SAES-422 Multistate Research Activity Accomplishments Report

Status: Approved

Basic Information

Participants

<b>Chair:</b> Lunney, Joan K. ( joan.lunney@ars.usda.gov ) USDA, ARS, BARC, APDL; <b>Secretary:</b> Meng, X.J. ( xjmeng@vt.edu )Virginia Polytechnic Institute and State University (VA Tech); Rowland, Raymond R.R. (browland@vet.k-state.edu) Kansas State University (KSU); Tompkins, S. Mark (smt@uga.edu) University of Georgia (UGA); Enjuanes, Luis (L.Enjuanes@cnb.csic.es) Centro Nacional de Biotecnología (CNB), CSIC; Zimmerman, Jeff (jjzimm@iastate.edu) Iowa State University (ISU); Schommer, Susan (schommers@missouri.edu ) University of Missouri (UMO); Christopher-Hennings, Jane (jane.hennings@sdstate.edu) SDSU; Goldberg, Tony (tgoldberg@vetmed.wisc.edu ) University of Wisconsin-Madison; Zuckermann, Federico A. (fazaaa@uiuc.edu) University of Illinois at Urbana-Champaign (UIUC); Faaberg, Kay (kay.faaberg@ars.usda.gov) NADC; Gourapura, Renukaradhya J.(gourapura.1@osu.edu ) The Ohio State University (OSU); Murtaugh, Michael P (murta001@umn.edu ) University of Minnesota (UMN); Osorio, Fernando A. (fosorio@unl.edu) University of Nebraska-Lincoln (UNL); Zhang, Yanjin (zhangyj@umd.edu ) University of Maryland (UMD); Pogranichniy, Roman (rmp@purdue.edu) Purdue (IN); Guillermo R. Risatti (guillermo.risatti@uconn.edu ) University of Connecticut (UCONN); Benfield, David (benfield.2@osu.edu) Ohio State University (OSU); Johnson, Peter (pjohnson@reeusda.gov )USDA, CSREES; <b>Other NC229 Scientists;</b> Garmendia, Antonio - UConn; Tripp, Ralph - UGA; Baker, RB - ISU; Halbur, Patrick - ISU; Holtkamp, Derald J - ISU; Harris, DL (Hank) - ISU; Johnson, John K - ISU; Karriker, Locke - ISU; Main, Rodger G - ISU; McKean, JD - ISU; Opriessnig, Tanja - ISU; Platt, Kenneth - ISU; Strait, Erin - ISU; Ramamoorthy, Sheila - ISU; Ramirez, Alejandro - ISU; Roth, JA - ISU; Yoon, Kyoung-Jin - ISU; Yoo, Dongwan - UIUC; Laegried, Will - UIUC; Wyatt, Carol - KSU; Hesse, Dick - KSU; Sang, Yongming - K-State; Chang, KC - KSU; Blecha, Frank - KSU; Zhu, Xiaoping -UMD; Davies, Peter - UMN; Dee, Scott - UMN; Deen, John - UMN; Joo, Han Soo - UMN; Molitor, Tom - UMN; Morrison, Robert - UMN; Rossow, Kurt - UMN; Rovira, Albert - UMN; Kerhli, Marcus Jr. - NADC; Lager, Kelly - NADC; Brockmeier, Susan - NADC; Miller, Laura - NADC; Loving, Crystal - NADC; Neill, John - NADC; John Butler - University of Iowa; Saif, Linda J - OSU; Fang, Ying - SDSU; Wang, Xiuqing - SDSU; Nelson, Eric - SDSU; Wysocki, Michal - BARC; Chen, Hongbo - BARC; Smith, Doug - Univ. MI; Ho, Sam - Univ. MI; Steibel, JP; Michigan State Univ. (MSU); Ernst, Cathy - Michigan State Univ. (MSU); LeRoith, Tanya - VA Tech; Roberts, P.C. - VA Tech; Elankumaran, S. - VA Tech; Mullarky, I.K. - VA Tech; Pattnaik, Asit - UNL; Ciobanu, Daniel C. - UNL; Johnson, Rodger - UNL;

NC229 Meeting Chicago, IL Friday, 12/04/2009 - 12/04/2009. 1. Meeting opened by Chair Joan Lunney; Welcome everyone. 2. Brief remarks by David Benfield, NC229 advisor. 3. Nominations of new officers and voting by station reps: Jane Christopher-Hennings (SDSU) was nominated and approved by station reps as the new NC229 Secretary, and X.J. Meng (Virginia Tech) rotated into the new NC229 chair. 4. Discussion of progress and plans by objective. The NC229 attendees split into groups according to the 3 new objectives, for informal discussions led by the respective objective coordinators and co-leaders. Objective 1: Elucidate the mechanisms of host-pathogen(s) interactions is led by Mike Murtaugh (co-leaders: Roman Pogranichniy, Ying Fang, Aradhya Gourapura) Objective 2: Understand the ecology and epidemiology of PRRSV and emerging viral diseases of swine is led by Scott Dee (co-leaders: Fred Leung, Jeff Zimmerman) Objective 3: Develop effective and efficient approaches for detection, prevention and control of PRRSV and emerging viral diseases of swine is led by Mark Tompkins (co-leaders: Luis Enjuanes, Antonio Garmendia). 5. Reports from each Objective team: Mike Murtaugh summarized and provided a brief on objective 1 (Elucidate the mechanisms of host-pathogen(s) interactions); Jeff Zimmerman summarized the discussion and progress on objective 2 (Understand the ecology and epidemiology of PRRSV and emerging viral diseases of swine); Mark Tompkins summarized the discussion and progress reports on objective 3 (Develop effective and efficient approaches for detection, prevention and control of PRRSV and emerging viral diseases of swine). Plans for sharing samples and protocols were presented by each group.; 6. Joan Lunney, closing remarks. The NC229 plans its next meeting for Friday December 3, 2010, prior to next year's PRRS Symposium. The leadership of NC229 was turned over to its new Chair X.J. Meng.

Accomplishments

Objective 1. Elucidate the mechanisms of host-pathogen(s) interactions. 1.1. Investigators (Risatti&339;s group) at UCONN studied the transcriptional activation in macrophages upon PRRSV infections. A sustained transcriptional activation was observed for interleukin-1± (IL-1±), IL-6, TNF-±, IFN-², IRF-7, PKR and Mx1, consistent with activation of an antiviral state within PAMs upon infection with virulent FL12v. At 24 hour post-infection (hpi), which corresponds to the peak logarithmic phase of FL12v infectious virus assembly and release, transcription of IL-1±, IL-10, IL-15, IRF-7, VCAM, and Mx-1 genes was significantly different in PAMs infected with cP5U.NSP12 or cPNSP3.8 relative to PAMs infected with FL12v. Live attenuated vaccine (LAV) SP infection induced a pattern of transcriptional activation at 24 hpi similar to that of FL12v. IL-1±, IL-1², IL-10, IL-15, TNF-±, MCP-2, IRF-7, and Mx1 mRNAs accumulation was significantly different in cells infected with the recombinant viruses or LAV SP relative to cells infected with FL12v. The data on induction obtained thus far, shows that PRRSV isolates do induce IFNb in PAM but at variable levels. 1.2. ISU scientists (Zimmerman) conducted studies that led to new estimates of PRRSV persistency for up to 175 days post-inoculation. They provided new information on virus evolution indicating that persistent PRRSV infection does not depend on mutations in ORFs 1b, 5, or 7. The ISU group also showed that among a variety of antibody assays and ELISPOT, the SVN antibody response was the best predictor of both level and duration of viremia. Antibody responses (IDEXX ELISA, N ELISA, and M 3' ELISA) predicted prior exposure to PRRSV, but provided little information regarding the ontogeny of the protective immune response. ELISPOT was a poor prognosticator of PRRSV infection status. 1.3. The UIUC station scientists (Zuckermann) analyzed the expression of CD163 on PAMs and macrophages derived from CD14 positive blood monocytes (MDMs), in correlation with PRRSV replication. By flow cytometric analysis, they showed that the levels of CD163 expression correlated well with the overall level of PRRSV replication. They further examined the effects of modulators of macrophage function, including 12-O-tetradecanoylphorbol-13-acetate (TPA), lipopolysaccharide (LPS), and interleukin (IL)-10 on the expression of CD163 and PRRSV replication. Pre-treatment of PAMs or MDMs with TPA or LPS resulted in decreased expression of CD163 and reduction in PRRSV replication. On the contrary, the incubation of CD14 positive monocytes with IL-10 during differentiation into MDMs resulted in up-regulated expression of CD163 with a corresponding increase in PRRSV infection. By utilizing a yeast two-hybrid screening, they identified that the inhibitor of MyoD family-a (I-mfa) domain-containing protein (HIC) is a cellular partner for PRRS virus (PRRSV) N protein. 1.4. Purdue scientists (Pogranichniy, IN) designed stealth RNAi antisense from specific PRRSV cellular receptor CD163 and co-receptor Siglec-1 (sialoadhesin, Sn, CD169) and 5-UTR region of viral genome and demonstrated significant inhibition of PRRSV infection and spread in MARC-145 cell culture line. PRRSV infection in MARC-145 cultured cell in the absence of endogenous Sn expression resulted in enhanced cellular expression of PTEN which is indicative of negative regulation of the Akt pathway leading to cell arrest and apoptosis. However, in the presence of endogenous Sn, the activation of the Akt pathway was demonstrated by the up regulation of tissue metalloproteinase -9 (MMP-9) mRNA leading to cell growth and survival. 1.5. Scientists at KSU (Sang, Blecha and Rowland) performed a study which identified 39 type I IFN genes. Recombinant IFN proteins expressed from these genes including some novel IFNs, showed a wide range of activities in controlling PRRSV replication. Some are quite effective while others have no activity against PRRSV infections. In addition, Hesse and Rowland are performing an analysis of cross-protection between diverse PRRSV strains. 1.6. Scientists at UMD (Zhang) continued their efforts on developing anti-PRRSV peptide-conjugated phosphorodiamidate morpholino oligomers (PPMOs). Compared with mock-infected controls, the expression of CXCL10, IFN-², and CCL2 in PRRSV-infected PAMs were 2157-, 1740-, and 108-fold higher, respectively. The expression of double-stranded RNA-activated protein kinase R (PKR), interferon regulatory factor 1 (IRF-1), signal transducers and activators of transcription 1 (STAT-1), tumor necrosis factor-± (TNF-±), toll-like receptor 3 (TLR-3), and nuclear factor-kB p65 (NF-kB) in infected PAM s were 7.5-, 6.4-, 6.4-, 8.3-, 2.8-, and 1.7-fold higher, respectively. 1.7. Scientists at the UMN (Murtaugh) demonstrated that pig age influences the pathogenesis of PRRSV infection. Clinical signs were markedly more severe and prolonged in young piglets than in finishers or sows. Viremia was prolonged in weaned pigs for both attenuated and virulent PRRSV. Viremia was reduced in magnitude and duration in finisher pigs and sows. Many sows did not show evidence of viremia following infection with attenuated PRRSV. They also showed that, based upon whole genome sequencing on field isolates, recombination occurs in the field. 1.8. UMN scientists also showed that IFN ³ secreting peripheral blood mononuclear cells were more abundant in sows but not specifically increased by PRRSV infection in any age group, and IL-10 levels in blood were not correlated with PRRSV infection status. These findings show that animal age, perhaps due to increased innate immune resistance, strongly influences the outcome of acute PRRSV infection, whereas an antibody response is triggered at a low threshold of infection that is independent of age. Prolonged infection was not due to IL-10-mediated immunosuppression, and PRRSV did not elicit a specific IFN ³ response, especially in non-adult animals. Equivalent antibody responses were elicited in response to virulent and attenuated viruses, indicating that the antigenic mass necessary for an immune response is produced at a low level of infection, and is not predicted by viremic status. Thus, viral replication was occurring in lung or lymphoid tissues even though viremia was not always observed. 1.9. OSU scientists (Gourapura, Saif) studied natural killer (NK) cell-mediated innate immune cytoxicity in PRRSV and porcine respiratory coronavirus (PRCV) infected pigs. The PRRSV/PRCV dual virus-infected pigs had significantly suppressed innate immune responses, as evidenced by reduced IFN-± level in lung and blood. In addition, they identified a significant reduction in systemic NK cell-mediated cytotoxicity in PRRSV alone infected pigs. Further, upon co-infection with PRCV, there was a synergistic suppression of NK cell-mediated cytotoxicity. Co-infection by PRRSV and PRCV led to enhanced PRRSV replication in lung and a trend towards increased serum Th1 (IFN³ and IL-12), but decreased Th2 (IL-4) cytokine responses, thus clinically exacerbating PRRSV pneumonia. These findings imply that a prior innate immune suppression by immunomodulating respiratory viruses (like that induced by PRRSV) may be a contributing factor to more severe pneumonia due to PRCV infection. 1.10. Scientists at SDSU (Fang) showed that the nonstructural protein 2 (nsp2) of PRRSV has a role in viral replication and may modulate host immunity. Each of the six identified immunodominant nsp2 B-cell epitopes (ES2 through ES7) was deleted from a Type I PRRSV cDNA infectious clone. Deletion of ES3, ES4, or ES7 allowed the generation of viable virus. The ?ES3 mutant showed increased cytolytic activity and more vigorous growth kinetics, while ?ES4 and ?ES7 mutants displayed decreased cytolytic activity and slower growth kinetics in vitro. In a nursery pig model, ?ES4 and ?ES7 mutants exhibited attenuated phenotypes and the ?ES3 mutant produced higher peak viral loads. IL-1² and TNF-± expression levels were down-regulated in cells stimulated (or infected) with the ?ES3 mutant. 1.11. At SDSU, scientists determined that the PRRSV nsp1 protein can antagonize beta interferon (IFN-²) responses. In PRRSV infected cells, we detected the presence of nsp1± and nsp1² and the cleavage sites between nsp1±/nsp1² and nsp1²/nsp2 were identified. Both nsp1± and nsp1² dramatically inhibited IFN-² expression and nsp1² inhibited nuclear translocation of STAT1 in the JAK-STAT signaling pathway. These results demonstrated that nsp1² inhibits both interferon synthesis and signaling, while nsp1± alone strongly inhibits the synthesis of interferon. 1.12. Scientists at USDA-BARC (Lunney) developed PRRS Host Genetics Consortium (PHGC) 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 NPB, USDA, universities and private companies; it represents the first-of-its-kind approach to food animal infectious disease research. The project uses a Nursery Pig Model to assess pig resistance/ susceptibility to primary PRRSV infection. Crossbred pigs were infected with PRRSV and followed for 42 days post infection (dpi). Blood samples were collected at 0,4,7,10,14,21,28,35 and 42 dpi and weekly weights recorded. DNA from all PHGC pigs has been prepared and is being genotyped with the PorcineSNP60 Genotyping BeadChip (containing over 60K single nucleotide polymorphisms or SNPs). Results from the first 5 trials of 200 pigs each have affirmed that all pigs become PRRSV infected; some pigs clear virus from serum quicker and weight effects are variable. 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 PRRS susceptible/reduced growth pigs. Overall, the PHGC project will enable researchers to verify important genotypes and phenotypes that predict resistance/susceptibility to PRRSV infection. 1.13. BARC scientists also identified host gene expression changes that are involved in regulating responses to PRRSV infection and vaccination. With samples collected McCaw at NC State Univ. BARC scientists are testing the effect of PRRSV infection or vaccination on pigs using RNA prepared from tracheobronchial lymph nodes (TBLN), the cranial and distal part of the lung, and tonsils. Pigs were either infected with Minnesota (MNW2B) or NC Powell strains of PRRSV or vaccinated with ATP or were non-treated controls. Mucosal tissue samples were collected from pigs between 3 and 6 days post treatment so that the early innate immune response could be evaluated. RNAs were prepared and hybridization to the swine long oligo array [www.pigoligoarray.org; Steibel et al. 2009] could be assessed. Analyses are underway at BARC with statistical assessment of gene expression patterns performed in collaboration with MSU scientists. Tests of the effect of samples collected after homologous or heterologous PRRS vaccination and challenge are also underway. 1.14. Scientists at VA Tech (Meng) identified and characterized a porcine monocytic cell line supporting PRRSV replication and progeny virion production by using an improved DNA-launched PRRSV reverse genetics system. We developed an improved DNA-launched (plasmid DNA transfection-based) reverse genetics system with reduced cost and labor for PRRSV by introduction of ribozyme elements at both termini of the viral genomic cDNA that were placed under the control of a eukaryotic hybrid promoter. The rescue efficacy of PRRSV with this system was approximately 10-50-fold higher than the in vitro-transcribed RNA-based system and the traditional DNA-launched system without the engineered ribozyme elements, as determined by reporter GFP level in transfected cells and the peak titer of the recovery virus. By using this new reverse genetics system, they identified and characterized a porcine monocytic cell line, 3D4/31, capable of supporting PRRSV replication, progeny virion production, and attachment on the cell surface. 1.15. The VA Tech group (Meng) described the molecular cloning, gene structure, tissue distribution and PRRSV binding characteristics of Porcine DC-SIGN. They cloned and characterized the cDNA and gene encoding porcine DC-SIGN (pDC-SIGN). The full-length pDC-SIGN cDNA encodes a type II transmembrane protein of 240 amino acids. Phylogenetic analysis revealed that pDC-SIGN, together with bovine, canis and equine DC-SIGN, are more closely related to mouse SIGNR7 and SIGNR8 than to human DC-SIGN. pDC-SIGN has the same gene structure as bovine, canis DC-SIGN and mouse SIGNR8 with eight exons. pDC-SIGN mRNA expression was detected in pig spleen, thymus, lymph node, lung, bone marrow and muscles. pDC-SIGN protein was found to express on the surface of monocyte-derived macrophages and dendritic cells, alveolar macrophages, lymph node sinusoidal macrophage-like, dendritic-like and endothelial cells but not of monocytes, peripheral blood lymphocytes or lymph node lymphocytes. A BHK cell line stably expressing pDC-SIGN binds to human ICAM-3 and ICAM-2 immunoadhesins in a calcium-dependent manner, and enhances the transmission of PRRSV to target cells in trans. 1.16. Scientists at UNL (Osorio, Pattnaik) studied the effects on innate and acquired immune responses. The group identified certain pathogenic mechanisms (such as decoy epitope deploying or glycan shielding) that would suggest that PRRSV employs diverse strategies to subvert and/or evade the hosts immune response, thus securing an abundant and unrestricted viral replication during the acute phase of infection and a long persistence in the host. They recently confirmed this weak IFN induction phenotype exhibited by PRRSV in monocyte-derived swine macrophages. They have recently screened all non-structural proteins (NSPs) of PRRSV identifying at least four (NSP1, NSP2, NSP4 and NSP11) having inhibitory activity towards IFN production. Of these, the strongest inhibitor of IFN production is NSP1², affecting primordially dsRNA signaling pathways. 1.17. UNL station scientists (Osorio, Pattnaik) also studied the role of viral genes in virulence and determinants. They initiated reverse genetics experiments to determine the molecular basis of attenuation of virulence in PRRSV. They have used the infectious clone for the development of chimeras between PRRSV strains of different degrees of virulence, and shown that a NSP-coding area of the PRRSV genome is a major cluster of virulence. Also ORF5 and ORF2 contain structural determinants of virulence. Likewise, site-specific mutagenesis of GP5 (product of ORF5) indicated that PRRSV evades the pigs immune system by means of a glycan-shielding mechanism. They have also studied epitopes in different PRRSV proteins that could be deleted or modified to be used as serologic differential markers. Objective 2. Understand the ecology and epidemiology of PRRSV and emerging viral diseases of swine. 2.1. ISU scientists (Zimmerman) performed studies that led to new estimates of R0 for PRRSV in different operations. They performed survival analysis of PRRSV transmission within herds and provided an overview of PRRSV routes of transmission and patterns of circulation. In addition, they continued work on PRRSV aerosols transmission, including methods of inactivating airborne viruses, and their research shows that PRRSV is difficult to transmit to susceptible pigs via consumption of meat from PRRSV-infected animals. 2.2. Scientists (Rowland and Hesse) at KSU in collaboration with Lunney (BARC) and others participated in the PRRS Host Genetics Consortium. The results for the year include the infection and sample collection of 400 pigs at K-State. The results reveal the appearance of stratified subpopulations which possessed wide variations in weight, virus load and growth performance. 2.3. At the UMN, diagnostic tests for PCV2 and differential PCR for subtypes 2a and 2b were refined, and ultrastructural studies were conducted to detect coinfecting agents in clinical samples. The tools were applied to epidemiologic studies of PCV2, including the ecology of PCV2 in boar studs, and associations of PCV2 subtypes with clinical disease. 2.4. Scientists at NADC continued to develop and provide materials and reagents to investigators. (Miller) Porcine modified Identitag (16bp tags) annotated database has been created with Dr Greg Harhay, USDA, ARS, USMARC, 2009; (Miller) MARC-145 cells were provided to Dr. Moiz Kitabwalla, Lipid Sciences; Dr S. Mark Tompkins, UGA; (Faaberg)- MARC-145 cells provided to Dr. Margo Brinton, Georgia State University; MARC-145, MA-104 and CL2621 cells provided to Dr. Jens Kuhn, Integrated Research Facility Frederick; (d) Faaberg group - Infectious clone pVR-V7 provided to Dr. Frederick Leung, Hong Kong University; Dr. Sergey Parinov, The National University of Singapore; Dr. István Kiss, National Veterinary Institute. 2.5. NADC scientists (Faaberg)- showed that nsp2 has been shown to be immunogenic, contains hypervariable segments, encodes a protease responsible for replicase cleavage and harbors B-cell epitopes. They studied the nature of the PL2 protease, when nsp2 was individually expressed in CHO cells and not associated with virus. They found that the PL2 cysteine protease domain possesses both trans- and cis-cleavage activities, and cleaved only at or near the G|G at nsp2 amino acids 1196|1197|1198. They also analyzed nsp2 when expressed from the viral genome in MARC-145 cells, and found that nsp2 was now found as at least 6 isomers, all containing the N-termini, but differing in size. They also discovered that heat shock 70kDa protein 5 (HSPA5) was bound to nsp2. 2.6. Scientists at NADC (Faaberg) also conducted in vivo study of two new isolates along with strains MN184 and SDSU-73. Serum and lavage samples are now being analyzed by virus isolation, qRT-PCR, IFN-gamma, ELISA, and lyphadenopathy. Also, previous results suggested that one section of strain VR-2332 nsp2, when deleted, resulted in virus that did not cause lymphadenopathy when infected into young swine. The NADC scientists are now examining that polypeptide when expressed in adenovirus. The viruses have been made, but no in vivo studies have been done. 2.7. Research at UWI (Goldberg) has begun to characterize genetic and antigenic diversity within PRRS virus, in an effort to identify a small number of representative viral genotypes for further testing, and that can be eventually incorporated into a polyvalent vaccine. Because of the large number of PRRSV sequences available (over 10,000 in the literature), the computational aspect of this work is challenging. The project has just begun, and data are currently being compiled and edited. Objective 3. Develop effective and efficient approaches for detection, prevention and control of PRRSV and emerging viral diseases of swine. 3.1. Investigators at the UGA (Tompkins, Tripp) intended to develop novel vaccine strategies for prevention of PRRSV, however all PRRSV efforts were set aside because of the influenza pandemic caused by H1N1 virus. The novel pandH1N1 virus was first identified in April 2009. UGA received the virus (A/CA/04/09) and initiated pathogenesis studies in swine and ferrets. The ferret serves as the primary model of human influenza virus infection. It is susceptible to infection with human influenza viruses without adaptation, sheds virus, and presents similar symptoms. Remarkably, the pandH1N1 replicated to very high titers in ferrets (>1e8 TCID50/ml of nasal wash), even when inoculated with very low 100 TCID50) doses of virus but failed to demonstrate significant clinical disease. Virus primarily replicated in the upper respiratory tract, but in some cases was detected in the lungs of infected ferrets. A/CA/04/09 infected pigs and caused clinical symptoms including loss of appetite, coughing, nasal discharge, and reduced activity. 3.2. ISU scientists (Zimmermann) conducted extensive studies on the use of oral fluids for the detection of PRRSV, anti-PRRSV antibody, and other pathogens - a new highly cost-effective approach for surveillance PRRSV and other pathogens in commercial settings. 3.3. Scientists at UIUC (Zuckermann) recently developed a porcine alveolar macrophage cell line. This cell line, named ZMAC, was found to efficiently support the replication of a number PRRS virus isolates, often achieving high titers (>107 TCID50/ml). Given the apparent high permissiveness of ZMAC cells to PRRS virus, they tested the proficiency of this cell line to isolate field PRRS virus from clinical samples. The ZMAC line proved highly efficient (>90%) at isolating PRRS virus within 72 hours after exposing ZMAC cells to pig serum samples known to be positive to PRRSV by real-time PCR, from which attempts to isolate PRRS virus in MARC-145, and even primary alveolar macrophages, had failed. The ZMAC cell line may prove useful for PRRSV vaccine development. 3.4. Scientists at UIUC engineered the viral genome to transcribe an additional subgenomic RNA initiating between non-structural and structural genes. Two unique restriction sites and a copy of the transcription regulatory sequence for ORF6 (TRS6) were inserted between ORFs 1b and 2a, yielding a general purpose expression vector. The enhanced green fluorescent protein (GFP) gene was cloned between the unique sites such that the inserted gene was transcribed from TRS2 which was located upstream within ORF1b, while the copy of TRS6 drives ORF2a/b transcription. Cells infected with P129-GFP produce virus progeny and showed fluorescence and the inserted gene was phenotypically stable for at least 37 serial in vitro passages. Subsequently, a capsid protein gene was cloned from PCV2 and inserted into the PRRSV infectious clone vector, generating virus "P129-PCV". Pigs immunized with either P129-GFP or P129-PCV2 produced antibodies specific for GFP or PCV2 capsid respectively. 3.5. Scientists at KSU (Rowland, Hesse) along with several others are incorporating Luminex for the detection of IgM and IgG antibodies to PRRSV and other pathogens. The results provide the opportunity to develop assays for the purpose of profiling multiple agents within a herd. 3.6. UMD scientists continued to develop anti-PRRSV PPMOs. PPMOs are single-stranded DNA analogs containing a modified backbone and cell-penetrating peptide. They examined PPMO-mediated inhibition of PRRSV replication in PAMs and found that (a) PAMs uptake PPMO efficiently. The uptake efficiency of PAMs of 72-hour pre-incubation was higher than PAMs of 24-h pre-incubation; (b) Treatment of PAMs with PPMO 5UP2 resulted in protection from PRRSV-induced cell death for at least seven days, and produced no elevation in the activity of the caspase 3, 7, 8 and 9; (c) 5UP2 treatment of PRRSV-infected PAMs also prevented the vigorous induction of interferon-² and chemokines observed in infected and mock-treated PAMs 3.7. Scientists at the UMN (Dee) completed the year 3 of the evaluation of air filtration as a means to reduce the risk of aerosol transmission of PRRSV at the SDEC production region model. A standardized means to validate the efficacy of biosecurity interventions to reduce the risk of airborne spread of PRRSV was developed. Assessment of air filtration to reduce the risk of airborne spread of PRRSV in large sow units in swine dense regions was initiated. Efficacy of regional control of PRRS was evaluated in Stevens County, Minnesota. Communication and implementation of best practices has reduced PRRS incidence to one infected herd in 89 existing sites. 3.8. Scientists at the OSU (Gourapura) evaluated the mucosal immune responses in the respiratory tract of pigs using innate immune cell specific agents as candidate adjuvants administered by the intranasal (IN) route. They initially determined the adjuvanticity of nine different bacterial preparations belongs to Mycobacterium tuberculosis, Streptoccocus pyogenes, and Vibrio cholera, administered intranasally (IN) to pigs. Based on the general mucosal immune responses elicited by the individual candidate adjuvants, we chose M. tuberculosis whole cell lysate (M. tb WCL) for further studies. Analyses are underway to explore the extended adjuvanticity of M. tb WCL, used with PRRSV modified live virus vaccine administered IN to PRRSV sero-negative pigs. Also they will perform challenge studies in mucosally immunized pigs using homologous (VR2332) and heterologous (MN184) PRRSV strains. 3.9. At SDSU station (Christopher-Hennings), a long-term objective is to provide a PRRSV-free semen supply for artificial insemination so mechanical and anti-viral methods were evaluated to reduce risk of transmission. A unilayer density gradient centrifugation method to purify PRRSV contaminated semen allowed for some risk reduction by eliminating PRRSV from 71% of semen samples tested. The antiviral chymostatin inhibited PRRSV infection in-vitro. However, further testing is needed to determine an effective animal dose and evaluate effects on sperm quality parameters. 3.10. Scientists at SDSU (Christopher-Hennings, Lawson) developed a multiplex assay to simultaneously quantify 9 porcine cytokines in serum using Luminex xMap" technology, and the assay was 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 will be of value in vaccine and challenge studies as well as for determining genetic resistance to PRRSV and immune responses to other swine pathogens. 3.11. Scientists at CSIC in Spain (Enjuanes) conducted animal experiments using the TGEV vector expressing PRRSV GP5 and M proteins, it was found that all animals present a high antibody response against TGEV, therefore, the vector infected target tissues as expected. Also, vaccinated animals showed a clear antibody response against the PRRSV antigens (i.e., GP5 and M proteins). After a challenge with a virulent PRRSV isolate, a fast recall response was observed, as vaccinated animals induced higher antibody titers against PRRSV antigens and earlier than control animals. Nevertheless, the immune response was not strong enough to provide full protection against PRRSV. That was likely due to the low levels of neutralizing antibodies produced before challenge. 3.12. Scientists at CSIC in Spain (Enjuanes) generated a set of rTGEV vectors expressing M protein and GP5 mutants with a modified glycosylation pattern. Just one of them was stable, a rTGEV expressing PRRSV M protein and a GP5 N46S mutant, lacking the glycosylation site overlapping neutralizing epitope. This rTGEV vector expressed high levels of GP5 and M PRRSV proteins in 74 % and 85% of the infected cells, respectively. A short in vivo immunization protocol was performed. One-week old piglets were inoculated with 1x108 pfu of the rTGEV by three routes: oral, nasal and intragastric. All animals present a high antibody response against TGEV, therefore, the vector infected target tissues as expected. Vaccinated animals showed a clear humoral response against PRRSV. A killed vaccine was also developed based on rTGEV expressing GP5-N46S mutant and M proteins. The protection conferred by this vaccine was tested. Vaccinated animals induced higher and faster antibody titers against PRRSV antigens than control animals. Neutralizing antibodies titers were also higher in the vaccinated animals when compared with non-vaccinated ones, suggesting that the elimination of glycosylation site close to the neutralizing epitope improves protective immune response against PRRSV. The presence of an immunodominant (decoy) epitope close to the neutralizing epitope in GP5 could be deleterious for a strong neutralizing immune response. Therefore, an rTGEV vector was constructed, expressing a GP5 protein lacking the non-neutralizing (decoy) epitope and the N46 glycosylation site. The virus was recovered with high titer and GP5 and M protein expression was stable in 65% and 90% of the infected cells, respectively.

Impacts

  1. Research advances over the last year continue to expand our understanding of PRRSV epidemiology, pathobiology, virology, and provide new ideas for countering and/or eliminating the infection. Extensive work has been done regarding the emergence of genetic and antigenic variation during replication in pigs and its role in persistence. Continued assessment and research in diagnostic technology contributes to the improvement and refinement of our ability to detect and diagnose PRRSV infection. On-going work on new methods of surveillance promise to provide new, cost-effective methods of tracking infection and implementing area elimination/eradication programs. Accomplishments in these areas linked with research in viral ecology/epidemiology, will lead to the development of tools that will result in the eventual elimination and eradication of PRRSV from individual farms and regions.
  2. The development of the porcine alveolar macrophage cell line ZMAC to isolate PRRS virus from field samples will aid better understand PRRSV as well as vaccine development.
  3. The use of PRRS virus as vector for foreign gene expression is a first, which demonstrates the potential use of PRRSV as a vaccine vector for swine pathogens. The studies on the modulation of CD163 receptor expression and the replication of PRRSV in porcine macrophages data indicated that the expression of CD163 on macrophages in different microenvironments, in vivo, may determine the replication efficiency and subsequent pathogenicity of PRRS virus.
  4. The interaction of PRRSV capsid with the cellular transcription factor implicates a possible regulation of host cell gene expression by the N protein during PRRSV infection
  5. Genetic analysis of host response has revealed the diverse negative impacts of PRRSV on a population. Decreased performance demonstrated by lack of weight gain is a loss to the producers bottom line. New IFN genes that possess potent anti-PRRSV can be incorporated into vaccines and other antiviral therapies.
  6. The development of Luminex system provides the means to 1) detect antibodies to multiple agents in a single small volume of sample, 2) increase sensitivity and specificity, 4) reduce the cost of testing, and 5) semi-quantitative output without need for serial dilution of a sample to an endpoint, and 5) test for agents in non-serum samples, such as oral fluids and meat juice.
  7. The studies of PPMOs against PRRSV have demonstrated that PPMOs inhibited PRRSV replication and protected the cells from PRRSV-induced cell death. Administration of PPMO 5UP2 to piglets that were experimentally infected with a PRRSV strain resulted in lower viremia and less lung lesion. Specific antiviral PPMOs can complement other approaches for PRRS prevention and control, because there are highly conserved target sequences among PRRSV strains. Application of the antiviral PPMOs will yield significant economic benefits to the swine industry, especially for breeding farms.
  8. The value of airborne transmission research results to stakeholders includes a comprehensive understanding of the airborne routes and significance for the spread of PRRSV between farms. The continued ability to demonstrate the efficacy of air filtration to reduce this risk, initially via the production region model and then under controlled field conditions provides producers and veterinarians with a tool to reduce this important risk factor.
  9. Regional elimination of PRRS demonstrates to stakeholders that the disease can be eliminated from a region and provides tools and methods that can be implemented in other regions.
  10. Immunity research informed stakeholders of significant age-dependent differences in the ability of pigs to resist PRRSV infection, providing important information on proper application of live vaccines in the field, and efficacy of serum inoculation in the control of PRRS disease in gestating sows and vertical transmission of PRRSV.
  11. The identification and potential use of bacterial preparations as candidate adjuvants to augment anti-PRRSV mucosal immune responses in pigs will advance in the area of mucosal vaccine production and in understanding innate immune responses to PRRSV.
  12. A multiplex assay to simultaneously quantify 9 porcine cytokines in serum using Luminex xMap" technology was developed and 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 will be of value in vaccine and challenge studies as well as for determining genetic resistance to PRRSV and immune responses to other swine pathogens.
  13. The demonstration that nsp1² inhibits both interferon synthesis and signaling, while nsp1± alone strongly inhibits the synthesis of interferon provides important insights into the mechanisms of how nsp1 contributes to PRRSV pathogenesis and how this may impact future vaccine development strategies.
  14. The PRRS Host Genetics Consortium (PHGC) has begun to determine the role of host genetics in resistance to PRRS and in effects on pig health and related growth effects. Using a Nursery Pig Model crossbred pigs from high health farms were infected with PRRSV and followed for 42 days. Results from the first 5 trials of 200 pigs each have affirmed that all pigs become PRRSV infected but pigs clear virus from serum at different rates; weight effects are variable. Overall, the PHGC project will enable researchers to verify important genotypes and phenotypes that predict resistance/susceptibility to PRRSV infection.
  15. The establishment of an improved reverse genetic system and the identification of a porcine monocytic cell line supporting PRRSV replication will aid future studies of host-virus interaction of PRRSV. The identification and characterization porcine DC-SIGN and demonstration of its binding to PRRSV will help better understand the biological role(s) of DC-SIGN family in innate immunity during the evolutionary process.

Publications

Ando A, Uenishi H, Kawata H, Tanaka M, Shigenari A, Flori L, Chardon P, Lunney JK, Kulski JK, Inoko H. 2008. Microsatellite diversity and crossover regions within homozygous and heterozygous SLA haplotypes of different pig breeds. Immunogenetics. 60: 399-407. Beura, L. K., Sarkar, S. N., Kwon, B. J., Subramaniam, S., Jones, C., Pattnaik, A. K., and Osorio, F. A. (2010). Porcine Reproductive and Respiratory Syndrome Virus nonstructural protein nsp1b modulates host immune response by antagonizing IRF3 activation. J. Virology In press. Boyd P, Hudgens E, Loftus JP, Tompkins D, Wysocki M, Kakach L, LaBresh J, Baldwin CL, Lunney JK. 2009. Expressed gene sequence and bioactivity of the IFN³-response chemokine CXCL11 of swine and cattle. Vet. Immunol. Immunopathol. Submitted. Brockmeier, S. L., K. M. Lager, M. J. Grubman, D. E. Brough, D. Ettyreddy, R. E. Sacco, P. C. Gauger, C. L. Loving, A. C. Vorwald, M. E. Kehrli, Jr., and H. D. Lehmkuhl. 2009. Adenovirus-mediated expression of interferon-alpha delays viral replication and reduces disease signs in swine challenged with porcine reproductive and respiratory syndrome virus. Viral Immunol. 22:173-180. Brown, E., S. Lawson, C. Welbon, M. P. Murtaugh, E. A. Nelson, J. J. Zimmerman, R. R. R. Rowland, Y. Fang. 2009. Antibody response of nonstructural proteins: implication for diagnostic detection and differentiation of Type I and Type II porcine reproductive and respiratory syndrome virus. Clinical and Vaccine Immunology. 16(5):628-35. Butler, J.E., P. Weber, N. Wertz and K.M. Lager. 2008. Porcine reproductive and respiratory syndrome virus (PRRSV) subverts development of adaptive immunity by proliferation of germline-encoded B cells with hydrophobic HCDR3s. J. Immunol. 180: 2347-2356. Calzada-Nova, G., Schnitzlein, W., Husmann, R., Zuckermann, F.A. 2009. Characterization of the cytokine and maturation responses. of pure populations of porcine plasmacytoid dendritic cells to porcine viruses and toll-like receptor agonists. Vet. Imm. Immunopath. doi: 10.1016/j.vetimm.2009.10.26. Cano JP, Dee SA Murtaugh MM, and Morrison RB. Infection dynamics and clinical manifestations following experimental inoculation of gilts at 90 days of gestation with porcine reproductive and respiratory syndrome virus. . Can J Vet Res (Accepted for publication). Cano, J.P., S.A. Dee, M.P. Murtaugh, A. Rovira, and R.B. Morrison. 2008. Infection dynamics and clinical manifestations following experimental inoculation of gilts at 90 days of gestation with a low dose of porcine reproductive and respiratory syndrome virus. Can. J. Vet. Res. 73:303-307. Chang C-C, Yoon K-J, Zimmerman JJ. 2009. Persistent porcine reproductive and respiratory syndrome virus (PRRSV) infection in pigs does not require significant genetic change. J Swine Health Prod 17(6):318324. Chen Z, Zhou X, Lunney JK, Lawson S, Sun Z, Brown E, Christopher-Hennings J, Knudsen D, Nelson EA. Fang Y. 2009. Immunodominant epitopes in nsp2 of porcine reproductive and respiratory syndrome virus are dispensable for replication but play an important role in viral pathogenesis. J Gen Virology. Epub. 11/18/09. Chen, Z, S. Lawson, Z. Sun, X. Zhou, X. Guan, J. Christopher-Hennings, E. A. Nelson, Y. Fang 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 (accepted). Chen, Z., X. Zhou, J. K. Lunney, S. Lawson, Z. Sun, E. Brown, J. Christopher-Hennings, D. Knudsen, E. Nelson, Y. Fang. Immunodominant epitopes in nsp2 of porcine reproductive and respiratory syndrome virus are dispensable for replication but play an important role in modulation of host immune response. J. Gen. Virology (accepted). Cruz, J.L.G., Zuñiga, S., Sanchez, C.M., Ceriani, J.E., Urniza, A., Plana-Duran, J. and Enjuanes L. Immunogenicity of a TGEV-based vector expressing porcine reproductive and respiratory syndrome virus GP5 and M proteins. Vaccine. Submitted. Das, P. B., Dinh, P. X., Ansari, I. H., de Lima, M., Osorio, F. A., and Pattnaik, A. K. (2010). The Minor Envelope Glycoproteins GP2a and GP4 of Porcine Reproductive and Respiratory Syndrome Virus Interact with the Receptor, CD163. J. Virology In press. Deendayal Patel, David A. Stein, and Yan-Jin Zhang: Morpholino Oligomer-Mediated Protection of Porcine Pulmonary Alveolar Macrophages from Arterivirus-Induced Cell Death. Antiviral Therapy 2009. In Press. de Lima, M. Ansari, I. H., Das, P. B., Ku, B., Martinez-Lobo, F. J., Pattnaik, A. K., and Osorio, F. A. (2009). GP3 is a Structural Component of the PRRSV Type II Virion. Virology, 390:31-36. de Abin, M.F, G. Spronk, M. Wagner, M. Fitzsimmons, J. Abrahante, and M.P. Murtaugh. 2009. Comparative infection efficiency of porcine reproductive and respiratory syndrome virus field isolates on MA104 cells and porcine alveolar macrophages. Can. J. Vet. Res. 73:200-204. Dee SA, Otake S, Oliviera S and Deen J. Evidence of long distance airborne spread of porcine reproductive and respiratory syndrome virus and Mycoplasma hyopnuemoniae. Vet Res 2009, 40(4)39. Dee SA, Pitkin AN and Deen J. Evaluation of alternative strategies to MERV 16-based air filtration systems for reduction of the risk of airborne spread of porcine reproductive and respiratory syndrome virus. Vet Microbiol doi:10.1016/j.vetmic.2009.03.019. Du, Y., Zuckermann, FA, and Yoo, D. 2009. Myristoylation of the small envelope protein of porcine reproductive and respiratory syndrome virus is non-essential for virus infectivity but negatively affects its growth. Virus Res. (submitted) Ellingson JS, Wang, Y., Layton, S., Ciacci-Zanella, J., Roof, M. B., and Faaberg, K. S. Vaccine efficacy of porcine reproductive and respiratory syndrome virus chimeras. Vaccine, submitted. Fang, Y., J. Christopher-Hennings, E. Brown, H. Liu, Z. Chen, S. Lawson, R. Breen, T. Clement, X. Gao, J. Bao, D. Knudsen, R. Daly and E.A. Nelson. 2008. Development of genetic markers in the non-structural protein 2 region of a US type 1 porcine reproductive and respiratory syndrome virus: implications for future recombinant marker vaccine development. J. Gen. Virol. 89:3086-3096. Gaudreault, N, RR Rowland, CR Wyatt. 2009. Factors affecting the permissiveness of porcine alveolar macrophages for porcine reproductive and respiratory syndrome virus. In press Archiv Virol. Gudmundsdottir I, Risatti GR. Infection of porcine alveolar macrophages with recombinant chimeric porcine reproductive and respiratory syndrome virus: effects on cellular gene transcription and virus growth. Virus Res. 2009 Oct;145(1):145-50. Lunney JK. 2008. Genetics of Infectious Disease Resistance in Animals. Proceedings American College of Veterinary Pathologists Meeting 2008. P. 240-242. Tuggle CK, Wang YF, Couture OP, Qu L, Uthe JJ, Kuhar D, Lunney JK, D. Nettleton D, J.C. Dekkers JC, Bearson SMD. 2008. Computational Integration of Structural and Functional Genomics Data across Species to Develop Information on Porcine Inflammatory Gene Regulatory Pathway. Dev Biol (Basel). 132: 105-13. Lunney JK, Ho C-S, Wysocki M, Smith DM. 2009. Molecular genetics of the swine major histocompatibility complex, the SLA complex. Dev Comp Immunol. 33: 362-74. Lunney JK, Fritz ER, Reecy JM, Kuhar D, Prucnal E, Molina R, Christopher-Hennings J, Zimmerman J, Rowland RRR. 2009. Interleukin-8, interleukin-1b and interferon-g levels are linked to PRRS virus clearance. Viral Immunology. In Revision. Han J, Rutherford, M. S., and K. S. Faaberg. 2009. Porcine reproductive and respiratory syndrome virus nsp2 cysteine protease domain possesses both trans- and cis-cleavage activities. J. Virol. 83:9449-9463. Han, J., M. S. Rutherford, and K. S. Faaberg. Proteolytic products of the porcine reproductive and respiratory syndrome virus nsp2 replicase protein. J. Virol., submitted. He D., C.C. Overend, R.J. Maganti, J. Ambrogio, , M.J. Grubman and A.E. Garmendia 2009. Marked differences between MARC-145 cells and alveolar macrophages in IFN ²-induced activation of antiviral state against PRRSV. Submitted to Vet Imm Immunopath Hermann JR, Brockmeier SL, Yoon KJ, Zimmerman JJ. 2008. Detection of respiratory pathogens in air samples from acutely infected pigs. Can J Vet Res 72:367-370. Hermann JR, Muñoz-Zanzi CA, Zimmerman JJ. 2009. A method to provide improved dose-response estimates for airborne pathogens in animals: An example using porcine reproductive and respiratory syndrome virus. Vet Microbiol 133:297-302. Hermann JR, Zimmerman JJ. 2008. Analytical sensitivity of air samplers based on uniform point source exposure to airborne porcine reproductive respiratory syndrome virus and swine influenza virus. Can J Vet Res 72:440-443. Ho C-S, Lunney JK, Franzo-Romain MH, Martens GW, Lee Y-J, Lee J-H, Wysocki M, Rowland RRR, Smith DM. 2009. Molecular characterization of swine leukocyte antigen (SLA) class I genes in outbred pig populations. Animal Genetics. 40: 468-78. Ho C-S, Lunney JK, Ando A, Rogel-Gaillard C, Lee J-H, Schook LB, Smith DM. 2009. Nomenclature for factors of the SLA system, update 2008. Tissue Antigens. 73: 307-315. Ho, C-S, Y-J Lee, JK Lunney, MH Franzo-Romain, GW Martens, J-H Lee, M Wysocki, RRR Rowland, DM Smith. 2009. Molecular characterization of swine leukocyte antigen (SLA) class I genes in outbred pig populations". Accepted for publication Anim Genetics. Holtkamp DJ, Yeske PE, Polson DD, Melody JL, Philips RC. 2009. A prospective cohort study evaluating duration of breeding herd PRRS virus-free status and its relationship with measured risk. Prev. Vet. Med. (submitted). Holtkamp DJ., Yu L, Polson DD, OConnor A. 2009. External and internal biosecurity factors associated with the occurrence of clinical PRRS breaks. Vet Res (submitted). Huang YW, Fang Y, Meng XJ. Identification and characterization of a porcine monocytic cell line supporting porcine reproductive and respiratory syndrome virus (PRRSV) replication and progeny virion production by using an improved DNA-launched PRRSV reverse genetics system. Virus Res. 2009 Oct;145(1):1-8. Huang YW, Dryman BA, Li W, Meng XJ. Porcine DC-SIGN: molecular cloning, gene structure, tissue distribution and PRRSV binding characteristics. Dev Comp Immunol. 2009 Apr;33(4):464-80. Jar, A.M., Osorio, F.A., Lopez, O.J. 2009 Mouse x pig chimeric antibodies expressed in baculovirus retain the same properties of their parent antibodies. Biotechnology Progress Mar-Apr;25(2):516-23. Jacobs AC, Hermann JR, Muñoz-Zanzi C, Prickett JR, Roof MB, Zimmerman JJ. 2010. Thermostability of porcine reproductive and respiratory syndrome virus in solution. J Vet Diagn Invest 22: (in press). Kim W-I, Kim J-J, Cha S-H, Yoon K-J. 2008. Different biological characteristics between wild-type PRRS viruses and vaccine viruses and identification of the corresponding genetic determinants. J Clin Microbiol 46:1758-1768. Kim W-I, Yoon K-J. 2008. Molecular assessment of the role of envelope-associated structural proteins in cross neutralization among different PRRS viruses. Virus Genes 37:380-391. Jung, K., G.J. Renukaradhya, K.P. Alekseev, Y. Fang, Y. Tang, and L.J. Saif (2009). 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. 90:2713-23. Kim, DY, TJ Kaiser, K Horlen, ML Keith, LP Taylor, R Jolie, JG Calvert, RR Rowland. 2008. Insertion and deletion in a non-essential region of the nonstructural protein 2 (nsp2) of porcine reproductive and respiratory syndrome (PRRS) virus: effects on virulence and immunogenicity. In press Virus Genes. Klinge, K.L., E.M. Vaughn, M.B. Roof, E.M. Bautista, and M.P. Murtaugh. 2009. Age-dependent resistance to Porcine reproductive and respiratory syndrome virus replication in swine. Virol. J. 6:177-187. Loving, C. L., S. L. Brockmeier, A. L. Vincent, K. M. Lager, and R. E. Sacco. 2008. Differences in clinical disease and immune response of pigs challenged with a high-dose versus low-dose inoculum of porcine reproductive and respiratory syndrome virus. Viral Immunol. 21:315-325. Lunney JK, Fritz ER, Reecy JM, Kuhar D, Prucnal E, Molina R, Christopher-Hennings J, Zimmerman J, Rowland RRR. 2010. Interleukin-8, interleukin 1² and interferon-³ levels are linked to PRRS virus clearance. Viral Immunol (in press). Metwally, S., F. Mohamed, T. Burrage, M. Prarat, K. Moran, A. Bracht, G. Mayr, M. Berninger, K. S. Faaberg, L. Koster, L.T. Thanh, V.L. Nguyen, M. Reising, S. Swenson, J. Lubroth and C. Carrillo. Pathogenicity and molecular characterization of emerging porcine reproductive and respiratory syndrome virus in Vietnam 2007. Transboundary and Emerging Diseases, submitted. Miller, L.C., Lager, K.M. and Kehrli Jr., M.E. 2009. Effect of porcine reproductive and respiratory syndrome virus infection of porcine alveolar macrophages on Toll-like receptors elicitation of type I interferon responses. Clinical and Vaccine Immunology 16:360-365. Molina, R.M., E.A. Nelson, J. Christopher-Hennings, R. Hesse, R.R. Rowland, J.J. Zimmerman, 2009. Evaluation of the risk of PRRSV transmission via ingestion of muscle from persistently-infected pigs. Transboundary and Emerging Diseases 56:1-8. Molina R, S.-H. Cha, W Chittick, S. Lawson, M.P. Murtaugh, E.A. Nelson, J Christopher-Hennings, K.-J. Yoon, R. Evans, R.R.R. Rowland and J.J. Zimmerman. 2008. Immune response against porcine reproductive and respiratory syndrome virus during acute and chronic infection. Veterinary Immunology and Immunopathology. Molina, R.M., W. Chittick, E.A. Nelson, J. Christopher-Hennings, R.R.R. Rowland and J.J. Zimmerman. 2008. Diagnostic performance of assays for the detection of anti-PRRSV antibodies in porcine muscle transudate (meat juice) samples. J. Vet. Diagn. Invest. 20:735-743. Mohammadi, H, S Sharif, RRR Rowland, D. Yoo. 2009. The lactate dehydrogenase-elevating virus capsid protein is a nuclear-cytoplasmic protein. Arch Virol. 154:1071-1080. Murtaugh, M.P., C.R. Johnson, Z. Xiao, R.W. Scamurra, and Y. Zhou. 2009. Species specialization in cytokine biology: is interleukin-4 central to the TH1-TH2 paradigm in swine. Develop. Comp. Immunol. 33:344-352. Opriessnig T, Patterson AR, Madson DM, PalM, Rothschild M, Kuhar D, Lunney JK, Juhan NM, Meng XJ, Halbur PG. 2009. Difference in severity of porcine circovirus type 2 (PCV2)-induced pathological lesions and disease between Landrace and Pietrain pigs. J. Animal Science. 87: 1582-90. Opriessnig T, Madson DM, Prickett JR, Kuhar D, Lunney JK, Elsener J, Halbur PG. 2008. Effect of porcine circovirus type 2 (PCV2) vaccination on porcine reproductive and respiratory syndrome virus (PRRSV) and PCV2 coinfection. Vet. Microbiol. 131: 103-14. Patton, J. B., R.R. Rowland, D. Yoo, and K.C. Chang. 2009. Modulation of CD163 receptor expression and replication of porcine reproductive and respiratory syndrome virus in porcine macrophages. Virus Res. 140:161-171. Pei, Y., D.C. Hodgins, J. Wu, S.K.W. Welch, J.G. Calvert, G. Li, Y. Du, C. Song, and Yoo, D.. 2009. Porcine reproductive and respiratory syndrome virus as a vector: Immunogenicity of green fluorescent protein and porcine circovirus type-2 capsid expressed from dedicated subgenomic RNAs. Virology 389:91-99. Pieters M, Dee SA, Fano E and Pijoan C. An assessment of the duration of Mycoplasma hyopneumoniae infection in an experimentally infected population of pigs. Vet Microbiol 2009;143:261-264. Pitkin AN, Deen J and Dee SA. Further assessment of fomites and personnel as vehicles for the mechanical transport and transmission of porcine reproductive and respiratory syndrome virus. Can J Vet Res (Accepted for publication). Pitkin AN, Deen J and Dee SA. Use of a production region model to assess the airborne spread of porcine reproductive and respiratory syndrome virus. Vet Microbiol 2009;136:1-7. Pitkin AN, Otake S, Deen J, Moon RD, Dee SA. Further assessment of houseflies (Musca domestica) as vectors for the mechanical transport and transmission of porcine reproductive and respiratory syndrome virus under field conditions. Can J Vet Res 2009;73:91-96. Prickett J, Kim W-I, Simmer R, Yoon K-J, Zimmerman JJ. 2008. Surveillance of commercial growing pigs for PRRSV and PCV2 infections using pen-based oral fluid sample: a pilot study. J Swine Health Prod 16(2):86-91. Prickett J, Simer R, Christopher-Hennings J, Yoon K-J, Evans RB, Zimmerman JJ. 2008. Detection of porcine reproductive and respiratory syndrome virus infection in porcine oral fluid samples: A longitudinal study under experimental conditions. J Vet Diagn Invest 20:156-163. Ran ZG, Chen XY, Guo X, Ge NX, Yoon K-J, Yang HC. 2008. Recovery of viable porcine reproductive and respiratory syndrome virus from an infectious clone containing partial deletion within Nsp2-encoding region. Arch Virol 153:899-907. Renukaradhya, G.J., K.P. Alekseev, K. Jung, Y. Tang, Y. Fang, and L.J. Saif (2009). Altered immune responses to porcine respiratory coronavirus in pigs previously infected with porcine reproductive and respiratory syndrome virus. Vet. Immunol. Immunopath. (In review). Rosenfeld, P., P.V. Turner, J.I. MacInnes, E. Nagy, and D. Yoo. 2009. Evaluation of porcine reproductive and respiratory syndrome virus replication in the laboratory rodents. Can. J. Vet. Res. 73: 313-318. Sang, Y, P Ruchala, R Lehrer, CR Ross, RRR Rowland, Frank Blecha. 2009. Antimicrobial Host Defense Peptides in an Arteriviral Infection: Differential Peptide Expression and Virus Inactivation. Viral Immun. Accepted for publication. Sang, Y, C Ross, RR Rowland, F Blecha. 2008. Toll-like receptor 3 (TLR3) activation decreases porcine arterivirus infection. Viral Immunol 21:303-313. Sang, Y, J Yang, C Ross, RRR Rowland, and F Blecha, 2008. Molecular identification and functional expression of porcine Toll-like receptor (TLR) 3 and TLR7. In press Vet Immunol Immunopath. Song, C., R. Lu, D. Bienzle, H.C. Liu, and D. Yoo. 2009. Interaction of porcine reproductive and respiratory syndrome virus nucleocapsid protein with the inhibitor of MyoD family-a domain containing protein. Biol. Chem. 390: 215-223. Spilman, M.S., C. Welbon, E.A. Nelson and T. Dokland. 2009. Cryo-electron tomography of porcine reproductive and respiratory syndrome virus (PRRSV): organization of the nucleocapsid. J. Gen. Virol. 90:527-535. Steibel JP, Wysocki M, Lunney JK, Ramos AM, Hu Z-L, Rothschild MF, Ernst CW. 2009. Validation of the Swine Protein-Annotated Oligonucleotide Microarray. Animal Genetics. 40: 883-893. Trible BR, Kerrigan, M., Faaberg, K. S., and R. R.R. Rowland. Identification of an immunodominant region the PCV2 capsid protein recognized by naturally infected and vaccinated pigs. Journal of General Virology, submitted. Vashisht, K., Erlandson, K.R., Firkins, L.D., Zuckermann, F.A., Goldberg, T.L. 2008. Evaluation of contact exposure as a method for acclimatizing growing pigs to porcine reproductive and respiratory syndrome virus. J Am Vet Med Assoc. 232:1530-5. Vashisht, K., Goldberg, T.L., Husmann, R.J., Schnitzlein, W., Zuckermann, F.A. 2008. Identification of immunodominant T-cell epitopes present in glycoprotein 5 of the North American genotype of porcine reproductive and respiratory syndrome virus. Vaccine. 26:4747-53. Vincent, A. L., Lager, K. M., Faaberg, K. S., Harland, M. L., Zanella, E., Ciacci-Zanella, J., Kehrli, Jr., M. E., Janke, B. H., Klimov, A. Susceptibility of Pigs to Pandemic 2009 A/H1N1 Influenza Virus. Plos Pathogens, submitted. Wenjun Ma, AL Vincent, KM Lager, BH Janke, SC Henry, RRR Rowland, RA Hesse, JA Richt. 2009. Identification and characterization of a highly virulent triple reassortant H1N1 swine influenza virus in the United States. Virus Genes, in press. Wu, J., J. Li, F. Tian, J. Shi, S. Ren, Z. Lan, X. Zhang, D. Yoo, and J. Wang. 2009. Porcine high fever disease: genetic variation and pathogenicity of porcine reproductive and respiratory syndrome virus in China. Arch. Virol. 154: 1579-1588. Xue Han, Sumin Fan, Deendayal Patel, and Yan-Jin Zhang: Enhanced Inhibitory Effect on PRRSV Replication by Combination of Two Morpholino oligomers. Antiviral Research 2009. 82:59-66. Yaeger M, Karriker L, Layman L, Halbur P, Huber G, Van Hulzen K. 2009. Survey of disease pressures in twenty-six niche herds in the Midwestern United States. J Swine Health Prod. 2009 17(5): 256-263. Yue F, Cui S, Zhang C, Yoon K-J. 2009. A multiplex PCR for rapid and simultaneous detection of porcine circovirus type 2, porcine parvovirus, porcine pseudorabies virus, and porcine reproductive and respiratory syndrome virus in clinical specimens. Virus Genes (in press) 2. List book chapters or monographs None 3. List abstracts or proceedings Beura L, Sarkar S , Kwon BJ, Subramaniam S, Jones C, Pattnaik AK, Osorio FA. Porcine reproductive and respiratory syndrome virus non structural protein 1 beta inhibits host innate immune response by antagonizing IRF3 activation. Proceedings of the 28th Annual Meeting American Society for Virology (25th ASV), Vancouver, BC, July 11-15, 2009 (Workshop 33-6). Butler, J.E. 2009 A comparative study of PRRSV, PCV-2 and SIV infections in germfree isolator piglets. Presented at 5th International Veterinary Vaccine Conference (5th IVVDC), Madison WI. July 23. Carley D, Ramamoorthy S, Opriessnig T, Wong C, Tobin G, Yoon KJ, Halbur PG, Messel R, Nara PL. 2009. Evaluation of the PRRSV antibody response following vaccination with a proprietary autogenous vaccine. Proceedings, Summer Scholar Program. Carley D, Ramamoorthy S, Opriessnig T, Wang C, Tobin G, Yoon KJ, Halbur PG, Messel R, Nara PL. August 2009. Evaluation of the PRRSV antibody response following vaccination with a proprietary autogenous vaccine. Research Day, College of Veterinary Medicine, Iowa State University. Ames, IA. Carmichael BA, Polson DD, Torremorell M, Holtkamp DJ. 2009. Pre-colostral stillborn piglet blood sampling procedure when a PRRSV positive sow herd is being monitored for time-to-negative interval. 40th AASV Annual Meeting. Dallas, Texas. Cha S-H, Dorman KS, Kim W-I, Yoon KJ. 2008. Viral recombination among field isolates of PRRSV type 2  implication for molecular epidemiology. PRRS Symposium. Chen Z, X Zhou, D Kuhar, S Lawson, J Lunney, Y Fang. 2008. Effect of PRRSV nsp2 epitope deletion mutants on the induction of cytokine response in porcine alveolar macrophages. 2008 CRWAD & 2008 PRRSV Symp. Chen Z, X Zhou, S Lawson, E Brown, R Breen J. Christopher-Hennings, E Nelson, Y Fang. 2008. Expression of foreign proteins in replicase gene regions of porcine reproductive and respiratory syndrome virus. CRWAD & 2008 PRRSV Symp. Chitko-McKown, C.G., Miller, L.C., Lager, K.M. and Kehrli Jr., M.E. Effects of PRRSV infection on TLR-dependent induction of NOS [abstract]. 2008 Conference of Research Workers in Animal Diseases (CRWAD), December 7-9, 2008, Chicago, IL. Chitko-McKown, C.G., Chapes, S.K., Miller, L.C., and Green, B.T. Characterization of the porcine monocyte-derived cell lines Cdelta2+ and Cdelta2-. 41st Annual Meeting of the Society for Leukocyte Biology, November 6-8, 2008, Denver, CO. Cutler T, Hoff S, Wang C, Warren K, Zhou F, Qin Q, Miller C, Ridpath J, Yoon K-J, Zimmerman J. 2009. UV254 Inactivation of Selected Viral Pathogens. 52st Annual Conference, American Association of Veterinary Laboratory Diagnosticians. San Diego, California, p. 124. Cutler T, Kittawornrat A, Hoff S, Wang C, Zimmerman J. 2009. Median infectious dose (ID50) of PRRSV isolate MN-184 for young pigs via aerosol exposure. 52st Annual Conference, American Association of Veterinary Laboratory Diagnosticians. San Diego, California, p. 110. Cruz, J. L. G., Zúñiga, S., Sánchez, C. M., Ceriani, J. E., Plana, J., and Enjuanes, L. 2008. Design of a recombinant TGEV vector to protect against porcine reproductive and respiratory syndrome. EuroPRRSnet Workshop. Combating PRRS in Europe. Cruz, J. L. G., Zúñiga, S., Sánchez, C. M., Urniza, A., Bru, T., Ceriani, J. E., Plana, J., and Enjuanes, L. 2008. Construction of a TGEV vector to protect against porcine reproductive and respiratory syndrome. 2008 PRRS Symposium. Cruz, J. L. G., Zúñiga, S., Sánchez, C. M., Ros, S., Juanola, S., Plana, J., and Enjuanes, L. 2009. Design of a TGEV vector to protect against porcine reproductive and respiratory syndrome. 2009 PRRS Symposium. Dee SA and Otake S. Investigation of alternative strategies for aerosol biosecurity for PRRSV. CRWAD, Chicago, Il, December 2008. Dee SA and Otake S. Use of a production region model to evaluate issues regarding the aerobiology of PRRSV and Mycoplasma hyopneumoniae. CRWAD, Chicago, Il, December 2008. Dee SA, AN Pitkin, Deen J. Alternative strategies for aerosol biosecurity for PRRSV. 2008 Pijoan Intl Symp on Swine Dis Erad St. Paul, September, 2008. Dee SA, Otake S, Deen J. Use of a production region model to evaluate the transmission and biosecurity of PRRS and Mycoplasma hyopneumoniae. AASV, Dallas, Tx, March 2009. Dee SA, Pitkin AN, Otake S and Deen J. Transmission of EP and PRRS. PIC Veterinary Conference, Stratford-upon-Avon, England, February 2009. Dion KR, Dau D, Delks AM, Hammer M, Holtkamp DJ. 2009. Feed medication protocol comparison in a PRRSv unstable nursery flow. 40th AASV Meeting. Dallas, Texas. Dixon PM, Yoon K-J. 2008. Estimating the mutation rate when not all mutations are detected. Proceedings, Iowa State University Fall Conference on Statistics in Biology. Dwivedi V, C. Manickam, R. Patterson, K. Dodson, and G. J. Renukaradhya. Steps towards development of a novel mucosal vaccine to PRRSV. Fourth International Scholar Research Exposition, The Ohio State University, November 19th, 2009. Dwivedi V, C. Manickam, R. Patterson, K. Dodson, and G. J. Renukaradhya. Development of a novel mucosal vaccine to protect against porcine reproductive and respiratory syndrome in pigs. 2009 PRRSV Symposium. Faaberg KS, J. Han, K. M. Lager, M. E. Kehrli, Jr., and M. S. Rutherford. 2008. PRRSV strain VR-2332 nsp2 deletion mutants attenuate clinical symptoms in swine. XIth International Symposium on Nidoviruses, P43, Oxford, Great Britain. Faaberg, K. S. 2008. State of the Art: Lessons learned through porcine reproductive and respiratory syndrome virus (PRRSV) recombinant technology. International Congress of Virology, 675, Istanbul, Turkey. Fang, Y. 2008. Structural and Function of PRRSV nonstructural proteins: where are we at? 2008 PRRSV Symp. Fritz E, Hu Z, Lunney J, Reecy J. 2008. The PHGC Database: management of large data sets. Intnl PRRS Symp. #324. 12/0. Guo, B., Faaberg, K. S., Lager, K., and M. E. Kehrli, Jr. 2009. Genetic stability of PRRSV VR-2332 nsp2 deletion mutants in swine. 28th Annual Meeting of the American Society for Virology, W33-12, Vancouver, Canada. Haley CA, Wagner B, Murtaugh MP. 2009. Estimating the sensitivity and specificity of a new ELISA test for porcine circovirus 2 exposure using a study pseudo gold standard and latent-class analysis. Proc AASV. pp 255-261. Harms PA, Holtkamp DJ, Quirk Z. 2009. Management of, and costs associated with, false positive when monitoring presumed PRRS-negative herds. 40th AASV Annual Meeting, Pre-Conference Seminar, Managing PRRS Introduction into High Risk Populations. Dallas, Texas. Holtkamp DJ, Melody JL, Burkgren TJ. 2008. Update On The AASV Production Animal Disease Risk Assessment Program (PADRAP) and the New Web-based Application. 20th IPVS Meetings. Durban, South Africa. June. p. 01.66. Holtkamp DJ, Melody JL, MacDougald D. 2008. A comparison of PRRSV risks for Canadian and U.S. breeding herds. 20th IPVS Meetings. Durban, South Africa. June. p. 01.77. Holtkamp DJ, Polson DD. 2008. PRRS herd classification: Can we speak the same language? Carlos Pijoan International Symposium: New Solutions to Old Problems. Pre-Conference Workshop 2006 Allen D. Leman Swine Conference. St. Paul, Minnesota. Hesse, R, R Rowland. 2008. Circovirus Vaccination Decisions: Herd Profiling and Next Generation Diagnostic Testing. 2008 Leman Swine Conference, St Paul, MN. Hiep Vu , Kwon BJ, Yoon KJ, Laegreid W, Pattnaik AK and Osorio FA. Analysis of the aberrant immune response induced by a PRRSV type 2 isolate naturally lacking glycan residues in two envelope glycoproteins. 2009 IPRRSS (#84) and 2009 CRWAD (poster # 64). Hicks, J., D. Yoo, and H.C. Liu. 2008. Identification of domains of PRRS virus GP5 and M proteins that interact with the host Snap-associated protein SNAPIN. 2008 IPRRSS. Hu J, Meng XJ, and Zhang C. 2008. Purification of native PRRSV virions from cell culture. 2008 PRRS Symposium. Huang YW, B. A. Dryman, X. J. Meng. 2008. Molecular cloning of porcine DC-SIGN and detection of its potential interaction with porcine reproductive and respiratory syndrome virus. 2008 International PRRS Symposium. 2008 PRRS Symposium. Huang YW, B. A. Dryman, X. J. Meng. 2008. Molecular cloning of porcine DC-SIGN and detection of its potential interaction with porcine reproductive and respiratory syndrome virus. 2008 International PRRS Symposium. 2008 CRWAD. Jung K, K. Alekseev, G.J. Renukaradhya, Y. Tang, Y. Fang, P. Lewis, X. Zhang, L.J. Saif. Altered pathogenesis of porcine respiratory coronavirus (PRCV) in the presence of PRRSV infection and their pathologic relationships: Potential effect of preexisting respiratory viral infections on SARS severity. XIV International Congress of Virology, ASV, Cornell University, Ithaca, NY, 11-15 August 2008. Jung K, K. Alekseev, G.J. Renukaradhya, Y. Tang, Y. Fang, P. Lewis, X. Zhang, L.J. Saif. Altered pathogenesis of porcine respiratory coronavirus (PRCV) subsequent to PRRSV infection: Model for effect of respiratory viral co-infections on SARS severity. 2008 CRWAD. Karriker L, Bowden J. 2009. PRRS virus vaccination strategies and efficacy. Proceedings of the George A. Young Swine Health and Management Conference. South Sioux City, Nebraska. Karriker L, Layman L, Yaeger M. 2008. Session 5531: Health challenges in niche production. Conference Notes CD of the 145th AVMA. New Orleans, Louisiana. Karriker L. 2008. Session 5543: PRRSV: management strategies. Current swine disease trends. Conference Notes CD of the 145th AVMA. New Orleans, Louisiana. Kim W-I, Bower L, Strait E, Harmon K, Yoon K-J. 2009. Simultaneous detection of multiple pathogens using high-throughput nanoliter real-time PCR. AAVLD. Kim W-I, Cho Y-I, Harmon K, Madson D, Opriessnig T, Yoon K-J. 2008. Comparison of three extraction methods for the detection of PCV2 and PRRSV in semen. AAVLD. Kim W-I, Sun D, Cho Y-I, Liu S, Cooper V, Yoon K-J. 2009. Identification of molecular markers for virulence of porcine reproductive and respiratory syndrome (PRRS) virus. AAVLD Kim W-I, Sun D, Cho Y-I, Liu S, Loynachan AT, Cooper VC, Yoon KJ. 2009. Genetic determinants associated with the virulence of PRRSV in pigs. AASV. Koziel JA, Yang X, Cutler T, Zhang S, Zimmerman J, Hoff S, Jenks WS, van Leeuwen JH, Laor Y, Ravid U, Armon R. 2008. Treatment of livestock odor and pathogens with ultraviolet photocatalysis. Proc AgEng 2008 International Conference on Agricultural Engineering and Industry Exhibition, (Abstr OP-575). Hersonissos, Greece. Koziel JA, Yang X, Zhang S, Cai L, Hoff SJ, Leeuwen HJ, Cutler T, Zimmerman J, Jenks WS, Laor Y, Ravid U, Armon R. October 2008. Treatment of livestock odor and pathogens with ultraviolet photocatalysis. Proc 3rd IWA Odour and VOCs Conference. Barcelona, Spain. Lawson S, Lunney JK, Fang Y, Nelson EA, Christopher-Hennings J. 2009. Development of a rapid, swine-specific microsphere assay to simultaneously detect multiple immune proteins (cytokines) affected by porcine reproductive and respiratory syndrome virus (PRRSV) infection. 2009 Intnl PRRS Symp. and CRWAD 12/09. Liu S, Kim W-I, Ramamoorthy S, Yoon K-J. 2009. Alternative assays and testing algorithm for confirmation of suspect false positives in a commercial ELISA for PRRSV. AAVLD. Lunney JK. 2008. Genetics of Infectious Disease Resistance in Animals: Pig and PRRS. Proc. Am Coll Vet Pathol. Meeting. 11/08 Lunney JK. 2009. PRRS Host Genetics Consortium: Current Progress and Potential for Canadian Involvement. Canadian Centre for Swine Improvement meeting, Quebec City, Canada 6/09 Lunney JK, Boyd P, LaBresh J, Kakach L, Wagner B, Tompkins D, Hudgens E, Baldwin C. 2009. Swine Toolkit progress for the US Veterinary Immune Reagent Network. 2009 Intnl PRRS Symp. and CRWAD 12/09. Lunney JK, Boyd P, Prucnal L, Zarlenga D, LaBresh J, Steffens C, Wagner B, Tompkins D, Hudgens T, C Baldwin C. 2008. Swine Toolkit progress for the US Veterinary Immune Reagent Network. Intnl PRRS Symp. #288 and CRWAD 95P. 12/08 Lunney JK, Reecy J, Rowland RRR. 2009. PRRS Host Genetics Consortium: Current Progress and Potential for Canadian Involvement. Canadian Swine Health Forum 2009: July 7-8, 2009, Saskatoon, SK, Canada. Lunney JK, Reecy J, Rowland RRR. 2009. Current Progress of US PRRS Host Genetics Consortium. Genomics for Animal Health: Outlook for the Future (EADGENE 2009) meeting, 10/09, Paris, France. Lunney JK, Rowland RRR, Chen Z, Zhou X, Lawson S, Sun Z, E. Brown E, J. Christopher-Hennings J, Nelson E, Fang Y. 2009. Genetic approaches to reveal immune response pathways and viral antigen targets for novel vaccine design. Intnl Vet Vaccines and Diagnostics Conference (IVVDC 2009), WI. 7/09. Lunney JK, Steibel JP, Reecy J, Rothschild M, Kerrigan M, Trible B, Rowland RRR. 2009. PRRS Host Genetics Consortium: Current Progress. 2009 Intnl PRRS Symp. and CRWAD 12/09. Lunney JK, Wysocki M, Steibel JP, Kuhar D, Ernst CW, McCaw M. 2009. Uncovering Genetic Components Involved In Regulating Early Immune Responses To Porcine Reproductive And Respiratory Syndrome (PRRS). PAG2009. PAG-XVII P640. 1/09. Loruzzo, A., Faaberg, K. S., Killian, M. L., Koster, L., Vincent, A. L. 2009. One step real-time RT-PCR for 2009 pandemic H1N1 matrix gene detection and quantitation in clinical samples. American Association of Swine Veterinarians 2010 Annual Meeting, March 6-9, 2010, Omaha, NE. Metwally S, C. Carrillo, F. Mohamed, K. Faaberg, M. McIntosh, L. Cox, L. Koster, S. Swenson, T. Burrage, T. Long, T. Beckham, E. Lautner, and J. Lubroth. 2008. Porcine High Fever Disease in Vietnam 2007; PRRS and Other Disease Agents. 51st Annual Meeting of the American Association of Veterinary Laboratory Diagnosticians, Greensboro, NC, USA. Miller, L.C., Harhay, G.P., Lager, K.M., Kehrli Jr., M.E., Laegreid, W.W. and Neill, J.D. In depth global analysis of gene expression levels in porcine alveolar macrophages following infection with porcine reproductive and respiratory syndrome virus [abstract]. ARK-Genomics Conference 2008: 3rd International Symposium on Animal Functional Genomics, April 7-9, 2008, Edinburgh, U.K. Paper No. ISAFG-P22.p. 38. Miller, L.C., Chitko-McKown, C.G., Lager, K.M. and Kehrli Jr., M.E. Differential roles of Toll-like receptors in the elicitation of type I interferon responses by alveolar macrophages [abstract]. 2008 International PRRS Symposium, December 5-6, 2008, Chicago, IL. Molina RM, Cha S-H, Rowland RRR, Christopher-Hennings J, Nelson E, Lunney J, Yoon K-J, Zimmerman JJ. 2008. Factores involucrados en la persistencia del virus de sindrome respiratorio porcino (PRRS). Memorias XXI Congreso Panamericano de Ciencias Veterinarias. Guadalajara, México, pp. 533- 534. Morrison RB, Davies PD and Dee SA. Regional and national eradication of PRRS. PIC Veterinary Conference, Stratford-upon-Avon, England, February 2009. Morrison, RB. Update on PRRS elimination in Stevens County, MN. Allen D Leman Swine Conference, preconference workshop. Pp. 67-74. Murtaugh M. 2009. Update on PRRSV immunology and viral genetics: from hopeless to hopeful. Proc AASV. pp 459-462. Osorio FA. Worldwide Research Efforts Towards a Broadly Protective and Effective Vaccine against PRRSV. Keynote presentation # 4 at the 2008 IPRRSS. Osorio FA. PRRSV immunology and vaccines Second Annual CVM Swine Health Initiatives Meeting UIUC, PRRSV protective immunity and immunization, Osorio FA presented at the XVIII Congreso Dia del Porcicultor, Navojoa, Sonora Mexico. 2008. Otake S, Deen J, Dee SA. New information aerosol transmission and biosecurity for Mycoplasma hyopneumoniae. 2008 Leman Swine Conference. Otake S, Deen J, Dee SA. Preliminary information from recent research on PRRSV and Mycoplasma hyopneumoniae transmission and biosecurity: Field application of air filters. 2008 Leman Swine Conference. Pires-Alves, M., Misra, A., Zuckermann, F.A., Laegreid, W. 2009. Comparison of two cell lines for the propagation of PRRSV. 2009 IPRRSS. Potter, ML, S Dritiz, R Hesse, R Rowland, J Nietfeld, R Oberst. 2008. Porcine Cirovirus Type 2 elimination study. 2008 AASV. Potter, ML, LM Tokach, SS Dritz, SC Henry, JM DeRouchey, MD Tokach, RD Goodband, JL Nelsen, RR Rowland, RD Hesse, RA Hesse. 2008. Genetic background influences pig growth rate responses to porcine circovirus type 2 (PCV2) vaccines. 2008 KSU Swine Day. Prickett J, Cutler S, Kinyon J, Naberhaus N, Stensland WR, Yoon K-J, Zimmerman J. 2008. PRRSV surveillance  Stability of diagnostic targets in oral fluid: sample storage and critical techniques for testing. Proc 89th CRWAD, Abstr #38. Prickett J, Cutler S, Kinyon J, Naberhaus N, Stensland WR, Yoon K-J, Zimmerman J. 2008. PRRSV surveillance  Stability of diagnostic targets in oral fluid: sample storage and critical techniques for testing. 2008 IPRRSS, p 26. Prickett J, Hoffmann P, Main R, Sornsen S, Johnson J, Zimmerman J. 2009. Cost-effective PRRS surveillance. AASV, pp. 467-469. Prickett J, Hoffmann P, Main R, Stensland W, Yoon K-J, Zimmerman J. 2008. Practical disease surveillance in growing pig populations. Proc 89th CRWAD, Abstr #39. Prickett J, Hoffmann P, Main R, Stensland W, Yoon K-J, Zimmerman J. 2008. Practical disease surveillance in growing pig populations. Proc 2008 IPRRSS, p 27. Prickett J, Zimmerman J. 2008. Practical disease surveillance in growing pig populations. Proceedings, Welfare and Epidemiology Conference: Across Species, Across Disciplines, and Across Borders. Ames, Iowa, p. 22. Prickett J, Zimmerman J. 2009. Practical disease surveillance in growing pig populations. J Appl Anim Welf Sci 12:156. Prickett JR, Cutler S, Kinyon J, Naberhaus N, Stensland W, Yoon KJ, Zimmerman J. 2008. PRRSV surveillance - Stability of diagnostic targets in oral fluid: sample storage and critical techniques for testing. Proc 51st Annual Conference, American Association of Veterinary Laboratory Diagnosticians. Greensboro, North Carolina, p. 140. Prickett JR, Cutler S, Kinyon J, Naberhaus N, Stensland W, Yoon K-J, Zimmerman J. 2008. PRRSV surveillance - stability of diagnostic targets in oral fluid: Sample storage and critical techniques for testing. Proc 47th North Central Conference of Veterinary Laboratory Diagnosticians. Madison, Wisconsin, pp. 8. Prickett JR, Cutler S, Kinyon J, Naberhaus N, Stensland WR, Yoon K-J, Zimmerman JJ. 2008. PRRSV surveillance  stability of diagnostic targets in oral fluid: sample storage and critical techniques for testing. AAVLD. Prickett JR, Cutler S, Kinyon J, Naberhaus N, Stensland WR, Yoon K-J, Zimmerman JJ. 2008. PRRS surveillance  Stability of diagnostic targets in oral fluid: Sample storage and critical techniques for testing. NCCVLD. Prickett JR, Hoffmann P, Main R, Sornsen S, Johnson J, Zimmerman J. 2008. Infectious disease surveillance in commercial swine populations. Proceedings 16th Annual Swine Disease Conference for Swine Practitioners, Iowa State University. Ames, Iowa, pp. 40-44. Prickett JR, Hoffmann P, Main R, Stensland W, Yoon K-J, Zimmerman J. June 2008. Practical disease surveillance in growing pig populations. Proc 47th North Central Conference of Veterinary Laboratory Diagnosticians. Madison, Wisconsin, pp. 10. Prickett JR, Hoffmann P, Stensland W, Yoon KJ, Zimmerman J. 2008. Practical disease surveillance in growing pig populations. Proc 51st Annual Conference, AAVLD, Greensboro, NC, p. 139. Renukaradhya GJ, Konstantin Alekseev, Kwonil Jung, and Linda J. Saif. Distorted immune responses in pigs to porcine respiratory coronavirus previously infected with porcine reproductive and respiratory syndrome virus: a respiratory viral co-infection model. 2009 CRWAD. Reister L, T Clement, E Nelson, J Christopher-Hennings. 2008. Potential mechanical and antiviral methods to insure PRRSV free semen. 2008 CRWAD & 2008 PRRSV Symp. Rovira A, Abrahante J, Murtaugh M. 2009. Detection of porcine reproductive and respiratory syndrome virus (PRRSV) by reverse transcriptase loop mediated isothermal amplification (RT-LAMP). Proc AASV. pp 109-110. Rowland RRR, Kerrigan M, Bujuru S, Trible B, Lunney JK. 2009. An infection model for the study of PRRS at the population level. 2009 IPRRSS. 12/09. Rowland, R, S Henry, S Dritz, R Hesse. 2008. Epidemiology of PCV2 and PCVAD. 2008 AASV. Rowland, R, R Hesse, K Horlen. 2008. Porcine circovirus vaccine trials: from the laboratory bench to the field. George Young Swine Conference, Sioux City. Rowland, R. 2008. PRRS vaccines. 2008 Leman Swine Conference, St Paul, MN. Rowland, R. 2008. Mapping host protective immunity in the PCV2 capsid protein. USDA NRI Prinicipal Investigators Meeting, Chicago. Sang, Y, P Ruchala, RI Lehrer, CR Ross, RRR Rowland, F Blecha. 2008. Antimicrobial host defense peptides in an arteriviral infection: differential expression and inactivation of PRRSV. 2008 IPRRSS. Sun D, Kim W, Cho Y, Cooper V, Cha S, Kim S, Choi E, Yoon K. 2009. Role of viral structural proteins in conferring protective immunity against PRRSV and its application to the development of vaccine candidates for broad cross-protection. 2009 AASV. Sun D, Kim W-I, Cho Y-I, Cha S-H, Kim S-H, Choi E-J, Yoon KJ. 2008. Cross-protection induced by chimeric mutant containing mixed structural genes of two different PRRS viruses. 2008 CRWAD. Sun D, Kim W-I, Cho Y-I, Yoon KJ. 2008. Attempt to achieve broader cross-protection among PRRS viruses by vaccination: use of chimeric mutant containing mixed structural genes of two different PRRS viruses. 2008 PRRS Symp. Trible, B, JG Calvert , RRR Rowland. 2008. Expression of enhanced green fluorescent protein (EGFP) in nonstructural protein 2 (nsp2) of PRRSV shows loss of fluorescence without affecting EGFP immunogenicity. Tuggle CK, Bearson SMD, Uthe JJ, Christian C, Couture O, Demirkale CY, Nettleton D, Lunney JK, Honavar V. 2009. Using transcriptomic data to develop tools for predicting shedding traits in growing pigs. CRWAD 12/09. Vincent, A. L., Lager, K. M., Faaberg, K. S., Harland, M. L., Zanella, E., Ciacci-Zanella, J., Kehrli, Jr., M. E., Janke, B. H., Klimov, A. Susceptibility of North American Swine to the Novel A/H1N1 Influenza A Virus. Center of Excellence Symposium, Minneapolis, MN. Vu HLX, M. Brito M, Kim WI, Yoon KJ, Laegreid W, Osorio FA. 2008. Sub-typing PRRSV isolates by means of measurement of cross neutralization reactions. CRWAD. Vu HLX, M. Brito M, Kim WI, Yoon KJ, Laegreid W, Osorio FA. 2008. Sub-typing PRRSV isolates by means of measurement of cross neutralization reactions. PRRS Symp. Waddell JM, Melody JL, Holtkamp DJ. 2009. Investigation of associations between risk factors, reported clinical PRRS breaks and reproductive performance in swine breeding herds. Proc. 40th AASV Annual Meeting. Dallas, Texas. Waddell JT, Polson DD, Holtkamp DJ. 2008. Assessment of changes in breeding herd PRRS site risk scores in a large production system over a three-year period. Proc 20th IPVS Meetings. Durban, South Africa. p. 01.135. Wong SJ, Lunney JK, Rowland RRR. 2009. Nucleocapsid protein-specific IgG and IgM responses in oral fluids during PRRSV infection. 2009 Intnl PRRS Symp. 12/09. Wong, SJ, R Hesse, R Rowland. Application of multiplex microsphere immunoassay techniques to the diagnosis of PRRSV and other infectious. 2008 International PRRS Symposium, Chicago. Wysocki M, SteibelJP, McCaw M, Kuhar D, Ernst CW, Lunney JK. 2008. Uncovering genetic components involved in early regulatory immune response during PRRSV infection. Intnl PRRS Symp. #285 and CRWAD #125. 12/08. Wu, J., Li, J., Tian, F., Shi, J., Ren, S., Lan, Z., Zhang, X., Niu, Z., Yoo, D., and Wang, J. 2008. Genetic variation and pathogenicity of porcine reproductive and respiratory syndrome virus in Shandong area of China. Intl PRRS Symposium, Chicago, IL. Yang X, Koziel JA, Cutler T, Zhang S, Zimmerman J, Hoff SJ, Jenks W, van Leeuwen J, Harmon J, Faulhaber C, Laor Y, Ravid U, Armon R. 2008. Treatment of livestock odor and pathogens with ultraviolet light. American Society of Agricultural and Biological Engineers (ASABE) Paper No. 085198. ASABE Annual International Meeting. Providence, Rhode Island. Yoon K-J. 2008. PRRS diagnosis as tool for evidence-based PRRS control. KASV. Zimmerman J, Prickett J, Johnson J. 2009. Oral fluid testing: Science-based applications. Carlos Pijoan International Symposium: New Approaches to Herd Diagnostics. 2009 Allen D. Leman Swine Conference. pp. 11-14. Zimmerman J, Prickett J, Johnson JH, Molina R. 2009. Evaluando fluidos orales: Base científica de su aplicación. XVIII Día del Porcicultor 2009. Asociación de Médicos Veterinarios Zootecnistas Especialistas en Ciencias Porcicolas del Sur de Sonora A.C. Navojoa, Sonora, Mexico (CD). Zimmerman J, Prickett J. 2008. Future epidemiologic / ecologic methods. 1st Annual Boehringer Ingelheim Swine Academy. Ames, Iowa, Vol 1, pp. 251-260. Zimmerman J, Prickett JR, Molina R, Hoffmann P, Main R, Sornsen S, Johnson J. 2008. Vigilancia epidemiológica de enfermedades infecciosasa en poblaciones porcinis comerciales utilizando fluidos orales. Memorias XXI Congreso Panamericano de Ciencias Veterinarias. Guadalajara, México, pp. 231-234. Zimmerman J. 2009. The Big Picture: Infectious disease, oral fluid testing, and swine health. 2nd Annual Boehringer Ingelheim Swine Academy. Ames, Iowa, pp. 181-200. Xiuqing Wang, Hanmo Zhang, Xueshui Guo. 2009. The interaction between PRRSV and type I interferon induction signaling pathways. 28th American Society for Virology. Yoo, D., Y. Sun, and N. Chen. 2008. One-step mutagenesis of the full-length infectious clone of PRRSV and generation of engineered viruses. Intl PRRS Symposium, Chicago, IL, Dec 6-7. Zuckermann, F.A. , Calzada-Nova, G., Schnitzlein, W., Husmann, R. 2009. Proficient isolation and titration of field PRRS virus from clinical samples using a porcine alveolar macrophage cell line. 2009 Intl PRRS Symposium, Chicago, IL, Dec 6-7. FUNDING SOURCES FOR PRRSV RESEARCH: Currently Funded Competitive Research Grants: Blecha, et al, USDA NRI, 2006-2009. Porcine antimicrobial peptides and Toll-like receptors in PRRS pathogenesis, $340,000. Christopher-Hennings J, Y Fang, J Lunney, EA Nelson. Development of a rapid, single tube, multiplex test to simultaneously detect immune parameters (cytokines) induced by PRRSV. National Pork Board, $101,107. Christopher-Hennings J, Y Fang, EA Nelson. Elimination of PRRSV from semen: On Farm Mechanical and Antiviral Methods. National Pork Board $94,558, 2007-2009. Enjuanes L. Induction of cross-protective immunity without exposure to live PRRSV (NPB #08-197). National Pork Board. 2008-2009. Enjuanes L. Mechanisms inducing protection against coronaviruses and arteriviruses. Fort Dodge, S.A. 2008-2009. Enjuanes L. Plant Production of Vaccines (PLAPROVA, EU 227056). European Communities. 2009-2011. Faaberg KS, Collins JE, Yoon K-J, Christopher-Hennings J, Crow JA, Leung FC. 09/01/07-08/30/08. Implementation of a PRRSV strain database. National Pork Board PRRS Initiative. $51,322. Faaberg, Guo, Miller: NPB, 11/01/09-11/01/10, $53,877, Molecular Identification of Type I Interferon Antagonistic Components of PRRSV Proteins. Faaberg: NRICGP, 10/1/06-6/30/10, $274,998, Biological Studies of Putative Nonstructural Protein 2 in Porcine Reproductive and Respiratory Syndrome Virus. Fang Y, JK Lunney, J Christopher-Hennings, E Nelson, A Young. The role of PRRSV non-structural proteins 1 and 2 in host immunity. USDA-NRI, $375,000, 1/08-12/2010. Fang Y, W Zhang, J Christopher-Hennings, EA Nelson, RB Baker. Development of an epitope-based vaccine against swine influenza A using a non-toxic enterotoxin as the carrier-adjuvant. National Pork Board. $49,993. Fang Y, Zimmerman J, Christopher-Hennings J, Nelson E, Murtaugh M, Lunney J. 01/01/10 to 12/31/11. Development of diagnostic assays for detecting PRRSV infection using oral fluid samples as an alternative to serum-based assays. National Pork Board - $99,989. Gourapura RJ. Evaluation of adjuvants at the mucosal area for the development of innovative mucosal vaccine against PRRS. National Pork Board. Nov. 2008 to May 1, 2010. Gourapura RJ. Study of mucosal immune responses in the respiratory tract of pigs infected with porcine reproductive and respiratory syndrome virus. OARDC Seed grant, The Ohio State University. March 2009 to February 2011. Gourapura RJ. Development of novel mucosal vaccines for the control of PRRSV outbreaks. National Pork Board. Dec. 2009 to May, 2011. Hesse and Rowland, 2009, Fort Dodge Animal Health, Heterotypic immunity as a platform for a new generation of modified live PRRS vaccines. $100,000 Holtkamp D, Ramirez A, O'Connor A, Zimmerman J. 01/01/09 to 12/31/09. Assessment of PRRS biosecurity in the field: Application of the American Assocation of Swine Veterinarians PRRS Risk Assessment Tool. USDA:CSREES National Research Initiative, Competitive Grants Program 230.1 Animal and Plant Biosecurity (subcontract: Kansas State University) - $24,300. Holtkamp D, Zimmerman J. 10/01/08 to 09/30/09. Quantifying risk factors for PRRSV introduction into swine herds through the use of the PRRS Risk Assessment Tool. National Pork Board - $89,875. Holtkamp D.J. A Cross-sectional Study Of PRRSV Positive Swine Breeding Herd Sites To Evaluate Associations Between Risk Factors And A Case Definition-based Number And Severity Of Clinical PRRS Episodes. Boehringer Ingelheim Vetmedica Inc. $115,000. Continuation of previous grant with same title. January 6, 2009. Kim W-I, Yoon K-J, Cooper VC. 11/01/07-10/30/08. Identification of protective epitopes toward developing a vaccine providing broad cross-protection among PRRS viruses. National Pork Board PRRS Initiative. $89,150. Kim W-I, Yoon K-J. 7/1/08-6/30/10. A new approach to PRRS vaccine that confers cross-protection against a broader range of PRRS viruses using chimeric mutant PRRS viruses. Iowa Healthy Livestock Initiative Research Grant. $39,980. Laegreid W., F. Osorio, T. Goldberg, J. Christopher-Hennings, E. Nelson. Immunological consequences of PRRSV Diversity. USDA-NRI, PRRSV CAP2. $947,885. Lunney J, J Christopher-Hennings, EA Nelson, Y Fang, JP Steibel, J Zimmerman. 01/01/10 to 12/31/11. Comparison of early immune responses of pigs which are genetically PRRS resistant/tolerant using a swine-specific immune protein (cytokine) multiplex assay. National Pork Board. $103,929. Lunney JK, J Dekkers, R Fernando, Z Jiang, H-C Liu, R Pogranichniy, JM Reecy, R Rekaya, M Rothschild, D Smith, JP Steibel, C Tuggle. PRRS CAP Host genetics: Characterization of host factors that contribute to PRRS disease resistance and susceptibility. USDA NIFA PRRS CAP2: Objective 3 Host Genetics. $560,000. 2009-2012. Lunney JK,C Ernst, V. Honavar, Z Jiang, R Pogranichniy, JP Steibel, C Tuggle. Identifying porcine genes and gene networks involved in effective response to PRRS virus using functional genomics and systems biology. USDA AFRI/NIFA Animal Genome, Genetics, and Breeding Program. $750,000. 2010-2012 Meng, X. J., Y. Fang, T. Opriessnig. Innovative approaches to develop a broadly protective and effective vaccine(s) against PRRSV. USDA PRRS CAP2, $100,000, 1/200912/2010. Miller, Harhay, Lager: NPB, 11/01/08-11/01/10, $139,152, Gene Expression in lymph nodes of PRRSV-infected pigs Murtaugh M (and MN stations). PRRS CAP 2, Minnesota Pork Board, National Pork Board, USDA, University of Minnesota Swine Disease Eradication Center, Minnesota Rapid Agricultural Response Fund. Murtaugh M, Gourapura RJ. Positive Prognosticators of Immune Protection and Prophylaxis against PRRSV in Swine Herds. PRRSV PRRS CAP 2. August 2009 to July 2013. Osorio FA: Immunologic Consequences of PRRSV Diversity, USDANRICGP CAP2 (Kansas State University subcontract), $74,368 August 2009-July 2010. Osorio FA: Development of a modified live vaccine against PRRSV with optimal DIVA marker potential, National Pork Board, Grant Period: 11/01/2008 - 12/31/2009 (extended at no cost) $125,700 Osorio FA: Porcine Reproductive and Respiratory Virus: role of viral genes in virulence/attenuation, USDA NRICGP Project No. No.2008-00903, Period: 09/01/2008 - 08/31/2011, $374,900. Pattnaik AK.; Molecular Structures of PRRSV that Contribute to PRRSV Protective Immunity. National Pork Board. $ 138,600; 12/01/2009-11/31/2010. Pattnaik AK.; Glycoproteins of Porcine Reproductive and Respiratory Syndrome Virus in Infection and Immunity?; States Department of Agriculture, AFRI (2009-01576), $371,230; 09/01/2009-08/31/2012. Pattnaik AK.: Role of All of PRRSV Glycoproteins in Protective Immune Response, 11/01/2008 - 10/31/2009 (extended at no cost) National Pork Board, $106,000. Rowland RRR, Lunney JK, Reecy J, Johnson, R, NPB, 2007-2008. PRRS host genetics consortium: A proposal to develop a consortium to study the role of host genetics and resistance to PRRSV. $300,000. Rowland RRR, Lunney JK, Reecy J, NPB, 2009-2010. PRRS host genetics consortium: A proposal to develop a consortium to study the role of host genetics and resistance to PRRSV. $247,000. Rowland et al., USDA NRI Coordinated Agricultural Program (CAP), 2008-2012. Integrated strategies to control and reduce the impact of PRRS virus control, $4.8 million. Wang, X. Interaction between PRRSV and interferon alpha/beta induction signaling pathways. USDA NRI 12/2008-11/2010 $ 100,000. Yoo D. Private industry, $90,000, Antiviral effects of tilmicosin on swine respiratory viruses. 2008-2009. Yoo D. USDA CSREES NRI, $375,000, Evasion strategies of PRRSV from the host defense. 2008-2011. Yoon K-J. 1/1/07-12/31/10. Development of surveillance program and vaccine for PRRS in Korea: Development of a differential test for PRRS vaccine virus and immunological study of viral factors for protective immunity. Korean Ministry of Agriculture and Forestry (c/o National Veterinary Research and Quarantine Service). $280,000. Zimmerman J, Dee SA, Davies PR, Holtkamp DJ, OConnor A. 08/01/09 to 07/31/10. Identifying ecologic and epidemiologic factors in the control of PRRS: A field-based approach. USDA:CSREES National Research Initiative, Competitive Grants Program 230.1 Animal and Plant Biosecurity (subcontract: Kansas State University) - $116,991. Zimmerman J, Hoff SJ. 07/01/09 to 06/30/10. Effect of temperature and humidity on ultraviolet (UV) inactivation of airborne PRRS virus. Innovative Swine Industry Enhancement Grant Program, Iowa Attorney Generals Office - $43,046. Zimmerman J. 05/01/09 to 04/31/10. PRRS CAP Graduate and Undergraduate Scholar Program Application - Iowa State University. USDA:CSREES National Research Initiative, Competitive Grants Program 230.1 Animal and Plant Biosecurity (subcontract: Kansas State University) - $23,999. Gold Sheet #97704. Zuckermann F. USDA NRI CPG, $369,064. In vivo analysis of PRRS virus immunopathogenesis. Tracking Number: GRANT00168948. 2007-2010.
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.