首页 | 本学科首页   官方微博 | 高级检索  
相似文献
 共查询到19条相似文献,搜索用时 171 毫秒
1.
目的:探究人髓细胞白血病基因-1(myeloid cell leukemia-1,Mcl-1)是否参与非小细胞肺癌对吉非替尼的耐药。方法:应用Western blot检测Mcl-1在吉非替尼敏感细胞PC-9和耐药细胞H1975表达差异;梯度浓度的吉非替尼作用于PC-9细胞后,Western blot实验检测B淋巴细胞瘤-2基因(B-cell lymphoma-2,Bcl-2)家族中抗凋亡蛋白的表达变化;应用Western blot实验检测Mcl-1在PC-9和H1975细胞内降解速度差异。结果:Mcl-1在PC-9细胞内的表达明显低于H1975细胞,差异有统计学意义(P0.05),并且随着吉非替尼作用浓度的升高,Mcl-1表达逐渐降低,而Bcl-2和Bcl-x L表达基本不变,并且PC-9细胞内Mcl-1降解更迅速,半衰期明显缩短。结论:非小细胞肺癌对吉非替尼耐药可能与Mcl-1的表达量上调,降解速度减慢,半衰期延长有关。  相似文献   

2.
目的:探讨抗凋亡蛋白Mcl-1在GCDA诱导的肝癌细胞耐药中的作用及其机制。方法:培养3种肝癌细胞系,用免疫荧光法和Western blot技术检测Mcl-1的表达;GCDA±CYC处理HepG2细胞,采用Western blot技术检测Mcl-1的半衰期变化;用抗癌药物Irinotecan与GCDA对HepG2细胞进行处理,采用MTT法和Western blot技术分别检测细胞增殖抑制率和Mcl-1的表达变化;用RNA干扰技术下调Mcl-1,检测化疗药物对HepG2细胞的敏感性。结果:Mcl-1在肝癌细胞中广泛表达;GCDA能延长Mcl-1的半衰期至6h以上,并明显减弱化疗药物对抗凋亡蛋白Mcl-1的抑制作用,降低癌细胞的药物敏感性;RNA干扰下调Mcl-1能增加癌细胞的药物敏感性。结论:胆盐(GCDA)能诱导HepG2细胞产生耐药性,其作用机制可能是通过延长Mcl-1半衰期增加其蛋白稳定性和抗凋亡作用来促使肝癌细胞抗药的。  相似文献   

3.
摘要 目的:探讨靶向抗凋亡蛋白Bcl-2克服非小细胞肺癌EGFR-TKIs耐药的作用及重定位Bcl-2靶向抑制剂用于克服耐药的可行性。方法:通过药物浓度梯度递增法构建非小细胞肺癌多代EGFR-TKIs耐药株,根据亲本细胞和多代EGFR-TKIs耐药的非小细胞肺癌细胞的RNA-seq数据筛选出潜在耐药相关基因Bcl-2,通过Western blot 检测其在耐药细胞中的蛋白水平。为了探讨Bcl-2诱导耐药的作用,采用siRNA干扰和使用Bcl-2的抑制剂ABT199抑制Bcl-2,通过CCK8法、IncuCyte实时监测和Western blot等方法检测其对亲本和耐药细胞的细胞活力、药物敏感性、增殖和凋亡的影响。随后使用奥希替尼分别处理亲本及耐药细胞,通过Western Blot检测NRF2受到药物作用后及在耐药细胞中的蛋白水平,并以临床公共数据库分析辅助验证。使用siRNA干扰或NRF2抑制剂ML385敲低或抑制NRF2功能,借助Western Blot和CCK8法检测其对Bcl-2表达水平及对EGFR-TKI敏感性的影响;通过加入NRF2的激动剂Ki696探究其对Bcl-2的诱导作用、对EGFR-TKI敏感性的影响及取消靶向Bcl-2逆转耐药的作用。结果:Bcl-2在EGFR-TKIs耐药细胞中上调;敲低或抑制Bcl-2后,可选择性抑制耐药细胞的生长和活力,并诱导凋亡,且能逆转包括第三代药物奥希替尼在内的多代EGFR-TKIs耐药;EGFR-TKI可在敏感细胞中诱导NRF2的上调,且耐药细胞中上调的Bcl-2受NRF2调控。结论:EGFR-TKIs耐药细胞通过上调抗凋亡分子Bcl-2获得耐药,该分子的上调受NRF2的调控,靶向Bcl-2则可以逆转耐药。  相似文献   

4.
研究抑制泛素特异性蛋白酶9X(ubiquitin-specific protease 9X,USP9X)对人肝癌(primary hepatocellular carcinoma,HCC)细胞SMMC7721和HepG2中髓细胞白血病-1(myeloid cell leukemia-1,Mcl-1)蛋白的表达调控及对细胞凋亡和生长活力的影响。实验分为USP9X-siRNA组和阴性对照NC组两组进行分析。通过Western blot技术分别检测USP9X在肝癌细胞SMMC7721、HepG2和正常人肝细胞株L02中的蛋白表达情况;应用化学合成USP9X-siRNA转染肝癌细胞SMMC7721和HepG2,通过Western blot、流式细胞仪和MTT检测转染前后Mcl-1的蛋白表达差异以及细胞凋亡和生长活力变化。结果表明,USP9X在肝癌细胞SMMC7721和HepG2中的蛋白表达水平均高于正常肝细胞L02(t=15.155,P=0.000;t=9.171,P=0.001);SMMC7721和HepG2细胞中抑制USP9X能明显下调Mcl-1的蛋白表达,并导致细胞凋亡增加和生长活力降低。提示,肝癌细胞SMMC7721和HepG2中USP9X表达上调;USP9X表达降低可能通过下调Mcl-1的蛋白表达进而诱导人肝癌细胞SMMC7721和HepG2的凋亡。  相似文献   

5.
目的:探讨抑制Mcl-1基因表达对淋巴瘤Raji细胞增殖和凋亡的影响及机制。方法:NC-siRNA、Mcl-1-siRNA转染Raji细胞,以不作任何处理的细胞作为空白对照组,48h后Western blot检测各组细胞中Mcl-1的蛋白表达;CCK8实验和流式细胞仪分别检测细胞的增殖和凋亡情况;Western blot检测Cleaved caspase3、Notch1、Hes1蛋白表达。结果:转染Mcl-1-siRNA后Mcl-1的蛋白表达显著降低;与对照组及NC-siRNA组比较,Mcl-1-siRNA组细胞存活率显著降低,细胞凋亡率显著升高,Cleaved caspase3蛋白显著上调表达,Notch1和Hes1蛋白显著下调表达。结论:RNA干扰抑制Mcl-1基因表达可显著降低Raji细胞增殖及诱导细胞凋亡,其机制与抑制Notch1信号通路有关。  相似文献   

6.
目的:研究PARP-1对高糖诱导的心肌细胞增殖的影响及可能机制。方法:用高糖处理H9C2细胞,qRT-PCR和Western blot检测细胞中PARP-1 m RNA和蛋白水平。H9C2细胞转染PARP-1 si RNA和si RNA control,q RT-PCR和Western blot检测细胞中PARP-1 m RNA和蛋白水平。用高糖处理转染PARP-1 si RNA后的H9C2细胞,CCK-8检测细胞增殖情况,硫代巴比妥酸法检测丙二醛(MDA)水平,黄嘌呤氧化酶法检测超氧化物歧化酶(SOD)水平,Western blot检测增殖细胞核抗原(PCNA)、p38丝裂原活化蛋白激酶(p38MAPK)、磷酸化的p38MAPK(p-p38MAPK)蛋白的表达。结果:高糖诱导的H9C2细胞中PARP-1 m RNA和蛋白水平明显高于正常培养的H9C2细胞(P0.05)。PARP-1 si RNA能够明显下调H9C2细胞中PARP-1 m RNA和蛋白水平。高糖处理后H9C2细胞存活率明显降低,细胞中MDA水平升高,细胞中SOD水平降低,细胞内的PCNA水平降低,p38MAPK磷酸化水平升高,与正常培养的H9C2细胞相比,差异均具有统计学意义(P0.05)。用高糖培养下调PARP-1的H9C2细胞,细胞存活率有所升高,细胞中MDA水平降低,细胞中SOD水平也升高,细胞中PCNA水平升高,细胞中p38MAPK磷酸化水平降低,与单纯高糖培养的细胞相比,差异均具有统计学意义(P0.05)。结论:PARP-1在高糖诱导的心肌细胞中表达上调,可能通过激活p38MAPK信号途径,增加细胞脂质氧化应激抑制心肌细胞增殖。  相似文献   

7.
目的:观察紫草素联合埃克替尼对肺腺癌耐药细胞H1975增殖的影响,探讨克服耐药可能的作用机制。方法:应用MTT法检测紫草素(1.25~20μmo/L)、埃克替尼(5~100μmol/L)及两药联合干预对H1975细胞生长的抑制作用;流式细胞术观察紫草素(1.25μmol/L)、埃克替尼(10μmol/L)及联合使用对H1975凋亡作用;Western blot检测不同干预对H1975细胞EGFR、p-EGFR、AKT、p-AKT、ERK、p-ERK和凋亡相关蛋白PARP表达水平的影响。结果:MTT检测结果显示,与单药组相比,联合用药组细胞H1975增殖能力明显减弱,差异有统计学意义(P0.05);流式细胞术结果显示,联合用药组细胞的凋亡率达到(52.45±3.04)%,较紫草素组细胞凋亡率(22±1.17)%和埃克替尼处理组细胞凋亡率(15.35±5.85)%明显提高,差异有统计学意义(P0.05)。Western blot结果显示,单药组下调了p-EGFR、p-Akt蛋白水平的表达,而联合用药组显著抑制了p-ERK、PARP蛋白水平的表达,差异有统计学意义(P0.05),EGFR、AKT、ERK蛋白表达无差异(P0.05)。结论:紫草素联合埃克替尼能明显抑制H1975细胞增殖,促进肿瘤细胞凋亡;抗肿瘤机制可能与调节EGFR信号通路相关蛋白表达有关。  相似文献   

8.
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   

9.
目的研究RNA干扰人pttg表达对肺癌SPC-A-1细胞恶性表型的影响。方法构建针对人pttg的RNA干扰真核表达载体,运用脂质体介导转染SPC-A-1细胞并用G418筛选出阳性克隆株;半定量PCR和Western blot检测转染后PTTG mRNA及蛋白的表达水平;人工计数法检测阳性克隆株SPC-A-1细胞染色体倍性的变化;氚-胸腺嘧啶核苷掺入法观察抑制人pttg表达后SPC-A-1细胞增殖能力的改变;TUNEL法检测细胞凋亡的变化情况。结果成功构建针对人pttg基因的RNA干扰真核表达载体(命名为Si-hPTTG),转染后筛选获得人pttg下调表达的SPC-A-1细胞株;Si-hPTTG转染使SPC-A-1细胞PTTG mRNA和蛋白的表达较未转染组和阴性对照质粒组均显著下调;Si-hPTTG转染后SPC-A-1细胞染色体众数和平均数增加,3H掺入量显著降低,细胞凋亡明显增加。结论人pttg下调表达可增加SPC-A-1细胞染色体众数和平均数,伴有减轻染色体结构畸变的趋势;显著抑制SPC-A-1细胞增殖,增加细胞凋亡。人pttg可能是肺癌治疗的新靶点。  相似文献   

10.
为了探讨Rh type C glycoprotein (RHCG)对非小细胞肺癌(non-small cell lung cancer,NSCLC)细胞增殖的影响及可能的作用机制,本研究使用荧光定量PCR法检测12对NSCLC及癌旁组织样本中RHCG mRNA的表达水平及pcDNA3.1-RHCG质粒对A549细胞RHCG m RNA的表达;采用CCK-8法检测细胞增殖能力;运用PI染色法检测细胞周期;使用免疫印迹法检p-PI3K、PI3K、p-AKT以及AKT蛋白表达水平。本研究发现,与癌旁组织比较,NSCLC中RHCG m RNA表达水平明显降低。RHCG过表达能抑制NSCLC细胞系A549细胞增殖能力。此外,RHCG过表达使A549细胞周期G1/S期转化发生阻滞。本研究还发现,RHCG过表达可下调A549细胞p-PI3K/PI3K和p-AKT/AKT水平。本研究表明,RHCG抑制NSCLC细胞增殖的作用与其抑制PI3K/AKT信号通路有关。  相似文献   

11.
Resistance to epidermal growth factor receptor-tyrosine kinase inhibitors (EGFR-TKIs), such as erlotinib and gefitinib, is a major clinical problem in the treatment of patients with non-small cell lung cancer (NSCLC). YM155 is a survivin small molecule inhibitor and has been demonstrated to induce cancer cell apoptosis and autophagy. EGFR-TKIs have been known to induce cancer cell autophagy. In this study, we showed that YM155 markedly enhanced the sensitivity of erlotinib to EGFR-TKI resistant NSCLC cell lines H1650 (EGFR exon 19 deletion and PTEN loss) and A549 (EGFR wild type and KRAS mutation) through inducing autophagy-dependent apoptosis and autophagic cell death. The effects of YM155 combined with erlotinib on apoptosis and autophagy inductions were more obvious than those of YM155 in combination with survivin knockdown by siRNA transfection, suggesting that YM155 induced autophagy and apoptosis in the NSCLC cells partially depend on survivin downregulation. Meanwhile, we found that the AKT/mTOR pathway is involved in modulation of survivin downregulation and autophagy induction caused by YM155. In addition, YM155 can induce DNA damage in H1650 and A549 cell lines. Moreover, combining erlotinib further augmented DNA damage by YM155, which were retarded by autophagy inhibitor 3MA, or knockdown of autophagy-related protein Beclin 1, revealing that YM155 induced DNA damage is autophagy-dependent. Similar results were also observed in vivo xenograft experiments. Therefore, combination of YM155 and erlotinib offers a promising therapeutic strategy in NSCLC with EGFR-TKI resistant phenotype.  相似文献   

12.
Epidermal growth factor receptor (EGFR) and c-MET receptors are expressed on many non-small cell lung cancer (NSCLC) cells. Current single agent therapeutic targeting of a mutant EGFR has a high efficacy in the clinic, but is not curative. Here, we investigated the combination of targeting EGFR and c-MET pathways in NSCLC cells resistant to receptor tyrosine kinase inhibitors (TKIs), using RNA interference and inhibition by TKIs. Different NSCLC cell lines with various genomic characteristics (H358, H1650 and H1975) were transfected with EGFR-specific-siRNA, T790M-specific-siRNA, c-MET siRNA or the combination. Subsequently EGFR TKIs (gefitinib, erlotinib or afatinib) or monoclonal antibody cetuximab were combined respectively with the c-MET-specific TKI su11274 in NSCLC cell lines. The cell proliferation, viability, caspase−3/7 activity and apoptotic morphology were monitored by spectrophotometry, fluorimetry and fluorescence microscopy. The combined effect of EGFR TKIs, or cetuximab and su11274, was evaluated using a combination index. The results showed that the cell lines that were relatively resistant to EGFR TKIs, especially the H1975 cell line containing the resistance T790M mutation, were found to be more sensitive to EGFR-specific-siRNA. The combination of EGFR siRNA plus c-MET siRNA enhanced cell growth inhibition, apoptosis induction and inhibition of downstream signaling in EGFR TKI resistant H358, H1650 and H1975 cells, despite the absence of activity of the c-MET siRNA alone. EGFR TKIs or cetuximab plus su11274 were also consistently superior to either agent alone. The strongest biological effect was observed when afatinib, an irreversible pan-HER blocker was combined with su11274, which achieved a synergistic effect in the T790M mutant H1975 cells. In a conclusion, our findings offer preclinical proof of principle for combined inhibition as a promising treatment strategy for NSCLC, especially for patients in whom current EGFR-targeted treatments fail due to the presence of the T790M-EGFR-mutation or high c-MET expression.  相似文献   

13.
Although non-small cell lung cancer (NSCLC) tumors with activating mutations in the epidermal growth factor receptor (EGFR) are highly responsive to EGFR tyrosine kinase inhibitors (TKIs) including gefitinib and erlotinib, development of acquired resistance is almost inevitable. Statins show antitumor activity, but it is unknown whether they can reverse EGFR-TKIs resistance in NSCLC with the T790M mutation of EGFR. This study investigated overcoming resistance to EGFR-TKI using simvastatin. We demonstrated that addition of simvastatin to gefitinib enhanced caspase-dependent apoptosis in T790M mutant NSCLC cells. Simvastatin also strongly inhibited AKT activation, leading to suppression of β-catenin activity and the expression of its targets, survivin and cyclin D1. Both insulin treatment and AKT overexpression markedly increased p-β-catenin and survivin levels, even in the presence of gefitinib and simvastatin. However, inhibition of AKT by siRNA or LY294002 treatment decreased p-β-catenin and survivin levels. To determine the role of survivin in simvastatin-induced apoptosis of gefitinib-resistant NSCLC, we showed that the proportion of apoptotic cells following treatment with survivin siRNA and the gefitinib–simvastatin combination was greater than the theoretical additive effects, whereas survivin up-regulation could confer protection against gefitinib and simvastatin-induced apoptosis. Similar results were obtained in erlotinib and simvastatin-treated HCC827/ER cells. These findings suggest that survivin is a key molecule that renders T790M mutant NSCLC cells resistant to apoptosis induced by EGFR-TKIs and simvastatin. Overall, these data indicate that simvastatin may overcome EGFR-TKI resistance in T790M mutant NSCLCs via an AKT/β-catenin signaling-dependent down-regulation of survivin and apoptosis induction.  相似文献   

14.
Cisplatin resistance of non-small-cell lung cancer (NSCLC) needs to be well elucidated. RING finger protein (RNF38) has been proposed as a biomarker of NSCLC poor prognosis. However, its role in drug resistance in NSCLC is poorly understood. RNF38 expression was detected in normal lung epithelial cell and four NSCLC cell lines. RNF38 was stably overexpressed in A549 and H460 cells or silenced in H1975 and cisplatin-resistant A549 cells (A549-CDDP resistant) using lentiviral vectors. RNF38 expression levels were determined using quantitative real-time polymerase chain reaction and western blotting analysis. Cell viability in response to different concentrations of cisplatin was evaluated by Cell Counting Kit-8 assay. RNF38 expression levels were markedly elevated in NSCLC cells and cells harboring high RNF38 were less sensitive to cisplatin. Overexpression of RNF38 reduced, while RNF38 silencing increased the drug sensitivity of cisplatin in NSCLC cells. Cisplatin-resistant cells expressed high RNF38 level. RNF38 silencing promoted cell apoptosis and enhanced the drug sensitivity of cisplatin in cisplatin-resistant NSCLC cells. These findings indicate that RNF38 might induce cisplatin resistance of NSCLC cells via promoting cell apoptosis and RNF38 could be a novel target for rectify cisplatin resistance in NSCLC cases.  相似文献   

15.
Twist1 is highly expressed in primary and metastatic non-small cell lung cancer (NSCLC), and thus acts as a critical target for lung cancer chemotherapy. In the current study, we investigated the underlying mechanism initiated by silencing of Twist1 that sensitizes NSCLC cells to cisplatin. Silencing of Twist1 triggered ATP depletion, leading to AMP-activated protein kinase (AMPK)-activated mammalian target of rapamycin (mTOR) inhibition in NSCLC cells. AMPK-induced mTOR inhibition, in turn, resulted in downregulation of ribosome protein S6 kinase 1 (S6K1) activity. Downregulation of mTOR/S6K1 reduced Mcl-1 protein expression, consequently promoting sensitization to cisplatin. Overexpression of Mcl-1 reduced PARP cleavage induced by cisplatin and Twist1 siRNA, suggesting that this sensitization is controlled through Mcl-1 expression. Interestingly, cells treated with Twist1 siRNA displayed upregulation of p21Waf1/CIP1, and suppression of p21Waf1/CIP1 with specific siRNA further enhanced the cell death response to cisplatin/Twist1 siRNA. In conclusion, silencing of Twist1 sensitizes lung cancer cells to cisplatin via stimulating AMPK-induced mTOR inhibition, leading to a reduction in Mcl-1 protein. To our knowledge, this is the first report to provide a rationale for the implication of cross-linking between Twist1 and mTOR signaling in resistance of NSCLC to anticancer drugs.  相似文献   

16.
Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) can induce both caspase-dependent apoptosis and kinase activation in tumor cells. Here, we examined the consequences and mechanisms of TRAIL-induced MAPKs p38 and JNK in non-small cell lung cancer (NSCLC) cells. In apoptosis sensitive H460 cells, these kinases were phosphorylated, but not in resistant A549 cells. Time course experiments in H460 cells showed that induction of p38 phosphorylation preceded that of JNK. To explore the function of these kinases in apoptosis activation by TRAIL, chemical inhibitors or siRNAs were employed to impair JNK or p38 functioning. JNK activation counteracted TRAIL-induced apoptosis whereas activation of p38 stimulated apoptosis. Notably, the serine/threonine kinase RIP1 was cleaved following TRAIL treatment, concomitant with detectable JNK phosphorylation. Further examination of the role of RIP1 by short hairpin (sh)RNA-dependent knockdown or inhibition by necrostatin-1 showed that p38 can be phosphorylated in both RIP1-dependent and -independent manner, whereas JNK phosphorylation occurred independent of RIP1. On the other hand JNK appeared to suppress RIP1 cleavage via an unknown mechanism. In addition, only the activation of JNK by TRAIL was caspase-8-dependent. Finally, we identified Mcl-1, a known substrate for p38 and JNK, as a downstream modulator of JNK or p38 activity. Collectively, our data suggest in a subset of NSCLC cells a model in which TRAIL-induced activation of p38 and JNK have counteracting effects on Mcl-1 expression leading to pro- or anti-apoptotic effects, respectively. Strategies aiming to stimulate p38 and inhibit JNK may have benefit for TRAIL-based therapies in NSCLC.  相似文献   

17.
摘要 目的:分析富含半胱氨酸的酸性分泌蛋白类似蛋白1(SPARCL1)对非小细胞肺癌(NSCLC)细胞增殖、凋亡、侵袭的影响,并探讨分裂原活化抑制剂(MEK)/细胞外调节蛋白激酶(ERK)通路在其中发挥的作用。方法:收集2019年9月~2021年6月期间本院接受手术治疗的84例NSCLC患者癌组织与相应癌旁组织,实时定量逆转录聚合酶链反应(qRT-PCR)法测定并比较各组织以及正常肺上皮细胞HBEpiC、NSCLC细胞A549、HCC827、H1299、H292中SPARCL1 信使RNA(mRNA)表达水平,选取A549、HCC827培养并分组,分为对照组、NC siRNA组、SPARCL1 siRNA组、U0126组(MEK/ERK特异性抑制剂)、SPARCL1 siRNA加U0126组,细胞计数法(CCK8)以及平板克隆法测定A549、HCC827细胞增殖,流式细胞仪测定A549、HCC827细胞凋亡,Transwell小室法测定A549、HCC827细胞侵袭能力,蛋白质印迹法(western blot)检测SPARCL1、p-MEK、MEK、p-ERK1/2、ERK1/2蛋白表达。结果:SPARCL1在NSCLC组织中mRNA表达水平低于癌旁组织(P<0.05);与HBEpiC细胞相比,NSCLC细胞A549、HCC827、H1299、H292细胞中SPARCL1 mRNA表达水平降低(P<0.05);与对照组相比,SPARCL1 siRNA组A549、HCC827细胞SPARCL1 mRNA表达水平与蛋白表达、凋亡率降低(P<0.05),OD450、克隆形成数、侵袭细胞数、p-MEK/MEK、p-ERK1/2/ERK1/2蛋白表达升高(P<0.05),U0126组A549、HCC827细胞SPARCL1 mRNA表达水平与蛋白表达、凋亡率升高(P<0.05),OD450、克隆形成数、侵袭细胞数、p-MEK/MEK、p-ERK1/2/ERK1/2蛋白表达降低(P<0.05);与SPARCL1 siRNA组相比,SPARCL1 siRNA加U0126组A549、HCC827细胞SPARCL1 mRNA表达水平与蛋白表达、凋亡率升高(P<0.05),OD450、克隆形成数、侵袭细胞数、p-MEK/MEK、p-ERK1/2/ERK1/2蛋白表达降低(P<0.05)。结论:SPARCL1可能通过调控MEK/ERK通路影响NSCLC A549、HCC827细胞增殖、侵袭与凋亡。  相似文献   

18.
Tyrosine kinase inhibitors for epidermal growth factor receptor (EGFR-TKIs) are used as molecular targeted therapy for non-small cell lung cancer (NSCLC) patients. The therapy is applied to the patients having EGFR-primary L858R mutation, but drug tolerance caused by EGFR-secondary mutation is occurred within one and half years. For the non-invasive detection of the EGFR-TKIs treatment positive patients by positron emission tomograpy (PET) imagaing, fluorine-18 labeled thienopyrimidine derivative, [18F]FTP2 was newly synthesized. EGFR inhibition assay, cell uptake study, and blocking study indicated [18F]FTP2 binds with high and selective affinity for EGFR with L858R mutation, and not with L858R/T790M dual mutations. On animal PET study using tumor bearing mice, H3255 cells expressing L858R mutated EGFR was more clearly visualized than H1975 cells expressing L858R/T790M dual mutated EGFR. [18F]FTP2 has potential for detecting NSCLC which is susceptible to EGFR-TKI treatment.  相似文献   

19.
Survivin is a member of the inhibitor of apoptosis protein family that is overexpressed in various tumors and is important in restricting apoptosis. Understanding the molecular events of apoptosis may provide information for developing novel therapeutic agents targeting non-small cell lung cancer (NSCLCs). This study used three human NSCLC cell lines, NCI-H1299, SK-MES-1, and NCI-H460. Changes in apoptosis, the mRNA and protein expression of survivin under normoxia and hypoxia, with or without rapamycin treatment were analyzed. In addition, siRNA and ChIP assay were further applied to demonstrate the role of hypoxia-inducible factor 1 (HIF-1)α in regulating survivin expression regulation under hypoxia during rapamycin induced NSCLC cell apoptosis. Treatment with rapamycin resulted in significantly increased NSCLC cells apoptosis under hypoxia. We demonstrated for the first time that rapamycin inhibited hypoxia-induced survivin expression in NSCLC cell lines. We further demonstrated that HIF-1α participated in hypoxia-induced survivin expression, and that rapamycin inhibited hypoxia-induced HIF-1α expression by enhancing its degradation. The results above collectively showed that rapamycin inhibits HIF-1α-induced survivin expression under hypoxia to induce NSCLC apoptosis.  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