scholarly journals RFX1 and RFX3 Transcription Factors Interact with the D Sequence of Adeno-Associated Virus Inverted Terminal Repeat and Regulate AAV Transduction

2018 ◽  
Vol 8 (1) ◽  
Author(s):  
Laura Julien ◽  
Julie Chassagne ◽  
Cécile Peccate ◽  
Stéphanie Lorain ◽  
France Piétri-Rouxel ◽  
...  
2020 ◽  
Vol 31 (3-4) ◽  
pp. 151-162 ◽  
Author(s):  
Lauriel F. Earley ◽  
Laura M. Conatser ◽  
Victoria M. Lue ◽  
Amanda L. Dobbins ◽  
Chengwen Li ◽  
...  

2011 ◽  
pp. 110805114044008 ◽  
Author(s):  
Christine Aurnhammer ◽  
Maren Haase ◽  
Nadine Muether ◽  
Martin Hausl ◽  
Christina Rauschhuber ◽  
...  

Virology ◽  
2004 ◽  
Vol 323 (2) ◽  
pp. 220-232 ◽  
Author(s):  
Priya Chikhlikar ◽  
Luciana Barros de Arruda ◽  
Shikha Agrawal ◽  
Barry Byrne ◽  
William Guggino ◽  
...  

2002 ◽  
Vol 76 (24) ◽  
pp. 12792-12802 ◽  
Author(s):  
Sergei Musatov ◽  
Jill Roberts ◽  
Donald Pfaff ◽  
Michael Kaplitt

ABSTRACT A novel pathway of adeno-associated virus (AAV) replication marked by the assembly of circular monomer duplex intermediates (cAAV) has been recently discovered. In the present report we identify a single AD domain of the inverted terminal repeat as a minimal origin of cAAV replication. A small internal palindrome (BB′), necessary for optimal Rep-inverted terminal repeat interaction, does not contribute to the efficiency of cAAV replication, while the terminal resolution site is an essential cis-acting element. Furthermore, recombinant cAAV vectors that encompass only the AD domain replicate exclusively in a circular form and no detectable linear duplex replicative intermediates are generated, suggesting that both pathways of AAV replication are independent and can be separated. In addition, we show that cAAVs are efficient templates for encapsidation of single-stranded DNA genomes, an observation that assigns a biological role for these novel replication species. Together, these findings shed new light on the current model of AAV replication and packaging.


2005 ◽  
Vol 79 (1) ◽  
pp. 364-379 ◽  
Author(s):  
Ziying Yan ◽  
Roman Zak ◽  
Yulong Zhang ◽  
John F. Engelhardt

ABSTRACT The relatively small package capacity (less than 5 kb) of adeno-associated virus (AAV) vectors has been effectively doubled with the development of dual-vector heterodimerization approaches. However, the efficiency of such dual-vector systems is limited not only by the extent to which intermolecular recombination occurs between two independent vector genomes, but also by the directional bias required for successful transgene reconstitution following concatemerization. In the present study, we sought to evaluate the mechanisms by which inverted terminal repeat (ITR) sequences mediate intermolecular recombination of AAV genomes, with the goal of engineering more efficient vectors for dual-vector trans-splicing approaches. To this end, we generated a novel AAV hybrid-ITR vector characterized by an AAV-2 and an AAV-5 ITR at opposite ends of the viral genome. This hybrid genome was efficiently packaged into either AAV-2 or AAV-5 capsids to generate infectious virions. Hybrid AV2:5 ITR viruses had a significantly lower capacity to form circular intermediates in infected cells than homologous AV2:2 and AV5:5 ITR vectors despite their similar capacity to express an encoded enhanced green fluorescent protein (EGFP) transgene. To examine whether the divergent ITR sequences contained within hybrid AV2:5 ITR vectors could direct intermolecular recombination in a tail-to-head fashion, we generated two hybrid ITR trans-splicing vectors (AV5:2LacZdonor and AV2:5LacZacceptor). Each delivered one exon of a β-galactosidase minigene flanked by donor or acceptor splice sequences. These hybrid trans-splicing vectors were compared to homologous AV5:5 and AV2:2 trans-splicing vector sets for their ability to reconstitute β-galactosidase gene expression. Results from this comparison demonstrated that hybrid ITR dual-vector sets had a significantly enhanced trans-splicing efficiency (6- to 10-fold, depending on the capsid serotype) compared to homologous ITR vectors. Molecular studies of viral genome structures suggest that hybrid ITR vectors provide more efficient directional recombination due to an increased abundance of linear-form genomes. These studies provide direct evidence for the importance of ITR sequences in directing intermolecular and intramolecular homologous recombination of AAV genomes. The use of hybrid ITR AAV vector genomes provides new strategies to manipulate viral genome conversion products and to direct intermolecular recombination events required for efficient dual-AAV vector reconstitution of the transgene.


Sign in / Sign up

Export Citation Format

Share Document