• 제목/요약/키워드: Caspase-1

검색결과 1,278건 처리시간 0.053초

Roles of ginsenosides in inflammasome activation

  • Yi, Young-Su
    • Journal of Ginseng Research
    • /
    • 제43권2호
    • /
    • pp.172-178
    • /
    • 2019
  • Inflammation is an innate immune response that protects the body from pathogens, toxins, and other dangers and is initiated by recognizing pathogen-associated molecular patterns or danger-associated molecular patterns by pattern-recognition receptors expressing on or in immune cells. Intracellular pattern-recognition receptors, including nucleotide-binding oligomerization domain-like receptors (NLRs), absent in melanoma 2, and cysteine aspartate-specific protease (caspase)-4/5/11 recognize various pathogen-associated molecular patterns and danger-associated molecular patterns and assemble protein complexes called "inflammasomes." These complexes induce inflammatory responses by activating a downstream effector, caspase-1, leading to gasdermin D-mediated pyroptosis and the secretion of proinflammatory cytokines, such as interleukin $(IL)-1{\beta}$ and IL-18. Ginsenosides are natural steroid glycosides and triterpene saponins found exclusively in the plant genus Panax. Various ginsenosides have been identified, and their abilities to regulate inflammatory responses have been evaluated. These studies have suggested a link between ginsenosides and inflammasome activation in inflammatory responses. Some types of ginsenosides, including Rh1, Rg3, Rb1, compound K, chikusetsu saponin IVa, Rg5, and Rg1, have been clearly demonstrated to inhibit inflammatory responses by suppressing the activation of various inflammasomes, including the NLRP3, NLRP1, and absent in melanoma 2 inflammasomes. Ginsenosides have also been shown to inhibit caspase-1 and to decrease the expression of $IL-1{\beta}$ and IL-18. Given this body of evidence, the functional relationship between ginsenosides and inflammasome activation provides new insight into the understanding of the molecular mechanisms of ginsenoside-mediated antiinflammatory actions. This relationship also has applications regarding the development of antiinflammatory remedies by ginsenoside-mediated targeting of inflammasomes, which could be used to prevent and treat inflammatory diseases.

Induction of caspase-dependent apoptosis in melanoma cells by the synthetic compound (E)-1-(3,4-dihydroxyphenethyl)-3-styrylurea

  • Kim, Ji-Hae;Jang, Young-Oh;Kim, Beom-Tae;Hwang, Ki-Jun;Lee, Jeong-Chae
    • BMB Reports
    • /
    • 제42권12호
    • /
    • pp.806-811
    • /
    • 2009
  • Recently, various phenolic acid phenethyl ureas (PAPUs) have been synthesized from phenolic acids by Curtius rearrangement for the development of more effective anti-oxidants. In this study, we examined the anti-tumor activity and cellular mechanism of the synthetic compound (E)-1-(3,4-dihydroxyphenethyl)-3-styrylurea (PAPU1) using melanoma B16/F10 and M-3 cells. Results showed that PAPU1 inhibited the cell proliferation and viability, but did not induce cytotoxic effects on primary cultured fibroblasts. PAPU1 induced apoptotic cell death rather than necrosis in melanoma cells, a result clearly proven by the shift of cells into sub-$G_1$ phase of the cell cycle and by the substantial increase in cells positively stained with TUNEL or Annexin V. Collectively, this study revealed that PAPU1 induced apoptosis in a caspase-dependent manner, suggesting a potential role as a cancer chemopreventive agent for melanoma cells.

Silibinin Inhibits Proliferation, Induces Apoptosis and Causes Cell Cycle Arrest in Human Gastric Cancer MGC803 Cells Via STAT3 Pathway Inhibition

  • Wang, Yi-Xin;Cai, Hong;Jiang, Gang;Zhou, Tian-Bao;Wu, Hai
    • Asian Pacific Journal of Cancer Prevention
    • /
    • 제15권16호
    • /
    • pp.6791-6798
    • /
    • 2014
  • Background: To investigate the effect of silibinin on proliferation and apoptosis in human gastric cancer cell line MGC803 and its possible mechanisms. Materials and Methods: Human gastric cancer cell line MGC803 cells were treated with various concentration of silibinin. Cellular viability was assessed by CCK-8 assay andapoptosis and cell cycle distribution by flow cytometry. Protein expression and mRNA of STAT3, and cell cycle and apoptosis regulated genes were detected by Western blotting and real-time polymerase chain reaction, respectively. Results: Silibinin inhibits growth of MGC803 cells in a dose- and time-dependent manner. Silibinin effectively induces apoptosis of MGC803 cells and arrests MGC803 cells in the G2/M phase of the cell cycle, while decreasing the protein expression of p-STAT3, and of STAT3 downstream target genes including Mcl-1, Bcl-xL, survivin at both protein and mRNA levels. In addition, silibinin caused an increase in caspase 3 and caspase 9 protein as well as mRNA levels. Silibinin caused G2/M phage arrest accompanied by a decrease in CDK1 and Cyclin B1 at protein and mRNA levels.. Conclusions: These results suggest that silibinin inhibits the proliferation of MGC803 cells, and it induces apoptosis and causes cell cycle arrest by down-regulating CDK1, cyclinB1, survivin, Bcl-xl, Mcl-1 and activating caspase 3 and caspase 9, potentially via the STAT3 pathway.

Ethanol Elicits Inhibitory Effect on the Growth and Proliferation of Tongue Carcinoma Cells by Inducing Cell Cycle Arrest

  • Le, Thanh-Do;Do, Thi Anh Thu;Yu, Ri-Na;Yoo, Hoon
    • The Korean Journal of Physiology and Pharmacology
    • /
    • 제16권3호
    • /
    • pp.153-158
    • /
    • 2012
  • Cellular effects of ethanol in YD-15 tongue carcinoma cells were assessed by MTT assay, caspase activity assay, Western blotting and flow cytometry. Ethanol inhibited the growth and proliferation of YD-15 cells in a dose- and time-dependent manner in an MTT assay. The effects of ethanol on cell cycle control at low percent range of ethanol concentration (0 to 1.5%), the condition not inducing YD-15 cell death, was investigated after exposing cells to alcohol for a certain period of time. Western blotting on the expression of cell cycle inhibitors showed that p21 and p27 was up-regulated as ethanol concentration increases from 0 to 1.5% whilst the cell cycle regulators, cdk1, cdk2, and cdk4 as well as Cyclin A, Cyclin B1 and Cyclin E1, were gradually down-regulated. Flow cytometric analysis of cell cycle distribution revealed that YD-15 cells exposed to 1.5% ethanol for 24 h was mainly arrested at G2/M phase. However, ethanol induced apoptosis in YD-15 cells exposed to 2.5% or higher percent of ethanol. The cleaved PARP, a marker of caspase-3 mediated apoptosis, and the activation of caspase-3 and -7 were detected by caspase activity assay or Western blotting. Our results suggest that ethanol elicits inhibitory effect on the growth and proliferation of YD-15 tongue carcinoma cells by mediating cell cycle arrest at G2/M at low concentration range and ultimately induces apoptosis under the condition of high concentration.

AGS 인체 위암세포에서 황흑산에 의한 ROS 생성 및 caspase 활성 의존적 apoptosis 유발 (Induction of Apoptosis by Hwangheuk-san in AGS Human Gastric Carcinoma Cells through the Generation of Reactive Oxygen Species and Activation of Caspases)

  • 홍수현;박철;김경민;최영현
    • 생명과학회지
    • /
    • 제25권11호
    • /
    • pp.1235-1243
    • /
    • 2015
  • 황흑산은 동의보감의 복강과 장옹의 처방을 위해 기록된 처방전으로 오랫동안 사용되어 왔으나, 항암 효능에 대한 구체적인 연구는 전혀 이루어진 바 없다. 본 연구에서는 AGS 인체 위암세포를 대상으로 황흑산 처리에 의한 증식억제와 연관된 apoptosis 유발 및 관련 기전 연구를 수행하였다. AGS 위암세포에 황흑산 추출물을 처리함에 처리 농도 의존적으로 증식이 억제되었으며, 이는 apoptosis 유발과 연관성이 있음을 핵의 형태적 변형과 sub-G1기 세포의 축적 등으로 확인하였다. 황흑산 추출물에 의한 apoptosis 유도에는 pro-apoptotic Bax 단백질의 발현 증가와 anti-apoptotic Bcl-2의 발현 감소 및 미토콘드리아에서 세포질로의 cytochrome c 유리와 연관성이 있었으며, 세포 내 활성산소종(reactive oxygen species, ROS)의 축적을 증가시켰다. 또한 황흑산 추출물에 의한 apoptosis 유발은 caspases (caspase-3, -8 및 -9)의 활성을 증가시켰으며, poly (ADP-ribose)-polymerase 단백질의 단편화를 초래하였다. 그러나 ROS scavenger 및 pan-caspases inhibitor는 황흑산 추출물에 의한 apoptosis의 유발을 거의 완벽하게 억제하였으며, 암세포의 증식억제도 차단하였다. 이상의 결과는 황흑산 추출물에 의한 apoptosis가 ROS 생성 및 caspase 활성 의존적으로 일어남을 의미하는 것으로 황흑산의 항암기전 해석을 이해하고 향후 지속적인 연구를 위한 유용한 자료로 사용될 것이다.

Evidence for the Association of Ce11u1ar Iron Loss in Nitric Oxide-induced Apoptosis of HL-60 Cells: Involvement of p38 Kinase, c-Jun N-terminal Kinase, Cytochrome C Release, and Caspases Pathways

  • Choi, Suck-Chei;Kim, Beom-Su;Yoon, Kwon-Ha;Song, Moon-Young;Oh, Hyun-Mee;Han, Weon-Cheol;Kim, Tae-Hyeon;Kim, Eun-Cheol;Jun, Chang Duk
    • Animal cells and systems
    • /
    • 제6권2호
    • /
    • pp.171-180
    • /
    • 2002
  • Nitric oxide has high affinity for iron, and thus it can cause intracellular iron loss. We tested the idea that intracellular iron can be the primary target of NO toxicity by comparing the signaling mechanisms involved in cell death caused by iron depletion and that caused by NO. Treatment of HL-60 cells with a NO donor, S-nitroso-N-acetyl-DL-penicillamine (SNAP), decreased the intracellular iron level rapidly as that observed with the iron chelator deferoxamine (DFO). Iron chelators such as DFO and mimosine could induce death of human leukemic HL-60 cells by a mechanism requiring activation of p38 kinase, c-Jun N-terminal kinase, caspase-3 and caspase-8. DFO and SNAP also caused release of cytochrome c from mitochondria. Inhibition of p38 kinase by a selective inhibitor, SB203580, abolished the NO and DFO-induced cell death, release of cytochrome c, and activation of caspase-3 and caspase-8, thus indicating that p38 kinase lies upstream in the cell death processes. In a parallel situation, the cells that are sensitive to NO showed similar sensitivity to DFO. Moreover, simultaneous addition of ferric citrate, an iron-containing compound, inhibited the SNAP and DFO-induced activation of caspases and also blocked the NO-mediated cell cycle arrest at $G_1$ phase. Collectively, our data implicate that the NO-induced cell death of tumor cells including HL-60 cells is mediated by depletion of iron and further suggest that activation of p38 kinase lies upstream of cytochrome c release and caspase activation involved in this apoptotic process.

NCI-H157 폐암 세포주에서 Caspase Cascade 활성을 통한 Arsenic Trioxide와 Sulindac 병합요법의 세포고사효과 (Inducing Apoptosis of NCI-H157 Human Lung Carcinoma Cells via Activation of Caspase Cascade by Combination Treatment with Arsenic Trioxide and Sulindac)

  • 김학렬;양세훈;정은택
    • Tuberculosis and Respiratory Diseases
    • /
    • 제56권4호
    • /
    • pp.381-392
    • /
    • 2004
  • 연구배경 : Arsenic trioxide($As_2O_3$)은 재발성 또는 불응성 급성전골수성백혈병의 치료제로 쓰이는 항암제로서 비소세포폐암을 포함한 다른 암세포주에도 효과가 있는 것으로 되어있다. NSAIDs는 항암 예방약제로 사용되고 있고, 세포고사를 통해 다른 항암제나 방사선치료의 반응성을 강화시키는 것으로 알려져 있다. 저자들은 NCI-H157 세포주에서 $As_2O_3$와 sulindac의 병합치료가 그것들의 세포고사를 배가시키는지 여부를 알아보고자 하였다. 방 법 : 세포 독성은 MTT 방법으로 측정하였고, 세포고사를 알아보기 위해 핵산 염색과 유식세포 분석을 시행하였다. 세포고사의 기전을 보기 위해 caspasefamily의 활성을 보았고, PARP와 ICAD의 분절을 western blotting으로 확인하였다. 또한 Fas와 Fas-L의 발현유무를 western blotting을 통해 관찰하였다. 결 과 : NCI-H157 폐암세포에 $As_2O_3$와 sulindac을 병합치료시 단독치료군에 비해 생존율이 의미 있게 감소하였고, 이러한 세포사는 핵산염색을 통한 염색사의 응축과 핵 분절 유도와 유식세포 분석에 의한 $sub-G_0/G_1$ DNA분획의 증가현상을 통해 세포고사에 의해 매개됨을 알 수 있었다. 세포고사의 유도에는 caspase 3, 8, 9를 통한 활성화와 이에 의한 PARP와 ICAD의 절단을 확인하였다. 또한 caspase-8 protease의 활성화에는 Fas와 Fas/L 단백질의 발현증가가 유도되었음을 알 수 있었다. 결 론 :NCI-H157 폐암세포주에 $As_2O_3$와 sulindac의 병합요법은 Fas/FasL 신호전달계의 활성화와 caspase 단백질 활성화 의해 세포고사가 유도되었다.

Up-Regulation of $p27^{Kip1}$ Protects hES Cells from Differentiation-Associated and Caspase 3-Dependent Apoptosis

  • Park, So-Hyun;Kim, Min Kyoung;Lee, Chul-Hoon
    • Journal of Microbiology and Biotechnology
    • /
    • 제22권12호
    • /
    • pp.1790-1794
    • /
    • 2012
  • Recently, it has been suggested that $p27^{Kip1}$, the cell cycle regulatory protein, plays a pivotal role in the progression of normal differentiation in murine embryonic stem (mES) cells. In the current study, we investigated the role of $p27^{Kip1}$ in the regulation of differentiation and apoptotic induction using Western blotting, quantitative real-time RT-PCR, and small interfering RNA (siRNA) assays and confocal laser scanning microscopic analysis of H9 human ES (hES) cells and H9-derived embryoid bodies (EBs) grown for 10 ($EB_{10}$) and 20 days ($EB_{20}$). Our results demonstrate that the proteins $p27^{Kip1}$ and cyclin D3 are strongly associated with cellular differentiation, and, for the first time, show that up-regulation of $p27^{Kip1}$ protects hES cells from inducing differentiation-associated and caspase 3-dependent apoptosis.