• Title/Summary/Keyword: poly (ADP-ribose) polymerase (PARP)

Search Result 263, Processing Time 0.022 seconds

Caspase-8 Potentiates Triglyceride (TG)-Induced Cell Death of THP-1 Macrophages via a Positive Feedback Loop (Caspase-8의 양성 피드백 방식을 통한 중성지방-유도 THP-1 대식세포 사멸 증가)

  • Jung, Byung Chul;Lim, Jaewon;Kim, Sung Hoon;Kim, Yoon Suk
    • Korean Journal of Clinical Laboratory Science
    • /
    • v.53 no.2
    • /
    • pp.158-164
    • /
    • 2021
  • Hypertriglyceridemia is the main risk factor for atherosclerosis. It is reported that triglyceride (TG) induces macrophage cell death, and is involved in the formation of plaques and development of atherosclerosis. We previously reported that TG-induced cell death of macrophages is mediated via pannexin-1 activation, which increases the extracellular ATP and subsequent increase in potassium efflux, thereby activating the caspase-2/caspase-1/apoptotic caspases, including the caspase-8 pathway. Contrarily, some studies have reported that caspase-8 is an upstream molecule of caspase-1 and caspase-2 in several cellular processes. Therefore, this study was undertaken to investigate whether caspase-8 influences its upstream molecules in TG-stimulated macrophage cell death. We first confirmed that caspase-8 induces caspase-3 activation and poly ADP-ribose polymerase (PARP) cleavage in TG-treated macrophages. Next, we determined that the inhibition of caspase-8 results in reduced caspase-1 and -2 activity, which are upstream molecules of caspase-8 in TG-induced cell death of macrophages. We also found that ATP treatment restores the caspase-8 inhibitor-induced caspase-2 activity, thereby implying that caspase-8 affects the upstream molecules responsible for increasing the extracellular ATP levels in TG-induced macrophage cell death. Taken together, these findings indicate that caspase-8 potentiates the TG-induced macrophage cell death by activating its upstream molecules.

Ginsenoside 20(S)-protopanaxadiol induces cell death in human endometrial cancer cells via apoptosis

  • Jo, Hantae;Jang, Dongmin;Park, Sun Kyu;Lee, Mi-Gi;Cha, Byungsun;Park, Chaewon;Shin, Yong Sub;Park, Hyein;Baek, Jin-myoung;Heo, Hyojin;Brito, Sofia;Hwan, Hyun Gyu;Chae, Sehyun;Yan, Shao-wei;Lee, Changho;Min, Churl K.;Bin, Bum-Ho
    • Journal of Ginseng Research
    • /
    • v.45 no.1
    • /
    • pp.126-133
    • /
    • 2021
  • Background: 20(S)-protopanaxadiol (20(S)-PPD), one of the aglycone derivatives of major ginsenosides, has been shown to have an anticancer activity toward a variety of cancers. This study was initiated with an attempt to evaluate its anti-cancer activity toward human endometrial cancer by cell and xenograft mouse models. Methods: Human endometrial cancer (HEC)-1A cells were incubated with different 20(S)-PPD concentrations. 20(S)-PPD cytotoxicity was evaluated using MTT assay. Apoptosis was detected using the annexin V binding assay and cell cycle analysis. Cleaved poly (ADP-ribose) polymerase (PARP) and activated caspase-9 were assessed using western blotting. HEC-1A cell tumor xenografts in athymic mice were generated by inoculating HEC-1A cells into the flank of BALB/c female mice and explored to validate 20(S)-PPD anti-endometrial cancer toxicity. Results: 20(S)-PPD inhibited HEC-1A cell proliferation in a dose-dependent manner with an IC50 value of 3.5 μM at 24 h. HEC-1A cells morphologically changed after 20(S)-PPD treatment, bearing resemblance to Taxol-treated cells. Annexin V-positive cell percentages were 0%, 10.8%, and 58.1% in HEC-1A cells when treated with 0, 2.5, and 5 μM of 20(S)-PPD, respectively, for 24 h. 20(S)-PPD subcutaneously injected into the HEC-1A cell xenograft-bearing mice three times a week for 17 days manifested tumor growth inhibition by as much as 18% at a dose of 80 mg/kg, which sharply contrasted to controls that showed an approximately 2.4-fold tumor volume increase. These events paralleled caspase-9 activation and PARP cleavage. Conclusion: 20(S)-PPD inhibits endometrial cancer cell proliferation by inducing cell death via a caspase-mediated apoptosis pathway. Therefore, the 20(S)-PPD-like ginsenosides are endowed with ample structural information that could be utilized to develop other ginsenoside-based anticancer agents.

Anti-proliferation, Cell Cycle Arrest, and Apoptosis Induced by Natural Liquiritigenin from Licorice Root in Oral Squamous Cell Carcinoma Cells (구강편평세포암종 세포에서 감초 유래 Liquiritigenin의 항증식, 세포주기 정지 및 세포사멸 유도)

  • Kwak, Ah-Won;Yoon, Goo;Chae, Jung-Il;Shim, Jung-Hyun
    • Journal of Life Science
    • /
    • v.29 no.3
    • /
    • pp.295-302
    • /
    • 2019
  • Liquiritigenin (LG) is a chiral flavonoid isolated from the roots of licorice. It exhibits multiple biological activities including anti-oxidant, anti-cancer, and anti-inflammatory effects. In particular though, the anti-cancer activity of LG in oral squamous cell carcinoma has yet to be elucidated, and LG-induced apoptosis in oral squamous cell carcinoma remains poorly understood. In the present study, we tested the role of LG in inducing apoptosis in oral squamous cell carcinoma cells. LG treatment of HN22 cells resulted in a dose-dependent inhibition of cell viability as detected by a 3-(4,5-dimethylthiazol-2-yl)-2,5 diphenyltetrazolium bromide assay. The induction of apoptosis in terms of Annexin V/7-Aminoactinomycin D staining, sub-G1 population, and multi-caspase activity were assessed with a $Muse^{TM}$ Cell Analyzer. Flow cytometric analysis revealed that LG treatment resulted in G2/M arrest in cell cycle progression and downregulation of cyclin B1 and CDC2 expression in a concentration-dependent manner. It also resulted in significant upregulation of p27. In addition, LG was seen to trigger the generation of reactive oxygen species and induce CCAAT/enhancer-binding protein homologous protein and 78-kDa glucose-regulated protein in concentration-dependent upregulation. The LG treatment of HN22 cells led to a loss of mitochondrial membrane potential (${\Delta}{\Psi}m$); it also reduced the levels of anti-apoptotic protein and increased the expression of apoptotic protease activating factor-1, cleaved poly (ADP-ribose)polymerase and Bax. Overall, our results indicate that the pro-apoptotic effects of LG in HN22 cells depend on the activation of both intrinsic and extrinsic signaling pathways. Thus, our results suggest that LG constitutes a natural compound with a potential role as an anti-tumor agent in oral squamous cell carcinoma.

A Study on Apoptotic Signaling Pathway in HL-60 Cells Induced by Radiation (급성전골수성백혈병 HL-60 세포주에서 방사선조사에 의한 세포고사기전)

  • Kim Hye Jung;Moon Sung Keun;Lee Jae Moon;Moon Sun Rock
    • Radiation Oncology Journal
    • /
    • v.19 no.2
    • /
    • pp.153-162
    • /
    • 2001
  • Purpose : The mechanical insights of death of cancer cells by ionizing radiation are not of yet clearly defined. Recent evidences have demonstrated that radiation therapy may induce cell death via activation of signaling pathway for apoptosis in target cells. This study is designed whether ionizing radiation may activate the signaling cascades of apoptosis including caspase family cysteine pretenses, $Bcl_2/Bax$, cytochrome c and Fas/Fas-L in target cells. Materials and Methods : HL-60 cells were irradiated in vitro with 6 MV X-ray at dose ranges from 2 Gy to 32 Gy. The cell viability was tested by M assay and the extent of apoptosis was determined using agarose gel electrophoresis. The activities of caspase proteases were measured by proteolytic cleavages of substrates. Western blot analysis was used to monitor PARP, Caspase-3, Cytochrome-c, Bcl-2, Bax, Fas and Fas-L. Results : Ionizing radiation decreases the viability of HL-60 cells in a time and dose dependent manner. Ionizing radiation-induced death in HL-60 cells is an apoptotic death which is revealed as characteristic ladder-pattern fragmentation of genomic DNA over 16 Gy at 4 hours. ionizing radiation induces the activation of caspase-2, 3, 6, 8 and 9 of HL-60 cells in a time-dependent manner. The activation of caspase-3 pretense is also evidenced by the digestion of poly (ADP-ribose) polymerase and procaspase 3 with 16Gy ionizing irradiation. Anti-apoptotic Bcl2 expression is decreased but apoptotic Bax expression is increased with mitochondrial cytochrome c release in a time- dependent manner. In addiiton, expression of Fas and Fas-L is also increased in a time dependent manner. Conclusion : These data suggest that ionizing radiation-induced apoptosis is mediated by the activation of various signaling pathways including caspase family cysteine proteases, $Bcl_2/Bax$, Fas and Fas-L in a time and dose dependent manner.

  • PDF

H9 Inhibits Tumor Growth and Induces Apoptosis via Intrinsic and Extrinsic Signaling Pathway in Human Non-Small Cell Lung Cancer Xenografts

  • Kim, Min-Je;Kwon, Sae-Bom;Ham, Seung Hoon;Jeong, Eui-Suk;Choi, Yang-Kyu;Choi, Kang Duk;Hong, Jin Tae;Jung, Seung Hyun;Yoon, Do-Young
    • Journal of Microbiology and Biotechnology
    • /
    • v.25 no.5
    • /
    • pp.648-657
    • /
    • 2015
  • H9, a novel herbal extract, demonstrated cytotoxicity in A549 non-small cell lung cancer (NSCLC) cell lines. In this study, we investigated whether H9, and/or co-treatment with an anticancer drug, pemetrexed (PEM), inhibited tumor growth in BALB/c nude mice models bearing A549 NSCLC cells. The mice were separated into groups and administered H9 and PEM for 2 weeks. Protein and mRNA levels were detected using western blotting and reverse transcription polymerase chain reaction, respectively; immunohistochemistry (IHC) was also performed on the tumor tissues. H9 and co-treatment with PEM induced the cleavage of proapoptotic factors, such as caspase-3, caspase-8, caspase-9, and poly(ADP)-ribose polymerase (PARP). Expression levels of cell-death receptors involving Fas/FasL, TNF-related apoptosisinducing ligands (TRAIL), and TRAIL receptors were increased by H9 and co-treatment with PEM. Furthermore, analysis of levels of cell-cycle modulating proteins indicated that tumor cells were arrested in the G1/S phase. In addition, the phosphatidylinositol-4,5-bisphosphate 3-kinase (PI3K)/Akt survival signaling pathways were inhibited by H9 and co-treatment with PEM. In conclusion, H9 and co-treatment with PEM inhibited tumor growth in BALB/c nude mice models bearing A549 NSCLC cells. These results indicate that H9 and co-treatment with PEM can be used as an anticancer therapy in NSCLC.

Anti-cancer Potentials of Rhus verniciflua Stokes, Ulmus davidiana var. japonica Nakai and Arsenium Sublimatum in Human Gastric Cancer AGS Cells (AGS 인체위암세포에서 건칠, 유근피 및 신석 추출물의 항암 활성 비교 연구)

  • Baek, Ilsung;Im, Lyeng-Hae;Park, Cheol;Cho, Yung Hyun
    • Journal of Life Science
    • /
    • v.25 no.8
    • /
    • pp.849-860
    • /
    • 2015
  • The anti-cancer activities of Rhus verniciflua Stokes (GC), Ulmus davidiana var. japonica Nakai (UGP) and arsenium sublimatum (SS) extracts, which have been used Oriental medicine therapy for various diseases, were investigated. The treatment of GC, UGP and SS alone, and combined treatment with GC, UGP and SS did not affect the cell viability in the mouse normal cell lines (RAW 264.7 macrophages and C2C12 myoblasts). However, co-treatment with GC, UGP and SS markedly induces apoptosis in human gastric cancer AGS cells, but not in other various cancer cell lines (human lung cancer A549, colon cancer HCT116, liver cancer Hep3B and bladder T24 cells) as evidenced by formation of apoptotic bodies, chromatin condensation, and accumulation of annexin-V positive cells. Co-treatment with GC, UGP and SS effectively induced the expression levels of Fas and Fas ligand, and inhibited the levels IAP family proteins such as XIAP, cIAP-1 and survivin, and anti-apoptotic Bcl-xL proteins compared with treatment with either agent alone. Combined treatment also significantly induced the loss of mitochondrial membrane potential, which was associated with the activation of caspases (-3, -8, and -9) and degradation of poly (ADP-ribose) polymerase. However, the cytotoxic effects induced by co-treatment with GC, UGP and SS were significantly attenuated by pan-caspases inhibitor, z-VAD-fmk, indicating an important role for caspases. These results indicated that the caspases were key regulators of apoptosis in response to co-treatment of GC, UGP and SS in human gastric cancer AGS cells and further studies will be needed to identify the active compounds.

Anticancer Effects of Typhae Pollen on HepG2 Human Hepatocellular Carcinoma

  • Joo, Jeong-Hyun;Kim, Kyung-Soon;Choi, Hong-Sik;Kim, Seung-Mo
    • Journal of Physiology & Pathology in Korean Medicine
    • /
    • v.32 no.4
    • /
    • pp.261-270
    • /
    • 2018
  • The aim of this study was to evaluate the antitumor activities of Typhae pollen (TP) by confirming in vitro cytotoxicity and in vivo anti-tumor and immune-modulatory effect with anti-cachexia effect. The MTT assay is used in HepG2 cell to detect potential cytotoxic activities of aqueous extract of Typhae pollen (TPe). After HepG2 tumor cell implantation, eight mice per groups were assigned to six groups. Three different dosages of TPe (500, 250 and 125 mg/kg) were orally administered in the amount of $10m{\ell}/kg$ and sorafenib also administered 20mg/kg, every day for 35 days from 28 days after the tumor cell implantation. We observed the changes on body weights, tumor volume and weights, lymphatic organ, serum interferon $(IFN)-{\gamma}$ levels, splenocytes and peritoneal NK cell activity, splenic tumor necrosis factor $(TNF)-{\alpha}$, interleukin $(IL)-1{\beta}$, IL-10 contents. Periovarian fat weights, serum IL-6 levels, thicknesses of deposited periovarian adipose tissue and mean diameters were also detected to monitor the tumor-related anticachexic effects. In tumor masses, the immunoreactivities of cleaved caspase-3 and cleaved poly (ADP-ribose) polymerase (cleaved PARP) - apoptotic marks, cyclooxygenase-2 (COX-2), inducible nitric oxide synthases (iNOS) and tumor necrosis factor $(TNF)-{\alpha}$ were additionally observed by immunohistochemistry. The results were compared with sorafenib. Decreases of COX-2 were demonstrated in sorafenib and TPe treated mice and also increases of iNOS in tumor masses were observed in TPe, not in sorafenib. TPe increased periovarian fat pad weights compared with tumor-bearing controls and sorafenib treated mice. TPe showed increases of splenic $TNF-{\alpha}$, IL-10 and $IL-1{\beta}$, serum $IFN-{\gamma}$ and NK cell activities corresponding to increases of spleen weights, lymph node weights and non-atrophic changes of lymph nodes. Our results show oral treatment of TPe 500, 250 and 125 mg/kg has potent in vitro and in vivo antitumor activities through modest cytotoxic effects, immunomodulatory effects and apoptotic activities in HepG2 tumor cells. In addition, TPe can prevent cancer related cachexia.

A Study on the Mechanisms by Which the Aqueous Extract of Inonotus obliquus Induces Apoptosis and Inhibits Proliferation in HT-29 Human Colon Cancer Cells (차가버섯 물추출물의 대장암세포 증식억제 및 Apoptosis 유도기전 연구)

  • Kim, Eun-Ji;Lee, Yong-Jin;Shim, Hyun-Kyung;YoonPark, Jung-Han
    • Journal of the Korean Society of Food Science and Nutrition
    • /
    • v.35 no.5
    • /
    • pp.516-523
    • /
    • 2006
  • The mushroom Inonotus obliquue (IO) has been traditionally used for the treatment of gastrointestinal cancer in Russia, Poland, and most of Baltic countries. To explore the possibility that IO has chemoprevention effects, we examined whether or not the aqueous extract of IO inhibits HT-29 cell growth and investigated tile mechanism for this effect. Cells were incubated in the presence of increasing concentrations of the aqueous extract of IO. The extract substantially inhibited the viable HT-29 cell number in a dose-dependent manner and inhibited 5-bromo-2'-deoxyuridine incorporation into DNA of HT-29 cells. Annexin-V staining followed by flow cytometry revealed that the extract induced apoptosis of HT-29 cells in a dose-dependent manner. Western blot analysis of total cell lysates revealed that the extract induced cleavage of caspase-8, -9 and -3 and poly (ADP-ribose) polymerase, but did not affect the protein levels of Bax and Bcl-2. In addition, the extract dose-dependently increased the activity of caspase-8, -9 and -3. We have demonstrated that the aqueous extract of IO inhibits cell proliferation and induces apoptosis in HT-29 cells, which may be mediated by its ability to activate the caspase pathway.

Herbal medicine In-Jin-Ho-Tang as a potential anti-cancer drug by induction of apoptosis in human hepatoma HepG2 cells. (사람 간암 세포주인 HepG2에 대한 인진호탕(茵陳蒿湯)의 항암 효과)

  • Yun, Hyun-Joung;Kim, Byung-Wan;Lee, Chang-Hyun;Jung, Jae-Ha;Heo, Sook-Kyung;Park, Won-Hwan;Park, Sun-Dong
    • The Korea Journal of Herbology
    • /
    • v.22 no.3
    • /
    • pp.27-37
    • /
    • 2007
  • Objectives: Hepatocellular carcinoma is the most common primary malignant tumor of the liver worldwide. In-Jin-Ho-Tang(IJHT) has been used as a traditional Chinese herbal medicine since ancient time. and today it is widely applied as a medication for jaundice which is associated with inflammation in liver. In this study, I investigated whether methanol extract of IJHT induced HepG2 cancer cell death. Methods: Cytotoxic activity of IJHT on HepG2 cells was using XTT assay. Apoptosis induction by Ros A in HCT116 cells was verified by the induction of cleavage of poly ADP-ribose polymerase (PARP). and activation of caspase-3, -8 and -9. The release of cytochrome c from mitochondria to cytosol. the level of Bcl-2 and Bax and the expression of p53 and p21 were examined by western blotting analysis. Furthermore, MAPKs activation was analyzed by western blotting analysis. Results: IJHT induced apoptosis in HepG2 cells. And treatment of IJHT resulted in the release of cytochrome c into cytosol, decreased anti-apoptotic Bcl-2, and increased pri-apoptotic Bax expression. IJHT markedly inactivated extracellular signal-regulated kinase (ERK1/2), and activated p38 mitogen-activated protein (MAP) kinase. Sodium orthovanadate (SOV), a phosphatase inhibitor, to reverse IJHT-induced ERK1/2 inactivation and SB203580, a specific p38 MAP Kinase inhibitor efficiently blocked apoptosis of HepG2. Thus, IJHT induces apoptosis in HepG2 cells via MAP kinase modulation. Conclusion: These results indicated that IJHT has some potential for use as an anti-cancer agent.

  • PDF

Sanguinarine Induces Apoptosis in Human Hepatocellular Carcinoma HepG2 Cells through the Generation of ROS and Modulation of Akt/ERK Signaling Pathways (HepG2 인체 간암세포의 ROS 생성 및 ERK/Akt 신호전달 경로 조절을 통한 sanguinarine의 apoptosis 유도)

  • Hwang, Ju Yeong;Cho, Yung Hyun
    • Journal of Life Science
    • /
    • v.25 no.9
    • /
    • pp.984-992
    • /
    • 2015
  • Sanguinarine is a benzophenanthridine alkaloid originally isolated from the roots of Sanguinaria canadensis. It has multiple biological activities (e.g., antioxidant and antiproliferative) and immune-enhancing potential. In this study, we explored the proapoptotic properties and modes of action of sanguinarine in human hepatocellular carcinoma HepG2 cells. Our results revealed that sanguinarine inhibited HepG2 cell growth and induced apoptosis in a dose-dependent manner. The induction of apoptosis by sanguinarine was associated with the up-regulation of Fas and Bax, the release of cytochrome c from the mitochondria to the cytosol, and the loss of the mitochondrial membrane potential. In addition, sanguinarine activated caspase-9 and -8, initiator caspases of the intrinsic and death extrinsic pathways, respectively, and caspase-3, accompanied by proteolytic degradation of poly (ADP-ribose) polymerase. Sanguinarine also triggered the generation of reactive oxygen species (ROS). The elimination of ROS by N-acetylcysteine reversed sanguinarine-induced apoptosis. Furthermore, sanguinarine induced the dephosphorylation of Akt and the phosphorylation of mitogen-activated protein kinases, including extracellular signal-regulated kinase (ERK), c-jun N-terminal kinase (JNK), and p38. The growth inhibition was enhanced by the combined treatment of sanguinarine with a phosphatidylinositol 3'-kinase (PI3K) inhibitor and an ERK inhibitor but not JNK and p38 inhibitors. Overall, our data indicate that the proapoptotic effects of sanguinarine in HepG2 cells depend on ROS production and the activation of both intrinsic and extrinsic signaling pathways, which is mediated by blocking PI3K/Akt and activating the ERK pathway. Thus, our data suggest that sanguinarine may be a natural compound with potential for use as an antitumor agent in liver cancer.