首页 | 本学科首页   官方微博 | 高级检索  
相似文献
 共查询到20条相似文献,搜索用时 62 毫秒
1.
2.
We examined the antitumor effect and safety of the adenovirus-mediated expression of antisense K-ras RNA in two peritoneal dissemination models of pancreatic cancer. First, we found that the infection of an adenovirus vector expressing antisense human K-ras RNA (AxCA-AS) induced significant apoptosis in vitro in human pancreatic cancer cells with K-ras mutation. Second, the intraperitoneal (ip) injection of AxCA-AS effectively suppressed the growth of human pancreatic cancer cells in the peritoneal cavity of nude mice. Third, in the hamster syngeneic peritoneal dissemination model, the ip injection of an adenovirus expressing antisense hamster K-ras RNA significantly suppressed the peritoneal growth of hamster pancreatic cancer cells, and no significant systemic toxicity was observed in the treated hamsters. This study suggests a feasibility of the development of a therapeutic strategy against pancreatic cancer based on the adenovirus-mediated transduction of an antisense K-ras construct.  相似文献   

3.
目的检测人胰腺癌细胞株PANC-1的K-ras基因点突变方式,明确干扰Ras基因靶点的碱基序列。方法针对K-ras基因第十二位点密码子点突变最常见方式CAT,CGT,GTT设计顺序特异性引物(SSP),对人胰腺癌细胞株PANC1进行聚合酶链反应(PCR),扩增产物用聚丙烯酰胺凝胶电泳判定该细胞株有无点突变及突变方式。结果人胰腺癌细胞株PANC-1存在K-ras基因的点突变,其突变方式为CAT。结论 PCR-SSP法快速简便,特异性高,结果准确,为应用RNAi技术研究胰腺癌的基因治疗奠定了基础。  相似文献   

4.
Jin K  Park S  Ewton DZ  Friedman E 《Cancer research》2007,67(15):7247-7255
The kinase Mirk is overexpressed in many resected pancreatic adenocarcinomas and is amplified in a subset of pancreatic cancer cell lines. Depletion of Mirk has been shown to lead to apoptosis in pancreatic cancer cell lines, and thus to inhibit their clonogenic growth. Mirk is activated by signaling from activated Rac1 to MKK3 in MDCK cells, but the mechanism of activation of Mirk in pancreatic cancers is unknown. In this report, Mirk is shown to be a novel effector of K-ras, a gene mutated in approximately 90% of pancreatic cancers. Activation of Mirk signaling from oncogenic K-ras through Rac1 was shown in transient expression systems and reporter assays. Mirk activation in pancreatic cancer cells was blocked by RNA interference using three different synthetic duplex RNAis to K-ras, or two RNAis to Rac1, by pharmacologic inhibition of Rac1, or by expression of dominant negative K-rasS17N. Rac1 was activated in four out of five pancreatic cancer cell lines, and was activated by signaling from oncogenic K-ras. Mirk knockout does not induce embryonic lethality, and depletion of Mirk had no effect on the survival of normal diploid fibroblasts. In contrast, the clonogenic ability of Panc1 and AsPc1 pancreatic cancer cell lines was reduced 8- to 12-fold by the depletion of Mirk, with a greater reduction seen following the depletion of K-ras or both genes. Mirk is a novel downstream effector of oncogenic K-ras and mediates some of the survival signals activated by ras signaling.  相似文献   

5.
KRAS mutations, which are the main cause of the pathogenesis of lethal pancreatic adenocarcinomas, impair the functioning of the GTPase subunit, thus rendering it constitutively active and signaling intracellular pathways that end with cell transformation. In the present study, the AsPC-1 cell line, which has a G12D-mutated KRAS gene sequence, was utilized as a cellular model to test peptide nucleic acid-based antisense technology. The use of peptide nucleic acids (PNAs) that are built to exhibit improved hybridization specificity and have an affinity for complementary RNA and DNA sequences, as well as a simple chemical structure and high biological stability that affords resistance to nucleases and proteases, enabled targeting of the KRAS-mutated gene to inhibit its expression at the translation level. Because PNA-based antisense molecules should be capable of binding to KRAS mRNA sequences, PNAs were utilized to target the mRNA of the mutated KRAS gene, a strategy that could lead to the development of a novel drug for pancreatic cancer. Moreover, it was demonstrated that introducing new PNA to cells inhibited the growth of cancer cells and induced apoptotic death and, notably, that it can inhibit G12D-mutated KRAS gene expression, as demonstrated by RT-PCR and western blotting. Altogether, these data strongly suggest that the use of PNA-based antisense agents is an attractive therapeutic approach to treating KRAS-driven cancers and may lead to the development of novel drugs that target the expression of other mutated genes.  相似文献   

6.
The K-ras oncogene is activated in more than 75% of pancreatic cancers and represents a compelling target for therapeutic intervention. We have examined the potential of K-ras antisense oligonucleotides in human pancreatic cancer cell lines. Dose-dependent antiproliferative effects are seen, but these do not correlate with reduced K-ras levels in the treated cells. Although patterns of uptake can be altered by treatment with cationic lipids, this does not enhance antiproliferative effects nor reduce K-ras expression. The antisense approach requires considerable development before useful anti-cancer agents can emerge and alternative approaches are needed for K-ras to be a target for genetic intervention therapy for human cancer.  相似文献   

7.
Molecular target-based therapy of pancreatic cancer   总被引:5,自引:0,他引:5  
Pancreatic cancer is genetically complex, and without effective therapy. Mutations in the Kirsten-ras (K-ras) oncogene occur early and frequently (approximately 90%) during pancreatic cancer development and progression. In this context, K-ras represents a potential molecular target for the therapy of this highly aggressive cancer. We now show that a bipartite adenovirus expressing a novel cancer-specific apoptosis-inducing cytokine gene, mda-7/interleukin-24 (IL-24), and a K-ras AS gene, but not either gene alone, promotes growth suppression, induction of apoptosis, and suppression of tumor development mediated by K-ras mutant pancreatic cancer cells. Equally, the combination of an adenovirus expressing mda-7/IL-24 and pharmacologic and genetic agents simultaneously blocking K-ras or downstream extracellular regulated kinase 1/2 signaling also promotes similar inhibitory effects on the growth and survival of K-ras mutant pancreatic carcinoma cells. This activity correlates with the reversal of a translational block in mda-7/IL-24 mRNA in pancreatic cancer cells that limits message association with polysomes, thereby impeding translation into protein. Our study provides support for a "dual molecular targeted therapy" involving oncogene inhibition and selective cancer apoptosis-inducing gene expression with potential for effectively treating an invariably fatal cancer.  相似文献   

8.
A human lung cancer cell line (H460a) with a homozygous spontaneous K-ras mutation was transfected with a recombinant plasmid that synthesizes a 2-kilobase genomic segment of the K-ras protooncogene in antisense orientation. Translation of the mutated K-ras mRNA in H460a cells was specifically inhibited, whereas expression of H-ras and N-ras was unchanged. A 3-fold growth inhibition occurred in H460a cells when expression of the mutated ras p21 protein was down-regulated by antisense RNA. However, cells remained viable despite the absence of K-ras expression. The growth of H460a tumors in nu/nu mice was substantially reduced by expressed K-ras antisense RNA.  相似文献   

9.
10.
Objective: To construct the small interfering RNA(siRNA) expression cassettes (SECs) targeting activated K-ras gene sequence and investigate the effects of SECs on K-ras gene in human pancreatic cancer cell line MIAPaCa-2. Methods: Three different sites of SECs were constructed by PCR. The K1/siRNA, K2/siRNA and K3/siRNA were located at the site 194, 491 and 327, respectively. They were transfected into MiaPaCa-2 cells by liposome to inhibit the expression of activated K-ras. In the interfering groups of site 194, 491, we observed the cytopathic effect of confluent MiaPaCa-2 cells after they were incubated for 48 hours, and detected the apoptosis in cells by FACS, then we tested the alternation of K-ras gene in confluent MiaPaCa-2 cells by RT-PCR, immunofluorescence and western blot, respectively. Results: Introductions of the K1/siRNA and K2/siRNA against K-ras into MiaPaCa-2 cells led to cytopathic effect, slower proliferation and increased apoptosis, while the appearances of control MiaPaCa-2 cells remained well. The number of apoptotic cells increased compared with control cells. RT-PCR, immunofluorescence and western blot showed the effects of inhibited expression of activated K-ras gene by RNA interference in the K1/siRNA and K2/siRNA groups. We also found that the introduction of K3/siRNA had no effect on MiaPaCa-2 cells. Conclusion: K1/siRNA and K2/siRNA can inhibit the expression of activated K-ras and decrease the growth of MiaPaCa-2 cells, while K3/siRNA has no such effect, demonstrating that the suppression of tumor growth by siRNA is sequence-specific. We conclude that K-ras is involved in maintenance of tumor growth of human pancreatic cancer, and SECs against K-ras expression may be a powerful tool to be used therapeutically against human pancreatic cancer.  相似文献   

11.
Cancer is a complex disease involving multiple oncogenes with diverse actions. Inhibiting only one oncogene is unlikely to eliminate the malignancy of cancer cells. The goal of this study was to investigate whether synergistic effects can be achieved by combined silencing of two oncogenes, K-ras and Akt2, which are key players in the Ras/MAPK and PI3K/Akt signaling pathways. The pancreatic cancer cell line, Panc-1, was selected for these studies as it has elevated expression of K-ras and Akt2. Compared with inhibiting each oncogene alone, simultaneously silencing the two oncogenes with RNA interference (RNAi) more effectively inhibited Panc-1 cell proliferation and colony formation, induced a significantly higher percentage of apoptosis and resulted in greater inhibition of c-myc expression in vitro. Furthermore, when delivered by polyethyleneimine into Panc-1 tumors in nude mice, RNAi simultaneously targeting K-ras and Akt2 inhibited tumor growth more efficiently than RNAi targeting the individual oncogenes. Therefore, RNAi simultaneously silencing the oncogenes K-ras and Akt2 may offer potential opportunities for pancreatic cancer gene therapy.  相似文献   

12.
13.
14.
The prognosis of pancreatic carcinoma is poor due to the difficulty in early diagnosis, insensitivity to routine therapies and limited understanding of its pathological mechanisms. Gene therapy is now becoming an important strategy for the treatment of pancreatic carcinoma, which includes antisense gene therapy. In this study, we investigated the effect of an antisense oligonucleotide specific to point mutated K-ras on the apoptosis of human pancreatic carcinoma cells in vitro. Human pancreatic carcinoma PC-2 cells were transfected with an antisense oligonucleotide specific to a K-ras point mutation by liposomes. The effect of the antisense oligonucleotide on the apoptosis of PC-2 cells was studied using flow cytometry, TUNEL, and phase contrast microscopy. An apoptotic peak was observed in the experimental group, and most cells were arrested at the G1 phase with few cells at the S phase. The numbers of apoptotic cells in the experimental group increased as indicated by TUNEL and phase contrast microscopy. An antisense oligonucleotide specific to a K-ras point mutation promotes apoptosis in PC-2 cells in vitro perhaps by inhibition of ras gene expression.  相似文献   

15.
目的:探讨miR-885-5p对胰腺癌细胞增殖、迁移和侵袭的影响,并阐明miR-885-5p和CTNNB1基因在胰腺癌细胞中的作用机制。方法:MTT、细胞划痕和Transwell实验测定各组细胞增殖、迁移与侵袭能力。双荧光素酶报告基因实验分析CTNNB1是否为miR-885-5p的靶基因。使用胰腺癌TCGA数据进行预后分析,并进行miR-885-5p与CTNNB1表达的相关性分析。结果:下调miR-885-5p明显促进胰腺癌细胞增殖、迁移与侵袭,而过表达miR-885-5p则相反。miR-885-5p可靶向CTNNB1 3' UTR并抑制其转录后表达。在下调miR-885-5p的细胞中进行回复性实验,结果表明,miR-885-5p对胰腺癌细胞增殖、迁移与侵袭的抑制作用部分依赖于CTNNB1。TCGA数据库结果显示,miR-885-5p高表达的胰腺癌患者有较好的预后,且与CTNNB1表达呈负相关。结论:miR-885-5p通过靶向CTNNB1抑制胰腺癌细胞增殖、迁移与侵袭,miR-885-5p有望成为胰腺癌治疗的新靶点。  相似文献   

16.
Z Li  W Xia  B Fang  D H Yan 《Cancer letters》2001,174(2):151-158
Overexpression of HER-2/neu proto-oncogene is found in many human cancers including 20-30% of breast cancer and is a predictor of poor prognosis. To target breast cancer cells that overexpress HER-2/neu mRNA, we previously described a novel strategy that combines the principle of antisense (AS) and translational inhibitory activity conferred by an iron-responsive element (IRE) (AS-IRE). Here, we showed that three potential AS-IREs, i.e. AS-IRE1, 4, and 5, derived from HER-2/neu antisense sequence could bind endogenous iron regulatory protein (IRP) and, when placed in 5' untranslated region (5'UTR) of a reporter gene, the gene expression could be translationally repressed by recombinant IRP in vitro. Using AS-IRE4 as our model, we demonstrated that it is regulated by iron, and importantly, such regulation is impaired in HER-2/neu-overexpressing breast cancer cells. Furthermore, we showed that AS-IRE4 could preferentially direct the expression of a reporter gene in HER-2/neu-overexpressing breast cancer cells. Interestingly, when AS-IRE4 was placed in 5'UTR of Bax gene, a pro-apoptotic protein in the Bcl-2 protein family, we observed a preferential cell killing in breast cancer cells that overexpress HER-2/neu. Taken together, our results suggest that AS-IRE behaves as a functional IRE and it may direct therapeutic gene expression to preferentially target HER-2/neu-overexpressing breast cancer cells.  相似文献   

17.
目的:探讨死亡相关蛋白激酶1(death-associated protein kinase 1,DAPK1)在胰腺癌(pancreatic cancer,PaC)细胞放射敏感性中的作用,验证miR-324-5p通过靶向调控DAPK1影响胰腺癌细胞放射敏感性的机制。方法:通过生物信息学预测靶向DAPK1的miRNAs,并利用双荧光素酶报告基因检测miR-324-5p对DAPK1的调控作用。在PANC-1和MIA PaCa-2细胞中过表达miR-324-5p和DAPK1或抑制miR-324-5p后,对各细胞株进行放射诱导,检测细胞增殖和凋亡情况,以及凋亡相关分子的表达情况。结果:GEO数据集结果显示,胰腺癌组织中miR-324-5p的表达水平高于正常组织。与正常胰腺导管上皮细胞系(HPDE6-C7)相比,胰腺癌细胞系(Capan-1、Bxpc-3、PANC-1和MIA PaCa-2)中miR-324-5p表达水平更高(P均<0.001)。双荧光素报告基因检测结果表明,miR-324-5p靶向DAPK1的3' UTR,并且可下调DAPK1的表达。细胞实验结果证实,过表达miR-324-5p通过靶向调控DAPK1降低放射诱导的细胞凋亡和DNA的损伤,进而降低了胰腺癌细胞的放射敏感性。结论:miR-324-5p通过负调控DAPK1降低胰腺癌细胞对放射的敏感性,从而影响DNA修复和细胞凋亡。miR-324-5p/DAPK1途径可能为胰腺癌的靶向治疗提供了潜在的治疗靶点。  相似文献   

18.
BACKGROUND: The gene expression profile of pancreatic cancer is significantly different from that of normal pancreas. Differences in gene expression are detectable using microarrays, but microarrays have traditionally been applied to pancreatic cancer tissue obtained from surgical resection. We hypothesized that gene expression alterations indicative of pancreatic cancer can be detected by profiling the RNA of pancreatic juice. METHODS: We performed oligonucleotide microarray analysis on RNA isolated from pancreatic juice obtained endoscopically after secretin stimulation from six patients with pancreatic cancer and ten patients with nonneoplastic diseases of the pancreas or upper gastrointestinal tract. Extracted RNA was subjected to two rounds of linear RNA amplification, and then hybridized with U133A or X3P gene chips (Affymetrix). RESULTS: Using the U133A or X3P chips, 37 and 133 gene fragments respectively, were identified as being at least 3-fold more abundant in the pancreatic juice of patients with pancreatic cancer compared to the noncancer controls (p<0.05, Mann-Whitney test). For example, pancreatic juice from patients with pancreatic cancer contained increased levels of IL8, IFITM1, fibrinogen, osteopontin, CXCR4, DAF and NNMT RNA, genes that have been previously reported as overexpressed in primary pancreatic cancers or pancreatic cancer cell lines relative to control tissues. CONCLUSIONS: These results demonstrate that RNA analysis of pancreatic juice can reveal some of the same RNA alterations found in invasive pancreatic cancers. RNA analysis of pancreatic juice deserves further investigation to determine its utility as a tool for the evaluation of pancreatic lesions.  相似文献   

19.
The ras mutation, which is observed in 20-30% of human nonsmall cell lung cancers (NSCLCs), is one of common genetic alterations and has been proposed to be a prognostic factor in lung cancer. Oncogene ras appears to be essential for tumor progression and maintenance. Several therapeutic agents have been developed to inhibit ras, such as FTIs and antisense oligonucleotides. A new tool for blocking oncogenes in cancer cells has emerged with the discovery that RNA interference can specifically silence expression of endogenous human genes. In the current study, we used small interfering RNA (siRNA) directed against mutant K-ras to determine the anti-tumor effects of decreasing the levels of this protein in lung cancer cell lines. Adenovirus-mediated siRNA (AdH1/siK-ras(V12)) against K-ras(V12) markedly decreased K-ras(V12) gene expression and inhibited cellular proliferation of NSCLC H441 cells that express the relevant mutation (K-ras codon 12 GGT --> GTT), but produced minimal growth inhibition on NSCLC H1650 cells that lack the relevant mutation. Pretreatment with AdH1/siK-ras(V12) completely abrogated subcutaneous engraftment of H441 cells, as compared with a 100% tumor take in animals that received control vector-treated tumor cells. The in vivo effect of AdH1/siK-ras(V12) treatment was further examined by intratumoral injections after tumor induction. Pre-existing tumor growth was reduced by 45% by a single intratumoral injection. Three or five repeat injections resulted in complete tumor regression in eight of ten nude mice. Further, 23.12% of AdH1/siK-ras(V12) treated H441 cells underwent apoptosis, as compared with 6.13%, and 8.27% in untreated and control vector-treated cells, respectively. These results indicate that adenovirus-mediated siRNA can specifically and efficiently target factors whose expression is altered in malignancy and may have the potential as a therapeutic modality to treat human lung cancer.  相似文献   

20.
Qian J  Niu J  Li M  Chiao PJ  Tsao MS 《Cancer research》2005,65(12):5045-5053
Genetic analysis of pancreatic ductal adenocarcinomas and their putative precursor lesions, pancreatic intraepithelial neoplasias (PanIN), has shown a multistep molecular paradigm for duct cell carcinogenesis. Mutational activation or inactivation of the K-ras, p16(INK4A), Smad4, and p53 genes occur at progressive and high frequencies in these lesions. Oncogenic activation of the K-ras gene occurs in >90% of pancreatic ductal carcinoma and is found early in the PanIN-carcinoma sequence, but its functional roles remain poorly understood. We show here that the expression of K-ras(G12V) oncogene in a near diploid HPV16-E6E7 gene immortalized human pancreatic duct epithelial cell line originally derived from normal pancreas induced the formation of carcinoma in 50% of severe combined immunodeficient mice implanted with these cells. A tumor cell line established from one of these tumors formed ductal cancer when implanted orthotopically. These cells also showed increased activation of the mitogen-activated protein kinase, AKT, and nuclear factor-kappaB pathways. Microarray expression profiling studies identified 584 genes whose expression seemed specifically up-regulated by the K-ras oncogene expression. Forty-two of these genes have been reported previously as differentially overexpressed in pancreatic cancer cell lines or primary tumors. Real-time PCR confirmed the overexpression of a large number of these genes. Immunohistochemistry done on tissue microarrays constructed from PanIN and pancreatic cancer samples showed laminin beta3 overexpression starting in high-grade PanINs and occurring in >90% of pancreatic ductal carcinoma. The in vitro modeling of human pancreatic duct epithelial cell transformation may provide mechanistic insights on gene expression changes that occur during multistage pancreatic duct cell carcinogenesis.  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