Cargando…

Efficient Transduction and Expansion of Ovine Macrophages for Gene Therapy Implementations

A number of bacteria provoking zoonotic diseases present intracellular survival and a host cell tropism limited to the monocyte/macrophage lineage. Thus, infection is rendered difficult to eradicate, causing chronic inflammatory reactions to the host and widespread prevalence. Although self-inactiva...

Descripción completa

Detalles Bibliográficos
Autores principales: Karponi, Garyfalia, Kritas, Spyridon, Petridou, Evanthia, Papanikolaou, Eleni
Formato: Online Artículo Texto
Lenguaje:English
Publicado: MDPI 2018
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6024765/
https://www.ncbi.nlm.nih.gov/pubmed/29912168
http://dx.doi.org/10.3390/vetsci5020057
_version_ 1783336128033587200
author Karponi, Garyfalia
Kritas, Spyridon
Petridou, Evanthia
Papanikolaou, Eleni
author_facet Karponi, Garyfalia
Kritas, Spyridon
Petridou, Evanthia
Papanikolaou, Eleni
author_sort Karponi, Garyfalia
collection PubMed
description A number of bacteria provoking zoonotic diseases present intracellular survival and a host cell tropism limited to the monocyte/macrophage lineage. Thus, infection is rendered difficult to eradicate, causing chronic inflammatory reactions to the host and widespread prevalence. Although self-inactivating lentiviral vectors have been successfully tested in the clinic against virally-induced human infectious diseases, little is known about the transduction susceptibility of ruminant animal phagocytes that play a critical role in the outbreak of zoonotic diseases such as brucellosis. In view of the development of a lentiviral vector-based platform targeting and inactivating specific genetic features of intracellular bacteria, we have tested the transducibility of ovine macrophages in terms of transgene expression and vector copy number (VCN). We show that ovine macrophages are relatively resistant to transduction even at a high multiplicity of infection with a conventional lentiviral vector expressing the green fluorescence protein and that addition of transduction enhancers, such as polybrene, increases transgene expression even after a one-week culture of the transduced cells in vitro. Overall, we demonstrate that ovine macrophages may be efficiently expanded and transduced in culture, thus providing the benchmark for gene therapy applications for zoonotic diseases.
format Online
Article
Text
id pubmed-6024765
institution National Center for Biotechnology Information
language English
publishDate 2018
publisher MDPI
record_format MEDLINE/PubMed
spelling pubmed-60247652018-07-08 Efficient Transduction and Expansion of Ovine Macrophages for Gene Therapy Implementations Karponi, Garyfalia Kritas, Spyridon Petridou, Evanthia Papanikolaou, Eleni Vet Sci Article A number of bacteria provoking zoonotic diseases present intracellular survival and a host cell tropism limited to the monocyte/macrophage lineage. Thus, infection is rendered difficult to eradicate, causing chronic inflammatory reactions to the host and widespread prevalence. Although self-inactivating lentiviral vectors have been successfully tested in the clinic against virally-induced human infectious diseases, little is known about the transduction susceptibility of ruminant animal phagocytes that play a critical role in the outbreak of zoonotic diseases such as brucellosis. In view of the development of a lentiviral vector-based platform targeting and inactivating specific genetic features of intracellular bacteria, we have tested the transducibility of ovine macrophages in terms of transgene expression and vector copy number (VCN). We show that ovine macrophages are relatively resistant to transduction even at a high multiplicity of infection with a conventional lentiviral vector expressing the green fluorescence protein and that addition of transduction enhancers, such as polybrene, increases transgene expression even after a one-week culture of the transduced cells in vitro. Overall, we demonstrate that ovine macrophages may be efficiently expanded and transduced in culture, thus providing the benchmark for gene therapy applications for zoonotic diseases. MDPI 2018-06-18 /pmc/articles/PMC6024765/ /pubmed/29912168 http://dx.doi.org/10.3390/vetsci5020057 Text en © 2018 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (http://creativecommons.org/licenses/by/4.0/).
spellingShingle Article
Karponi, Garyfalia
Kritas, Spyridon
Petridou, Evanthia
Papanikolaou, Eleni
Efficient Transduction and Expansion of Ovine Macrophages for Gene Therapy Implementations
title Efficient Transduction and Expansion of Ovine Macrophages for Gene Therapy Implementations
title_full Efficient Transduction and Expansion of Ovine Macrophages for Gene Therapy Implementations
title_fullStr Efficient Transduction and Expansion of Ovine Macrophages for Gene Therapy Implementations
title_full_unstemmed Efficient Transduction and Expansion of Ovine Macrophages for Gene Therapy Implementations
title_short Efficient Transduction and Expansion of Ovine Macrophages for Gene Therapy Implementations
title_sort efficient transduction and expansion of ovine macrophages for gene therapy implementations
topic Article
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6024765/
https://www.ncbi.nlm.nih.gov/pubmed/29912168
http://dx.doi.org/10.3390/vetsci5020057
work_keys_str_mv AT karponigaryfalia efficienttransductionandexpansionofovinemacrophagesforgenetherapyimplementations
AT kritasspyridon efficienttransductionandexpansionofovinemacrophagesforgenetherapyimplementations
AT petridouevanthia efficienttransductionandexpansionofovinemacrophagesforgenetherapyimplementations
AT papanikolaoueleni efficienttransductionandexpansionofovinemacrophagesforgenetherapyimplementations