Skip to main content

Evaluation of immune responses to porcine reproductive and respiratory syndrome virus in pigs during early stage of infection under farm conditions

Abstract

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.

Background

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 [3–5]. 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 [12–14]. 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 [21–24]. Foxp3-expressing 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, 26–30]. 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.

Results

PRRSV-infected and contact pigs had suppressed NK cell-mediated cytotoxicity

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.

Figure 1
figure 1

Active replication of PRRSV in infected pigs. Plasma collected from pigs on day 0 (n = 50) (pre-infection) and day 2 post-infection of infected (n = 25) and contact (n = 25) pigs was analyzed to determine the PRRSV titer by quantitative RT-PCR. (A) Results of PRRSV qRT-PCR in each ml of plasma (RNA copies in log10 values) and (B) viral load in plasma in log10 TCID50/ml are shown.

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 PRRSV-infected 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).

Figure 2
figure 2

Reduced NK cell-mediated cytotoxicity in PRRSV infected pigs. Percent NK-specific cytotoxicity was measured in pig PBMC (effectors). (A) Day 0 pre-infection and (B) day 2 post-infection against K-562 target cells. Effectors and targets at indicated E:T ratios were co-cultured and the supernatant harvested after 24 h was analyzed spectrophotometrically (OD490 nm) for released LDH from the lysed cells using a LDH substrate. Each line corresponds to NK cell-specific lysis from one pig at the four different E:T ratios. Statistical analysis was performed using a paired t-test by comparing the percent specific NK cell lysis between day 0 and day 2 post-infection at respective E:T ratios: P = 0.0115 at 1:100; P = 0.0001 at 1:50; P = 0.0007 at 1:25; P = 0.0069 at 1:12.5.

Table 1 Rapid modulation in the frequency of lymphoid and myeloid immune cells in PRRSV-infected pigs.

Early increase in IFN-α, IL-4, and IL-10 production in PRRSV infected pigs

In plasma samples, a 1-fold increase in IFN-α production in 7 out of 25 PRRSV-infected and 6 out of 25 contact pigs carrying replicating virus at day 2 post-infection 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).

Figure 3
figure 3

A moderate increase in secretion of the cytokine IFN-α PRRSV in infected pigs. Plasma samples collected from pigs on day 0 pre-infection (n = 50) and day 2 post-infection of infected (n = 25) and contact (n = 25) pigs were analyzed by ELISA to determine IFN-α levels: (A) PRRSV infected; (B) contact neighbor pigs. Each bar represents the amount of cytokines secreted by individual pigs, and numbers shown above horizontal lines indicate the average amount of cytokines from 25 pigs +/- standard deviation. Statistical analysis was performed using a paired t-test by comparing day 0 and day 2 cytokine responses.

Figure 4
figure 4

Rapid increase in secretion of the cytokines IL-4 and IL-10 in PRRSV infected pigs. Plasma samples collected from pigs on day 0 pre-infection (n = 50) and day 2 post-infection of infected (n = 25) and contact (n = 25) pigs were analyzed by ELISA to evaluate cytokine levels: (A and C) IL-4; (B and D) IL-10 secreted by indicated pig groups at day 0 and day 2 post-infection. Each bar represents the amount of cytokines secreted by individual pigs, and numbers shown above horizontal lines indicate the average amount of cytokines from 25 pigs +/- standard deviation. Statistical analysis was performed using a paired t-test by comparing day 0 and day 2 cytokine responses.

A rapid increase in IL-12 but not IFN-γ 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, PRRSV-infected 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).

Figure 5
figure 5

Secretion of the Th1 cytokines IL-12 and IFN-γ in PRRSV infected pigs. Plasma samples collected from pigs on day 0 pre-infection (n = 50) and day 2 post-infection of infected (n = 25) and contact (n = 25) pigs were analyzed by ELISA to determine cytokine levels: (A and C) IL-12; (B and D) IFN-γ secreted by indicated pig groups at day 0 and day 2 post-infection. Each bar represents the amount of cytokines secreted by individual pigs, and numbers shown above the horizontal lines indicate the average amount of cytokine from 25 pigs +/- standard deviation. Statistical analysis was performed using a paired t-test by comparing day 0 and day 2 cytokine responses.

Modulation in the frequency of immune cells

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).

Discussion

Modulation in host innate immunity

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 PRRSV-infected 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 [41–46]. 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.

PRRSV rapidly alters the cytokine response

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 PRRSV-infected pigs was detected early post-infection in our study, secretion of IFN-γ remained low. 2 days post-infection 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 post-infection [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 [56–58]. 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.

PRRSV rapidly modulates the frequency of immune cells

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 [26–28]. 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.

Conclusions

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.

Methods

Pigs and Inoculations

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 (HerdChek 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 × 104 TCID50 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.

Collection of blood samples and isolation of PBMC

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 [65–67].

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.

Detection of PRRSV in blood

PRRSV RNA was extracted from plasma using MagMAX-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 log10 values. In addition, a standard curve was developed by preparing 10-fold dilutions of PRRSV VR2332 stock starting at 106 TCID50 per ml for the viral RNA quantification and the results are reported in log10 TCID50 per ml of the plasma as previously described [69].

Pig NK cell cytotoxic assay

The NK cell assay to determine the generic pig NK cell-mediated 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 CO2 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.

Analysis of cytokine response

Plasma samples collected after initial centrifugation of unclotted blood were analyzed for the IFN-α, Th1 (IFN-γ, IL-12), Th2 (IL-4), and immunosuppressive (IL-10) cytokines by cytokine sandwich ELISA as described previously [71].

Flow cytometry

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 anti-rat 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+CD8-cells; 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.

Statistical analyses

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).

Abbreviations

PRRS:

Porcine reproductive and respiratory syndrome

PRRSV:

PRRS virus

NK:

Natural Killer

Tregs:

T-regulatory cells

IFN:

Interferon

IL:

Interleukin.

References

  1. Holtkamp D, Kliebenstein J: PRRS Costs Industry $664 Million Annually. Pork Checkoff Study 2011. [http://www.pork.org/News/1265/PRRSCostsIndustry664Million.aspx]

    Google Scholar 

  2. Koyama S, Ishii KJ, Coban C, Akira S: Innate immune response to viral infection. Cytokine 2008, 43: 336-341. 10.1016/j.cyto.2008.07.009

    Article  PubMed  CAS  Google Scholar 

  3. Van Reeth K, Labarque G, Nauwynck H, Pensaert M: Differential production of proinflammatory cytokines in the pig lung during different respiratory virus infections: correlations with pathogenicity. Res Vet Sci 1999, 67: 47-52. 10.1053/rvsc.1998.0277

    Article  PubMed  CAS  Google Scholar 

  4. Van Reeth K, Nauwynck H: Proinflammatory cytokines and viral respiratory disease in pigs. Vet Res 2000, 31: 187-213. 10.1051/vetres:2000113

    Article  PubMed  CAS  Google Scholar 

  5. Van Reeth K, Van Gucht S, Pensaert M: Correlations between lung proinflammatory cytokine levels, virus replication, and disease after swine influenza virus challenge of vaccination-immune pigs. Viral Immunol 2002, 15: 583-594. 10.1089/088282402320914520

    Article  PubMed  CAS  Google Scholar 

  6. Biron CA, Nguyen KB, Pien GC, Cousens LP, Salazar-Mather TP: Natural killer cells in antiviral defense: function and regulation by innate cytokines. Annu Rev Immunol 1999, 17: 189-220. 10.1146/annurev.immunol.17.1.189

    Article  PubMed  CAS  Google Scholar 

  7. Gerner W, Kaser T, Saalmuller A: Porcine T lymphocytes and NK cells-An update. Dev Comp Immunol 2009, 33: 310-320. 10.1016/j.dci.2008.06.003

    Article  PubMed  CAS  Google Scholar 

  8. Duncan IA, Binns RM, Duffus WP: The null T cell in pig blood is not an NK cell. Immunology 1989, 68: 392-395.

    PubMed  CAS  PubMed Central  Google Scholar 

  9. Raymond C, Wilkie BN: 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. Nat Immun 1998, 16: 18-26.

    Article  PubMed  CAS  Google Scholar 

  10. Renukaradhya GJ, Alekseev K, Jung K, Fang Y, Saif LJ: Porcine reproductive and respiratory syndrome virus-induced immunosuppression exacerbates the inflammatory response to porcine respiratory coronavirus in pigs. Viral Immunol 2010, 23: 457-466. 10.1089/vim.2010.0051

    Article  PubMed  CAS  PubMed Central  Google Scholar 

  11. Dwivedi V, Manickam C, Patterson R, Dodson K, Murtaugh M, Torrelles JB, Schlesinger LS, Renukaradhya GJ: Cross-protective immunity to porcine reproductive and respiratory syndrome virus by intranasal delivery of a live virus vaccine with a potent adjuvant. Vaccine 2011, 29: 4058-4066. 10.1016/j.vaccine.2011.03.006

    Article  PubMed  CAS  Google Scholar 

  12. Albina E, Carrat C, Charley B: Interferon-alpha response to swine arterivirus (PoAV), the porcine reproductive and respiratory syndrome virus. J Interferon Cytokine Res 1998, 18: 485-490. 10.1089/jir.1998.18.485

    Article  PubMed  CAS  Google Scholar 

  13. Murtaugh MP, Xiao Z, Zuckermann F: Immunological responses of swine to porcine reproductive and respiratory syndrome virus infection. Viral Immunol 2002, 15: 533-547. 10.1089/088282402320914485

    Article  PubMed  CAS  Google Scholar 

  14. Jung K, Renukaradhya GJ, Alekseev KP, Fang Y, Tang Y, Saif LJ: 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. J Gen Virol 2009, 90: 2713-2723. 10.1099/vir.0.014001-0

    Article  PubMed  CAS  PubMed Central  Google Scholar 

  15. Xiao Z, Batista L, Dee S, Halbur P, Murtaugh MP: 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. J Virol 2004, 78: 5923-5933. 10.1128/JVI.78.11.5923-5933.2004

    Article  PubMed  CAS  PubMed Central  Google Scholar 

  16. Molina RM, Cha SH, Chittick W, Lawson S, Murtaugh MP, Nelson EA, Christopher-Hennings J, Yoon KJ, Evans R, Rowland RR, et al.: Immune response against porcine reproductive and respiratory syndrome virus during acute and chronic infection. Vet Immunol Immunopathol 2008, 126: 283-292. 10.1016/j.vetimm.2008.08.002

    Article  PubMed  CAS  Google Scholar 

  17. Burstein HJ, Tepper RI, Leder P, Abbas AK: Humoral immune functions in IL-4 transgenic mice. J Immunol 1991, 147: 2950-2956.

    PubMed  CAS  Google Scholar 

  18. Paludan SR: Interleukin-4 and interferon-gamma: the quintessence of a mutual antagonistic relationship. Scand J Immunol 1998, 48: 459-468. 10.1046/j.1365-3083.1998.00435.x

    Article  PubMed  CAS  Google Scholar 

  19. Murtaugh MP, Johnson CR, Xiao Z, Scamurra RW, Zhou Y: Species specialization in cytokine biology: is interleukin-4 central to the T(H)1-T(H)2 paradigm in swine? Dev Comp Immunol 2009, 33: 344-352. 10.1016/j.dci.2008.06.014

    Article  PubMed  CAS  Google Scholar 

  20. Zhou Y, Lin G, Baarsch MJ, Scamurra RW, Murtaugh MP: Interleukin-4 suppresses inflammatory cytokine gene transcription in porcine macrophages. J Leukoc Biol 1994, 56: 507-513.

    PubMed  CAS  Google Scholar 

  21. De Bruin TG, Van Rooij EM, De Visser YE, Bianchi AT: Cytolytic function for pseudorabies virus-stimulated porcine CD4+ CD8dull+ lymphocytes. Viral Immunol 2000, 13: 511-520. 10.1089/vim.2000.13.511

    Article  PubMed  CAS  Google Scholar 

  22. Franco MA, Greenberg HB: Role of B cells and cytotoxic T lymphocytes in clearance of and immunity to rotavirus infection in mice. J Virol 1995, 69: 7800-7806.

    PubMed  CAS  PubMed Central  Google Scholar 

  23. Ober BT, Summerfield A, Mattlinger C, Wiesmuller KH, Jung G, Pfaff E, Saalmuller A, Rziha HJ: Vaccine-induced, pseudorabies virus-specific, extrathymic CD4+CD8+ memory T-helper cells in swine. J Virol 1998, 72: 4866-4873.

    PubMed  CAS  PubMed Central  Google Scholar 

  24. Murray PD, McGavern DB, Pease LR, Rodriguez M: Cellular sources and targets of IFN-gamma-mediated protection against viral demyelination and neurological deficits. Eur J Immunol 2002, 32: 606-615. 10.1002/1521-4141(200203)32:3<606::AID-IMMU606>3.0.CO;2-D

    Article  PubMed  CAS  Google Scholar 

  25. Kaser T, Gerner W, Hammer SE, Patzl M, Saalmuller A: Phenotypic and functional characterisation of porcine CD4(+)CD25(high) regulatory T cells. Vet Immunol Immunopathol 2008, 122: 153-158. 10.1016/j.vetimm.2007.08.002

    Article  PubMed  Google Scholar 

  26. Silva-Campa E, Cordoba L, Fraile L, Flores-Mendoza L, Montoya M, Hernandez J: European genotype of porcine reproductive and respiratory syndrome (PRRSV) infects monocyte-derived dendritic cells but does not induce Treg cells. Virology 2009, 396: 264-271.

    Article  PubMed  Google Scholar 

  27. Silva-Campa E, Flores-Mendoza L, Resendiz M, Pinelli-Saavedra A, Mata-Haro V, Mwangi W, Hernandez J: Induction of T helper 3 regulatory cells by dendritic cells infected with porcine reproductive and respiratory syndrome virus. Virology 2009, 387: 373-379. 10.1016/j.virol.2009.02.033

    Article  PubMed  CAS  Google Scholar 

  28. Wongyanin P, Buranapraditkun S, Chokeshai-Usaha K, Thanawonguwech R, Suradhat S: Induction of inducible CD4+CD25+Foxp3+ regulatory T lymphocytes by porcine reproductive and respiratory syndrome virus (PRRSV). Vet Immunol Immunopathol 2010, 133: 170-182. 10.1016/j.vetimm.2009.07.012

    Article  PubMed  CAS  Google Scholar 

  29. Leroith T, Hammond S, Todd SM, Ni Y, Cecere T, Pelzer KD: A modified live PRRSV vaccine and the pathogenic parent strain induce regulatory T cells in pigs naturally infected with Mycoplasma hyopneumoniae. Vet Immunol Immunopathol 2011.

    Google Scholar 

  30. Dwivedi V, Manickam C, Patterson R, Dodson K, Weeman M, Renukaradhya GJ: 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. Vaccine 2011, 29: 4067-4076. 10.1016/j.vaccine.2011.03.005

    Article  PubMed  CAS  Google Scholar 

  31. Suradhat S, Thanawongnuwech R, Poovorawan Y: Upregulation of IL-10 gene expression in porcine peripheral blood mononuclear cells by porcine reproductive and respiratory syndrome virus. J Gen Virol 2003, 84: 453-459. 10.1099/vir.0.18698-0

    Article  PubMed  CAS  Google Scholar 

  32. Klinge KL, Vaughn EM, Roof MB, Bautista EM, Murtaugh MP: Age-dependent resistance to Porcine reproductive and respiratory syndrome virus replication in swine. Virol J 2009, 6: 177. 10.1186/1743-422X-6-177

    Article  PubMed  PubMed Central  Google Scholar 

  33. Akbari O, DeKruyff RH, Umetsu DT: Pulmonary dendritic cells producing IL-10 mediate tolerance induced by respiratory exposure to antigen. Nat Immunol 2001, 2: 725-731. 10.1038/90667

    Article  PubMed  CAS  Google Scholar 

  34. Kimman TG, Cornelissen LA, Moormann RJ, Rebel JM, Stockhofe-Zurwieden N: Challenges for porcine reproductive and respiratory syndrome virus (PRRSV) vaccinology. Vaccine 2009, 27: 3704-3718. 10.1016/j.vaccine.2009.04.022

    Article  PubMed  CAS  Google Scholar 

  35. Mateu E, Diaz I: The challenge of PRRS immunology. Vet J 2008, 177: 345-351. 10.1016/j.tvjl.2007.05.022

    Article  PubMed  CAS  Google Scholar 

  36. Dimova T, Mihaylova A, Spassova P, Georgieva R: Superficial implantation in pigs is associated with decreased numbers and redistribution of endometrial NK-cell populations. Am J Reprod Immunol 2008, 59: 359-369. 10.1111/j.1600-0897.2007.00579.x

    Article  PubMed  Google Scholar 

  37. Toka FN, Nfon C, Dawson H, Golde WT: Natural killer cell dysfunction during acute infection with foot-and-mouth disease virus. Clin Vaccine Immunol 2009, 16: 1738-1749. 10.1128/CVI.00280-09

    Article  PubMed  CAS  PubMed Central  Google Scholar 

  38. Buddaert W, Van Reeth K, Pensaert M: In vivo and in vitro interferon (IFN) studies with the porcine reproductive and respiratory syndrome virus (PRRSV). Adv Exp Med Biol 1998, 440: 461-467. 10.1007/978-1-4615-5331-1_59

    Article  PubMed  CAS  Google Scholar 

  39. Chen Z, Lawson S, Sun Z, Zhou X, Guan X, Christopher-Hennings J, Nelson EA, Fang Y: 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. Virology 2010, 398: 87-97. 10.1016/j.virol.2009.11.033

    Article  PubMed  CAS  Google Scholar 

  40. Delputte PL, Van Breedam W, Barbe F, Van Reeth K, Nauwynck HJ: IFN-alpha treatment enhances porcine Arterivirus infection of monocytes via upregulation of the porcine Arterivirus receptor sialoadhesin. J Interferon Cytokine Res 2007, 27: 757-766. 10.1089/jir.2007.0001

    Article  PubMed  CAS  Google Scholar 

  41. Nguyen KB, Salazar-Mather TP, Dalod MY, Van Deusen JB, Wei XQ, Liew FY, Caligiuri MA, Durbin JE, Biron CA: Coordinated and distinct roles for IFN-alpha beta, IL-12, and IL-15 regulation of NK cell responses to viral infection. J Immunol 2002, 169: 4279-4287.

    Article  PubMed  CAS  Google Scholar 

  42. Liang S, Wei H, Sun R, Tian Z: IFNalpha regulates NK cell cytotoxicity through STAT1 pathway. Cytokine 2003, 23: 190-199. 10.1016/S1043-4666(03)00226-6

    Article  PubMed  CAS  Google Scholar 

  43. Ostensen M, Forre O: Modulation of human natural killer cell function by cytokines and rheumatic disease. Scand J Rheumatol Suppl 1988, 76: 183-188.

    Article  PubMed  CAS  Google Scholar 

  44. Hervas-Stubbs S, Perez-Gracia JL, Rouzaut A, Fernandez de Sanmamed M, Le Bon A, Melero IJ: Direct Effects of type I IFNs on cells of the immune system. Clin Cancer Res 2011.

    Google Scholar 

  45. Trinchieri G, Santoli D, Granato D, Perussia B: Antagonistic effects of interferons on the cytotoxicity mediated by natural killer cells. Fed Proc 1981, 40: 2705-2710.

    PubMed  CAS  Google Scholar 

  46. Lee CK, Rao DT, Gertner R, Gimeno R, Frey AB, Levy DE: Distinct requirements for IFNs and STAT1 in NK cell function. J Immunol 2000, 165: 3571-3577.

    Article  PubMed  CAS  Google Scholar 

  47. Hagberg N, Berggren O, Leonard D, Weber G, Bryceson YT, Alm GV, Eloranta ML, Ronnblom L: 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. J Immunol 2011, 186: 5085-5094. 10.4049/jimmunol.1003349

    Article  PubMed  CAS  Google Scholar 

  48. Knoblock KF, Canning PC: Modulation of in vitro porcine natural killer cell activity by recombinant interleukin-1 alpha, interleukin-2 and interleukin-4. Immunology 1992, 76: 299-304.

    PubMed  CAS  PubMed Central  Google Scholar 

  49. Foss DL, Zilliox MJ, Meier W, Zuckermann F, Murtaugh MP: Adjuvant danger signals increase the immune response to porcine reproductive and respiratory syndrome virus. Viral Immunol 2002, 15: 557-566. 10.1089/088282402320914502

    Article  PubMed  CAS  Google Scholar 

  50. Kobayashi M, Fitz L, Ryan M, Hewick RM, Clark SC, Chan S, Loudon R, Sherman F, Perussia B, Trinchieri G: Identification and purification of natural killer cell stimulatory factor (NKSF), a cytokine with multiple biologic effects on human lymphocytes. J Exp Med 1989, 170: 827-845. 10.1084/jem.170.3.827

    Article  PubMed  CAS  Google Scholar 

  51. Stern AS, Podlaski FJ, Hulmes JD, Pan YC, Quinn PM, Wolitzky AG, Familletti PC, Stremlo DL, Truitt T, Chizzonite R, et al.: Purification to homogeneity and partial characterization of cytotoxic lymphocyte maturation factor from human B-lymphoblastoid cells. Proc Natl Acad Sci USA 1990, 87: 6808-6812. 10.1073/pnas.87.17.6808

    Article  PubMed  CAS  PubMed Central  Google Scholar 

  52. Manetti R, Parronchi P, Giudizi MG, Piccinni MP, Maggi E, Trinchieri G, Romagnani S: 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. J Exp Med 1993, 177: 1199-1204. 10.1084/jem.177.4.1199

    Article  PubMed  CAS  Google Scholar 

  53. Hsieh CS, Macatonia SE, Tripp CS, Wolf SF, O'Garra A, Murphy KM: Development of TH1 CD4+ T cells through IL-12 produced by Listeria-induced macrophages. Science 1993, 260: 547-549. 10.1126/science.8097338

    Article  PubMed  CAS  Google Scholar 

  54. Gillessen S, Carvajal D, Ling P, Podlaski FJ, Stremlo DL, Familletti PC, Gubler U, Presky DH, Stern AS, Gately MK: Mouse interleukin-12 (IL-12) p40 homodimer: a potent IL-12 antagonist. Eur J Immunol 1995, 25: 200-206. 10.1002/eji.1830250133

    Article  PubMed  CAS  Google Scholar 

  55. Xing Z, Zganiacz A, Santosuosso M: 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. J Leukoc Biol 2000, 68: 897-902.

    PubMed  CAS  Google Scholar 

  56. Delputte PL, Nauwynck HJ: Porcine arterivirus infection of alveolar macrophages is mediated by sialic acid on the virus. J Virol 2004, 78: 8094-8101. 10.1128/JVI.78.15.8094-8101.2004

    Article  PubMed  CAS  PubMed Central  Google Scholar 

  57. Van Gorp H, Van Breedam W, Delputte PL, Nauwynck HJ: Sialoadhesin and CD163 join forces during entry of the porcine reproductive and respiratory syndrome virus. J Gen Virol 2008, 89: 2943-2953. 10.1099/vir.0.2008/005009-0

    Article  PubMed  CAS  Google Scholar 

  58. Van Breedam W, Van Gorp H, Zhang JQ, Crocker PR, Delputte PL, Nauwynck HJ: The M/GP(5) glycoprotein complex of porcine reproductive and respiratory syndrome virus binds the sialoadhesin receptor in a sialic acid-dependent manner. PLoS Pathog 2010, 6: e1000730. 10.1371/journal.ppat.1000730

    Article  PubMed  PubMed Central  Google Scholar 

  59. Yang H, Parkhouse RM: Characterization of the porcine gammadelta T-cell receptor structure and cellular distribution by monoclonal antibody PPT27. Immunology 2000, 99: 504-509. 10.1046/j.1365-2567.2000.00019.x

    Article  PubMed  CAS  PubMed Central  Google Scholar 

  60. Christianson W, Joo H: Porcine reproductive and respiratory syndrome: a review. Swine Health Prod 1994, 10-28.

    Google Scholar 

  61. Olin MR, Batista L, Xiao Z, Dee SA, Murtaugh MP, Pijoan CC, Molitor TW: Gammadelta lymphocyte response to porcine reproductive and respiratory syndrome virus. Viral Immunol 2005, 18: 490-499. 10.1089/vim.2005.18.490

    Article  PubMed  CAS  Google Scholar 

  62. Ezquerra A, Revilla C, Alvarez B, Perez C, Alonso F, Dominguez J: Porcine myelomonocytic markers and cell populations. Dev Comp Immunol 2009, 33: 284-298. 10.1016/j.dci.2008.06.002

    Article  PubMed  CAS  Google Scholar 

  63. Otake S, Dee S, Corzo C, Oliveira S, Deen J: Long-distance airborne transport of infectious PRRSV and Mycoplasma hyopneumoniae from a swine population infected with multiple viral variants. Vet Microbiol 2010, 145: 198-208. 10.1016/j.vetmic.2010.03.028

    Article  PubMed  Google Scholar 

  64. Murtaugh MP, Yeske P: Epidemiology of a new PRRS virus isolate and Outbreak. In Proceedings of the Allen D Leman Swine Conference St Paul Edited by: Smith T. 2008, 11-15.

    Google Scholar 

  65. VanCott JL, Brim TA, Simkins RA, Saif LJ: Isotype-specific antibody-secreting cells to transmissible gastroenteritis virus and porcine respiratory coronavirus in gut- and bronchus-associated lymphoid tissues of suckling pigs. J Immunol 1993, 150: 3990-4000.

    PubMed  CAS  Google Scholar 

  66. Khatri M, Dwivedi V, Krakowka S, Manickam C, Ali A, Wang L, Qin Z, Renukaradhya GJ, Lee CW: Swine influenza H1N1 virus induces acute inflammatory immune responses in pig lungs: a potential animal model for human H1N1 influenza virus. J Virol 2010.

    Google Scholar 

  67. Mulupuri P, Zimmerman JJ, Hermann J, Johnson CR, Cano JP, Yu W, Dee SA, Murtaugh MP: Antigen-specific B-cell responses to porcine reproductive and respiratory syndrome virus infection. J Virol 2008, 82: 358-370. 10.1128/JVI.01023-07

    Article  PubMed  CAS  PubMed Central  Google Scholar 

  68. Wasilk A, Callahan JD, Christopher-Hennings J, Gay TA, Fang Y, Dammen M, Reos ME, Torremorell M, Polson D, Mellencamp M, et al.: 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. J Clin Microbiol 2004, 42: 4453-4461. 10.1128/JCM.42.10.4453-4461.2004

    Article  PubMed  CAS  PubMed Central  Google Scholar 

  69. Prickett J, Simer R, Christopher-Hennings J, Yoon KJ, Evans RB, Zimmerman JJ: Detection of Porcine reproductive and respiratory syndrome virus infection in porcine oral fluid samples: a longitudinal study under experimental conditions. J Vet Diagn Invest 2008, 20: 156-163. 10.1177/104063870802000203

    Article  PubMed  Google Scholar 

  70. Korzeniewski C, Callewaert DM: An enzyme-release assay for natural cytotoxicity. J Immunol Methods 1983, 64: 313-320. 10.1016/0022-1759(83)90438-6

    Article  PubMed  CAS  Google Scholar 

  71. Azevedo MS, Yuan L, Pouly S, Gonzales AM, Jeong KI, Nguyen TV, Saif LJ: Cytokine responses in gnotobiotic pigs after infection with virulent or attenuated human rotavirus. J Virol 2006, 80: 372-382. 10.1128/JVI.80.1.372-382.2006

    Article  PubMed  CAS  PubMed Central  Google Scholar 

  72. Renukaradhya GJ, Manickam C, Khatri M, Rauf A, Li X, Tsuji M, Rajashekara G, Dwivedi V: Functional invariant NKT cells in pig lungs regulate the airway hyperreactivity: a potential animal model. J Clin Immunol 2010, in press.

    Google Scholar 

  73. Kaser T, Gerner W, Hammer SE, Patzl M, Saalmuller A: Detection of Foxp3 protein expression in porcine T lymphocytes. Vet Immunol Immunopathol 2008, 125: 92-101. 10.1016/j.vetimm.2008.05.007

    Article  PubMed  Google Scholar 

Download references

Acknowledgements

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.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Gourapura J Renukaradhya.

Additional information

Competing interests

The authors declare that they have no competing interests.

Authors' contributions

MPM, DL, and GJR conceived and designed the study and wrote the paper. VD, CM, BB, and GJR performed the experiments and analyzed the data. All authors read and approved the final manuscript.

Authors’ original submitted files for images

Rights and permissions

Open Access This article is published under license to BioMed Central Ltd. This is an Open Access article is distributed under the terms of the Creative Commons Attribution License ( https://creativecommons.org/licenses/by/2.0 ), which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.

Reprints and permissions

About this article

Cite this article

Dwivedi, V., Manickam, C., Binjawadagi, B. et al. Evaluation of immune responses to porcine reproductive and respiratory syndrome virus in pigs during early stage of infection under farm conditions. Virol J 9, 45 (2012). https://doi.org/10.1186/1743-422X-9-45

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/1743-422X-9-45

Keywords