15
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      Adeno-associated Virus (AAV) Serotypes Have Distinctive Interactions with Domains of the Cellular AAV Receptor

      research-article

      Read this article at

      Bookmark
          There is no author summary for this article yet. Authors can add summaries to their articles on ScienceOpen to make them more accessible to a non-specialist audience.

          ABSTRACT

          Adeno-associated virus (AAV) entry is determined by its interactions with specific surface glycans and a proteinaceous receptor(s). Adeno-associated virus receptor (AAVR) (also named KIAA0319L) is an essential cellular receptor required for the transduction of vectors derived from multiple AAV serotypes, including the evolutionarily distant serotypes AAV2 and AAV5. Here, we further biochemically characterize the AAV-AAVR interaction and define the domains within the ectodomain of AAVR that facilitate this interaction. By using a virus overlay assay, it was previously shown that the major AAV2 binding protein in membrane preparations of human cells corresponds to a glycoprotein with a molecular mass of 150 kDa. By establishing a purification procedure, performing further protein separation by two-dimensional electrophoresis, and utilizing mass spectrometry, we now show that this glycoprotein is identical to AAVR. While we find that AAVR is an N-linked glycosylated protein, this glycosylation is not a strict requirement for AAV2 binding or functional transduction. Using a combination of genetic complementation with deletion constructs and virus overlay assays with individual domains, we find that AAV2 functionally interacts predominantly with the second Ig-like polycystic kidney disease (PKD) repeat domain (PKD2) present in the ectodomain of AAVR. In contrast, AAV5 interacts primarily through the first, most membrane-distal, PKD domain (PKD1) of AAVR to promote transduction. Furthermore, other AAV serotypes, including AAV1 and -8, require a combination of PKD1 and PKD2 for optimal transduction. These results suggest that despite their shared dependence on AAVR as a critical entry receptor, different AAV serotypes have evolved distinctive interactions with the same receptor.

          IMPORTANCE Over the past decade, AAV vectors have emerged as leading gene delivery tools for therapeutic applications and biomedical research. However, fundamental aspects of the AAV life cycle, including how AAV interacts with host cellular factors to facilitate infection, are only partly understood. In particular, AAV receptors contribute significantly to AAV vector transduction efficiency and tropism. The recently identified AAV receptor (AAVR) is a key host receptor for multiple serotypes, including the most studied serotype, AAV2. AAVR binds directly to AAV2 particles and is rate limiting for viral transduction. Defining the AAV-AAVR interface in more detail is important to understand how AAV engages with its cellular receptor and how the receptor facilitates the entry process. Here, we further define AAV-AAVR interactions, genetically and biochemically, and show that different AAV serotypes have discrete interactions with the Ig-like PKD domains of AAVR. These findings reveal an unexpected divergence of AAVR engagement within these parvoviruses.

          Related collections

          Most cited references44

          • Record: found
          • Abstract: found
          • Article: not found

          High resolution two-dimensional electrophoresis of proteins.

          A technique has been developed for the separation of proteins by two-dimensional polyacrylamide gel electrophoresis. Due to its resolution and sensitivity, this technique is a powerful tool for the analysis and detection of proteins from complex biological sources. Proteins are separated according to isoelectric point by isoelectric focusing in the first dimension, and according to molecular weight by sodium dodecyl sulfate electrophoresis in the second dimension. Since these two parameters are unrelated, it is possible to obtain an almost uniform distribution of protein spots across a two-diminsional gel. This technique has resolved 1100 different components from Escherichia coli and should be capable of resolving a maximum of 5000 proteins. A protein containing as little as one disintegration per min of either 14C or 35S can be detected by autoradiography. A protein which constitutes 10 minus 4 to 10 minus 5% of the total protein can be detected and quantified by autoradiography. The reproducibility of the separation is sufficient to permit each spot on one separation to be matched with a spot on a different separation. This technique provides a method for estimation (at the described sensitivities) of the number of proteins made by any biological system. This system can resolve proteins differing in a single charge and consequently can be used in the analysis of in vivo modifications resulting in a change in charge. Proteins whose charge is changed by missense mutations can be identified. A detailed description of the methods as well as the characteristics of this system are presented.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            Novel adeno-associated viruses from rhesus monkeys as vectors for human gene therapy.

            Tissues from rhesus monkeys were screened by PCR for the presence of sequences homologous to known adeno-associated virus (AAV) serotypes 1-6. DNA spanning entire rep-cap ORFs from two novel AAVs, called AAV7 and AAV8, were isolated. Sequence comparisons among these and previously described AAVs revealed the greatest divergence in capsid proteins. AAV7 and AAV8 were not neutralized by heterologous antisera raised to the other serotypes. Neutralizing antibodies to AAV7 and AAV8 were rare in human serum and, when present, were low in activity. Vectors formed with capsids from AAV7 and AAV8 were generated by using rep and inverted terminal repeats (ITRs) from AAV2 and were compared with similarly constructed vectors made from capsids of AAV1, AAV2, and AAV5. Murine models of skeletal muscle and liver-directed gene transfer were used to evaluate relative vector performance. AAV7 vectors demonstrated efficiencies of transgene expression in skeletal muscle equivalent to that observed with AAV1, the most efficient known serotype for this application. In liver, transgene expression was 10- to 100-fold higher with AAV8 than observed with other serotypes. This improved efficiency correlated with increased persistence of vector DNA and higher number of transduced hepatocytes. The efficiency of AAV8 vector for liver-directed gene transfer of factor IX was not impacted by preimmunization with the other AAV serotypes. Vectors based on these novel, nonhuman primate AAVs should be considered for human gene therapy because of low reactivity to antibodies directed to human AAVs and because gene transfer efficiency in muscle was similar to that obtained with the best known serotype, whereas, in liver, gene transfer was substantially higher than previously described.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              Clades of Adeno-associated viruses are widely disseminated in human tissues.

              The potential for using Adeno-associated virus (AAV) as a vector for human gene therapy has stimulated interest in the Dependovirus genus. Serologic data suggest that AAV infections are prevalent in humans, although analyses of viruses and viral sequences from clinical samples are extremely limited. Molecular techniques were used in this study to successfully detect endogenous AAV sequences in 18% of all human tissues screened, with the liver and bone marrow being the most predominant sites. Sequence characterization of rescued AAV DNAs indicated a diverse array of molecular forms which segregate into clades whose members share functional and serologic similarities. One of the most predominant human clades is a hybrid of two previously described AAV serotypes, while another clade was found in humans and several species of nonhuman primates, suggesting a cross-species transmission of this virus. These data provide important information regarding the biology of parvoviruses in humans and their use as gene therapy vectors.
                Bookmark

                Author and article information

                Contributors
                Role: Editor
                Journal
                J Virol
                J. Virol
                jvi
                jvi
                JVI
                Journal of Virology
                American Society for Microbiology (1752 N St., N.W., Washington, DC )
                0022-538X
                1098-5514
                5 July 2017
                24 August 2017
                15 September 2017
                24 August 2017
                : 91
                : 18
                : e00391-17
                Affiliations
                [a ]Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, California, USA
                [b ]Department of Microbiology, Molecular Genetics and Immunology, University of Kansas Medical Center, Kansas City, Kansas, USA
                [c ]Department of Biochemistry and Molecular Biology, School of Medicine, Oregon Health & Science University, Portland, Oregon, USA
                [d ]Department of Anatomy and Cell Biology, University of Iowa, Iowa City, Iowa, USA
                [e ]Virus Reference Department, Public Health England, Colindale, London, United Kingdom
                International Centre for Genetic Engineering and Biotechnology
                Author notes
                Address correspondence to Jianming Qiu, jqiu@ 123456kumc.edu , or Jan E. Carette, carette@ 123456stanford.edu .

                S.P., W.Z., J.Q., and J.E.C. contributed equally.

                Citation Pillay S, Zou W, Cheng F, Puschnik AS, Meyer NL, Ganaie SS, Deng X, Wosen JE, Davulcu O, Yan Z, Engelhardt JF, Brown KE, Chapman MS, Qiu J, Carette JE. 2017. Adeno-associated virus (AAV) serotypes have distinctive interactions with domains of the cellular AAV receptor. J Virol 91:e00391-17. https://doi.org/10.1128/JVI.00391-17.

                Article
                00391-17
                10.1128/JVI.00391-17
                5571256
                28679762
                365e0376-04bc-4499-8464-6e8f1e9b5054
                Copyright © 2017 Pillay et al.

                This is an open-access article distributed under the terms of the Creative Commons Attribution 4.0 International license.

                History
                : 8 March 2017
                : 27 June 2017
                Page count
                Figures: 8, Tables: 0, Equations: 0, References: 56, Pages: 17, Words: 9858
                Funding
                Funded by: Stanford SPARK program
                Award ID: Pilot
                Award Recipient : Sirika Pillay Award Recipient : Andreas S. Puschnik
                Funded by: Weston Havens Foundation
                Award ID: Pilot
                Award Recipient : Sirika Pillay Award Recipient : Andreas S. Puschnik
                Funded by: HHS | National Institutes of Health (NIH) https://doi.org/10.13039/100000002
                Award ID: RO1GM066875
                Award ID: R35GM122564
                Award ID: DP2AI104557
                Award ID: U19AI109662
                Award ID: RO1AI070723
                Award ID: R21AI112803
                Award ID: PO1HL051670
                Award Recipient : John Engelhardt Award Recipient : Michael S. Chapman Award Recipient : Jianming Qiu Award Recipient : Jan E. Carette
                Funded by: Cystic Fibrosis Foundation (CF Foundation) https://doi.org/10.13039/100000897
                Award ID: YAN15XX0
                Award Recipient : Ziying Yan
                Funded by: David and Lucile Packard Foundation (David & Lucile Packard Foundation) https://doi.org/10.13039/100000008
                Award ID: Fellow
                Award Recipient : Jan E. Carette
                Categories
                Virus-Cell Interactions
                Spotlight
                Custom metadata
                September 2017

                Microbiology & Virology
                aavr,adeno-associated virus,gene therapy,receptor-ligand interaction,viral receptor,virus overlay assay,virus-host interactions

                Comments

                Comment on this article