• Title/Summary/Keyword: Bax inhibitor

Search Result 173, Processing Time 0.027 seconds

Olibanum-induced Apoptosis Signaling in Human Cervical Carcinoma HeLa Cells (자궁경부암세포(子宮經部癌細胞)(HeLa cell)에서 유향(乳香) 세포고사(細胞枯死) 기전(機轉) 연구(硏究))

  • Park, Kyung-Mi;Kong, Bok-Cheul;Lee, Su-Jung;Choe, Chang-Min;Yoo, Sim-Keun
    • The Journal of Korean Obstetrics and Gynecology
    • /
    • v.19 no.2
    • /
    • pp.92-106
    • /
    • 2006
  • Purpose : To address the ability of Olibanum to induce cell death, we investigated the effect of olibanum on cell apoptosis. Twenty-four hours later, apoptosis occurred following olibanum exposure in a dose-dependent manner. Methods : We culture HeLa cell which is human metrocarcinoma cell in D-MEM included 10% fetal bovine serum(Hyclone Laboratories) below $37^{\circ}C$, 5% CO2. Then we observed apoptosis of log phage cell which is changed cultivation liquid 24 Hours periodically. Results : The treatment of BAPTA-AM regulated olibanum-induced apoptosis in HeLa human cervical carcinoma cells. The 24 hr-earlier -thapsigargin-pretreated cell showed the resistance against olibanum-induced apoptosis and the Ru360-mitochondrial uniporter-inhibited olibanum-induced apoptosis, too. It means that olibanum leads to the accumulation of calcium and the resultant apoptosis in HeLa cells. Immunoblotting data also shows that the expression of GRP78, ER stress marker protein, was induced by the olibanum. Bcl-2, anti-apototic protein, was decreased and that the expression of Bax, pro-apoptotic protein, was increased by the addition of olibanum. Interestingly, the olibanum increased the activity of caspase-8 as well as calpain cysteine pretense in HeLa cervical carcinoma cells. Calpain inhibitor-calpastatin as well as caspase-8C/A expression abrogated olibanum-induced apoptosis in the carcinoma cells. The inhibition of caspase-8 regulated olibanum-induced calpain activation but the inhibition of calpain did not have any effect on the caspase-8 activation in HeLa human cervical carcinoma cells. Conclusion : We conclude that olibanum induces the accumulation of calcium and the resultant apoptosis in which caspase-8 and calpain are involved.

  • PDF

Role of STAT3-Interacting Protein (STIP1) in ${\Delta}^{12}-Prostaglandin$ $J_2-Induced$ Cell Death

  • Kim, Seong-Mook;Lee, Sun;Kwak, Hwan-Jong;Kim, Bo-Eun;Kim, Dong-Jin;Kim, In-Kyung;Jeong, Seong-Whan
    • The Korean Journal of Physiology and Pharmacology
    • /
    • v.8 no.1
    • /
    • pp.27-31
    • /
    • 2004
  • ${\Delta}^{12}-Prostaglandin$ $J_2\;({\Delta}^{12}-PGJ_2)$ is one of cyclopentenone prostaglandins. The ${\Delta}^{12}-PGJ_2$ is known to induce apoptosis of tumor cells, however, it's action mechanism is not clear. It has recently been reported that STAT3 is involved in tumorigenesis. In the present study, we investigated the role of STAT3-interacting protein (STIP1) in the cytotoxicity of ${\Delta}^{12}-PGJ_2$, since STIP1 was recently reported as a modulator of STAT3 activation by specifically binding to inactive (unphosphorylated) STAT3. The effect of ${\Delta}^{12}-PGJ_2$ was observed in stably overexpressing Neuro-2A cells transfected with full cDNA of STIP1, and cytotoxicity of ${\Delta}^{12}-PGJ_2$ in the transfected cells was increased, compared with the vector control cells. The cytotoxicity of ${\Delta}^{12}-PGJ_2$ treatment was significantly accentuated by pretreatment of the STIP1-transfected cells with protein kinase inhibitor, genistein, and less activation of STAT3 in STIP1-transfected cells was shown, compared with the vector control cells. Expression of bax was also increased in the STIP1-transfected cells. These data suggest that STIP1 inhibits cell growth via inhibition of STAT3 activation in ${\Delta}^{12}-PGJ_2$ treatment.

Anthocyanins from Hibiscus syriacus L. Inhibit Oxidative Stress-mediated Apoptosis by Activating the Nrf2/HO-1 Signaling Pathway

  • Molagoda, Ilandarage Menu Neelaka;Karunarathne, Wisurumuni Arachchilage Hasitha Maduranga;Lee, Kyoung Tae;Choi, Yung Hyun;Jayasooriya, Rajapaksha Gedara Prasad Tharanga;Kim, Gi-Young
    • Proceedings of the Plant Resources Society of Korea Conference
    • /
    • 2019.10a
    • /
    • pp.91-91
    • /
    • 2019
  • Hibiscus syriacus L. is widely distributed throughout Eastern and Southern Asia and its root bark has been used as a traditional remedy. Recently, the extracts of H. syriacus L. exerts anti-cancerous, anti-microbial, and anti-inflammatory activities. However, the effect of anthocyanin-rich fraction of H. syriacus L. petals (PS) has not been studied under excessive oxidative stress. In this study, we evaluated the cellular protective effect of PS in HaCaT human skin keratinocytes under hydrogen peroxide ($H_2O_2$)-induced oxidative stress conditions. PS at below $400{\mu}g/ml$ did not show any cell death; however, over $800{\mu}g/ml$ of PS gradually increased cell death. PS at below $400{\mu}g/ml$ significantly inhibited $H_2O_2$-induced apoptosis in HaCaT cells concomitant with downregulation of Bax and upregulation of pro-PARP and p-Bcl-2. Additionally, PS remarkably reversed $H_2O_2$-induced excessive reactive oxygen species (ROS) production and apoptosis, and also significantly inhibited mitochondrial ROS production concomitant with suppression of $H_2O_2$-induced mitochondrial depolarization. $H_2O_2$-mediated ratio of Bax to Bcl-2, and caspase-3 activation were markedly abolished in the presence of PS. Moreover, the inhibition of HO-1 function using zinc protoporphyrin, an HO-1 inhibitor, significantly attenuated the cellular protective effects of PS against $H_2O_2$, indicating the significance of HO-1 in PS mediated cytoprotective effect, which was mediated by activating nuclear factor erythroid 2-related factor-2 (Nrf2). Taken together, our results suggest that cytoprotective effect of PS in HaCaT keratinocytes against oxidative stress-induced apoptosis is mediated by inhibiting cellular and mitochondrial ROS production, which is downregulated by activating Nrf2/HO-1 axis.

  • PDF

LncRNA LINC01232 Enhances Proliferation, Angiogenesis, Migration and Invasion of Colon Adenocarcinoma Cells by Downregulating miR-181a-5p

  • Yu Yuan;Zhou Long
    • Journal of Microbiology and Biotechnology
    • /
    • v.33 no.3
    • /
    • pp.398-409
    • /
    • 2023
  • LncRNAs play crucial roles in the progression of colon adenocarcinoma (COAD), but the role of LINC01232 in COAD has not received much attention. The present study was designed to explore the related mechanisms of LINC01232 in the progression of COAD. LINC01232, miR-181a-5p, p53, c-myc, Bcl-2, cyclin D1, p16, Bax, VEGF, E-cadherin, vimentin, N-cadherin and SDAD1 expressions were determined by western blot and qRT-PCR. CCK-8, tubule formation, and Transwell assays were employed to detect proliferation, angiogenesis, and migration/invasion of COAD cells, respectively. The relationship between LINC01232 and miR-181a-5p was predicted by LncBase Predicted v.2, and then verified through dual luciferase reporter gene assay. According to the results, LINC01232 was highly expressed in COAD cells and enhanced proliferation, angiogenesis, migration, and invasion of COAD cells. Downregulated LINC01232 promoted expression of p53 and p16, and inhibited c-myc, Bcl-2 and cyclin D1 expressions in COAD cells, while upregulation of LINC01232 generated the opposite effects. LINC01232 was negatively correlated with miR-181a-5p while downregulated miR181a-5p could reverse the effects of siLINC01232 on cell proliferation, angiogenesis, migration, and invasion. Similarly, miR-181a-5p mimic could also offset the effect of LINC01232 overexpression. SiLINC01232 increased the expressions of Bax and E-cadherin, and decreased the expressions of VEGF, vimentin, N-cadherin and SDAD1, which were partially attenuated by miR-181a-5p inhibitor. Collectively, LINC01232 enhances the proliferation, migration, invasion, and angiogenesis of COAD cells by decreasing miR-181a-5p expression.

Apoptotic Cell Death by Pectenotoxin-2 in p53-Deficient Human Hepatocellular Carcinoma Cells (종양억제유전자 p53 결손 인체간암세포에서 Pectenotoxin-2에 의한 Apoptosis 유도)

  • Shin, Dong-Yeok;Kim, Gi-Young;Choi, Byung-Tae;Kang, Ho-Sung;Jung, Jee-H.;Choi, Yung-Hyun
    • Journal of Life Science
    • /
    • v.17 no.10
    • /
    • pp.1447-1451
    • /
    • 2007
  • Through the screening of marine natural compounds that inhibit cancer cell proliferation, we previously reported that pectenotoxin-2 (PTX-2) isolated from marine sponges exhibits selective cytotoxicity against several cell lines in p53-deficient tumor cells compared to those with functional p53. However, the molecular mechanisms of its anti-proliferative action on malignant cell growth are not completely known. To further explore the mechanisms of its anti-cancer activity and to test whether the status of p53 in liver cancer cells correlates with their chemo-sensitivities to PTX-2, we used two well-known hepatocarcinoma cell lines, p53-deficient Hep3B and p53-wild type HepG2. We have demonstrated that PTX-2 markedly inhibits Hep3B cell growth and induces apoptosis whereas HepG2 cells are much more resistant to PTX-2 suggesting that PTX-2 seems to act by p53-independent cytotoxic mechanism. The apoptosis induced by PTX-2 in Hep3B cells was associated with the modulation of DNA fragmentation factor (DFF) family proteins, up-regulation of pro-apoptotic Bcl-2 family members such as Bax and Bcl-xS and activation of caspases (caspase-3, -8 and -9). Blockade of the caspase-3 activity by caspase-3 inhibitor, z-DEVD-fmk, prevented the PTX-2-induced growth inhibition in Hep3B cells. Moreover, treatment with PTX-2 also induced phosphorylation of AKT and extracellular-signal regulating kinase (ERK), but not c-Jun N-terminal kinase (JNK) and p38 mitogen-activated protein kinase (MARK). Specific inhibitors of PI3K inhibitor (LY294002) and ERK1/2 inhibitor (PD98059) significantly blocks PTX-2-induced-anti-proliferative effects, whereas a JNK inhibitor (SP600125) and a p38 MAPK inhibitor (SB203580) have no significant effects demonstrating that the pro-apoptotic effect of PTX-2 mediated through activation of AKT and ERK signal pathway in Hep3B cells.

Effects of Sodium Butyrate, a Histone Deacetylase Inhibitor, on TRAIL-mediated Apoptosis in Human Bladder Cancer Cells (인체 방광암세포에서 histone deacetylase 억제제인 sodium butyrate이 TRAIL에 의한 apoptosis 유도에 미치는 영향)

  • Han, Min-Ho;Choi, Yung Hyun
    • Journal of Life Science
    • /
    • v.26 no.4
    • /
    • pp.431-438
    • /
    • 2016
  • The tumour necrosis factor (TNF)-related apoptosis-inducing ligand (TRAIL) is considered a promising anticancer agent due to its unique ability to induce cancer cell death having only negligible effects on normal cells. However, many cancer cells tend to be resistant to TRAIL. In this study, we investigated the effects and molecular mechanisms of sodium butyrate (SB), a histone deacetylase inhibitor, in sensitizing TRAIL-induced apoptosis in 5637 human bladder cancer cells. Our results indicated that co-treatment with SB and TRAIL significantly increased the apoptosis induction, compared with treatment with either agent alone. Co-treatment with SB and TRAIL effectively increased the cell-surface expression of death receptor (DR) 5, but not DR4, which was associated with the inhibition of cellular Fas-associated death domain (FADD)-like interleukin-1β-converting enzyme (FLICE) inhibitory protein (c-FLIP). Furthermore, the activation of caspases (caspase-3, -8 and -9) and degradation of poly(ADP-ribose) were markedly increased in 5637 cells co-treated with SB and TRAIL; however, the synergistic effect was perfectly attenuated by caspase inhibitors. We also found that combined treatment with SB and TRAIL effectively induced the expression of pro-apoptotic Bax, cytosolic cytochrome c and cleave Bid to truncated Bid (tBid), along with down-regulation of anti-apoptotic Bcl-xL expression. These results collectively suggest that a combined regimen of SB plus TRAIL may offer an effective therapeutic strategy for safely and selectively treating TRAIL-resistant bladder cancer cells.

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.

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

  • Hong, Su Hyun;Park, Cheol;Kim, Kyoung Min;Choi, Yung Hyun
    • Journal of Life Science
    • /
    • v.25 no.11
    • /
    • pp.1235-1243
    • /
    • 2015
  • Hwangheuk-san (HHS) is a Korean multi-herb formula comprising four medicinal herbs. HHS, which was recorded in “Dongeuibogam,” has been used to treat patients with inflammation syndromes and digestive tract cancer for hundreds of years. However, little is known about its anti-tumor efficacy. The present study investigated the pro-apoptotic effect and mode of action of HHS against AGS human gastric carcinoma cells. HHS inhibited the cell growth of AGS cells in a dose-dependent manner, which was associated with the induction of apoptotic cell death, as evidenced by the formation of apoptotic bodies, chromatin condensation, and an accumulation of cells in the sub-G1 phase. HHS-induced apoptotic cell death was associated with the up-regulation of pro-apoptotic Bax protein expression, down-regulation of antiapoptotic Bcl-2 protein, and the release of cytochrome c from mitochondria to the cytosol. The treatment of AGS cells with HHS significantly elevated the generation of reactive oxygen species (ROS). Additionally, apoptosis-inducing concentrations of HHS induced the activation of both caspase-9 and -8, initiator caspases of the mitochondrial-mediated intrinsic and death receptor-mediated extrinsic pathways, respectively, and caspase-3, accompanied by proteolytic degradation of poly (ADP-ribose)-polymerase. However, ROS scavenger and pan-caspases inhibitor significantly blocked HHS-induced growth inhibition and apoptosis. Taken together, these findings suggest that HHS induces apoptosis through ROS- and caspase-dependent mechanisms and that HHS may be a potential chemotherapeutic agent for the control of human gastric cancer.

The Involvement of p38 MAPK and JNK Activation in Palmitic Acid-Induced Apoptosis in Rat Hepatocytes (Palmitic acid에 의한 간세포 사멸효과에 대한 p38 MAPK 및 JNK 관련성)

  • Bae, Chun-Sik;Park, Soo-Hyun
    • Journal of Life Science
    • /
    • v.19 no.8
    • /
    • pp.1119-1124
    • /
    • 2009
  • Hyperlipidemia has been reported to be associated with the development of fatty liver. Palmitic acid, a major saturated fatty acid, is involved in the development of diverse diseases. The activation of mitogen activated protein kinases (MAPKs), such as Jun N-terminal kinase (INKs) and p38 MAPK is implicated in the apoptosis in diverse cells. Thus, this study was conducted to investigate the effects of palmitic acid on apoptosis and its relationship between JNK and p38 MAPK in cultured rat hepatocytes. In the present study, palmitic acid (>50 uM) decreased cell proliferation and increased lactate dehydrogenase activity in hepatocytes, which was blocked by the treatment of SP600125 (a JNK inhibitor) and SB203580 (a p38 MAPK inhibitor). Indeed, palmitic acid decreased Bcl-2 expression but increased Bax expression in rat hepatocytes, which was blocked by the treatment of SP600125 and SB203580. In addition, palmitic acid decreased glutathione (GSH) content and increased lipid peroxide formation, which was blocked by the treatment of SP600125 and SB203580. Western immunoblotting analysis also revealed that palmitic acid increased JNK and p38 MAPK. In conclusion, palmitic acid induced apoptosis through oxidative stress via JNK and p38 MAPK activation in rat hepatocytes.