The signal peptide of BmNPV GP64 activates the ERAD pathway to regulate heterogeneous secretory protein expression.

IF 4.3 2区 生物学 Q1 BIOTECHNOLOGY & APPLIED MICROBIOLOGY Microbial Cell Factories Pub Date : 2024-10-18 DOI:10.1186/s12934-024-02534-7
Na Liu, Ying Xu, Luping Sun, Mengmeng Li, Jinshan Huang, Bifang Hao
{"title":"The signal peptide of BmNPV GP64 activates the ERAD pathway to regulate heterogeneous secretory protein expression.","authors":"Na Liu, Ying Xu, Luping Sun, Mengmeng Li, Jinshan Huang, Bifang Hao","doi":"10.1186/s12934-024-02534-7","DOIUrl":null,"url":null,"abstract":"<p><p>As a powerful eukaryotic expression vector, the baculovirus expression vector system (BEVS) is widely applied to the production of heterogeneous proteins for research and pharmaceutical purposes, while optimization of BEVS remains a work in progress for membrane or secreted protein expression. In this study, the impact of the signal peptide (SP) derived from Bombyx mori nucleopolyhedrovirus (BmNPV) GP64 protein on protein expression, secretion, and the endoplasmic reticulum-associated degradation (ERAD) pathway were investigated in BmN cells and BEVS. Transient expression studies in BmN cells revealed that SP alters the localization and expression levels of recombinant proteins, reducing intracellular accumulation while enhancing secretion efficiency. Quantitative analysis demonstrated that SP-mediated secretion was markedly higher compared to controls, albeit with lower total expression levels. Further exploration into SP-mediated ERAD pathway activation showed increased expression of BiP and other ERAD-associated genes (PDI, UFD1, S1P, and ASK1), correlating with higher SP-driven protein expression levels. RNA interference (RNAi) experiments elucidated that knockdown of ERAD-associated genes enhances both the secretion efficiency of SP-guided proteins and the infectivity of BmNPV. Particularly, interference with BiP demonstrated the most pronounced effect on protein secretion enhancement. Viral infection experiments further supported these findings, showing upregulated ERAD-associated genes during BmNPV infection, indicating their role in viral protein processing and infectivity. In conclusion, this study elucidates the complex interplay between SP-mediated protein secretion, ERAD pathway activation, and viral infectivity in BmNPV-infected cells. These insights suggest strategies for optimizing recombinant protein production and viral protein processing in baculovirus expression systems, with potential implications for biotechnological and biomedical applications. Further research could refine our understanding and manipulation of protein secretion pathways in insect cell-based expression systems.</p>","PeriodicalId":18582,"journal":{"name":"Microbial Cell Factories","volume":null,"pages":null},"PeriodicalIF":4.3000,"publicationDate":"2024-10-18","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11487928/pdf/","citationCount":"0","resultStr":null,"platform":"Semanticscholar","paperid":null,"PeriodicalName":"Microbial Cell Factories","FirstCategoryId":"5","ListUrlMain":"https://doi.org/10.1186/s12934-024-02534-7","RegionNum":2,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":null,"EPubDate":"","PubModel":"","JCR":"Q1","JCRName":"BIOTECHNOLOGY & APPLIED MICROBIOLOGY","Score":null,"Total":0}
引用次数: 0

Abstract

As a powerful eukaryotic expression vector, the baculovirus expression vector system (BEVS) is widely applied to the production of heterogeneous proteins for research and pharmaceutical purposes, while optimization of BEVS remains a work in progress for membrane or secreted protein expression. In this study, the impact of the signal peptide (SP) derived from Bombyx mori nucleopolyhedrovirus (BmNPV) GP64 protein on protein expression, secretion, and the endoplasmic reticulum-associated degradation (ERAD) pathway were investigated in BmN cells and BEVS. Transient expression studies in BmN cells revealed that SP alters the localization and expression levels of recombinant proteins, reducing intracellular accumulation while enhancing secretion efficiency. Quantitative analysis demonstrated that SP-mediated secretion was markedly higher compared to controls, albeit with lower total expression levels. Further exploration into SP-mediated ERAD pathway activation showed increased expression of BiP and other ERAD-associated genes (PDI, UFD1, S1P, and ASK1), correlating with higher SP-driven protein expression levels. RNA interference (RNAi) experiments elucidated that knockdown of ERAD-associated genes enhances both the secretion efficiency of SP-guided proteins and the infectivity of BmNPV. Particularly, interference with BiP demonstrated the most pronounced effect on protein secretion enhancement. Viral infection experiments further supported these findings, showing upregulated ERAD-associated genes during BmNPV infection, indicating their role in viral protein processing and infectivity. In conclusion, this study elucidates the complex interplay between SP-mediated protein secretion, ERAD pathway activation, and viral infectivity in BmNPV-infected cells. These insights suggest strategies for optimizing recombinant protein production and viral protein processing in baculovirus expression systems, with potential implications for biotechnological and biomedical applications. Further research could refine our understanding and manipulation of protein secretion pathways in insect cell-based expression systems.

查看原文
分享 分享
微信好友 朋友圈 QQ好友 复制链接
本刊更多论文
作为一种功能强大的真核表达载体,杆状病毒表达载体系统(BEVS)被广泛应用于生产用于研究和制药的异构蛋白,而对于膜蛋白或分泌蛋白的表达,BEVS的优化工作仍在进行中。本研究研究了从蚕核多角体病毒(BmNPV)GP64蛋白中提取的信号肽(SP)在BmN细胞和BEVS中对蛋白表达、分泌和内质网相关降解(ERAD)途径的影响。在 BmN 细胞中进行的瞬时表达研究表明,SP 改变了重组蛋白的定位和表达水平,减少了细胞内的积累,同时提高了分泌效率。定量分析表明,与对照组相比,SP 介导的分泌量明显增加,尽管总表达水平较低。对 SP 介导的 ERAD 通路激活的进一步研究表明,BiP 和其他 ERAD 相关基因(PDI、UFD1、S1P 和 ASK1)的表达量增加,这与 SP 驱动的蛋白质表达量增加有关。RNA 干扰(RNAi)实验表明,敲除 ERAD 相关基因可提高 SP 引导蛋白的分泌效率和 BmNPV 的感染性。尤其是对 BiP 的干扰对蛋白质分泌的增强作用最为明显。病毒感染实验进一步证实了这些发现,在 BmNPV 感染过程中,ERAD 相关基因上调,表明它们在病毒蛋白加工和感染性中的作用。总之,本研究阐明了 BmNPV 感染细胞中 SP 介导的蛋白质分泌、ERAD 通路激活和病毒传染性之间复杂的相互作用。这些见解提出了优化重组蛋白生产和杆状病毒表达系统中病毒蛋白处理的策略,对生物技术和生物医学应用具有潜在影响。进一步的研究可以完善我们对基于昆虫细胞的表达系统中蛋白质分泌途径的理解和操作。
本文章由计算机程序翻译,如有差异,请以英文原文为准。
求助全文
约1分钟内获得全文 去求助
来源期刊
Microbial Cell Factories
Microbial Cell Factories 工程技术-生物工程与应用微生物
CiteScore
9.30
自引率
4.70%
发文量
235
审稿时长
2.3 months
期刊介绍: Microbial Cell Factories is an open access peer-reviewed journal that covers any topic related to the development, use and investigation of microbial cells as producers of recombinant proteins and natural products, or as catalyzers of biological transformations of industrial interest. Microbial Cell Factories is the world leading, primary research journal fully focusing on Applied Microbiology. The journal is divided into the following editorial sections: -Metabolic engineering -Synthetic biology -Whole-cell biocatalysis -Microbial regulations -Recombinant protein production/bioprocessing -Production of natural compounds -Systems biology of cell factories -Microbial production processes -Cell-free systems
期刊最新文献
No two clones are alike: characterization of heterologous subpopulations in a transgenic cell line of the model diatom Phaeodactylum tricornutum. Investigation on taxonomy, secondary metabolites and antibacterial activity of Streptomyces sediminicola sp. nov., a novel marine sediment-derived Actinobacteria. Cell immobilization for enhanced milk clotting enzyme production from Bacillus amyloliquefacien and cheese quality. The signal peptide of BmNPV GP64 activates the ERAD pathway to regulate heterogeneous secretory protein expression. A growth-coupling strategy for improving the stability of terpenoid bioproduction in Escherichia coli.
×
引用
GB/T 7714-2015
复制
MLA
复制
APA
复制
导出至
BibTeX EndNote RefMan NoteFirst NoteExpress
×
×
提示
您的信息不完整,为了账户安全,请先补充。
现在去补充
×
提示
您因"违规操作"
具体请查看互助需知
我知道了
×
提示
现在去查看 取消
×
提示
确定
0
微信
客服QQ
Book学术公众号 扫码关注我们
反馈
×
意见反馈
请填写您的意见或建议
请填写您的手机或邮箱
已复制链接
已复制链接
快去分享给好友吧!
我知道了
×
扫码分享
扫码分享
Book学术官方微信
Book学术文献互助
Book学术文献互助群
群 号:481959085
Book学术
文献互助 智能选刊 最新文献 互助须知 联系我们:info@booksci.cn
Book学术提供免费学术资源搜索服务,方便国内外学者检索中英文文献。致力于提供最便捷和优质的服务体验。
Copyright © 2023 Book学术 All rights reserved.
ghs 京公网安备 11010802042870号 京ICP备2023020795号-1