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 31 毫秒
1.
Although a lot of progress has been made in development of lentiviral vectors for gene therapy, the interactions of these vectors with cellular factors have not been explored adequately. Here we show that lentivirus infection phosphorylates JNK and that blocking the kinase activity of JNK decreases gene transfer in a dose-dependent manner, regardless of the viral envelope glycoprotein. Knockdown by small interfering RNA (siRNA) revealed that JNK1 but not JNK2 was required for productive gene transfer. The effect of JNK on gene transfer was not due to changes in the cell cycle, as JNK knockdown did not affect the cell cycle profile of target cells and even increased cell proliferation. In addition, confluent cell monolayers also exhibited JNK phosphorylation upon lentivirus infection and a dose-dependent decrease in gene transfer efficiency upon JNK inhibition. On the other hand, JNK activation was necessary for lentivirus internalization into the cell cytoplasm, while inhibition of JNK activity decreased virus entry without affecting binding to the cell surface. These experiments suggest that JNK is required for lentivirus entry into target cells and may have implications for gene transfer or for development of antiviral agents.  相似文献   

2.
Genetic modification of hematopoietic stem cells holds great promise in the treatment of hematopoietic disorders. However, clinical application of gene delivery has been limited, in part, by low gene transfer efficiency. To overcome this problem, we investigated the effect of retronectin (RN) on lentiviral-mediated gene delivery into hematopoietic progenitor cells (HPCs) derived from bone marrow both in vitro and in vivo. RN has been shown to enhance transduction by promoting colocalization of lentivirus and target cells. We found that RN enhanced lentiviral transfer of the VENUS transgene into cultured c-Kit+ Lin? HPCs. As a complementary approach, in vivo gene delivery was performed by subjecting mice to intra-bone marrow injection of lentivirus or a mixture of RN and lentivirus. We found that co-injection with RN increased the number of VENUS-expressing c-Kit+ Lin? HPCs in bone marrow by 2-fold. Further analysis of VENUS expression in colony-forming cells from the bone marrow of these animals revealed that RN increased gene delivery among these cells by 4-fold. In conclusion, RN is effective in enhancing lentivirus-mediated gene delivery into HPCs.  相似文献   

3.
Human immunodeficiency virus (HIV, lentivirus) vector has attractive features for gene therapy, including the ability to transduce non-dividing cells and long-term transgene expression. We have already reported that lentivirus vector can transduce well-differentiated rat cardiac myocytes. Endothelial cells (EC) are an attractive target for gene therapy, both for the treatment of cardiovascular disease and for the systemic delivery of recombinant gene products directly into the circulation. There are several reports regarding application of adenovirus and retrovirus based vectors to EC. However, there have been few reports which show the effect to lentivirus-mediated gene transfer efficiency, compared with adenovirus and retrovirus. In this study, bovine aortic endothelial cells (BAECs) were infected, in vitro, with these virus vectors. Transduction efficiency (TE) of beta-Gal gene transfer in BAECs by adenovirus, lentivirus, or retrovirus at MOI10 (Multiplicity of infection) (determined on Hela cells) is 69+/-11, 33+/-8, or 22+/-6% respectively. In adenovirus and lentivirus, almost 100% of BAECs were transduced at MOI 50. However, in retrovirus, TE showed only 48+/-6% at MOI 50 and no increase at MOI 100. The percentage of beta-Gal positive cells was decreased rapidly at longer passage of cells after being transduced by adenovirus. However, lentivirus and retrovirus showed sustained higher percentage of positive cells. Furthermore, transduction by lentiviral vectors had no significant effect on viability of BAECs. Our results indicate that lentivirus showed high-level and long term gene expression in BAECs. Lentivirus can be an effective vector for the ex vivo, genetically modified EC implantation and in vivo gene therapy.  相似文献   

4.
慢病毒载体是目前基因治疗中研究较多的载体, 与通常使用的逆转录病毒载体和腺病毒载体比较, 它有感染非分裂期细胞及容纳大片段外源性目的基因等优点.本文应用基因重组的方法,构建了1种新型的慢病毒载体,并将转录因子HBP1基因插入到这一载体上,成为pITA-HBP1.检测其在人的肿瘤细胞和正常细胞中的转染效率,发现转染效率明显增加,Western 印迹证实外源基因得到高效表达.这一结果,对今后实验室提高基因的转染效率、表达水平,以及研究基因的表达调控,都提供了重要的技术方案.  相似文献   

5.
During the past decade, lentiviral vectors based on the HIV-1 genome have been developed to become highly useful tools for efficient and stable delivery of transgenes to dividing and nondividing cells in a variety of experimental protocols. The vector system has been progressively and substantially improved, mainly to meet growing concerns over safety issues. However, the actual design and size of the lentiviral transfer vector often makes transgene cloning and DNA preparation a troublesome task. In this study, the pHR transfer vector used for lentivirus production in many laboratories was modified to contain a more versatile polylinker than the one present in the original pHR vector. In addition, the vector was significantly reduced in size from 12 to 7 kb, by replacing the original vector backbone with sequence from the multipurpose pUC18 vector. These modifications allowed for easier cloning and higher DNA yields without compromising the fundamental ability of this vector system to transduce cells in vitro and in vivo. Finally, the trimmed vector sequence was fully characterized by sequencing the vector in its entirety. In both cultured cells and directly into the rat striatum, transduction with this lentivirus, based on the modified pHsCXW vector, was as efficient and durable as with the pHR vector-based virus. In conclusion, the modified lentiviral transfer vector pHsCXW holds promise as a new valuable tool for the research community in the field of gene transfer.  相似文献   

6.
7.
The murine embryonal stem (ES) cell virus (MESV) can express transgenes from the long terminal repeat (LTR) promoter/enhancer in undifferentiated ES cells, but expression is turned off upon differentiation to embryoid bodies (EBs) and hematopoietic cells in vitro. We examined whether a human immunodeficiency virus type 1-based lentivirus vector pseudotyped with the vesicular stomatitis virus G protein (VSV-G) could transduce ES cells efficiently and express the green fluorescent protein (GFP) transgene from an internal phosphoglycerate kinase (PGK) promoter throughout development to hematopoietic cells in vitro. An oncoretrovirus vector containing the MESV LTR and the GFP gene was used for comparison. Fluorescence-activated cell sorting analysis of transduced CCE ES cells showed 99.8 and 86.7% GPF-expressing ES cells in the VSV-G-pseudotyped lentivirus (multiplicity of infection [MOI] = 59)- and oncoretrovirus (MOI = 590)-transduced cells, respectively. Therefore, VSV-G pseudotyping of lentiviral and oncoretrovirus vectors leads to efficient transduction of ES cells. Lentivirus vector integration was verified in the ES cell colonies by Southern blot analysis. When the transduced ES cells were differentiated in vitro, expression from the oncoretrovirus LTR was severely reduced or extinct in day 6 EBs and ES cell-derived hematopoietic colonies. In contrast, many lentivirus-transduced colonies, expressing the GFP gene in the undifferentiated state, continued to express the transgene throughout in vitro development to EBs at day 6, and many continued to express in cells derived from hematopoietic colonies. This experimental system can be used to analyze lentivirus vector design for optimal expression in hematopoietic cells and for gain-of-function experiments during ES cell development in vitro.  相似文献   

8.
9.
In this study, we explored the therapeutic potential of microRNA (miR) analogs against non–small-cell lung cancer (NSCLC) using lentiviral delivery of short hairpin RNA (shRNA). By using A549 as a model cell line, we used analogs and mimics of miR-4319/miR-125-5p to target the tumorigenic RAF1 gene. Lentiviral vectors carrying shRNA of a highly efficient miRNA analog of miR-4319/miR-125-5p, Analog2, were constructed to infect A549 cells. Our results showed that, compared with the noncancerous bronchial epithelial cell line 16HBE, lentivirus delivering Analog2 shRNA induced significant G2/M arrest and subsequent apoptosis in A549 cells, but not in 16HBE cells. Western blot analysis revealed that key factors regulating cell cycle were downregulated following RAF1 inhibition. In vivo xenograft experiments showed that lentivirus carrying Analog2 shRNA markedly decreased tumor size. Therefore, lentiviral delivery of Analog2 shRNA is a valid RNA interference-based treatment against NSCLC with high potency and specificity.  相似文献   

10.
目标:提供一种能够显著提高慢病毒稳定转染人多能干细胞的方法,并建立一种简便无损的转染细胞筛选方法.方法:在慢病毒转染人多能干细胞过程,分别比较添加与不添加Y-27632情况下细胞形态的动态变化规律,以及细胞不同形态下对慢病毒颗粒的摄入能力差异,优化建立高效的慢病毒转染方法.随后,设计并研制可视化的简便显微操作装置,探索...  相似文献   

11.
为实现基因治疗过程中的有效药物筛选及体内检测, 首次利用核糖体内部进入位点(IRES)构建了同时携带O6-烷基鸟嘌呤-DNA烷基转移酶(MGMT)的突变型P140K基因和荧光素酶(Luciferase)基因的慢病毒载体pBobi-MIL。RT-PCR、免疫荧光、药物筛选克隆形成及化学发光检测等实验结果表明感染重组慢病毒L-MIL的细胞能同时表达MGMT及Luciferase。构建成功的新型慢病毒载体为今后的基因治疗奠定了基础, 也为慢病毒滴度的确定提供了一种新的可能。  相似文献   

12.
为实现基因治疗过程中的有效药物筛选及体内检测, 首次利用核糖体内部进入位点(IRES)构建了同时携带O6-烷基鸟嘌呤-DNA烷基转移酶(MGMT)的突变型P140K基因和荧光素酶(Luciferase)基因的慢病毒载体pBobi-MIL。RT-PCR、免疫荧光、药物筛选克隆形成及化学发光检测等实验结果表明感染重组慢病毒L-MIL的细胞能同时表达MGMT及Luciferase。构建成功的新型慢病毒载体为今后的基因治疗奠定了基础, 也为慢病毒滴度的确定提供了一种新的可能。  相似文献   

13.
14.
Yamada K  McCarty DM  Madden VJ  Walsh CE 《BioTechniques》2003,34(5):1074-8, 1080
Recombinant lentiviral vectors stably transduce both dividing and nondividing cells. Virus pseudotyping with vesicular stomatitis virus envelope G (VSV-G) protein broadens the host range of lentiviral vector and enables vector concentration by ultra-centrifugation. However, as a result of virus vector concentration, contaminating protein debris derived from vector-producing cell culture media is toxic to target cells and reduces the transduction efficiency. Here we report a new and rapid technique for purifying lentivirus vector using the strong anion exchange column that significantly improves gene transfer rates. We purified VSV-G pseudotyped self-inactivating lentivirus vector and obtained two protein elution peaks (Peak 1 and Peak 2) corresponding to transducing activity. Peak 1 viral particles were 4-8 times more effective in transducing target cells than Peak 2 or non-purified (pre-HPLC) viral particles. We used purified lentivirus vector expressing the human Fanconi anemia group A (FANCA) gene to transduce murine hematopoietic stem/progenitor cells. We observed a consistent 2- to 3-fold increase in gene transfer rates using Peak 1 purified virus compared with non-purified virus. We conclude that the purification method using the HPLC system provides the highly purified virus vector that reduces cell toxicity and significantly improves gene transfer in primary cells.  相似文献   

15.
目的构建针对N-乙酰氨基葡萄糖转移酶(GnT)Ⅲ、Ⅳa和Ⅴ的RNA干扰(RNAi)慢病毒系统,并检测干扰慢病毒在体外小鼠肝癌细胞中对不同GnT表达的抑制作用。方法针对三种基因序列设计合成特异的shRNA序列,并构建干扰慢病毒表达载体,利用病毒包装细胞293T包装生产病毒,感染靶细胞Hca-F后,应用RT-PCR和免疫印迹检测干扰慢病毒对三种N-乙酰氨基葡萄糖转移酶表达的抑制。结果经测序证实三种干扰慢病毒表达载体构建成功,并获得高滴度的感染慢病毒。干扰慢病毒感染靶细胞后能够有效下调三种N-乙酰氨基葡萄糖转移酶的表达。结论干扰慢病毒可有效地抑制三种N-乙酰氨基葡萄糖转移酶GnT-Ⅲ、GnT-Ⅳa和GnT-Ⅴ的表达。  相似文献   

16.
Viral gene delivery is hailed as a great milestone in gene-based therapeutic approaches. The human immunodeficiency virus-derived lentiviral vectors (LVs) are advantageous in infecting both dividing and non-dividing cells leading to continuous expression of transgenes. A variety of protocols are available for concentration of LVs. We primarily generated our internal ribosome entry site (IRES)-based LVs. Virus titration and transduction efficiency were compared between various strategies that included sucrose cushion centrifugation (SCC), protein column ultrafiltration and polyethylene glycol precipitation. Among these approaches, SCC resulted in concentration of high-titer EGFP-expressing lentivirus (1.4 ± 0.3 × 109 TU/ml) with the lowest protein impurities. Further, we examined transduction strengths of our three methods on two challenging stem cells. Both human NT2 and mouse bone marrow-derived mesenchymal stem cells demonstrated high transduction using SCC of 65 ± 2.8 and 49 ± 0.8%, respectively. Finally, lentivirus particles harboring IRES-based transfer vectors of specific genes, concentrated by SCC, integrated into host genome. Taken together, development of cost-effective and efficient concentration strategies such as our SCC method is yet highly demanded to broaden the horizons of lentivirus application in clinical and translational research.  相似文献   

17.
The SCID-beige/Alb-uPA mouse model is currently the best small animal model available for viral hepatitis infection studies [1]. But the construction procedure is often costly and time-consuming due to logistic and technical difficulties. Thus, the widespread application of these chimeric mice has been hampered [2]. In order to optimize the procedure, we constructed a single lentiviral vector containing modified tetracycline-regulated system to control Alb-uPA gene expression in the cultured hepatocytes. The modified albumin promoter controlled by tetracycline (Tet)-dependent transactivator rtTA2S-M2 was integrated into a lentiviral vector. The full-length uPA cDNA was inserted into another lentiviral vector containing PTight, a modified Tet-responsive promoter. Two vectors were then digested by specific enzymes and ligated by DNA ligase 4. The ligated DNA fragment was inserted into a modified pLKO.1 cloning vector and the final lentiviral vector was then successfully constructed. H2.35 cell, Lewis lung carcinoma, primary kidney, primary hepatic interstitial and CT26 cells were infected with recombinant lentivirus at selected MOI. The expression of uPA induced by DOX was detectable only in the infected H2.35 cells, which was confirmed by real-time PCR and Western blot analysis. Moreover, DOX induced uPA expression on the infected H2.35 cells in a dose-dependent manner. The constructed single lentiviral vector has many biological advantages, including that the interested gene expression under “Tet-on/off” system is controlled by DOX in a dose-depending fashion only in murine liver cells, which provides an advantage for simplifying generation of conditional transgenic animals.  相似文献   

18.
目的:构建并制备能够有效表达Semaphorin 4D的重组慢病毒。方法:从人急性T细胞白血病Jurkat细胞DNA 扩增人Semaphorin 4D基因,克隆至pWPI GW慢病毒载体上,与pVSVG及pSPAX质粒共转染人胚肾293T细胞,包装出重组慢病毒,将纯化后的重组病毒直接感染293T和HUVEC细胞,通过免疫印迹、免疫荧光染色和血管内皮细胞迁移分析等方法检测Semaphorin 4D的表达和诱导血管内皮细胞迁移的作用。结果: 重组慢病毒介导Semaphorin 4D在293T和HUVEC内获得表达,能介导血管内皮细胞迁移。结论:成功构建了表达Semaphorin 4D的重组慢病毒载体。  相似文献   

19.
目的:DPC4/Smad4基因RNA干扰靶点的设计和RNA干扰靶点慢病毒载体制备。方法:针对DPC4/Smad4基因序列,并利用网站设计程序,依据RNA干扰序列设计的原则,设计多个RNA干扰靶点序列。根据设计经验和设计软件将其进行评估测定,选择最佳动力学参数靶点进入其后续的实验流程;生工生物合成含干扰序列的DNAoligo,具有严格的检测体系(PAGE纯化体系),其两端含酶切位点粘端,直接连入酶切后的RNA干扰载体上。将连接好的产物转入制备好的细菌感受态细胞,并且对长出的克隆进行酶切鉴定。然后挑选出阳性克隆测序,进行测序比对后,鉴定阳性的克隆即为构建成功的目的基因RNA干扰慢病毒载体。将构建的慢病毒载体以及辅助包装载体质粒共转染到293T细胞。收获含有病毒的细胞培养上清,浓缩后进行滴度测定,并检测其感染性。另外应用荧光实时定量PCR检测在感染的293T细胞中敲减效果。结果:成功构建DPC4/Smad4shRNA的慢病毒载体LVshSmad4,并成功制备DPC4/Smad4shRNA慢病毒,三株病毒感染细胞后均具有有效的敲减效应,其中SHl最为显著。结论:DPC4/Smad4基因RNA干扰靶点的成功设计和RNA干扰靶点慢病毒制备,为以后探讨DPC4/Smad4基因与肿瘤的相关性治疗提供了实验基础。  相似文献   

20.
Efficient delivery of therapeutic proteins into the pancreas represents a major obstacle to gene therapy of pancreatic disorders. The current study compared the efficiency of recombinant lentivirus and adeno-associated virus (AAV) serotypes 1, 2, 5, 8 vectors delivered by intrapancreatic injection for gene transfer in vivo. Our results indicate that lentivirus and AAV 1, 2, 8 are capable of transducing pancreas with the order of efficiency AAV8 >>AAV1 > AAV2 ≥ lentivirus, whereas AAV5 was ineffective. AAV8 resulted in an efficient, persistent (150 days) and dose-dependent transduction in exocrine acinar cells and endocrine islet cells. Pancreatic ducts and blood vessels were also transduced. Extrapancreatic transduction was restricted to liver. Leukocyte infiltration was not observed in pancreas and blood glucose levels were not altered. Thus, AAV8 represents a safe and effective vehicle for therapeutic gene transfer to pancreas in vivo.  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