中国神经再生研究(英文版) ›› 2021, Vol. 16 ›› Issue (5): 851-855.doi: 10.4103/1673-5374.299272

• 原著:神经损伤修复保护与再生 • 上一篇    下一篇

AAV8的转导能力因事先暴露于内体样pH条件而降低

  

  • 出版日期:2021-05-15 发布日期:2020-12-29
  • 基金资助:

    美国国立卫生研究院(NS100531),克雷格·尼尔森基金会(598684)和迈阿密治愈麻痹计划。

AAV8 transduction capacity is reduced by prior exposure to endosome-like pH conditions

Jeffrey A. Lowell1, Kar Men Mah1, John L. Bixby1, 2, 3, Vance P. Lemmon1, 2,*   

  1. 1 Miami Project to Cure Paralysis, University of Miami, Miami, FL, USA;  2 Department of Neurological Surgery, University of Miami, Miami, FL, USA;  3 Department of Molecular and Cellular Pharmacology, University of Miami, Miami, FL, USA
  • Online:2021-05-15 Published:2020-12-29
  • Contact: Vance P. Lemmon, PhD, VLemmon@med.miami.edu.
  • Supported by:
    This work was supported by grants to JLB and VPL from the National Institutes of Health (NS100531), the Craig H. Neilsen Foundation (598684), and the Miami Project to Cure Paralysis. VPL holds the Walter G. Ross Distinguished Chair in Developmental Neuroscience. 

摘要:

腺相关病毒(Adeno associated virus,AAV)是神经科学家工具箱中的一个重要工具,它允许DNA的传递提供荧光蛋白的标记或基因指令来调节基因的表达。在神经再生领域,神经元的转导可以观察和调控轴突的生长和再生,未来有望成为一种提供分子疗法促进中枢神经系统损伤后再生和再生的机制。传统的AAV制剂配方允许在生理条件下进行有效的病毒转导,但对AAV转导机制局限性的进一步了解可能有助于生产更具弹性的AAV菌株,用于研究和治疗目的。我们研究了AAV8血清型(AAV8)在内质体内吞作用(pH7.4~pH4.4)所遇到的环境pH值范围内(pH7.4~pH4.4)环境中对AAV的转导进行了研究,在这种环境中,低pH触发病毒衣壳的结构和自蛋白水解变化被认为是内体逃逸和病毒脱壳所必需的。由于这些过程的基本性质,我们假设AAV8颗粒过早暴露于酸性pH会降低体外HT1080细胞的病毒转导,这是通过使用高含量成像分析的荧光报告基因表达来测量的。我们发现,越来越酸性的孵育条件伴随着转导效率的降低,并且报告基因在转化细胞中的表达水平也同样降低。在pH7.4和pH6.4之间,信号转导的最大减少,这表明pH相关事件和病毒失活可能同时发生。综上所述,这些发现表明AAV8暴露在酸性pH下1小时,对转导能力有害。未来的研究有必要了解pH相关的致病机制。

https://orcid.org/0000-0003-3550-7576 (Vance P. Lemmon)

关键词: 腺相关病毒, 自溶, 衣壳, 低pH, 蛋白酶, 蛋白水解, 温度, 运输, 转导, 病毒灭活

Abstract: Adeno-associated virus (AAV) is an essential instrument in the neuroscientist’s toolkit, which allows delivery of DNA to provide labeling with fluorescent proteins or genetic instructions to regulate gene expression. In the field of neural regeneration, the transduction of neurons enables the observation and regulation of axon growth and regeneration, and in the future will likely be a mechanism for delivering molecular therapies to promote sprouting and regeneration after central nervous system injury. Traditional formulations of AAV preparations permit efficient viral transduction under physiologic conditions, but an improved understanding of the mechanistic limitations of AAV transduction may facilitate production of more resilient AAV strains for investigative and therapeutic purposes. We studied AAV transduction in the context of prior exposure of AAV serotype 8 (AAV8) to environmental pH within the range encountered during endosomal endocytosis (pH 7.4 to pH 4.4), during which low pH-triggered structural and autoproteolytic changes to the viral capsid are believed to be necessary for endosome escape and virus uncoating. Due to the fundamental nature of these processes, we hypothesized that premature exposure of AAV8 particles to acidic pH would decrease viral transduction of HT1080 cells in vitro, as measured by fluorescent reporter gene expression using high-content imaging analysis. We found that increasingly acidic incubation conditions were associated with concomitant reductions in transduction efficiency, and that quantitative levels of reporter gene expression in transduced cells were similarly decreased. The biggest decrease in transduction occurred between pH 7.4 and pH 6.4, suggesting the possible co-occurrence of a pH-associated event and viral inactivation within that range. Taken together, these findings indicate that exposure of AAV8 to acidic pH for as little as 1 hour is deleterious to transduction ability. Future studies are necessary to understand the pH-associated causative mechanisms involved. This study was approved by the University of Miami Institutional Animal Care and Use Committee, USA (Protocol #18-108-LF) on July 12, 2018.

Key words: adeno-associated virus, autolytic, capsid, low pH, protease, proteolytic, temperature, trafficking, transduction, viral inactivation