Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










Publication year range
1.
Vet Res ; 53(1): 57, 2022 Jul 08.
Article in English | MEDLINE | ID: mdl-35804440

ABSTRACT

Peste des petits ruminants (PPR) is an acute disease of small ruminants caused by a morbillivirus. Clinical observation of the disease in the field revealed that several species of small ruminants are affected to varying degrees. This difference in disease-related effects could depend either on the host or on the virulence of the virus strain. A previous study highlighted the difference in virulence between two strains of PPRV used to infect Saanen goats. For this breed, PPRV Morocco 2008 strain (MA08) was highly virulent while PPRV Côte d'Ivoire 1989 (IC89) strain induced mild disease. Experimental studies generally based on healthy and young animals do not permit exploration of the natural variability of the host susceptibility to PPRV. Therefore, building on the previous study on Saanen goats, the current study focussed on this breed of goat and used commercially available animals with an unknown history of infection with other pathogens. Results confirmed the previous disease pattern for PPRV IC89 and MA08 strains. Viral RNA detection, macroscopic and histological lesions were stronger for the highly virulent MA08 strain. We show here for the first time that viral RNA can be detected in the tissues of vaccinated animals. Viral RNA was also detected for the first time in serum samples, which is in agreement with the role of circulating immune cells in transporting the virus into host target organs. Thus, this study provides insight into the pathogenesis of strains of different virulence of PPRV and will help to better understand the onset of the disease.


Subject(s)
Goat Diseases , Peste-des-Petits-Ruminants , Peste-des-petits-ruminants virus , Animals , Goat Diseases/virology , Goats , Peste-des-Petits-Ruminants/virology , Peste-des-petits-ruminants virus/genetics , Peste-des-petits-ruminants virus/pathogenicity , RNA, Viral/genetics , Virulence/genetics
2.
Front Immunol ; 12: 745315, 2021.
Article in English | MEDLINE | ID: mdl-34671358

ABSTRACT

Peste des petits ruminants (PPR) is an acute transboundary infectious viral disease of small ruminants, mainly sheep and goats. Host susceptibility varies considerably depending on the PPR virus (PPRV) strain, the host species and breed. The effect of strains with different levels of virulence on the modulation of the immune system has not been thoroughly compared in an experimental setting so far. In this study, we used a multi-omics approach to investigate the host cellular factors involved in different infection phenotypes. Peripheral blood mononuclear cells (PBMCs) from Saanen goats were activated with a T-cell mitogen and infected with PPRV strains of different virulence: Morocco 2008 (high virulence), Ivory Coast 1989 (low virulence) and Nigeria 75/1 (live attenuated vaccine strain). Our results showed that the highly virulent strain replicated better than the other two in PBMCs and rapidly induced cell death and a stronger inhibition of lymphocyte proliferation. However, all the strains affected lymphocyte proliferation and induced upregulation of key antiviral genes and proteins, meaning a classical antiviral response is orchestrated regardless of the virulence of the PPRV strain. On the other hand, the highly virulent strain induced stronger inflammatory responses and activated more genes related to lymphocyte migration and recruitment, and inflammatory processes. Both transcriptomic and proteomic approaches were successful in detecting viral and antiviral effectors under all conditions. The present work identified key immunological factors related to PPRV virulence in vitro.


Subject(s)
Goats/immunology , Leukocytes, Mononuclear/immunology , Peste-des-Petits-Ruminants/immunology , Peste-des-petits-ruminants virus/pathogenicity , Virulence/immunology , Animals , Gene Expression Profiling , Goats/virology , Peste-des-Petits-Ruminants/virology , Peste-des-petits-ruminants virus/immunology , Proteomics
3.
Front Immunol ; 11: 1429, 2020.
Article in English | MEDLINE | ID: mdl-32733474

ABSTRACT

The present study investigated the transcriptomic response of porcine dendritic cells (DC) to innate stimulation in vitro and in vivo. The aim was to identify DC subset-specialization, suitable Toll-like receptor (TLR) ligands targeting plasmacytoid DC (pDC), and the DC activation profile during highly and low virulent classical swine fever virus (CSFV, strain Eystrup and Pinar del Rio, respectively) infection, chosen as model for a virus causing a severe immunopathology. After identification of porcine conventional DC (cDC) 1, cDC2, pDC and a monocyte-derived subset in lymphoid tissues, we characterized DC activation using transcriptomics, and focused on chemokines, interferons, cytokines, as well as on co-stimulatory and inhibitory molecules. We demonstrate that porcine pDC provide important signals for Th1 and interferon responses, with CpG triggering the strongest responses in pDC. DC isolated early after infection of pigs with either of the two CSFV strains showed prominent upregulation of CCL5, CXCL9, CXCL10, CXCL11, and XCL1, as well as of the cytokines TNFSF13B, IL6, IL7, IL12B, IL15, IL27. Transcription of IL12B and many interferon genes were mostly restricted to pDC. Interestingly, the infection was associated with a prominent induction of inhibitory and cell death receptors. When comparing low and highly virulent CSFV strains, the latter induced a stronger inflammatory and antiviral response but a weaker cell cycle response, and reduced antigen presentation functions of DC. Taken together, we provide high-resolution information on DC activation in pigs, as well as information on how DC modulation could be linked to CSFV immunopathology.


Subject(s)
Classical Swine Fever/immunology , Dendritic Cells/immunology , Immunity, Innate/immunology , Swine/immunology , Animals , Classical Swine Fever Virus/immunology , Swine/virology
4.
J Gen Virol ; 101(2): 156-167, 2020 02.
Article in English | MEDLINE | ID: mdl-31922948

ABSTRACT

Several pigeon paramyxovirus-1 (PPMV-1) outbreaks in feral pigeons were described recently in Switzerland. The potential of PPMV-1 to induce the notifiable Newcastle disease in chickens is discussed controversially. Therefore, in order to study epidemiologically relevant parameters such as the kinetics of PPMV-1 replication and shedding as well as seroconversion after infection, chickens were infected experimentally with a Swiss PPMV-1 isolate. This generated also defined sample material for the comparison of diagnostic tests. The infectivity of the Swiss PPMV-1 isolate for chickens was demonstrated successfully by virus shedding after experimental inoculation. Our data suggest that long-lasting shedding for up to 60 days can occur in chickens infected with PPMV-1. The isolate used here was of low pathogenicity for chickens. Different quantitative reverse transcription PCR assays were evaluated with a set of Swiss PPMV-1 isolates, and various samples from experimentally infected chickens were analysed with respect to their suitability for viral RNA detection. At 14 days post-infection, virus genome was detected mainly in spleen, caecal tonsils, heart, cloacal swabs, liver, proventriculus, duodenum and kidney tissue samples. Overall, the level of virus replication was low. Not all assays used routinely in diagnostics were capable of detecting viral genome from the isolates tested. Possible explanations are the genetic divergence of PPMV-1 and the low level of viral RNA in the samples. In contrast, two methods that are not used routinely proved more suitable for virus-genome detection. Importantly, the collection of material from various different organs is recommended, in addition to the kidney and brain analysed routinely. In conclusion, this study shows that there is a need to reconsider the type of samples and the protocols used for the detection of PPMV-1 RNA in chickens.


Subject(s)
Avulavirus Infections/diagnosis , Avulavirus , Newcastle Disease/diagnosis , Animals , Avulavirus/genetics , Avulavirus/growth & development , Avulavirus/isolation & purification , Avulavirus/pathogenicity , Avulavirus Infections/pathology , Chickens , Columbidae/virology , Genome, Viral , Newcastle Disease/pathology , Newcastle Disease/virology , Newcastle disease virus/genetics , Newcastle disease virus/growth & development , Newcastle disease virus/isolation & purification , Newcastle disease virus/pathogenicity , Poultry Diseases/virology , Switzerland , Virus Diseases/veterinary , Virus Replication , Virus Shedding
6.
NPJ Vaccines ; 3: 41, 2018.
Article in English | MEDLINE | ID: mdl-30302283

ABSTRACT

Inactivated vaccines lack immunogenicity and therefore require potent adjuvants. To understand the in vivo effects of adjuvants, we used a system immunology-based analysis of ovine blood transcriptional modules (BTMs) to dissect innate immune responses relating to either antibody or haptoglobin levels. Using inactivated foot-and-mouth disease virus as an antigen, we compared non-adjuvanted to liposomal-formulated vaccines complemented or not with TLR4 and TLR7 ligands. Early after vaccination, BTM relating to myeloid cells, innate immune responses, dendritic cells, and antigen presentation correlated positively, whereas BTM relating to T and natural killer cells, as well as cell cycle correlated negatively with antibody responses. Interestingly, similar BTM also correlated with haptoglobin, but in a reversed manner, indicating that acute systemic inflammation is not beneficial for early antibody responses. Analysis of vaccine-dependent BTM modulation showed that liposomal formulations induced similar responses to those correlating to antibody levels. Surprisingly, the addition of the TLR ligands appeared to reduce early immunological perturbations and mediated anti-inflammatory effects, despite promoting antibody responses. When pre-vaccination BTM were analyzed, we found that high vaccine responders expressed higher levels of many BTM relating to cell cycle, antigen-presenting cells, and innate responses as compared with low responders. In conclusion, we have transferred human BTM to sheep and identified early vaccine-induced responses associated with antibody levels or unwanted inflammation in a heterogeneous and small group of animals. Such readouts are applicable to other veterinary species and very useful to identify efficient vaccine adjuvants, their mechanism of action, and factors related to low responders.

7.
Dev Comp Immunol ; 84: 181-192, 2018 07.
Article in English | MEDLINE | ID: mdl-29408047

ABSTRACT

Considering that macrophage functions are strongly impacted by the local tissue environment and the type of immune response, the aim of this study was to carefully set the methodological baseline for phenotype and functions of polarized porcine monocyte-derived macrophages. To this end, macrophages were generated in autologous serum alone or with colony-stimulating factor (CSF)-1 or CSF-2, and subsequently polarized with interferon (IFN)γ, interleukin-4 or IFNß. IFNγ promoted expression of MHC class I, MHC class II, CD11a, and CD40 as well as LPS-induced IL-6 and IL-12. A hallmark of interleukin-4 was Arginase 1 and CD203a upregulation, without abrogating pro-inflammatory cytokine production. IFNß induced CD169, MHC class I, CD40, CD80/86, but suppressed IL-6, IL-12 and tumor-necrosis-factor secretion. CSF-2 alone altered macrophage differentiation and promoted an IFNγ-like polarization. Altogether, the results provide a comprehensive overview of porcine macrophage polarization, and demonstrate commonalities with other species as well as peculiarities of the pig.


Subject(s)
Interferon-beta/metabolism , Interferon-gamma/metabolism , Macrophages/immunology , Monocytes/immunology , Swine/immunology , Animals , Antigens, CD/metabolism , Arginase/metabolism , Cell Differentiation , Cells, Cultured , Colony-Stimulating Factors/metabolism , Cytokines/metabolism , Histocompatibility Antigens/metabolism , Immunophenotyping , Inflammation Mediators/metabolism , Interleukin-4/metabolism , Species Specificity
8.
Sci Rep ; 7(1): 16379, 2017 11 27.
Article in English | MEDLINE | ID: mdl-29180817

ABSTRACT

Studies in the mouse model indicate that the nucleoprotein of influenza A virus represents an interesting vaccine antigen being well conserved across subtypes of influenza virus but still able to induce protective immune responses. Here we show that immunizations of pigs with vesicular stomatitis virus- and classical swine fever virus-derived replicon (VRP) particles expressing the nucleoprotein (NP) of H1N1 A/swine/Belzig/2/01 induced potent antibody and T-cell responses against influenza A virus. In contrast to a conventional whole inactivated virus vaccine, the VRP vaccines induced both NP-specific CD4 and CD8 T cells responses, including interferon-γ and tumor-necrosis-factor dual-secreting cell. Although T-cells and antibody responses were cross-reactive with the heterologous H1N2 A/swine/Bakum/R757/2010 challenge virus, they did not provide protection against infection. Surprisingly, vaccinated pigs showed enhanced virus shedding, lung inflammation and increased levels of systemic and lung interferon-α as well as elevated lung interleukin-6. In conclusion, our study shows that NP, although efficacious in the mouse model, appears not to be a promising stand-alone vaccine antigen for pigs.


Subject(s)
Influenza A virus/immunology , Influenza Vaccines/immunology , Orthomyxoviridae Infections/immunology , Virion/immunology , Animals , Antibodies, Viral/immunology , Cell Line , Cytokines/metabolism , Genetic Vectors/genetics , Influenza A virus/genetics , Influenza Vaccines/genetics , Orthomyxoviridae Infections/pathology , Orthomyxoviridae Infections/prevention & control , Orthomyxoviridae Infections/virology , Swine , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Viral Load , Virion/genetics
9.
Front Immunol ; 8: 1044, 2017.
Article in English | MEDLINE | ID: mdl-28890720

ABSTRACT

Toll-like receptors (TLR) triggering of B cells are known to promote B cell expansion, differentiation of B cells into antibody-producing and memory cells, but the TLR responses of porcine B cells is poorly characterized. Therefore, this study investigated the response pattern of porcine B cell subsets to a large collection of TLR ligands and demonstrates that the TLR2 ligand Pam3Cys-SK4 and the TLR7/8 ligands gardiquimod and resiquimod are particularly efficient at inducing proliferation, CD25 and CCR7. This activation was also determined in B-cell subpopulations including a CD21+IgM+ subset, an IgG+ subset and two putative B1-like subsets, defined as CD21-IgMhighCD11R1+CD11c+CD14+ and CD21-IgMhigh CD11R1-CD11c+CD14- B cells. The latter two were larger and expressed higher levels of CD80/86 and spontaneous phospholipase C-γ2 phosphorylation. All porcine B-cell subsets were activated by TLR2, TLR7, and TLR9 ligands. Naïve and memory conventional B cells responded similar to TLR ligands. The CD11R1+ B1-like subset had the highest proliferative responses. While both B1-like subsets did not spontaneously secrete IgM, they were the only subsets to produce high level of TLR-induced IgM. Similar to polyclonal IgM responses, memory B cells were efficiently induced to produce specific antibodies by CpG oligodinucleotide, resiquimod, and to a weaker extend by Pam3Cys-SK4. Depletion of plasmacytoid dendritic cells (pDCs) enhanced TLR-induced antibodies. The same set of TLR ligands also induced CD40 on cDCs, pDCs, and monocytes with the exception of TLR4 ligand being unable to activate pDCs. Gardiquimod and resiquimod were particularly efficient at inducing CCR7 on pDCs. Porcine B cells expressed high levels of TLR7, but relatively little other TLR mRNA. Nevertheless, TLR2 on B cells was rapidly upregulated following stimulation, explaining the strong responses following stimulation. Subset-specific analysis of TLR expression demonstrated a comparable expression of TLR2, TLR7, and TLR9 in all B cell subsets, but TLR3 was restricted to B1-like cells, whereas TLR4 was only expressed on conventional B cells, although both at low levels. Altogether, our data describe porcine innate B1-like cells, and how different B cell subsets are involved in innate sensing.

10.
Viruses ; 9(5)2017 05 22.
Article in English | MEDLINE | ID: mdl-28531165

ABSTRACT

The Japanese encephalitis virus (JEV) is responsible for zoonotic severe viral encephalitis transmitted by Culex mosquitoes. Although birds are reservoirs, pigs play a role as amplifying hosts, and are affected in particular through reproductive failure. Here, we show that a lentiviral JEV vector, expressing JEV prM and E proteins (TRIP/JEV.prME), but not JEV infection induces strong antibody-dependent enhancement (ADE) activities for infection of macrophages. Such antibodies strongly promoted infection via Fc receptors. ADE was found at both neutralizing and non-neutralizing serum dilutions. Nevertheless, in vivo JEV challenge of pigs demonstrated comparable protection induced by the TRIP/JEV.prME vaccine or heterologous JEV infection. Thus, either ADE antibodies cause no harm in the presence of neutralizing antibodies or may even have protective effects in vivo in pigs. Additionally, we found that both pre-infected and vaccinated pigs were not fully protected as low levels of viral RNA were found in lymphoid and nervous system tissue in some animals. Strikingly, the virus from the pre-infection persisted in the tonsils throughout the experiment. Finally, despite the vaccination challenge, viral RNA was detected in the oronasal swabs in all vaccinated pigs. These latter data are relevant when JEV vaccination is employed in pigs.


Subject(s)
Antibodies, Viral/blood , Encephalitis Virus, Japanese/immunology , Encephalitis, Japanese/immunology , Encephalitis, Japanese/prevention & control , Viral Vaccines/immunology , Animals , Antibodies, Neutralizing/blood , Cell Line , Culex/immunology , Disease Models, Animal , Encephalitis Virus, Japanese/genetics , Encephalitis Virus, Japanese/pathogenicity , Encephalitis, Japanese/virology , Lentivirus/immunology , Lymphocytes/virology , Macrophages/virology , Nervous System/virology , RNA, Viral/isolation & purification , Receptors, IgG , Sus scrofa , Swine , Vaccination
11.
J Immunol ; 197(12): 4791-4806, 2016 12 15.
Article in English | MEDLINE | ID: mdl-27837108

ABSTRACT

Porcine dendritic cells (DCs) are relatively well characterized, but a clear-cut identification of all DC subsets combined with full transcriptional profiling was lacking, preventing an unbiased insight into the functional specializations of DC subsets. Using a large panel of Abs in multicolor flow cytometry, cell sorting, and RNA sequencing we identified and characterized the porcine equivalent of conventional DCs (cDC) 1 and cDC2 as well as plasmacytoid DCs (pDCs) in the peripheral blood of pigs. We demonstrate that cDC1 are CD135+CD14-CD172alowCADM1+wCD11R1+ cells, cDC2 are CD135+CD14-CD172a+CADM1+CD115+wCD11R1+CD1+ cells and pDCs are CD4+CD135+CD172a+CD123+CD303+ cells. As described in other species, only cDC1 express BATF3 and XCR1, cDC2 express FCER1A and FCGR2B, and only pDCs express TCF4 and NRP1 Nevertheless, despite these cross-species conserved subset-specific transcripts, porcine pDCs differed from the species described so far in many expressed genes and transcriptomic profiling clustered pDCs more distantly from cDCs than monocytes. The response of porcine DC subsets to TLR ligands revealed that pDCs are by far the most important source of TNF-α, IL-12p40, and of course IFN-α, whereas cDCs are most efficient in MHC and costimulatory molecule expression. Nevertheless, upregulation of CD40 and CD86 in cDCs was critically influenced or even dependent on the presence of pDCs, particularly for TLR 7 and 9 ligands. Our data demonstrate that extrapolation of data on DC biology from one species to another has to be done with care, and it shows how functional details have evolved differentially in different species.


Subject(s)
Blood Cells/physiology , Dendritic Cells/physiology , Species Specificity , Swine/immunology , Transcriptome , Animals , Antigens, CD/metabolism , Cell Differentiation , Cell Separation , Cells, Cultured , Flow Cytometry , Gene Expression Profiling , Interferon-gamma/metabolism , Interleukin-12 Subunit p40/metabolism , Toll-Like Receptor 7/metabolism , Toll-Like Receptor 9/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Transcriptome/immunology , Tumor Necrosis Factor-alpha/metabolism
12.
Front Immunol ; 7: 253, 2016.
Article in English | MEDLINE | ID: mdl-27446083

ABSTRACT

This work was initiated by previous reports demonstrating that mismatched influenza A virus (IAV) vaccines can induce enhanced disease, probably mediated by antibodies. Our aim was, therefore, to investigate if a vaccine inducing opsonizing but not neutralizing antibodies against the hemagglutinin (HA) of a selected heterologous challenge virus would enhance disease or induce protective immune responses in the pig model. To this end, we immunized pigs with either whole inactivated virus (WIV)-vaccine or HA-expressing virus replicon particles (VRP) vaccine based on recombinant vesicular stomatitis virus (VSV). Both types of vaccines induced virus neutralizing and opsonizing antibodies against homologous virus as shown by a highly sensitive plasmacytoid dendritic cell-based opsonization assay. Opsonizing antibodies showed a broader reactivity against heterologous IAV compared with neutralizing antibodies. Pigs immunized with HA-recombinant VRP vaccine were partially protected from infection with a mismatched IAV, which was not neutralized but opsonized by the immune sera. The VRP vaccine reduced lung lesions, lung inflammatory cytokine responses, serum IFN-α responses, and viral loads in the airways. Only the VRP vaccine was able to prime IAV-specific IFNγ/TNFα dual secreting CD4(+) T cells detectable in the peripheral blood. In summary, this work demonstrates that with the virus pair selected, a WIV vaccine inducing opsonizing antibodies against HA which lack neutralizing activity, is neither protective nor does it induce enhanced disease in pigs. In contrast, VRP-expressing HA is efficacious vaccines in swine as they induced both potent antibodies and T-cell immunity resulting in a broader protective value.

13.
Vet Res ; 47: 34, 2016 Feb 24.
Article in English | MEDLINE | ID: mdl-26911997

ABSTRACT

Pigs are considered to be the main amplifying host for Japanese encephalitis virus (JEV), and their infection can correlate with human cases of disease. Despite their importance in the ecology of the virus as it relates to human cases of encephalitis, the pathogenesis of JEV in pigs remains obscure. In the present study, the localization and kinetics of virus replication were investigated in various tissues after experimental intravenous infection of pigs. The data demonstrate a rapid and broad spreading of the virus to the central nervous system (CNS) and various other organs. A particular tropism of JEV in pigs not only to the CNS but also for secondary lymphoid tissue, in particular the tonsils with the overall highest viral loads, was observed. In this organ, even 11 days post infection, the latest time point of the experiment, no apparent decrease in viral RNA loads and live virus was found despite the presence of a neutralizing antibody response. This was also well beyond the clinical and viremic phase. These results are of significance for the pathogenesis of JEV, and call for further experimental studies focusing on the cellular source and duration of virus replication in pigs.


Subject(s)
Central Nervous System/virology , Encephalitis Virus, Japanese/physiology , Encephalitis, Japanese/veterinary , Lymphoid Tissue/virology , Swine Diseases/virology , Viremia/veterinary , Animals , Encephalitis Virus, Japanese/growth & development , Encephalitis, Japanese/virology , Palatine Tonsil/virology , Swine , Tropism , Viral Load/veterinary , Viremia/virology
14.
Nat Commun ; 7: 10832, 2016 Feb 23.
Article in English | MEDLINE | ID: mdl-26902924

ABSTRACT

Japanese encephalitis virus (JEV), a main cause of severe viral encephalitis in humans, has a complex ecology, composed of a cycle involving primarily waterbirds and mosquitoes, as well as a cycle involving pigs as amplifying hosts. To date, JEV transmission has been exclusively described as being mosquito-mediated. Here we demonstrate that JEV can be transmitted between pigs in the absence of arthropod vectors. Pigs shed virus in oronasal secretions and are highly susceptible to oronasal infection. Clinical symptoms, virus tropism and central nervous system histological lesions are similar in pigs infected through needle, contact or oronasal inoculation. In all cases, a particularly important site of replication are the tonsils, in which JEV is found to persist for at least 25 days despite the presence of high levels of neutralizing antibodies. Our findings could have a major impact on the ecology of JEV in temperate regions with short mosquito seasons.


Subject(s)
Encephalitis Virus, Japanese , Encephalitis, Japanese/transmission , Animals , Brain/pathology , Encephalitis, Japanese/pathology , Female , Male , Palatine Tonsil/virology , Swine , Virus Shedding
15.
J Immunol ; 193(1): 364-71, 2014 Jul 01.
Article in English | MEDLINE | ID: mdl-24899499

ABSTRACT

Cathelicidins constitute potent antimicrobial peptides characterized by a high cationic charge that enables strong interactions with nucleic acids. In fact, the only human cathelicidin LL-37 triggers rapid sensing of nucleic acids by plasmacytoid dendritic cells (pDC). Among the porcine cathelicidins, phylogenetic analysis of the C-terminal mature peptide showed that porcine myeloid antimicrobial peptide (PMAP)-36 was the most closely related of the 11 porcine cathelicidins to human LL-37. Despite several investigations evaluating potent antimicrobial functions of porcine cathelicidins, nothing is known about their ability to promote pDC activation. We therefore investigated the capacity of the proline-arginine-rich 39-aa peptide, PMAP-23, PMAP-36, and protegrin-1 to complex with bacterial DNA or synthetic RNA molecules and facilitate pDC activation. We demonstrate that these peptides mediate a rapid and efficient uptake of nucleic acids within minutes, followed by robust IFN-α responses. The highest positively charged cathelicidin, PMAP-36, was found to be the most potent peptide tested for this effect. The peptide-DNA complexes were internalized and also found to associate with the cell membranes of pDC. The amphipathic conformation typical of PMAP-36 was not required for IFN-α induction in pDC. We also demonstrate that PMAP-36 can mediate IFN-α induction in pDC stimulated by Escherichia coli, which alone fail to activate pDC. This response was weaker with a scrambled PMAP-36, relating to its lower antimicrobial activity. Collectively, our data suggest that the antimicrobial and nucleic acid-complexing properties of cathelicidins can mediate pDC activation-promoting adaptive immune responses against microbial infections.


Subject(s)
Adaptive Immunity , Antimicrobial Cationic Peptides/immunology , Bacterial Infections/immunology , DNA, Bacterial/immunology , Dendritic Cells/immunology , Interferon-alpha/immunology , Plasma Cells/immunology , Animals , Bacterial Infections/pathology , Dendritic Cells/pathology , Humans , Plasma Cells/pathology , Protein Structure, Secondary , Swine , Cathelicidins
16.
PLoS Pathog ; 9(6): e1003412, 2013.
Article in English | MEDLINE | ID: mdl-23785283

ABSTRACT

Plasmacytoid dendritic cells (pDC) have been shown to efficiently sense HCV- or HIV-infected cells, using a virion-free pathway. Here, we demonstrate for classical swine fever virus, a member of the Flaviviridae, that this process is much more efficient in terms of interferon-alpha induction when compared to direct stimulation by virus particles. By employment of virus replicon particles or infectious RNA which can replicate but not form de novo virions, we exclude a transfer of virus from the donor cell to the pDC. pDC activation by infected cells was mediated by a contact-dependent RNA transfer to pDC, which was sensitive to a TLR7 inhibitor. This was inhibited by drugs affecting the cytoskeleton and membrane cholesterol. We further demonstrate that a unique viral protein with ribonuclease activity, the viral E(rns) protein of pestiviruses, efficiently prevented this process. This required intact ribonuclease function in intracellular compartments. We propose that this pathway of activation could be of particular importance for viruses which tend to be mostly cell-associated, cause persistent infection, and are non-cytopathogenic.


Subject(s)
Classical Swine Fever Virus/immunology , Classical Swine Fever/immunology , Dendritic Cells/immunology , Endoribonucleases/immunology , Plasma Cells/immunology , Viral Proteins/immunology , Animals , Cell Line , Classical Swine Fever/pathology , Plasma Cells/pathology , RNA, Viral/immunology , Swine , Toll-Like Receptor 7/immunology
18.
Vet Res ; 43: 64, 2012 Aug 30.
Article in English | MEDLINE | ID: mdl-22934974

ABSTRACT

Foot-and-mouth disease virus (FMDV) is a highly infectious member of the Picornaviridae inducing an acute disease of cloven-hoofed species. Vaccine-induced immune protection correlates with the presence of high levels of neutralizing antibodies but also opsonising antibodies have been proposed as an important mechanism of the immune response contributing to virus clearance by macrophages and leading to the production of type-I interferon (IFN) by plasmacytoid dendritic cells (pDC). The present study demonstrates that the opsonising antibody titres mediating enhanced IFN-α responses in pDC were similar to neutralizing titres, when antigenically related viruses from the same serotype were employed. However, sera cross-reacted also with non-neutralized isolates of multiple serotypes, when tested in this assay. Both uncomplexed virus and immune complexed virus stimulated pDC via Toll-like receptor 7. An additional finding of potential importance for strain-specific differences in virulence and/or immunogenicity was that pDC activation by FMDV strongly differed between viral isolates. Altogether, our results indicate that opsonising antibodies can have a broader reactivity than neutralizing antibodies and may contribute to antiviral responses induced against antigenically distant viruses.


Subject(s)
Antibodies, Viral/metabolism , Dendritic Cells/immunology , Foot-and-Mouth Disease Virus/immunology , Foot-and-Mouth Disease/immunology , Swine Diseases/immunology , Animals , Antibodies, Neutralizing/blood , Antibodies, Neutralizing/metabolism , Antibodies, Viral/blood , Dendritic Cells/virology , Enzyme-Linked Immunosorbent Assay/veterinary , Foot-and-Mouth Disease/virology , Foot-and-Mouth Disease Virus/genetics , Interferon Type I/genetics , Interferon Type I/metabolism , Interferon-alpha/biosynthesis , Opsonin Proteins/metabolism , Swine , Swine Diseases/virology , Toll-Like Receptor 7/genetics , Toll-Like Receptor 7/metabolism
19.
Virology ; 427(1): 1-9, 2012 May 25.
Article in English | MEDLINE | ID: mdl-22365327

ABSTRACT

The 2009 influenza A virus (IAV) pandemic resulted from reassortment of avian, human and swine strains probably in pigs. To elucidate the role of viral genes in host adaptation regarding innate immune responses, we focussed on the effect of genes from an avian H5N1 and a porcine H1N1 IAV on infectivity and activation of porcine GM-CSF-induced dendritic cells (DC). The highest interferon type I responses were achieved by the porcine virus reassortant containing the avian polymerase gene PB2. This finding was not due to differential tropism since all viruses infected DC equally. All viruses equally induced MHC class II, but porcine H1N1 expressing the avian viral PB2 induced more prominent nuclear NF-κB translocation compared to its parent IAV. The enhanced activation of DC may be detrimental or beneficial. An over-stimulation of innate responses could result in either pronounced tissue damage or increased resistance against IAV reassortants carrying avian PB2.


Subject(s)
Dendritic Cells/virology , Host-Pathogen Interactions , Influenza A Virus, H1N1 Subtype/immunology , Influenza A Virus, H5N1 Subtype/immunology , Interferon Type I/metabolism , Orthomyxoviridae Infections/immunology , RNA-Dependent RNA Polymerase/immunology , Reassortant Viruses/immunology , Swine Diseases/virology , Viral Proteins/immunology , Animals , Birds , Cell Line , Chick Embryo , Dendritic Cells/immunology , Dogs , Genes, Viral , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Humans , Influenza A Virus, H1N1 Subtype/genetics , Influenza A Virus, H5N1 Subtype/genetics , Major Histocompatibility Complex , Mice , NF-kappa B/metabolism , Orthomyxoviridae Infections/veterinary , Orthomyxoviridae Infections/virology , Pandemics , Promoter Regions, Genetic/genetics , Reassortant Viruses/genetics , Swine , Swine Diseases/immunology , Virus Replication/genetics
20.
J Virol ; 85(16): 8002-11, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21680532

ABSTRACT

Viruses are detected by different classes of pattern recognition receptors that lead to the activation of interferon regulatory factors (IRF) and consequently to the induction of alpha/beta interferon (IFN-α/ß). In turn, efficient viral strategies to escape the type I IFN-induced antiviral mechanisms have evolved. Previous studies established that pestivirus N(pro) antagonizes the early innate immune response by targeting the transcription factor IRF3 for proteasomal degradation. Here, we report that N(pro) of classical swine fever virus (CSFV) interacts also with IRF7, another mediator of type I IFN induction. We demonstrate that the Zn-binding domain of N(pro) is essential for the interaction of N(pro) with IRF7. For IRF3 and IRF7, the DNA-binding domain, the central region, and most of the regulatory domain are required for the interaction with N(pro). Importantly, the induction of IRF7-dependent type I IFN responses in plasmacytoid dendritic cells (pDC) is reduced after wild-type CSFV infection compared with infection with virus mutants unable to interact with IRF7. This is associated with lower levels of IRF7 in pDC. Consequently, wild-type but not N(pro) mutant CSFV-infected pDC show reduced responses to other stimuli. Taken together, the results of this study show that CSFV N(pro) is capable of manipulating the function of IRF7 in pDC and provides the virus with an additional strategy to circumvent the innate defense.


Subject(s)
Classical Swine Fever Virus/immunology , Dendritic Cells/immunology , Endopeptidases/immunology , Endopeptidases/metabolism , Interferon Regulatory Factor-7/metabolism , Interferon Type I/biosynthesis , Viral Proteins/immunology , Viral Proteins/metabolism , Animals , Blotting, Western , Cell Line , Classical Swine Fever Virus/metabolism , Dendritic Cells/metabolism , Dendritic Cells/virology , Electrophoresis, Polyacrylamide Gel , Endopeptidases/chemistry , HEK293 Cells , Humans , Immunity, Innate , Immunoprecipitation , Interferon Type I/immunology , Polymerase Chain Reaction , Protein Structure, Tertiary , Receptors, Pattern Recognition/immunology , Receptors, Pattern Recognition/metabolism , Signal Transduction , Swine , Viral Proteins/chemistry , Zinc/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...