scholarly journals CCL2 and CCL5 driven attraction of CD172a+ monocytic cells during an equine herpesvirus type 1 (EHV-1) infection in equine nasal mucosa and the impact of two migration inhibitors, rosiglitazone (RSG) and quinacrine (QC)

2017 ◽  
Vol 48 (1) ◽  
Author(s):  
Jing Zhao ◽  
Katrien C. K. Poelaert ◽  
Jolien Van Cleemput ◽  
Hans J. Nauwynck
2010 ◽  
Vol 84 (22) ◽  
pp. 11602-11613 ◽  
Author(s):  
Helga Hofmann-Sieber ◽  
Jens Wild ◽  
Nicole Fiedler ◽  
Karsten Tischer ◽  
Jens von Einem ◽  
...  

ABSTRACT Heterologous gene transfer by viral vector systems is often limited by factors such as preexisting immunity, toxicity, low packaging capacity, or weak immunogenic potential. A novel viral vector system derived from equine herpesvirus type 1 (EHV-1) not only overcomes some of these obstacles but also promotes the robust expression of a delivered transgene and the induction of antigen-specific immune responses. Regarding an enhanced safety profile, we assessed the impact of the gene encoding the sole essential tegument protein, ETIF, on the replication and immunogenicity of recombinant EHVs. The deletion of ETIF severely attenuates replication in permissive RK13 cells and a human lung epithelial cell line but without influencing transgene expression. Whereas the intranasal administration of a recombinant luciferase EHV in BALB/c mice resulted in transgene expression in nasal cavities and lungs for 5 to 6 days, the ETIF deletion limited expression to 2 days and resulted in 30-fold-less luminescence. Attenuated replication was accompanied by a decreased capacity to induce CD8+ T cells against a delivered HIV Gag transgene in BALB/c mice following repeated intranasal application. However, a single subcutaneous immunization with a gag DNA vaccine primed specific T cells for substantial expansion by two subsequent intranasal booster immunizations with either the gag recombinant ETIF mutant or the parental virus. In addition to inducing Gag-specific serum antibodies, this prime-boost strategy clearly outperformed three sequential immunizations with the parental or EHV-ΔETIF virus or repeated DNA vaccination by inducing substantial specific secretory IgA (sIgA) titers.


Author(s):  
Kathlyn Laval ◽  
Jolien Van Cleemput ◽  
Katrien C. Poelaert ◽  
Ivy K. Brown ◽  
Hans J. Nauwynck

2015 ◽  
Vol 89 (21) ◽  
pp. 10912-10923 ◽  
Author(s):  
Kathlyn Laval ◽  
Herman W. Favoreel ◽  
Katrien C. K. Poelaert ◽  
Jolien Van Cleemput ◽  
Hans J. Nauwynck

ABSTRACTEquine herpesvirus type 1 (EHV-1) is a main cause of respiratory disease, abortion, and encephalomyelopathy in horses. Monocytic cells (CD172a+) are the main carrier cells of EHV-1 during primary infection and are proposed to serve as a “Trojan horse” to facilitate the dissemination of EHV-1 to target organs. However, the mechanism by which EHV-1 is transferred from CD172a+cells to endothelial cells (EC) remains unclear. The aim of this study was to investigate EHV-1 transmission between these two cell types. We hypothesized that EHV-1 employs specific strategies to promote the adhesion of infected CD172a+cells to EC to facilitate EHV-1 spread. Here, we demonstrated that EHV-1 infection of CD172a+cells resulted in a 3- to 5-fold increase in adhesion to EC. Antibody blocking experiments indicated that α4β1, αLβ2, and αVβ3integrins mediated adhesion of infected CD172a+cells to EC. We showed that integrin-mediated phosphatidylinositol 3-kinase (PI3K) and ERK/MAPK signaling pathways were involved in EHV-1-induced CD172a+cell adhesion at early times of infection. EHV-1 replication was enhanced in adherent CD172a+cells, which correlates with the production of tumor necrosis factor alpha (TNF-α). In the presence of neutralizing antibodies, approximately 20% of infected CD172a+cells transferred cytoplasmic material to uninfected EC and 0.01% of infected CD172a+cells transmitted infectious virus to neighboring cells. Our results demonstrated that EHV-1 infection induces adhesion of CD172a+cells to EC, which enhances viral replication, but that transfer of viral material from CD172a+cells to EC is a very specific and rare event. These findings give new insights into the complex pathogenesis of EHV-1.IMPORTANCEEquine herpesvirus type 1 (EHV-1) is a highly prevalent pathogen worldwide, causing frequent outbreaks of abortion and myeloencephalopathy, even in vaccinated horses. After primary replication in the respiratory tract, EHV-1 disseminates via cell-associated viremia in peripheral blood mononuclear cells (PBMC) and subsequently infects the endothelial cells (EC) of the pregnant uterus or central nervous system, leading in some cases to abortion and/or neurological disorders. Recently, we demonstrated that CD172a+monocytic carrier cells serve as a “Trojan horse” to facilitate EHV-1 spread from blood to target organs. Here, we investigated the mechanism underlying the transmission of EHV-1 from CD172a+cells to EC. We demonstrated that EHV-1 infection induces cellular changes in CD172a+cells, promoting their adhesion to EC. We found that both cell-to-cell contacts and the secretion of soluble factors by EC activate EHV-1 replication in CD172a+cells. This facilitates transfer of cytoplasmic viral material to EC, resulting mainly in a nonproductive infection. Our findings give new insights into how EHV-1 may spread to EC of target organs in vaccinated horses.


Vaccine ◽  
2008 ◽  
Vol 26 (19) ◽  
pp. 2335-2343 ◽  
Author(s):  
Cristina Rosas ◽  
Gerlinde R. Van de Walle ◽  
Stephan M. Metzger ◽  
Karin Hoelzer ◽  
Edward J. Dubovi ◽  
...  

Sign in / Sign up

Export Citation Format

Share Document