• 제목/요약/키워드: Tumor necrosis factor-related apoptosis-inducing ligand

검색결과 42건 처리시간 0.025초

Vanillin oxime inhibits lung cancer cell proliferation and activates apoptosis through JNK/ERK-CHOP pathway

  • Shen, Jie;Su, Zhixiang
    • The Korean Journal of Physiology and Pharmacology
    • /
    • 제25권4호
    • /
    • pp.273-280
    • /
    • 2021
  • Lung cancer despite advancement in the medical field continues to be a major threat to human lives and accounts for a high proportion of fatalities caused by cancers globally. The current study investigated vanillin oxime, a derivative of vanillin, against lung cancer cells for development of treatment and explored the mechanism. Cell viability changes by vanillin oxime were measured using MTT assay. Vanillin oxime-mediated apoptosis was detected in A549 and NCI-H2170 cells at 48 h of exposure by flow cytometry. The CEBP homologous protein (CHOP) and death receptor 5 (DR5) levels were analysed by RT-PCR and protein levels by Western blotting. Vanillin oxime in concentration-dependent way suppressed A549 and NCI-H2170 cell viabilities. On exposure to 12.5 and 15 μM concentrations of vanillin oxime elevated Bax, caspase-3, and -9 levels in A549 and NCI-H2170 cells were observed. Vanillin oxime exposure suppressed levels of Bcl-2, survivin, Bcl-xL, cFLIP, and IAPs proteins in A549 and NCI-H2170 cells. It stimulated significant elevation in DR4 and DR5 levels in A549 and NCI-H2170 cells. In A549 and NCI-H2170 cells vanillin oxime exposure caused significant (p < 0.05) enhancement in CHOP and DR5 mRNA expression. Vanillin oxime exposure of A549 and NCI-H2170 cells led to significant (p < 0.05) enhancement in levels of phosphorylated extracellular-signal-regulated kinase and c-Jun N-terminal kinase. Thus, vanillin oxime inhibits pulmonary cell proliferation via induction of apoptosis through tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) mediated pathway. Therefore, vanillin oxime may be studied further to develop a treatment for lung cancer.

Microfluidic System Based High Throughput Drug Screening System for Curcumin/TRAIL Combinational Chemotherapy in Human Prostate Cancer PC3 Cells

  • An, Dami;Kim, Kwangmi;Kim, Jeongyun
    • Biomolecules & Therapeutics
    • /
    • 제22권4호
    • /
    • pp.355-362
    • /
    • 2014
  • We have developed a fully automated high throughput drug screening (HTDS) system based on the microfluidic cell culture array to perform combinational chemotherapy. This system has 64 individually addressable cell culture chambers where the sequential combinatorial concentrations of two different drugs can be generated by two microfluidic diffusive mixers. Each diffusive mixer has two integrated micropumps connected to the media and the drug reservoirs respectively for generating the desired combination without the need for any extra equipment to perfuse the solution such as syringe pumps. The cell array is periodically exposed to the drug combination with the programmed LabVIEW system during a couple of days without extra handling after seeding the cells into the microfluidic device and also, this device does not require the continuous generation of solutions compared to the previous systems. Therefore, the total amount of drug being consumed per experiment is less than a few hundred micro liters in each reservoir. The utility of this system is demonstrated through investigating the viability of the prostate cancer PC3 cell line with the combinational treatments of curcumin and tumor necrosis factor-alpha related apoptosis inducing ligand (TRAIL). Our results suggest that the system can be used for screening and optimizing drug combination with a small amount of reagent for combinatorial chemotherapy against cancer cells.

Anti-inflammatory Activity of Sambucus Plant Bioactive Compounds against TNF-α and TRAIL as Solution to Overcome Inflammation Associated Diseases: The Insight from Bioinformatics Study

  • Putra, Wira Eka;Salma, Wa Ode;Rifa'i, Muhaimin
    • Natural Product Sciences
    • /
    • 제25권3호
    • /
    • pp.215-221
    • /
    • 2019
  • Inflammation is the crucial biological process of immune system which acts as body's defense and protective response against the injuries or infection. However, the systemic inflammation devotes the adverse effects such as multiple inflammation associated diseases. One of the best ways to treat this entity is by blocking the tumor necrosis factor alpha ($TNF-{\alpha}$) and TNF-related apoptosis-inducing ligand (TRAIL) to avoid the proinflammation cytokines production. Thus, this study aims to evaluate the potency of Sambucus bioactive compounds as anti-inflammation through in silico approach. In order to assess that, molecular docking was performed to evaluate the interaction properties between the $TNF-{\alpha}$ or TRAIL with the ligands. The 2D structure of ligands were retrieved online via PubChem and the 3D protein modeling was done by using SWISS Model. The prediction results of the study showed that caffeic acid (-6.4 kcal/mol) and homovanillic acid (-6.6 kcal/mol) have the greatest binding affinity against the $TNF-{\alpha}$ and TRAIL respectively. This evidence suggests that caffeic acid and homovanillic acid may potent as anti-inflammatory agent against the inflammation associated diseases. Finally, this study needs further examination and evaluation to validate the potency of Sambucus bioactive compounds.

Genistein과 TRAIL의 복합처리에 의한 U937 인체 혈구암 세포의 Apoptosis 유도 (Induction of Apoptosis by Combined-treatment with Genistein and TRAIL in U937 Human Leukemia Cells)

  • 최영현;한민호
    • 한국식품영양과학회지
    • /
    • 제40권9호
    • /
    • pp.1201-1207
    • /
    • 2011
  • TRAIL은 최근 암세포의 apoptosis 유도를 위한 효율적인 도구로 제시되었으나 많은 암세포들이 TRAIL 저항성을 획득한 것으로 알려져 TRAIL 저항성 극복을 위한 새로운 방법론의 제시가 요구되어지고 있다. Genistein은 대두의 대표적인 생리활성 물질인 isoflavonoid의 일종으로 많은 암세포에서 G2/M arrest를 유발하면서 apoptosis를 유도하는 것으로 알려져 있다. 본 연구에서는 U937 인체백혈병세포를 대상으로 genistein에 의한 TRAIL 유도 apoptosis의 감수성 증대 여부를 조사하였다. 본 연구의 결과에 의하면 U937 세포에서 세포독성이 없는 범위의 genistein 처리는 TRAIL에 의한 apoptosis 유도를 매우 증진시켰으며, 이는 tBid의 발현 증가와 cFLIPL의 발현 감소와 연계된 caspase의 활성 증가와 연관성이 있었다. 또한 caspase의 활성 저해제는 genistein과 TRAIL의 복합처리에 의한 apoptosis를 유의적으로 감소시켜 복합 처리에 의한 apoptosis의 유도에 caspase의 활성 증대가 필수적임을 알 수 있었다. 따라서 genistein은 TRAIL 저항성을 획득한 암세포의 효율적인 combined therapy approach를 위해 유용하게 사용될 수 있음을 알 수 있었다.

TRAIL Suppresses Human Breast Cancer Cell Migration via MADD/CXCR7

  • Wang, Rui;Li, Jin-Cheng
    • Asian Pacific Journal of Cancer Prevention
    • /
    • 제16권7호
    • /
    • pp.2751-2756
    • /
    • 2015
  • Background: Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) can specifically induce apoptosis limited to various cancer cells, so this reagent is considered a promising medicine for cancer therapy. TRAIL also exerts effects on non-apoptotic signals, relevant to processes such as metastasis, autophagy and proliferation in cancer cells. However, the mechanisms of TRAIL-regulated non-apoptotic signals are unclear. The purpose of this study was to investigate MADD/CXCR7 effects in TRAIL-mediated breast cancer cell migration. Materials and Methods: The ability of MADD/CXCR7 to regulate MVP signaling in TRAIL-mediated breast cancer cells migration was evaluated by transwell migration assay, quantitative RT-PCR, Western blotting and knock down experiments. Results: In this study, we found that treatment with TRAIL resulted in induced expression levels of MADD and CXCR7 in breast cancer cells. Knock down of MADD followed by treatment with TRAIL resulted in increased cell migration compared to either treatment alone. Similarly, through overexpression and knockdown experiments, we demonstrated that CXCR7 also positively regulated TRAIL-inhibited migration. Surprisingly, knock down of MADD lead to inhibition of TRAIL-induced CXCR7 mRNA and protein expression and overexpression of CXCR7 lead to the reduction of MADD expression, indicating that MADD is an upstream regulatory factor of TRAIL-triggered CXCR7 production and a negative feedback mechanism between MADD and CXCR7. Furthermore, we showed that CXCR7 is involved in MADD-inhibited migration in breast cancer cells. Conclusions: Our work defined a novel signaling pathway implicated in the control of breast cancer migration.

동충하초 유래 cordycepin에 의한 AGS 인체 위암세포의 apoptosis 유발 (Induction of Apoptotic Cell Death by Cordycepin, an Active Component of the Fungus Cordyceps militaris, in AGS Human Gastric Cancer Cells)

  • 이혜현;정진우;최영현
    • 생명과학회지
    • /
    • 제26권7호
    • /
    • pp.847-854
    • /
    • 2016
  • Nucleoside adenosine 유도체의 하나인 cordycepin (3′-deoxyadenosine)은 Cordyceps 속에서 유래된 활성 물질 중의 하나로서 항염증, 항산화 및 항암활성을 포함한 다양한 약리학적 효능이 있는 것으로 잘 알려져 있다. 본 연구에서는 AGS 인체 위암세포의 증식에 미치는 cordycepin의 영향과 관련 기전 연구를 시도하였다. Cordycepin의 처리에 따라 AGS 세포의 생존율이 처리 농도 의존적으로 감소되었으며, DNA 단편화 및 flow cytometery 분석에 따른 apoptosis 유발 또한 유의적으로 증가하였음을 확인하였다. 이러한 cordycepin 처리에 따른 AGS 세포의 apoptosis 유도에는 TRAIL, DR5 및 FasL의 mRNA 및 단백질의 발현 증가가 연관되어 있었다. 아울러 cordycepin은 Bcl-2 family 중 pro-apoptotic 인자인 Bax의 발현은 증가시켰으며, anti-apoptotic 인자인 Bcl-2 및 Bcl-xL의 발현은 전사 및 번역 수준에서 억제시켰다. 이러한 현상들은 extrinsic 및 intrinsic apoptosis의 initiator caspase (caspase-8 및 -9) 뿐만 아니라 effector caspase인 caspase-3의 활성과 PARP 단백질의 절단 증가와 연관성이 있었다. 따라서 AGS 세포에서 cordycepin에 의한 apoptosis의 유발은 death receptor 활성과 mitochondria 기능 손상을 포함한 multiple apoptotic pathway가 관여할 것으로 생각된다. 비록 좀 더 세심한 기전 연구의 결과가 뒤따라야 되겠지만, 본 연구의 결과는 cordycepin의 항암작용을 이해하는데 중요한 자료가 될 것이며 향후 수행될 추가 실험을 위한 기초 자료로서 그 가치가 매우 높을 것으로 생각된다.

Withaferin A의 다양한 항암 효과 및 분자생화학적 기전 (Anti-cancer Effects and Molecular Mechanisms of Withaferin A)

  • 우선민;민경진;권택규
    • 생명과학회지
    • /
    • 제23권3호
    • /
    • pp.462-469
    • /
    • 2013
  • Withaferin A는 Withania somnifera에서 추출한 천연물질로 스테로이드성 락톤(steroidal lactone)으로 항암, 항염증, 면역억제기능을 가진다. 본 연구에서는 withaferin A의 다양한 기능 중 항암효과에 대하여 논하고자 한다. Withaferin A는 암세포에서 세포의 분열, 전이, 침투 및 혈관생성을 억제함으로써 항암작용을 나타내는 것으로 알려져 있다. 또한, 기존에 사용되고 있던 항암요법인 방사선 용법과 저농도의 항암제와 withaferin A를 함께 병합 처리하면 암세포의 세포사멸을 현저하게 증가시키는 약물 민감화 작용을 한다. 이러한 withaferin A에 의한 항암작용에는 다양한 신호전달체계가 수반된다. 우선, withaferin A는 세포 내 활성산소의 양을 증가시키고, ER stress와 미토콘드리아 매개의 세포사멸을 유도한다. 둘째로, withaferin A는 세포의 성장과 분열, 전이에 중요한 Jak/STAT, Akt, Notch, 그리고 c-Met의 신호전달을 억제한다. 셋째, withaferin A는 prostate apoptosis protein-4의 발현을 증가시켜 세포사멸을 유도하거나 세포의 이동을 억제한다. 마지막으로, withaferin A는 proteasome의 활성을 억제하여 세포사멸 유도단백질의 발현을 증가시킴으로써 암세포사멸을 증가시킨다. 이러한 결과를 바탕으로 withaferin A는 새로운 항암제로서의 가능성을 가지고 있다.

인체간암세포에서 genistein의 TRAIL에 의한 apoptosis 유도 상승효과에서 미치는 p38 MAPK signaling pathway의 영향 (Enhancement of TRAIL-Mediated Apoptosis by Genistein in Human Hepatocellular Carcinoma Hep3B Cells: Roles of p38 MAPK Signaling Pathway)

  • 김성윤;박철;박상은;홍상훈;최영현
    • 생명과학회지
    • /
    • 제21권11호
    • /
    • pp.1549-1557
    • /
    • 2011
  • TRAIL은 다양한 암세포에서 apoptosis를 유발하는 것으로 알려져 있으나 간암세포를 포함한 일부 암세포에서 TRAIL 저항성이 획득된 것으로 보고되어지고 있다. 대두의 대표적인 생리활성 물질인 isoflavonoid계열 genistein은 이미 많은 암세포에서 apoptotic 효능을 가진 것으로 알려져 있으나 TRAIL에 의한 apoptosis 유도에 미치는 영향과 기전에 대한 연구는 여전히 미비한 실정이다. 본 연구에서는 TRAIL 저항성을 가진 Hep3B 간암세포에서 TRAIL에 의한 apoptosis 유도를 genistein이 더욱 상승시킬 수 있음을 보고하고자 한다. 본 연구의 결과에 의하면, Hep3B 세포에 세포독성을 보이지 않는 범위의 genistein에 의한 TRAIL 유도 apoptosis 상승효과는 미토콘드리아의 기능 손상과 연관성이 있었다. 또한 genistein과 TRAIL 복합처리에 의한 apoptosis 유도는 p38 MAPK 활성 저하로 더욱 상승하였으며, 이는 Bid의 truncation 증가, pro-apoptotic 단백질인 Bax의 발현 증가와 anti-apoptotic Bcl-2의 발현 감소 및 미토콘드리아에서 세포질로의 cytochrome c 유출의 증가와 연관성이 있었다. 또한 p38 MAPK 억제제는 genistein 및 TRAIL 복합처리에 의한 caspase의 활성 증가와 PARP 단백질의 단편화를 촉진시켰으며, 이는 미토콘드리아의 기능적 손상 증가에 의한 것임을 알 수 있었다. 따라서 본 연구의 결과는 genistein이 TRAIL에 의한 apoptosis 유도를 효과적으로 증가시킬 수 있으며, 이러한 과정이 p38 MAPK 의존적으로 이루어짐을 알 수 있었다.

Over Expression of BCL2 and Low Expression of Caspase 8 Related to TRAIL Resistance in Brain Cancer Stem Cells

  • Qi, Ling;Ren, Kuang;Fang, Fang;Zhao, Dong-Hai;Yang, Ning-Jiang;Li, Yan
    • Asian Pacific Journal of Cancer Prevention
    • /
    • 제16권12호
    • /
    • pp.4849-4852
    • /
    • 2015
  • Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) has been investigated as an effective agent to treat various cancers. Cancer stem cells are resistant to TRAIL treatment, but the mechanism of TRAIL resistance remains unknown. In this study, brain cancer stem cells were isolated by CD133 magnetic sorting, and the number of CD133 positive cells detected by flow cytometry. The self-renewing capacity of brain cancer stem cells was examined by a neurosphere formation assay, and the percentage of cell death after TRAIL treatment was examined by an MTS assay. Expression of DR5, FADD, caspase 8 and BCL2 proteins was detected by western blot. The amount of CD133 positive cells was enriched to 71% after CD133 magnetic sorting. Brain cancer stem cell neurosphere formation was significantly increased after TRAIL treatment. TRAIL treatment also reduced the amount of viable cells and this decrease was inhibited by a caspase 8 inhibitor or by the pan-caspase inhibitor z-VAD (P<0.05). Brain cancer stem cells expressed lower levels caspase 8 protein and higher levels of BCL2 protein when compared with CD133 negative cells (P<0.05). Our data suggest that TRAIL resistance is related to overexpression of BCL2 and low expression of caspase 8 which limit activation of caspase 8 in brain cancer stem cells.

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.