• Title/Summary/Keyword: Lung cancer cells

Search Result 998, Processing Time 0.031 seconds

Antitumor profiles and cardiac electrophysiological effects of aurora kinase inhibitor ZM447439

  • Lee, Hyang-Ae;Kwon, Miso;Kim, Hyeon-A;Kim, Ki-Suk
    • The Korean Journal of Physiology and Pharmacology
    • /
    • v.23 no.5
    • /
    • pp.393-402
    • /
    • 2019
  • Aurora kinases inhibitors, including ZM447439 (ZM), which suppress cell division, have attracted a great deal of attention as potential novel anti-cancer drugs. Several recent studies have confirmed the anti-cancer effects of ZM in various cancer cell lines. However, there have been no studies regarding the cardiac safety of this agent. We performed several cytotoxicity, invasion and migration assays to examine the anti-cancer effects of ZM. To evaluate the potential effects of ZM on cardiac repolarisation, whole-cell patch-clamp experiments were performed with human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) and cells with heterogeneous cardiac ion channel expression. We also conducted a contractility assay with rat ventricular myocytes to determine the effects of ZM on myocardial contraction and/or relaxation. In tests to determine in vitro efficacy, ZM inhibited the proliferation of A549, H1299 (lung cancer), MCF-7 (breast cancer) and HepG2 (hepatoma) cell lines with $IC_{50}$ in the submicromolar range, and attenuated the invasive and metastatic capacity of A549 cells. In cardiac toxicity testing, ZM did not significantly affect $I_{Na}$, $I_{Ks}$ or $I_{K1}$, but decreased $I_{hERG}$ in a dose-dependent manner ($IC_{50}$: $6.53{\mu}M$). In action potential (AP) assay using hiPSC-CMs, ZM did not induce any changes in AP parameters up to $3{\mu}M$, but it at $10{\mu}M$ induced prolongation of AP duration. In summary, ZM showed potent broad-spectrum anti-tumor activity, but relatively low levels of cardiac side effects compared to the effective doses to tumor. Therefore, ZM has a potential to be a candidate as an anti-cancer with low cardiac toxicity.

Suppressor of Variegation 3-9 Homolog 2, a Novel Binding Protein of Translationally Controlled Tumor Protein, Regulates Cancer Cell Proliferation

  • Kim, A-Reum;Sung, Jee Young;Rho, Seung Bae;Kim, Yong-Nyun;Yoon, Kyungsil
    • Biomolecules & Therapeutics
    • /
    • v.27 no.2
    • /
    • pp.231-239
    • /
    • 2019
  • Suppressor of Variegation 3-9 Homolog 2 (SUV39H2) methylates the lysine 9 residue of histone H3 and induces heterochromatin formation, resulting in transcriptional repression or silencing of target genes. SUV39H1 and SUV39H2 have a role in embryonic development, and SUV39H1 was shown to suppress cell cycle progression associated with Rb. However, the function of human SUV39H2 has not been extensively studied. We observed that forced expression of SUV39H2 decreased cell proliferation by inducing $G_1$ cell cycle arrest. In addition, SUV39H2 was degraded through the ubiquitin-proteasomal pathway. Using yeast two-hybrid screening to address the degradation mechanism and function of SUV39H2, we identified translationally controlled tumor protein (TCTP) as an SUV39H2-interacting molecule. Mapping of the interacting regions indicated that the N-terminal 60 amino acids (aa) of full-length SUV39H2 and the C-terminus of TCTP (120-172 aa) were critical for binding. The interaction of SUV39H2 and TCTP was further confirmed by co-immunoprecipitation and immunofluorescence staining for colocalization. Moreover, depletion of TCTP by RNAi led to up-regulation of SUV39H2 protein, while TCTP overexpression reduced SUV39H2 protein level. The half-life of SUV39H2 protein was significantly extended upon TCTP depletion. These results clearly indicate that TCTP negatively regulates the expression of SUV39H2 post-translationally. Furthermore, SUV39H2 induced apoptotic cell death in TCTP-knockdown cells. Taken together, we identified SUV39H2, as a novel target protein of TCTP and demonstrated that SUV39H2 regulates cell proliferation of lung cancer cells.

Expression of COX-2 and IDO by Uteroglobin Transduction in NSCLC Cell Lines (비소세포폐암 세포주에서 Uteroglobin Transduction이 COX-2 및 IDO의 발현에 미치는 영향)

  • Park, Gun Min;Lee, Sang-Min;Yim, Jae-Joon;Yang, Seok-Chul;Yoo, Chul Gyu;Lee, Choon-Taek;Han, Sung Koo;Sim, Young-Soo;Kim, Young Whan
    • Tuberculosis and Respiratory Diseases
    • /
    • v.66 no.4
    • /
    • pp.274-279
    • /
    • 2009
  • Background: Uteroglobin (UG) is a secretary protein that has strong immunomodulatory properties, and which is synthesized in most epithelia including lung tissue. Overexpression of UG is associated with decreased expression of cyclooxygenase (COX)-2 and suppression of cancer cell growth. Indoleamine 2,3-dioxygenase (IDO) catalyzes tryptophan along the kynurenine pathway, and both the reduction in local tryptophan and the production of tryptophan metabolites contribute to the immunosuppressive effects of IDO. Methods: In this study, we investigated the pattern of expression of COX-2 and IDO, and the effect of UG transduction in the expression of COX-2 and IDO in several non-small cell lung cancer cell lines, especially A549. Results: Both COX-2 and IDO were constitutionally expressed in A549 and H460 cells, and was reduced by UG transduction. In A549 cells, the slightly increased expression of COX-2 and IDO with the instillation of interferon-gamma (IFN-$\gamma$) was reduced by UG transduction. However, the reduced expression of COX-2 and IDO by UG transduction was not increased with IFN-$\gamma$ instillation in A549 cells. In both the A549 COX-2 sense and the A549 COX-2 anti-sense small interfering RNA (siRNA)-transfected cells, IDO was expressed; expression was reduced by UG transduction, irrespective of the expression of COX-2. Conclusion: The results suggest that the anti-proliferative function of UG may be associated with the immune tolerance pathway of IDO, which is independent of the COX-2 pathway.

Expression of CD40, CD86, and HLA-DR in CD1c+ Myeloid Dendritic Cells Isolated from Peripheral Blood in Primary Adenocarcinoma of Lung (원발성 폐선암환자의 말초혈액에서 분리한 CD1c+ 골수성 수지상 세포에서의 CD40, CD86 및 HLA-DR의 발현)

  • Kang, Moon-Chul;Kang, Chang-Hyun;Kim, Young-Tae;Kim, Joo-Hyun
    • Journal of Chest Surgery
    • /
    • v.43 no.5
    • /
    • pp.499-505
    • /
    • 2010
  • Background: There have been several reports using animal experiments that CD1-restricted T-cells have a key role in tumor immunity. To address this issue, we studied the expression of markers for CD1c+ myeloid dendritic cells (DCs) isolated from peripheral blood in the clinical setting. Material and Method: A total of 24 patients with radiologically suspected or histologically confirmed lung cancer who underwent pulmonary resection were enrolled in this study. The patients were divided according to histology findings into three groups: primary adenocarcinoma of lung (PACL), primary squamous cell carcinoma of lung (PSqCL) and benign lung disease (BLD). We obtained 20 mL of peripheral venous blood from patients using heparin-coated syringes. Using flow-cytometry after labeling with monoclonal antibodies, data acquisition and analysis were done. Result: The ratio of CD1c+CD19- dendritic cells to CD1c+ dendritic cells were not significantly different between the three groups. CD40 (p=0.171), CD86 (p=0.037) and HLA-DR (p=0.036) were less expressed in the PACL than the BLD group. Expression of CD40 (p=0.319), CD86 (p=0.036) and HLA-DR (p=0.085) were less expressed in the PACL than the PSqCL group, but the differences were only significant for CD86. Expression of co-stimulatory markers was not different between the PSqCL and BLD groups. Expression of markers for activated DCs were dramatically lower in the PACL group than in groups with other histology (CD40 (p=0.005), CD86 (p=0.013) HLA-DR (p=0.004). Conclusion: These results suggest the possibility that CD1c+ myeloid DCs participate in control of the tumor immunity system and that low expression of markers results in lack of an immune response triggered by dendritic cells in adenocarcinoma of the lung.

EFFECT OF RED GINSENG ON NATURAL KILLER CELL ACTIVITY IN MICE WITH LUNG ADENOMA INDUCED BY URETHAN AND BENZO(A)PYRENE (홍삼이 Urethan 및 Benzo(a)pyrene에 의하여 폐선종이 유발된 마우스에서 Natural Killer 세포활성도에 미치는 영향)

  • Yun Yeon-Sook;Jo Sung-Kee;Moon Hae-Sun;Kim Young-Ju;Oh Yeong-Ran;Yun Taik-Koo
    • Proceedings of the Ginseng society Conference
    • /
    • 1984.09a
    • /
    • pp.27-36
    • /
    • 1984
  • It was previously reported that red ginseng extract inhibited carcinogenesis by urethan, DMBA and aflatoxin $B_1E (Cancer Detection and Prevention, 6: 515-525, 1983). In an attempt to investigate the mechanism of the anticarcinogenic effect of ginseng, we assayed natural killer (N.K) activity in mice treated with urethan and benzo(a)pyrene. In our experiment newly born Swiss Webster mice, less than 24 hrs. old, were given a single subcutaneous injection of lmg of ure-than and 40ug of benzo(a)pyrene. The mice had been administered with ginseng since weaning, and sacrificed at various intervals. Major organs were examined both, with the naked eye and microscopically. N.K. activity of spleen cells was analyzed in a 12-hour $^{51}Cr^-release$ assay against YAC-1 cells. Administration of ginseng resulted in an increase of N.K. activity by $18\%$ at 4 weeks, $20\%$ (P < 0.05) at 6, $29\%$ (P < 0.05) at 12, and $13\%$ at 24 following a single injection of urethan. At the same time, significantly lower incidences of lung adenoma were noted at 6 weeks $(50\%)$ and 12 weeks $(27\%)$ following the administration of ginseng to urethan-injected mice. This result indicates that the enhancement of N.K. activity by ginseng makes a contribution to its anticarcinogenic effect. On the hand, N.K. activity was suppressed by benzo(a)pyrene during the time span of this experiment and it almost returned to the level of controls following the adminsitration of ginseng. However, the lung adenoma induced by benzo(a)pyrene began to occur at 48 weeks in which N.K. activity had naturally declined to a very low level in all experimental mice, and administration of ginseng did not decrease the incidence. In explanation of this result, we might propose that the recovery of the N.K. activity by ginseng had little effect on the incidence of lung adenoma because of the long latent period of carcinogenesis by benzo(a)pyrene. In conclusion, these results suggest that the anticarcinogenic effect of ginseng in urethan-treated mice may be related to the augmentation of N.K. activity.

  • PDF

Implications of NQO1 in cancer therapy

  • Oh, Eun-Taex;Park, Heon Joo
    • BMB Reports
    • /
    • v.48 no.11
    • /
    • pp.609-617
    • /
    • 2015
  • NAD(P)H:quinone oxidoreductase (NQO1), an obligatory two-electron reductase, is a ubiquitous cytosolic enzyme that catalyzes the reduction of quinone substrates. The NQO1- mediated two-electron reduction of quinones can be either chemoprotection/detoxification or a chemotherapeutic response, depending on the target quinones. When toxic quinones are reduced by NQO1, they are conjugated with glutathione or glucuronic acid and excreted from the cells. Based on this protective effect of NQO1, the use of dietary compounds to induce the expression of NQO1 has emerged as a promising strategy for cancer prevention. On the other hand, NQO1-mediated two-electron reduction converts certain quinone compounds (such as mitomycin C, E09, RH1 and β-lapachone) to cytotoxic agents, leading to cell death. It has been known that NQO1 is expressed at high levels in numerous human cancers, including breast, colon, cervix, lung, and pancreas, as compared with normal tissues. This implies that tumors can be preferentially damaged relative to normal tissue by cytotoxic quinone drugs. Importantly, NQO1 has been shown to stabilize many proteins, including p53 and p33ING1b, by inhibiting their proteasomal degradation. This review will summarize the biological roles of NQO1 in cancer, with emphasis on recent findings and the potential of NQO1 as a therapeutic target for the cancer therapy.

Anti-cancer Activities of Ginseng Extract Fermented with Phellinus linteus

  • Lee, Jong-Jin;Kwon, Ho-Kyun;Jung, In-Ho;Cho, Yong-Baik;Kim, Kyu-Joong;Kim, Jong-Lae
    • Mycobiology
    • /
    • v.37 no.1
    • /
    • pp.21-27
    • /
    • 2009
  • In the present study, the anti-cancer effects of ginseng fermented with Phellinus linteus (GFPL) extract were examined through in vitro and in vivo assays. GFPL was produced by co-cultivating ginseng and Phellinus linteus together. Ginsenoside Rg3, Rh1 and Rh2 are important mediators of anti-angiogenesis and their levels in GFPL were enriched 24, 19 and 16 times, respectively, more than that of ginseng itself through the fermentation. GFPL exhibited distinct anti-cancer effects, including growth inhibition of the human lung carcinoma cell line A549, and promotion of immune activation by stimulating nitric oxide (NO) production in Raw 264.7 cells. Further evidence supporting anti-cancer effects of GFPL was its significant prolongment of the survival of B16F10 cancer cell-implanted mice. These results suggest that the GFPL may be a candidate for cancer prevention and treatment through immune activation and anti-angiogenic effects by enriching Rg3, Rh1 and Rh2.

Spontaneous Lymphosarcoma in a Formosan Sika Deer (Cervus nippon taiouanus) (꽃사슴에서 자연발생한 림프육종)

  • Han Jeong-hee;Jin Hee-kyung;Kim Jong-taek
    • Journal of Veterinary Clinics
    • /
    • v.11 no.2
    • /
    • pp.573-578
    • /
    • 1994
  • A lymphosarcoma was diagnosed in a 4-year-old female Formosan sika deer presented with persistent reproductive failure, anorexia, depression and diarrhea. Characteristic pathological findings were infiltration of neoplastic lymphoid cells and cancer emboli in the lymph nodes, heart, lung, kidney, urinary bladder, ovary, uterus and peritoneal fat tissue.

  • PDF