Cargando…

Pseudotyped AAV Vector-Mediated Gene Transfer in a Human Fetal Trachea Xenograft Model: Implications for In Utero Gene Therapy for Cystic Fibrosis

BACKGROUND: Lung disease including airway infection and inflammation currently causes the majority of morbidities and mortalities associated with cystic fibrosis (CF), making the airway epithelium and the submucosal glands (SMG) novel target cells for gene therapy in CF. These target cells are relat...

Descripción completa

Detalles Bibliográficos
Autores principales: Keswani, Sundeep G., Balaji, Swathi, Le, Louis, Leung, Alice, Katz, Anna B., Lim, Foong-Yen, Habli, Mounira, Jones, Helen N., Wilson, James M., Crombleholme, Timothy M.
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Public Library of Science 2012
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC3427158/
https://www.ncbi.nlm.nih.gov/pubmed/22937069
http://dx.doi.org/10.1371/journal.pone.0043633
_version_ 1782241574637273088
author Keswani, Sundeep G.
Balaji, Swathi
Le, Louis
Leung, Alice
Katz, Anna B.
Lim, Foong-Yen
Habli, Mounira
Jones, Helen N.
Wilson, James M.
Crombleholme, Timothy M.
author_facet Keswani, Sundeep G.
Balaji, Swathi
Le, Louis
Leung, Alice
Katz, Anna B.
Lim, Foong-Yen
Habli, Mounira
Jones, Helen N.
Wilson, James M.
Crombleholme, Timothy M.
author_sort Keswani, Sundeep G.
collection PubMed
description BACKGROUND: Lung disease including airway infection and inflammation currently causes the majority of morbidities and mortalities associated with cystic fibrosis (CF), making the airway epithelium and the submucosal glands (SMG) novel target cells for gene therapy in CF. These target cells are relatively inaccessible to postnatal gene transfer limiting the success of gene therapy. Our previous work in a human-fetal trachea xenograft model suggests the potential benefit for treating CF in utero. In this study, we aim to validate adeno-associated virus serotype 2 (AAV2) gene transfer in a human fetal trachea xenograft model and to compare transduction efficiencies of pseudotyping AAV2 vectors in fetal xenografts and postnatal xenograft controls. METHODOLOGY/PRINCIPAL FINDINGS: Human fetal trachea or postnatal bronchus controls were xenografted onto immunocompromised SCID mice for a four-week engraftment period. After injection of AAV2/2, 2/1, 2/5, 2/7 or 2/8 with a LacZ reporter into both types of xenografts, we analyzed for transgene expression in the respiratory epithelium and SMGs. At 1 month, transduction by AAV2/2 and AAV2/8 in respiratory epithelium and SMG cells was significantly greater than that of AAV2/1, 2/5, and 2/7 in xenograft tracheas. Efficiency in SMG transduction was significantly greater in AAV2/8 than AAV2/2. At 3 months, AAV2/2 and AAV2/8 transgene expression was >99% of respiratory epithelium and SMG. At 1 month, transduction efficiency of AAV2/2 and AAV2/8 was significantly less in adult postnatal bronchial xenografts than in fetal tracheal xenografts. CONCLUSIONS/SIGNIFICANCE: Based on the effectiveness of AAV vectors in SMG transduction, our findings suggest the potential utility of pseudotyped AAV vectors for treatment of cystic fibrosis. The human fetal trachea xenograft model may serve as an effective tool for further development of fetal gene therapy strategies for the in utero treatment of cystic fibrosis.
format Online
Article
Text
id pubmed-3427158
institution National Center for Biotechnology Information
language English
publishDate 2012
publisher Public Library of Science
record_format MEDLINE/PubMed
spelling pubmed-34271582012-08-30 Pseudotyped AAV Vector-Mediated Gene Transfer in a Human Fetal Trachea Xenograft Model: Implications for In Utero Gene Therapy for Cystic Fibrosis Keswani, Sundeep G. Balaji, Swathi Le, Louis Leung, Alice Katz, Anna B. Lim, Foong-Yen Habli, Mounira Jones, Helen N. Wilson, James M. Crombleholme, Timothy M. PLoS One Research Article BACKGROUND: Lung disease including airway infection and inflammation currently causes the majority of morbidities and mortalities associated with cystic fibrosis (CF), making the airway epithelium and the submucosal glands (SMG) novel target cells for gene therapy in CF. These target cells are relatively inaccessible to postnatal gene transfer limiting the success of gene therapy. Our previous work in a human-fetal trachea xenograft model suggests the potential benefit for treating CF in utero. In this study, we aim to validate adeno-associated virus serotype 2 (AAV2) gene transfer in a human fetal trachea xenograft model and to compare transduction efficiencies of pseudotyping AAV2 vectors in fetal xenografts and postnatal xenograft controls. METHODOLOGY/PRINCIPAL FINDINGS: Human fetal trachea or postnatal bronchus controls were xenografted onto immunocompromised SCID mice for a four-week engraftment period. After injection of AAV2/2, 2/1, 2/5, 2/7 or 2/8 with a LacZ reporter into both types of xenografts, we analyzed for transgene expression in the respiratory epithelium and SMGs. At 1 month, transduction by AAV2/2 and AAV2/8 in respiratory epithelium and SMG cells was significantly greater than that of AAV2/1, 2/5, and 2/7 in xenograft tracheas. Efficiency in SMG transduction was significantly greater in AAV2/8 than AAV2/2. At 3 months, AAV2/2 and AAV2/8 transgene expression was >99% of respiratory epithelium and SMG. At 1 month, transduction efficiency of AAV2/2 and AAV2/8 was significantly less in adult postnatal bronchial xenografts than in fetal tracheal xenografts. CONCLUSIONS/SIGNIFICANCE: Based on the effectiveness of AAV vectors in SMG transduction, our findings suggest the potential utility of pseudotyped AAV vectors for treatment of cystic fibrosis. The human fetal trachea xenograft model may serve as an effective tool for further development of fetal gene therapy strategies for the in utero treatment of cystic fibrosis. Public Library of Science 2012-08-24 /pmc/articles/PMC3427158/ /pubmed/22937069 http://dx.doi.org/10.1371/journal.pone.0043633 Text en © 2012 Keswani et al http://creativecommons.org/licenses/by/4.0/ This is an open-access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are properly credited.
spellingShingle Research Article
Keswani, Sundeep G.
Balaji, Swathi
Le, Louis
Leung, Alice
Katz, Anna B.
Lim, Foong-Yen
Habli, Mounira
Jones, Helen N.
Wilson, James M.
Crombleholme, Timothy M.
Pseudotyped AAV Vector-Mediated Gene Transfer in a Human Fetal Trachea Xenograft Model: Implications for In Utero Gene Therapy for Cystic Fibrosis
title Pseudotyped AAV Vector-Mediated Gene Transfer in a Human Fetal Trachea Xenograft Model: Implications for In Utero Gene Therapy for Cystic Fibrosis
title_full Pseudotyped AAV Vector-Mediated Gene Transfer in a Human Fetal Trachea Xenograft Model: Implications for In Utero Gene Therapy for Cystic Fibrosis
title_fullStr Pseudotyped AAV Vector-Mediated Gene Transfer in a Human Fetal Trachea Xenograft Model: Implications for In Utero Gene Therapy for Cystic Fibrosis
title_full_unstemmed Pseudotyped AAV Vector-Mediated Gene Transfer in a Human Fetal Trachea Xenograft Model: Implications for In Utero Gene Therapy for Cystic Fibrosis
title_short Pseudotyped AAV Vector-Mediated Gene Transfer in a Human Fetal Trachea Xenograft Model: Implications for In Utero Gene Therapy for Cystic Fibrosis
title_sort pseudotyped aav vector-mediated gene transfer in a human fetal trachea xenograft model: implications for in utero gene therapy for cystic fibrosis
topic Research Article
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC3427158/
https://www.ncbi.nlm.nih.gov/pubmed/22937069
http://dx.doi.org/10.1371/journal.pone.0043633
work_keys_str_mv AT keswanisundeepg pseudotypedaavvectormediatedgenetransferinahumanfetaltracheaxenograftmodelimplicationsforinuterogenetherapyforcysticfibrosis
AT balajiswathi pseudotypedaavvectormediatedgenetransferinahumanfetaltracheaxenograftmodelimplicationsforinuterogenetherapyforcysticfibrosis
AT lelouis pseudotypedaavvectormediatedgenetransferinahumanfetaltracheaxenograftmodelimplicationsforinuterogenetherapyforcysticfibrosis
AT leungalice pseudotypedaavvectormediatedgenetransferinahumanfetaltracheaxenograftmodelimplicationsforinuterogenetherapyforcysticfibrosis
AT katzannab pseudotypedaavvectormediatedgenetransferinahumanfetaltracheaxenograftmodelimplicationsforinuterogenetherapyforcysticfibrosis
AT limfoongyen pseudotypedaavvectormediatedgenetransferinahumanfetaltracheaxenograftmodelimplicationsforinuterogenetherapyforcysticfibrosis
AT hablimounira pseudotypedaavvectormediatedgenetransferinahumanfetaltracheaxenograftmodelimplicationsforinuterogenetherapyforcysticfibrosis
AT joneshelenn pseudotypedaavvectormediatedgenetransferinahumanfetaltracheaxenograftmodelimplicationsforinuterogenetherapyforcysticfibrosis
AT wilsonjamesm pseudotypedaavvectormediatedgenetransferinahumanfetaltracheaxenograftmodelimplicationsforinuterogenetherapyforcysticfibrosis
AT crombleholmetimothym pseudotypedaavvectormediatedgenetransferinahumanfetaltracheaxenograftmodelimplicationsforinuterogenetherapyforcysticfibrosis