Identification of adeno-associated virus variants for gene transfer into human neural cell types by parallel capsid screening

Abstract Human brain cells generated by in vitro cell programming provide exciting prospects for disease modeling, drug discovery and cell therapy. These applications frequently require efficient and clinically compliant tools for genetic modification of the cells. Recombinant adeno-associated virus...

Full description

Bibliographic Details
Main Authors: Lea Jessica Flitsch, Kathleen Börner, Christian Stüllein, Simon Ziegler, Vera Sonntag-Buck, Ellen Wiedtke, Vesselina Semkova, Si Wah Christina Au Yeung, Julia Schlee, Mohamad Hajo, Mona Mathews, Beatrice Stefanie Ludwig, Susanne Kossatz, Horst Kessler, Dirk Grimm, Oliver Brüstle
Format: Article
Language:English
Published: Nature Portfolio 2022-05-01
Series:Scientific Reports
Online Access:https://doi.org/10.1038/s41598-022-12404-0
_version_ 1811250849634058240
author Lea Jessica Flitsch
Kathleen Börner
Christian Stüllein
Simon Ziegler
Vera Sonntag-Buck
Ellen Wiedtke
Vesselina Semkova
Si Wah Christina Au Yeung
Julia Schlee
Mohamad Hajo
Mona Mathews
Beatrice Stefanie Ludwig
Susanne Kossatz
Horst Kessler
Dirk Grimm
Oliver Brüstle
author_facet Lea Jessica Flitsch
Kathleen Börner
Christian Stüllein
Simon Ziegler
Vera Sonntag-Buck
Ellen Wiedtke
Vesselina Semkova
Si Wah Christina Au Yeung
Julia Schlee
Mohamad Hajo
Mona Mathews
Beatrice Stefanie Ludwig
Susanne Kossatz
Horst Kessler
Dirk Grimm
Oliver Brüstle
author_sort Lea Jessica Flitsch
collection DOAJ
description Abstract Human brain cells generated by in vitro cell programming provide exciting prospects for disease modeling, drug discovery and cell therapy. These applications frequently require efficient and clinically compliant tools for genetic modification of the cells. Recombinant adeno-associated viruses (AAVs) fulfill these prerequisites for a number of reasons, including the availability of a myriad of AAV capsid variants with distinct cell type specificity (also called tropism). Here, we harnessed a customizable parallel screening approach to assess a panel of natural or synthetic AAV capsid variants for their efficacy in lineage-related human neural cell types. We identified common lead candidates suited for the transduction of directly converted, early-stage induced neural stem cells (iNSCs), induced pluripotent stem cell (iPSC)-derived later-stage, radial glia-like neural progenitors, as well as differentiated astrocytic and mixed neuroglial cultures. We then selected a subset of these candidates for functional validation in iNSCs and iPSC-derived astrocytes, using shRNA-induced downregulation of the citrate transporter SLC25A1 and overexpression of the transcription factor NGN2 for proofs-of-concept. Our study provides a comparative overview of the susceptibility of different human cell programming-derived brain cell types to AAV transduction and a critical discussion of the assets and limitations of this specific AAV capsid screening approach.
first_indexed 2024-04-12T16:10:54Z
format Article
id doaj.art-4875817cf7bc4d6497c93a0dff7c11d4
institution Directory Open Access Journal
issn 2045-2322
language English
last_indexed 2024-04-12T16:10:54Z
publishDate 2022-05-01
publisher Nature Portfolio
record_format Article
series Scientific Reports
spelling doaj.art-4875817cf7bc4d6497c93a0dff7c11d42022-12-22T03:25:54ZengNature PortfolioScientific Reports2045-23222022-05-0112111210.1038/s41598-022-12404-0Identification of adeno-associated virus variants for gene transfer into human neural cell types by parallel capsid screeningLea Jessica Flitsch0Kathleen Börner1Christian Stüllein2Simon Ziegler3Vera Sonntag-Buck4Ellen Wiedtke5Vesselina Semkova6Si Wah Christina Au Yeung7Julia Schlee8Mohamad Hajo9Mona Mathews10Beatrice Stefanie Ludwig11Susanne Kossatz12Horst Kessler13Dirk Grimm14Oliver Brüstle15Institute of Reconstructive Neurobiology, University of Bonn Medical Faculty and University Hospital BonnCenter for Infectious Diseases, Virology, Medical Faculty, Heidelberg UniversityCLADIAC GmbHCLADIAC GmbHCenter for Infectious Diseases, Virology, Medical Faculty, Heidelberg UniversityCenter for Infectious Diseases, Virology, Medical Faculty, Heidelberg UniversityInstitute of Reconstructive Neurobiology, University of Bonn Medical Faculty and University Hospital BonnInstitute of Reconstructive Neurobiology, University of Bonn Medical Faculty and University Hospital BonnInstitute of Reconstructive Neurobiology, University of Bonn Medical Faculty and University Hospital BonnInstitute of Reconstructive Neurobiology, University of Bonn Medical Faculty and University Hospital BonnLIFE and BRAIN GmbHDepartment of Nuclear Medicine, School of Medicine, Technical University Munich (TUM), University Hospital Klinikum Rechts der Isar and Central Institute for Translational Cancer Research (Transla TUMDepartment of Nuclear Medicine, School of Medicine, Technical University Munich (TUM), University Hospital Klinikum Rechts der Isar and Central Institute for Translational Cancer Research (Transla TUMInstitute for Advanced Study, Department Chemie, Technical University Munich (TUM)Center for Infectious Diseases, Virology, Medical Faculty, Heidelberg UniversityInstitute of Reconstructive Neurobiology, University of Bonn Medical Faculty and University Hospital BonnAbstract Human brain cells generated by in vitro cell programming provide exciting prospects for disease modeling, drug discovery and cell therapy. These applications frequently require efficient and clinically compliant tools for genetic modification of the cells. Recombinant adeno-associated viruses (AAVs) fulfill these prerequisites for a number of reasons, including the availability of a myriad of AAV capsid variants with distinct cell type specificity (also called tropism). Here, we harnessed a customizable parallel screening approach to assess a panel of natural or synthetic AAV capsid variants for their efficacy in lineage-related human neural cell types. We identified common lead candidates suited for the transduction of directly converted, early-stage induced neural stem cells (iNSCs), induced pluripotent stem cell (iPSC)-derived later-stage, radial glia-like neural progenitors, as well as differentiated astrocytic and mixed neuroglial cultures. We then selected a subset of these candidates for functional validation in iNSCs and iPSC-derived astrocytes, using shRNA-induced downregulation of the citrate transporter SLC25A1 and overexpression of the transcription factor NGN2 for proofs-of-concept. Our study provides a comparative overview of the susceptibility of different human cell programming-derived brain cell types to AAV transduction and a critical discussion of the assets and limitations of this specific AAV capsid screening approach.https://doi.org/10.1038/s41598-022-12404-0
spellingShingle Lea Jessica Flitsch
Kathleen Börner
Christian Stüllein
Simon Ziegler
Vera Sonntag-Buck
Ellen Wiedtke
Vesselina Semkova
Si Wah Christina Au Yeung
Julia Schlee
Mohamad Hajo
Mona Mathews
Beatrice Stefanie Ludwig
Susanne Kossatz
Horst Kessler
Dirk Grimm
Oliver Brüstle
Identification of adeno-associated virus variants for gene transfer into human neural cell types by parallel capsid screening
Scientific Reports
title Identification of adeno-associated virus variants for gene transfer into human neural cell types by parallel capsid screening
title_full Identification of adeno-associated virus variants for gene transfer into human neural cell types by parallel capsid screening
title_fullStr Identification of adeno-associated virus variants for gene transfer into human neural cell types by parallel capsid screening
title_full_unstemmed Identification of adeno-associated virus variants for gene transfer into human neural cell types by parallel capsid screening
title_short Identification of adeno-associated virus variants for gene transfer into human neural cell types by parallel capsid screening
title_sort identification of adeno associated virus variants for gene transfer into human neural cell types by parallel capsid screening
url https://doi.org/10.1038/s41598-022-12404-0
work_keys_str_mv AT leajessicaflitsch identificationofadenoassociatedvirusvariantsforgenetransferintohumanneuralcelltypesbyparallelcapsidscreening
AT kathleenborner identificationofadenoassociatedvirusvariantsforgenetransferintohumanneuralcelltypesbyparallelcapsidscreening
AT christianstullein identificationofadenoassociatedvirusvariantsforgenetransferintohumanneuralcelltypesbyparallelcapsidscreening
AT simonziegler identificationofadenoassociatedvirusvariantsforgenetransferintohumanneuralcelltypesbyparallelcapsidscreening
AT verasonntagbuck identificationofadenoassociatedvirusvariantsforgenetransferintohumanneuralcelltypesbyparallelcapsidscreening
AT ellenwiedtke identificationofadenoassociatedvirusvariantsforgenetransferintohumanneuralcelltypesbyparallelcapsidscreening
AT vesselinasemkova identificationofadenoassociatedvirusvariantsforgenetransferintohumanneuralcelltypesbyparallelcapsidscreening
AT siwahchristinaauyeung identificationofadenoassociatedvirusvariantsforgenetransferintohumanneuralcelltypesbyparallelcapsidscreening
AT juliaschlee identificationofadenoassociatedvirusvariantsforgenetransferintohumanneuralcelltypesbyparallelcapsidscreening
AT mohamadhajo identificationofadenoassociatedvirusvariantsforgenetransferintohumanneuralcelltypesbyparallelcapsidscreening
AT monamathews identificationofadenoassociatedvirusvariantsforgenetransferintohumanneuralcelltypesbyparallelcapsidscreening
AT beatricestefanieludwig identificationofadenoassociatedvirusvariantsforgenetransferintohumanneuralcelltypesbyparallelcapsidscreening
AT susannekossatz identificationofadenoassociatedvirusvariantsforgenetransferintohumanneuralcelltypesbyparallelcapsidscreening
AT horstkessler identificationofadenoassociatedvirusvariantsforgenetransferintohumanneuralcelltypesbyparallelcapsidscreening
AT dirkgrimm identificationofadenoassociatedvirusvariantsforgenetransferintohumanneuralcelltypesbyparallelcapsidscreening
AT oliverbrustle identificationofadenoassociatedvirusvariantsforgenetransferintohumanneuralcelltypesbyparallelcapsidscreening