首页 | 本学科首页   官方微博 | 高级检索  
相似文献
 共查询到18条相似文献,搜索用时 156 毫秒
1.
摘要目的:研究内毒素对体外培养非小细胞肺癌(NSCLC)细胞株A549 细胞增殖的影响及其机制。方法:不同浓度脂多糖(LPS) 进行8-48h 干预,MTT 及细胞计数法检测其对A549 细胞增殖的影响;EGFR中和抗体或COX-2 抑制剂与LPS联合干预,检测其 对A549 细胞增殖及PGE2 的影响。结果:LPS 可引发A549 细胞MTT 活性和细胞计数显著增加,且呈现时间和剂量依赖性。LPS 还可诱发PGE2 水平显著升高。药物干预结果显示,抑制COX-2 或EGFR 可明显逆转LPS 所引发的细胞增殖和PGE2 水平升高 趋势。结论:LPS 可能通过激活EGFR 和COX-2 信号途径,诱导体外培养的非小细胞肺癌细胞增殖分化。肺部感染可能会加速非 小细胞肺癌进展,并可能造成不良预后。  相似文献   

2.
《生命科学研究》2016,(5):418-423
紫铆因抗多种肿瘤的生物学活性已被广泛研究,但其抑制非小细胞肺癌(non-small cell lung cancer,NSCLC)的分子机制还不清楚。研究中,通过MTS和软琼脂集落实验检测紫铆因对非小细胞肺癌细胞停泊依赖和停泊非依赖增殖的影响,利用免疫印迹检测紫铆因处理后非小细胞肺癌表皮生长因子受体(epidermal growth factor receptor,EGFR)信号通路的磷酸化状态,同时采用流式细胞术检测紫铆因对非小细胞肺癌细胞周期演进的影响。研究发现紫铆因剂量依赖性抑制A549和H1650细胞增殖,在抑制EGFR信号通路磷酸化活化的同时下调EGFR总蛋白及EGFR在胞膜和胞核的表达,而且紫铆因下调AuroraA/B和H3-S10磷酸化,促进A549细胞G2/M期阻滞。结果表明紫铆因可通过靶向EGFR和AuroraA/B信号通路抑制非小细胞肺癌。  相似文献   

3.
目的:研究表没食子儿茶素-3-没食子酸酯(epigallocatechin-3-gallate,EGCG)对炎性刺激的人肺腺癌A549细胞增殖和凋亡的影响及与CUGBP1表达的关系。方法:MTT法检测EGCG和LPS刺激A549细胞增殖活性的影响;流式细胞仪检测细胞凋亡;免疫细胞化学检测EGCG对LPS刺激人肺腺癌A549细胞内CUGBP1蛋白的表达。结果:与对照组相比,LPS体外显著促进A549细胞增殖,其胞核胞质内CUGBP1表达明显增强(P〈0.01)。加入EGCG可拮抗LPS促A549细胞增殖的作用,促进其凋亡,明显抑制LPS刺激的A549细胞内CUGBP1的表达(P〈0.01)。CUGBP1蛋白定量分析可知EGCG和LPS共同孵育A549细胞4h、24h时,细胞中的CUGBP1蛋白表达量较单纯LPS作用时降低。但EGCG和LPS共同孵育A549细胞24h,A549细胞中胞核CUGBP1蛋白表达量(1210.565±3.46)较4h时胞核CUGBP1蛋白表达量(67.344±3.68)高,差异有统计学意义(t=927.164,P〈0.001)。结论:EGCG可能通过干扰CUGBP1基因的表达抑制炎症刺激人肺腺癌细胞A549的增殖,促进其凋亡。  相似文献   

4.
目的:探讨褪黑素(MLT)对体外培养的肺腺癌A549细胞增殖的影响及作用机制。方法:体外培养人肺腺癌A549细胞,通过不同浓度的褪黑素(0.1、1.0、2.5、5.0mmol/L)干预24、48、72h,通过噻唑蓝(MTT)法测定细胞增殖,DNA末端原位标记染色法(Tunel)检测细胞凋亡情况,蛋白印迹(Western-blot)法检测褪黑素对A549细胞核内核转录因子Bp65(NF-Bp65)蛋白水平的影响。结果:褪黑素能够抑制人肺腺癌A549细胞增殖,呈剂量、时间依赖性性;高浓度褪黑素作用后凋亡细胞比例明显升高,同时细胞核内NF-Bp65蛋白量明显减少。结论:褪黑素能够呈剂量、时间依赖性抑制人肺腺癌A549细胞的增殖,抑制核因子Bp65的核移位诱导细胞凋亡是可能作用路径之一。  相似文献   

5.
目的:研究表没食子儿茶素-3-没食子酸酯(epigallocatechin-3-gallate,EGCG)对炎性刺激的人肺腺癌A549细胞增殖和凋亡的影响及与CUGBP1表达的关系。方法:MTT法检测EGCG和LPS刺激A549细胞增殖活性的影响;流式细胞仪检测细胞凋亡;免疫细胞化学检测EGCG对LPS刺激人肺腺癌A549细胞内CUGBP1蛋白的表达。结果:与对照组相比,LPS体外显著促进A549细胞增殖,其胞核胞质内CUGBP1表达明显增强(P0.01)。加入EGCG可拮抗LPS促A549细胞增殖的作用,促进其凋亡,明显抑制LPS刺激的A549细胞内CUGBP1的表达(P0.01)。CUGBP1蛋白定量分析可知EGCG和LPS共同孵育A549细胞4h、24h时,细胞中的CUGBP1蛋白表达量较单纯LPS作用时降低。但EGCG和LPS共同孵育A549细胞24h,A549细胞中胞核CUGBP1蛋白表达量(1210.565±3.46)较4h时胞核CUGBP1蛋白表达量(67.344±3.68)高,差异有统计学意义(t=927.164,P0.001)。结论:EGCG可能通过干扰CUGBP1基因的表达抑制炎症刺激人肺腺癌细胞A549的增殖,促进其凋亡。  相似文献   

6.
该文研究了重组蛋白r Lj-112对人非小细胞肺癌A549细胞增殖、迁移和凋亡的影响及其作用机制。采用MTT(四甲基偶氮唑蓝)法检测r Lj-112对人非小细胞肺癌A549细胞增殖的影响,采用Transwell方法检测r Lj-112对A549细胞迁移和侵袭的影响,Hoechst和吉姆萨染色的方法检测r Lj-112对A549细胞凋亡的影响;采用Western blot法检测r Lj-112对A549细胞信号通路相关蛋白质水平的影响。结果表明,r Lj-112能够抑制A549细胞的增殖,半抑制浓度IC50值为1.64μmol/L;r Lj-112能抑制A549细胞的迁移和侵袭并呈剂量依赖性;r Lj-112能诱导A549细胞的凋亡;r Lj-112处理过的细胞cleaved-caspase3蛋白质和cleaved-PARP蛋白质水平升高,证明r Lj-112通过caspase3/PARP途径执行对A549细胞的凋亡的诱导,p-AKT、p-PI3K和p-Erk1/2的水平下降,提示r Lj-112的作用方式与PI3K/AKT相关信号通路有关。  相似文献   

7.
该研究探讨了IQGAP1(IQ domain GTPase-activating protein 1)对非小细胞肺癌(nonsmall cell lung cancer,NSCLC)细胞增殖的影响及其对ERK信号通路的调节作用。将内源性IQGAP1低表达的A549细胞中分为空白组、空载组和IQGAP1过表达组;将内源性IQGAP1高表达的H1299细胞中分为空白组、阴性对照si RNA组和IQGAP1 si RNA组;采用ERK1/2磷酸化抑制剂U0126处理上述两株细胞。MTT法检测细胞增殖能力,Western blot法检测ERK1/2和p-ERK1/2蛋白质水平。结果显示,在A549细胞中,过表达IQGAP1能促进细胞增殖并促进ERK1/2磷酸化;在H1299中,敲低IQGAP1表达能够抑制细胞增殖并下调ERK1/2磷酸化水平。用U0126处理后能抑制IQGAP1对细胞增殖的促进作用。研究结果表明,IQGAP1可通过ERK信号通路促进体外非小细胞肺癌细胞增殖。  相似文献   

8.
目的:研究RGD肽对肺癌A549细胞增殖凋亡及侵袭迁移的影响,并探讨其作用机制。方法:不同浓度RGD肽处理肺癌A549细胞后,MTT检测肺癌细胞的增殖能力,流式细胞仪检测肺癌细胞凋亡及周期分布,Transwell检测其迁移及侵袭能力的变化,Western blot检测RGD肽对肺癌A549细胞MMP2、MMP9的表达水平影响。结果:当RGD肽浓度增加至50 mg/L时,肺癌A549细胞增殖明显受到抑制,且这种抑制作用呈剂量依赖关系;RGD肽组A549细胞G0/G1期细胞比例增高,细胞凋亡率由(6.1±0.1)%增至(15.2±0.5)%;在迁移和侵袭试验中,RGD肽组A549细胞的穿膜细胞数分别由123±10和43±10降至45±5和18±5;RGD肽组A549细胞MMP2、MMP9表达水平显著降低。结论:RGD肽对肺癌A549细胞的增殖有明显抑制作用,并促进其凋亡,可能与RGD肽改变其周期分布有关,RGD肽可明显抑制A549细胞的迁移及侵袭,可能与其下调MMP2、MMP9的表达相关。  相似文献   

9.
目的:探讨组蛋白去乙酰化酶抑制剂曲古霉素A(trichostatin A,TSA)对人非小细胞肺癌(NSCLC)A549细胞增殖抑制作用及机制.方法:以不同剂量TSA(0.1μM,0.5pM和1μM)处理A549细胞.MTT法检测细胞增殖情况,碘化丙啶(PI)染色结合流式细胞仪检测细胞周期,Westem blot法检测P21蛋白表达,流式细胞仪检测细胞线粒体膜电位和细胞凋亡.结果:TSA剂量依赖性抑制肺癌A549细胞增殖,表现为细胞周期阻滞于G2/M期,同时P21蛋白表达增高;此外,TSA还可以剂量依赖性的促进A549细胞凋亡,伴有线粒体膜电位下降.结论:TSA促进NSCLCA549细胞周期阻滞和凋亡,从而抑制其增殖.  相似文献   

10.
为探究三种沙棘总黄酮(TFH)对非小细胞肺癌A549细胞增殖及迁移的影响,并探讨其分子作用机制,选择不同浓度的西藏沙棘(Hippophae tibetana Schlecht)、中国沙棘(H.rhamnoides L.subsp.sinensis Rousi)、肋果沙棘(H.neurocarpa)总黄酮作用于A549细胞。通过MTT检测细胞相对活力,平板克隆形成实验及软琼脂克隆形成实验检测细胞克隆形成能力,流式细胞仪AnnexinV/PI双染法检测细胞凋亡比例。选择效果最佳的西藏沙棘总黄酮应用细胞划痕实验及Transwell实验分析该化合物对肺癌细胞迁移侵袭能力的影响。Western blot检测MMP9、E-cadherin等侵袭迁移相关蛋白表达。敲低E-cadherin基因检测沙棘总黄酮对细胞迁移能力的影响。结果显示,三种沙棘总黄酮均对A549细胞系具有增殖抑制作用,抑制作用依次为:西藏沙棘中国沙棘肋果沙棘。西藏沙棘总黄酮可显著性抑制非小细胞肺癌A549细胞侵袭迁移能力(P0.05)。实验组中MMP9、MMP2、TGF-β、N-cadherin表达水平显著降低,E-cadherin表达水平上调。我们发现在A549细胞中敲低E-cadherin,西藏沙棘总黄酮可逆转迁移增加。以上研究表明西藏沙棘总黄酮对肺癌A549增殖抑制作用具有明显的优势,且西藏沙棘总黄酮可明显抑制肺癌A549细胞的侵袭迁移能力,并可能与下调细胞中的TGF-β抑制MMP9表达并阻止肺癌EMT有关。  相似文献   

11.
Lung cancer is frequently complicated by pulmonary infections which may impair prognosis of this disease. Therefore, we investigated the effect of bacterial lipopolysaccharides (LPS) on tumor proliferation in vitro in the non-small cell lung cancer (NSCLC) cell line A549, ex vivo in a tissue culture model using human NSCLC specimens and in vivo in the A549 adenocarcinoma mouse model. LPS induced a time- and dose-dependent increase in proliferation of A549 cells as quantified by MTS activity and cell counting. In parallel, an increased expression of the proliferation marker Ki-67 and cyclooxygenase (COX)-2 was detected both in A549 cells and in ex vivo human NSCLC tissue. Large amounts of COX-2-derived prostaglandin (PG)E2 were secreted from LPS-stimulated A549 cells. Pharmacological interventions revealed that the proliferative effect of LPS was dependent on CD14 and Toll-like receptor (TLR)4. Moreover, blocking of the epidermal growth factor receptor (EGFR) also decreased LPS-induced proliferation of A549 cells. Inhibition of COX-2 activity in A549 cells severely attenuated both PGE2 release and proliferation in response to LPS. Synthesis of PGE2 was also reduced by inhibiting CD14, TLR4 and EGFR in A549 cells. The proliferative effect of LPS on A549 cells could be reproduced in the A549 adenocarcinoma mouse model with enhancement of tumor growth and Ki-67 expression in implanted tumors. In summary, LPS induces proliferation of NSCLC cells in vitro, ex vivo in human NSCLC specimen and in vivo in a mouse model of NSCLC. Pulmonary infection may thus directly induce tumor progression in NSCLC.  相似文献   

12.
Non-small cell lung carcinoma (NSCLC) accounts for most of all lung cancers, which is the leading cause of mortality in human beings. High level of cyclooxygenase-2 (COX-2) is one of the features of NSCLC and related to the low survival rate of NSCLC. However, whether extracellular nucleotides releasing from stressed resident tissues contributes to the expression of COX-2 remains unclear. Here, we showed that stimulation of A549 cells by adenosine 5'-O-(3-thiotriphosphate) (ATPγS) led to an increase in COX-2 gene expression and prostaglandin E(2) (PGE(2)) synthesis, revealed by Western blotting, RT-PCR, promoter assay, and enzyme-linked immunosorbent assay. In addition, ATPγS induced intracellular reactive oxygen species (ROS) generation through the activation of NADPH oxidase. The increase of ROS level resulted in activation of the c-Src/epidermal growth factor receptor (EGFR)/phosphatidylinositol 3-kinase (PI3K)/protein kinase B (Akt)/nuclear factor (NF)-κB cascade. We also found that activated Akt was translocated into the nucleus and recruited with NF-κB and p300 to form a complex. Thus, activation of p300 modulated the acetylation of histone H4 via the NADPH oxidase/c-Src/EGFR/PI3K/Akt/NF-κB cascade stimulated by ATPγS. Our results are the first to show a novel role of NADPH oxidase-dependent Akt/p65/p300 complex formation that plays a key role in regulating COX-2/PGE(2) expression in ATPγS-treated A549 cells. Taken together, we demonstrated that ATPγS stimulated activation of NADPH oxidase, resulting in generation of ROS, which then activated the downstream c-Src/EGFR/PI3K/Akt/NF-κB/p300 cascade to regulate the expression of COX-2 and synthesis of PGE(2) in A549 cells. Understanding the regulation of COX-2 expression and PGE(2) release by ATPγS on A549 cells may provide potential therapeutic targets of NSCLC.  相似文献   

13.
We investigated the formation and pharmacology of prostaglandin E(3) (PGE(3)) derived from fish oil eicosapentaenoic acid (EPA) in human lung cancer A549 cells. Exposure of A549 cells to EPA resulted in the rapid formation and export of PGE(3.) The extracellular ratio of PGE(3) to PGE(2) increased from 0.08 in control cells to 0.8 in cells exposed to EPA within 48 h. Incubation of EPA with cloned ovine or human recombinant cyclooxygenase 2 (COX-2) resulted in 13- and 18-fold greater formation of PGE(3), respectively, than that produced by COX-1. Exposure of A549 cells to 1 microM PGE(3) inhibited cell proliferation by 37.1% (P < 0.05). Exposure of normal human bronchial epithelial (NHBE) cells to PGE(3), however, had no effect. When A549 cells were exposed to EPA (25 microM) or a combination of EPA and celecoxib (a selective COX-2 inhibitor), the inhibitory effect of EPA on the growth of A549 cells was reversed by the presence of celecoxib (at both 5 and 10 microM). This effect appears to be associated with a 50% reduction of PGE(3) formation in cells treated with a combination of EPA and celecoxib compared with cells exposed to EPA alone. These data indicate that exposure of lung cancer cells to EPA results in a decrease in the COX-2-mediated formation of PGE(2), an increase in the level of PGE(3), and PGE(3)-mediated inhibition of tumor cell proliferation.  相似文献   

14.
目的:在二氧化硅(SiO2)刺激下可引起肺部一系列的炎症反应及其伴随相关的成纤维细胞增殖,然而EGFR信号通路可维持细胞增殖、分化和凋亡的平衡,因此,我们可以设想EGFR信号通路是否在肺纤维化的发生发展中起到重要的作用。本实验探讨SiO2是否能诱导人肺上皮细胞(A549)发生上皮间质转化,并且研究EGFR信号通路在矽肺纤维化中的作用机制。方法:以A549为研究对象,用0(对照组)、50、100、200μg/mlSiO2孵育A549,作用48h后于倒置显微镜观察细胞形态学改变,并收集不同时段细胞,采用实时荧光定量PCR(RT-PCR)检测E-钙黏蛋白(E-cadherin)和α-平滑肌肌动蛋白(α-SMA)mRNA表达变化,细胞免疫荧光方法检测E-cadllerin、α-SMA及信号转导蛋白EGFR表达的变化。结果:倒置显微镜观察A549经SiO2处理后细胞形态由鹅卵石状转变为纺锤型或梭型,形态似成纤维细胞,随着SiO2浓度的升高,E-cadmRNA和蛋白表达逐渐下调,在200μg/ml组表达最低,α-SMAmRNA和蛋白表达逐渐上调,200μg/ml组α-SMA表达最高;EGFR蛋白表达上调;50、100、200μg/ml与对照组的差异具有统计学学意义(P〈0.05)。结论:SiO2可诱导肺上皮细胞向间质细胞转化,其机制可能与EGFR信号通路有关。  相似文献   

15.
Blockade of epidermal growth factor receptor (EGFR) activity has been a primary therapeutic target for non-small cell lung cancers (NSCLC). As patients with wild-type EGFR have demonstrated only modest benefit from EGFR tyrosine kinase inhibitors (TKIs), there is a need for additional therapeutic approaches in patients with wild-type EGFR. As a key component of downstream integrin signalling and known receptor cross-talk with EGFR, we hypothesized that targeting focal adhesion kinase (FAK) activity, which has also been shown to correlate with aggressive stage in NSCLC, would lead to enhanced activity of EGFR TKIs. As such, EGFR TKI-resistant NSCLC cells (A549, H1299, H1975) were treated with the EGFR TKI erlotinib and FAK inhibitors (PF-573,228 or PF-562,271) both as single agents and in combination. We determined cell viability, apoptosis and 3-dimensional growth in vitro and assessed tumor growth in vivo. Treatment of EGFR TKI-resistant NSCLC cells with FAK inhibitor alone effectively inhibited cell viability in all cell lines tested; however, its use in combination with the EGFR TKI erlotinib was more effective at reducing cell viability than either treatment alone when tested in both 2- and 3-dimensional assays in vitro, with enhanced benefit seen in A549 cells. This increased efficacy may be due in part to the observed inhibition of Akt phosphorylation when the drugs were used in combination, where again A549 cells demonstrated the most inhibition following treatment with the drug combination. Combining erlotinib with FAK inhibitor was also potent in vivo as evidenced by reduced tumor growth in the A549 mouse xenograft model. We further ascertained that the enhanced sensitivity was irrespective of the LKB1 mutational status. In summary, we demonstrate the effectiveness of combining erlotinib and FAK inhibitors for use in known EGFR wild-type, EGFR TKI resistant cells, with the potential that a subset of cell types, which includes A549, could be particularly sensitive to this combination treatment. As such, further evaluation of this combination therapy is warranted and could prove to be an effective therapeutic approach for patients with inherent EGFR TKI-resistant NSCLC.  相似文献   

16.
Bufalin, a naturally occurring small-molecule compound from Traditional Chinese Medicine (TCM) Chansu showed inhibitory effects against human prostate, hepatocellular, endometrial and ovarian cancer cells, and leukemia cells. However, whether or not bufalin has inhibitory activity against the proliferation of human non–small cell lung cancer (NSCLC) cells is unclear. The aim of this study is to study the effects of bufalin on the proliferation of NSCLC and its molecular mechanisms of action. The cancer cell proliferation was measured by MTT assay. The apoptosis and cell cycle distribution were analyzed by flow cytometry. The protein expressions and phosphorylation in the cancer cells were detected by Western blot analysis. In the present study, we have demonstrated that bufalin suppressed the proliferation of human NSCLC A549 cell line in time- and dose-dependent manners. Bufalin induced the apoptosis and cell cycle arrest by affecting the protein expressions of Bcl-2/Bax, cytochrome c, caspase-3, PARP, p53, p21WAF1, cyclinD1, and COX-2 in A549 cells. In addition, bufalin reduced the protein levels of receptor expressions and/or phosphorylation of VEGFR1, VEGFR2, EGFR and/or c-Met in A549 cells. Furthermore, bufalin inhibited the protein expressions and phosphorylation of Akt, NF-κB, p44/42 MAPK (ERK1/2) and p38 MAPK in A549 cells. Our results suggest that bufalin inhibits the human lung cancer cell proliferation via VEGFR1/VEGFR2/EGFR/c-Met–Akt/p44/42/p38-NF-κB signaling pathways; bufalin may have a wide therapeutic and/or adjuvant therapeutic application in the treatment of human NSCLC.  相似文献   

17.
Cyclooxygenase (COX) inhibitors were regarded as anticarcinogenic agents for lung cancer at least partly via PGE2; but these were based on cytokin stimulation experiment on A549 cell. In order to clarify whether COX inhibitors directly inhibit A549 cell, three COX inhibitors, NS398 (selective COX-2 inhibitor), SC560 (selective COX-1 inhibitor), and acetyl salicylic acid (ASA, non-selective COX inhibitor), were studied. NS398, and ASA, can inhibit PGE2 generation via COX-2 inhibition. The viability of A549 cell was assayed by MTT. However, without cytokin stimulation, all the three inhibitors (NS398 0.2-20 microM; SC560 1.0-100 nM; ASA 0.01-1.0 mM) were not able to inhibit A549 cell proliferation, in the other way round, NS398 promoted cell growth. And arachidonic acid (AA) and lipopolysaccharide (LPS) did not disturb the property of its growth. These data suggested that without cytokin stimulation, COX and PGE2 may not be the kernel molecules involved in A549 cell proliferation, and COX inhibitors could not inhibit A549 cell growth directly.  相似文献   

18.
BACKGROUND: Cyclooxygenase-2 (COX-2) over-expression and subsequent prostaglandin E2 (PGE2) production are frequently associated with human non-small-cell lung cancer (NSCLC) and are involved in tumor proliferation, invasion, angiogenesis, and resistance to apoptosis. Here, we report that ciglitazone downregulates PGE2 in NSCLC cells. METHODS: PGE2 ELISA assay and COX-2 ELISA assay were performed for measuring PGE2 and COX-2, respectively, in NSCLC. The mRNA level of COX-2 was measured by semi-quantitative RT-PCR. The transient transfection experiments were performed to measure COX-2 and peroxisome proliferator-response element (PPRE) promoter activity in NSCLC. Western blots were unitized to measure PGE synthase (PGES) and 15-hydroxyprostaglandin dehydrogenase (15-PGDH) protein expression. RESULTS: COX-2 ELISA assays suggested that ciglitazone-dependent inhibition of PGE2 occurs through the suppression of COX-2. Ciglitazone treatment suppressed COX-2 mRNA expression and COX-2 promoter activity while upregulating PPRE promoter activity. Ciglitazone did not modify the expression of enzymes downstream of COX-2 including PGES and 15-PGDH. Utilization of a dominant-negative PPARgamma showed that the suppression of COX-2 and PGE2 by ciglitazone is mediated via non-PPAR pathways. CONCLUSION: Taken together, our findings suggest that ciglitazone is a negative modulator of COX-2/PGE2 in NSCLC.  相似文献   

设为首页 | 免责声明 | 关于勤云 | 加入收藏

Copyright©北京勤云科技发展有限公司  京ICP备09084417号