• Title/Summary/Keyword: non-small-cell lung cancer (H460)

Search Result 27, Processing Time 0.032 seconds

Anticancer effect of mountain ginseng Pharmacopuncture to the nude mouse of lung carcinoma induced by NCI-H460 human non-small cell lung cancer cells

  • Kwon, Ki-Rok
    • Journal of Pharmacopuncture
    • /
    • v.13 no.1
    • /
    • pp.5-14
    • /
    • 2010
  • Objectives : This study was performed to examine the anticancer effect of mountain ginseng Pharmacopuncture(MGP) to the nude mouse of lung carcinoma induced by NCI-H460 human nonsmall lung cancer cells. Methods : Human lung cancer (NCI-H460) cells were cultured and applied to evaluate anti-tumor activity in nude mice. After confirmed tumor growth in mice, MGP was treated per 0.1ml/kg dose to intraperitoneal and intravenous injection everyday for four weeks. And checked the changes in body weights, tumor volume, mean survival time and percent, increase in life span, histo-pathological findings, organ weights, and blood chemistry levels. Results : The results of in vivo study showed that MGP may have potential as growth inhibitor of solid tumor induced NCI-H460 without marked side effects. MGP inhibited dosage-dependently the growth of NCI-H460 cell-transplanted solid tumor compared with the control group. And mean survival time of MGP treated group was prolonged comparing with control group. Generally the group of intravenous injection is more effective than intraperitoneal injection. Conclusion : These results were suggested that MGP may be a useful anticancer agent for therapy of human lung cancer. And follow study need for the certain evidence.

Induction of Apoptosis by Samgibopae-tang in Human Non-small-cell Lung Cancer Cells (인체폐암세포 NCI-H460 및 A549의 apoptosis 유발에 미치는 삼기보배탕의 영향)

  • Heo, Man-Kyu;Heo, Tae-Yool;Kim, Ki-Tak;Byun, Mi-Kwon;Kim, Jin-Young;Sim, Sung-Heum;Kim, Koang-Lock;Kam, Cheol-Woo;Park, Dong-Il
    • The Journal of Internal Korean Medicine
    • /
    • v.28 no.3
    • /
    • pp.473-491
    • /
    • 2007
  • Objectives : This study was designed to investigate the antiproliferative activity of the water extract of Samgibopae-tang (SGBPT) in NCI-H460 and A549 non-small-cell lung cancer cell lines Methods : In this study, we measured the subsistence, form of NCI-H460 and A549 non-small-cell lung cancer cell by hemocytometer and DAPI staining. In each cell, we analyzed DNA fragmentation. reverse transcription-polymerase chain reaction and measured activity of caspase-3, caspase-8 and caspase-9. Results and Conclusions : We found that exposure of A549 cells to SGBPT resulted in growth inhibition in a dose-dependent manner. butSGBPT did not affect the growth of NCI-H460 cells. The antiproliferative effect by SGBPT treatment in A549 cells was associated with morphological changes. SGBPT treatment partially induced the expression of DR5 cells and the expression of Faswas markedly increased in both transcriptional and translational levels in A549 cells. SGBPT treatment partially induced the expression of Bcl-2, Bcl-XL and the expression of Bid was markedly decreased in translational levels in A549 cells. However, SGBPT treatment did not affect the expression of IAP family in A549 orNCI-H460 cells. SGBPT treatment partially induced the expression of caspase-3, caspase-8, caspase-9 activity which markedly increased in a dose-dependent manners in A549 cells. The fragmental development of PARP and ${\beta}$-catenin protein was observed in A549 cells by SGBPT treatment. SGBPT treatment induced the expression of PLC-${\gamma}1$ protein which decreased in A549 cells. SGBPT treatment partially induced the expression of DFF45/ICAD which markedly increased in a dose-dependent manner in A549 cells. Taken together. these findings suggested that SGBPT-induced inhibition of human lung carcinoma did not affect NCI-H460 cell growth. However, SGBPT-induced inhibition of human lung carcinoma A549 cell growth was associated with the induction of death receptor and mitochondrial pathway. The results provided important new insights into the possible molecular mechanisms of the anti-cancer activity of SGBPT.

  • PDF

Anticancer Effect of Activated Natural Killer Cells on Human Non-small Cell Lung Cancer (비소세포성폐암에 대한 자연살해세포의 항암효능)

  • Park, Min-Gyeong;Sung, Hye-Ran;Park, Ji-Sung;Kim, Jee-Youn;Han, Sang-Bae;Lee, Chong-Kil;Yun, Byung-Kui;Song, Suk-Gil
    • YAKHAK HOEJI
    • /
    • v.55 no.3
    • /
    • pp.267-272
    • /
    • 2011
  • Human NK cells, identified 30 years ago based on their ability to spontaneously kill tumor cells, constitute a subset of lymphocytes, which play an important role in the first line of immune defense and the effective function of these cells are enhanced by cytokines. Lung carcinoma has been one of the most commonly diagonosed cancer as well as the leading cause of cancer death in male. Here we provide the evidence that human natural killer cells has inhibitory effects on tumor growth of human lung cancer cell NCI-H460 (non-small cell lung cancer). Enriched NK cell population was obtained by 2 weeks cultivation in interleukin-2(IL-2)-containing medium. The resulting population comprised 26% CD3$^+$ cells, 9% CD3$^+$CD4$^+$ cells, 16% CD3$^+$CD8$^+$ cells, 76% CD56$^+$ cells, 6% CD3$^+$CD56$^+$ cells and 70% CD3$^-$CD56$^+$ cells. Activated NK cells at doese of 2.5, 5, and 10 million cells per mouse inhibited 2%, 12% and 45% of NCI-H460-induced tumor growth in nude mouse xenograft assays, repectively. This result suggests that NK cell-based immunotherapy may be used as an adoptive immunotherapy for lung cancer patients.

Relative Apoptosis-inducing Potential of Homeopathic Condurango 6C and 30C in H460 Lung Cancer Cells In vitro -Apoptosis-induction by homeopathic Condurango in H460 cells-

  • Sikdar, Sourav;Saha, Santu Kumar;Khuda-Bukhsh, Anisur Rahman
    • Journal of Pharmacopuncture
    • /
    • v.17 no.1
    • /
    • pp.59-69
    • /
    • 2014
  • Objectives: In homeopathy, it is claimed that more homeopathically-diluted potencies render more protective/curative effects against any disease condition. Potentized forms of Condurango are used successfully to treat digestive problems, as well as esophageal and stomach cancers. However, the comparative efficacies of Condurango 6C and 30C, one diluted below and one above Avogadro's limit (lacking original drug molecule), respectively, have not been critically analyzed for their cell-killing (apoptosis) efficacy against lung cancer cells in vitro, and signalling cascades have not been studied. Hence, the present study was undertaken. Methods: 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assays were conducted on H460-non-small-cell lung cancer (NSCLC) cells by using a succussed ethyl alcohol vehicle (placebo) as a control. Studies on cellular morphology, cell cycle regulation, generation of reactive oxygen species (ROS), changes in mitochondrial membrane potential (MMP), and DNA-damage were made, and expressions of related signaling markers were studied. The observations were done in a "blinded" manner. Results: Both Condurango 6C and 30C induced apoptosis via cell cycle arrest at subG0/G1 and altered expressions of certain apoptotic markers significantly in H460 cells. The drugs induced oxidative stress through ROS elevation and MMP depolarization at 18-24 hours. These events presumably activated a caspase-3-mediated signalling cascade, as evidenced by reverse transcriptase-polymerase chain reaction (RT-PCR), western blot and immunofluorescence studies at a late phase (48 hours) in which cells were pushed towards apoptosis. Conclusion: Condurango 30C had greater apoptotic effect than Condurango 6C as claimed in the homeopathic doctrine.

Dual Inhibition of PI3K/Akt/mTOR Pathway and Role of Autophagy in Non-Small Cell Lung Cancer Cells

  • Jeong, Eun-Hui;Choi, Hyeong-Sim;Lee, Tae-Gul;Kim, Hye-Ryoun;Kim, Cheol-Hyeon
    • Tuberculosis and Respiratory Diseases
    • /
    • v.72 no.4
    • /
    • pp.343-351
    • /
    • 2012
  • Background: The phosphoinositide 3-kinase (PI3K)/Akt/mammalian target of rapamycin (mTOR) signaling axis has emerged as a novel target for cancer therapy. Agents that inhibit this pathway are currently under development for lung cancer treatment. In the present study, we have tested whether dual inhibition of PI3K/Akt/mTOR signaling can lead to enahnced antitumor effects. We have also examined the role of autophagy during this process. Methods: We analyzed the combination effect of the mTOR inhibitor, temsirolimus, and the Akt inhibitor, GSK690693, on the survival of NCI-H460 and A549 non-small cell lung cancer cells. Cell proliferation was determined by MTT assay and apoptosis induction was evaluated by flow cytometry and terminal deoxynucleotidyl transferase dUTP nick end labeling assay. Autophagy induction was also evaluated by acridine orange staining. Changes of apoptosis or autophagy-related proteins were evaluated by western blot analysis. Results: Combination treatment with temsirolimus and GSK690693 caused synergistically increased cell death in NCI-H460 and A549 cells. This was attributable to increased induction of apoptosis. Caspase 3 activation and poly(ADP-ribose) polymerase cleavage accompanied these findings. Autophagy also increased and inhibition of autophagy resulted in increased cell death, suggesting its cytoprotective role during this process. Conclusion: Taken together, our results suggest that the combination of temsirolimus and GSK690693 could be a novel strategy for lung cancer therapy. Inhibition of autophagy could also be a promising method of enhancing the combination effect of these drugs.

Resistance to Cisplatin Renders High Metastatic Potential in Human Non-Small Cell Lung Cancer Cell Line (Cisplatin 내성을 보이는 비소세포폐암 세포주에서의 전이 능력 증가)

  • 차대원;김진국;손동섭;조대윤;양기민
    • Journal of Chest Surgery
    • /
    • v.34 no.5
    • /
    • pp.377-385
    • /
    • 2001
  • 배경: Cisplain과 같은 세포돗성 약제에 대한 내성은 폐암 치료 실패의 중요한 원인이다. 이러한 항암제에 대한 내성의 발생기전은 복잡하고 아직 완전히 알려져 있지 않지만 불량한 예후의 원인으로 생각된다. 특히 약제 내성이 발생한 환자의 경우 기존의 종양의 급속한 성장뿐 아니라 새로운 전이 병소가 급속히 발생 및 진단됨은 약제 내성을 가진 종양이 전이에의 용이성을 획득하는게 아닌가 의심케한다. 이를 규명하기 위해 Cisplatin에 내성을 지닌 비소세포폐암 세포주 H460/CISm이 전이 능력을 Cisplatin에 민감한 비소세포폐암 세로주 H460과 비교하고자 한다. 대상 및 방법: 약제 내성 세포주를 확보하기 위하여 H460세포에 cisplatin을 점차적으로 증가시켜 처리한 후 배양하였다. H460 세포와 H460/CIS 세로에서의 혈관신생인자와 성장관련인 자의 발현양상, gelatin zymography 분석 그리고 in vivo 실험으로 nude 마우스에서의 자발적 전이 능력의 차이를 비교하였다. 결과: H460 세포를 이식한 마우스에 폐에서는 종양이 형성되지 않았으나 H460/CIS세포를 이식한 마우스 10마리중 8마리에서 종양이 형성되었다. 또한 H460/CIS 세포주에서 전이 관련 유전자로 알려진 angiopoietin-1, vascular endothelial growth factor, basic fibroblast growth factor, matrix metalloproteinase 2 등이 더 발현되었고, 전이의 침습성을 유발하는 gelatinase의 활성이 증가된 것을 확인할 수 있었다. 결론: 본 여구 결과를 통해 cisplatin에 내성을 가진 비소세포폐암세포에서 전이 능력이 증가될 수 있다고 여겨지며 이러한 사실을 토대로 초기 비소세포폐암 환자의 수술 전 항암약물요법의 타당성에 대해서 이야기 하기 위해서는 많은 임상적 연구가 필요하리라 생각된다.

  • PDF

Induction of Apoptosis by Gamisamgibopae-tang in A549 Human Lung Cancer Cells through Modulation of Bcl-2 Family and Activation of Caspases (Bcl-2 family 발현 변화 및 caspases의 활성을 통한 가미삼기보폐탕의 A549 인체폐암세포 apoptosis 유도)

  • Kim, Hyun-Joong;Kim, Hong-Gi;Kim, Jin-Young;Kam, Cheol-Woo;Park, Dong-Il
    • Journal of Physiology & Pathology in Korean Medicine
    • /
    • v.22 no.3
    • /
    • pp.630-641
    • /
    • 2008
  • Gamisamgibopae-tang (GMSGBPT) is a traditional Korean medicine, which has been used for patients suffering from a lung disease in Oriental medicine. In the present study, we examined the biochemical mechanisms of apoptosis by GMSGBPT in NCI-H460 and A549 human non-small-cell lung cancer cell lines. It was found that GMSGBPT could inhibit the cell proliferation of A549 cells in a concentration-dependent manner, however GMSGBPT did not affect the cell proliferation of NCI-H460 cells. Apoptotic cell death in A549 cells were detected using DAPI staining and annexin V fluorescein methods. The induction of apoptotic cell death by GMSGBPT was connected with a down-regulation of anti-apoptotic Bcl-2 and Bcl-xL expression, and proteolytic activation of caspase-3 and caspase-9 in A549 cells. However, GMSGBPT did not affect the levels of pro-apoptotic Bax and Bad expression, and activity of caspase-8. GMSGBPT treatment also concomitant degradation and/or inhibition of poly (ADP-ribose) polymerase (PARP), ${\beta}$-catenin, phospholipase C-1 (PLC${\gamma}$1) and DNA fragmentation factor 45/inhibitor of caspase-activated DNase (DFF45/ICAD). Taken together, these findings suggest that GMSGBPT may be a potential chemotherapeutic agent for the control of human non-small-cell lung cancer cells and further studies will be needed to identify the active compounds that confer the anti-cancer activity of GMSGBPT.

Plumbagin from Plumbago Zeylanica L Induces Apoptosis in Human Non-small Cell Lung Cancer Cell Lines through NF-κB Inactivation

  • Xu, Tong-Peng;Shen, Hua;Liu, Ling-Xiang;Shu, Yong-Qian
    • Asian Pacific Journal of Cancer Prevention
    • /
    • v.14 no.4
    • /
    • pp.2325-2331
    • /
    • 2013
  • Objective: To detect effects of plumbagin on proliferation and apoptosis in non-small cell lung cancer cell lines, and investigate the underlying mechanisms. Materials and Methods: Human non-small cell lung cancer cell lines A549, H292 and H460 were treated with various concentrations of plumbagin. Cell proliferation rates was determined using both cell counting kit-8 (CCK-8) and clonogenic assays. Apoptosis was detected by annexin V/propidium iodide double-labeled flow cytometry and TUNEL assay. The levels of reactive oxygen species (ROS) were detected by flow cytometry. Activity of NF-${\kappa}B$ was examined by electrophoretic mobility shift assay (EMSA) and luciferase reporter assay. Western blotting was used to assess the expression of both NF-${\kappa}B$ regulated apoptotic-related gene and activation of p65 and $I{\kappa}B{\kappa}$. Results: Plumbagin dose-dependently inhibited proliferation of the lung cancer cells. The IC50 values of plumbagin in A549, H292, and H460 cells were 10.3 ${\mu}mol/L$, 7.3 ${\mu}mol/L$, and 6.1 ${\mu}mol/L$ for 12 hours, respectively. The compound concentration-dependently induced apoptosis of the three cell lines. Treatment with plumbagin increased the intracellular level of ROS, and inhibited the activation of NK-${\kappa}B$. In addition to inhibition of NF-${\kappa}B$/p65 nuclear translocation, the compound also suppressed the degradation of $I{\kappa}B{\kappa}$. ROS scavenger NAC highly reversed the effect of plumbagin on apoptosis and inactivation of NK-${\kappa}B$ in H460 cell line. Treatment with plumbagin also increased the activity of caspase-9 and caspase-3, downregulated the expression of Bcl-2, upregulated the expression of Bax, Bak, and CytC. Conclusions: Plumbagin inhibits cell growth and induces apoptosis in human lung cancer cells through an NF-${\kappa}B$-regulated mitochondrial-mediated pathway, involving activation of ROS.

Induction of Apoptosis by Samgibopae-tang in Human Non-small-cell Lung Cancer Cells (인체폐암세포 NCI-H460 및 A549의 증식에 미치는 삼기보폐탕의 영향 비교)

  • Heo, Man-Kyu;Park, Cheol;Choi, Young-Hyun;Kam, Cheol-Woo;Park, Dong-Il
    • Journal of Physiology & Pathology in Korean Medicine
    • /
    • v.21 no.4
    • /
    • pp.973-981
    • /
    • 2007
  • In the present study, we investigated the antiproliferative activity of the water extract of Samgibopae-tang (SGBPT) in NCI-H460 and A549 non-small-cell lung cancer cell lines. We found that exposure of A549 cells to SGBPT resulted in the growth inhibition in a dose-dependent manner as measured by MTT assay, however SGBPT did not affect the growth of NCI-H460 cells. The antiproliferative effect by SGBPT treatment in A549 cells was associated with morphological changes such as membrane shrinking and cell rounding up. SGBPT treatment did not induce the cell cycle arrest in both cell lines, however the frequency of sub-G1 population was concentration-dependently increased by SGBPT treatment in A549 cells. SGBPT treatment partially induced the expression of tumor suppressor p53 in A549 cells and the expression of cyclin-dependent kinase inhibitor p21(WAF1/CIP1) was markedly increased in both transcriptional and translational levels in A549 cells. The up-regulation of p21 by SGBPT occurred in a similar a concentration dependent manner to that observed with the inhibition of cell viability and induction of sub-G1 population of the cell cycle. However SGBPT treatment did not affect other growth regulation-related genes such as early growth response-1 (Egr-1), nonsteroidal anti-inflammatory drug-activated gene-1 (NAG-1), inducible nitric oxide synthease (iNOS), cyclooxygenases (COXs), telomere-regulatory factors in A549 as well as NCI-H460 cells. Taken together, these findings suggested that SGBPT-induced inhibition of human lung carcinoma A549 cell growth was aoosciated with the induction of p21 and the results provided important new insights into the possible molecular mechanisms of the anti-cancer activity of SGBPT.

Enhanced Sensitivity to Gefitinib after Radiation in Non-Small Cell Lung Cancer Cells

  • Choi, Yun-Jung;Rho, Jin-Kyung;Back, Dae-Hyun;Kim, Hye-Ryoun;Lee, Jae-Cheol;Kim, Cheol-Hyeon
    • Tuberculosis and Respiratory Diseases
    • /
    • v.71 no.4
    • /
    • pp.259-265
    • /
    • 2011
  • Background: Epidermal growth factor receptor (EGFR) tyrosine kinase inhibitors, gefitinib and erlotinib, are effective therapies for non-small cell lung cancer (NSCLC) patients whose tumors harbor somatic mutations in EGFR. The mutations are, however, only found in about 30% of Asian NSCLC patients and all patients ultimately develop resistance to these agents. Ionizing radiation has been shown to induce autophosphorylation of EGFR and activate its downstream signaling pathways. In the present study, we have tested whether the effect of gefitinib treatment can be enhanced after ionizing radiation. Methods: We compared the PC-9 and A549 cell line with its radiation-resistant derivatives after gefitinib treatment with cell proliferation and apoptosis assay. We also analyzed the effect of gefitinib after ionizing radiation in PC-9, A549, and NCI-H460 cells. Cell proliferation was determined by MTT assay and induction of apoptosis was evaluated by flow cytometry. Caspase 3 activation and PARP cleavage were evaluated by western blot analysis. Results: PC-9 cells having mutated EGFR and their radiation-resistant cells showed no significant difference in cell viability. However, radiation-resistant A549 cells were more sensitive to gefitinib than were their parental cells. This was attributable to an increased induction of apoptosis. Gefitinib-induced apoptosis increased significantly after radiation in cells with wild type EGFR including A549 and NCI-H460, but not in PC-9 cells with mutated EGFR. Caspase 3 activation and PARP cleavage accompanied these findings. Conclusion: The data suggest that gefitinib-induced apoptosis could increase after radiation in cells with wild type EGFR, but not in cells with mutated EGFR.