lentiviral vector

慢病毒载体
  • 文章类型: Journal Article
    近年来,基因治疗取得了长足的进步。4000多个蛋白质编码基因与6000多种遗传疾病有关;下一代测序极大地彻底改变了遗传疾病的诊断。大多数遗传疾病被认为是非常罕见的或超罕见的,这里定义为少于1:100,000的案例,但在12种已批准的基因疗法(不包括RNA疗法)中,只有一种针对的是超微病变.本文探讨了三种适用于多种罕见遗传病的基因补充治疗方法:慢病毒载体修饰的自体CD34+造血干细胞移植,将腺相关病毒(AAV)载体全身递送至肝脏,和局部AAV递送到脑脊液和大脑。连同RNA疗法,我们为这些基因疗法提出了一个潜在的商业模式。
    Gene therapy has made considerable strides in recent years. More than 4000 protein-coding genes have been implicated in more than 6000 genetic diseases; next-generation sequencing has dramatically revolutionized the diagnosis of genetic diseases. Most genetic diseases are considered very rare or ultrarare, defined here as having fewer than 1:100,000 cases, but only one of the 12 approved gene therapies (excluding RNA therapies) targets an ultrarare disease. This article explores three gene supplementation therapy approaches suitable for various rare genetic diseases: lentiviral vector-modified autologous CD34+ hematopoietic stem cell transplantation, systemic delivery of adeno-associated virus (AAV) vectors to the liver, and local AAV delivery to the cerebrospinal fluid and brain. Together with RNA therapies, we propose a potential business model for these gene therapies.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    慢病毒载体(LVV)已被用作临床试验中基因治疗的常见载体之一。已经报道了LVV介导的临床试验成功治疗了数百个β-地中海贫血病例。这些LVV带有反向放置的β-血红蛋白(HBB)基因表达盒,用于在病毒RNA包装期间保存内含子。因此,这些LVV通常产生少量由其内部基因启动子驱动的负取向转录物,并且通过与病毒骨架互补的负链降低病毒滴度。为了克服这个问题,我们设计了特异性靶向由LVV内部启动子驱动的负链RNA的shRNA,其导致病毒滴度显著增加.该报告证明了提高LVV系统基因治疗有效性的简单而积极的手段。
    Lentiviral vector (LVV) has been used as one of the common carriers for gene therapy in clinical trials. LVV-mediated clinical trials have being reported in successfully treating hundreds of β-thalassemia cases. These LVVs bear an inversely placed β-hemoglobin (HBB) gene expression cassette for preserving introns during the viral RNA packaging. Consequently, these LVVs often produce a small amount of negatively orientated transcript driven by its internal gene promoter and would lower the viral titer by the minus-strand complemented with the viral backbone. To overcome this problem, we designed shRNAs specifically target the minus-strand RNA driven by the LVV internal promoter that resulted in a notable increase in the viral titer. This report demonstrates a simple and positive mean for increasing the effectiveness for gene therapy with the LVV system.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    杜氏肌营养不良症(DMD)是一种由肌营养不良蛋白基因突变引起的无法治愈的X连锁隐性遗传病。许多研究人员旨在通过病毒载体恢复截短的肌营养不良蛋白。然而,载体的低包装能力和免疫原性阻碍了其临床应用。在这里,我们构建了四个慢病毒载体,这些载体具有由肌肉特异性启动子驱动的截短和序列优化的肌营养不良蛋白基因。四种慢病毒载体在体外C2C12肌细胞中稳定表达小肌营养不良蛋白。为了评估体内的治疗效果,我们通过局部注射将慢病毒载体转移到新生C57BL/10ScSn-Dmdmdx小鼠中。修饰的肌营养不良蛋白表达水平增加,它们的分布在治疗小鼠中也得到了恢复。同时,它们表现出拉力的恢复和单核细胞数量的减少。缓解在体内持续3-6个月。此外,没有载体的整合位点分布到癌基因中。总之,本研究初步证明了微肌营养不良蛋白慢病毒载体用于DMD基因治疗的可行性和安全性,并为修复截短的肌营养不良蛋白提供了新的策略。
    Duchenne muscular dystrophy (DMD) is an incurable X-linked recessive genetic disease caused by mutations in the dystrophin gene. Many researchers aim to restore truncated dystrophin via viral vectors. However, the low packaging capacity and immunogenicity of vectors have hampered their clinical application. Herein, we constructed four lentiviral vectors with truncated and sequence-optimized dystrophin genes driven by muscle-specific promoters. The four lentiviral vectors stably expressed mini-dystrophin in C2C12 muscle cells in vitro. To estimate the treatment effect in vivo, we transferred the lentiviral vectors into neonatal C57BL/10ScSn-Dmdmdx mice through local injection. The levels of modified dystrophin expression increased, and their distribution was also restored in treated mice. At the same time, they exhibited the restoration of pull force and a decrease in the number of mononuclear cells. The remissions lasted 3-6 months in vivo. Moreover, no integration sites of vectors were distributed into the oncogenes. In summary, this study preliminarily demonstrated the feasibility and safety of lentiviral vectors with mini-dystrophin for DMD gene therapy and provided a new strategy to restore truncated dystrophin.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    背景:慢球蛋白BB305是携带用于治疗β-地中海贫血的人β-球蛋白表达盒的自我失活慢病毒载体。最初,cHS4的2×250bp鸡基因座控制区片段,起到绝缘子的作用,被置于其ΔU3,在由法国团队领导的避免异常剪接的第一次临床试验后被移除,等。这种作用可能导致由β-珠蛋白启动子驱动的转录通读速率增加到显著水平的风险。在临床试验中构成生物安全风险。
    方法:在本研究中,设计了一种通读还原剂(C-U或WPRE),将其置于β-珠蛋白基因的3UTR处。检查mRNA水平的增强子活性和/或转录通读(EATRT)率和关于慢病毒制剂滴度的蛋白质表达水平。
    结果:我们发现元素(C-U或WPRE)的插入有效地将EATRT降低了53%或41%,分别。C-U+对病毒封装效率影响较小。此外,C-U+或WPRE插入后蛋白表达水平无显著差异。
    结论:本研究的结果表明,在BB305的polyA序列之前插入C-U+或WPRE将降低EATRT率,而不损害其表达功效和病毒制备滴度。因此,我们提出了一种用于β-地中海贫血临床试验的更安全的慢病毒载体的替代改进。
    BACKGROUND: LentiGlobin BB305 is a self-inactivating lentiviral vector carrying a human β-globin expressing cassette for treating β-thalassemia. Initially, a 2 × 250 bp chicken Locus Control Region fragment of cHS4, functioning as an insulator, was placed at its ΔU3, which was removed after the first clinical trial led by a French team to avoid abnormal splicing, etc. This action could potentially lead to an increasing risk of the transcriptional read-through rate driven by the β-globin promoter to a significant level, posing a biosafety risk in clinical trials.
    METHODS: In the present study, a read-through reducing agent (C-U+ or WPRE) was designed to be placed at the 3\' UTR of the β-globin gene. The Enhancer Activities and/or Transcriptional Read-Through (EATRT) rate at the mRNA level and the protein expression level regarding lentiviral preparation titer were examined.
    RESULTS: We found that the insertion of the element (C-U+ or WPRE) reduced the EATRT effectively by 53% or 41%, respectively. C-U+ has less impact on virus package efficiency. Furthermore, there was no significant difference in the protein expression level after the C-U+ or WPRE insertion.
    CONCLUSIONS: The results of the present study show that inserting C-U+ or WPRE before the polyA sequence of the BB305 would reduce the EATRT rate at no cost of its expressing efficacy and viral preparation titers. Thus, we present an alternative improvement for a safer lentiviral vector for β-thalassemia clinical trials.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    狂犬病仍然是对公众健康的巨大威胁。狂犬病病毒包膜糖蛋白(RABVG)是一种主要的狂犬病病毒抗原,含有中和表位,它们是亚单位疫苗和诊断抗原的主要候选者。然而,rRABVG的生产和纯化同时保留其抗原性和免疫原性仍然是一个挑战。这里,我们的目标是建立一个rRABVG生产和纯化的平台,并确定rRABVG的免疫原性和抗原性。合成了编码RABVG可溶形式的cDNA片段,并将其克隆到慢病毒表达载体中。重组慢病毒载体LV-CMV-RABVG-eGFP包装,tideleted,然后转导到HEK293T细胞中。使用镍亲和层析纯化细胞培养上清液,随后通过蛋白质印迹分析和间接酶联免疫吸附测定(ELISA)确认。ELISA利用从已经接种人商业纯化Vero细胞狂犬病疫苗(PVRV)的个体获得的人血清。值得注意的是,我们在免疫的猪而不是小鼠中观察到中和抗体反应。这种差异可能归因于诸如rRABVG蛋白的不稳定性等因素。宿主反应的变化,和所用佐剂的差异。考虑到所有这些发现,本研究中产生的rRABVG蛋白有望成为预防狂犬病的潜在候选疫苗.
    Rabies continues to be a huge threat to public health. The rabies virus envelope glycoprotein (RABV G) is a major rabies virus antigen and contains neutralizing epitopes, which are primary candidates for subunit vaccines and diagnostic antigens. However, the production and purification of rRABV G while retaining its antigenic and immunogenic remains to be a challenge. Here, we aimed to establish a platform for rRABV G production and purification, and determine the immunogenicity and antigenicity of rRABV G. The cDNA fragment encoding the soluble form of RABV G was synthesized and cloned into a lentiviral expressing vector. Recombinant lentiviral vector LV-CMV-RABV G-eGFP was packaged, titered, and then transduced into HEK 293T cells. The cell culture supernatant was purified using nickel affinity chromatography and subsequently confirmed through Western Blot analysis and indirect enzyme-linked immunosorbent assay (ELISA). The ELISA utilized human sera obtained from individuals who had been vaccinated with the human commercial Purified Vero Cells Rabies Vaccine (PVRV). Notably, we observed a neutralizing antibody response in immunized pigs rather than in mice. This discrepancy could potentially be attributed to factors such as the instability of the rRABV G protein, variations in host responses, and variances in the adjuvant used. Taking all these findings into account, the rRABV G protein generated in this study exhibits promise as a potential vaccine candidate for the prevention of rabies.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    这里,我们首次提出评估磁敏clMagR/clCry4作为磁共振成像(MRI)报告基因,可赋予真核生物对内源性对比的敏感性。使用慢病毒载体,我们将clMagR/clCry4引入C57BL/6小鼠来源的骨髓间充质干细胞(mBMSCs),可以与铁特异性结合,显著影响MRI横向松弛,并且在体内产生容易检测到的对比度而没有不利影响。具体来说,clMagR/clCry4使mBMSCs有利于增强MRI-R2对铁颗粒的敏感性,其中细胞募集外源性铁并将这些储存转化为MRI可检测的造影剂;这对于对照细胞是无法实现的。此外,与TEM一起进行普鲁士蓝染色以提供在MRI上可检测到天然铁颗粒的直接证据。因此,推断MRI检测的敏感性应与clMagR/clCry4和外源性铁相关。一起来看,clMagR/clCry4作为MRI报告基因具有巨大的潜力。重要声明::在这项研究中,我们建议评估磁敏clMagR/clCry4作为MRI报告基因,赋予真核mBMSCs内源性对比的检测灵敏度。在这一点上,clMagR和clCry4位于细胞质内,可能相互影响。clMagR/clCry4使mBMSCs有利于增强MRI-R2对天然铁颗粒的敏感性,其中蛋白质可以与铁特异性结合,并将这些储存转化为MRI可检测的造影剂;这不是由对照细胞实现的。该观点推测clMagR/clCry4与外源铁互补。此外,普鲁士蓝染色与TEM观察一起进行,以提供铁颗粒对MRI敏感的直接证据。
    Here, we propose for the first time the evaluation of magnetosensitive clMagR/clCry4 as a magnetic resonance imaging (MRI) reporter gene that imparts sensitivity to endogenous contrast in eukaryotic organisms. Using a lentiviral vector, we introduced clMagR/clCry4 into C57BL/6 mice-derived bone marrow mesenchymal stem cells (mBMSCs), which could specifically bind with iron, significantly affected MRI transverse relaxation, and generated readily detectable contrast without adverse effects in vivo. Specifically, clMagR/clCry4 makes mBMSCs beneficial for enhancing the sensitivity of MRI-R2 for iron-bearing granules, in which cells recruit exogenous iron and convert these stores into an MRI-detectable contrast; this is not achievable with control cells. Additionally, Prussian blue staining was performed together with ultrathin cell slices to provide direct evidence of natural iron-bearing granules being detectable on MRI. Hence, it was inferred that the sensitivity of MRI detection should be correlated with clMagR/clCry4 and exogenous iron. Taken together, the clMagR/clCry4 has great potential as an MRI reporter gene. STATEMENT OF SIGNIFICANCE: In this study, we propose the evaluation of magnetosensitive clMagR/clCry4 as an MRI reporter gene, imparting detection sensitivity to eukaryotic mBMSCs for endogenous contrast. At this point, the clMagR and clCry4 were located within the cytoplasm and possibly influence each other. The clMagR/clCry4 makes mBMSCs beneficial for enhancing the sensitivity of MRI-R2 for iron-bearing granules, in which protein could specifically bind with iron and convert these stores into MRI-detectable contrast; this is not achieved by control cells. The viewpoint was speculated that the clMagR/clCry4 and exogenous iron were complementary to each other. Additionally, Prussian blue staining was performed together with TEM observations to provide direct evidence that the iron-bearing granules were sensitive to MRI.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    基因疗法通过编辑肿瘤细胞的有害基因来治愈癌症,但是,缺乏载体系统来有效地将遗传物质递送到体内特定的肿瘤位点,这限制了其在癌症基因治疗中的全部治疗潜力。在过去的二十年里,越来越多的研究表明,慢病毒载体(LV)用不同的糖蛋白从捐赠病毒修饰,一个被称为假型化的过程,在转导中具有改变的向性并显示出细胞类型特异性,导致选择性肿瘤细胞杀伤。LV的这一特征以及它们在体内分裂和非分裂哺乳动物细胞中实现高效基因递送的能力使得它们成为未来癌症基因治疗中的有吸引力的工具。本文旨在总结一些典型的LV假型的现状及其在许多恶性肿瘤基础抗癌研究中的应用。还讨论了将假型LV转化为癌症治疗的临床应用的机会。
    Gene therapy holds great promise for curing cancer by editing the deleterious genes of tumor cells, but the lack of vector systems for efficient delivery of genetic material into specific tumor sites in vivo has limited its full therapeutic potential in cancer gene therapy. Over the past two decades, increasing studies have shown that lentiviral vectors (LVs) modified with different glycoproteins from a donating virus, a process referred to as pseudotyping, have altered tropism and display cell-type specificity in transduction, leading to selective tumor cell killing. This feature of LVs together with their ability to enable high efficient gene delivery in dividing and non-dividing mammalian cells in vivo make them to be attractive tools in future cancer gene therapy. This review is intended to summarize the status quo of some typical pseudotypings of LVs and their applications in basic anti-cancer studies across many malignancies. The opportunities of translating pseudotyped LVs into clinic use in cancer therapy have also been discussed.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    背景:血友病A(HA)是由内源性凝血级联中因子VIII(FVIII)基因缺乏引起的X连锁单基因疾病。目前HA的蛋白质替代疗法(PRT)有许多局限性,包括短期有效性,高成本,和终身治疗要求。基因治疗已成为HA的一种有希望的治疗方法。原位功能性FVIII生物合成对其凝血活性至关重要。
    方法:为了研究FVIII的靶向表达,我们开发了一系列先进的慢病毒载体(LV)携带通用启动子(EF1α)或多种组织特异性启动子,包括内皮特异性(VEC),内皮和上皮特异性(KDR),和巨核细胞特异性(Gp和ITGA)启动子。
    结果:为了检查组织特异性,在人内皮细胞和巨核细胞系中测试B结构域缺失的人F8(F8BDD)基因的表达。功能测定表明LV-VEC-F8BDD和LV-ITGA-F8BDD在转导的内皮细胞和巨核细胞的治疗范围内的FVIII活性,分别。在F8敲除小鼠(F8KO小鼠,F8null小鼠),静脉内(iv)注射LV说明了不同载体的不同程度的表型校正以及抗FVIII免疫应答。LV-VEC-F8BDD和LV-Gp-F8BDD的静脉给药在180天内实现了80%和15%的治疗性FVIII活性,分别。与其他LV结构不同,LV-VEC-F8BDD在处理的F8null小鼠中显示出低FVIII抑制反应。
    结论:LV-VEC-F8BDD具有较高的LV包装和交付效率,在F8null小鼠中具有内皮特异性和低免疫原性,具有很大的临床应用潜力。
    Hemophilia A (HA) is an X-linked monogenic disorder caused by deficiency of the factor VIII (FVIII) gene in the intrinsic coagulation cascade. The current protein replacement therapy (PRT) of HA has many limitations including short term effectiveness, high cost, and life-time treatment requirement. Gene therapy has become a promising treatment for HA. Orthotopic functional FVIII biosynthesis is critical to its coagulation activities.
    To investigate targeted FVIII expression, we developed a series of advanced lentiviral vectors (LVs) carrying either a universal promoter (EF1α) or a variety of tissue-specific promoters, including endothelial-specific (VEC), endothelial and epithelial-specific (KDR), and megakaryocyte-specific (Gp and ITGA) promoters.
    To examine tissue specificity, the expression of a B-domain deleted human F8 (F8BDD) gene was tested in human endothelial and megakaryocytic cell lines. Functional assays demonstrated FVIII activities of LV-VEC-F8BDD and LV-ITGA-F8BDD in the therapeutic range in transduced endothelial and megakaryocytic cells, respectively. In F8 knockout mice (F8 KO mice, F8null mice), intravenous (iv) injection of LVs illustrated different degrees of phenotypic correction as well as anti-FVIII immune response for the different vectors. The iv delivery of LV-VEC-F8BDD and LV-Gp-F8BDD achieved 80% and 15% therapeutic FVIII activities over 180 days, respectively. Different from the other LV constructs, the LV-VEC-F8BDD displayed a low FVIII inhibitory response in the treated F8null mice.
    The LV-VEC-F8BDD exhibited high LV packaging and delivery efficiencies, with endothelial specificity and low immunogenicity in the F8null mice, thus has a great potential for clinical applications.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    乙型肝炎病毒(HBV)感染是主要的公共卫生问题。HBV的清除可能涉及细胞毒性T淋巴细胞(CTL)活性和T辅助1型反应。由树突状细胞(DC)产生的外来体可以诱导能够根除病毒的免疫应答。然而,载有抗原的外泌体尚未证明在HBV感染中的治疗潜力。因此,本研究旨在研究负载泛素化HBV核心抗原(Dexs-Ub-HBcAg)的DC衍生外泌体(Dexs)的抗病毒作用。用编码HBcAg的泛素化形式的重组lentivector装载鼠骨髓来源的DC。使用差速离心产生高纯度Dexs。用Dexs-Ub-HBcAg刺激脾T淋巴细胞,并检查特异性T细胞介导的免疫反应。使用酶联免疫吸附测定分析细胞因子表达。使用细胞计数试剂盒-8测定法检测T淋巴细胞增殖,并使用乳酸脱氢酶释放测定法测定HBcAg特异性CTL活性。结果表明,Dexs-Ub-HBcAg可有效刺激T细胞增殖,并诱导抗原特异性CTL的激活,从而在体外表现出HBcAg特异性CTL免疫反应。这些结果表明Dexs-Ub-HBcAg作为消除HBV的未来治疗选择的发展潜力。
    Hepatitis B virus (HBV) infection is a major public health concern. The clearance of HBV may involve cytotoxic T-lymphocyte (CTL) activity and T helper type 1 reactions. Exosomes generated from dendritic cells (DCs) can induce immunological responses capable of eradicating viruses. However, exosomes loaded with antigens have not yet demonstrated therapeutic potential in HBV infection. Therefore, the present study aimed to investigate the antiviral effects of DC-derived exosomes (Dexs) loaded with ubiquitinated HBV core antigen (Dexs-Ub-HBcAg). Murine bone marrow-derived DCs were loaded with a recombinant lentivector encoding the ubiquitinated form of HBcAg. High-purity Dexs were generated using differential velocity centrifugation. Splenic T-lymphocytes were stimulated with Dexs-Ub-HBcAg and the specific T-cell-mediated immune responses were examined. Cytokine expression was analyzed using enzyme-linked immunosorbent assays. T-lymphocyte proliferation was detected using a Cell Counting Kit-8 assay and HBcAg-specific CTL activity was determined using a lactate dehydrogenase release assay. The results revealed that Dexs-Ub-HBcAg effectively stimulated T-cell proliferation and induced the activation of antigen-specific CTLs to exhibit HBcAg-specific CTL immune responses in vitro. These results suggest the potential of Dexs-Ub-HBcAg for development as a future therapeutic option for the elimination of HBV.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    过继嵌合抗原受体(CAR)修饰的T或NK细胞(CAR-T/NK)已成为一种新型的疾病治疗方法。慢病毒载体(LV)通常用于工程化NK细胞以有效表达CAR。本研究报道了单启动子和双启动子LV对工程化NK细胞的CAR表达和细胞毒性的影响。我们构建了第三代基于NKG2D的CAR,通过靶向多达八种应激诱导配体(NKG2DL)来杀死癌细胞。我们的结果表明,通过利用优化的单启动子pCDHsp而不是原始的双启动子pCDHdp,CAR在HEK-293T或NK92细胞中表现出与GFP报道分子更高的表达水平和更高的共表达一致性。选择嘌呤霉素后,pCDHsp产生强大的CAR表达并增强工程化NK细胞的体外细胞毒性。因此,推荐使用单启动子pCDHsp慢病毒感染制备CAR工程NK细胞。这项研究有助于优化CAR-NK细胞的生产并增强其功能活性,提供质量更好、更均匀的CAR-NK细胞产品。
    Adoptive chimeric antigen receptor (CAR)-modified T or NK cells (CAR-T/NK) have emerged as a novel form of disease treatment. Lentiviral vectors (LVs) are commonly employed to engineer NK cells for the efficient expression of CARs. This study reported the influence of single-promoter and dual-promoter LVs on the CAR expression and cytotoxicity of engineered NK cells. We constructed a third-generation NKG2D-based CAR that kills cancer cells by targeting up to eight stress-induced ligands (NKG2DLs). Our results demonstrated that the CAR exhibits both a higher expression level and a higher coexpression concordance with the GFP reporter in HEK-293T or NK92 cells by utilizing the optimized single-promoter pCDHsp rather than the original dual-promoter pCDHdp. After puromycin selection, the pCDHsp produces robust CAR expression and enhanced in vitro cytotoxicity of engineered NK cells. Therefore, infection with a single-promoter pCDHsp lentivector is recommended to prepare CAR-engineered NK cells. This research helps to optimize the production of CAR-NK cells and enhance their functional activity, to provide CAR-NK cell products with better and more uniform quality.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

公众号