Elsevier

Current Opinion in Virology

Volume 18, June 2016, Pages 89-96
Current Opinion in Virology

Engineering AAV receptor footprints for gene therapy

https://doi.org/10.1016/j.coviro.2016.05.001Get rights and content

Highlights

  • Recognition of host receptors is a crucial determinant of AAV cell entry and tropism.

  • Several AAV capsid–glycan interactions have been identified and characterized.

  • Structural studies have mapped glycan receptor footprints on the AAV capsid surface.

  • Engineering glycan receptor footprints can yield new, synthetic AAV strains with altered tropisms and favorable transduction profiles.

Adeno-associated viruses (AAV) are currently at the forefront of human gene therapy clinical trials as recombinant vectors. Significant progress has been made in elucidating the structure, biology and tropisms of different naturally occurring AAV isolates in the past decade. In particular, a spectrum of AAV capsid interactions with host receptors have been identified and characterized. These studies have enabled a better understanding of key determinants of AAV cell recognition and entry in different hosts. This knowledge is now being applied toward engineering new, lab-derived AAV capsids with favorable transduction profiles. The current review conveys a structural perspective of capsid–glycan interactions and provides a roadmap for generating synthetic strains by engineering AAV receptor footprints.

Introduction

The last decade has witnessed the transition of Adeno-associated viruses (AAV) from gene transfer vectors to clinically viable, therapeutic candidates. Discovered as a contaminant in Adenovirus preparations during the mid 1960s [1], at first AAV failed to garner much clinical interest due to its complete lack of pathogenicity [2]. AAV is helper-dependent, meaning its replication relies on the presence of factors from a helper virus, such as adenovirus or herpes simplex virus [3, 4]. As a member of the Dependovirus genus of Parvoviridae, AAV has a T = 1 icosahedral capsid, ∼25 nm in diameter. The 4.7 kb single-stranded DNA genome contains two open reading frames: Rep and Cap [5]. Rep encodes four alternatively spliced proteins for viral genome packaging and replication, while Cap, encodes the capsid structural proteins VP1, VP2, and VP3 along with the Assembly Activating Protein (AAP) [5, 6, 7]. VP1, VP2, and VP3 share a common beta barrel domain, but differ in their N-terminal extension [5, 6, 7].

The AAV genome is flanked by inverted terminal repeats (ITRs), which are crucial for packaging, replication, and integration into the host genome. The ITRs represent the only essential cis-acting element of the AAV genome, enabling the incorporation of different transgenes between the ITRs, which can then be packaged into AAV capsids to generate recombinant AAV vectors (rAAV) [8]. Recombinant AAVs can be generated via transfection of plasmids containing the ITR-flanked transgene, an AAV capsid gene, and necessary helper genes, followed by harvest of transfected cells and purification of virus [5, 6, 7]. The application of rAAVs in gene therapy applications has rapidly mushroomed over the past decade. During this time, significant improvements to rAAV technology have been made with the goal of improving gene transfer efficiency, altering tropism and/or reducing antigenicity [9, 10]. Structural studies of AAV capsids combined with rational mutagenesis as well as combinatorial protein engineering strategies combined with directed evolution have yielded several lab-derived AAV strains [9, 10]. Here, we specifically review AAV capsid engineering strategies focused on exploiting knowledge of AAV capsid structure and interactions with host cell surface glycan receptors. Potential future studies focused on re-engineering other AAV–receptor footprints are also discussed.

Section snippets

AAV and host receptors

Several distinct AAV serotypes and variants have been discovered and isolated from different animal species including humans over the past decade [9, 11, 12, 13, 14]. It is now well known that different AAV isolates can utilize a variety of cell surface glycans for cell surface binding, including heparan sulfate (HS), N-linked sialic acids (SIA), O-linked sialic acids or mucins (MUC), or galactose (GAL) [5, 15, 16, 17, 18, 19]. Some naturally occurring AAV serotypes recognize two different

Structural determinants of AAV glycan interactions

The VP3 capsid protein subunit of AAV consists of 8 antiparallel beta strands forming a barrel. The beta strands are interconnected by variable loops, which are localized on the capsid surface. The VP1, VP2 and VP3 monomers assemble with twofold, threefold, and fivefold axes of symmetry to create the 60-subunit AAV capsid [5, 7, 34, 35]. The twofold axis of symmetry is characterized by depressions, where the variable loops form the barrier between these depressions [5]. The variable loops also

Re-engineering AAV capsid glycan interactions

A thorough structural understanding of AAV capsid glycan interactions has enabled rational manipulation of glycan footprints on the AAV capsid surface. This re-engineering approach has yielded novel, synthetic AAV strains with potential applications in therapeutic gene transfer. Specifically, structure-inspired design has been utilized to abrogate capsid binding to glycan receptors, alter binding affinity, and more recently engineer orthogonal glycan receptor interactions. Structural diagrams

Conflict of interest

Aravind Asokan is a co-founder at Stridebio LLC and an inventor on patents owned by UNC-CH.

References and recommended reading

Papers of particular interest, published within the period of review, have been highlighted as:

  • • of special interest

  • •• of outstanding interest

Acknowledgements

We would like to acknowledge funding support from the NIH (R01HL089221; P01HL112761 awarded to AA and training grant T32GM007092 to VM).

References (59)

  • F. Zhang et al.

    Characterization of interactions between heparin/glycosaminoglycan and adeno-associated virus

    Biochemistry

    (2013)
  • T.F. Lerch et al.

    The structure of adeno-associated virus serotype 3B (AAV-3B): insights into receptor binding and immune evasion

    Virology

    (2010)
  • M.A. Dimattia et al.

    Structural insight into the unique properties of adeno-associated virus serotype 9

    J Virol

    (2012)
  • C. Xiao et al.

    Interpretation of electron density with stereographic roadmap projections

    J Struct Biol

    (2007)
  • R.W. Atchison et al.

    Adenovirus-associated defective virus particles

    Science

    (1965)
  • N. Alazard-Dany et al.

    Definition of herpes simplex virus type 1 helper activities for adeno-associated virus early replication events

    PLoS Pathog

    (2009)
  • M.C. Geoffroy et al.

    Helper functions required for wild type and recombinant adeno-associated virus growth

    Curr Gene Ther

    (2005)
  • M. Agbandje-McKenna et al.

    AAV capsid structure and cell interactions

  • K. Van Vliet et al.

    The role of the adeno-associated virus capsid in gene transfer

  • M. Salganik et al.

    Adeno-associated virus as a mammalian DNA vector

    Microbiol Spectr

    (2015)
  • E. Hastie et al.

    Adeno-associated virus at 50: a golden anniversary of discovery, research, and gene therapy success  a personal perspective

    Hum Gene Ther

    (2015)
  • S. Mori et al.

    Two novel adeno-associated viruses from cynomolgus monkey: pseudotyping characterization of capsid protein

    Virology

    (2004)
  • I. Bossis et al.

    Cloning of an avian adeno-associated virus (AAAV) and generation of recombinant AAAV particles

    J Virol

    (2003)
  • A. Bello et al.

    Isolation and evaluation of novel adeno-associated virus sequences from porcine tissues

    Gene Ther

    (2009)
  • L.J. Ströh et al.

    Glycan engagement by viruses: receptor switches and specificity

    Annu Rev Virol

    (2014)
  • Z. Wu et al.

    α2,3 and α2,6 N-linked sialic acids facilitate efficient binding and transduction by adeno-associated virus types 1 and 6

    J Virol

    (2006)
  • S. Shen et al.

    Terminal N-linked galactose is the primary receptor for adeno-associated virus 9

    J Biol Chem

    (2011)
  • R.W. Walters et al.

    Binding of adeno-associated virus type 5 to 2,3-linked sialic acid is required for gene transfer

    J Biol Chem

    (2001)
  • K. Quinn et al.

    Intranasal administration of adeno-associated virus type 12 (AAV12) leads to transduction of the nasal epithelia and can initiate transgene-specific immune response

    Mol Ther

    (2009)
  • Cited by (0)

    View full text