Abstract

Rabies pseudotyped lentiviral vectors have great potential in gene therapy, not least because of their ability to transduce neurons following their distal axonal application. However, very little is known about the molecular processes that underlie their retrograde transport and cell transduction. Using multiple labeling techniques and confocal microscopy, we demonstrated that pseudotyping with rabies virus envelope glycoprotein (RV-G) enabled the axonal retrograde transport of two distinct subtypes of lentiviral vector in motor neuron cultures. Analysis of this process revealed that these vectors trafficked through Rab5-positive endosomes and accumulated within a non-acidic Rab7 compartment. RV-G pseudotyped vectors were co-transported with both the tetanus neurotoxin-binding fragment and the membrane proteins thought to mediate rabies virus endocytosis (neural cell adhesion molecule, nicotinic acetylcholine receptor, and p75 neurotrophin receptor), thus demonstrating that pseudotyping with RV-G targets lentiviral vectors for transport along the same pathway exploited by several toxins and viruses. Using motor neurons cultured in compartmentalized chambers, we demonstrated that axonal retrograde transport of these vectors was rapid and efficient; however, it was not able to transduce the targeted neurons efficiently, suggesting that impairment in processes occurring after arrival of the viral vector in the soma is responsible for the low transduction efficiency seen in vivo, which suggests a novel area for improvement of gene therapy vectors.

Highlights

  • Trafficking pathways underlying retrograde transduction of neurons with lentiviral vectors carrying the rabies G glycoprotein are uncharted, and barriers to high transduction efficiencies are undefined

  • Using multiple labeling techniques and confocal microscopy, we demonstrated that pseudotyping with rabies virus envelope glycoprotein (RV-G) enabled the axonal retrograde transport of two distinct subtypes of lentiviral vector in motor neuron cultures

  • Using motor neurons cultured in compartmentalized chambers, we demonstrated that axonal retrograde transport of these vectors was rapid and efficient; it was not able to transduce the targeted neurons efficiently, suggesting that impairment in processes occurring after arrival of the viral vec

Read more

Summary

Background

Trafficking pathways underlying retrograde transduction of neurons with lentiviral vectors carrying the rabies G glycoprotein are uncharted, and barriers to high transduction efficiencies are undefined. Using multiple labeling techniques and confocal microscopy, we demonstrated that pseudotyping with rabies virus envelope glycoprotein (RV-G) enabled the axonal retrograde transport of two distinct subtypes of lentiviral vector in motor neuron cultures. Analysis of this process revealed that these vectors trafficked through Rab5-positive endosomes and accumulated within a non-acidic Rab compartment. 2 Present address: School of Medical Sciences, College of Life Sciences and Medicine, University of Aberdeen, Inst. Use of this in vitro system, which parallels the in vivo intramuscular injection route, strongly suggests that the major block to efficient viral vector transduction is at a stage following axonal transport perhaps between endosomal escape and nuclear transport of the vector capsid

EXPERIMENTAL PROCEDURES
RESULTS
B EIAV gag MA
DISCUSSION
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