IDEAS home Printed from https://ideas.repec.org/a/nat/natcom/v11y2020i1d10.1038_s41467-020-19230-w.html
   My bibliography  Save this article

Identification of a myotropic AAV by massively parallel in vivo evaluation of barcoded capsid variants

Author

Listed:
  • Jonas Weinmann

    (Heidelberg University Hospital, Dept. of Infectious Diseases/Virology, Cluster of Excellence CellNetworks
    University of Heidelberg)

  • Sabrina Weis

    (Heidelberg University Hospital, Dept. of Infectious Diseases/Virology, Cluster of Excellence CellNetworks
    University of Heidelberg)

  • Josefine Sippel

    (Heidelberg University Hospital, Dept. of Infectious Diseases/Virology, Cluster of Excellence CellNetworks
    University of Heidelberg)

  • Warut Tulalamba

    (Vrije Universiteit Brussel, Department of Gene Therapy & Regenerative Medicine)

  • Anca Remes

    (University Hospital Schleswig-Holstein, Campus Kiel, Innere Medizin III
    German Center for Infection Research (DZIF) and German Center for Cardiovascular Research (DZHK), partner site Hamburg/Kiel/Lübeck)

  • Jihad El Andari

    (Heidelberg University Hospital, Dept. of Infectious Diseases/Virology, Cluster of Excellence CellNetworks
    University of Heidelberg)

  • Anne-Kathrin Herrmann

    (Heidelberg University Hospital, Dept. of Infectious Diseases/Virology, Cluster of Excellence CellNetworks
    University of Heidelberg)

  • Quang H. Pham

    (Vrije Universiteit Brussel, Department of Gene Therapy & Regenerative Medicine)

  • Christopher Borowski

    (University Hospital Schleswig-Holstein, Campus Kiel, Innere Medizin III
    German Center for Infection Research (DZIF) and German Center for Cardiovascular Research (DZHK), partner site Hamburg/Kiel/Lübeck)

  • Susanne Hille

    (University Hospital Schleswig-Holstein, Campus Kiel, Innere Medizin III
    German Center for Infection Research (DZIF) and German Center for Cardiovascular Research (DZHK), partner site Hamburg/Kiel/Lübeck)

  • Tanja Schönberger

    (Boehringer Ingelheim Pharma GmbH & Co. KG, Drug Discovery Sciences)

  • Norbert Frey

    (University Hospital Schleswig-Holstein, Campus Kiel, Innere Medizin III
    German Center for Infection Research (DZIF) and German Center for Cardiovascular Research (DZHK), partner site Hamburg/Kiel/Lübeck)

  • Martin Lenter

    (Boehringer Ingelheim Pharma GmbH & Co. KG, Drug Discovery Sciences)

  • Thierry VandenDriessche

    (Vrije Universiteit Brussel, Department of Gene Therapy & Regenerative Medicine
    University of Leuven, Center for Molecular & Vascular Biology, Department of Cardiovascular Sciences)

  • Oliver J. Müller

    (University Hospital Schleswig-Holstein, Campus Kiel, Innere Medizin III
    German Center for Infection Research (DZIF) and German Center for Cardiovascular Research (DZHK), partner site Hamburg/Kiel/Lübeck)

  • Marinee K. Chuah

    (Vrije Universiteit Brussel, Department of Gene Therapy & Regenerative Medicine
    University of Leuven, Center for Molecular & Vascular Biology, Department of Cardiovascular Sciences)

  • Thorsten Lamla

    (Boehringer Ingelheim Pharma GmbH & Co. KG, Drug Discovery Sciences)

  • Dirk Grimm

    (Heidelberg University Hospital, Dept. of Infectious Diseases/Virology, Cluster of Excellence CellNetworks
    University of Heidelberg
    German Center for Infection Research (DZIF) and German Center for Cardiovascular Research (DZHK), partner site Heidelberg)

Abstract

Adeno-associated virus (AAV) forms the basis for several commercial gene therapy products and for countless gene transfer vectors derived from natural or synthetic viral isolates that are under intense preclinical evaluation. Here, we report a versatile pipeline that enables the direct side-by-side comparison of pre-selected AAV capsids in high-throughput and in the same animal, by combining DNA/RNA barcoding with multiplexed next-generation sequencing. For validation, we create three independent libraries comprising 183 different AAV variants including widely used benchmarks and screened them in all major tissues in adult mice. Thereby, we discover a peptide-displaying AAV9 mutant called AAVMYO that exhibits superior efficiency and specificity in the musculature including skeletal muscle, heart and diaphragm following peripheral delivery, and that holds great potential for muscle gene therapy. Our comprehensive methodology is compatible with any capsids, targets and species, and will thus facilitate and accelerate the stratification of optimal AAV vectors for human gene therapy.

Suggested Citation

  • Jonas Weinmann & Sabrina Weis & Josefine Sippel & Warut Tulalamba & Anca Remes & Jihad El Andari & Anne-Kathrin Herrmann & Quang H. Pham & Christopher Borowski & Susanne Hille & Tanja Schönberger & No, 2020. "Identification of a myotropic AAV by massively parallel in vivo evaluation of barcoded capsid variants," Nature Communications, Nature, vol. 11(1), pages 1-12, December.
  • Handle: RePEc:nat:natcom:v:11:y:2020:i:1:d:10.1038_s41467-020-19230-w
    DOI: 10.1038/s41467-020-19230-w
    as

    Download full text from publisher

    File URL: https://www.nature.com/articles/s41467-020-19230-w
    File Function: Abstract
    Download Restriction: no

    File URL: https://libkey.io/10.1038/s41467-020-19230-w?utm_source=ideas
    LibKey link: if access is restricted and if your library uses this service, LibKey will redirect you to where you can use your library subscription to access this item
    ---><---

    Citations

    Citations are extracted by the CitEc Project, subscribe to its RSS feed for this item.
    as


    Cited by:

    1. Marco Thürkauf & Shuo Lin & Filippo Oliveri & Dirk Grimm & Randall J. Platt & Markus A. Rüegg, 2023. "Fast, multiplexable and efficient somatic gene deletions in adult mouse skeletal muscle fibers using AAV-CRISPR/Cas9," Nature Communications, Nature, vol. 14(1), pages 1-16, December.
    2. Trevor J. Gonzalez & Katherine E. Simon & Leo O. Blondel & Marco M. Fanous & Angela L. Roger & Maribel Santiago Maysonet & Garth W. Devlin & Timothy J. Smith & Daniel K. Oh & L. Patrick Havlik & Ruth , 2022. "Cross-species evolution of a highly potent AAV variant for therapeutic gene transfer and genome editing," Nature Communications, Nature, vol. 13(1), pages 1-17, December.
    3. Markus Grosch & Laura Schraft & Adrian Chan & Leonie Küchenhoff & Kleopatra Rapti & Anne-Maud Ferreira & Julia Kornienko & Shengdi Li & Michael H. Radke & Chiara Krämer & Sandra Clauder-Münster & Emer, 2023. "Striated muscle-specific base editing enables correction of mutations causing dilated cardiomyopathy," Nature Communications, Nature, vol. 14(1), pages 1-15, December.

    More about this item

    Statistics

    Access and download statistics

    Corrections

    All material on this site has been provided by the respective publishers and authors. You can help correct errors and omissions. When requesting a correction, please mention this item's handle: RePEc:nat:natcom:v:11:y:2020:i:1:d:10.1038_s41467-020-19230-w. See general information about how to correct material in RePEc.

    If you have authored this item and are not yet registered with RePEc, we encourage you to do it here. This allows to link your profile to this item. It also allows you to accept potential citations to this item that we are uncertain about.

    We have no bibliographic references for this item. You can help adding them by using this form .

    If you know of missing items citing this one, you can help us creating those links by adding the relevant references in the same way as above, for each refering item. If you are a registered author of this item, you may also want to check the "citations" tab in your RePEc Author Service profile, as there may be some citations waiting for confirmation.

    For technical questions regarding this item, or to correct its authors, title, abstract, bibliographic or download information, contact: Sonal Shukla or Springer Nature Abstracting and Indexing (email available below). General contact details of provider: http://www.nature.com .

    Please note that corrections may take a couple of weeks to filter through the various RePEc services.

    IDEAS is a RePEc service. RePEc uses bibliographic data supplied by the respective publishers.