key: cord-0792882-pukh8zqt authors: Dwivedi, Varun; Manickam, Cordelia; Binjawadagi, Basavaraj; Linhares, Daniel; Murtaugh, Michael P; Renukaradhya, Gourapura J title: Evaluation of immune responses to porcine reproductive and respiratory syndrome virus in pigs during early stage of infection under farm conditions date: 2012-02-16 journal: Virol J DOI: 10.1186/1743-422x-9-45 sha: 9151f784207180e12703758211d31c6d0ee84423 doc_id: 792882 cord_uid: pukh8zqt BACKGROUND: Porcine reproductive and respiratory syndrome virus (PRRSV) causes chronic, economically devastating disease in pigs of all ages. Frequent mutations in the viral genome result in viruses with immune escape mutants. Irrespective of regular vaccination, control of PRRSV remains a challenge to swine farmers. In PRRSV-infected pigs, innate cytokine IFN-α is inhibited and the adaptive arm of the immunity is delayed. To elucidate both cellular and innate cytokine responses at very early stages of PRRSV infection, seven weeks old pigs maintained on a commercial pig farm were infected and analyzed. RESULTS: One pig in a pen containing 25 pigs was PRRSV infected and responses from this pig and one penmate were assessed two days later. All the infected and a few of the contact neighbor pigs were viremic. At day 2 post-infection, approximately 50% of viremic pigs had greater than 50% reduction in NK cell-mediated cytotoxicity, and nearly a 1-fold increase in IFN-α production was detected in blood of a few pigs. Enhanced secretion of IL-4 (in ~90%), IL-12 (in ~40%), and IL-10 (in ~20%) (but not IFN-γ) in PRRSV infected pigs was observed. In addition, reduced frequency of myeloid cells, CD4(-)CD8(+ )T cells, and CD4(+)CD8(+ )T cells and upregulated frequency of lymphocytes bearing natural T regulatory cell phenotype were detected in viremic pigs. Interestingly, all viremic contact pigs also had comparable immune cell modulations. CONCLUSION: Replicating PRRSV in both infected and contact pigs was found to be responsible for rapid modulation in NK cell-meditated cytotoxicity and alteration in the production of important immune cytokines. PRRSV-induced immunological changes observed simultaneously at both cellular and cytokine levels early post-infection appear to be responsible for the delay in generation of adaptive immunity. As the study was performed in pigs maintained under commercial environmental conditions, this study has practical implications in design of protective vaccines. Porcine reproductive and respiratory syndrome (PRRS) is a chronic respiratory and reproductive viral disease of pigs that is responsible for huge economic losses to the swine industry worldwide. In the US alone, PRRS is estimated to cause losses of $664 million every year [1] . As per the Animal and Plant Health Inspection Service report of 2009, 49.8% of unvaccinated pigs in the US are seropositive to PRRS virus (PRRSV), suggesting PRRS an endemic disease in the US, and pig producers have to constantly battle against outbreaks. At present we lack a good understanding of early immunological mechanisms in PRRSV-infected pigs and elucidation of such information could guide us in the development of improved preventive or therapeutic measures. The innate immune system is an important arm of defense to prevent viral invasion and replication to initiate the adaptive arm of the immune system. Adequate early activation of the innate immune system is critical to initiate generation of protective adaptive immunity to achieve complete viral clearance [2] . The quantities of important cytokines secreted in pigs infected by PRRSV appeared to be significantly lower than pigs infected with a swine influenza virus or porcine respiratory coronavirus [3] [4] [5] . Natural killer (NK) cell, a lymphocyte subpopulation, provides a first line of innate defense against virus infection [6] . In pigs, NK cells are small to medium sized lymphocytes and they lack adequate intracellular granules [7, 8] . Therefore, although younger pigs possess a higher frequency of NK cells, they have reduced NK cytolytic activity [9] . Unfortunately, PRRSV further suppresses the NK cell-mediated cytotoxicity in infected pigs [10, 11] . So far, studies addressing cytokine profiles and NK cell cytotoxic functions have been performed in pigs from 1 week post-PRRSV infection and under controlled experimental conditions. PRRSV is known to suppress production of an important innate antiviral cytokine, interferon (IFN)-α [12] [13] [14] . IFN-γ response in PRRSV-infected pigs appears to be dampened and delayed [13, 15, 16] . The Th1 and Th2 cytokine profiles provide an elegant model of directed response to infectious pathogens and are indicative of immune regulation, protective immunity, and vaccine efficacy. The Th2 cytokine IL-4 is involved in suppression of pathogen-specific Th1 immune responses [17, 18] , but the role of IL-4 in the pig immune system appears to be different [19, 20] . Lymphocytes expressing markers CD4 or CD8 alone and CD4 and CD8 together are important in viral clearance by secreting IFN-γ and mediating pathogen specific cytotoxicity [21] [22] [23] [24] . Foxp3expressing CD4 + CD25 + cells with immunosuppressive properties, called "T-regulatory cells (Tregs)", have been identified in pigs [25] . PRRSV-mediated proliferation of Tregs in infected and vaccinated pigs suggests the involvement of Tregs in disease progression and immune modulation [11, [26] [27] [28] [29] [30] . The mechanism of immune suppression in PRRSV-infected pigs appears to be governed by enhanced production of interleukin (IL)-10 [10, 31, 32] , which drives the generation of IL-10-producing Tregs [33] . However, it has also been shown that IL-10 expression varies with infection using different strains of the PRRSV (Diaz et al., 2006); thus, it is unclear if Treg-mediated suppression of immune response occurs with all the strains of PRRSV. The purpose of our study was to elucidate innate immunological mediators' modulated early post-PRRSV infection in infected and contact pigs maintained under field conditions. In each pen (n = 25 pigs) only 2 pigs were studied, the pig infected and 1 of the other penmate (contact control). All 25 PRRSV-infected pigs in 25 pens were viremic with detectable RNA and viral titer by quantitative RT-PCR at 2 days post-infection ( Figure 1A and 1B) . Interestingly, seven of 25 contact pigs also were viremic (Figure 1 ), indicating the rapid transmission of PRRSV to penmates. We observed pigs twice daily for clinical PRRS symptoms such as fever, inappetence, respiratory distress, cough, etc., but did not see any such symptoms in infected or contact pigs until day 2 post-infection. Prior to PRRSV infection, NK cell cytotoxicity was analyzed by a colorimetric assay using peripheral blood mononuclear cells (PBMC) from all 50 pigs and appreciable NK cell cytotoxicity was detected in 13 infected pigs and 12 contact control pigs ( Figure 2A ). Two days after infection, 10 of the 13 NK cell competent PRRSVinfected pigs had more than 50% reduction in NK cell cytotoxicity, whereas only three of the 12 contact pigs had a similar reduction in NK cell cytotoxicity ( Figure 2B ). The reduction in NK cytolytic activity in the 13 NK cell competent infected pigs was statistically significant at tested effector cell: target cell (E:T) ratios compared to day 0. Flow cytometric analysis detected an increased frequency of NK cells rich fraction at day 2 post-infection in both infected and contact pigs (Table 1) . Early increase in IFN-a, IL-4, and IL-10 production in PRRSV infected pigs CD3 + and b CD3cells were gated to enumerate CD4 and CD8α expression and grouped as follows: total T lymphocytes (CD3 + T cells); CD4 + T cells (CD3 + CD4 + CD8α -); CD8 + T cells (CD3 + CD4 -CD8α + ); Th/memory cells (CD3 + CD4 + CD8α + ); NK cells rich fraction (CD3 -CD4 -CD8α + ). Other immune cells: c γδ T cells (CD8α + TcR1N4 + ); d Myeloid cells (CD172 + ). e CD25 + cells were gated to enumerate CD4 and Foxp3 expressing cells and the percent of total CD4 + CD25 + Foxp3 + cells are shown. PBMCs of all the seven contact viremic pigs were used in the study. f Each number is an average percent of immune cells from 7 to 13 pigs +/-standard deviation. Symbols indicate statistically significant difference (P < 0.05) between: *-day 0 and day 2 contact; #-day 0 and day 2 infected; $-day 2 contact and day 2 infected pig groups, analyzed by Wilcoxon t-test. contact pigs carrying replicating virus at day 2 postinfection was detected ( Figure 3A and 3B). Similarly, plasma samples of 22 out of 25 infected pigs at day 2 post-infection had increased secretion of IL-4 ( Figure 4A ), but none of the contact pigs produced detectable levels of IL-4 ( Figure 4C ). Increased IL-10 production was also observed in 5 out of 25 infected and 4 out of 25 of contact control pigs ( Figure 4B and 4D ). Production of IL-4 and IL-10 was statistically significant in infected animals at day 2 post-infection compared to their secretion preinfection (P < 0.05). A rapid increase in IL-12 but not IFN-g secretion in PRRSV infected pigs Eleven out of 25 PRRSV-infected pigs and 7 out of 25 contact pigs at day 2 post-infection had a significant increase in secretion of IL-12 compared to their preinfection levels ( Figure 5A and 5B). In contrast, PRRSVinfected pigs did not secrete increased IFN-γ ( Figure 5C ). Five of the 25 contact pigs produced increased IFN-γ but the results were not statistically significant ( Figure 5D ). The frequency of lymphoid and myeloid immune cells was analyzed by flow cytometry. The population of total lymphocytes was markedly increased in infected pigs, but the frequency of CD8 + T cells was significantly reduced in infected pigs (Table 1) . Surprisingly, contact pigs had a significantly higher frequency of CD8 + T cells (Table 1) . A decrease in the frequency of CD4 + CD8 + T cells was statistically significant at day 2 post-infection in infected pigs compared to their levels at preinfection ( Table 1 ). The frequency of myeloid cells in PBMC was significantly reduced by greater than two-fold in both infected and contact pigs (Table 1 ). In contrast, the frequency of lymphocytes with a natural Treg phenotype was significantly higher in both infected and contact pigs at day 2 post-infection (Table 1 ). Our rationale for conducting this study was to investigate how PRRSV rapidly modulates host innate immune mediators at an early time point post-infection, because the virus-induced adaptive immune response is always weak and delayed [34, 35] . It is known that younger pigs suffer severely from PRRS than adult animals [32] , likely resulting from poorly developed innate immune system or an efficient immune evasion strategy adapted by the virus. Innate NK cells are the lymphocyte subpopulation known for their ability to provide the first line of defense against viral infections [6] . Younger pigs have underdeveloped NK cell-mediated cytotoxic function [7, 9] , and in our study, approximately 50% of pigs at day 0 (preinfection) did not have detectable NK cell cytotoxicity. Moreover, in pigs with satisfactory NK cell cytotoxicity, PRRSV significantly suppresses the NK function by 50-80% from post-infection day 7-24 [10] . In the current study, as early as day 2 post-infection, a significant reduction in NK cell-cytolytic activity was observed. In addition, an increased frequency in NK cells rich fraction in virus-infected pigs did not result in rescued NK cytotoxicity, suggesting PRRSV-induced modulation in NK cell function. This could be one of the innate immune evasion mechanisms of PRRSV. All pigs with reduced NK cell cytotoxicity were viremic with titers greater than 2 logs, suggesting replicating PRRSV mediated suppression of NK cell function. In pigs, a precise selection marker for phenotypic analysis of NK cells is not available, however, CD56 is used in some studies [36, 37] . Recently, we identified comparable frequency of CD56 + and CD3 -CD4 -CD8α + cells in the pig PBMC (data not shown). PRRSV is a poor inducer of IFN-α and its level remains low throughout the course of infection [12, 13, 38] . Suppression of IFN-α production in PRRSV infected pigs is mediated by viral nsp1β [39] . In vitro stimulation of porcine monocytes and macrophages with low levels of IFN-α (10 units/ml) stimulates increased expression of sialoadhesin, the receptor required for PRRSV internalization in macrophages. Interestingly, such a stimulation of macrophages for the initial 2-3 days is sufficient to enhance the efficiency of PRRSV infection by nearly 20-fold [40] . Consistent with that, our results identified a slight increase in IFN-α production early post-infection in greater than 50% of PRRSVinfected pigs which maybe sufficient to augment the yield of replicating PRRSV. Critical and coordinated functions of IFN-α/β, IL-12, and IL-15 in regulating NK cell responses during viral infections have been reported [41] . There have been many studies demonstrating the important role played by IFN-α in modulating NK cell cytotoxicity [41] [42] [43] [44] [45] [46] . Moreover, reduction in PRRSV-induced pig NK cell cytotoxicity is independent of NK cell population observed in this and earlier reports [10, 30] . Impaired basal NK cell cytolytic activity, despite the presence of normal NK cell numbers, was observed due to impairment in the STAT1 pathway [46] . Recently, more than 1000-fold increase in IFN-α production in mice by NK cells mediated through MIP-1β and LFA was reported [47] . Thus, PRRSV-mediated NK cell modulation in pigs would have affected both its cytotoxic function as well as IFN-α production. Cytokine IL-4 mediates reduction in IL-2-stimulated pig NK cell function [48] . Enhanced production of IL-4 in greater than 90% of PRRSV-infected pigs detected in our study appears to be having an important immune modulatory role on NK cell function. In both mice and humans, IL-4 is essential for antibody production and is a soluble diagnostic marker of Th2 immune response. In contrast, IL-4 is not a stimulatory factor for porcine B cells, and in fact in pigs, IL-4 blocks antibody and IL-6 secretion and also suppresses antigen-stimulated proliferation of B cells [19] . IL-4 suppresses the transcriptional activity of all the inflammatory cytokines and plays an important role in regulation of inflammatory activity of pig alveolar macrophages in respiratory disease conditions [20] . Therefore, the role of IL-4 in pigs is different compared to mice and humans. Overall, our study has demonstrated the correlative evidences on PRRSV-mediated suppression of NK cell function by coordinated modulation of multiple cytokines. Studies have demonstrated the adjuvant role of IL-12 to live PRRSV vaccine in pigs by induction of IFN-γ production resulting in a protective immune response [49] . Although, increased secretion of IL-12 in PRRSVinfected pigs was detected early post-infection in our study, secretion of IFN-γ remained low. 2 days postinfection maybe too early to expect IFN-γ production, but PRRSV-induced upregulation of IL-12 did not increase the IFN-γ secretion even after 3 weeks postinfection [10] . Generally, IL-12 is a heterodimer consisting of p40 and p35 subunits [50, 51] , and it is a critical regulator of T and NK cell functions [52, 53] . But IL-12 can also exist as a p40 homodimer which is a potent antagonist of immune responses [54] . The cytokine IL-12 mediates multiple roles in the immune system which include activation of macrophages during intracellular infection [55] . At present, reagents to differentiate homo-and heterodimers of porcine IL-12 are not available. Interestingly, our data further confirmed that PRRSV-induced enhanced IL-12 is not helping to elicit protective immunity against PRRSV infection. PRRSV has restricted cell tropism, with differentiated macrophages expressing various receptor molecules such as heparin sulphate, sialoadhesin, and CD163, which all aid in PRRSV invasion [56] [57] [58] . Therefore, PRRSV-induced upregulation of IL-12 might be involved in differentiation of macrophages which aids viral infection. However, targeted approach to investigate such a mechanism of action of IL-12 in PRRSV-infected pigs is important. Lymphocytes expressing the CD8 marker are important in viral clearance by secreting IFN-γ and mediating pathogen-specific cytotoxicity [21, 22] . Pigs have abundant CD4 + CD8 + T cells and these cells possess memory, T-helper, and cytolytic properties, and are called as "Th/memory cells," and they too secrete IFN-γ [21, 23] . The CD4 + CD8 + T cells were found to be associated with protection in pigs vaccinated against pseudorabies virus [21, 23] . Interestingly, we detected a rapid downregulation of both CD8 + and CD4 + CD8 + T cells, suggesting one mechanism of the PRRSV-mediated delay in initiation of virus-specific adaptive immunity is by altering the function and frequency of important lymphocytes early post-infection. The reason behind an increased frequency of CD8 + T cells detected in contact pigs needs further investigation. Swine contain a large population of γδ T cells in the periphery, capable of acting in both innate and adaptive immunity [59, 60] . In PRRSV-infected pigs, γδ T cells secrete IFN-γ, indicating their role in antiviral defense [61] . In our study, the population of γδ T cells at 2 days post-infection was decreased in contact pigs and remained unaltered in infected pigs compared to this population at preinfection, suggesting that the data on PRRSV-mediated effect on γδ T cells is not conclusive at day 2 post-infection to make any meaningful conclusion. In pigs, the cell surface marker CD172 is expressed on myeloid cells and granulocytes [62] . A rapid reduction in the total myeloid cell pool in PBMC of infected pigs indicates their migration to the lungs, a primary site of infection. Like in mice and humans, Foxp3 + CD4 + CD25 + Tregs are present in pigs and have comparable immunosuppressive properties [25] . PRRSV-mediated upregulation of Tregs bearing a natural Treg phenotype in infected pigs indicates the role of Tregs in viral pathogenesis [26] [27] [28] . Recently, a significant decrease in the frequency of Tregs in pigs vaccinated with a modified live PRRSV vaccine co-administered with a potent mucosal adjuvant and challenged with a virulent heterologous PRRSV MN184 has suggested the cross-protective immunity [11] . Increased frequency of Tregs in unvaccinated PRRSV challenged pigs was associated with an increased secretion of immunosuppressive cytokines, IL-10 and TGF-β [11] . Our study has demonstrated that the increased population of Tregs in PRRSV-infected pigs at day 2 post-infection is associated with an increased production of IL-10. Within the contact pig group, a significant modulation in the cytokine secretion profile and the frequency of immune cells was observed principally in pigs which were viremic. A rapid reduction in NK cell cytolytic activity in PRRSV infected pigs was associated with high titered replicating PRRSV and reduced innate IFN-α production. In addition, enhanced production of the cytokines, IL-4, IL-12, and IL-10, and reduction in the population immune cells (CD8, + Th/memory, and myeloid cells) were also mediated by replicating PRRSV. Our results have practical relevance as the study was performed in pigs maintained under natural environmental conditions. This knowledge may benefit PRRSV researchers to consider innovative strategies to circumvent the virus induced suppressive mechanisms during design of effective vaccines. This study was performed on a subset of nursery pigs (n = 50), approximately 7 weeks of age, in a commercial research setting containing two rooms with 40 pens per room, and 25 healthy pigs per pen. Nursery pigs were from a herd free of PRRSV and experimental pigs were seronegative at the beginning of the experiment (Herd-Chek PRRS ELISA, IDEXX). Twenty-five pens were selected at random out of 40 pens in a room for this study. In each pen, two pigs were selected at random and ear-tagged. The 50 pigs from 25 pens were bled and one of the two tagged pigs in each pen was injected with 2 × 10 4 TCID 50 of PRRSV strain MN 1-18-2 [63, 64] , intramuscularly. The other pig was an uninoculated (mock) contact control. Pigs were bled again 2 days after infection and samples were paired by animal to facilitate statistical analysis. All the pigs were maintained and samples collected as per the protocol approved by the institutional animal care and use committee (IACUC), University of Minnesota. For the isolation of peripheral blood mononuclear cells (PBMC), blood was collected in EDTA from pigs on both day 0 and day 2 post-PRRSV infection. Blood samples were transported on ice overnight to the Food Animal Health Research Program, Ohio Agricultural Research and Development Center (OARDC), The Ohio State University, Wooster, Ohio. Samples were processed and PBMC were isolated from a total of 100 samples; on day 0 (n = 50) and day 2, infected (n = 25) and contact (n = 25), as described previously [65] [66] [67] . Samples collected from all the contact pigs were included together irrespective of a few of the pigs had detectable viremia, because our main purpose of this study was to understand early immune responses in known infected pigs at day 2 post-infection. In addition, as we did not know when the contact control pigs get infected within our 2 day post-infection study period, we grouped them as one unit for the immunological evaluation. PRRSV RNA was extracted from plasma using Mag-MAX-96 viral RNA Isolation Kit (Ambion/Applied Biosystems, Carlsbad, California) as per the manufacturer's protocol. A commercial real-time PCR assay kit (Tetracore Inc., Gaithersburg, MD) was used for the quantification of PRRSV as previously described [68] . Results of PRRSV qRT-PCR in each ml of plasma are reported as RNA copies in log 10 values. In addition, a standard curve was developed by preparing 10-fold dilutions of PRRSV VR2332 stock starting at 10 6 TCID 50 per ml for the viral RNA quantification and the results are reported in log 10 TCID 50 per ml of the plasma as previously described [69] . The NK cell assay to determine the generic pig NK cellmediated cyotoxicity in PRRSV-infected pigs was followed as described previously [48, 70] with a few modifications. Briefly, PBMC were used as the source of NK cells (effectors) and K-562 human myeloblastoid leukemia cells were the targets. Effectors were incubated in RPMI supplemented with FBS with a fixed number of targets for 24 h at 37C in a CO 2 incubator and the amount of released lactate dehydrogenase (LDH) into the supernatant was measured by a colorimetric assay. Released LDH is directly proportional to the NK-specific lysis of target cells. The percent of NK cell-specific killing was calculated after subtracting the spontaneously released LDH due to nonspecific cell lysis of targets. Plasma samples collected after initial centrifugation of unclotted blood were analyzed for the IFN-α, Th1 (IFNγ, IL-12), Th2 (IL-4), and immunosuppressive (IL-10) cytokines by cytokine sandwich ELISA as described previously [71] . Flow cytometric analysis was performed to determine the phenotype and the frequency of immune cell populations in a multicolor immunoassay as described previously [10, 66, 72] . Briefly, PBMC were treated with 2% pig serum to block Fc receptors. Cells were then stained with an appropriate mAb which was either directly conjugated to a specific fluorochrome or biotinylated or with a purified antibody to pig-specific immune cell surface markers [CD3ε, CD172 (SouthernBiotech, Birmingham, Alabama), CD4α, CD8α, CD11c (BD PharMingen), CD25 (Serotec, Raleigh, NC), TcR1N4 (VMRD, Pullman, WA), Foxp3 (eBioscience, San Diego, CA)] or with their respective isotype control mAb. Labeled cells were treated with streptavidin-conjugated fluorochrome or respective anti-species isotype-specific secondary antibody conjugated with fluorochrome. Finally, cells were fixed with 1% paraformaldehyde. For intracellular Foxp3 staining, cells were surface stained for CD4 and CD25 as described above and incubated overnight at 4C in permeabilization buffer. Cells were then stained with fluorochrome-conjugated pig Foxp3 cross-reactive antirat Foxp3 mAb for detection [25, 73] . Immunostained PBMC were acquired using a FACS AriaII flow cytometer (BD Biosciences, San Jose, CA). Analysis was performed using FlowJo software (Tree Star, Inc. OR, USA) to enumerate immune cell populations based on cell surface marker expression as follows: Total T lymphocytes (CD3 + cells); NK cells (CD3 -CD4 -CD8α + ) [7] ; CD3 + CD4 + CD8cells; CD3 + CD4 -CD8α + cells; CD4 CD8 double positive T cells (CD3 + CD4 + CD8α + ) also called as "T-helper/memory cells" [21, 23] ; γδ T cells (CD8α + TcR1N4 + ); phenotype compatible to natural T-regulatory cells (CD4 + CD25 + FOXP3 + ); myeloid cells (CD172 + ). Frequency of individual lymphoid and myeloid cell subsets was analyzed from a total 50,000 events. Individual pig immune response data are shown in all figures. Average values from 25 pigs +/-standard deviation for cytokines and for immune cells from 7-13 pigs are shown. Statistical analyses were performed using a paired t-test or Wilcoxon t-test when the sample number was more than 10 or less than 10, respectively(SAS software, SAS Institute Inc., Cary, NC). PRRS Costs Industry $664 Million Annually Innate immune response to viral infection Differential production of proinflammatory cytokines in the pig lung during different respiratory virus infections: correlations with pathogenicity Proinflammatory cytokines and viral respiratory disease in pigs Correlations between lung proinflammatory cytokine levels, virus replication, and disease after swine influenza virus challenge of vaccination-immune pigs Natural killer cells in antiviral defense: function and regulation by innate cytokines Porcine T lymphocytes and NK cells-An update The null T cell in pig blood is not an NK cell Natural killer cell frequency and function in pigs selectively bred for high or low antibody and cell-mediated immune response: response to vaccination with modified live transmissible gastroenteritis virus Porcine reproductive and respiratory syndrome virus-induced immunosuppression exacerbates the inflammatory response to porcine respiratory coronavirus in pigs Cross-protective immunity to porcine reproductive and respiratory syndrome virus by intranasal delivery of a live virus vaccine with a potent adjuvant Interferon-alpha response to swine arterivirus (PoAV), the porcine reproductive and respiratory syndrome virus Immunological responses of swine to porcine reproductive and respiratory syndrome virus infection Porcine reproductive and respiratory syndrome virus modifies innate immunity and alters disease outcome in pigs subsequently infected with porcine respiratory coronavirus: implications for respiratory viral co-infections The level of virus-specific T-cell and macrophage recruitment in porcine reproductive and respiratory syndrome virus infection in pigs is independent of virus load Immune response against porcine reproductive and respiratory syndrome virus during acute and chronic infection Humoral immune functions in IL-4 transgenic mice Interleukin-4 and interferon-gamma: the quintessence of a mutual antagonistic relationship Species specialization in cytokine biology: is interleukin-4 central to the T(H)1-T (H)2 paradigm in swine? Interleukin-4 suppresses inflammatory cytokine gene transcription in porcine macrophages Cytolytic function for pseudorabies virus-stimulated porcine CD4+ CD8dull+ lymphocytes Role of B cells and cytotoxic T lymphocytes in clearance of and immunity to rotavirus infection in mice Vaccine-induced, pseudorabies virus-specific, extrathymic CD4+CD8+ memory T-helper cells in swine Cellular sources and targets of IFN-gamma-mediated protection against viral demyelination and neurological deficits Phenotypic and functional characterisation of porcine CD4(+)CD25(high) regulatory T cells European genotype of porcine reproductive and respiratory syndrome (PRRSV) infects monocyte-derived dendritic cells but does not induce Treg cells Induction of T helper 3 regulatory cells by dendritic cells infected with porcine reproductive and respiratory syndrome virus Induction of inducible CD4+CD25+Foxp3+ regulatory T lymphocytes by porcine reproductive and respiratory syndrome virus (PRRSV) A modified live PRRSV vaccine and the pathogenic parent strain induce regulatory T cells in pigs naturally infected with Mycoplasma hyopneumoniae Intranasal delivery of whole cell lysate of Mycobacterium tuberculosis induces protective immune responses to a modified live porcine reproductive and respiratory syndrome virus vaccine in pigs Upregulation of IL-10 gene expression in porcine peripheral blood mononuclear cells by porcine reproductive and respiratory syndrome virus Agedependent resistance to Porcine reproductive and respiratory syndrome virus replication in swine Pulmonary dendritic cells producing IL-10 mediate tolerance induced by respiratory exposure to antigen Challenges for porcine reproductive and respiratory syndrome virus (PRRSV) vaccinology. Vaccine The challenge of PRRS immunology Superficial implantation in pigs is associated with decreased numbers and redistribution of endometrial NK-cell populations Natural killer cell dysfunction during acute infection with foot-and-mouth disease virus In vivo and in vitro interferon (IFN) studies with the porcine reproductive and respiratory syndrome virus (PRRSV) Identification of two auto-cleavage products of nonstructural protein 1 (nsp1) in porcine reproductive and respiratory syndrome virus infected cells: nsp1 function as interferon antagonist IFNalpha treatment enhances porcine Arterivirus infection of monocytes via upregulation of the porcine Arterivirus receptor sialoadhesin Coordinated and distinct roles for IFNalpha beta, IL-12, and IL-15 regulation of NK cell responses to viral infection IFNalpha regulates NK cell cytotoxicity through STAT1 pathway Modulation of human natural killer cell function by cytokines and rheumatic disease Direct Effects of type I IFNs on cells of the immune system Antagonistic effects of interferons on the cytotoxicity mediated by natural killer cells Distinct requirements for IFNs and STAT1 in NK cell function IFN-{alpha} Production by Plasmacytoid Dendritic Cells Stimulated with RNA-Containing Immune Complexes Is Promoted by NK Cells via MIP-1{beta} and LFA-1 Modulation of in vitro porcine natural killer cell activity by recombinant interleukin-1 alpha, interleukin-2 and interleukin-4 Adjuvant danger signals increase the immune response to porcine reproductive and respiratory syndrome virus Identification and purification of natural killer cell stimulatory factor (NKSF), a cytokine with multiple biologic effects on human lymphocytes Purification to homogeneity and partial characterization of cytotoxic lymphocyte maturation factor from human B-lymphoblastoid cells Natural killer cell stimulatory factor (interleukin 12 [IL-12]) induces T helper type 1 (Th1)-specific immune responses and inhibits the development of IL-4-producing Th cells Development of TH1 CD4+ T cells through IL-12 produced by Listeriainduced macrophages Mouse interleukin-12 (IL-12) p40 homodimer: a potent IL-12 antagonist Role of IL-12 in macrophage activation during intracellular infection: IL-12 and mycobacteria synergistically release TNF-alpha and nitric oxide from macrophages via IFN-gamma induction Porcine arterivirus infection of alveolar macrophages is mediated by sialic acid on the virus Sialoadhesin and CD163 join forces during entry of the porcine reproductive and respiratory syndrome virus The M/GP(5) glycoprotein complex of porcine reproductive and respiratory syndrome virus binds the sialoadhesin receptor in a sialic acid-dependent manner Characterization of the porcine gammadelta Tcell receptor structure and cellular distribution by monoclonal antibody PPT27 Porcine reproductive and respiratory syndrome: a review. Swine Health Prod Gammadelta lymphocyte response to porcine reproductive and respiratory syndrome virus Porcine myelomonocytic markers and cell populations Long-distance airborne transport of infectious PRRSV and Mycoplasma hyopneumoniae from a swine population infected with multiple viral variants Epidemiology of a new PRRS virus isolate and Outbreak Isotype-specific antibodysecreting cells to transmissible gastroenteritis virus and porcine respiratory coronavirus in gut-and bronchus-associated lymphoid tissues of suckling pigs Swine influenza H1N1 virus induces acute inflammatory immune responses in pig lungs: a potential animal model for human H1N1 influenza virus Antigen-specific B-cell responses to porcine reproductive and respiratory syndrome virus infection Detection of U.S., Lelystad, and European-like porcine reproductive and respiratory syndrome viruses and relative quantitation in boar semen and serum samples by real-time PCR Detection of Porcine reproductive and respiratory syndrome virus infection in porcine oral fluid samples: a longitudinal study under experimental conditions An enzyme-release assay for natural cytotoxicity Cytokine responses in gnotobiotic pigs after infection with virulent or attenuated human rotavirus Functional invariant NKT cells in pig lungs regulate the airway hyperreactivity: a potential animal model Detection of Foxp3 protein expression in porcine T lymphocytes Submit your next manuscript to BioMed Central and take full advantage of: • Convenient online submission • Thorough peer review • No space constraints or color figure charges • Immediate publication on acceptance • Inclusion in PubMed, CAS, Scopus and Google Scholar • Research which is freely available for redistribution This research is supported in whole by USDA-NIFA PRRS CAP2 award 2008-55620-19132 to MPM and GJR. Salaries were provided by the state and federal funds appropriated to OARDC and PHPID, The Ohio State University. We thank Ms. Ruthi Patterson for processing the blood samples. We also thank Bert Bishop for statistics, and Dr. Michele Williams for editing the manuscript.