• 제목/요약/키워드: Caspase 1

검색결과 1,285건 처리시간 0.033초

Curcumin과 resveratrol에 의한 두경부암 유래의 HN-4 세포의 세포주기, 세포사 및 전이관련 단백질의 발현 조절 (EFFECT OF CURCUMIN AND RESVERATROL ON THE CELL CYCLE REGULATION, APOPTOSIS AND INHIBITION OF METASTASIS RELATED PROTEINS IN HN-4 CELLS)

  • 김사엽;이상한;권택규
    • Journal of the Korean Association of Oral and Maxillofacial Surgeons
    • /
    • 제29권5호
    • /
    • pp.272-281
    • /
    • 2003
  • 본 연구에서는 두경부암 HN-4 세포에서 cancer chemopreventive agent인 curcumin과 resveratrol를 처리하여 HN-4 세포의 생육 억제의 원인이 apoptotic cell death에 의하여 일어나며 세포 주기 조절 단백질의 발현 및 암 전이에 관련된 MMP 활성 저해 기전에 대하여 이해하고자 하였다. HN-4 세포에 다양한 농도(10-100 ${\mu}M$) curcumin을 처리하여 세포주기 조절 단백질의 발현을 측정한 결과 cdk1과 cdk4 단백질이 농도 의존적으로 발현이 감소하였으며, resveratrol 처리에서는 cyclin A 단백질의 특이적인 감소 현상을 확인하였다. Curcumin에 의한 apoptosis 유도 기전을 조사한 결과 anti-apoptotic 기능이 있는 Bcl-2 및 Bcl-xL 단백질 발현 감소 현상은 없었으나, caspase 저해 IAP family 단백질중 cIAP1과 survivin 단백질 발현 현상이 처리 농도 의존적으로 감소하였다. Resveratrol을 처리한 경우 Bcl-2 및 survivin 단백질 발현 감소현상을 확인하였다. Curcumin과 resveratrol에 의한 apoptosis 과정은 caspase-3 의존적인 apoptosis 유도 기전을 보였다. Curcumin에 의한 apoptosis 과정은 항산화제인 NAC 처리에 의해서 저해되었다. Curcumin과 NAC 동시 처리는 cytochrome c 유리, caspase-3 활성화 및 Bax 단백질 분절 현상을 억제하였다. 그러나 resveratrol에 의한 apoptosis 과정은 NAC 처리에 의하여 억제되지 않았다. Curcumin과 resveratrol에 의한 암 전이 관련 단백질인 MMP2와 9의 활성 저해효과도 확인하였다. 결론적으로, curcumin에 의한 항암효과는 세포주기 조절 및 apoptosis 유도 및 전이 관련 단백질의 활성 억제를 통하여 야기되는 것으로 생각되어 진다.

폐암 세포에서 Proteasome Inhibitor에 의한 Apoptosis의 기전 (The Mechanism of Proteasome Inhibitor-Induced Apoptosis in Lung Cancer Cells)

  • 김철현;이경희;이춘택;김영환;한성구;심영수;유철규
    • Tuberculosis and Respiratory Diseases
    • /
    • 제54권4호
    • /
    • pp.403-414
    • /
    • 2003
  • 연구배경 : Proteasome 억제가 세포의 apoptosis에 미치는 영향은 세포 종류에 따라 차이를 보이고 있다. 그러나 아직까지 폐암 세포에서 proteasome 억제가 미치는 효과 및 그 기전에 관해서는 확실하게 규명되어 있지 못한 상태이다. 본 연구에서는 폐암 세포주에서 proteasome 억제에 의한 apoptosis의 유도 유무를 관찰하고, 그 기전을 규명하고자 하였다. 방 법 : 폐암 세포주인 A549와 NCI-H157 세포에 proteasome 억제제인 MG132와 PS-341을 투여하고 세포의 생존율을 MTT 분석으로 평가하였고, apoptosis 유무를 PARP 단백에 대한 Western 분석으로 확인하였다. Proteasome 억제가 caspase 3와 JNK의 활성화에 미치는 효과를 각각 Western 분석과 immunocomplex kinase 분석으로 평가하였다. Proteasome 억제제를 투여하고 항 apoptosis 경로인 ERK와 cIAP1에 미치는 효과를 Western 분석으로 평가하였다. 결 과 : A549와 NCI-H157 세포에 MG132를 투여하였을 때, 세포생존율의 감소가 관찰되었고, 이는 apoptosis의 유도에 의한 것으로 확인되었다. MG132와 PS-341 처치로 caspase 3와 JNK가 활성화되었다. 반면에 활성화된 ERK의 발현과 cIAP1의 발현은 감소하였다. 결 론 : 폐암 세포에서 proteasome 억제에 의한 apoptosis에는 caspase 3, JNK와 같은 apoptosis 유도 경로의 활성화와 함께 항 apoptosis 경로의 억제가 관여할 것으로 생각된다.

A Combination of PG490 and Lipopolysaccharide Induce Apoptosis through Activation of Casapase-3 and Downregulation of cIAP1 and XIAP in Human Astroglioma Cell

  • Lee, Tae-Jin;Woo, Kyung-Jin;Park, Jong-Wook;Kwon, Taeg-Kyu
    • IMMUNE NETWORK
    • /
    • 제5권2호
    • /
    • pp.99-104
    • /
    • 2005
  • Background: Malignant gliomas are the most common primary tumors in the central nervous system. Methods: We investigated the combined effect of PG490 and LPS on the induction of the apoptotic pathway in human astroglioma cells. Results: Treatment of U87 cells with combination of 50nM of PG490 and $50{\mu}g/ml$ of LPS resulted in increased internucleosomal DNA fragmentation, cleavage of PLC-${\gamma}1$, and downregulation of cIAP1 and XIAP. The combination of LPS and PG490 treatment-induced apoptosis is mediated through the activation of caspase, which is inhibited by the caspase inhibitor, z-VAD-fmk. Also, release of cytochrome c was found in PG490 and LPS-cotreated U87 cell. Conclusion: Taken together, combination of PG490 and LPS appears to be a potent inducer of apoptosis in astrogliaoma cells, and might have some benefit in the treatment of glioma patients.

Glycyrrhizin Attenuates MPTP Neurotoxicity in Mouse and $MPP^+$-Induced Cell Death in PC12 Cells

  • Kim, Yun-Jeong;Lee, Chung-Soo
    • The Korean Journal of Physiology and Pharmacology
    • /
    • 제12권2호
    • /
    • pp.65-71
    • /
    • 2008
  • The present study examined the inhibitory effect of licorice compounds glycyrrhizin and a metabolite $18{\beta}$-glycyrrhetinic acid on the neurotoxicity of 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) in the mouse and on the 1-methyl-4-phenylpyridinium ($MPP^+$)-induced cell death in differentiated PC12 cells. MPTP treatment increased the activities of total superoxide dismutase, catalase and glutathione peroxidase and the levels of malondialdehyde and carbonyls in the brain compared to control mouse brain. Co-administration of glycyrrhizin (16.8 mg/kg) attenuated the MPTP effect on the enzyme activities and formation of tissue peroxidation products. In vitro assay, licorice compounds attenuated the $MPP^+$-induced cell death and caspase-3 activation in PC12 cells. Glycyrrhizin up to $100{\mu}M$ significantly attenuated the toxicity of $MPP^+$. Meanwhile, $18{\beta}$-glycyrrhetinic acid showed a maximum inhibitory effect at $10{\mu}M$; beyond this concentration the inhibitory effect declined. Glycyrrhizin and $18{\beta}$-glycyrrhetinic acid attenuated the hydrogen peroxide- or nitrogen species-induced cell death. Results from this study indicate that glycyrrhizin may attenuate brain tissue damage in mice treated with MPTP through inhibitory effect on oxidative tissue damage. Glycyrrhizin and $18{\beta}$-glycyrrhetinic acid may reduce the $MPP^+$ toxicity in PC12 cells by suppressing caspase-3 activation. The effect seems to be ascribed to the antioxidant effect.

신경교(神經膠) 성상세포(星狀細胞)에서 쥬니퍼오일에 의한 세포자멸사(細胞自滅死)의 억제(抑制) 효과(效果) (Inhibitory Effect of Apoptosis of Human Astrocytes by Juniper Oil)

  • 김태형;김태헌;이성률;류영수
    • 동의신경정신과학회지
    • /
    • 제11권2호
    • /
    • pp.1-9
    • /
    • 2000
  • In previous studies, heat shock has been reported to induce the apoptosis or programmed cell death through the activation of caspase-3. 1 investigated the effect of juniper pure essential oil on the heat shock-induced apoptosis in human astrocyte cell line CCF-STTGI. Treatment of the astrocytes with heat shock markedly induced apoptotic cell death. However, pretreatment of the astrocytes with juniper oil ingibited the heat shock-induced apoptosis. To determine whether juniper inhibits the heat shock-induced activation of these apoptotic proteases, activation of CPP32 was assessed by Western blotting. Consistent with flow cytometry. DNA fragmentation and giemsa staining, heat shock-induced activation of CPP32 was blocked by juniper oil. Poly(ADP-ribose) polymerase (PARP), cysteine protease substrates were fragmented as a consequence of apoptosis by heat shock. Juniper oil inhibited the PARP fragmentation. This juniper oil also inhibited the heat shock-induced activation of caspase-3. These results suggest that juniper oil may modulate the apoptosis through the activation of the interleukin-1-converting enzyme-like protease.

  • PDF

육울탕(六鬱湯)에 의한 인체자궁경부암세포의 증식억제에 관한 연구 (Induction of Apoptosis by Yukwool-tang in Human Cervical Carcinoma HeLa Cells)

  • 최영현;최병태;이용태
    • 동의생리병리학회지
    • /
    • 제21권6호
    • /
    • pp.1513-1519
    • /
    • 2007
  • Yukwool-tang (YWT) is a traditional Chinese medicine, which has been used for patients suffering from a uterine disease in Oriental medicine. In the present study, it was examined the biochemical mechanisms of apoptosis by YWT in human cervical carcinoma HeLa cells. It was found that YWT could inhibit the cell growth of HeLa cells in a dose-dependent manner, which was associated with apoptotic cell death such as formation of apoptotic bodies and DNA fragmentation. Flow cytometry analysis confirmed that YWT treatment increased populations of apoptotic-sub-G1 phase of the cell cycle. We observed the p53-independent induction of p21 proteins, down-regulation of anti apoptotic Bcl-2 expression and proteolytic activation of caspase-3 in YWT-treated HeLa cells. YWT treatment also concomitant degradation and/or inhibition of poly (ADP-ribose) polymerase (PARP), phospholipase C-1 ($PLC{\gamma}1$), ${\beta}-catenin$ and DNA fragmentation factor 45/inhibitor of caspase-activated DNase (DFF45/ICAD). Taken together, these findings partially provide novel insights into the possible molecular mechanism of the anti-cancer activity of YWT.

Tannic acid-induced apoptosis in FaDu hypopharyngeal squamous cell carcinoma

  • Ta, Loan Thi;Nguyen, Trang Thi Kieu;Yoo, Hoon
    • International Journal of Oral Biology
    • /
    • 제44권2호
    • /
    • pp.43-49
    • /
    • 2019
  • Tannic acid (TA) is a water-soluble polyphenol compound found in various herbal plants. We investigated the chemopreventive effects of TA on FaDu hypopharyngeal squamous carcinoma cells. In an 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay, TA showed dose-dependent cytotoxicity with a half maximal inhibitory concentration (IC50) of 50 ?M. Cell cycle analysis and immunofluorescence imaging demonstrated that under low-dose ($25{\mu}M$) treatment, FaDu cells were arrested in G2/M phase, and as the dose of TA was increased, apoptosis was induced with the increase of cell population at sub-G1 phase. The expressions of various cyclins, including cyclin D1 and cyclin-dependent kinases (CDK-1 and CDK-2), were down-regulated at low doses of TA, whereas apoptotic effectors such as cleaved caspase 3, cleaved caspase 7, and poly (ADP-ribose) polymerase (PARP) were expressed in a dose-dependent manner in Western blotting. In addition, TA-induced apoptosis of FaDu cells might be mediated by the extracellular signal-regulated kinase (ERK)/mitogen-activated protein kinase pathway, with the upregulation of p-AKT/p-PKB (phosphorylated protein kinase B) and p-ERK. Overall, our data support the hypothesis that TA is a potential candidate agent for the treatment of hypopharyngeal cancer.

Decursin induces apoptosis in glioblastoma cells, but not in glial cells via a mitochondria-related caspase pathway

  • Oh, Seung Tack;Lee, Seongmi;Hua, Cai;Koo, Byung-Soo;Pak, Sok Cheon;Kim, Dong-Il;Jeon, Songhee;Shin, Boo Ahn
    • The Korean Journal of Physiology and Pharmacology
    • /
    • 제23권1호
    • /
    • pp.29-35
    • /
    • 2019
  • Decursin is a major biological active component of Angelica gigas Nakai and is known to induce apoptosis of metastatic prostatic cancer cells. Recently, other reports have been commissioned to examine the anticancer activities of this plant. In this study, we evaluated the inhibitory activity and related mechanism of action of decursin against glioblastoma cell line. Decursin demonstrated cytotoxic effects on U87 and C6 glioma cells in a dose-dependent manner but not in primary glial cells. Additionally, decursin increased apoptotic bodies and phosphorylated JNK and p38 in U87 cells. Decursin also down-regulated Bcl-2 as well as cell cycle dependent proteins, CDK-4 and cyclin D1. Furthermore, decursin-induced apoptosis was dependent on the caspase activation in U87 cells. Taken together, our data provide the evidence that decursin induces apoptosis in glioblastoma cells, making it a potential candidate as a chemotherapeutic drug against brain tumor.

Crataegus pinnatifida Bunge root extract induces apoptosis of murine lung carcinoma cells in vitro

  • Minjeong Kwon;Jongbeom Chae;Ju-Ock Nam
    • Journal of Applied Biological Chemistry
    • /
    • 제66권
    • /
    • pp.299-304
    • /
    • 2023
  • This study sought to evaluate the anticancer effects of Crataegus pinnatifida Bunge root extract (CPE) on murine Lewis lung carcinoma cells (LLC1) in vitro. CPE treatment (2.5, 5, 10 ㎍/mL, 24 h) of LLC cells led to a dose-dependent decrease in cell viability, while CPE treatment did not have a cytotoxic effect on non-cancer cells (NIH/3T3). CPE affects LLC by flipping the plasma membrane and making the membrane more permeable; by flow cytometry, CPE-induced annexin V and propidium iodide positivity, indicating induction of apoptosis in LLC cells. In addition, CPE enhanced the expression of apoptotic proteins caspase-3 and poly (ADP-ribose) polymerase 1 (PARP-1). CPE upregulated the proapoptotic protein BCL-2-associated X while downregulating the anti-apoptotic protein B-cell lymphoma 2 (BCL-2), suggesting that CPE induces apoptosis via the mitochondrial pathway. Furthermore, CPE upregulated the phosphorylation of the mitogen activated protein kinase p38. In conclusion, the results suggest that CPE has an anticancer effect in LLC cells by inducing apoptosis via p38.

Cigarette Smoke Extract-Treated Mouse Airway Epithelial Cells-Derived Exosomal LncRNA MEG3 Promotes M1 Macrophage Polarization and Pyroptosis in Chronic Obstructive Pulmonary Disease by Upregulating TREM-1 via m6A Methylation

  • Lijing Wang;Qiao Yu;Jian Xiao;Qiong Chen;Min Fang;Hongjun Zhao
    • IMMUNE NETWORK
    • /
    • 제24권2호
    • /
    • pp.3.1-3.23
    • /
    • 2024
  • Cigarette smoke extract (CSE)-treated mouse airway epithelial cells (MAECs)-derived exosomes accelerate the progression of chronic obstructive pulmonary disease (COPD) by upregulating triggering receptor expressed on myeloid cells 1 (TREM-1); however, the specific mechanism remains unclear. We aimed to explore the potential mechanisms of CSE-treated MAECs-derived exosomes on M1 macrophage polarization and pyroptosis in COPD. In vitro, exosomes were extracted from CSE-treated MAECs, followed by co-culture with macrophages. In vivo, mice exposed to cigarette smoke (CS) to induce COPD, followed by injection or/and intranasal instillation with oe-TREM-1 lentivirus. Lung function and pathological changes were evaluated. CD68+ cell number and the levels of iNOS, TNF-α, IL-1β (M1 macrophage marker), and pyroptosis-related proteins (NOD-like receptor family pyrin domain containing 3, apoptosis-associated speck-like protein containing a caspase-1 recruitment domain, caspase-1, cleaved-caspase-1, gasdermin D [GSDMD], and GSDMD-N) were examined. The expression of maternally expressed gene 3 (MEG3), spleen focus forming virus proviral integration oncogene (SPI1), methyltransferase 3 (METTL3), and TREM-1 was detected and the binding relationships among them were verified. MEG3 increased N6-methyladenosine methylation of TREM-1 by recruiting SPI1 to activate METTL3. Overexpression of TREM-1 or METTL3 negated the alleviative effects of MEG3 inhibition on M1 polarization and pyroptosis. In mice exposed to CS, EXO-CSE further aggravated lung injury, M1 polarization, and pyroptosis, which were reversed by MEG3 inhibition. TREM-1 overexpression negated the palliative effects of MEG3 inhibition on COPD mouse lung injury. Collectively, CSE-treated MAECs-derived exosomal long non-coding RNA MEG3 may expedite M1 macrophage polarization and pyroptosis in COPD via the SPI1/METTL3/TREM-1 axis.