IC-BEVS

  • 文章类型: Journal Article
    昆虫细胞-杆状病毒表达载体(IC-BEV)平台使得能够进行小规模研究和大规模商业生产重组蛋白和治疗性生物制品,包括基于重组腺相关病毒(rAAV)的基因递送载体。由于其简单性,该平台的广泛使用可与其他基于哺乳动物细胞系的平台相媲美。高产,可比较的质量属性,和强大的生物处理功能。在这一章中,我们描述了使用One-Bac平台的最新修改之一的rAAV生产方案,其由携带AAVRep2/Cap5基因的稳定转化的Sf9细胞系组成,所述AAVRep2/Cap5基因在感染后被单个重组杆状病毒表达载体诱导,所述载体含有感兴趣的转基因(rAAV基因组)。总体方案包括基本步骤,包括rBEV工作库存准备,rAAV生产,和基于离心的细胞培养裂解物的澄清。相同的协议也可以应用于使用传统的Three-Bac的rAAV载体生产,两个Bac,和Mono-Bac平台,无需进行重大更改。
    The insect cell-baculovirus expression vector (IC-BEV) platform has enabled small research-scale and large commercial-scale production of recombinant proteins and therapeutic biologics including recombinant adeno-associated virus (rAAV)-based gene delivery vectors. The wide use of this platform is comparable with other mammalian cell line-based platforms due to its simplicity, high-yield, comparable quality attributes, and robust bioprocessing features. In this chapter, we describe a rAAV production protocol employing one of the recent modifications of the One-Bac platform that consists of a stable transformed Sf9 cell line carrying AAV Rep2/Cap5 genes that are induced upon infection with a single recombinant baculovirus expression vector harboring the transgene of interest (rAAV genome). The overall protocol consists of essential steps including rBEV working stock preparation, rAAV production, and centrifugation-based clarification of cell culture lysate. The same protocol can also be applied for rAAV vector production using traditional Three-Bac, Two-Bac, and Mono-Bac platforms without requiring significant changes.
    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
    自适应实验室进化(ALE)是增强细胞系在特定应用中的适应性的强大工具,包括重组蛋白生产。通过适应非标准培养条件,细胞可以发展特定的特性,使它们成为高生产者。尽管被广泛用于微生物和,在较小程度上,哺乳动物细胞,ALE对昆虫细胞的利用程度很低。这里,我们描述了通过ALE方法使昆虫HighFive和Sf9细胞适应非标准培养条件的方法。为了证明ALE提高昆虫细胞生产力的潜力,展示了两个案例研究。在第一,我们将昆虫HighFive细胞从标准pH(6.2)调整到中性pH(7.0);这种适应使流感病毒样颗粒(VLP)的产量提高了三倍,使用瞬时杆状病毒表达载体系统。在第二个,我们使昆虫Sf9细胞从其标准培养温度(27°C)适应低温生长(22°C);这种适应使流感VLP的产量提高了六倍,使用稳定的细胞系。这些实施例证明了ALE通过操纵不同的培养条件来提高不同昆虫细胞宿主和表达系统内的生产力的潜力。
    Adaptive laboratory evolution (ALE) is a powerful tool for enhancing the fitness of cell lines in specific applications, including recombinant protein production. Through adaptation to nonstandard culture conditions, cells can develop specific traits that make them high producers. Despite being widely used for microorganisms and, to lesser extent, for mammalian cells, ALE has been poorly leveraged for insect cells. Here, we describe a method for adapting insect High Five and Sf9 cells to nonstandard culture conditions via an ALE approach. Aiming to demonstrate the potential of ALE to improve productivity of insect cells, two case studies are demonstrated. In the first, we adapted insect High Five cells from their standard pH (6.2) to neutral pH (7.0); this adaptation allowed to improve production of influenza virus-like particles (VLPs) by threefold, using the transient baculovirus expression vector system. In the second, we adapted insect Sf9 cells from their standard culture temperature (27 °C) to hypothermic growth (22 °C); this adaptation allowed to improve production of influenza VLPs by sixfold, using stable cell lines. These examples demonstrate the potential of ALE for enhancing productivity within distinct insect cell hosts and expression systems by manipulating different culture conditions.
    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
    The insect cell-baculovirus expression vector system (IC-BEVS) has been widely used to produce recombinant protein at high titers, including complex virus-like particles (VPLs). However, cell-to-cell variability upon infection is yet one of the least understood phenomena in virology, and little is known about its impact on production of therapeutic proteins. This study aimed at dissecting insect cell population heterogeneity during production of influenza VLPs in IC-BEVS using single-cell RNA-seq (scRNA-seq). High Five cell population was shown to be heterogeneous even before infection, with cell cycle being one of the factors contributing for this variation. In addition, infected insect cells were clustered according to the timing and level of baculovirus genes expression, with each cluster reporting similar influenza VLPs transgenes (i.e., hemagglutinin and M1) transcript counts. Trajectory analysis enabled to track infection progression throughout pseudotime. Specific pathways such as translation machinery, protein folding, sorting and degradation, endocytosis and energy metabolism were identified as being those which vary the most during insect cell infection and production of Influenza VLPs. Overall, this study lays the ground for the application of scRNA-seq in IC-BEVS processes to isolate relevant biological mechanisms during recombinant protein expression towards its further optimization.
    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
    严重急性呼吸综合征冠状病毒2(SARS-CoV-2)同源三聚体刺突(S)蛋白通过与血管紧张素转换酶2(ACE2)受体结合来介导宿主细胞进入,因此是冠状病毒病19(COVID-19)疫苗中的关键病毒抗原。尽管有COVID-19疫苗,低疫苗覆盖率以及未接种疫苗和免疫功能受损的受试者导致了值得关注的SARS-CoV-2变体的出现。因此,新的和/或更新的疫苗的持续开发对于防止此类新变体是必不可少的。在这项研究中,我们使用昆虫细胞-杆状病毒表达载体系统(IC-BEVS)开发了一种可扩展的生物过程,以生产高质量的S蛋白,融合前构象稳定,纳入基于病毒体的COVID-19候选疫苗。通过探索不同的生物过程工程策略(即,信号肽,杆状病毒转移载体,细胞系,感染策略和制剂缓冲液),我们能够获得~4mg/L的纯化S蛋白,which,据我们所知,是迄今为止使用昆虫细胞获得的最高值。此外,昆虫细胞来源的S蛋白表现出与哺乳动物细胞相似的聚糖加工和储存时的中期稳定性(在-80和4°C下或在5个冻融循环后长达90天)。值得注意的是,S蛋白的抗原性,作为单一抗原或显示在病毒体表面,通过ELISA证实,与ACE2受体结合,泛SARS抗体CR3022和S蛋白上各种表位簇的中和抗体。在4°C下储存1个月的病毒体-S上也保持结合能力。这项工作证明了使用IC-BEVS产生高度糖基化和复杂S蛋白的潜力,在不损害其完整性和抗原性的情况下,纳入基于病毒体的COVID-19候选疫苗。
    The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) homotrimeric spike (S) protein is responsible for mediating host cell entry by binding to the angiotensin-converting enzyme 2 (ACE2) receptor, thus being a key viral antigen to target in a coronavirus disease 19 (COVID-19) vaccine. Despite the availability of COVID-19 vaccines, low vaccine coverage as well as unvaccinated and immune compromised subjects are contributing to the emergence of SARS-CoV-2 variants of concern. Therefore, continued development of novel and/or updated vaccines is essential for protecting against such new variants. In this study, we developed a scalable bioprocess using the insect cells-baculovirus expression vector system (IC-BEVS) to produce high-quality S protein, stabilized in its pre-fusion conformation, for inclusion in a virosome-based COVID-19 vaccine candidate. By exploring different bioprocess engineering strategies (i.e., signal peptides, baculovirus transfer vectors, cell lines, infection strategies and formulation buffers), we were able to obtain ~4 mg/L of purified S protein, which, to the best of our knowledge, is the highest value achieved to date using insect cells. In addition, the insect cell-derived S protein exhibited glycan processing similar to mammalian cells and mid-term stability upon storage (up to 90 days at -80 and 4 °C or after 5 freeze-thaw cycles). Noteworthy, antigenicity of S protein, either as single antigen or displayed on the surface of virosomes, was confirmed by ELISA, with binding of ACE2 receptor, pan-SARS antibody CR3022 and neutralizing antibodies to the various epitope clusters on the S protein. Binding capacity was also maintained on virosomes-S stored at 4 °C for 1 month. This work demonstrates the potential of using IC-BEVS to produce the highly glycosylated and complex S protein, without compromising its integrity and antigenicity, to be included in a virosome-based COVID-19 vaccine candidate.
    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
    Bone Marrow Tyrosine kinase in the chromosome X (BMX) is a TEC family kinase associated with numerous pathological pathways in cancer cells. Covalent inhibition of BMX activity holds promise as a therapeutic approach against cancer. To screen for potent and selective covalent BMX inhibitors, large quantities of highly pure BMX are normally required which is challenging with the currently available production and purification processes. Here, we developed a scalable production process for the human recombinant BMX (hrBMX) using the insect cell-baculovirus expression vector system. Comparable expression levels were obtained in small-scale shake flasks (13 mL) and in stirred-tank bioreactors (STB, 5 L). A two-step chromatographic-based process was implemented, reducing purification times by 75% when compared to traditional processes, while maintaining hrBMX stability. The final production yield was 24 mg of purified hrBMX per litter of cell culture, with a purity of > 99%. Product quality was assessed and confirmed through a series of biochemical and biophysical assays, including circular dichroism and dynamic light scattering. Overall, the platform herein developed was capable of generating 100 mg purified hrBMX from 5 L STB in just 34 days, thus having the potential to assist in-vitro covalent ligand high-throughput screening for BMX activity inhibition.
    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