melatonin and FSS treatment activated the ERK/MAPK pathway but not COX-2. Furthermore, combined melatonin and FSS treatment significantly decreased $C/EBP{\beta}$ and $PPAR{\gamma}$ compared to other groups. However, caspase-3 and caspase-8 did not result in significant changes. In summary, combined melatonin and FSS appears to have the potential to inhibit adipogenesis and treat obesity.