• Title/Summary/Keyword: Anti-tumor action

Search Result 183, Processing Time 0.029 seconds

Molecular Mechanisms of Cell Cycle Arrest and Apoptosis by Dideoxypetrosynol A, a Polyacetylene from the Sponge Petrosia sp., in Human Monocytic Leukemia Cells

  • Choi, Yung Hyun
    • Journal of Marine Bioscience and Biotechnology
    • /
    • v.1 no.4
    • /
    • pp.243-251
    • /
    • 2006
  • Dideoxypetrosynol A, a polyacetylene from the marine sponge Petrosia sp., is known to exhibit significant selective cytotoxic activity against a small panel of human tumor cell lines, however, the mechanisms of which are poorly understood. In the present study, it was investigated the further possible mechanisms by which dideoxytetrosynol A exerts its anti-proliferative action in cultured human leukemia cell line U937. We observed that the proliferation-inhibitory effect of dideoxypetrosynol A was due to the induction of G1 arrest of the cell cycle and apoptosis, which effects were associated with up-regulation of cyclin D1 and down-regulation of cyclin E without any change in cyclin-dependent-kinases (Cdks) expression. Dideoxypetrosynol A markedly induced the levels of Cdk inhibitor p16/INK4a expression. Furthermore, down-regulation of phosphorylation of retinoblastoma protein (pRB) by this compound was associated with enhanced binding of pRB and the transcription factor E2F-1. The increase in apoptosis was associated with a dose-dependent up-regulation in pro-apoptotic Bax expression and activation of caspase-3 and caspase-9. Dideoxytetrosynol A decreased the levels of cyclooxygenase (COX)-2 mRNA and protein expression without significant changes in the levels of COX-1, which was correlated with a decrease in prostaglandin E2 (PGE2) synthesis. Furthermore, dideoxytetrosynol A treatment markedly inhibited the activity of telomerase, and the expression of human telomerase reverse transcriptase (hTERT), a main determinant of the telomerase enzymatic activity, was progressively down-regulated by dideoxytetrosynol A treatment in a dose-dependent fashion. Taken together, these findings provide important new insights into the possible molecular mechanisms of the anti-cancer activity of dideoxytetrosynol A.

  • PDF

Modulation of Sarcodon Aspratus on lon Currents-induced by Excitatory Neurotransmitters in Rat Periaqueductal Gray Neurons

  • Kim, Sung-Tae;Sung, Yun-Hee;Kim, Chang-Ju;Joo, Kwan-Joong;Han, Seung-Ho;Lee, Choong-Yeol;Kim, Youn-Sub
    • Journal of Physiology & Pathology in Korean Medicine
    • /
    • v.20 no.6
    • /
    • pp.1672-1677
    • /
    • 2006
  • Sarcodon aspratus is the mushroom of Telephoracea which was been classified into Alphllophorales. The aqueous extract of Sarcodon aspratus in known to have anti-tumor activity, immune modulatory effect, and anti-oxidative action. The descending pain control system consists of three major components: the periaqueductal gray (PAG) of the midbrain, the rostroventral medulla including the nucleus raphe magnus, and the spinal dorsal horn. Glutamate is the primary excitatory neurotransmitter in the brain. Glutamate ionotropic receptors are classified as N-methyl-D-aspartate (NMDA) receptor, ${\alpha}$-amino-3-hydroxy-5-methyl-4-isoxazolepropionate (AMPA) receptor, and kainate receptor. In the present study, the modulation of Sarcodon aspratus on the ion currents activated by glutamate, NMDA, AMPA, and kainate in the acutely dissociated PAG neurons was investigated by nystatin-perforated patch-clamp technique under boltage-clamp condition. Sarcodon aspratus increased glutamate- and NMDA-induced ion currents were not increased by Sarcodon aspratus. The present results show that Sarcodon aspratus may activate the descending pain control system in rat PAG neurons through NMDA receptor.

Inhibitory Effects of Chestnut Inner Shell Cytokine Production from Human Mast Cell Line (율피추출물의 사람 비만세포주 사이토카인 발현 억제 효과)

  • Jun, Dong-Ha;Jang, Young-Ah;Kim, Hui-Yeong;Kim, Su-Jin;Kim, Jin-Chul;Kim, Sea-Hyun;Kwoen, Dae-Jun;Han, Sang-Ik;Lee, Jin-Tae
    • The Korea Journal of Herbology
    • /
    • v.28 no.2
    • /
    • pp.55-60
    • /
    • 2013
  • Objectives : Atopic dermatitis (AD) is a chronic inflammatory skin disease characterized by eczematous inflammtion of the skin. The chestnut inner shell extracts (CI) has been used as a cosmetic material for a long time in Korea. However, the precise anti-allergy effects of CI have yet to be clearly elucidated. In this study, we attempted to evaluate the effect of CI on mast cell-mediated allergy inflammation. Methods : To find the anti-allergy and inflammatory effect of CI, we investigated the inhibitory effect of CI on the production of inflammatory mediators using by enzyme-linked immunosorbent assay in phorbol 12-myristate 13-acetate (PMA) plus calcium ionophore (A23187) stimulated-human mast cell (HMC-1). Results : In this study, we found that CI did not show cytotoxic effect at up to 10 ug/ml on HMC-1. CI inhibited the production of tumor necrosis factor (TNF)-${\alpha}$, interleukin (IL)-6 and IL-8 in stimulated HMC-1. Maximal rate of TNF-${\alpha}$, IL-6 and IL-8 inhibition by CI (10 ug/ml) were about 47.6%, 44.1% and 22.5% respectively. In addition, we showed that Fr.3 isolated from n-Butyl alcohol layer of CI attenuated the production of TNF-${\alpha}$, IL-6 and IL-8 in HMC-1. Conclusion : Taken together, the findings of this study provide us with a novel insight action of CI as a potential molecule for use in the treatment of allergic inflammation diseases.

Sanghuangporus sanghuang extract inhibits the proliferation and invasion of lung cancer cells in vitro and in vivo

  • Weike Wang;Jiling Song;Na Lu;Jing Yan;Guanping Chen
    • Nutrition Research and Practice
    • /
    • v.17 no.6
    • /
    • pp.1070-1083
    • /
    • 2023
  • BACKGROUND/OBJECTIVES: Sanghuangporus sanghuang (SS) has various medicinal effects, including anti-inflammation and anticancer activities. Despite the extensive research on SS, its molecular mechanisms of action on lung cancer are unclear. This study examined the impact of an SS alcohol extract (SAE) on lung cancer using in vitro and in vivo models. MATERIALS/METHODS: Different concentrations of SAE were used to culture lung cancer cells (A549 and H1650). A cell counting kit-8 assay was used to detect the survival ability of A549 and H1650 cells. A scratch assay and transwell cell invasion assay were used to detect the migration rate and invasive ability of SAE. Western blot analysis was used to detect the expression of B-cell lymphoma-2 (Bcl-2), Bcl2-associated X (Bax), cyclin D1, cyclin-dependent kinases 4 (CDK4), signal transducer and activator of transcription 3 (STAT3), and phosphorylated STAT3 (p-STAT3). Lung cancer xenograft mice were used to detect the inhibiting ability of SAE in vivo. Hematoxylin and eosin staining and immunohistochemistry were used to detect the effect of SAE on the structural changes to the tumor and the expression of Bcl-2, Bax, cyclin D1, CDK4, STAT3, and p-STAT3 in lung cancer xenograft mice. RESULTS: SAE could inhibit lung cancer proliferation significantly in vitro and in vivo without cytotoxicity. SAE suppressed the viability, migration, and invasion of lung cancer cells in a dose and time-dependent manner. The SAE treatment significantly decreased the proapoptotic Bcl-2/Bax ratio and the expression of pro-proliferative proteins Cyclin D1 and CDK4 in vitro and in vivo. Furthermore, SAE also inhibited STAT3 expression. CONCLUSIONS: SAE reduced the cell viability and suppressed cell migration and invasion in human lung cancer cells. Moreover, SAE also exhibited anti-proliferation effects in vivo. Therefore, SAE may have benefits in cancer therapy.

Anti-inflammatory effect of (-)-epigallocatechin-3-gallate on Porphyromonas gingivalis lipopolysaccharide-stimulated fibroblasts and stem cells derived from human periodontal ligament

  • Jung, Im-Hee;Lee, Dong-Eun;Yun, Jeong-Ho;Cho, Ah-Ran;Kim, Chang-Sung;You, Yoon-Jeong;Kim, Sung-Jo;Choi, Seong-Ho
    • Journal of Periodontal and Implant Science
    • /
    • v.42 no.6
    • /
    • pp.185-195
    • /
    • 2012
  • Purpose: (-)-epigallocatechin-3-gallate (EGCG) has been reported to exert anti-inflammatory and antibacterial effects in periodontitis. However, its exact mechanism of action has yet to be determined. The present in vitro study evaluated the anti-in-flammatory effects of EGCG on human periodontal ligament fibroblasts (hPDLFs) and human periodontal ligament stem cells (hPDLSCs) affected by bacterial lipopolysaccharide (LPS) extracted from Porphyromonas gingivalis. Methods: hPDLFs and hPDLSCs were extracted from healthy young adults and were treated with EGCG and/or P. gingivalis LPS. After 1, 3, 5, and 7 days from treatment, cytotoxic and proliferative effects were evaluated using a 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assay and bromodeoxyuridine assay, respectively. And then, the gene expressions of hPDLFs and hPDLSCs were observed for interleukin (IL)-$1{\beta}$, IL-6, tumor necrosis factor (TNF)-${\alpha}$, osteoprotegerin (OPG), receptor activator of nuclear factor kappa-B ligand (RANKL), and RANKL/OPG using real-time polymerase chain reaction (PCR) at 0, 6, 24, and 48 hours after treatment. The experiments were performed with the following groups for hPDLFs and hPDLSCs; 1) No treat, 2) EGCG alone, 3) P. gingivalis LPS alone, 4) EGCG+P. gingivalis LPS. Results: The 20 ${\mu}M$ of EGCG and 20 ${\mu}g/mL$ of P. gingivalis LPS had the lowest cytotoxic effects, so those concentrations were used for further experiments. The proliferations of hPDLFs and hPDLSCs increased in all groups, though the 'EGCG alone' showed less increase. In real-time PCR, the hPDLFs and hPDLSCs of 'EGCG alone' showed similar gene expressions to those cells of 'no treat'. The gene expressions of 'P. gingivalis LPS alone' in both hPDLFs and hPDLSCs were highly increased at 6 hours for IL-$1{\beta}$, IL-6, TNF-${\alpha}$, RANKL, and RANKL/OPG, except the RANKL/OPG in hPDLSCs. However, those increased gene expressions were down-regulated in 'EGCG+P. gingivalis LPS' by the additional treatment of EGCG. Conclusions: Our results demonstrate that EGCG could exert an anti-inflammatory effect in hPDLFs and hPDLSCs against a major pathogen of periodontitis, P. gingivalis LPS.

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

  • Lee, Hye Hyeon;Jeong, Jin-Woo;Choi, Yung Hyun
    • Journal of Life Science
    • /
    • v.26 no.7
    • /
    • pp.847-854
    • /
    • 2016
  • Cordycepin, a derivative of the nucleoside adenosine, is one of the active components extracted from fungi of genus Cordyceps, and has been shown to have many pharmacological activities. In this study, we investigated the effects of cordycepin on proliferation and apoptosis of human gastric cancer AGS cells, and its possible mechanism of action. Treatment of cordycepin resulted in significant decrease in cell viability of AGS cells in a concentration-dependent manner. A concentration-dependent apoptotic cell death was also measured by agarose gel electrophoresis and flow cytometery analysis. Molecular mechanistic studies of apoptosis unraveled cordycepin treatment resulted in an enhanced expression of tumor necrosis factor-related apoptosis-inducing ligand, death receptor 5 and Fas ligand. Furthermore, up-regulation of pro-apoptotic Bax, and down-regulation of anti-apoptotic Bcl-2 and Bcl-xL expression were also observed in cordycepin-treated AGS cells. These were followed by activation of caspases (caspase-9, -8 and -3), subsequently leading to poly (ADP-ribose) polymerase cleavage. Taken together, these findings indicate that cordycepin induces apoptosis in AGS cells through regulation of multiple apoptotic pathways, including death receptor and mitochondria. Although further mechanical studies are needed, our results revealed that cordycepin can be regarded as a new effective and chemopreventive compound for human gastric cancer treatment.

Anti-proliferation Effects of Isorhamnetin on Lung Cancer Cells in Vitro and in Vivo

  • Li, Qiong;Ren, Fu-Qiang;Yang, Chun-Lei;Zhou, Li-Ming;Liu, Yan-You;Xiao, Jing;Zhu, Ling;Wang, Zhen-Grong
    • Asian Pacific Journal of Cancer Prevention
    • /
    • v.16 no.7
    • /
    • pp.3035-3042
    • /
    • 2015
  • Background: Isorhamnetin (Iso), a novel and essential monomer derived from total flavones of Hippophae rhamnoides that has long been used as a traditional Chinese medicine for angina pectoris and acute myocardial infarction, has also shown a spectrum of antitumor activity. However, little is known about the mechanisms of action Iso on cancer cells. Objectives: To investigate the effects of Iso on A549 lung cancer cells and underlying mechanisms. Materials and Methods: A549 cells were treated with $10{\sim}320{\mu}g/ml$ Iso. Their morphological and cellular characteristics were assessed by light and electronic microscopy. Growth inhibition was analyzed by MTT, clonogenic and growth curve assays. Apoptotic characteristics of cells were determined by flow cytometry (FCM), DNA fragmentation, single cell gel electrophoresis (comet) assay, immunocytochemistry and terminal deoxynucleotidyl transferase nick end labeling (TUNEL). Tumor models were setup by transplanting Lewis lung carcinoma cells into C57BL/6 mice, and the weights and sizes of tumors were measured. Results: Iso markedly inhibited the growth of A549 cells with induction of apoptotic changes. Iso at $20{\mu}g/ml$, could induce A549 cell apoptosis, up-regulate the expression of apoptosis genes Bax, Caspase-3 and P53, and down-regulate the expression of Bcl-2, cyclinD1 and PCNA protein. The tumors in tumor-bearing mice treated with Iso were significantly smaller than in the control group. The results of apoptosis-related genes, PCNA, cyclinD1 and other protein expression levels of transplanted Lewis cells were the same as those of A549 cells in vitro. Conclusions: Iso, a natural single compound isolated from total flavones, has antiproliferative activity against lung cancer in vitro and in vivo. Its mechanisms of action may involve apoptosis of cells induced by down-regulation of oncogenes and up-regulation of apoptotic genes.

Tumor Suppressor Protein p53 Promotes 2-Methoxyestradiol-Induced Activation of Bak and Bax, Leading to Mitochondria-Dependent Apoptosis in Human Colon Cancer HCT116 Cells

  • Lee, Ji Young;Jee, Su Bean;Park, Won Young;Choi, Yu Jin;Kim, Bokyung;Kim, Yoon Hee;Jun, Do Youn;Kim, Young Ho
    • Journal of Microbiology and Biotechnology
    • /
    • v.24 no.12
    • /
    • pp.1654-1663
    • /
    • 2014
  • To examine the effect of tumor suppressor protein p53 on the antitumor activity of 2-methoxyestradiol (2-MeO-$E_2$), 2-MeO-$E_2$-induced cell cycle changes and apoptotic events were compared between the human colon carcinoma cell lines HCT116 ($p53^{+/+}$) and HCT116 ($p53^{-/-}$). When both cell types were exposed to 2-MeO-$E_2$, a reduction in the cell viability and an enhancement in the proportions of $G_2/M$ cells and apoptotic sub-$G_1$ cells commonly occurred dose-dependently. These 2-MeO-$E_2$-induced cellular changes, except for $G_2/M$ arrest, appeared to be more apparent in the presence of p53. Immunofluorescence microscopic analysis using anti-${\alpha}$-tubulin and anti-lamin B2 antibodies revealed that after 2-MeO-$E_2$ treatment, impaired mitotic spindle network and prometaphase arrest occurred similarly in both cell types. Following 2-MeO-$E_2$ treatment, only HCT116 ($p53^{+/+}$) cells exhibited an enhancement in the levels of p53, p-p53 (Ser-15), $p21^{WAF1/CIP1}$, and Bax; however, the Bak level remained relatively constant in both cell types, and the Bcl-2 level decreased only in HCT116 ($p53^{+/+}$) cells. Additionally, mitochondrial apoptotic events, including the activation of Bak and Bax, loss of ${\Delta}{\psi}m$, activation of caspase-9 and -3, and cleavage of lamin A/C, were more dominantly induced in the presence of p53. The Bak-specific and Bax-specific siRNA approaches confirmed the necessity of both Bak and Bax activations for the 2-MeO-$E_2$-induced apoptosis in HCT116 cells. These results show that among 2-MeO-$E_2$-induced apoptotic events, including prometaphase arrest, up-regulation of Bax level, down-regulation of Bcl-2 level, activation of both Bak and Bax, and mitochondria-dependent caspase activation, the modulation of Bax and Bcl-2 levels is the target of the pro-apoptotic action of p53.

Bee Venom-induced Growth Inhibition of Human Lung Cancer Cells was Associated with Inhibition of Prostagladin E2 Production and Telomerase Activity. (인체폐암세포에서 봉독에 의한 prostagladin E2 생성 및 telomerase 활성 저하)

  • Kim, Jong-Hwan;Hwang, Won-Deuk;Kim, Byung-Woo;Choi, Yung-Hyun
    • Journal of Life Science
    • /
    • v.19 no.4
    • /
    • pp.502-507
    • /
    • 2009
  • In modern oriental medicine, bee venom therapy is being used for aqua-acupuncture to relieve pain and to cure inflammatory diseases such as rheumatoid arthritis, osteoarthritis, and gout. Bee venom therapy has been processed and reported in many experimental studies, with regard to its effects on pain alleviation, anti-inflammation, removal of fever, anti-convulsion, suppression of tumor and immunity strengthening, etc., however, its mechanism of action, molecular targeting on prostaglandin $E_2$ ($PGE_2$) production and telomere length regulation in human cancer remains unclear. In this study, we investigated the effect of bee venom on the levels of cyclooxygenases (COXs) and telomere regulatory components of A549 human lung cancer cells. Bee venom-induced anti-proliferative effects of A549 cells were associated with the inhibition of human telomerase reverse transcriptase (hTERT) as well as human telomerase RNA (hTR), transcription factor c-myc and the activity of telomerase. In addition, bee venom treatment markedly decreased the levels of COX-2 mRNA and protein expression without significant changes in the expression of COX-1, which was correlated with a decrease in $PGE_2$ synthesis. Taken together, these findings provide important new insights into the possible molecular mechanisms of the anti-cancer activity of bee venom.

Anti-inflammaory effects of the MeOH extract of Humulus japonicus in vivo (율초(葎草)가 항염 효과에 미치는 영향)

  • Hwang, Sun-Yi;Jo, Mi-Jeong;Kim, Sang-Chan;Jee, Seon-Young
    • The Journal of Korean Medicine Ophthalmology and Otolaryngology and Dermatology
    • /
    • v.22 no.2
    • /
    • pp.92-103
    • /
    • 2009
  • Objectives : The present study was examined to evaluate the anti-inflammatory effects of the Humulus japonicus MeOH extracts (HJE) in vivo. Methods : The effects of HJE on anti-inflammation were measured by production of NO, iNOS (inducible Nitric Oxide Synthase), COX-2, I$\kappa$B$\alpha$ (Inhibitor kappa B alpha), NF$\kappa$B (Nuclear Factor kappa B), TNF-$\alpha$ (Tumor Necrosis Factor-alpha) and IL-1$\beta$ (Interleukin-1$\beta$), IL-6 in Raw 264.7 macrophage cells stimulated with LPS. Results : 1. All concentrations of HJE(0.03 and 0.10 mg/ml) had no significant cytotoxicity in Raw 264.7 cell during the entire experimental period. 2. The level of NO and iNOS in culture medium was dramatically increased by LPS application. However, these increases were dose-dependently(0.03 and 0.10 mg/ml) attenuated by treatment with HJE. 3. HJE extract reduced PGE2 levels in a dose-dependent manner as a consequence of inhibition of COX-2 protein expression in Raw 264.7 macrophage cells stimulated with LPS. 4. 0.10 mg/ml HJE significantly inhibited the phosphorylation of I$\kappa$B$\alpha$ indicating the suppression of NF-$\kappa$B pathway in Raw 264.7 macrophage cells stimulated with LPS. 5. 0.10 mg/ml HJE significantly inhibited the production of TNF-$\alpha$ in Raw 264.7 macrophage cells stimulated with LPS. 6. All concentrations of HJE significantly inhibited the production of IL-1$\beta$, IL-6 in Raw 264.7 macrophage cells stimulated with LPS. Conclusions : These results provide evidences that therapeutic effect of HJE on heat syndrome, especially due to the acute inflammation, are partly due to the reduction of some of inflammatory factors by inhibiting iNOS and COX-2 through the suppression of p-I$\kappa$B$\alpha$. Moreover, it suggests that the mechanism of action of HJE comes from the suppression of inflammatory mediators, such as NO, PGE$_2$ and pro-inflammatory cytokines.

  • PDF