• Title/Summary/Keyword: Anti-tumor

Search Result 2,595, Processing Time 0.03 seconds

Antitumor and Anti-metastatic Effect of Mineral Powder (광물석분의 항암 및 항전이 효과에 관한 연구)

  • Song Gyu Yong;Bae Ki Whan;Myung Chang-Seon;Jin Wen Yi;Park Yong Jin;Yun Mi Young;Kim Dong Hee
    • Journal of Physiology & Pathology in Korean Medicine
    • /
    • v.19 no.4
    • /
    • pp.1011-1015
    • /
    • 2005
  • Antitumor and anti-metastatic effects of mineral powder(MP) were studied. In the present study, MP did not exhibit the any cytotoxic activity against leukemic cells such as L1210 and U937 tumor cell lines in vitro. Also, MP did not exhibit the any cytotoxic activity against solid cells such as A549 and B16-BL6 tumor cell lines in vitro. However, in vivo, MP exhibit a significant antitumor activity in BDF1 mice bearing Lewis lung carcinoma cells(LLC) with inhibition rates of 46 and $23\%$ at 200 and 100 mg/kg/day, respectively. Furthermore, in pulmonary colonization assay, MP exhibit the inhibitory effect of tumor metastasis. From these results, it was concluded that MP had antitumor and anti-metastatic activity suggesting its application for the prevention and treatment of cancer.

Ginsenoside Rp1 Inhibits Proliferation and Migration of Human Lung Cancer Cells

  • Hong, Sam-Yeol;Cho, Jae-Youl;Seo, Dong-Wan
    • Biomolecules & Therapeutics
    • /
    • v.19 no.4
    • /
    • pp.411-418
    • /
    • 2011
  • Ginsenoside Rp1 (G-Rp1) is a novel ginseng saponin derivative with anti-tumor activity. However, the biochemical and molecular mechanisms of G-Rp1 on anti-tumor activity are not fully understood. In the present study, we report that G-Rp1 inhibits lung cancer cell proliferation, migration and adhesion in p53 wild-type A549 and p53-defi cient H1299 cells. Anti-proliferative activity of G-Rp1 in lung cancer cells is mediated by enhanced nuclear localization of cyclin-dependent kinase inhibitors including $p27^{Kip1}$ and $p21^{WAF1/Cip1}$, and subsequent inhibition of pRb phosphorylation. We also show that these anti-tumor activities of G-Rp1 in both A549 and H1299 cells appear to be mediated by suppression of mitogenic signaling pathways such as ERK, Akt and $p70^{S6K}$. Taken together, these findings suggest further development and evaluation of G-Rp1 for the treatment of lung cancers with mutated p53 as well as wild-type p53.

Isolation of a Quinone-rich Fraction from Ardisia crispa Roots and its Attenuating Effects on Murine Skin Tumorigenesis

  • Yeong, Looi Ting;Hamid, Roslida Abdul;Yazan, Latifah Saiful;Khaza'ai, Huzwah
    • Asian Pacific Journal of Cancer Prevention
    • /
    • v.14 no.4
    • /
    • pp.2301-2305
    • /
    • 2013
  • Ardisia crispa (Family: Myrsinaceae) is an evergreen, fruiting shrub that has been traditionally used as folklore medicine. Despite a scarcity of research publications, we have succeeded in showing suppressive effects on murine skin papillomagenesis. In extension, the present research was aimed at determining the effect of a quinone-rich fraction (QRF) isolated from the same root hexane extract on both initiation and promotion stages of carcinogenesis, at the selected dose of 30 mg/kg. Mice (groups I-IV) were initiated with a single dose of 7,12-dimethylbenz(${\alpha}$)anthracene (DMBA, $100{\mu}g/100{\mu}l$) followed by repeated promotion of croton oil (1%) twice weekly for 20 weeks. In addition, group I (anti-initiation) received QRF 7 days before and after DMBA; group II (anti-promotion) received QRF 30 minutes before each croton oil application; group III (anti-initiation/promotion) was treated with QRF as a combination of group I and II. A further two groups served as vehicle control (group V) and treated control (group VI). As carcinogen control, group IV showed the highest tumor volume ($8.79{\pm}5.44$) and tumor burden ($3.60{\pm}1.17$). Comparatively, group III revealed only 20% of tumor incidence, tumor burden ($3.00{\pm}1.00$) and tumor volume ($2.40{\pm}1.12$), which were significantly different from group IV. Group II also showed significant reduction of tumor volume (3.11), tumor burden (3.00) and tumor incidence (11.11%), along with prominent increase of latency period of tumor formation (week 12). Group I, nonetheless, demonstrated marked increment of tumor incidence by 40% with prompted latency period of tumor formation (week 7). No tumor formation was observed in groups V and VI. This study provided clear evidence of inhibitory effects of QRF during promotion period which was in agreement with our previous findings. The mechanism(s) underlying such effects have yet to be elucidated.

Identification of CM1 as a Pathogenic Factor in Inflammatory Diseases and Cancer

  • Bae, Se-Yeon;Kim, Hyem-In;Yu, Yeon-Sil;Lee, Na-Eun;Kong, Joo-Myoung;Kim, Hang-Rae;Hwang, Young-Il;Song, Yeong-Wook;Kang, Jae-Seung;Lee, Wang-Jae
    • IMMUNE NETWORK
    • /
    • v.11 no.3
    • /
    • pp.175-181
    • /
    • 2011
  • Background: CM1 (centrocyte/-blast marker 1) was defined by a mAb against concavabalin-A (ConA) activated PBMC. It is expressed in germinal center of human tonsil and on the surface of activated PBMC as well as cancer cells. Recently, increased productions of pro-inflammatory mediators were detected from activated PBMC by CM1 ligation. Methods: However, there is a limitation to explain the exact role of CM1 on inflammation and its related mechanisms, since the identity of CM1 is still not clarified. In our previous study, we have already confirmed that soluble form of CM1 was produced by Raji. Therefore, we performed Q-TOF analysis after immunoprecipitation of concentrated Raji culture supernatant using anti-CM1 mAbs. Results: As a result, we found that CM1 is identical to enolase-1(ENO1), a glycolytic enzyme, and we confirmed that results by silencing ENO1 using siRNA. It was also confirmed through competition assay between anti-CM1 and anti-ENO1 mAbs. Finally, we investigated the possible role of CM1 in inflammatory response and cancer. The ligation of CM1 on Raji cells with anti-CM1 mAbs induces the extensive production of prostaglandin $E_2(PGE_2)$. In addition, the increased activity of matrix metalloproteinase (MMP)-2/9 was shown in NCI-N87, stomach cancer cell line by CM1 stimulation. Conclusion: CM1 is identical to ENO1 and it might be an important role in the regulation of inflammatory responses.

CM1 Ligation Induces Apoptosis via Fas-FasL Interaction in Ramos Cells, but via Down-regulation of Bcl-2 and Subsequent Decrease of Mitochondrial Membrane Potential in Raji Cells

  • Lee, Young-Sun;Kim, Yeong-Seok;Kim, Dae-Jin;Hur, Dae-Young;Kang, Jae-Seung;Kim, Young-In;Hahm, Eun-Sil;Cho, Dae-Ho;Hwang, Young-Il;Lee, Wang-Jae
    • IMMUNE NETWORK
    • /
    • v.6 no.2
    • /
    • pp.59-66
    • /
    • 2006
  • Background: CM1 (Centrocyte/-blast Marker I) defined by a mAb developed against concanavalin-A activated PBMC, is expressed specifically on a subpopulation of centroblasts and centrocytes of human germinal center (GC) B cells. Burkitt lymphoma (BL) is a tumor consisting of tumor cells with the characteristics of GC B cell. Previously we reported that CM1 ligation with anti-CM1 mAb induced apoptosis in Ramos $(IgM^{high})$ and Raji $(IgM^{low})$ cells. Methods & Results: In the present study, we observed that CM1 ligation with anti-CM1 mAb induced Fas ligand and Fas expression in Ramos cells, but not in Raji cells. Furthermore, anti-Fas blocking antibody, ZB4, blocked CM1-mediated apoptosis effectively in Ramos cells, but not in Raji cells. Increased mitochondrial membrane permeabilization, which was measured by $DiOC_6$, was observed only in Raji cells. In contrast to no significant change of Bax known as pro-apoptotic protein, anti-apoptotic protein Bcl-2 was significantly decreased in Raji cells. In addition, we observed that CM1 ligation increased release of mitochondrial cytochrome c and upregulated caspase-9 activity in Raji cells. Conclusion: These results suggest that apoptosis induced by CM1-ligation is mediated by Fas-Fas ligand interaction in Ramos cells, whereas apoptosis is mediated by down-regulation of Bcl-2 and subsequent decrease of mitochondrial membrane potential in Raji cells.

Co-expression of Survivin and Bcl-2 in Primary Brain Tumors : Their Potential Effect on Anti-apoptosis

  • Ryu, Je-Il;Kim, Choong-Hyun;Cheong, Jin-Hwan;Bak, Koang-Hum;Kim, Jae-Min;Oh, Suck-Jun
    • Journal of Korean Neurosurgical Society
    • /
    • v.40 no.1
    • /
    • pp.1-5
    • /
    • 2006
  • Objective : Survivin is an inhibitor of apoptosis protein[IAP], which inhibits apoptosis through a pathway distinct from the Bcl-2 family members. Overexpression of survivin and Bcl-2 have been commonly reported in human neoplasms. The authors investigate whether there is a synergistic effect on the anti-apoptosis rate of primary brain tumors "in situ" based on the co-expression of survivin and Bcl-2. Methods : One hundred and two brain tumor patients who had been resected were included in this study. Survivin tin and Bcl-2 were detected by Western blotting analysis, while apoptosis was examined by DNA fragmentation analysis. An anti-apoptotic rate was assessed in these brain tumor samples based on the expression of survivin and Bcl-2 or co-expression of both. Results : Survivin and Bcl-2 were expressed in 57[55.9%] and 53[52.0%] of 102 brain tumor samples studied respectively, and co-expressed in 31[30.4%]. The percentage of astrocytic and meningeal tumors expressing survivin was significantly correlated with histological grades; however, Bcl-2 was not correlated [p=0.106]. The anti-apoptotic rate in primary brain tumors with survivin, Bcl-2, and both was detected in 49[86.0%] of 57 samples, 42[79.9%] of 53 samples, and 27[87.1%] of 31 samples, respectively. Their difference in the frequency of anti-apoptosis was not significant. Conclusion : Survivin or Bcl-2 is involved in the anti-apoptosis. However, it suggests that co-expression of survivin and Bcl-2, together, have no synergistic effect on the anti-apoptotic properties of the primary brain tumors.

Biological activities and single oral dose toxicity in rat of fermented Rhus verniciflua extract (발효 옻 추출물의 생리활성 및 단회 경구 투여 독성시험)

  • Choi, Myung-Jin;Lee, Seung-Jin;Jang, Seung-Hee;Reza, Md. Ahsanur;Hong, Joo-Heon;Jung, Hee-Kyoun;Park, Seung-Chun
    • Korean Journal of Veterinary Research
    • /
    • v.50 no.3
    • /
    • pp.187-195
    • /
    • 2010
  • In this study, we investigated the biological activities such as anti-tumor, anti-oxidant and antiinflammatory activities as well as single oral dose toxicity of fermented Rhus verniciflua extract (FRVE). In order to examine anti-tumor activity of FRVE, the sarcoma 180 cells were treated with FRVE at various concentrations (0.03, 0.3, 3 and 30 mg/mL) in microtetrazolium (MTT) assay. In MTT assay, all the cells treated with FRVE at various concentrations have shown a significant difference compared with control (p < 0.05). In xanthine oxidase inhibition assay to examine the antioxidant activity, the xanthine oxidase inhibition rate of FRVE at 1.5 mg/mL and 15 mg/mL was $85{\pm}15.01%$ and $99{\pm}16.02%$, respectively. Nitric oxide production in RAW 264.7 cells showed that FRVE showed a significant anti-inflammation effect at 3 mg/mL (p < 0.05). In single oral dose toxicity study, no differences were observed between control and treated groups in clinical signs, body weight gains, feed and water consumptions. The results indicated that lethal dose 50 ($LD_{50}$) of FRVE was found to be higher than 5,000 mg/kg in this experiment. From the above results, we may suggest that FRVE might have useful as a material for functional food and/or animal pharmaceutics.

Physico-chemical Properties and In Vivo Anti-cancer Effects of Potato Kimchi Prepared by adding Hot Water Extracts of Potato (생감자의 열탕 추출물을 첨가한 감자 김치의 이화학적 특성 및 In Vivo에서의 항암 효과)

  • Chang, Sang-Keun;Kim, Hee-Joo
    • Journal of the East Asian Society of Dietary Life
    • /
    • v.18 no.3
    • /
    • pp.302-310
    • /
    • 2008
  • In this study, potato kimchi was prepared by applying heat to raw potatoes, and then the physico-chemical properties and anti-cancer effects of the kimchi were analyzed. The texture results indicated the potato kimchi had very good hardness and springiness attributes. During th late storage period, total vitamin C content of the kimchi slowly increased. In addition, the potato kimchi had non-volatile organic acid changes that promoted early aging; however, after the complete aging period, it was comparatively similar to other types of kimchi. Using the methanol extracts of various kimchi samples, the potato kimchi(solid 100%) showed the highest anti-carcinogenic effects in terms of anti-tumor activity in tumor bearing Balb/c mice with sarcoma-180 cells. In addition, the effects of the methanol extracts on hepatic glutathione S-transferase content were $289.76\;{\mu}mol/mg$ protein/min, $250.97\;{\mu}mol/mg$ protein/min, $251.20\;{\mu}mol/mg$ protein/min, $219.53\;{\mu}mol/mg$ protein/min, $183.79\;{\mu}mol/mg$ protein/min, for control kimchi, mul kimchi, and two potato kimchis [(solid 100%) and(solid 60%+kimchi juice 40%)], respectively. The in vivo anti-cancer effects of the potato kimchi were investigated using AGS human gastric adenocarcionoma cells and HT-29 human colon adenocarcionoma cells. Overall, an MTT assay revealed that the methanol extract of the potato kimchi showed the highest anti-carcinogenic effects.

  • PDF

Improving Combination Cancer Therapy by Acetaminophen and Romidepsin in Non-small Cell Lung Cancer Cells

  • Lee, Seong-Min;Park, James S.;Kim, Keun-Sik
    • Biomedical Science Letters
    • /
    • v.25 no.4
    • /
    • pp.293-301
    • /
    • 2019
  • Combination chemotherapy is more effective than mono-chemotherapy and is widely used in clinical practice for enhanced cancer treatment. In this study, we investigated the potential synergistic effects of acetaminophen, a common component in many cold medicines, and romidepsin, a histone deacetylase (HDAC) inhibitor, in the A549 non-small cell lung cancer (NSCLC) cell line. The combination of acetaminophen and romidepsin also exerted significant cytotoxicity and apoptosis induced by activation of caspase-3 on tumor cells in vitro. Moreover, combination therapy significantly induced increased production of chemokines that stimulate migration of activated T-cells into tumor cells. This mechanism can lead to active T-cell mediated anti-tumor immunity in addition to the direct cytotoxic chemotherapeutic effect. Activated T-cells led to enhanced cytotoxicity in drug-treated A549 cells through interaction with tumor cells. These results suggested that the interaction between the two drugs is synergistic and significant. In conclusion, our data showed that the use of romidepsin and low concentrations acetaminophen could induce effective anti-tumor effects via enhanced tumor immune and direct cytotoxic chemotherapeutic responses. The combination of acetaminophen with romidepsin should be considered as a promising strategy for the treatment of lung cancer.

Nrf2 in TIME: The Emerging Role of Nuclear Factor Erythroid 2-Related Factor 2 in the Tumor Immune Microenvironment

  • Jialin Feng;Oliver J. Read;Albena T. Dinkova-Kostova
    • Molecules and Cells
    • /
    • v.46 no.3
    • /
    • pp.142-152
    • /
    • 2023
  • Nuclear factor erythroid 2-related factor 2 (Nrf2) mediates the cellular antioxidant response, allowing adaptation and survival under conditions of oxidative, electrophilic and inflammatory stress, and has a role in metabolism, inflammation and immunity. Activation of Nrf2 provides broad and long-lasting cytoprotection, and is often hijacked by cancer cells, allowing their survival under unfavorable conditions. Moreover, Nrf2 activation in established human tumors is associated with resistance to chemo-, radio-, and immunotherapies. In addition to cancer cells, Nrf2 activation can also occur in tumor-associated macrophages (TAMs) and facilitate an anti-inflammatory, immunosuppressive tumor immune microenvironment (TIME). Several cancer cell-derived metabolites, such as itaconate, L-kynurenine, lactic acid and hyaluronic acid, play an important role in modulating the TIME and tumor-TAMs crosstalk, and have been shown to activate Nrf2. The effects of Nrf2 in TIME are context-depended, and involve multiple mechanisms, including suppression of proinflammatory cytokines, increased expression of programmed cell death ligand 1 (PD-L1), macrophage colony-stimulating factor (M-CSF) and kynureninase, accelerated catabolism of cytotoxic labile heme, and facilitating the metabolic adaptation of TAMs. This understanding presents both challenges and opportunities for strategic targeting of Nrf2 in cancer.