Abstract

Dual vector AAV systems are being utilised to enable gene therapy for disorders in which the disease gene is too large to fit into a single capsid. Fragmented adeno-associated viral (fAAV) vectors containing single inverted terminal repeat truncated transgenes have been considered as one such gene replacement strategy. Here we aim to add to the current understanding of the molecular mechanisms employed by fAAV dual vector systems. Oversized (>8kb) transgene constructs containing ABCA4 coding sequence were packaged as truncated fragments <5kb in size into various AAV serotypes. In vitro transductions with these fAAV vector preparations were conducted with mRNA and protein expression products assessed by way of RT-PCR, qPCR and western blot techniques. Transductions with fAAV vector preparations yielded ABCA4 mRNA, but did not generate detectable levels of protein. Sequencing of the transcript population revealed the presence of full length ABCA4 CDS with additional hybrid ABCA4 variants, indicating truncated transgenes without regions of overlap were joining and forming stable hybrid transgenes. In contrast, an ABCA4 overlapping dual vector system (OV) with a defined complementary region generated only full length mRNA transcripts plus detectable ABCA4 protein. Despite previous success shown with the fAAV approach, the lack of repeatability and identification of stable hybrid transcripts capable of protein production suggests there is more refinement required before considering this approach in a clinical setting.

Highlights

  • IntroductionAdeno-associated virus (AAV) vectors are known to have an optimal packaging capacity of ~4.7 kb (the size of their native genome) but can be encouraged to package slightly larger transgenes [1]

  • Adeno-associated virus (AAV) vectors are known to have an optimal packaging capacity of ~4.7 kb but can be encouraged to package slightly larger transgenes [1]

  • Our results suggest that despite the success seen by other research groups employing this dual vector strategy, the lack of control in generating fragmented AAV (fAAV) dual vectors leads to unwanted hybrid expression products

Read more

Summary

Introduction

Adeno-associated virus (AAV) vectors are known to have an optimal packaging capacity of ~4.7 kb (the size of their native genome) but can be encouraged to package slightly larger transgenes [1]. With AAV showing encouraging safety and efficacy in human clinical trials, there is a desire to utilize this vector for the treatment of diseases for which the causative gene is larger than the packaging capacity. The replicated transgenes are truncated at the 5’ end at an undefined point (Figure 1a). This method provides no designated limit of packaging at the 5’ end of either the plus or minus strands of the transgene and results in a collection of AAV particles carrying transgenes of different length containing only a single ITR, referred to as fragmented AAV (fAAV) [5,6].

Methods
Results
Conclusion
Full Text
Published version (Free)

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