Abstract

We evaluate gene editing of HSV in a well-established mouse model, using adeno-associated virus (AAV)-delivered meganucleases, as a potentially curative approach to treat latent HSV infection. Here we show that AAV-delivered meganucleases, but not CRISPR/Cas9, mediate highly efficient gene editing of HSV, eliminating over 90% of latent virus from superior cervical ganglia. Single-cell RNA sequencing demonstrates that both HSV and individual AAV serotypes are non-randomly distributed among neuronal subsets in ganglia, implying that improved delivery to all neuronal subsets may lead to even more complete elimination of HSV. As predicted, delivery of meganucleases using a triple AAV serotype combination results in the greatest decrease in ganglionic HSV loads. The levels of HSV elimination observed in these studies, if translated to humans, would likely significantly reduce HSV reactivation, shedding, and lesions. Further optimization of meganuclease delivery and activity is likely possible, and may offer a pathway to a cure for HSV infection.

Highlights

  • We evaluate gene editing of Herpes simplex virus (HSV) in a well-established mouse model, using adeno-associated virus (AAV)-delivered meganucleases, as a potentially curative approach to treat latent HSV infection

  • We took advantage of a relatively simple mouse model of HSV infection to perform a set of iterative studies to increase the efficiency of AAV-delivered gene editing enzymes targeting HSV

  • While HSV does not reactivate spontaneously from ganglia of living mice, the virus does reactivate from mouse neurons after explantation, and our results demonstrate 95% (SCG) to 55% (TG) reduction in viral genomes produced de novo in ganglionic explants after meganuclease treatment of latently infected mice

Read more

Summary

Introduction

We evaluate gene editing of HSV in a well-established mouse model, using adeno-associated virus (AAV)-delivered meganucleases, as a potentially curative approach to treat latent HSV infection. We show that AAV-delivered meganucleases, but not CRISPR/Cas[9], mediate highly efficient gene editing of HSV, eliminating over 90% of latent virus from superior cervical ganglia. Gene editing using CRISPR/Cas[9], meganucleases, or similar enzymes offers the possibility of directly targeting latent genomes for disruption or elimination while preserving neurons, eliminating the possibility of viral reactivation and pathogenesis[13,14,15,16,17,18,19,20]. In vivo gene editing of latent HSV genomes within TG sensory neurons of mice has been previously demonstrated using HSV-specific meganucleases delivered via adenoassociated virus (AAV) vectors, but the levels of gene editing were modest (

Methods
Results
Conclusion
Full Text
Paper version not known

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call

Disclaimer: All third-party content on this website/platform is and will remain the property of their respective owners and is provided on "as is" basis without any warranties, express or implied. Use of third-party content does not indicate any affiliation, sponsorship with or endorsement by them. Any references to third-party content is to identify the corresponding services and shall be considered fair use under The CopyrightLaw.