SAES-422 Multistate Research Activity Accomplishments Report

Status: Approved

Basic Information

Participants

Chair: Meng, X.J. - Virginia Polytechnic Institute and State University (VA Tech) - xjmeng@vt.edu; Secretary: Christopher-Hennings, Jane - SDSU - jane.hennings@sdstate.edu; Rowland, Raymond R.R. - Kansas State University (KSU)- browland@vet.k-state.edu; Benfield, David - Ohio State University (OSU)- benfield.2@osu.edu; Faaberg, Kay, - National Animal Disease Center (NADC)- kay.faaberg@ars.usda.gov; Goldberg, Tony - University of Wisconsin-Madison - tgoldberg@vetmed.wisc.edu; Gourapura, Renukaradhya J. - The Ohio State University (OSU)- gourapura.1@osu.edu; Johnson, Peter - USDA, CSREES - pjohnson@reeusda.gov; Lunney, Joan - USDA-ARS, BARC - joan.lunney@ars.usda.gov; Murtaugh, Michael P - University of Minnesota (UMN)- murta001@umn.edu; Osorio, Fernando A.- University of Nebraska-Lincoln (UNL) - fosorio@unl.edu; Pogranichniy, Roman - (Purdue), IN - rmp@purdue.edu; Risatti, Guillermo - University of Connecticut - guillermo.risatti@uconn.edu.; Tompkins, S. Mark - University of Georgia (UGA)- smt@uga.edu; Yuan, Shishan - Shanghai Veterinary Res. Inst. (SHVRI), SHVRI - shishanyuan@hotmail.com; Zhang, Yanjin - University of Maryland - zhangyj@umd.edu; Zimmerman, Jeff - Iowa State University (ISU)- jjzimm@iastate.edu; Zuckermann, Federico - University of Illinois at Urbana-Champaign (UIUC) - fazaaa@illinois.edu;<p> <b>Other NC229 Scientists: </b>; Abrams, Sam - BARC; Anderson, Tavis - UWI; Araujo, Karla - BARC; Araujo, Karla - BARC; Arceo M - Purdue; Baker, RB - ISU; Blecha, Frank - KSU; Boddicker, Nick - ISU; Brockmeier, Susan - NADC; Calvert, Jay - Pfizer Animal health; Carman, Susy - University of Guelph, Canada; Chang, KC - KSU; Chen, Hongbo - USDA-BARC; Choi, Igseo - BARC; Ciobanu, Dan - UNL; Clark, A. - Purdue University; Davies, Peter -UMN; Dee, Scott - UMN; Dekkers, Jack - ISU; Ernst, Cathy - MSU; Fang, Ying - SDSU; Garmendia, Antonio - UCONN; Garrick, Dorian - ISU; Gourapura, Aradhya - OSU; Gramer, Marie - UMN; Halbur, Patrick -ISU; Haley, Charles - USDA-APHIS; Harhay, Greg - NADC; Harris, DL (Hank) - ISU; Hause, Ben - Newport Labs, MN; Hesse, Dick - KSU; Holtkamp, Derald J - ISU; Huang T - Purdue; Huang, Tinghua - ISU; Jiang, Zhihua - WSU; Johnson, John K - ISU; Joo, Han Soo - UMN; Karriker, Locke - ISU; Kerhli, Marcus Jr. - NADC; Kerrigan M. - KSU; Kittawornrat Apisit - ISU; Kuhar, D. - USDA-USDA-BARC; Laegried, Will - UIUC; Lager, Kelly - NADC; Lawson, Steve - SDSU; Lazar V -Purdue ; Leung, Frederick - Hong Kong University; Loving, Crystal - NADC; Lunney, Joan - BARC; Main, Rodger G - ISU; McCaw, Monte B. (deceased) - NCSU; McKean, JD - ISU; Moore, B - Purdue; Morrison, Robert - UMN; Nelson, Eric - SDSU; Nerem, Joel Pipestone - Vet Clinic, MN; Nicholson, Tracy - NADC; Opriessnig, Tanja - ISU; Pattnaik, Asit - UNL; Prickett, J. - ISU; Ramamoorthy, Sheila - UGA; Ramirez, Alejandro - ISU; Ramirez-Nieto, Gloria - Universidad Nacional de Colombia; Raney NE - Purdue; Raney, Nancy - MSU; Reecy, Jim - ISU; Rossow, Kurt - UMN; Roth, JA - ISU; Rothschild, Max - ISU; Rovira, Albert - UMN; Rowland, R.R.R. - KSU; Sang, Yongming - KSU; Schwartz, Kent J. - ISU; Sina, R - Purdue; Souza, Carlos - BARC; Steibel, J.P. - MSU; Stevenson, Greg W. - ISU; Strait, Erin - ISU; Srinivas, Jay - CVB-PEL/APHIS/USDA; Torremorell, Montserrat - UMN; Trible B. - KSU; Tripp, Ralph - UGA; Tuggle, Chris - ISU; Waide, Emily - ISU; Wang, Chong -ISU; Wang, Xiuqing -SDSU; Wyatt, Carol - KSU; Wysocki, Michal -USDA-BARC; Yoo, Dongwan -UIUC; Yoon, Kyoung-Jin -ISU; Zhang, C. - VA Tech; Zhu, Xiaoping - UMD; Zimmerman, Jeff - ISU;

NC229 Meeting Chicago, IL, 12/02/2010 Brief Summary of Minutes of Annual Meeting. (Held on Friday, Dec. 2, at 1:00 PM -3:00 PM) 1:00 XJ Meng, opening remarks 1:05 Update of NC229, Dr. David Benfield, Administrative Advisor ; 2012 appears to be flat for NC229; 2013 budget unknown due to election For NC229 funding , current funding expires Sept.. 30th, 2014, so a new project would need to be written in 2013 for 2014. In 2012, there is a mid-year review. The original NC229 project was written for PRRSV research primarily (with a push for the PRRSV CAP programs) and then expanded to PRRSV + emerging viral diseases of swine. With the CAP program ending, for the next written proposal, may want to consider emphasis on other viruses since some stations do not get any funding from their station directors (eg. to leverage other opportunities for funding). 1:15 Election of new NC-229 officer (Dr. Fernando Osorio was elected Secretary for NC229. Dr. Jane Christopher-Hennings is the Chair starting 2012, replacing Dr. XJ Meng who was chair for last 2 years) 1:20 Update from USDA-NIFA (Drs. Peter Johnson/Margo Holland): Dr. Margo Holland: FY 2012 funding is flat. AFRI Indirect costs will increase to 30% for 2012 funding. No less than 40% for applied research No less than 2% for equipment grants 60% is for basic research (with 30% focused on multi-disciplinary teams) 2012 Foundation Programs to be released in early 2012 (need letter of intent) Areas of interest include Animal Health and Production; Agricultural systems (may want to put emphasis on nanotechnology) Grants generally $500,000 total 2011 Animal Health Foundation program had > 200 letters of intent with 160 proposals received; 12 proposals recommended for funding (8.3% success rate) with 4 awards for PRRSV research (3 standard research proposals and 1 seed grant) representing vaccine, health surveillance and host-pathogen interactions. PRRSV CAP has been $1.2 million/year and is considered a success story of a multi-disciplinary group. $ 3 million has been leveraged (eg. NPB, University, Fed. Gov. private companies) NIFA Fellowships: Pre-docs $75,000 (for 2 years) Post-docs $130,000 (for 2 years) Dr. Peter Johnson: NEW funding on Ecology and evolution of disease (NIFA, NSF, NIH) Due 1st Wed in Dec (this year Dec. 7th). Need to appeal to all agencies, http://nsf.gov/pubs/2011/nsf11580/nsf11580.htm

Accomplishments

Objective 1. Elucidate the mechanisms of host-pathogen(s) interactions. 1.1 (NADC, Faaberg/Brockmeier/Loving/Miller) Compared pathogenesis in swine after challenge with 3 new Type 2 PRRSV field isolates in vivo and in vitro. Differences seen in viral growth kinetics and pathogenesis. 1.2 (NADC, Faaberg/Brockmeier/Loving/Miller) Compared growth and disease in swine after vaccination with nsp2 deletion mutant viruses and challenge with novel Type 2 isolate. 1.3 (NADC, Kehrli/Miller/Faaberg) Adenovirus expression of a region of PRRSV nsp 2. 1.4 (NADC Faaberg/Lager/Miller/Brockmeier/Kerhli/Nicholson) Compared high and low dose challenge of US swine with Chinese and Vietnamese HP-PRRSV. 1.5 (NADC, Faaberg)Development of infectious clone of a new vaccine strain. 1.6 (NADC, Faaberg) Examined the interaction of nsp2 with host genes. 1.7 (NADC, Miller/Kehrli) Developed PRRSV infected tissue culture assay to screen for polyclonal B-cell activation. 1.8 (NADC, Miller/Kehrli/Faaberg)Assessed PRRSV strains that have a reduced capacity for the induction of polyclonal B-cell activation for potential vaccine candidates. 1.9 (NADC, Miller; G Rohrer USMARC) Sample collection and genomic DNA purification for SNP chip analysis to examine genotype in host susceptibility to PCV2. 1.10 (NADC, Miller) Compared the transcript expression of tracheobronchial lymph nodes of pigs infected with PRV/PRRSV/SIV/PCV-2 in vivo. 1.11 (NADC, Miller) Determine the transcriptomic immune response to contemporary US and Asian PRRSV in vivo. 1.12 (NADC, Faaberg/Miller; B. Guo, Visiting Scientist) Developed type 1 IFN bioassay for examination of PRRSV strain specific pathology. 1.13 (NADC, Faaberg) Developing chimeric vaccine to Asian HP-PRRSV. 1.14 (UNL) Understanding the role of the nsp1 in PRRSV--Using nsp1², a proteolytically processed functional product of nsp1 as bait, we have identified the cellular poly (C)-binding proteins 1 and 2 (PCBP1 and PCBP2) as 2 of its interaction partners. Interactions of PCBP1 and 2 with nsp1² was confirmed. In MARC cells, the cytoplasmic PCBP1 and 2 partially co-localize to the viral replication-transcription complexes. Recombinant purified PCBP1 and 2 bound to the viral 5 UTR. SiRNA-mediated silencing of PCBP1 and 2 in cells resulted in significantly reduced replication and transcription without effects on initial polyprotein synthesis. ID of cellular factors involved in PRRSV lifecycle give a better understanding of virus biology and has potential for development of anti-viral therapeutics. 1.15 (UMN) discovered a novel PRRSV protein expressed in infected cells that may have a role in cellular pathogenesis. 1.16 (UMN) characterized the molecular pathogenesis of PCV2 infection by transcriptome profiling. Discovered a pronounced gene expression induction profile characteristic of a classical IFN response to viral infection. 1.17 (PURDUE) Host immune responses to PRRSV infection may be correlated with genetic control. The PRRS Host Genetics Consortium (PHGC) studies are aimed at identifying genes and pathways associated with pigs that clear PRRS virus while continuing to gain weight. Analyses of data from each PHGC trial [viral load from 0-21 days post infection (dpi) and weight gain from 0-42 dpi] were used to statistical identify 4 groups of pigs: those with the best phenotype, low virus and high growth (LvHg), high virus and high growth (HvHg), high virus and low growth (HvLg), and, the worst, low virus and low growth (LvLg). Real time PCR was used with primers corresponding to markers important for immune system activation. Markers included: transcription factors TBX21 (T-bet), GATA3 (presumed Th1 and Th2 regulators, respectively), FOXP3, cytokines IL10, IFNg, CD163, the PRRSV receptor, and CD69, the early marker for T cell activation and proliferation. LvHg animals at 4-10 DPI exhibited increased ratio of TBX21/GATA3. LvHg animals had a robust increase in expression of IL10 and IFNG within 4-14 DPI, with high basal expression of CD69 relative to other groups at 0 DPI. In HvHg, TBX21/GATA3 was the highest during PRRSV infection, up regulation and expression of TBX21 and GATA3 was minimal, along with other markers. HvLg animals had high base line expression of GATA3 and FOXP3 transcription factors and delayed increase of TBX21/GATA3. In LvLg animals a high basal expression of GATA3 and FOXP3 was detected with delayed increased of TBX21/GATA3. In LvLg animals higher baseline expression for CD163 and cytokine IL10 markers was detected. The HvLg and LvLg animals exhibited up regulation of the markers responsible for Th2 immunity during infection. Our data correlated basal expression of CD69 as a factor leading to up regulation and activation of markers responsible for Th1 immune response. Basal expression of GATA3 and FOXP3 demonstrated activation of the markers responsible for Th2 pathway during infection. 1.18 (KSU) Sang, Blecha,Rowland characterized the expression 39 type I IFN genes and related receptors in the PRRSV-infected fetus. 1.19 (KSU) Hesse, Rowland performed an analysis of cross-protection between diverse PRRSV strains. 1.20 (KSU) Rowland identified a decoy epitope CP (169-180) in PCV2 capsid protein produced to immunization with capsid monomer. Immunization with whole capsid offered protection. This explains why production of large quantities of virus during infection are non-protective. 1.21 (SHVRI) report viable chimeric viruses in which the envelope protein genes from ORF2a to ORF5 of vSHE (type 1) were swapped into the genetic backbone of vAPRRS (type 2). Found that the envelope proteins of type 1 were fully functional in type 2 PRRSV and the rescued chimeric progeny viruses showed robust genetic stability and similar replication properties to the parental strains in vitro. For PRRSV N protein, we found that type 1 N protein was functional in the type 2 PRRSV backbone. All cysteines in the N protein are non-essential for type 1 and 2 PRRSV viability. NEM treatment prevents disulfide-linked N dimerization in cells but not in extracellular virions. 1.22 (UCONN; Garmendia): Aims are to determine the sensitivity to and induction of IFNb by PRRSV isolates, to identify mechanisms of evasion of host's innate immune responses by PRRSV and to determine correlations with virulence. Field viruses were previously tested in their sensitivity to IFNb in vitro. Significant differences in sensitivity to IFNb among different PRRSV isolates and between MARC-145 cells and porcine alveolar macrophages (PAM) were demonstrated. The induction of IFNb by PRRSV was tested in PAMs using field isolates and a series of chimeric viruses derived from infectious clones of FL12 (virulent isolate) and a vaccine virus (attenuated), provided by Dr. Osorio, UNL. The induction tests demonstrated that PRRSV isolates do induce IFNb in PAMs but such induction is variable. The IFNb induced was bioactive as shown by a dose-dependent reduction of VSV infectivity in cell culture. Mx and TNF were measured in PRRSV-infected PAMs. Mx is expressed at significant levels that generally correlate with the corresponding levels of IFNb produced. TNF production is detectable but levels are not significant and do not follow a pattern related to IFNb. 1.23 (ISU) Studies on enhanced pneumonia and disease in pigs vaccinated with an inactivated human-like (´-cluster) H1N2 vaccine and challenged with 2009 pH1N1 influenza virus. 1.24 (ISU) Studies on cytokine and chemokine mRNA expression profiles in tracheobronchial LN from pigs singularly infected or coinfected with PCV2 and Mycoplasma hyopneumoniae. 1.25 (ISU) Studies on genetic and phenotypic characterization of a 2006 US PRRSV isolate associated with high morbidity and mortality in the field. 1.26 (ISU) Studies on the establishment of a DNA-launched infectious clone for a highly pneumovirulent type 2 PRRSV: Identification and in vitro and in vivo characterization of a large spontaneous deletion in the nsp2 region. 1.27 (OSU) To elucidate both cellular and innate cytokine response in growing pigs at very early stages of PRRSV infection in a commercial pig herd premises, 7 wk old pigs were infected. 1 in a pen of 25 was PRRSV infected and responses were assessed 2 DPI. All the infected and a few of the contact neighbor pigs were viremic. A majority of viremic pigs had more than 50% reduction in NK cell-cytotoxicity. At 2 DPI 1 fold increase in innate IFNa production was detected in plasma. Enhanced secretion of IL4, IL10 and IL12 (but not IFNg) in a majority of infected pigs was seen. A reduced frequency of myeloid cells, CD8+ and CD4+CD8+ T cells and increased T-regulatory cell population was detected in all the viremic pigs. Our results suggest that PRRSV modulate the innate and adaptive immune mediators from 2 DPI, resulting in subversion of host innate immunity from early stages post-infection. 1.28 (VA) We identified a large spontaneous deletion of 435-bp in the nsp2 gene of a highly pneumovirulent PRRSV, VR2385 . We established a DNA-launched infectious clone (passage 14) containing the 435-bp nsp2 deletion (pIR-VR2385-CA) and another DNA-launched infectious clone, pIR-VR2385-R, in which we restored the deleted 435-bp nsp2 sequence back to the pIR-VR2385-CA backbone. The growth characteristics of the 2 rescued viruses were compared, and results showed that the VR2385-CA virus with the nsp2 deletion replicated more efficiently in vitro than the VR2385-R virus with the restored nsp2 sequence but, VR2385-CA virus had a significantly reduced serum viral RNA load in vivo. In pigs, the nsp2 deletion had no effect on virulence. The spontaneous nsp2 deletion has a role for enhanced virus replication in vitro but has no effect on pathogenicity. 1.29 (VA) To determine whether cellular microRNAs (miRNAs) play a role in host response to PRRSV infection, we performed a global profiling of both cellular miRNA and mRNA in MARC cells infected with type 1 (SD01-08) or type 2 (VR2385) PRRSV. Results showed that the expressions of approximately 240 miRNAs were significantly altered with infection by PRRSV type 1 or 2 (114 for type 1, and 82 for type 2), and at least 15 specific miRNAs were shared by both types. Approximately 4,500 genes showed differential expression with infection by either virus type (p<0.05). We conducted a global human/bovine/porcine miRNA and porcine gene expression microarray analyses using a pool of lung homogenates of 10 SPF pigs at 14 DPI. Compared to the negative control, PRRSV infection resulted in significant changes of the expression level (>2-fold) of 17 miRNAs (p<0.05) and 3,713 mRNAs (p<0.01) including genes involved in host innate and adaptive immune responses. We found 270 unique miRNAs that were differentially up- (151 miRNAs) or down-regulated (119 miRNAs) in response to PRRSV infection. Deep sequencing data showed 1,892 novel putative porcine miRNAs that do not align to any known Sus scrofa miRNAs. We have correlated inverse regulation between miRNAs and putative target genes to build a miRNA-gene network. 1.30 (SDSU: Z Sun, Y Li, Y Fang) investigated ISG15 and PRRSV nsp2 OTU domain mediated deisgylation function. IFN-stimulated gene 15 (ISG15) is an ubiquitin-like protein which is stimulated by type I IFN ±/² or induced by viral or bacterial infection. Over-expression of ISG15 in cells significantly reduced the PRRSV titer and was confirmed using small interfering RNAs against ISG15-conjugating enzymes. IFN-induced antiviral activity is significantly alleviated by inhibiting ISG15 conjugation. In vitro deISGylation assay showed the N-terminal OTU domain of nsp2 has deconjugating activity towards ISGylated products. A 19 AA deletion plus a single AA mutation partially relieved the nsp2 de-ISGylation function. This showed that ISG15 conjugation has an important role in PRRSV infection and modifying certain regions of nsp2 could reduce the deISGylation ability of the virus. 1.31 (SDSU, KSU Lawson,Li, Patton, Langenhorst, Sun, Jiang, Hennings, Nelson, Knudsen, Fang, Chang). Constructed a recombinant PRRSV that encodes swine IL-1b as a separate subgenomic mRNA inserted between ORFlb and ORF2. MARC cells infected with recombinant virus secreted ILlb and had a similar growth rate to parental virus. No clinical signs were observed in the recombinant virus-infected nursery pigs and IL-lb, IL4 and IFNg were up-regulated in PBMCs. 1.32 (SDSU, LUMC, NL: Li, Tas, Sun, Snijder, Fang). MAbl and polyclonal antibodies were generated against PRRSV ORF1a-encoded nsps. Using these antibodies, we identified and characterized these ORFla-encoded nsps in infected MARC cells. This study confirmed the existence of proteolytic processing products of PRRSV ORFla encoded polyprotein in virus infected cells and provides a basis for both applied and basic research on the role of PRRSV nsps in viral replication and pathogenesis. 1.33 (BARC) The PRRS Host Genetics Conso rtium (PHGC) has expanded efforts to determine the role of host genetics in resistance to PRRS and in effects on pig health and related growth effects. The PHGC is a multi-year project that is funded by a US consortium representing the US National Pork Board (NPB), USDA ARS and NIFA, universities and private companies; it represents the first-of-its-kind approach to food animal infectious disease research. The project has used a Nursery Pig Model to assess pig resistance/susceptibility to primary PRRSV infection. Ten sets of 200 crossbred pigs from high health farms were donated by commercial sources and transported KSU. Pigs were infected with PRRSV and followed for 42 DPI. Blood was collected at 0,4,7,10,14,21,28,35 and 42 DPIi and weekly weights recorded. All 10 trials have been completed; each trial has affirmed that all pigs become PRRSV infected but some pigs clear virus from serum quicker with variable weight effects. Data is being stored in the PHGC database at ISU www.animalgenome.org/lunney. 1.34 (BARC) Genome wide association studies have identified genomic regions that determine PRRS resistance/susceptibility. DNA from PHGC pigs has been genotyped with the PorcineSNP60 Genotyping BeadChip (> 60K single nt polymorphisms or SNPs). (Data stored at ISU www.animalgenome.org/lunney. Multivariate analyses of viral load and weight data have identified PHGC pigs in different virus/weight categories, so that ongoing serum cytokine and gene expression studies can compare data from PRRS resistant/maximal growth pigs to susceptible/reduced growth pigs. PRRS CAP, NRSP8 Swine Genome Coordinator and Genome Alberta have supported SNP chip analyses and PRRS CAP the state-of-the-art genome wide association studies (GWAS) to identify genetic determinants of PRRS resistance/susceptibility. Based on evaluation of phenotypic traits, viral load [VL, area under the curve 0-21 dpi], and 0-42dpi weight gain (WG), response to PRRSV challenge was shown to be moderately heritable at 0.30 each. Regions on swine chromosomes 4 (SSC4) and SSCX appear to be associated with VL, and on SSC1, 2, 3, 4, 7, and 17 with WG. Regions on SSC4 for VL and WG are almost perfectly negatively correlated. Investigations revealed that the BB genotype for this region of SSC4 is the desirable genotype and has a low frequency (0.02), suggesting that genetic progress can be made by selective breeding. Functional gene and protein transcriptomic analyses are ongoing to ID gene networks and resistance associated biomarkers that differ in high versus low VL pigs. 1.35 (BARC) Identifying host gene expression changes that are involved in regulating responses to PRRSV infection. Grants from USDA NIFA and Genome Alberta are supporting whole blood gene expression analyses using microarrays and RNAseq (next generation transciptomics), respectively. The goal is to assess differential expression of individual genes and to discover networks and pathways enriched for those genes, in pigs showing different responses to PRRSV infection. RNA from 3 pigs per group was hybridized to the 20K 70-mer oligonucleotide Pigoligoarray following a blocked reference design with time 0 of each individual animal as a reference sample. Expression levels for 491 genes showed significant viral level-growth interaction for all time-points. Differentially expressed (DE) genes at early time-points (4, 7, 14 dpi) were evaluated by enrichment analysis with Ingenuity Pathways Analysis software www.ingenuity.com; for comparisons of viral level, 308 genes were DE with 16 significant gene networks (p d0.0001); for growth level, 367 genes were DE with 17 significant gene networks. Confirmatory qPCR work will explore these DE genes and their roles in PRRS control. The more significant biological functions identified (FDRd 5%) were those related to cell death, cellular function, maintenance and compromise, and inflammatory disease. For the growth comparison at 4 dpi in Lv animals, the antigen presentation pathway was over-represented (FDRd5%). 1.36 (UMD) found that PRRSV inhibits IFN downstream signaling and continued the studies to identify the interference mechanism. We found that NSP1² of PRRSV VR2385 interferes with IFN signaling pathway. VR2385 is a virulent strain whereas NSP1² of Ingelvac PRRS MLV does not affect IFN signaling. NSP1² of VR2385 blocks nuclear translocation of the IFN-induced STAT1/STAT2/p48 heterotrimer. IFN-induced phosphorylation of both STAT1 and STAT2 and their heterodimer formation were not affected. The NSP1² of VR2385 interferes with the interaction of STAT1 and importin. NSP1² may be the viral protein that interferes with the activation and signaling of type IFN. 1.37 (UMD) During studies of IFN signaling, we noticed 1 lab mutant induces synthesis of type I IFNs in cultured cells and has no effect on IFN downstream signaling. This mutant was plaque purified and named A2MC2. A2MC2-infected MARC cells resulted in strong inhibition of replication of an IFN-sensitive virus, Newcastle disease virus (NDV). Analysis of A2MC2-infected MARC cells showed that the transcripts of IFN-stimulated genes were elevated. Inhibition of A2MC2 replication abolished its capability to induce IFNs. A2MC2 does not affect IFN downstream signaling and induces IFN in PAMS. Infection of PAMs leads to little cytopathic effect. 1.38 (UIUC D Yoo) found that the suppression of NF-kB activation and sumoylation of PRRSV Nsp1± is mediated by protein inhibitor of activated STAT1 (PAIS1). We examined the role of PRRSV Nsp11, and endoribonuclease, on virus replication and IFN regulation. 1.39 (UIUC, Zuckermann) found that the influence of PRSV on the IFNa response of macrophages to infection with PRRSV is strain-dependent. Macrophages appeared non-responsive in the presence of either of 2 wild-type PRRSV, copious amounts of this cytokine was released by ZMAC cells exposed to a 3rd individual. This unconventional isolate was unique since it induced type I gene transcription and stimulated the phosphorylation of IRF3 to a greater extent than ZMAC cells infected with either conventional virus. Since the phosphorylation of a 2nd transcriptional factor, NFºß, also involved in the initiation of IFNa/ß gene transcription was comparatively unaffected in any of the virus-infected cells, the conventional viruses may be blocking IFNa production by interfering with a step(s) in the pathway leading to IRF3 activation. Objective 2. Understand the ecology and epidemiology of PRRSV and emerging viral diseases of swine. 2.1 (UGA) Studied ability of swine, human and avian influenza viruses to reassort on the TRIG backbone in swine and primary swine and human epithelial cells with the goal to elucidate the potential for reassortment. We also explored the evolution of AIVs in poultry and waterfowl. 2.2 (UGA) Explored the potential for avian and swine origin influenza viruses to infect and transmit in mice and ferrets. 2.3 (UGA) Explored receptor specificity of avian, human and swine influenza viruses. 2.4 (UGA) Explored the potential for avian influenza viruses to infect felines and tested for exposure of feral cats to AIVs to test their potential as an alternate reservoir/vector. 2.5 (UMN, Guelph, Hong Kong). PRRSV diversity based on sequencing/RFLP typing was described for type 2 PRRSV. 2.6 (UMN). Influenza Transmission dynamics and transmission parameters were evaluated in pigs with passive immunity induced by influenza vaccination. Homologous passive immunity decreased transmission but did not prevent it. Transmission was similar in pigs without passive immunity and in pigs with heterologous immunity. Influenza ecology studies showed that influenza infections in closed grow-finish populations were prolonged (70 DPI). Oral fluids were a sensitive method to detect influenza infections in populations. Weaned pigs are a source of virus introduction for grow-finish populations. Methods to study aerosol transmission of influenza virus in pigs were validated to detect influenza virus from aerosols generated from infected pigs. Detection of infectious aerosols may be related to the number of pigs shedding virus in a population. Positive aerosols could be detected when just a few pigs were known to be shedding. Detection of infectious aerosols has been shown in the field, inside pig barns and exhaust fans. 2.7 (UMN, ISU, SDSU) showed long-distance airborne spread of PRRSV and identified climactic conditions which support it. Multiple air filtration interventions were validated to prevent airborne spread of PRRSV and M hyopneumoniae. 2.8 (UMN, Pipestone) PRRSV shedding was decreased in pigs that had been vaccinated with a MLV vaccine compared to non-vaccinates. Decrease shedding was seen in oral fluids at 36 DPI and in air samples collected in exhaust fans. PRRSV was detected in air samples from non-vaccinated pigs for up to 70 days and 45 days for vaccinates. 2.9 (KSU, BARC, others) participated in the PHGC. Results include a genome wide association study of the first 600 pigs. The results show the ID of genomic markers that are linked to virus load and weight gain. To date, 2000 pigs are in the study. 2.10 (SHVRI) Viral RNA synthesis regulatory elements identification. The 5' untranslated region (UTR) of the genomic RNA is believed to be vital for the replication of PRRSV yet its functional mechanism remains largely unknown. Using a full-length cDNA clone we found SL1 was essential for infectivity of PRRSV. SL2 was a key regulatory structural element for PRRSV replication, particularly sg mRNA synthesis. 2.11 (ISU) Studies on the systematic review of factors that influence the persistence of influenza in environmental matrices. 2.12 (ISU) PCV2: Studies on commercially produced spray dried porcineplasma contains high levels of PCV2 DNA but did not transmit PCV2 when fed to naïve pigs. Establishment and maintenance of a PCV2-free breeding herd on a site that experienced a natural outbreak of PCV2-associated reproductive disease. Studies on high prevalence of PCV viremia in newborn piglets in 5 clinically normal swine breeding herds in North America. PRRSV influences infection dynamics of PCV2 subtypes PCV2a and PCV2b by prolonging PCV2 viremia and shedding. Shedding and infection dynamics of PCV2 after experimental infection and after natural exposure. 2.13 (ISU) Median infectious dose (ID50) of PRRSV isolate MN-184 via aerosol exposure. 2.14 (OSU) To potentiate the effect of killed PRRSV vaccine, poly(lactide-co-glycolide) (PLGA)- nanoparticles were prepared to encapsulate killed-PPRSV antigens. In nanoparticle-killed-PRRSV vaccinated pigs a reduction in viremia with complete viral clearance by day 15 was seen. In the lungs of nanoparticle- PRRSV vaccinated MN184 challenged pigs a significant reduction in PRRSV antigen load and a reduction in inflammatory cells infiltration was observed. Immunologically, increased frequency of immune cells which initiate Th1response associated with production of IFN±, IL12, IFN³ and IL6 cytokines was detected. Enhanced titers of PRRSV specific total and virus neutralizing antibodies were detected in nanoparticle-killed-PRRSV vaccinates. 2.15 (UW) developed novel analytical approaches to identify a small number of representative viral genotypes from among the diversity of viral sequences available in GenBank and PRRSVdb. We adapted techniques from network theory to rank PRRSV sequences in terms of their importance and applied these methods to a highly-curated PRRSV database that combines high-quality, non-recombinant sequences from both GenBank and PRRSVdb. Viruses represented by the top ranking sequences are valuable targets for future study and can be eventually incorporated into a polyvalent vaccine. Objective 3. Develop effective and efficient approaches for detection, prevention and control of PRRSV and emerging viral diseases of swine. 3.1 (NADC, Faaberg/Nicholson) Analyzed efficacy of using a diagnostic microarray to detect different PRRSV isolates. 3.2 (NADC, Lager/Miller; E. Zanella, Visiting Scientist) Validation of a real-time PCR for PRV. 3.3 (UGA) Studied prophylactic and therapeutic application of PRRSV-specific swine mAbs. 3.4 (UGA) Explored aerosol vaccination for SIV vaccines in mice and ferrets. 3.5 (UGA) Established primary normal swine bronchoepithelial cell cultures to measure innate responses to swine respiratory viruses. 3.6 (UGA) Tested a variety of PIV5-based live-attenuated vaccines against influenza virus. 3.7 (UGA) Developed new method for rapid detection of influenza virus: 3.8 (UGA) Developed assay for simultaneous serological detection of PRRSV and PCV2. 3.9 (UNL) previously identified immunodominant B-cell linear epitopes in the proteins of a type 2 PRRSV strain FL12 (infectious cDNA clone-derived PRRSV pathogenic strain) with the epitope number 201 (EP-201) of the M protein as a marker candidate (conserved epitope). A triple mutant (TM) carrying 3 AA substitutions in the epitope 201 of PRRSV FL12 was generated. The TM was no longer recognized by anti-201 mAb, was stable in vivo, did not elicit antibodies to peptide 201 and can be used as a DIVA marker vaccine strain (with current development of a companion ELISA). We are pursuing the development of DIVA vaccines using PRRSV mutants devoid of the only dispensable structural gene of PRRSV (ORF5a) and by developing mutants devoid of antigenic reactivity in conserved epitopes of structural genes. 3.10 (UMN, ISU) Development of methods for viral and serological monitoring of PCV2 in oral fluids using an experimental model. Prevalence of PCV2 in US finishing herds was characterized. MN participated with IA, IN, KA, NC, Canada and WV to compare PCV2 serological detection methods for diagnostics. 3.11 (UMN) PCR for detection of cytomegalovirus and lymphotrophic herpesvirus. 3.12 (UMN, OH) Novel vaccine development against PRRSV. 3.13 (UMN, ISU) Multiplex methods for serological detection of PCV2 and PRRSV. 3.14 (UMN, IA, SD, Guelph, Newport Labs) identified new PRRSV RFLP types in D-lab submissions. 3.15 (UMN) discovered a novel PRRSV protein, ORF5a, that is immunogenic and induces antibody responses in pigs. 3.16 (KSU, ISU, SDSU) Rowland, Zimmerman, Fang, Opriessnig developed a multiplex Luminex assay for the detection of antibodies against PRRSV, PCV2 and SIV and initiated a multi-lab validation of Luminex assays for serum and oral fluids. 3.17 (KSU Wyatt) characterized a T cell epitope in the PCV2 capsid protein. 3.18 (SHVRI) DIVA vaccine development used JX143 (parental virus HP-PRRSV). After serial psg., 88 extra AA deletions were found in the Nsp2 region and was genetically stable up to the 100th psg. (JXM100). An Nsp2-88aa epitope-based ELISA was developed. 9 pigs were divided into 3 groups and inoculated with parental JX143 (A), JXM100 (B) or were mock-infected (C). Grp. A were immunized with JXM100 virus from which antibodies could not be detected against the corresponding 88 AA deleted epitope until JX143 challenge. This demonstrated that the recombinant marker virus with the diagnostic test, enables serological differentiation between marker virus-infected and wild-type infected pigs. 3.19 (ISU, USDA, AASV, NPB) produced an instructional video and poster on collection and processing of swine oral fluids for disease monitoring and was distributed to AASV members by NPB. 3.20 (ISU) Multiple publications on PCV2 vaccines. 3.21 (ISU) Disinfection protocols reduce the amount of PCV2-contaminated livestock transport vehicles. 3.22 (ISU) Prolonged detection of PCV2 and anti-PCV2 antibody in oral fluids following experimental inoculation. 3.23 (ISU) Comparison of RNA extraction and PCR methods for the detection of PRRSV in oral fluid. 3.24 (ISU) Inhibition of PRRSV infection in piglets by a peptide-conjugated morpholino oligomer. 3.25 (ISU) Kinetics of UV254 inactivation of selected viral pathogens in a static system. 3.26 (ISU) Multiplex method for the simultaneous serological detection of PRRSV and PCV2. 3.27 (ISU) Terminology for classifying swine herds by PRRSV status. 3.28 (SDSU: Langenhorst, Lawson, Sun, Li, Hennings, Nelson, Fang) developed an FMIA for detection of PRRSV antibodies in oral fluid and serum. Recombinant nucleocapsid and nsp7 from genotypes 1 and 2 were used as antigens. 3.29 (SDSU: Li, Chen, Sun, Fang). NSP1b is a strong IFN antagonist. Site-specific mutations were introduced into nsp1b and these had reduced ability to inhibit IFNb activation. Recombinant viruses with these mutations were produced for vaccine development. 3.30 (BARC) A multiplex FMIA was developed to simultaneously quantify porcine cytokines in serum or oral fluids (eg. IL1b, IL6, IL8, IFNa, TNF-a, IL-10, IL-12, IFNg, IL4, CCL2). The assay will be of value in immunity, vaccine, challenge studies, determining genetic resistance to PRRSV and responses to other swine pathogens. 3.31 (UIUC, Laegreid; UNL, Osorio, Pattnaik) confirmed that N-glycan moieties in GP5 of type 2 PRRSV are important for the virus to escape neutralizing antibodies and that the N-glycan in GP3 is important in protecting the virus from antibody neutralization.

Impacts

  1. Evaluation of PRRSV strains in vivo. USDA-ARS. Examination of several PRRSV strains allow us to survey the viral growth properties, the disease in swine, the commensal bacteria that may arise during infection, the innate response, the adaptive immune response and the host gene expression patterns that differ between PRRSV strains. We use the gained knowledge to better understand PRRSV pathogenesis. With this knowledge, we can then develop better vaccines and vaccination strategies.
  2. Construction of a chimeric vaccine to protect against Asian HP-PRRSV. USDA-ARS. HP-PRRSV strains have proven to be a serious threat to our nations pork industry. Utilizing an infectious clone to a vaccine strain in combination with several structural genes from the Asian lineage, along with an identifiable foreign marker, we are constructing a vaccine to be used to protect swine in case such HP-PRRSV strains appear in the U.S.
  3. Gene expression in lymph nodes of PRRSV-infected pigs. USDA-ARS. The goal of this discovery project is to identify changes that occur in gene expression in porcine lung lymph nodes following PRRSV infection. Knowledge derived from this study will more clearly define the negative effect of PRRSV on the pig immune system, and it may be used to design better cross-protective vaccines.
  4. Progress in this reporting period continues to focus on non-PRRS viruses (e.g. influenza). While not directly related to PRRSV, this work is contributing directly to development of swine reagents and resources (e.g. primary swine epithelial cell lines). As we develop these tools for other (re-)emerging swine diseases, we plan to apply many of these approaches directly to PRRSV projects (UGA).
  5. In regard in influenza as an emerging (re-emerging) disease of swine, we have made extensive advances in understanding features of the virus and host that influence infection, tropism, and potentially reassortment. We have also explored a number of vaccine and anti-viral therapies for influenza and developed a novel approach for rapid and sensitive detection of influenza virus; all of which may directly impact swine and/or human health (UGA).
  6. Four PRRSV-related refereed papers involving our laboratories have been published in refereed journals during the period covered in this report (UNL).
  7. US and European Patent Title: Methods and Compositions for Vaccination of Animals with PRRSV Antigens with Improved Immunogenicity. Inventors: Ansari, I, Osorio FA, and Pattnaik, AK Serial No. 12/064, Issued: October 27, 2009. During 2011 this patent is being explored by a veterinary biologics company (UNL)
  8. . Provisional claim for invention: A reverse genetics method to develop a PRRSV attenuated live vaccine strain with DIVA differential capacity that would permit distinguishing naturally infected from non-infected, just vaccinated animals (Osorio, FA, Pattnaik, AK, Kwon, BJ, and Vu H.) filed on March 4, 2011, EFS ID: 9585692 Application No. 61449138 (UNL)
  9. Elucidation of neonatal infection with minimal impact of maternal immunity illuminated the critical need to control and eliminate early influenza virus infection (UMN).
  10. Commercial vaccines are not likely to benefit influenza control since homologous protection is required to prevent transmission.(UMN)
  11. PRRSv MLV vaccines and air filtration interventions can aid in regional elimination of PRRSv (UMN).
  12. New protein identification provides novel targets for immune protection (UMN).
  13. PCV2 pathogenesis characterization increases knowledge of key pathogenic features.
  14. Genomic markers for improved response to PRRS creates the opportunity to conduct marker-selected breeding of pigs (KSU).
  15. Identification of a decoy epitope in PCV2 capsid is being used for assays that assess protective immunity following infection or vaccination (KSU).
  16. Luminex is being developed as a substitute for standard ELISA approaches (KSU)
  17. PRRS is a more complicated swine disease in China. A lot of work for completely controlling PRRS needs to be done in China. Vaccination is the primary choice for the majority of pig products for preventing and controlling PRRS. The viral replication and transcription mechanism and related foundation research should be conducted and a new generation of DIVA vaccines should be developed (SHVRI)
  18. Shishan Yuan, Jian Lv, Xiangjian Li, Jianwu Zhang, Dandan Yu, Zhi Sun, Fei Gao, Zuzhang Wei, Jinshan Zhuang, Tao Tan, Haihong Zheng, Feifei Tan, Changlong Liu, Jiaqi Lu, Yanfang Cong, Xiaoming Wang, Hao Zheng. The construction of highly pathogenic PRRSV recombinant plasmid and genetic engineering vaccine. Patent NO. 200710172364.8; Announcement No.: CN101205539A; Publication No.: 2010062500313230
  19. Output for project: Assessment of Virulence of PRRSV Isolates Based Both on their Sensitivity to IFNb and Ability to Induce Type I IFN Responses. Mr. Christopher Overend a doctoral candidate in Pathobiology and Veterinary Science working on the project has successfully completed his degree. Data obtained in this project are routinely discussed with our collaborator Dr. Marvin Grubman, a scientist from Plum Island Animal Disease, Center who shares interests in the area of type I IFN (UCONN).
  20. Epidemiology/pathobiology/diagnostic studies provide ideas for detecting, preventing and eliminating viruses. Work has been done on genetic/antigenic variation during replication and persistence and new methods of surveillance for cost-effective methods of tracking infection and use in elimination/eradication. Advances in these areas linked with research in viral ecology/epidemiology and improvements in vaccines leads to possible elimination/eradication of viruses from farms and regions (ISU).
  21. Presence of replicating PRRSV in pigs from very early days post-infection modulates the innate immune function resulting in subversion of immunity. As this study was performed in pigs maintained in natural commercial environmental settings, the outcome of this study has more practical implications in development of new generation protective vaccines. Our results on nanoparticles-based PRRSV-killed vaccine suggested that mucosal immunization has the potential to induce protective immunity to PRRS (OSU).
  22. Understanding the role nsp2 in PRRSV virulence has important implication in developing better vaccines against PRRSV (VA).
  23. Understanding the PRRSV-host miRNAs interaction provides new insight into the role of miRNAs in PRRSV pathogenesis. (VA)
  24. A panel of monoclonal and polyclonal antibodies to non-structural proteins of PRRSV will be important tools in studies of PRRSV replication and pathogenesis. (SDSU)
  25. Recombinant PRRSV can be produced whereby Interleukin 1 B expression or modifications in nsp2 can enhance viral specific immunity. These are important modifications to further vaccine design. (SDSU)
  26. A multiplex FMIA for antibodies against PRRSV differentiates types I and II and can be used for multiplexing for serological profiling (SDSU).
  27. The PRRS Host Genetics Consortium (PHGC) is helping to dissect the role of host genetics in resistance to PRRS and in effects on pig health and related growth effects. Results using a Nursery Pig Model of commercial pigs infected with PRRSV and followed for 42 days have affirmed that all pigs become PRRSV infected but they clear virus from serum at different rates with variable weight effects. Genome wide association studies (GWAS) have mapped pig PRRS responses (viral load and weight gain during infection) to multiple swine chromosomes. This result suggests that genetic progress can be made by selective breeding.(BARC)
  28. Functional gene and protein transcriptomic analyses are underway with PHGC samples to identify gene networks and resistance associated biomarkers that differ in high versus low VL or WG PHGC pigs. Pathways have been identified and are being validated. Overall, the PHGC project will enable researchers to verify important genotypes and phenotypes that predict resistance/susceptibility to PRRSV infection.(BARC).
  29. An FMIA has been developed to simultaneously quantify multiple porcine cytokines in serum using Luminex xMap" technology. It has been optimized to detect innate (IL-1b, IL-6, IL-8, IFN-a, TNF-a); regulatory (IL-10), T helper 1 (Th1) (IL-12, IFN-g) and Th2 (IL-4) cytokines. The assay has been tested for levels of porcine cytokines in oral fluids with positive results. This assay will be a useful tool to determine cytokine involved in genetic resistance to PRRSV using PHGC samples (BARC).
  30. Antigenic/genetic variation in PRRSV is a major impediment to vaccine development. By distilling this diversity down to a manageable unit, we are hoping to provide guidance for the development of next-generation polyvalent vaccines that have maximum broad efficacy (BARC).
  31. Our studies on PRRSV inhibition of interferon signaling showed that NSP1² of virulent VR2385 may be the viral protein that interferes with IFN signaling, while NSP1² of Ingelvac MLV has no effect. This result has a biological relevance on PRRS vaccine design (UMD).
  32. Our finding of a lab mutant A2MC2 inducing interferons in cultured cells may be beneficial for vaccine development to induce protective immunity against PRRS. This isolate is expected to induce higher titer of neutralizing antibody in pigs (UMD).
  33. PRRSV expresses proteins that circumvent the type I IFN response and other cellular processes and to compensate the small coding capacity of PRRSV, these proteins are multifunctional. Studies for Dr. Yoos laboratory suggest that PRRSV Nsp1± and NSP11 are a multifunctional nuclear protein participating in the modulation of the host IFN system (UIUC).
  34. Studies from Dr. Zuckermanns lab suggest that PRRSV is inhibiting the ability of porcine macrophages to produce IFNa in response to infection by interfering with the activation of the transcription factor IRF-3 but not NFkB (UIUC).
  35. . The data presented by Dr. Laegreids lab firmly confirms the important notion that GP3 may be involved in inducing neutralizing antibodies. Collectively, our work aimed at deciphering the transcriptional and cytokine response of cells as we as porcine alveolar macrophages to PRRSV infection will likely lead to the development of strategies to developed better vaccine against this costly disease.

Publications

Abrahante JE, Martins K, Papas KK, Hering VJ, Schuurman H-J, Murtaugh MP. 2011. Microbiological safety of porcine islets: comparison with source pig. Xenotransplantation. 18:88-93. Baker SA, Mondaca E, Polson D and Dee SA. Evaluation of a needle-free injection device (AcuShot) for reduction of hematogenous transmission of PRRS virus. Swine Health Prod (accepted for publication). Baker SR, ONeil K, Gramer M and Dee SA. An estimate of seroprevalence of production-limiting diseases in wild swine. Vet Rec 2011;168:564 Beura LK, Dinh PX, Osorio FA, Pattnaik AK.(2011) Cellular Poly(C) Binding Proteins 1 and 2 Interact with Porcine Reproductive and Respiratory Syndrome Virus Non-Structural Protein 1{beta} and Support Viral Replication. J Virol. 2011 Oct 5. In press [Epub ahead of print] Binjawadagi B, V. Dwivedi, C. Manickam, J.B. Torrelles, and G.J. Renukaradhya (2011) Intranasal delivery of an adjuvanted modified live porcine reproductive and respiratory syndrome virus vaccine reduces the ROS production. Viral Immunol., In press. Bishop SC, Lunney JK, Pinard-van der Laan MH, Gay CG. 2011. Report from the Second International Symposium on Animal Genomics for Animal Health: Critical Needs, Challenges and Potential Solutions. Proc. Intnl. Symp. Animal Genomics for Animal Health (AGAH 2010). BMC Proceedings 5 Suppl 4:S1. Boddicker, N, EH Waide, RRR Rowland, JK Lunney, DJ Garrick1, JM Reecy, JCM Dekkers. Major QTL associated with host response to porcine reproductive and respiratory syndrome virus challenge. J Anim Sci. In Revision. Bradley, K.C., Jones, C.A., Tompkins, S.M., Tripp, R.A., Russell, R.J., Gramer, M.R., Heimburg-Molinaro, J., Smith, D.F., Cummings R.D., and D.A. Steinhauer. 2011 Comparison of the receptor binding properties of contemporary swine isolates and early human pandemic H1N1 isolates (novel 2009 H1N1). Virology. 413(2):169-82. PMID: 21353280 Brar MS, Shi M, Carman S, Ge L, Murtaugh MP, Leung FC-C. 2011. Porcine reproductive and respiratory syndrome virus in Ontario, Canada 1999-2010: diversity and restriction fragment length polymorphism. J Gen Virol. 92:1391-1397. PMID: 21346028. Calzada-Nova, G., Schnitzlein, W., Husmann, R., Zuckermann, F.A. 2011. North American porcine reproductive and respiratory viruses suppress the type I interferon response of activated porcine plasmacytoid dendritic cells. J. Virol. 85:2703-13. Chittick WA, Stensland WR, Prickett JR, Strait EL, Harmon K, Yoon K-J, Wang C, Zimmerman JJ. 2011. Comparison of RNA extraction and RT-PCR methods for the detection of porcine reproductive and respiratory syndrome virus (PRRSV) in porcine oral fluid specimens. J Vet Diagn Invest 23:248-253.C Cino-Ozuna, AG, S Henry, R Hesse, JC Nietfeld, J Bai, HM Scott, RRR Rowland.2011. Characterization of a new disease syndrome associated with porcine circovirus type 2 (PCV2) in previously vaccinated herds. J Clin Micro. In press. Corzo C, Mondaca E, Wayne S, Torremorell M, Dee S, Davies P, Morrison B (2010). Control and elimination of porcine respiratory syndrome virus. Virus Res, 154:185-192, doi:10.1016/j.virusres.2010.08.016 Cutler T, Wang C, Qin Q, Zhou F, Warren K, Yoon K-J, Hoff SJ, Zimmerman J. 2011. Kinetics of UV254 inactivation of selected viral pathogens in a static system. J Appl Microbiol 111:389-395. Cutler TD, Wang C, Hoff SJ, Kittawornrat A, Zimmerman JJ. 2011. Median infectious dose (ID50) of porcine reproductive and respiratory syndrome virus isolate MN-184 via aerosol exposure. Vet Microbiol 151:229-237. Cutler TD, Zimmerman JJ. 2011. Ultraviolet irradiation and the mechanisms underlying its inactivation of infectious agents. Anim Health Res Rev 12:15-23. Das PB, Vu HL, Dinh PX, Cooney JL, Kwon B, Osorio FA, Pattnaik AK.(2011)Glycosylation of minor envelope glycoproteins of porcine reproductive and respiratory syndrome virus in infectious virus recovery, receptor interaction, and immune response. Virology. 2011 Feb 20; 410(2):385-94. Epub 2010 Dec 30. Debin Tian, Haihong Zheng, Rong Zhang, Jinshan Zhuang, Shishan Yuan. 2011. Chimeric Porcine Reproductive and Respiratory Syndrome Viruses reveal full function of genotype 1 envelope proteins in the backbone of genotype 2. Virology. YVIRO-06057; No. of page: 8; 4C: 4. Dee S, Spronk G, Reicks D, Ruen P and Deen J. Further assessment of air filtration for preventing PRRSV infection in large breeding herds. Vet Rec 2010;167:976-977. Dee SA, Otake S and Deen J. An evaluation of ultraviolet light (UV 254) as a means to inactivate porcine reproductive and respiratory syndrome virus on common farm surfaces and materials. Vet Microbiol 2011;150:96-99 Dee SA, Otake S and Deen J. Use of a production region model to assess the efficacy of various air filtration systems for preventing the airborne transmission of porcine reproductive and respiratory syndrome virus and Mycoplasma hyopneumoniae. Results of a 2-year study. Virus Res 2010. 154:177-184. Dee SA, Otake S, Pitkin A and Deen J. A 4-year summary of air filtration system efficacy for preventing airborne spread of porcine reproductive and respiratory syndrome virus and Mycoplasma hyopneumoniae. Swine Health Prod 2011;19:292-294. Dlugolenski, D., Jones, L., Saavedra, G., Tompkins. S.M., Tripp R.A., and E. Mundt. 2011 Passage of low-pathogenic avian influenza (LPAI) viruses mediates rapid genetic adaptation of a wild-bird isolate in poultry. Arch Virol. 156(4):565-76. PMID: 21197555 Driskell, E.A., Jones, C.A., Berghaus, R.D., Stallknecht, D.E., Howerth, E.W., and S.M. Tompkins. 2011 Domestic cats are susceptible to infection with low pathogenic avian influenza viruses from shorebirds. In press, Veterinary Pathology. Driskell, E.A., Jones, C.A., Stallknecht, D.E., Howerth, E.W., and S.M. Tompkins. 2010 Avian influenza isolates from wild birds replicate and cause disease in a mouse model of infection. Virology. 399(2):280-9. PMID: 20123144 Driskell, J.D., Jones, C.A., Tompkins, S.M., and R.A. Tripp. 2011 One-Step Assay for Detecting Influenza Using Dynamic Light Scattering and Gold Nanoparticles. Analyst 136(15):3083-90. PMID: 21666913 Cover Article. Du, Y., D. Yoo, M. A. Paradis, and G.Scherba. 2011. Antiviral activity of tilmicosin for type 1 and 2 porcine reproductive and respiratory syndrome virus in cultured porcine alveolar macrophages. J. Antivirals Antiretrovirals 3:28-33. Dwivedi V, C. Manickam, R. Patterson, K. Dodson, and G.J. Renukaradhya (2011). 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, 29(23):4067-76. Dwivedi V, Manickam C, Patterson R, Dodson K, Murtaugh M, Torrelles JB, Schlesinger LS, Renukaradhya GJ. 2011. Cross-protective immunity to porcine reproductive and respiratory syndrome virus by intranasal delivery of a live virus vaccine with a potent adjuvant. Vaccine 29:4058-4066.PMID: 21419162 Faaberg, K. S., Kehrli, Jr., M. E., Lager, K. M., Guo, B., and J. Han. 2010. In vivo growth of porcine reproductive and respiratory syndrome virus engineered nsp2 deletion mutants. Virus Res. 154:77-85. Fei Gao, Jiaqi Lu, Huochun Yao, Zuzhang Wei, Qian Yang, Shishan Yuan, Cis- Acting Structural Element in 5 UTR Is Essential for Replication of Porcine Reproductive and Respiratory Syndrome Virus. Virus Research, [J] 2011. 10.1016/j.virusres.2011.08.018. Feifei Tan, Zuzhang Wei, Yanhua Li, Rong Zhang, Jinshan Zhuang, Zhi Sun, Shishan Yuan. 2011. Identification of non-essential regions in nucleocapsid protein of Porcine and Respiratory Syndrome Virus for replication in cell culture. Virus Research. Volume 158, issues 1-2, June 2011, page 62-71. Gauger PC, Faaberg KS, Guo B, Kappes MA, Opriessnig T. 2011. Genetic and phenotypic characterization of a 2006 United States porcine reproductive and respiratory virus isolate associated with high morbidity and mortality in the field. Virus Res [Epub ahead of print] Gauger PC, Lager KM, Vincent AL, Opriessnig T, Cheung AK, Butler JE, Kehrli ME, Jr (2011) Leukogram abnormalities in gnotobiotic pigs infected with porcine circovirus type 2. Veterinary Microbiology 154:185-190 Gauger PC, Lager KM, Vincent AL, Opriessnig T, Kehrli ME, Jr, Cheung AK (2011) Postweaning multisystemic wasting syndrome produced in gnotobiotic pigs following exposure to various amounts of porcine circovirus type 2a or type 2b. Veterinary Microbiology 153:229-239 Gauger PC, Vincent AL, Loving CL, Lager KM, Janke BH, Kehrli Jr. M.E, Roth JA. 2011. Enhanced pneumonia and disease in pigs vaccinated with an inactivated human-like (´-cluster) H1N2 vaccine and challenged with pandemic 2009 H1N1 influenza virus. Vaccine 24:2712-2719. Gordy, J.T., Jones,C.A., Rue, J., Crawford, P.C., Levy, J.K., Stallknecht, D.E., Tripp, R.A., and S.M. Tompkins. 2011 Surveillance of Feral Cats for Influenza A Virus. In Press, Influenza and Other Respiratory Pathogens. Gorres JP, Lager KM, Kong W-P, Royals M, Todd JP, Vincent AL, Wei C-J, Loving CL, Zanella EL, Janke B, Marcus E. Kehrli J, Nabel GJ, Rao SS (2011) DNA Vaccination Elicits Protective Immune Responses against Pandemic and Classic Swine Influenza Viruses in Pigs. Clinical and Vaccine Immunology 18:1987-1995 Guo, B., Vorwald, A.C., Alt, D.P., Lager, K.M., Bayles, D.O. and Faaberg, K.S. 2011. Large scale parallel pyrosequencing technology: PRRSV strain VR-2332 nsp2 deletion mutant stability in swine. Virus Res. 161:162-169 Haley C, Wagner B, Puvanendiran S, Abrahante J, Murtaugh MP. 2011. Diagnostic performance measures of ELISA and quantitative PCR tests for porcine circovirus type 2 exposure using Bayesian latent class analysis. Prev Vet Med. 101:79-88. PMID: 21632130 Hamir AN, Greenlee JJ, Stanton T, Smith JD, Doucette S, Kunkle RA, Stasko JA, Richt JA, Kehrli ME, Jr (2011) Experimental inoculation of raccoons (Procyon lotor) with Spiroplasma mirum and transmissible mink encephalopathy (TME) Canadian Journal of Veterinary Research 75:18-24 Hamir AN, Kehrli ME, Jr, Kunkle RA, Greenlee JJ, Nicholson EM, Richt JA, Miller JM, Cutlip RC (2011) Experimental interspecies transmission studies of the transmissible spongiform encephalopathies to cattle: comparison to bovine spongiform encephalopathy in cattle. J Vet Diagn Invest 23:407-420 He D, C. Overend, J. Ambrogio, R.J. Maganti, M.J Grubman, A.E Garmendia 2011. Marked differences between MARC-145 cells and swine alveolar macrophages in IFN²-induced activation of antiviral state against PRRSV. Vet Immunol Immunopathol 139(1):57-60. Hiep, L. X. Vu., Byungjoon, K., Kyoung-Jin Y., LAegreid, W., Pattnaik, A.K., Osorio, F.A. 2011. Immune Evasion of Porcine Reproductive and Respiratory Syndrome Virus through Glycan Shielding Involves both Glycoprotein 5 as Well as Glycoprotein 3. J. Virol. 85:5555-64. Holtkamp DJ, Polson DD, Torremorell M (2011). Terminology for classifying swine herds by porcine reproductive and respiratory syndrome virus status. J Swine Health and Prod 19(1):44-56. Holtkamp DJ, Polson DD, Torremorell M (2011). Terminology for classifying swine herds by porcine reproductive and respiratory syndrome virus status. Tierärztliche Praxis Großtiere 39 2: 101-112 Huang YW, Meng XJ. Novel strategies and approaches to develop the next generation of vaccines against porcine reproductive and respiratory syndrome virus (PRRSV). Virus Research. 2010;154(1-2):141-9. Irwin CK, Yoon K-J, Wang C, Hoff SJ, Zimmerman JJ, Denagamage T, O'Connor AM. 2011. Using the systematic review methodology to evaluate factors that influence the persistence of influenza in environmental matrices. Appl Environ Microbiol 77:1049-1060. Jiaqi Lu, Fei Gao, Zuzhang Wei, Ping Liu, Changlong Liu, Haihong Zheng, Yanhua Li, Tao Lin, Shishan Yuan. 2011. A 5-Proximal Stem-loop Structure of 5 Untranslated Region of Porcine Reproductive and Respiratory Syndrome Virus Genome Is Key for Virus Replication. Virology Journal 2011, 8: 172, doi: 10. 1186/ 1743-422X-8-172. Johnson CR, Griggs TF, Gnanandarajah J, Murtaugh MP. 2011. Novel structural protein in Porcine reproductive and respiratory syndrome virus encoded in an alternative open reading frame 5 present in all arteriviruses. J Gen Virol 92:1107-1116. PMID: 21307222 Kappes MA, Sandbulte MR, Platt R, Wang C, Lager KM, Henningson JN, Lorusso A, Vincent AL, Loving CL, Roth JA, Kehrli ME, Jr. (2011) Vaccination with NS1-truncated H3N2 swine influenza virus primes T cells and confers cross-protection against an H1N1 heterosubtypic challenge in pigs. Vaccine (e-pub ahead of print 07Nov2011) Kittawornrat A, Prickett J, Wang C, Panyasing Y, Ballagi A, Rice A, Main R, Johnson J, Rademacher C, Hoogland M, Rowland R, Zimmerman J. Detection of porcine reproductive and respiratory syndrome virus (PRRSV) antibodies in oral fluid specimens using a commercial PRRSV serum antibody ELISA. J Vet Diagn Invest (in press). Langenhorst Robert J., Steven Lawson, Apisit Kittawornrat, Jeffrey J. Zimmerman, Zhi Sun, Yanhua Li, Jane Christopher-Hennings, Eric A. Nelson, Ying Fang. Development of a fluorescent microsphere immunoassay for detection of antibodies against PRRSV using oral fluid samples as an alternative to serum-based assays. Journal of Clinical Vaccine and Immunology. Lawson, Steve, Yanhua Li, John Patton, Robert J. Langenhorst, Zhi Sun, Zhiyong Jiang, Jane Christopher-Hennings, Eric A. Nelson, David Knudsen, Ying Fang, Kyeong-Ok Chang. Interleukin-1² expression by a recombinant porcine reproductive and respiratory syndrome virus. Virus Research, in preparation. Lin K, Wang C, Murtaugh MP, Ramamoorthy S. 2011. Multiplex method for simultaneous serological detection of porcine reproductive and respiratory syndrome virus and porcine circovirus type 2. J Clin Micro 49:3184-3190. Linhares D, Rovira A, Torremorell M (2011). Evaluation of FTA cards for collection and transport of samples for PRRS virus diagnostics. Accepted for publication Linhares D, Torremorell M, Cano JP and Dee SA. Effect of modified live porcine reproductive and respiratory syndrome (PRRS) vaccine on the shedding of wild-type virus from an endemically infected population of growing pigs. Vaccine (accepted for publication). Lorusso A, Vincent AL, Harland ML, Alt D, Bayles DO, Swenson SL, Gramer MR, Russell CA, Smith DJ, Lager KM, Lewis NS (2011) Genetic and antigenic characterization of H1 influenza viruses from United States swine from 2008. The Journal of general virology 92:919-930 Lunney JK, DA Benfield DA, RR Rowland. 2010. Porcine reproductive and respiratory syndrome virus: an update on an emerging and re-emerging viral disease of swine. Virus Res. 154:1-6. Lunney JK, Rowland RRR. 2011. Understanding Genetic Disease Resistance. National Hog Farmer. Blueprint Immunology 101. Apr. 15, 2011. p.30-42. Lunney JK, Steibel JP, Reecy J, Rothschild M, Kerrigan M, Trible B, Rowland RRR. 2011. Probing genetic control of swine responses to PRRSV infection: Current Progress of the PRRS Host Genetics Consortium. Proceedings of the International Symposium on Animal Genomics for Animal Health (AGAH 2010). BMC Proceedings. 5 Suppl 4:S30. Nfon CK, Berhane Y, Hisanaga T, Zhang S, Handel K, Kehler H, Labrecque O, Lewis NS, Vincent AL, Copps J, Alexandersen S, Pasick J (2011) Characterization of H1N1 swine influenza viruses circulating in Canadian pigs in 2009. Journal of virology 85:8667-8679 Ni YY, Huang YW, Cao D, Opriessnig T, Meng XJ. Establishment of a DNA-launched infectious clone for a highly pneumovirulent strain of type 2 porcine reproductive and respiratory syndrome virus: identification and in vitro and in vivo characterization of a large spontaneous deletion in the nsp2 region. Virus Research. 2011;160 (1-2):264-73. Nicholson, T. L., D. Kukielka, A. L. Vincent, S. L. Brockmeier, Miller, L. C. and K. S. Faaberg. 2011. Utility of a Panviral Microarray for Detection of Swine Respiratory Viruses in Clinical Samples. J. Clin. Microbiol. 49:1542-1548. ONeill K, Shen HG, Lin K, Hemann M, Beach NM, Meng XJ, Halbur PG, Opriessnig T. 2011. Studies on PCV2 vaccination of 5-day-old piglets. Clin Vaccine Immunol [Epub ahead of print] Opriessnig T, Gomes-Neto J, Hemann M, Shen HG, Huang YW, Halbur PG, Meng XJ. 2011. An experimental live chimeric PCV1-2a vaccine is efficacious at decreasing PCV2b viremia when administered intramuscularly or orally in a PCV2b and PRRSV dual-challenge model. Microbiol Immunol [Epub ahead of print] Opriessnig T, Madson DM, Roof M, Layton S, Ramamoorthy S, Meng XJ, Halbur PG. 2011. Experimental reproduction of porcine circovirus type 2 (PCV2)-associated enteritis in pigs infected with PCV2 alone or concurrently with Lawsonia intracellularis or Salmonella typhimurium. J Comp Pathol (in press). Opriessnig T, Madson DM, Schalk S, Brockmeier S, Shen HG, Beach NM, Meng XJ, Baker, RB, Zanella EL, Halbur PG. 2011. Porcine circovirus type 2 (PCV2) vaccination is effective in reducing disease and PCV2 shedding in semen of boars concurrently infected with PCV2 and Mycoplasma hyopneumoniae. Theriogenology 76:351-360. Opriessnig T, Patel D, Halbur PG, Meng XJ, Stein DA, Zhang YJ. 2011. Inhibition of porcine reproductive and respiratory syndrome virus infection in piglets by a peptide-conjugated morpholino oligomer. Antiviral Res 91:36-42. Opriessnig T, Patterson AR, Madson DM, Pal N, Ramamoorthy S, Meng XJ, Halbur PG: 2010. Comparison of the effectiveness of passive (dam) versus active (piglet) immunization against porcine circovirus type 2 (PCV2) and impact of passively-derived PCV2 vaccine-induced immunity on vaccination. Vet Microbiol 142:177-183. Opriessnig T, Shen H-G, Pal N, Ramamoorthy S, Huang Y-W, Lager KM, Halbur PG, Meng X-J. 2011. A live-attenuated chimeric vaccine based on the new porcine circovirus subtype 2b (PCV2b) is safe and efficacious in a triple challenge co-infection model with PCV2b, porcine reproductive and respiratory syndrome virus (PRRSV), and porcine parvovirus. (in press). Opriessnig T, Shen HG, Pal N, Ramamoorthy S, Huang YW, Lager KM, Beach NM, Halbur PG, Meng XJ. 2011. A live-attenuated chimeric porcine circovirus Type 2 (PCV2) vaccine is transmitted to contact pigs but is not upregulated by concurrent infection with porcine parvovirus (PPV) and porcine reproductive and respiratory syndrome virus (PRRSV) and is efficacious in a PCV2b-PRRSV-PPV challenge model. Clin Vaccine Immunol 18:1261-1268. Opriessnig, T., Shen, H.G., Pal, N., Ramamoorthy, S., Huang, Y.W., Lager, K.M., Beach, N.M., Halbur, P.G., and Meng, X.J., A live-attenuated chimeric PCV2 vaccine is transmitted to contact pigs but is not upregulated by concurrent infection with PPV and PRRSV and is efficacious in a PCV2a-PRRSV-PPV challenge model. Clinical and Vaccine Immunology. 29(15):P.2712-9 Opriessnig, T., Shen, H.G., Pal, N., Ramamoorthy, S., Huang, Y.W., Lager, K.M., Beach, N.M., Halbur, P.G., and Meng, X.J. 2011. A Live-Attenuated Chimeric Porcine Circovirus Type 2 (PCV2) Vaccine Is Transmitted to Contact Pigs but Is Not Upregulated by Concurrent Infection with Porcine Parvovirus (PPV) and Porcine Reproductive and Respiratory Syndrome Virus (PRRSV) and Is Efficacious in a PCV2b-PRRSV-PPV Challenge Model. Clin Vaccine Immunol 18, 1261-1268. PMID: 21653745 Oshansky, C.M., Pickens, J.A., Bradley, K.C., Jones, L.P., Saavedra-Ebner, G.M., Barber, J.P., Crabtree, J.M., Steinhauer, D.A., Tompkins, S.M., and R.A. Tripp. 2011 Avian influenza viruses infect primary human bronchial epithelial cells unconstrained by sialic acid ±2, 3 residues. PLoS ONE, 6(6):e21183. PMID: 21731666 Otake S, Dee S, Corzo C, Oliveira S and Deen J. Long distance airborne transport of viable PRRSV and Mycoplasma hyopneumoniae from a swine population infected with multiple viral variants. Vet Microbiol 2010; 145: 198-208. Patterson AR, Baker RB, Madson DM, Pintar AL, Opriessnig T. 2011. Disinfection protocols reduce the amount of porcine circovirus type 2 contaminated livestock transport vehicles. J Swine Health Prod 19:156-164. Patterson AR, Johnson JK, Ramamoorthy S, Hesse RA, Murtaugh MP, Puvanendiran S, Pogranichniy RM, Erickson GA, Carman S, Hause B, Meng XJ, Opriessnig T. 2011. Interlaboratory comparison of Porcine circovirus-2 indirect immunofluorescent antibody test and enzyme-linked immunosorbent assay results on experimentally infected pigs. J Vet Diagn Invest. 23:206-212.. Patterson AR, Madson DM, Schalk SD, Halbur PG, Opriessnig T. 2011. Establishment and maintenance of a porcine circovirus type 2 (PCV2)-free breeding herd on a site that experienced a natural outbreak of PCV2-associated reproductive disease. J Swine Health Prod 19:165-174. Patterson AR, Ramamoorthy S, Madson DM, Meng XJ, Halbur PG, Opriessnig T. 2011. Shedding and infection dynamics of porcine circovirus type 2 (PCV2) after experimental infection. Vet Microbiol 149:91-98. Pena L, Vincent AL, Ye J, Ciacci-Zanella JR, Angel M, Lorusso A, Gauger PC, Janke BH, Loving CL, Perez DR (2011) Modifications in the polymerase genes of a swine-like triple-reassortant influenza virus to generate live attenuated vaccines against 2009 pandemic H1N1 viruses. Journal of virology 85:456-469 Pitkin A, Otake S and Dee SA. A one-night downtime period prevents the spread of PRRSV and Mycoplasma hyopneumoniae by personnel and fomites (boots and coveralls)". Swine Health Prod (accepted for publication). Platt R, Vincent AL, Gauger PC, Loving CL, Zanella EL, Lager KM, Kehrli ME, Jr., Kimura K, Roth JA (2011) Comparison of humoral and cellular immune responses to inactivated swine influenza virus vaccine in weaned pigs. Veterinary Immunology and Immunopathology 142:252-257 Prickett JR, Johnson J, Murtaugh MP, Puvanendiran S, Wang C, Zimmerman JJ, Opriessnig T. 2011. Prolonged detection of PCV2 and anti-PCV2 antibody in oral fluids following experimental inoculation. Transbound Emerg Dis. 58:121-127. PMID: 21223532 Puvanendiran S, Stone S, Yu W, Johnson CR, Abrahante J, Garcia Jimenez L, Griggs T, Haley C, Wagner B, Murtaugh MP. 2011. High prevalence of porcine circovirus type 2 exposure and infection in swine finisher herds. Virus Res. 157:92-98. PMID:21352865 Ramirez A, Wang C, Prickett JR, Pogranichniy R, Yoon K-J, Main R, Rademacher C, Hoogland M, Hoffmann P, Johnson JK, Kurtz A, Kurtz E, Zimmerman J. 2011. Efficient surveillance of pig populations using oral fluids. Prev Vet Med (in press). Romagosa A, Gramer M, Joo HS, Torremorell M (2011). Sensitivity of oral fluids for detecting influenza A virus in populations of vaccinated and non-vaccinated pigs. J of influenza and other respiratory viruses. DOI:10.1111/j.1750-2659.2011.00276.x Rowland, RRR. 2010. The interaction between PRRSV and the late gestation pig fetus . Virus Res., 154:114-122. Shen H, Wang C, Madson DM, Opriessnig T. 2010. High prevalence of porcine circovirus viremia in newborn piglets in five clinically normal swine breeding herds in North America. Prev Vet Med 97:228236. Shen HG, Beach NM, Huang YW, Halbur PG, Meng XJ, Opriessnig T. 2011. Comparison of commercial and experimental porcine circovirus type 2 (PCV2) vaccines using a triple challenge with PCV2, porcine reproductive and respiratory syndrome virus (PRRSV), and porcine parvovirus (PPV). Vaccine 28:5960-5966. Shen HG, Schalk S, Halbur PG, Campbell J, Russell L, Opriessnig T. 2011. Commercially produced spray dried porcine plasma contains high levels of PCV2 DNA but did not transmit PCV2 when fed to naïve pigs. J Anim Science 89:1930-1938. Shi, M., T. T. Lam, C. C. Hon, R. K. Hui, K. S. Faaberg, T. Wennblom, M. P. Murtaugh, T. Stadejek, and F. C. Leung. 2010. Molecular epidemiology of PRRSV: A phylogenetic perspective. Virus Res. 154:7-17. Sinha A, Schalk S, Lager K M, Wang C, Opriessnig T. 2011. Singular PCV2a or PCV2b infection results in apoptosis of hepatocytes 3 in clinically affected gnotobiotic pigs. Res Vet Sci DOI:10.1016/j.rvsc.2010.10.013 Sinha A, Shen HG, Schalk S, Beach NM, Huang YW, Halbur PG, Meng XJ, Opriessnig T. 2010. Porcine reproductive and respiratory syndrome virus (PRRSV) infection at the time of porcine circovirus type 2 (PCV2) vaccination has no impact on vaccine efficacy. Clin Vaccine Immunol 17:19040-1945. Sinha A, Shen HG, Schalk S, Beach NM, Huang YW, Meng XJ, Halbur PG, Opriessnig T. 2011. Porcine reproductive and respiratory syndrome virus (PRRSV) influences infection dynamics of porcine circovirus type 2 (PCV2) subtypes PCV2a and PCV2b by prolonging PCV2 viremia and shedding. Vet Microbiol 152:235-246. Smith, J.H., Brooks, P., Johnson, S., Tompkins, S.M., Custer, K.M., Haas, D.L., Mair, R., Papania, M., and R.A. Tripp. 2010 Aerosol Vaccination Induces Robust Protective Immunity to Homologous and Heterologous Influenza Infection in Mice. Vaccine 29(14): 2568-75. PMID: 21300100 Smith, J.H., Nagy, T., Driskell, E.A., Brooks, P., Tompkins, S.M., and R.A. Tripp. 2011 Comparative Pathology in Ferrets Infected with H1N1 Influenza A Viruses Isolated from Different Hosts. J Virol. 85(15):7572-81. PMID: 21593156 Smith, J.H., Papania, M., Knaus, D., Brooks, P., Hass, D.L., Mair, R., Barry, J., Tompkins, S.M., and R.A. Tripp. Nebulized Live-Attenuated Influenza Vaccine Provides Protection in Ferrets at a Reduced Dose. In Press, Vaccine. Spronk G, Otake S and Dee S. Prevention of PRRSV infection in large breeding herds using air filtration. Vet Rec 2010; 166:758-759. Subramaniam S, Sur JH, Kwon B, Pattnaik AK, Osorio FA.(2011) A virulent strain of porcine reproductive and respiratory syndrome virus does not up-regulate interleukin-10 levels in vitro or in vivo.Virus Res. 2011 Feb;155(2):415-22. Epub 2010 Dec 17. Tiawsirisup S, Blitvich BJ, Tucker BJ, Halbur PG, Bartholomay LC, Rowley WA, Platt KB. 2010. Susceptibility of fox squirrels (Sciurus niger) to West Nile virus by oral exposure. Vector Borne Zoonotic Dis 10:207-209. Trible, BR, M Kerrigan, N Crossland, M Potter, K Faaberg, R Hesse, RRR Rowland. Porcine circovirus type 2 capsid protein epitopes associated with vaccination, infection and disease. Clin Vacc Immunol. In press. Trible, Kerrigan, M., Faaberg, K. S., and R. R.R. Rowland. 2011. Identification of an immunodominant region the PCV2 capsid protein recognized by naturally infected and vaccinated pigs. Clinical and Vaccine Immunology, 18:749-757. Vincent, A. L., K. M. Lager, K. S. Faaberg, M. Harland, E. L. Zanella, J. R. Ciacci-Zanella, M. E. Kehrli, Jr., B. H. Janke, and A. Klimov. 2010. Experimental inoculation of pigs with pandemic H1N1 2009 virus and HI cross-reactivity with contemporary swine influenza virus antisera. Influenza Other Respiratory Viruses. 4:53-60. Vu HL, Kwon B, Yoon KJ, Laegreid WW, Pattnaik AK, Osorio FA. (2011) Immune evasion of porcine reproductive and respiratory syndrome virus through glycan shielding involves both glycoprotein 5 as well as glycoprotein 3. J Virol. 2011 Jun;85(11):5555-64. Epub 2011 Mar 16. Wagner J, Kneucker A, Liebler-Tenorio E, Fachinger V, Glaser M, Pesch S, Murtaugh MP, Reinhold P. 2011. Respiratory function and pulmonary lesions in pigs infected with porcine reproductive and respiratory syndrome virus. Vet. J. 187:310-319. PMID: 20089425 Zhang H, Lunney JK, Baker RB, Opriessnig T. 2011. Cytokine and chemokine mRNA expression profiles in tracheobronchial lymph nodes from pigs singularly infected or coinfected with porcine circovirus type 2 (PCV2) and Mycoplasma hyopneumoniae (MHYO). Vet Immunol Immunopathol 140:152-158. Zhang H, Mohn U, Prickett JR, Schalk S, Motz M, Halbur PG, Feagins AR, Meng X-J, Opriessnig T. 2011. Different capabilities of enzyme immunoassays to detect anti-hepatitis E virus immunoglobin G in sera from pigs infected experimentally with hepatitis E genotype 3 or 4 and in sera from field pigs with unknown status of HEV infection. J Virol Methods (in press).
Log Out ?

Are you sure you want to log out?

Press No if you want to continue work. Press Yes to logout current user.

Report a Bug
Report a Bug

Describe your bug clearly, including the steps you used to create it.