• Title/Summary/Keyword: intrinsic apoptosis

Search Result 154, Processing Time 0.024 seconds

Induction of Apoptosis with Moringa oleifera Fruits in HCT116 Human Colon Cancer Cells Via Intrinsic Pathway

  • Guon, Tae-Eun;Chung, Ha Sook
    • Natural Product Sciences
    • /
    • v.23 no.4
    • /
    • pp.227-234
    • /
    • 2017
  • Moringa oleifera Lam (M. oleifera, Moringaceae) is a tree of the Moringaceae family that can reach a height of between 5 and 10 m. The current paper presents cytotoxic effect of M. oleifera fruits and its flavonoids 1 and 2. The viability of HCT116 human colon cancer cells were 38.5% reduced by $150{\mu}g/mL$ of ethanolic extracts in a concentration-dependent manner; in addition, we observed the apoptotic features of cell shrinkage and decreased cell size. Bcl-2 family proteins were regulated as determined by Western blotting analysis, suggesting that M. oleifera fruits and their flavonoids 1 and 2 induced apoptosis through an intrinsic pathway. Based on our findings, 70% ethanolic extracts of M. oleifera fruits and flavonoids 1 and 2 might be useful as cytotoxic agents in colorectal cancer therapy.

Peroxisome Proliferator-Activated Receptor-Gamma Agonist 4-O-Methylhonokiol Induces Apoptosis by Triggering the Intrinsic Apoptosis Pathway and Inhibiting the PI3K/Akt Survival Pathway in SiHa Human Cervical Cancer Cells

  • Hyun, Seungyeon;Kim, Man Sub;Song, Yong Seok;Bak, Yesol;Ham, Sun Young;Lee, Dong Hun;Hong, Jintae;Yoon, Do Young
    • Journal of Microbiology and Biotechnology
    • /
    • v.25 no.3
    • /
    • pp.334-342
    • /
    • 2015
  • 4-O-Methylhonokiol (MH), a bioactive compound derived from Magnolia officinalis, is known to exhibit antitumor effects in various cancer cells. However, the precise mechanism of its anticancer activity in cervical cancer cells has not yet been studied. In this study, we demonstrated that MH induces apoptosis in SiHa cervical cancer cells by enhancing peroxisome proliferator-activated receptor-gamma (PPARγ) activation, followed by inhibition of the PI3K/Akt pathway and intrinsic pathway induction. MH upregulated PPARγ and PTEN expression levels while it decreased p-Akt in the MH-induced apoptotic process, thereby supporting the fact that MH is a PPARγ activator. Additionally, MH decreased the expression of Bcl-2 and Bcl-XL, inducing the intrinsic pathway in MH-treated SiHa cells. Furthermore, MH treatment led to the activation of caspase-3/caspase-9 and proteolytic cleavage of polyADP ribose polymerase. The expression levels of Fas (CD95) and E6/E7 oncogenes were not altered by MH treatment. Taken together, MH activates PPARγ/PTEN expression and induces apoptosis via suppression of the PI3K/Akt pathway and mitochondria-dependent pathways in SiHa cells. These findings suggest that MH has potential for development as a therapeutic agent for human cervical cancer.

H9 Induces Apoptosis via the Intrinsic Pathway in Non-Small-Cell Lung Cancer A549 Cells

  • Kwon, Sae-Bom;Kim, Min-Je;Sun Young, Ham;Park, Ga Wan;Choi, Kang-Duk;Jung, Seung Hyun;Do-Young, Yoon
    • Journal of Microbiology and Biotechnology
    • /
    • v.25 no.3
    • /
    • pp.343-352
    • /
    • 2015
  • H9 is an ethanol extract prepared from nine traditional/medicinal herbs. This study was focused on the anticancer effect of H9 in non-small-cell lung cancer cells. The effects of H9 on cell viability, apoptosis, mitochondrial membrane potential (MMP; ${\Delta}\psi_{m}$), and apoptosisrelated protein expression were investigated in A549 human lung cancer cells. In this study, H9-induced apoptosis was confirmed by propidium iodide staining, expression levels of mRNA were determined by reverse transcriptase polymerase chain reaction, protein expression levels were checked by western blot analysis, and MMP (${\Delta}\psi_{m}$) was measured by JC-1 staining. Our results indicated that H9 decreased the viability of A549 cells and induced cell morphological changes in a dose-dependent manner. H9 also altered expression levels of molecules involved in the intrinsic signaling pathway. H9 inhibited Bcl-xL expression, whereas Bax expression was enhanced and cytochrome C was released. Furthermore, H9 treatment led to the activation of caspase-3/caspase-9 and proteolytic cleavage of poly(ADP-ribose) polymerase; the MMP was collapsed by H9. However, the expression levels of extrinsic pathway molecules such as Fas/FasL, TRAIL/TRAIL-R, DR5, and Fas-associated death receptor were downregulated by H9. These results indicated that H9 inhibited proliferation and induced apoptosis by activating intrinsic pathways but not extrinsic pathways in human lung cancer cells. Our results suggest that H9 can be used as an alternative remedy for human non-small-cell lung cancer.

Induction of Apoptosis by Pachymic Acid in T24 Human Bladder Cancer Cells (T24 인체방광암 세포에서 pachymic acid에 의한 apoptosis 유발)

  • Jeong, Jin-Woo;Baek, Jun Young;Kim, Kwang Dong;Choi, Yung Hyun;Lee, Jae-Dong
    • Journal of Life Science
    • /
    • v.25 no.1
    • /
    • pp.93-100
    • /
    • 2015
  • Pachymic acid (PA) is a lanostane-type triterpenoid derived from the Poria cocos mushroom. Several beneficial biological features of PA provide medicine with a wide variety of valuable effects, such as anticancer and anti-inflammatory activity; it also has antioxidant effects against oxidative stress. Nonetheless, the biological properties and mechanisms that produce this anti-cancer action of PA remain largely undetermined. In this study, we investigated the pro-apoptotic effects of PA in T24 human bladder cancer cells. It was found that PA could inhibit the cell growth of T24 cells in a dose-dependent manner, which was associated with the induction of apoptotic cell death, as evidenced by the formation of apoptotic bodies and chromatin condensation and accumulation of cells in the sub-G1 phase. The induction of apoptotic cell death by PA was connected with an up-regulation of pro-apoptotic Bax and Bad protein expression and down-regulation of anti-apoptotic Bcl-2 and Bcl-xL proteins, and inhibition of apoptosis family proteins. In addition, apoptosis-inducing concentrations of PA induced the activation of caspase-9, an initiator caspase of the mitochondrial-mediated intrinsic pathway, and caspase-3, accompanied by proteolytic degradation of poly (ADP-ribose)-polymerase. PA also induced apoptosis via a death receptor-mediated extrinsic pathway by caspase-8 activation, resulting in the truncation of Bid and suggesting the existence of cross-talk between the extrinsic and intrinsic pathways. Taken together, the present results suggest that PA may be a potential chemotherapeutic agent for the control of human bladder cancer cells.

ALEX1 Regulates Proliferation and Apoptosis in Breast Cancer Cells

  • Gao, Yue;Wu, Jia-Yan;Zeng, Fan;Liu, Ge-Li;Zhang, Han-Tao;Yun, Hong;Song, Fang-Zhou
    • Asian Pacific Journal of Cancer Prevention
    • /
    • v.16 no.8
    • /
    • pp.3293-3299
    • /
    • 2015
  • Background: Arm protein lost in epithelial cancers, on chromosome X (ALEX) is a novel subgroup within the armadillo (ARM) family, which has one or two ARM repeat domains as opposed to more than six-thirteen repeats in the classical Armadillo family members. Materials and Methods: In the study, we explore the biological functions of ALEX1 in breast cancer cells. Overexpression of ALEX1 and silencing of ALEX1 were performed with SK-BR3 and MCF-7 cell lines. Cell proliferation and colony formation assays, along with flow cytometry, were carried out to evaluate the roles of ALEX1. Results: ALEX1 overexpression in SK-BR3 breast cancer cells inhibited proliferation and induced apoptosis. Furthermore, depletion of ALEX1 in MCF-7 breast cancer cells increased proliferation and inhibited apoptosis. Additional analyses demonstrated that the overexpression of ALEX1 activated the intrinsic apoptosis cascades through up-regulating the expression of Bax, cytosol cytochrome c, active caspase-9 and active caspase-3 and down-regulating the levels of Bcl-2 and mitochondria cytochrome c. Simultaneouly, silencing of ALEX1 inhibited intrinsic apoptosis cascades through down-regulating the expression of Bax, cytosol cytochrome c, active caspase-9, and active caspase-3 and up-regulating the level of Bcl-2 and mitochondria cytochrome c. Conclusions: Our data suggest that ALEX1 as a crucial tumor suppressor gene has been involved in cell proliferation and apoptosis in breast cancer, which may serve as a novel candidate therapeutic target.

Apicidin Induces Apoptosis via Cytochrome c-Mediated Intrinsic Pathway in Human Ovarian Cancer Cells

  • Ahn, Mee-Young;Na, Yong-Jin;Lee, Jae-Won;Lee, Byung-Mu;Kim, Hyung-Sik
    • Biomolecules & Therapeutics
    • /
    • v.17 no.1
    • /
    • pp.17-24
    • /
    • 2009
  • Histone deacetylase (HDAC) inhibitors are a promising class of anticancer agents that inhibit cancer cell growth in vitro and in vivo. Previous report has shown that apicidin inhibited SK-OV-3 cells proliferation and down-regulation of cyclin B1 and CDK1, and up-regulation of $p21^{WAF1}$ and p27. However, the mechanism of apicidin-mediated apoptotic cell death is not clearly understood. For this study, we investigated the mechanism of apoptotic pathway induced by apicidin in human ovarian cancer cell. We found that SK-OV-3 cells treated with apicidin caused an increase in the percentage of cells in the G2/M phase, which preceded apoptosis characterized by the appearance of cells with sub-G1 population. To further investigate the mechanism of apoptosis induction by apicidin, we measured TUNEL assay, poly-ADP ribose polymerase (PARP) cleavage, and caspase activity in SK-OV-3 cells treated with apicidin for 48 h. Apicidin significantly enhanced apoptosis as measured by TUNEL positive apoptotic cells, PARP cleavage, and increased Bax/Bcl-2 ratio. Induction of apoptosis was confirmed by the release of cytochrome c to cytosol. Our data suggest that apicidin-induced apoptosis in SK-OV-3 cells was accompanied by caspase-3 activation and the increase in Bax/Bcl-2 ratio. These data suggest that apicidin may be effective in the treatment of ovarian cancer through activation of intrinsic apoptotic pathway.

Apigenin Sensitizes Huh-7 Human Hepatocellular Carcinoma Cells to TRAIL-induced Apoptosis

  • Kim, Eun-Young;Kim, An-Keun
    • Biomolecules & Therapeutics
    • /
    • v.20 no.1
    • /
    • pp.62-67
    • /
    • 2012
  • TNF-related apoptosis-inducing ligand (TRAIL) is a promising agent for management of cancer because of its selective cytotoxicity to cancer cells. However, some cancer cells have resistance to TRAIL. Accordingly, novel treatment strategies are required to overcome TRAIL resistance. Here, we examined the synergistic apoptotic effect of apigenin in combination with TRAIL in Huh-7 cells. We found that combined treatment of TRAIL and apigenin markedly inhibited Huh-7 cell growth compared to either agent alone by inducing apoptosis. Combined treatment with apigenin and TRAIL induced chromatin condensation and the cleavage of poly (ADP-ribose) polymerase (PARP). In addition, enhanced apoptosis by TRAIL/apigenin combination was quantified by annexin V/PI flow cytometry analysis. Western blot analysis suggested that apigenin sensitizes cells to TRAIL-induced apoptosis by activating both intrinsic and extrinsic apoptotic pathway-related caspases. The augmented apoptotic effect by TRAIL/apigenin combination was accompanied by triggering mitochondria-dependent signaling pathway, as indicated by Bax/Bcl-2 ratio up-regulation. Our results demonstrate that combination of TRAIL and apigenin facilitates apoptosis in Huh-7 cells.

E1/E2 of Hepatitis C Virus Genotype-4 and Apoptosis

  • Zekri, Abdel-Rahman N;Sobhy, Esraa;Hussein, Nehal;Ahmed, Ola S;Hussein, Amira;Shoman, Sahar;Soliman, Amira H;El-Din, Hanaa M Alam
    • Asian Pacific Journal of Cancer Prevention
    • /
    • v.17 no.7
    • /
    • pp.3131-3138
    • /
    • 2016
  • Several studies have addressed the possible role of hepatitis C virus genotype-4 (HCV GT4) in apoptosis. However, this still not fully understood. In the current study a re-constructed clone of E1/E2 polyprotein region of the HCV GT4 was transfected into the Huh7 cell line and a human apoptotic PCR array of 84 genes was used to investigate its possible significance for apoptosis. Out of the 84 genes, only 35 showed significant differential expression, 12 genes being up-regulated and 23 down-regulated. The highest-up regulated genes were APAF1 (apoptotic peptidase-activating factor 1), BID (BH3 interacting domain death agonist) and BCL 10 (B-cell CLL/lymphoma protein 10) with fold regulation of 33.2, 30.1 and 18.9, respectively. The most down-regulated were FAS (TNF receptor super family), TNFRSF10B (tumor necrosis factor receptor super-family member 10b) and FADD (FAS-associated death domain) with fold regulation of -30.2, -27.7 and -14.9, respectively. These results suggest that the E1/E2 proteins may be involved in HCV-induced pathogenesis by modulating apoptosis through the induction of the intrinsic apoptosis pathway and disruption of the BCL2 gene family.

Effects of 8-week Exercise on Bcl-2, Bax, Caspase-8, Caspase-3 and HSP70 in Mouse Gastrocnemius Muscle (8주간 운동이 생쥐의 gastrocnemius에서 Bcl-2, Bax, caspase-8, caspase-3와 HSP70에 미치는 영향)

  • Kim, Ki-Bum;Kim, Yong-An;Park, Jung-Jun
    • Journal of Life Science
    • /
    • v.20 no.9
    • /
    • pp.1409-1414
    • /
    • 2010
  • The aim of this study was to investigate the effects of exercise on intrinsic and extrinsic apoptosis signaling pathways in skeletal muscle. ICR-type white male mice were divided into a control group (CON: n=10) and an exercise training group (EX: n=10) after a 1 week adaptation period. EX performed treadmill running at 16.4 m/min with a 4% incline, 40 min/day and 5 days/week for 8 weeks. Cervical dislocation was performed at 48 hours after the last bout of exercise, after which gastrocnemius skeletal muscles were immediately collected. The results of verifying the intrinsic apoptosis pathway showed that there were no significant differences in Bcl-2, Bax, or the ratio of Bax/Bcl-2 proteins between EX and CON. On the other hand, the results of verifying the extrinsic apoptosis pathway showed that caspase-8 proteins were significantly lower in EX than in CON (p<0.05). Apoptosis suppressing protein HSP70 was higher in EX than in CON. In addition, caspase-3, which is the final factor for apoptosis, was not activated. These results indicate that apoptosis did not develop since caspase-3 is non-cleaved by the effects of caspase-8 and HSP70 extrinsic pathways rather than Bcl-2 and Bax intrinsic pathways among signal pathways for apoptosis.

A non-replicating oncolytic vector as a novel therapeutic tool against cancer

  • Kaneda, Yasufumi
    • BMB Reports
    • /
    • v.43 no.12
    • /
    • pp.773-780
    • /
    • 2010
  • Cancers are still difficult targets despite recent advances in cancer therapy. Due to the heterogeneity of cancer, a single-treatment modality is insufficient for the complete elimination of cancer cells. Therapeutic strategies from various aspects are needed. Gene therapy has been expected to bring a breakthrough to cancer therapy, but it has not yet been successful. Gene therapy also should be combined with other treatments to enhance multiple therapeutic pathways. In this view, gene delivery vector itself should be equipped with intrinsic anti-cancer activities. HVJ (hemagglutinating virus of Japan; Sendai virus) envelope vector (HVJ-E) was developed to deliver therapeutic molecules. HVJ-E itself possessed anti-tumor activities such as the generation of anti-tumor immunities and the induction of cancer-selective apoptosis. In addition to the intrinsic anti-tumor activities, therapeutic molecules incorporated into HVJ-E enabled to achieve multi-modal therapeutic strategies in cancer treatment. Tumor-targeting HVJ-E was also developed. Thus, HVJ-E will be a novel promising tool for cancer treatment.