NC_old229: Detection and Control of Porcine Reproductive and Respiratory Syndrome Virus and Emerging Viral Diseases of Swine

(Multistate Research Project)

Status: Active

SAES-422 Reports

Annual/Termination Reports:

[12/22/2019] [01/01/1970] [01/05/2022] [01/27/2023] [01/08/2024]

Date of Annual Report: 12/22/2019

Report Information

Annual Meeting Dates: 11/02/2019 - 11/03/2019
Period the Report Covers: 12/02/2018 - 11/02/2019

Participants

Listed in the meeting minutes

Brief Summary of Minutes

Accomplishments

<p><strong><span style="text-decoration: underline;">Objective 1. CONTROL OF PRRSV</span></strong></p><br /> <p>&nbsp;<strong>1.1. PRRS immunology/vaccinology</strong><strong>:</strong></p><br /> <p>Correlation of protection and neutralizing antibodies <strong>(UMN)</strong>, differences in antibody response based in genetic host background <strong>(UMN)</strong>, immunity related with quasispecies and cross protection between different PRRSV linages <strong>(UMN), </strong>and genetic effects on PRRSV persistence <strong>(BARC). </strong>Studies were performed to understand heterologous PRRSV protection <strong>(UConn, UNL)</strong>, immune response against to synthetic vaccines<strong> (UNL)</strong>, antiviral activity by over-expression of IFITM3 and ZMPSTE 24 <strong>(SDSU)</strong>, viral infection interaction with interferon-activated JAK/STAT pathway <strong>(UMD)</strong>, reduction in stress granules <strong>(SDSU)</strong>, and effect of virus expression of interferons <strong>(NACD)</strong></p><br /> <p><strong>&nbsp;</strong><strong>1.2. PRRS epidemiology:</strong></p><br /> <p>Studies were performed on genome variation in highly pathogenic PRRSV, epidemiological factors associate with PRRSV elimination, role of animal movement networks in PRRS epidemiology, airborne exposure, biosecurity methods for PRRSV inactivate, sampling method and processing fluids for farrowing barns, and spatiotemporal cluster of ORF5 <strong>(UMN)</strong>.</p><br /> <p><strong>&nbsp;</strong><strong>1.3. PRRS Surveillance and Diagnostics</strong>:</p><br /> <p>Development of the Swine Pathogen Database (<a href="https://swinepathogendb.org">https://swinepathogendb.org</a>) <strong>(NACD)</strong>. Phylogenetic analysis of diagnostic PRRSV samples was examined for <strong>SDSU</strong>, <strong>ISU</strong> and <strong>KSU</strong> <strong>NACD</strong> and evaluation of quasispecies in re-emergent populations <strong>(UMN)</strong>. First detection and characterization in Peru <strong>(SDSU)</strong></p><br /> <p>&nbsp;<strong><span style="text-decoration: underline;">Objective 2 Developing effective and efficient approaches for detection, prevention and control of pressing viral diseases of swine of recent emergence</span></strong></p><br /> <p><strong>2.1: ASFV</strong></p><br /> <p>Epidemiological status of African swine fever in in Republic of Armenia <strong>(Uconn)</strong> and genetic Characterization of ASFV in Asia and Africa <strong>(Uconn)</strong>. Survivability of ASFV in feed, effect in effect in foreign food suppliers, and risk for viral introduction on US <strong>(UMN)</strong></p><br /> <p><strong>2.2: Swine Influenza Virus:</strong></p><br /> <p><strong>Vaccine research: Subunit vaccines base on Alphavirus vectors (NACD), nanoparticle vaccine (OSU)</strong> cross-protective efficacy of intranasal nanovaccine by incorporating CpG-ODN <strong>(OSU)</strong>, hemagglutinin vaccine immunogen against H3 influenza A viruses of swine <strong>(UNL)</strong>, heterologous prime-boost using whole inactivated virus <strong>(UMN)</strong>, effect of sow vaccines in pigs at weaning <strong>(UMN)</strong>, and transmission of live attenuated influenza vaccine to non-vaccinated pigs <strong>(UMN), </strong>polymer-based vaccine delivery system for broadly protective peptide vaccine for SIV<strong> (NDSU)</strong></p><br /> <p><strong>Epidemiology research:</strong> Influenza transmission and persistency <strong>(UMN)</strong>, risk of transmission of Avian influenza to humans <strong>(UMN)</strong> and zoonotic potential of human derived influenza <strong>(UMN)</strong>. Antigenic evolution of H3N2 influenza A in viruses United States <strong>(NACD)</strong>, human-origin influenza A (H3N2) reassortant viruses in swine in Mexico <strong>(UMN)</strong>. Role of Influenza D in Swine Influenza other agricultural animals <strong>(SDSU)</strong>.</p><br /> <p><strong>Diagnostic research:</strong> Automated classification tool for influenza viruses <strong>(NACD)</strong>, validated novel sampling methods <strong>(UMN)</strong>, developed a GMR biosensor chip <strong>(UMN)</strong>, monoclonal antibodies specific for Influenza D virus <strong>(SDSU)</strong>, primary porcine respiratory epithelial cells to study swine influenza viruses <strong>(SDSU).</strong></p><br /> <p><strong>2.3 Porcine Circovirus: </strong></p><br /> <p>Evaluation of nucleotide variation that affects PRRSV vaccination and clinical response against PCV2 and PRRSV challenge <strong>(BARC), </strong>characterization of new linear B cell epitopes of the capsid protein<strong> (NDSU)</strong></p><br /> <p><strong>2.4 Swine Pestiviruses:</strong></p><br /> <p><strong>2.5: Senecavirus:</strong></p><br /> <p>Studies were conducted to estimate the Senecavirus A (SVA) seroprevalence in US swine herds <strong>(UMN).</strong> Characterization of SVA pathogenesis <strong>(NADC, SDSU)</strong>, immune response <strong>(SDSU)</strong>, persistent infection, and clinical response to a potential live attenuated vaccine <strong>(SDSU)</strong>. New diagnostic tools that include Mab specific antibodies for IHC detection of SVA on lesions <strong>(SDSU)</strong>.</p><br /> <p><strong>2.6:&nbsp; Sapelovirus:&nbsp;&nbsp;</strong></p><br /> <p><strong>2.7: Viruses with potential interest to Xeno-transplantation science:</strong></p><br /> <p><strong>2.8: Porcine Coronaviruses</strong></p><br /> <p><strong>Studies were conducted to evaluate PEDV immune repose and infection effect on gilts post farrowing (NADC). Development and characterization of a </strong>recombinant porcine delta coronavirus virus (PDCoV)<strong> (NADC)</strong>. Development of rapid response vaccines using PEDV as a model <strong>(NDSU)</strong></p><br /> <p><strong>&nbsp;</strong><strong>Publications/funding sources:</strong> (see attached &ldquo;2019 NC229 Publications&rdquo;).</p><br /> <p><strong>Authorization</strong>:&nbsp; Submission by an AES or CES director or administrative advisor through NIMSS constitutes signature authority for this information.</p><br /> <p>*Limited to three pages or less exclusive</p>

Publications

Impact Statements

  1. As listed under accomplishments and publications
Back to top

Date of Annual Report: 01/01/1970

Report Information

Annual Meeting Dates: 12/05/2020 - 12/05/2020
Period the Report Covers: 01/01/1970 - 01/01/1970

Participants

Eric, Nelson (eric.nelson@sdstate.edu)-South Dakota State University; Osorio, Fernando A (fosorio@unl.edu)- and Vu, Hiep (hiepvu@unl.edu)University of Nebraska-Lincoln; Rowland, Raymond (browland@vet.k-state.edu)-Kansas State University; Benfield, David, (benfield.2@osu.edu) Ohio State University; Timothy, Sullivan (timothy.sullivan@usda.gov)-USDA-NIFA/Kansas: USDA; Lunney, Joan (joan.lunney@ars.usda.gov)-USDA-ARS, BARC; Gourapura, Renukaradhya (gourapura.1@osu.edu)-The Ohio State University (OSU); Schroeder, Declan (deschroe@umn.edu)-University of Minnesota; Pogranichniy Roman (rmp@vet.k-state.edu); Kansas State University-Ramamoorthy, S (sheela.ramamoorthy@ndsu.edu)- North Dakota St Univ (N.D.); Zhang, Yanjin (zhangyj@umd.edu)- University of Maryland; Pablo Pineyro (pablop@iastate.edu)-Iowa State University; Zuckermann, Federico (fazaaa@illinois.edu)- University of Illinois at Urbana-Champaign.

Brief Summary of Minutes

The 2020 NC229 meeting was held December 5, 2020, as a viirtual meeting on the zoom platform. The meeting was open to all NC229 members. Annaul reports from 15 stations were shared with the audience by speakers selected by the station rpresentatives for each station. More than 50 participants  accessed the meeting online. The agenda is presented in Table 1. The business meeting centered on the topics noted below:



  • Ramamoorthy (Chair) and Dr. Fernando Osorio (academic advisor) inaugurated the annual NC229 scientific meeting.

  • Ramamoorthy (Chair) and Roman Pogranichniy (Vice-Chair), and the scientific program committee were recognized for their outstanding efforts in organizing the scientific program for the online meeting.

  • Fernando Osorio discussed two major accomplishments, publication of the manuscript entitled "The NC229 multi-station research consortium on emerging viral diseases of swine: Solving stakeholder problems through innovative science and research” for a special edition on multi-state consortia in Virus Research, describing the history and accomplishments of NC229. He also informed the audience about the successful renwal of the project for the next 5 years.

  • Tim Sullivan & Dr. Mark Mirando presented updates regarding funding opportunities for the upcoming year. Two significant changes were announced: 1) update on minimum funding amount. 2) New investigators seed grants.

  • Additional funding opportunities were discussed related to Interagency funding opportunities: 1) Ecology and evolution on infectious diseases, and 2) Dual Purpose with Dual Benefits: Research in Biomedical and Agriculture Using Agriculturally important Domestic animals which might be renwed

  • Raymond Rowland, University of Illinois Urbana Champaign , who is a long standing member of NC229 and currently as an admisntrator, agreed to replace Dr. Fernando Osorio as the academic advisor of NC229, as Dr. Osorio had consented to serve as the interim advisor after the departure of Dr. Benfield. Drs. Rowland and Ramamoorthy expressed deep appreciation for Dr. Osorio’s contribution and leadership for the project.

  • Ying Fang led a discussion on seeking input from the group on changes to the format and location for the joint meeting of the North American PRRSV symposium and NC229 and separation of this satellite meeting from CRWAD. This scientific meeting is a flagship multi-state activity of NC229, bringing together researchers from academia, industry and other stakeholders from the U.S and other countries for the dissemination of scientific information relevant to NC229 goals and objectives.


 


Table: 1


























































Meeting Agenda Time



Presenter/ Station/Title



8.00am – 8.05am



Dr. Fernando Osorio –Advisor; Sheela Ramamoorthy –/Chair


Opening remarks



8.05am-8.45am



Dr. Tim Sullivan & Dr. Mark Mirando


National Program Leaders, Animal Production Systems, USDA- NIFA


Updates on funding and opportunities



8.45am-8.55am



Linhares D; Iowa State University; Overview of studies on PRRS


monitoring and control under field conditions



8.55am – 9.05am



Nelli R; Iowa State University; 3D culture systems as infection Models for swine diseases



9.05am-9.15am



Shi J and Niederwerder M, Kansas State University;Update on PRRS, ASF andCSF research



9.15am-9.25am



Wang X; South Dakota State University; Role of host restriction factors in  PRRSV replication



9.25am-9.35am



Pasternak A; Purdue University; A genome-wide association study (GWAS) of thyroid hormone levels In piglets challenged with PRRSV



9.35am-9.45am



Van Goor A; BARC USDA. Understanding the impact of RRSV infection on the Fetus



9.45am-9.50am



Miller LC; NADC; An overview of research at the National Animal Disease Center on endemic and emerging viral diseases of swine



9.50am -10.00am



VanderWaal K; University of Minnesota; From Pathogen to Populations:


Building a multi-scale understanding of how porcine viruses evolve, adapt, spread, and persist



10.00am-10.10am



Vu H; University of Nebraska-Lincoln; A simple and reliable method for


quantification of swine antibody response to PRRSV infection



10.10am-10.20am



Osorio F; University of Nebraska-Lincoln; Novel characterization of tropism PRRSV for swine epithelial Testicular cells


Accomplishments

<p>The introduction of foreign animal diseases (FADs) such as ASF, CSF, FMD into the U.S is a looming threat to the pork industry. Monitoring and preventing current endemic pathogens such as PRRSV, IAV, and PCV still remains a challenge. Collectively the major areas of progress include animal feed safety, ASFV detection/ prevention, molecular diagnostic test development for FADs. In addition, establishment of the swine health information center (SHIC) a multidisciplinary and multistate monitoring system for endemic diseases provides real-time information to producers and veterinaries.</p><br /> <p>&nbsp;</p><br /> <p><strong>Short-term Outcomes: </strong>No outcomes to report at this time.</p><br /> <p><strong>Outputs:</strong> The Swine Disease Reporting System (SDRS) was launched to aggregate real time data from participating veterinary diagnostic laboratories (VDLs) in the United States of America (ISU, U of M, SDSU, and KSU) and reports the significant findings to the swine industry</p><br /> <p>A cell line for the laboratory culture of AFSV was established</p><br /> <p>The survival and transmissibility of AFSV in animal feed was determined, along with information on specific feed ingrediants that either promote or inhibit viral replication.</p><br /> <p><strong>Activities:</strong></p><br /> <ul><br /> <li>The Swine Disease Reporting System (SDRS) has been providing monthly reports through multiple platforms (newsletters, podcasts, and YouTube videos) of the major disease trends. The collated cumulative data from the major Veterinary diagnostic laboratories also provides the opportunity to forecast the behavior of different diseases in specific production areas of the country</li><br /> <li>The NC 229 multistate annual meeting helped to disseminate the scientific advances contemplated within this project's objectives. More than 15 researchers share the advances on PRRSV immunology/vaccinology and epidemiology.</li><br /> </ul><br /> <p><strong>&nbsp;</strong></p><br /> <p><strong>Milestones:</strong></p><br /> <p><strong>Impacts: </strong>In the 2<sup>nd</sup> year of this 5 year project renewal, major impacts from multi-state efforts included improvements in dissemination of information regarding trends in animal infections, &nbsp;where data generated through SDRS is used daily by practitioners to make relevant decisions regarding biosecurity and prevention strategies of endemic pathogens, and development of models and generating data to inform best practices on formulation and transport of animal feed to reduce the risk of FAD introduction via feed and feed ingredients.</p>

Publications

<ol><br /> <li>Gilbert P; Megan CN; Gordon S; Scott AD. Quantification of soya-based feed ingredient entry from ASFV-positive countries to the United States by ocean freight shipping and associated seaports. <strong><em>Transbound Emerg Dis</em></strong> . 2020 October 16. doi: 10.1111/tbed.13881.</li><br /> <li>Niederwerder MC; Dee&nbsp; S; Diel DG; Stoian AMM; Constance LA;&nbsp; Olcha&nbsp; M; Petrovan V; Patterson G; Cino-Ozuna&nbsp; AG; Rowland&nbsp; Mitigating the risk of African swine fever virus in feed with anti-viral chemical additives. <strong><em>Transbound Emerg Dis</em></strong>&nbsp; 2020 July 2.&nbsp; doi: 10.1111/tbed.13699.</li><br /> <li>Dee S; Shah A; Cochrane R; Clement&nbsp; T; Singrey&nbsp; A; Edler&nbsp; R; Spronk&nbsp; G; Niederwerder&nbsp; M; Nelson&nbsp; E.Use of a demonstration project to evaluate viral survival in feed: Proof of concept. <strong><em>Transbound Emerg Dis</em></strong> . 2020 June 14.&nbsp; doi: 10.1111/tbed.13682. Online ahead of print.</li><br /> <li>Ricker N; Trachsel&nbsp; J; Colgan&nbsp; P; Jones&nbsp; J; Choi&nbsp; J; Lee&nbsp; J; Coetzee&nbsp; JF; Howe&nbsp; A; Brockmeier&nbsp; SL; Loving&nbsp; CL; Allen&nbsp; HK.Toward Antibiotic Stewardship: Route of Antibiotic Administration Impacts the Microbiota and Resistance Gene Diversity in Swine Feces . <strong><em> Vet. Sci.,</em></strong> May 19 2020 | https://doi.org/10.3389/fvets.2020.00255</li><br /> <li>Lerner AB; Cochrane RA; Gebhardt&nbsp; JT; Dritz S.S.; Jones&nbsp; CK; DeRouchey&nbsp; JM; Tokach&nbsp; M; Goodband&nbsp; RD; Bai&nbsp; J; Porter&nbsp; E; Anderson&nbsp; J; Gauger&nbsp; PC, Magstadt DR; Zhang J; Bass B; Karnezos T; de Rodas B; Woodworth JC <strong>. </strong>Effects of medium chain fatty acids as a mitigation or prevention strategy against porcine epidemic diarrhea virus in swine feed. <strong><em>J Anim Sci</em></strong>&nbsp; 2020 June 1;98(6):skaa159. doi: 10.1093/jas/skaa159.</li><br /> <li>Dee SA; Niederwerder MC; Patterson&nbsp; G; Cochrane&nbsp; R; Jones&nbsp; C; Diel&nbsp; D; Brockhoff&nbsp; E; Nelson&nbsp; E; Spronk G; Sundberg&nbsp; P.The risk of viral transmission in feed: What do we know, what do we do?. <strong><em>Transbound Emerg Dis</em></strong>.&nbsp;2020 Nov;67(6):2365-2371.&nbsp;doi: 10.1111/tbed.13606.</li><br /> <li>Jackman JA; Boyd RD; Elrod&nbsp; CC<strong>. </strong>Medium-chain fatty acids and monoglycerides as feed additives for pig production: towards gut health improvement and feed pathogen mitigation. <strong><em>J Animal Sci Biotechnol</em></strong>&nbsp;11<strong>,&nbsp;</strong>44 (2020). https://doi.org/10.1186/s40104-020-00446-1</li><br /> <li>Stoian AMM; Petrovan&nbsp; V; Constance&nbsp; LA; Olcha&nbsp; M; Dee&nbsp; S; Diel&nbsp; DG; Sheahan&nbsp; MA; Rowland&nbsp; RRR; Patterson&nbsp; G; Niederwerder&nbsp; Stability of classical swine fever virus and pseudorabies virus in animal feed ingredients exposed to transpacific shipping conditions. <strong><em>Transbound Emerg Dis</em></strong> 2020 Jul;67(4):1623-1632. doi: 10.1111/tbed.13498. Epub 2020 Feb</li><br /> <li>Gebhardt JT; Thomson&nbsp; KA; Woodworth&nbsp; JC; Dritz S.S.; Tokach&nbsp; MD; DeRouchey&nbsp; JM; Goodband RD; Jones&nbsp; CK; Cochrane&nbsp; RA; Niederwerder&nbsp; MC; Fernando&nbsp; S; Abbas&nbsp; W; Burkey&nbsp; TE. Effect of dietary medium-chain fatty acids on nursery pig growth performance, fecal microbial composition, and mitigation properties against porcine epidemic diarrhea virus following storage. <strong><em>J Anim Sci</em></strong>.&nbsp;2020 January 1;98(1):skz358. &nbsp;doi: 10.1093/jas/skz358.</li><br /> <li>Wang Y; Yim-Im&nbsp; W; Porter&nbsp; E; Lu&nbsp; N; Anderson&nbsp; J; Noll&nbsp; L; Fang&nbsp; Y; Zhang&nbsp; J; Bai&nbsp; J. Development of a bead-based assay for detection and differentiation of field strains and four vaccine strains of type 2 porcine reproductive and respiratory syndrome virus (PRRSV-2) in the USA. <strong><em>Transbound Emerg Dis</em></strong>. 2020 August 20. doi: 10.1111/tbed.13808. Online ahead of print.</li><br /> <li>Shang P; Yuan&nbsp; F; Misra&nbsp; S; Li Y; Fang&nbsp; Y. Hyper-phosphorylation of nsp2-related proteins of porcine reproductive and respiratory syndrome virus. <strong><em>Virology</em></strong>. 2020 Apr;543:63-75. doi: 10.1016/j.virol.2020.01.018. Epub 2020 February 4.</li><br /> <li>Jara M; Rasmussen&nbsp; DA; Corzo CA; Machado&nbsp; G. Porcine reproductive and respiratory syndrome virus dissemination across pig production systems in the United States. <strong><em>Transbound Emerg Dis. </em></strong>2020 July 13. doi: 10.1111/tbed.13728. Online ahead of print.</li><br /> <li>Trevisan G; Linhares LCM; Crim&nbsp; B; Dubey&nbsp; P; Schwartz&nbsp; KJ; Burrough ER; Wang&nbsp; C; Main R.G.; Sundberg P; Thurn &nbsp;M; Lages PTF; Corzo&nbsp; CA; Torrison&nbsp; J; Henningson&nbsp; J; Herrman&nbsp; E; Hanzlicek&nbsp; GA; Raghavan&nbsp; R; Marthaler&nbsp; D; Greseth J;&nbsp; Clement&nbsp; T; Christopher-Hennings J <sup>5</sup>; Muscatello D<sup> 6</sup>; Linhares&nbsp; DC<sup>1</sup>. Prediction of seasonal patterns of porcine reproductive and respiratory syndrome virus RNA detection in the U.S. swine industry. <strong><em>J Vet Diagn Invest</em></strong>. 2020 May;32(3):394-400. doi: 10.1177/1040638720912406. Epub 2020 April 10.</li><br /> <li>Benfield D; Lunney JK; Murtaugh M; Nelson E; Osorio&nbsp; F; Pogranichniy&nbsp; R; Ramamoorthy S; Rowland&nbsp; RRR; Zimmerman&nbsp; JJ<sup>;</sup> Zuckermann&nbsp; The NC229 multi-station research consortium on emerging viral diseases of swine: Solving stakeholder problems through innovative science and research. <strong><em>Virus Res</em></strong>. 2020 April 15;280:197898. doi: 10.1016/j.virusres.2020.197898. Epub 2020 February 28</li><br /> <li>Benfield D; Lunney JK; Murtaugh M; Nelson E; Osorio&nbsp; F; Pogranichniy&nbsp; R; Ramamoorthy S; Rowland&nbsp; RRR; Zimmerman&nbsp; JJ<sup>; </sup>Zuckermann&nbsp; The NC229 multi-station research consortium on emerging viral diseases of swine: Solving stakeholder problems through innovative science and research. <strong><em>IPVS 2020 Proceedings </em></strong>Rio de janeiro page 761. Base on paper No. 14.</li><br /> </ol>

Impact Statements

Back to top

Date of Annual Report: 01/05/2022

Report Information

Annual Meeting Dates: 12/04/2021 - 12/04/2021
Period the Report Covers: 12/05/2020 - 12/04/2021

Participants

Brief Summary of Minutes

The 2021 NC229 meeting was held December 4th, 2021 in conjunction with the NA PRRS Symposium in Chicago Marriott Downtown Magnificent Mile Hotel. The meeting has the honor to host 19 presenters from multiple stations and 37 poster presentation. The business meeting centered on the topics noted below:


 



  • NIFA representative could not attend the meeting due to USDA restrictions.

  • Dr. Rowland was approved as advisor of the NC229 group.

  • Dr. Sheela Ramamoorthy from North Dakota State University completed 2-year term as chair of NC229, and Dr. Roman Pogranichniy from Kansas State University will be becoming chair of NC229 for a 2-years term.

  • Dr. Pablo Pineyro (Iowa State University) assumed the position of vice-chair and Dr. Hiep Vu (Nebraska state university) secretary of the NC229 committee. 

  • Andreia Arruda (The Ohio State University) was elected as the new member at large

  • A short retirement ceremony was conducted in honor of Dr. Osorio led by Dr. Bob Rowland 

  • Dr. Ying Fang and Dr. Bob Rowland announced the results of the vote cast by the NC229 station representatives on whether to move the meeting to a new location or date, with the majority of station reps voting for a change of venue and time. 

  • The meeting was adjourned.

Accomplishments

<ol><br /> <li>Influenza vaccine efficacy studies demonstrated that intranasal delivery of Nano-11 and Poly(I:C) based inactivated SwIAV vaccine induce polyfunctional and cross-protective cell-mediated immunity. (<strong>The Ohio State University, Purdue University)</strong></li><br /> <li>PRRSV detection studies showed a higher probability of detection in processing fluids compared in breeding farms and tonsil scrapings in growing pig farms. Collaborators: <strong>The Ohio State University</strong>, <strong>North Carolina State University</strong>, and <strong>University of Minnesota</strong>).</li><br /> <li>We demonstrated during fetal infection, PRRSV produce an endocrine disruption, reducing animal performance. Additional studies showed that thyroid hormone levels may be promising biomarkers for genetic improvement of resilience during PRRSV challenge <strong>(ARS, USDA, Purdue University, University of Saskatchewan)</strong></li><br /> <li>We demonstrated effective protection induced by an experimental subunit DIVA vaccine against PRRS Virus <strong>(University of Illinois, University of Nebraska)</strong>. In addition, monoclonal antibodies were developed for a DIVA-based, diagnostic ELISA for detection of vaccinated versus naturally infected animals <strong>(USDA, (South Dakota State University)</strong></li><br /> <li>Susceptibility studies demonstrated SARS-CoV2 lack of susceptibility on swine and cattle, but not in whitetail deer <strong>(USDA, </strong><strong>Cornell University)</strong></li><br /> <li>Based on a reverse genetics system we created a virulent and pathogenic infectious clone of Senecavirus A <strong>(Cornell University, South Dakota State University, Universidade Federal de Pelotas)</strong></li><br /> <li>Targeting suicidal replication we created a PCV2 vaccine that enhance the safety of attenuated vaccines. <strong>(North Dakota State University, Iowa State University, South Dakota State University)</strong></li><br /> <li>We development of a blocking ELISA for detection of antibodies against ASFV <strong>(University of Illinois, Kansas State University, Iowa State University)</strong></li><br /> <li>Intestinal microbiota studies identified gut microbes associated with improved weight gain on pigs after immunization with PRRS MLV vaccine and co-challenge with PRRSV/PCV2, which may increase the efficacy of PRRS vaccination. <strong>(Kansas State University, Lawrence Livermore National Laboratory, Iowa State University)</strong></li><br /> <li>The first United States Swine Pathogen Database platform to integrate veterinary diagnostic laboratory sequence data was create to monitor emerging pathogens of swine and to combine and compare with sequences deposited in GenBank. <strong>(ARS, Iowa State University, Kansas State University, South Dakota State University, and Cornell University)</strong></li><br /> </ol>

Publications

<ol><br /> <li>Chaudhari J., Liew CS, Riethoven JJ, Sillman S., and Vu H., 2021. Porcine reproductive and respiratory syndrome virus infection upregulates negative immune regulators and T cell exhaustion markers. <em>J Virol. 2021 Oct 13;95(21):e0105221. doi: 10.1128/JVI.01052-21. Epub 2021 Aug 11.</em></li><br /> <li>Rakibuzzaman, A.; Pineyro, P.; Pillatzki, A.; Ramamoorthy, S. Harnessing the genetic plasticity of PCV2 to target suicidal replication. <em>Viruses </em> 13, 1676. <a href="https://doi.org/10.3390/v13091676">https://doi.org/10.3390/v13091676</a></li><br /> <li>Magnus R. Campler, Ting-Yu Cheng, Declan C. Schroeder, My Yang, Sunil K. Mor, Juliana B. Ferreira, Andr&eacute;ia. G. Arruda. A longitudinal study on PRRSV detection in swine herds with different demographics and PRRSV management strategies. Transboundary and Emerging Diseases. 2021 https://doi.org/10.1111/tbed.14386</li><br /> <li>Wang Y, Yim-im W, Porter E, Lu N, Anderson J, Noll L, Fang Y, Zhang J, Bai J. (2021). Development of a bead-based assay for detection and differentiation of field strains and four vaccine strains of type 2 porcine reproductive and respiratory syndrome virus (PRRSV-2) in the USA. <em>Transboundary and Emerging Diseases</em>. 68: 1414-1423.</li><br /> <li>Yuan F, Petrovan V, Gim&eacute;nez-Lirola L, Zimmerman J, Rowland RRR, Fang Y. (2021). Development of a Blocking Enzyme-Linked Immunosorbent Assay for Detection of Antibodies Against African Swine Fever Virus. Pathogens. 2021 Jun 17;10(6):760. doi: 10.3390/pathogens10060760.</li><br /> <li>Constance LA, Thissen JB, Jaing CJ, McLoughlin KS, Rowland RRR, Ser&atilde;o NVL,Cino-Ozuna AG, Niederwerder MC. Gut microbiome associations with outcome following co-infection with porcine reproductive and respiratory syndrome virus (PRRSV) and porcine circovirus type 2 (PCV2) in pigs immunized with a PRRS modified live virus vaccine. Vet Microbiol. 2021 Mar;254:109018. doi: 10.1016/j.vetmic.2021.109018. Epub 2021 Feb 16. PMID: 33639341.</li><br /> </ol><br /> <p>&nbsp;</p><br /> <p><strong><em>Full list of publications in the supplementary&nbsp; material section</em></strong></p><br /> <p>&nbsp;</p><br /> <p>&nbsp;</p><br /> <p><strong>Funding Leveraging via collaborative grants between stations and members.</strong></p><br /> <ol><br /> <li>Dr. RJ. Gourapura from <strong>The Ohio State University</strong> collaborated with Dr. D. Diel from <strong>Cornell University</strong> on: A Multi-Species Vaccine Delivery Platform for Infectious Disease Prevention and Control in Livestock. USDA-AFRI, 2017-67015-26909.</li><br /> <li>RJ . Gourapura and Dr. SP. Kenny from <strong>The Ohio State University, </strong>Dr. JK. Lunney and Dr. C. Loving from the <strong>USDA</strong>, and Dr. JM LaBresh collaborated on: Development of new swine reagents to broaden our understanding of immune correlates of protection and microbial pathogenesis. USDA-AFRI. ($500,000)</li><br /> <li>H. HogenEsch from <strong>Purdue University</strong> and Dr. RJ. Gourapura from <strong>The Ohio State University</strong>, collaborate on<strong>:</strong> Improving vaccine performance with novel phytoglycogen nanoparticle adjuvants</li><br /> <li>A. Arruda and M. Pairis-Garcia from <strong>The Ohio State University</strong>, Drs G. Almond and J. Ferreira from <strong>North Carolina State University</strong>, and Dr Declan Schroeder, C. Vilalta and SK. Mor from <strong>University of Minnesota</strong> collaborate on: Assessing within-herd PRRS variability and its impact on production parameters. National Pork Board. ($106,959)</li><br /> <li>S. Ramamoorthy, Dr. B. Webb from <strong>North Dakota State University </strong>and Dr. A. Pillatzki and from <strong>South Dakota State University</strong> collaborate on: Integrating vaccine efficacy and safety by directed suicidal replication. USDA NIFA AFRI &ndash; Animal Health and Disease. ( $500,000)</li><br /> <li>.S. Dee and Dr R. Cochrane from <strong>Pipestone Veterinary Service, </strong>A Shah from <strong>SAM Nutrition&rsquo;s</strong>, Dr. M. Niederwerder from <strong>Kansas State, University</strong>, Dr. E. Nelson, C. Jones, and D. Hanson from <strong>South Dakota State University</strong> collaborate on: Using a demonstration project to validate laboratory-based viral survival in feed: Phase 2. Swine Health Information Center. ($15,000)</li><br /> <li>CL. Miller from <strong>USDA</strong> and Dr. Y. Sang, Dr B. Lepenies and Dr. D. Fleming from <strong>Tennessee State University</strong> collaborate on: Validation of A Live-Virus Vaccine Candidate for Efficient Attenuation and Better Protection. USDA, 5030-32000-230-068-R. ($640,000)</li><br /> <li>Niederwerder M and Dr. Hefley T from <strong>Kansas State University</strong> and Dr G. Cino from <strong>Oklahoma State University</strong> collaborate on a National Pork Board and State of Kansas National Bio and Agro-defense Facility Fund ($178,808)</li><br /> <li>S. Ramamoorthy and Dr. B. Webb from <strong>North Dakota State University</strong> and Dr. A. Pillatzki from <strong>South Dakota State University</strong>, collaborate on: first response vaccines for emergency preparedness. USDA-NIFA. ($337,425)</li><br /> <li>S. Ramamoorthy and Dr. B. Webb from <strong>North Dakota State University</strong> and Dr. A. Pillatzki from <strong>South Dakota State University</strong>, collaborate on: first response vaccines for emergency preparedness. NIH-NIAID R21. ($398,749)</li><br /> <li>S. Ramamoorthy and Dr. B. Webb from <strong>North Dakota State University</strong> and Dr. A. Pillatzki from <strong>South Dakota State University</strong>, collaborate on: Integrating vaccine safety and efficacy by directed suicidal replication. USDA-NIFA. ($500,000)</li><br /> <li>D. Diel from <strong>Cornell University</strong> and Dr R. Gourapura form <strong>The Ohio State University</strong> collaborate on: Novel broadly protective swine influenza vaccine platforms. NIFA (Proposal 2021-06981. ($642,000)</li><br /> <li>H. Vu from <strong>University of Nebraska-Lincoln</strong> and Dr. P. Gauger from <strong>Iowa State University</strong> and Dr. H. Ly from <strong>University of Minnesota</strong> collaborate on: Development of a broadly protective vaccine against swine influenza virus. USDA-NIFA ($500,000)</li><br /> </ol><br /> <p>&nbsp;</p>

Impact Statements

  1. The major impacts from multi-state efforts included improvements in dissemination of information regarding trends in animal infections and diseases, role of the SARS-Cov2 in swine as pathogen, data generated through SDRS is used daily by practitioners to make relevant decisions regarding biosecurity and prevention strategies of endemic pathogens, and development of models and generating data to inform best practices on formulation and transport of animal feed to reduce the risk of FAD introduction via feed and feed ingredients, development of the PCV2 vaccine.
Back to top

Date of Annual Report: 01/27/2023

Report Information

Annual Meeting Dates: 12/02/2022 - 12/02/2022
Period the Report Covers: 12/05/2021 - 12/04/2022

Participants

NC229 Executive Meeting attendees (total = 17)
Roman Pogranichniy (Kansas State University), Diego Diel (Cornell University), Scott Kenney, Andreia Arruda, Renukaradhya Gourapura (The Ohio State University), Ying Fang, Federico Zuckermann, Dongwan Yoo, Raymond Rowland (University of Illinois), Hiep Vu (University of Nebraska), Pablo Pineyro-Pineiro, Phillip Gauger (Iowa State University), Laura Miller (USDA ARS Nation Animal Disease Center), Joan K. Lunney, (USDA-ARS Beltsville Agricultural Research Center), Declan Schroeder, Kim VanderWaal (University of Minnesota), Jonathan Pasternak (Purdue University).

Brief Summary of Minutes

 


Brief Summary of Minutes of Annual Business Meeting from 3:30 pm – 5:30 pm on 12/02/2022:


 


The 2022 NC229 Business Meeting was held on December 2nd, 2022, in conjunction with the NAPRRS/NC229: International Conference of Swine Viral Diseases at the Intercontinental Hotel. The meeting was open to all NC229 members. Annual reports from 15 stations were shared with the audience by representatives from each station. Fifty-three people attended the Business Meeting. The business meeting centered on the topics noted below-(detailed agenda in table 1 of report appendix):



  1. Pogranichniy (Chair) and Dr. Rowland (Academic Advisor) inaugurated the annual NC229 scientific meeting.

  2. Pogranichniy (Chair), Pineyro-Pineiro (Vice-Chair), and the scientific program committee were recognized for their outstanding efforts in organizing the scientific program.

  3. Pogranichniy (Chair) discussed the need to form a core group that will be in charge of writing the next renewal and the possibility of writing a sustainability grant proposal.

  4. Colby presented updates regarding funding opportunities for the upcoming year.

  5. Colby announced the renewal of the Dual Purpose with Dual Benefits: Research in Biomedical and Agriculture Using Agriculturally Important Domestic animals.

Accomplishments

<p>&nbsp;</p><br /> <h1>1. Accomplishment</h1><br /> <h1>Objective 1. CONTROL OF PRRSV:</h1><br /> <h2>1.1. PRRS immunology/vaccinology</h2><br /> <ol><br /> <li>Ribosome profiling of porcine reproductive and respiratory syndrome virus reveals novel features of viral gene expression.</li><br /> <li>Assessment of the potential role of the gut microbiota in the response of pigs to PRRSV-killed virus vaccination. This knowledge may pave the road for developing novel strategies to enhance vaccine efficacy<strong>.</strong></li><br /> <li>Developed a machine learning algorithm to predict cross-reactivity from genetic sequence data.</li><br /> <li>Defined mechanisms of immune evasion that contribute to PRRSV disease pathogenicity, which can be targeted through recombinant vaccines to improve vaccine efficacy.</li><br /> <li>Demonstrated that PRRSV-induced hypothyroidism is not directly responsible for changes in fetal developmental processes.</li><br /> </ol><br /> <h2>1.2. PRRS epidemiology</h2><br /> <ol><br /> <li>Evaluation of the influence of partial immunity in quasispecies evolution.</li><br /> <li>Investigation of risk factors associated with the incidence of wild-type PRRSV introductions into wean-to-finish herds located in the Midwest with an effort to improve biosecurity practices.</li><br /> <li>Investigation of PRRSV microevolution and diversity over time in PRRSV-positive</li><br /> <li>Assessed and modeled additional transmission routes (vehicles and feed) for PRRSV</li><br /> </ol><br /> <h2>1.3. PRRS Surveillance and Diagnostics.</h2><br /> <ol><br /> <li>Collaborative project among multiple VDLs, with the goal to aggregate swine diagnostic data and report in an intuitive format (web dashboards and monthly PDF report), describing <em>dynamics of pathogen detection by PCR-based assays over time, specimen, age group, and geographical area</em>.</li><br /> <li>Developed methods to rapidly detect and characterize the etiology of new and emerging viruses that may impact swine health.</li><br /> </ol><br /> <p>&nbsp;</p><br /> <h1>Objective 2 Developing effective and efficient approaches for detection, prevention, and control of pressing viral diseases of swine of recent emergence:&nbsp;</h1><br /> <h2>2.1. African Swine Fever Virus</h2><br /> <ol><br /> <li>Four Universities have been approved the Select Agent status by USDA-APHIS to conduct research on ASFV (University of Nebraska-Lincoln, The Ohio State University, Cornell University, and University of Minnesota).</li><br /> <li>Development of several vaccine candidates for African swine fever virus.</li><br /> <li>Developed a risk-free in situ non-animal surrogate assay to validate ASFV mitigation protocols.</li><br /> <li>Evaluated characteristics of supply chains for the transmission of foreign viral animal diseases and application of block-chain technology to trace imported ingredients.</li><br /> <li>In collaboration with the swine industry, monitoring the evolution of the global spread of ASF through the Swine Disease Global Surveillance project.</li><br /> <li>Validation of a simple and reliable method for profiling antibody response to ASFV.</li><br /> </ol><br /> <h2>2.2. Swine Influenza Virus</h2><br /> <ol><br /> <li>Development of new swine influenza vaccine candidates and establish the pregnant sow-fetus models to assess the safety and efficacy of influenza vaccines.</li><br /> <li>Investigated farm workers' roles in introducing seasonal influenza viruses into swine farms.</li><br /> <li>Investigated the genetic diversity of the influenza A virus in vaccinated pigs.</li><br /> <li>Conducted genetic and antigenic characterization of new and emerging swine IAVs, including phylogenetics and network analysis.</li><br /> <li>Identified the molecular mechanisms by which viruses infect and adapt to swine.</li><br /> <li>Evaluate and improve existing and new diagnostic tests and testing strategies for swine IAV surveillance, detection, and recovery from disease outbreaks.</li><br /> <li>Characterize swine innate and adaptive immune responses to swine IAVs and determine correlates of protection.</li><br /> </ol><br /> <h2>2.3&nbsp;&nbsp; Porcine Circovirus</h2><br /> <ol><br /> <li>Developed an infectious clone for PCV3.</li><br /> <li>Developed a co-infection model of PCV3 and PRRSV to test PCV3 and PRRSV vaccines in a dual-challenge</li><br /> </ol><br /> <h2>2.4 Swine Pestiviruses</h2><br /> <ol><br /> <li>Developed a new real-time PCR to detect atypical porcine pestivirus (APPV).</li><br /> </ol><br /> <h2>2.5. Senecavirus A (SVA)</h2><br /> <ol><br /> <li>Estimated the seroprevalence of ASV and assessed risk factors in the US swine industry.</li><br /> <li>Determined the minimum infectious dose of SVA in both neonates and market-weight pigs.</li><br /> <li>Assessed dynamics and duration of antibodies response to SVA in a breeding herd.</li><br /> <li>Identified viral genes associated with virulence and mechanisms of viral pathogenesis.</li><br /> <li>Conducted molecular characterization to predict the evolution of new SVA strains.</li><br /> <li>Evaluated new SVA vaccine platforms and determined whether vaccines against SVA would cross-react with FMDV or interfere with FMDV serological surveillance.</li><br /> </ol>

Publications

<ol><br /> <li>Barrera-Zarate, J., Detmer, S.E., Pasternak, J.A., Hamonic, G., MacPhee, D.J., Harding, J.C.S., 2022a. Detection of PRRSV-2 alone and co-localized with CD163 positive macrophages in porcine placental areolae. Vet Immunol Immunopathol 250, 110457.</li><br /> <li>Barrera-Zarate, J.A., Detmer, S.E., Pasternak, J.A., Hamonic, G., MacPhee, D.J., Harding, J.C.S., 2022b. Effect of porcine reproductive and respiratory syndrome virus 2 on angiogenesis and cell proliferation at the maternal-fetal interface. Vet Pathol 59, 940-949.</li><br /> <li>Chaudhari, J., Leme, R.A., Durazo-Martinez, K., Sillman, S., Workman, A.M., Vu, H.L.X., 2022a. A Single Amino Acid Substitution in Porcine Reproductive and Respiratory Syndrome Virus Glycoprotein 2 Significantly Impairs Its Infectivity in Macrophages. Viruses 14.</li><br /> <li>Chaudhari, J., Nguyen, T.N., Vu, H.L.X., 2022b. Identification of Cryptic Promoter Activity in cDNA Sequences Corresponding to PRRSV 5' Untranslated Region and Transcription Regulatory Sequences. Viruses 14.</li><br /> <li>Guidoni, P.B., Pasternak, J.A., Hamonic, G., MacPhee, D.J., Harding, J.C.S., 2022. Effect of porcine reproductive and respiratory syndrome virus 2 on tight junction gene expression at the maternal-fetal interface. Theriogenology 184, 162-170.</li><br /> <li>Yan X, Shang P, Yim-Im W, Sun Y, Zhang J, Firth AE, Lowe JF, <strong>Fang Y*</strong>. 2022. Molecular characterization of emerging variants of PRRSV in the United States: new features of the -2/-1 programmed ribosomal frameshifting signal in the nsp2 region. Virology. 573:39-49.</li><br /> <li>Yuan F, Sharma J, Nanjappa SG, Gaulke CA*,&nbsp;<strong>Fang Y*</strong>. 2022. Effect of Killed PRRSV Vaccine on Gut Microbiota Diversity in Pigs. Viruses. 14(5):1081.</li><br /> <li>Cook G. M., Brown, P. Shang,&nbsp; Y. Li,&nbsp; L. Soday,&nbsp; A. M. Dinan, C. Tumescheit, A. P. Mockett,&nbsp; Y. <strong>Fang*</strong>,&nbsp; A. E. Firth*,&nbsp; I. Brierley*. 2022. Ribosome profiling of porcine reproductive and respiratory syndrome virus reveals novel features of viral gene expression. Elife, 11:e75668.</li><br /> <li><strong>Zuckermann FA</strong>, Husmann R, Chen W, Roady P, Pfeiff J, Leistikow KR, Duersteler M, Son S, King MR, Augspurger NR.&nbsp;<em>Bacillus</em>-Based Direct-Fed Microbial Reduces the Pathogenic Synergy of a Coinfection with Salmonella enterica Serovar Choleraesuis and Porcine Reproductive and Respiratory Syndrome Virus. Infect Immun. 2022 Mar 7:e0057421.</li><br /> <li>Campler, M. R., Cheng, T., Schroeder, D. C., Yang, M., Mor, S. K., Ferreira, J. B., &amp; Arruda, A. G. (2022). A longitudinal study on PRRSV detection in swine herds with different demographics and PRRSV management strategies. <em>Transboundary and Emerging Diseases</em>. <a href="http://dx.doi.org/10.1111/tbed.14386">doi: 10.1111/tbed.14386</a></li><br /> <li>Schroeder, D. C., Odogwu, N. M., Kevill, J., Yang, M., Krishna, V. D., Kikuti, M., . . . Torremorell, M. (2021). Phylogenetically Distinct Near-Complete Genome Sequences of Porcine Reproductive and Respiratory Syndrome Virus Type 2 Variants from Four Distinct Disease Outbreaks at U.S. Swine Farms over the Past 6 Years. <em>Microbiology Resource Announcements, 10</em>(33). <a href="http://dx.doi.org/10.1128/mra.00260-21">doi: 10.1128/mra.00260-21</a></li><br /> <li>Paploski, I. A., Pamornchainavakul, N., Makau, D. N., Rovira, A., Corzo, C. A., Schroeder, D. C., . . . VanderWaal, K. (2021). Phylogenetic Structure and Sequential Dominance of Sub-Lineages of PRRSV Type-2 Lineage 1 in the United States. <em>Vaccines, 9</em>(6), 608. <a href="http://dx.doi.org/10.3390/vaccines9060608">doi: 10.3390/vaccines9060608</a></li><br /> <li>Pamornchainavakul N, Kikuti M, Paploski IAD, Makau DN, Rovira A, Corzo CA, et al. Measuring How Recombination Re-shapes the Evolutionary History of PRRSV-2: A Genome-Based Phylodynamic Analysis of the Emergence of a Novel PRRSV-2 Variant. Frontiers in Veterinary Science. 2022;9.</li><br /> <li>Ouyang H, Qiao Y, Yang M,&nbsp;Marabella IA,&nbsp;Hogan CJ,&nbsp;Torremorell M,&nbsp;Olson BA (2022). Single pass wind tunnel testing for recirculating virus aerosol control technologies. J of Aerosol Sciences, 165(2022) 106045.&nbsp;<a href="https://doi.org/10.1016/j.jaerosci.2022.106045">https://doi.org/10.1016/j.jaerosci.2022.106045</a></li><br /> <li>Kikuti M, Vilalta C, Sanhueza J, Melini CM, <strong>Corzo CA</strong>. Porcine reproductive and respiratory syndrome prevalence and processing fluids use for diagnosis in United States breeding herds. <em>Front Vet Sci</em>. Accepted for publication. 2022.</li><br /> <li>Kanankege KST, Graham K, <strong>Corzo C</strong>, VanderWaal K, Perez A, Durr P. Adapting an atmospheric dispersion model to assess the risk of windborne transmission of Porcine Reproductive and Respiratory Syndrome virus between swine farms. <em>Viruses</em>. Accepted for publication. 2022.</li><br /> <li>Moeller J, Mount J, Geary E, Campler MR, <strong>Corzo CA</strong>, Morrison RB, Arruda A. Investigation of the distance to slaughterhouses and weather parameters in the occurrence of porcine reproductive and respiratory syndrome outbreaks in U.S. swine breeding herds. <em>Can Vet J</em>. 2022. 63(5):528-534.</li><br /> <li>Galvis JA, <strong>Corzo CA</strong>, Machado G. Modeling and assessing additional transmission routes for porcine reproductive and respiratory syndrome virus: vehicle movement and feed ingredients. <em> Emerg. Dis. </em>2022. Doi: 10.111/tbed.14488.</li><br /> <li>Pamornchainavakul N, Kikuti M, Paploski IAD, Makau DN, Rovira A, <strong>Corzo CA</strong>, VanderWaal K. Measuring how recombination re-shapes the evolutionary history of PRRSV-2: a genome-based phylodynamic analysis of the emergence of a novel PRRSV-2 variant. <em>Front Vet Sci</em>. 2022. 9:846904. doi. 10.3389/fvets.2022.846904.</li><br /> <li>Kikuti M, Sanhueza J, Vilalta C, Paploski IAD, VanderWaal K, <strong>Corzo CA</strong>. Porcine reproductive and respiratory syndrome virus 2 (PRRSV-2) genetic diversity and occurrence of wild type and vaccine-like strains in the United States swine industry. <em>PLoS One</em>. 2021. 16(11). doi. 10.1371/journal.pone.0259531.</li><br /> <li>Kikuti M, Paploski IAD, Pamornchainavakul N, Picasso-Risso C, Schwartz M, Yeske P, Leuwerke B, Bruner L, Murray D, Roggow BD, Thomas P, Feldmann L, Allerson M, Hensch M, Bauman T, Sexton B, Rovira, VanderWaal K, <strong>Corzo CA</strong>. Emergence of a new lineage 1C variant of porcine reproductive and respiratory syndrome virus 2 in the United States. <em>Front Vet Sci</em>. 2021. 8:752938. doi. 10.3389/fvets.2021.752938.</li><br /> <li>Holtkamp D, Torremorell M, <strong>Corzo CA</strong>, Linhares DCL, Almeida MN, Yeske P, Polson DD, Becton L, Snelson H, Donovan T, Pittman J, Johnson C, Vilalta C, Silva GS, Sanhueza J. Proposed modifications to porcine reproductive and respiratory syndrome virus herds classification. <em>J Swine Health Prod</em>. 2021. 29(5):261-270.</li><br /> <li>Paploski IAD, Bhojwani RK, Sanhueza JM, <strong>Corzo CA</strong>, VanderWaal K. Forecasting viral disease outbreaks at the farm-level for commercial sow farms in the U.S. <em>Prev Vet Med</em>. 2021. 29. doi: 10.1016/j.prevetmed.2021.105449.</li><br /> <li>Almeida M, Zhang M, Lopez WAL, Vilalta C, Sanhueza J, <strong>Corzo CA</strong>, Zimmerman JJ, Linhares DCL. A comparison of three sampling approaches for detecting PRRSV in suckling piglets. <em>Prev Vet Med</em>. 2021. 194. doi: 10.1016/j.prevetmed.2021.105427.</li><br /> <li>Almeida M, <strong>Corzo CA</strong>, Zimmerman JJ, Linhares DCL. Longitudinal piglet sampling in commercial sow farms highlights the challenge of PRRSV detection. <em>Porcine Health Management</em>. 2021. 7:31. doi: 10.1186/s40813-021-00210-5.</li><br /> <li>Trevisan G, Linhares LCM, Schwartz KJ, Burrough ER, Magalhaes ES, Crim B, Dubey P, Main RG, Gauger P, Thurn M, Lages PTF,&nbsp;<strong>Corzo&nbsp;CA</strong>, Torrison J, Henningson J, Herrman E, McGaughey R, Cino G, Greseth J, Clement T, Christopher-Hennings J, Linhares DCL. Data standardization implementation and applications within and among diagnostic laboratories: integrating and monitoring enteric coronaviruses. <em>J Vet Diagn Invest</em>. 2021. Doi: 10.1177/10406387211002163.jvdi.sagepub.com</li><br /> <li>Galvis JA, Prada JM, <strong>Corzo CA</strong>, Machado G. Modeling the transmission and vaccination strategy for porcine reproductive and respiratory syndrome virus. <em> Emerg. Dis. </em>2021. Doi: 10.111/tbed.14007.</li><br /> </ol><br /> <ol><br /> <li>Fleming, D.S., Miller, L.C., Li, J., Lager, K.M., Van Geelen, A., Sang, Y. 2022. Transcriptomic analysis of liver indicates novel vaccine to porcine reproductive and respiratory virus promotes homeostasis in T-Cell and inflammatory immune responses compared to commercial vaccine in pigs. Frontiers in Veterinary Science. 9. Article 791034. <a href="https://doi.org/10.3389/fvets.2022.791034">https://doi.org/10.3389/fvets.2022.791034</a>.</li><br /> <li>Cheng, T.Y., Campler, M.R., Schroeder, D.C., Yang, M., Mor, S.K., Ferreira, J.B., Arruda, A.G., 2022. Detection of Multiple Lineages of PRRSV in Breeding and Growing Swine Farms. Front Vet Sci 9, 884733.</li><br /> <li>Moeller, J., Mount, J., Geary, E., Campler, M.R., Corzo, C.A., Morrison, R.B., Arruda, A.G., 2022. Investigation of the distance to slaughterhouses and weather parameters in the occurrence of porcine reproductive and respiratory syndrome outbreaks in U.S. swine breeding herds. Can Vet J 63, 528-534.</li><br /> <li>Guidoni, P.B., Pasternak, J.A., Hamonic, G., MacPhee, D.J., Harding, J.C.S., 2022. Effect of porcine reproductive and respiratory syndrome virus 2 on tight junction gene expression at the maternal-fetal interface. Theriogenology 184, 162-170.</li><br /> <li>Ison, E.K., Hopf-Jannasch, A.S., Harding, J.C.S., Alex Pasternak, J., 2022. Effects of porcine reproductive and respiratory syndrome virus (PRRSV) on thyroid hormone metabolism in the late gestation fetus. Veterinary research 53, 74.</li><br /> <li>Katwal, P., Aftab, S., Nelson, E., Hildreth, M., Li, S., Wang, X., 2022. Role of zinc metalloprotease (ZMPSTE24) in porcine reproductive and respiratory syndrome virus (PRRSV) replication in vitro. Archives of virology 167, 2281-2286.</li><br /> <li>Cui X, Xia D, Huang X, Sun Y, Shi M, Zhang J, Li G, Yang Y, Wang H, Cai X, An T. 2022. Recombinant characteristics based on 949 PRRSV-2 genomic sequences in 1991-2021 revealed viral multiplication ability contribute to the dominant recombination. <em>Microbiology Spectrum</em>. Sep 8: e02934-22.</li><br /> <li>Yim-im W, Huang H, Zheng Y, Li G, Rawal G, Gauger P, Krueger K, Main R, Zhang J. 2022. Characterization of PRRSV in clinical samples and the corresponding cell culture isolates. <em>Transboundary and Emerging Diseases</em>. 69: e3045-e3059.</li><br /> <li>Trevisan G, Zeller M, Li G, Zhang J, Gauger P, Linhares D. 2022. Implementing a user-friendly format to analyze PRRSV next-generation sequencing results and associating breeding herd production performance with a number of PRRSV strains and recombination events. <em>Transboundary and Emerging Diseases</em>. 69: e2214-e2229.</li><br /> <li>L&oacute;pez W, Zimmerman J, Gauger P, Harmon K, Magtoto R, Bradner L, Holtkamp D, Zhang M, Zhang J, Ramirez A, Linhares D, Gim&eacute;nez-Lirola L. 2022. Considerations in the use of processing fluids for the detection of PRRSV RNA and antibody. <em>Journal of Veterinary Diagnostic Investigation</em>. 34(5): 859-863.</li><br /> <li>Yuan X, Shang P, Yim-im W, Sun Y, Zhang J, Firth A, Lowe J, Fang Y. 2022. Molecular characterization of emerging variants of PRRSV in the United States: new features of the -2/-1 programmed ribosomal frameshifting signal in the nsp2 region. <em>Virology</em>. 573: 39-49.</li><br /> <li>Li P, Koziel JA, Zimmerman JJ, Zhang J, Cheng TY, Yim-im W, Jenks WS, Lee M, Chen B, Hoff SJ. 2022. Correction: Li, et al., Mitigation of airborne PRRSV transmission with UV light treatment: proof-of-concept. Agriculture 2021, 11, 259. <em>Agriculture</em>. 12(5): 680.</li><br /> <li>Rawal G, Yim-im W, Chamba F, Smith C, Okones J, Francisco C, Zhang J. 2022. Development and validation of a reverse transcription real-time PCR assay for specific detection of PRRSGard vaccine-like virus. <em>Transboundary and Emerging Diseases</em>. 69: 1212-1226.</li><br /> <li>Rupasinghe R, Lee K, Liu X, Gauger PC, Zhang J, Mart&iacute;nez-L&oacute;pez B. (2022). Molecular evolution of porcine reproductive and 1 respiratory syndrome virus field strains from 2 two swine production systems in the midwestern United States from 2001 to 2020. <em>Microbiology</em> <em>Spectrum</em>. 10(3): e0263421.</li><br /> </ol><br /> <p>&nbsp;</p><br /> <h2>African Swine Fever Virus</h2><br /> <ol start="42"><br /> <li>Shurson, G. C., Palowski, A., Ligt, J. L., Schroeder, D. C., Balestreri, C., Urriola, P. E., &amp; Sampedro, F. (2022). New perspectives for evaluating relative risks of African swine fever virus contamination in global feed ingredient supply chains. <em>Transboundary and Emerging Diseases, 69</em>(1), 31-56. <a href="http://dx.doi.org/10.1111/tbed.14174">doi: 10.1111/tbed.14174</a>&nbsp;</li><br /> <li>Shurson, G.C., Urriola, P.E., &amp; van de Ligt, J.L.G. 2021. Can we effectively manage parasites, prions, and pathogens in the global feed industry to achieve One Health? <em>Transboundary and Emerging Diseases 69</em>(1), 4-30. DOI:&nbsp;<a href="https://doi.org/10.1111/tbed.14205">1111/tbed.14205</a></li><br /> <li>Schambow, R., Sampedro, F., Urriola, P.E., van de Ligt, J.L.G., Perez, A., &amp; Shurson, G.C. 2021. Rethinking the uncertainty of African swine fever virus contamination in feed ingredients and risk of introduction into the United States. <em>Transboundary and Emerging Diseases</em> <em>69</em>(1),157-175. <a href="https://doi.org/10.1111/tbed.14358">https://doi.org/10.1111/tbed.14358</a></li><br /> <li>Dee, N., Havas, K., Shah, A., Singrey, A., Spronk, G., Niederwerder, M., Nelson, E., Dee, S., 2022a. Evaluating the effect of temperature on viral survival in plant-based feed during storage. Transbound Emerg Dis 69, e2105-e2110.</li><br /> <li>Dee, S., Shah, A., Jones, C., Singrey, A., Hanson, D., Edler, R., Spronk, G., Niederwerder, M., Nelson, E., 2022b. Evidence of viral survival in representative volumes of feed and feed ingredients during long-distance commercial transport across the continental United States. Transbound Emerg Dis 69, 149-156.</li><br /> <li>Luong, H.Q., Lai, H.T., Do, L.D., Ha, B.X., Nguyen, G.V., Vu, H.L., 2022. Differential antibody responses in sows and finishing pigs naturally infected with African swine fever virus under field conditions. Virus research 307, 198621.</li><br /> <li>Havas K, Gogin AE, Basalaeva JV, Sindryakova IP, Kolbasova OL, Titov IA, Lyska VM, Morgunov SY, Vlasov ME, Sevskikh TA, Pivova EY, Kudrjashov DA, Zimmerman S, Witbeck W, Gim&eacute;nez-Lirola LG, Nerem J, Spronk GD, Zimmerman JJ, Sereda AD. (2022).&nbsp; An Assessment of Diagnostic Assays and Sample Types in the Detection of an Attenuated Genotype 5 African Swine Fever Virus in European Pigs over a 3-Month Period.&nbsp; 2022 Mar 26;11(4):404. doi: 10.3390/pathogens11040404.</li><br /> </ol><br /> <p><strong>&nbsp;</strong></p><br /> <h2>Influenza</h2><br /> <ol start="49"><br /> <li>Joshi LR, Knudsen D, Pi&ntilde;eyro P, Dhakal S, Renukaradhya GJ, Diel DG. Protective Efficacy of an Orf Virus-Vector Encoding the Hemagglutinin and the Nucleoprotein of Influenza A Virus in Swine. Front Immunol. 2021 Nov 5;12:747574. <a href="https://doi.org/10.3389/fimmu.2021.747574">https://doi.org/10.3389/fimmu.2021.747574</a></li><br /> <li>Kumari, S., Chaudhari, J., Huang, Q., Gauger, P., De Almeida, M.N., Liang, Y., Ly, H., Vu, H.L.X., 2022. Immunogenicity and Protective Efficacy of a Recombinant Pichinde Viral-Vectored Vaccine Expressing Influenza Virus Hemagglutinin Antigen in Pigs. Vaccines (Basel) 10.</li><br /> <li>Li, C., Culhane, M. R., Schroeder, D. C., Cheeran, M. C.-J., Galina Pantoja, L., Jansen, M. L., &amp; Torremorell, M. (2022). Vaccination decreases the risk of influenza A virus reassortment but not genetic variation in pigs. <em>eLife, 11</em>. <a href="http://dx.doi.org/10.7554/elife.78618">doi: 10.7554/elife.78618</a>&nbsp;</li><br /> </ol><br /> <p>&nbsp;</p><br /> <ol start="52"><br /> <li>Lopez-Moreno G, Davies P, Yang M, Culhane MR, Corzo CA, Li C, Rendahl A, Torremorell M (2022). Evidence of influenza A infection and risk of transmission between pigs and farmworkers. Zoonoses and Public Health. Apr 20. doi: 10.1111/zph.12948. Epub ahead of print. PMID: 35445551.</li><br /> <li>de Lara AC, Garrido-Mantilla J, Lopez-Moreno G, Yang M, Barcellos DESN, Torremorell M (2022). Effect of pooling udder skin wipes on the detection of influenza A virus in preweaning pigs. Journal of Veterinary Diagnostic Investigation. 2022; 34(1):133-135. doi:10.1177/10406387211039462</li><br /> <li>Lopez-Moreno G, Garrido-Mantilla J, Sanhueza JM, Rendahl A, Davies P, Culhane M, McDowell E, Fano E, Goodell C, Torremorell M (2022). Evaluation of dam parity and interanal biosecurity practices in influenza infections in piglets prior to weaning. Prev Vet Med, 208:105764. doi: 10.1016/j.prevetmed.2022.105764.</li><br /> </ol><br /> <ol><br /> <li>Arendsee, Z.W., Chang, J., Hufnagel, D.E., Markin, A., Baker, A.L., Anderson, T.K. 2021. octoFLUshow: an interactive tool describing spatial and temporal trends in the genetic diversity of influenza A virus in U.S. swine. Microbiology Resource Announcements. 10(50). Article e01081-21. <a href="https://doi.org/10.1128/MRA.01081-21">https://doi.org/10.1128/MRA.01081-21</a>.</li><br /> <li>Neveau, M.M., Zeller, M.A., Kaplan, B.S., Souza, C.K., Gauger, P.C., Baker, A.L., Anderson, T.K. 2022. Genetic and antigenic characterization of an expanding H3 influenza A virus clade in US swine visualized by Nextstrain. mSphere. 7(3):e0994-21. <a href="https://doi.org/10.1128/msphere.00994-21">https://doi.org/10.1128/msphere.00994-21</a></li><br /> <li>Sharma, A., Zeller, M.A., Souza, C.K., Anderson, T.K., Baker, A.L., Harmon, K., Li, G., Zhang, J., Gauger, P.C. 2022. Characterization of a 2016-17 human seasonal H3 influenza A virus spillover now endemic to U.S. swine. mSphere. 7(1). Article e00809-21. <a href="https://doi.org/10.1128/msphere.00809-21">https://doi.org/10.1128/msphere.00809-21</a>.</li><br /> <li>Souza, C.K., Anderson, T.K., Chang, J., Venkatesh, D., Lewis, N.S., Pekosz, A., Shaw-Saliba, K., Rothman, R.E., Chen, K., Baker, A.L. 2022. Antigenic distance between North American swine and human seasonal H3N2 influenza A viruses as an indication of zoonotic risk to humans. Journal of Virology. 96(2). Article e01374-21. <a href="https://doi.org/10.1128/JVI.01374-21">https://doi.org/10.1128/JVI.01374-21</a>.</li><br /> <li>Markin, A., Wagle, S., Anderson, T.K., Eulenstein, O. 2022. RF-Net 2: Fast inference of virus reassortment and hybridization networks. Bioinformatics. 38(8). Pages 2144-2152. <a href="https://doi.org/10.1093/bioinformatics/btac075">https://doi.org/10.1093/bioinformatics/btac075</a>.</li><br /> <li>Kimble, B.J., Brand, M.W., Kaplan, B.S., Coyle, E.M., Chilcote, K., Gauger, P., Khurana, S., Baker, A.L. 2022. Vaccine-associated enhanced respiratory disease following influenza virus infection in ferrets recapitulates the model in pigs. Journal of Virology. 96(5). <a href="https://doi.org/10.1128/jvi.01725-21">https://doi.org/10.1128/jvi.01725-21</a>.</li><br /> <li>Sharma A, Zeller M, Souza C, Anderson T, Vincent A, Harmon K, Li G, Zhang J, Gauger P. 2022. <strong>Characterization of a 2016-2017 human-seasonal H3 influenza A virus spillover now endemic in United States swine.</strong> <em>mSphere</em>. 7(1): e00809-21.</li><br /> </ol><br /> <p>&nbsp;</p><br /> <h2>Porcine pestiviruses</h2><br /> <ol><br /> <li>Sutton, K.M., Eaton, C.W., Borza, T., Burkey, T.E., Mote, B.E., Loy, J.D., Ciobanu, D.C., 2022. Genetic diversity and detection of atypical porcine pestivirus infections. J Anim Sci 100. DOI:&nbsp;<a href="https://doi.org/10.1093/jas/skab360">10.1093/jas/skab360</a></li><br /> <li>&nbsp;Arruda B, Falkenberg S, Mora-D&iacute;az JC, Matias Ferreyra F, Magtoto R, Gim&eacute;nez-Lirola L.&nbsp; (2022).&nbsp; Development and evaluation of antigen-specific dual matrix Pestivirus K ELISAs using longitudinal known infectious status samples.&nbsp; J Clin Microbiol. 2022 Oct 12;e0069722. doi: 10.1128/jcm.00697-22.</li><br /> </ol><br /> <p>&nbsp;</p><br /> <h2>Senecavirus</h2><br /> <ol start="64"><br /> <li>Preis G, Sanhueza JM, Vilalta C, Vannucci F, Culhane MR, <strong>Corzo CA</strong>. National Senecavirus A seroprevalence and risk factors assessment for seropositivity. <em>Front Vet Sci</em>. Accepted for publication. 2022.</li><br /> </ol><br /> <ol><br /> <li>Devries, A.C., Lager, K.M. 2022. Efficacy of an inactivated Senecavirus A vaccine in weaned pigs and mature sows. Vaccine. 40(12):1747-1754. <a href="https://doi.org/10.1016/j.vaccine.2022.02.018">https://doi.org/10.1016/j.vaccine.2022.02.018</a>.</li><br /> <li>Hoffman, K., Humphrey, N., Korslund, J., Anderson, T.K., Faaberg, K., Lager, K.M., Devries, A.C. 2022. Characterization of Senecavirus A isolates collected from the environment of U.S. sow slaughter plants. Frontiers in Veterinary Science. 9. Article 923878. <a href="https://doi.org/10.3389/fvets.2022.923878">https://doi.org/10.3389/fvets.2022.923878</a>.&nbsp;</li><br /> <li>Devries, A.C., Lager, K.M. 2022. Infectious dose of Senecavirus A in market weight and neonatal pigs. PLoS ONE. 17(4). Article e0267145. <a href="https://doi.org/10.1371/journal.pone.0267145">https://doi.org/10.1371/journal.pone.0267145</a></li><br /> </ol><br /> <ol start="68"><br /> <li>Devries, A.C., Lager, K.M. 2022. Senecavirus A: Frequently asked questions. Swine Health and Production. 30(3):149-159. <a href="https://doi.org/10.54846/jshap/1270">https://doi.org/10.54846/jshap/1270</a>.</li><br /> <li>L, C.C., JC, G.N., Singrey, A., Niederwerder, M.C., Dee, S., Nelson, E.A., Diel, D.G., 2022. Stability of Senecavirus A in animal feed ingredients and infection following consumption of contaminated feed. Transbound Emerg Dis 69, 88-96.</li><br /> </ol><br /> <p><strong>&nbsp;</strong></p><br /> <h2>Coronavirus</h2><br /> <ol start="70"><br /> <li>Galvis JA, <strong>Corzo CA</strong>, Prada JM, Machado G. Modeling between-farm transmission dynamics of porcine epidemic diarrhea virus: characterizing the dominant transmission routes.<em> Prev Vet Med</em>. 2022. doi.org/10.1016/j.prevetmed.2022.105759.</li><br /> <li>Galvis JA, Prada JM, <strong>Corzo CA</strong>, Machado G. The between-farm transmission dynamics of Porcine Epidemic Diarrhea Virus: A short-term forecast modeling comparison and the effectiveness of control strategies.<em> Emerg. Dis. </em>2021. Doi: 10.111/tbed.13997.</li><br /> </ol><br /> <ol><br /> <li>Alhamo, M.A., Boley, P.A., Liu, M., Niu, X., Yadav, K.K., Lee, C., Saif, L.J., Wang, Q., Kenney, S.P., 2022. Characterization of the Cross-Species Transmission Potential for Porcine Deltacoronaviruses Expressing Sparrow Coronavirus Spike Protein in Commercial Poultry. Viruses 14.</li><br /> <li>Cruz-Pulido, D., Ouma, W.Z., Kenney, S.P., 2022. Differing coronavirus genres alter shared host signaling pathways upon viral infection. Sci Rep 12, 9744.</li><br /> <li>Kong, F., Wang, Q., Kenney, S.P., Jung, K., Vlasova, A.N., Saif, L.J., 2022. Porcine Deltacoronaviruses: Origin, Evolution, Cross-Species Transmission and Zoonotic Potential. Pathogens 11.</li><br /> <li>Martins, M., Boggiatto, P.M., Buckley, A., Cassmann, E.D., Falkenberg, S.M., Caserta, L.C., Fernandes, M.H., Kanipe, C.R., Lager, K.M., Palmer, M.V., Diel, D.G. 2022. From Deer-to-Deer: SARS-CoV-2 is efficiently transmitted and presents broad tissue tropism and replication sites in white-tailed deer. PLoS Pathogens. 18(3). Article e1010197. <a href="https://doi.org/10.1371/journal.ppat.1010197">https://doi.org/10.1371/journal.ppat.1010197</a>.</li><br /> <li>More-Bayona, J.A., Ramirez-Velasquez, M., Hause, B., Nelson, E., Rivera-Geronimo, H., 2022. First isolation and whole genome characterization of porcine deltacoronavirus from pigs in Peru. Transbound Emerg Dis 69, e1561-e1573.</li><br /> <li>Yen L, Mora-Diaz JC, Rauh R, Nelson W, Ye F, Zhang J, Baum D, Zimmerman J, Nelli R, Gim&eacute;nez-Lirola LG. (2022). Characterization of the subclinical infection of porcine deltacoronavirus in grower pigs under experimental conditions. <em>Viruses</em>. 14: 2144.</li><br /> <li>Schumacher L, Chen Q, Fredericks L, Gauger P, Bandrick M, Keith M, Gim&eacute;nez-Lirola LG, Magstadt D, Yim-im W, Welch M, Zhang J*. (2022). Evaluation of the efficacy of an S-INDEL PEDV strain, administered to pregnant gilts, against a virulent non-S-INDEL PEDV challenge in newborn piglets. <em>Viruses</em>. 14: 1801.</li><br /> <li>Yen L, Magtoto R, Mora-Diaz JC, Carrillo-&Aacute;vila JA, Zhang J, Cheng TY, Magtoto P, Nelli RK, Baum DH, Zimmerman JJ, Gim&eacute;nez-Lirola LG. (2022). The N-terminal subunit of the porcine deltacoronavirus spike recombinant protein (S1) does not serologically cross-react with other porcine coronaviruses. <em>Pathogens</em>. 11: 910.</li><br /> <li>&nbsp;Zhu J, Rawal G, Aljets E, Yim-im W, Yang YL, Huang YW, Krueger K, Gauger P, Main R, Zhang J*. (2022). Development and clinical applications of a 5-plex real-time RT-PCR for swine enteric coronaviruses. <em>Viruses</em>. 14: 1536.</li><br /> <li>Saeng-Chuto K; Madapong A; Kaeoket K; Pi&ntilde;eyro PE; Tantituvanont A; Nilubol D. Co-infection of porcine deltacoronavirus and porcine epidemic diarrhea virus induces early TRAF6-mediated NF-&kappa;B and IRF7 signaling pathways through TLRs. Scientific Reports 2022 Vol. 12 Issue 1. DOI: 10.1038/s41598-022-24190-w</li><br /> </ol><br /> <p>&nbsp;</p><br /> <h2>Other viruses</h2><br /> <ol start="82"><br /> <li>Li Y, Yuan F, Yan X, Matta T, Cino-Ozuna GA, <strong>Fang Y*.</strong> Characterization of an emerging porcine respirovirus 1 isolate in the US: A novel viral vector for expression of foreign antigens. Virology. 570:107-116.</li><br /> <li>Wu X, Hu Y, Sui C, Pan L, <strong>Yoo D</strong>, Miller LC, Lee C, Cong X, Li J, Du Y, Qi J. Multiple-Site SUMOylation of FMDV 3C Protease and Its Negative Role in Viral Replication. J Virol. 2022 Sep 14;96(17):e0061222.</li><br /> <li>Makau, D.N., Lycett, S., Michalska-Smith, M.&nbsp;<em>et al.</em>Ecological and evolutionary dynamics of multi-strain RNA viruses.&nbsp;<em>Nat Ecol Evol</em>&nbsp;<strong>6</strong>, 1414&ndash;1422 (2022). <a href="https://doi.org/10.1038/s41559-022-01860-6">https://doi.org/10.1038/s41559-022-01860-6</a></li><br /> </ol><br /> <ol><br /> <li>Cheng TY, Magtoto R, Henao-D&iacute;az A, Poonsuk K, Devries A, Van Geelen A, Lager K, Zimmerman J, Gim&eacute;nez-Lirola L. Detection of pseudorabies virus antibody in swine serum and oral fluid specimens using a recombinant gE glycoprotein dual-matrix indirect ELISA. J Vet Diagn Invest. 2021 Nov;33(6):1106-1114. <a href="about:blank">http://doi:10.1177/10406387211040755</a></li><br /> <li>Manirarora, J.N., Walker, K.E., Patil, V., Renukaradhya, G.J., LaBresh, J., Sullivan, Y., Francis, O., Lunney, J.K., 2022. Development and Characterization of New Monoclonal Antibodies Against Porcine Interleukin-17A and Interferon-Gamma. Frontiers in immunology 13, 786396.</li><br /> <li>Nelsen, A., Lager, K.M., Stasko, J., Nelson, E., Lin, C.M., Hause, B.M., 2022. Identification of Pulmonary Infections With Porcine Rotavirus A in Pigs With Respiratory Disease. Front Vet Sci 9, 918736.</li><br /> <li>Zhang Q, Rawal G, Qian J, Ibrahim H, Zhang J*, Liang D*, Lu M. (2022). An integrated magneto-opto-fluidic biosensor for rapid on-chip assay of respiratory viruses of livestock. <em>Lab on a Chip</em>. 22(17): 3236-3244.</li><br /> <li>Welch M, Harmon K, Zhang J, Pi&ntilde;eyro P, Magtoto R, Wang C, Gim&eacute;nez-Lirola LG, Strait E, Mogler M, Gauger P. (2022). Detection of porcine parainfluenza virus type-1 antibody in swine serum using a whole-virus ELISA, indirect fluorescence antibody and virus neutralizing assays. <em>BMC Veterinary Research</em>. 18(1): 110.</li><br /> <li>Shen H, Zhang J, Gauger P, Burrough E, Zhang J, Harmon K, Wang L, Zheng Y, Petznick T, Li G. (2022). Genetic characterization of porcine sapoviruses identified from pigs during a diarrhea outbreak in Iowa, 2019. <em>Transboundary and Emerging Diseases</em>. 69: 1246-1255.</li><br /> <li>Anderson TK, Inderski B, Diel DG, Hause BM, Porter E, Clement T, Nelson EA, Bai J, Christopher-Hennings J, Gauger PC, Zhang J, Harmon KM, Main R, Lager KM, Faaberg KS. (2021). The United States Swine Pathogen Database: integrating veterinary diagnostic laboratory sequence data to monitor emerging pathogens of swine. <em>DATABASE</em>. 2021: 1-9.</li><br /> <li>&nbsp;Kroeger M; Temeeyasen G; Pi&ntilde;eyro PE. Five years of porcine circovirus 3: what have we learned about the clinical disease, immune pathogenesis, and diagnosis. Virus Res 2022. 314: 198764. DOI: 10.1016/j.virusres.2022.198764</li><br /> <li>Welch M; Krueger K; Zhang J; Pi&ntilde;eyro P; Magtoto R; Wang C; Gim&eacute;nez-Lirola L; Strait E; Mogler M; Gauger P. Detection of porcine parainfluenza virus type-1 antibody in swine serum using whole-virus ELISA, indirect fluorescence antibody and virus neutralizing assays. BMC Veterinary Research 2022. 18 (1):110. DOI: 10.1186/s12917-022-03196-6</li><br /> <li>Cheng T, Zimmerman J, Gim&eacute;nez-Lirola LG.&nbsp; (2022).&nbsp; Internal reference genes with the potential for normalizing quantitative PCR results for oral fluid specimens.&nbsp; Anim Health Res Rev. 2022 Nov 4:1-10. doi: 10.1017/S1466252322000044. Epub ahead of print. PMID: 36330795.</li><br /> </ol><br /> <p>&nbsp;</p><br /> <p>&nbsp;</p><br /> <h2>Book Chapters or Monographs</h2><br /> <ol><br /> <li>Fang Y., Snijder E. J., &amp; Balasuriya U.B. 2022. Chapter 5. Arterivirus. In P. Howley, &amp; D. Knipe (Ed.). Fields Virology: RNA Viruses (7th Edition), Wolters Kluwer Production.</li><br /> <li>Circoviridae. Pablo Pi&ntilde;eyro and Sheela Ramamoorthy. In: Veterinary Microbiology, 4th Edition. Ed. D. Scott McVey, Melissa Kennedy, M.M. Chengappa, and Rebecca Wilkes. 2022.</li><br /> <li>Kennedy M, Delhon G, McVey DS, <strong>Vu H</strong>, and Borca M. 2021. Chapter 49: Asfarviridae and Iridoviridae. In <em>Veterinary Microbiology</em>, Fourth ed.; McVey, S., Kennedy, M., M.M. Chengappa, M.M., Wilkes, R., Eds. Wiley Blackwell: 2022.</li><br /> </ol><br /> <p>&nbsp;</p>

Impact Statements

  1. The major impacts from multi-state efforts included improvements in dissemination of information regarding trends in animal viral infections and diseases, highlighted topics such as PRRSV, African swine fever, ASV, IAV, risk assessment and biosecurity role in swine production systems. The data generated through MSHMP is used daily by practitioners to make relevant decisions regarding biosecurity and prevention strategies of endemic pathogens, and development of models and generating data. New strategies have been developed for ASF, ASV and PCV3 vaccines and tested in the field.
Back to top

Date of Annual Report: 01/08/2024

Report Information

Annual Meeting Dates: 11/30/2023 - 11/30/2023
Period the Report Covers: 12/05/2022 - 12/04/2023

Participants

Brief Summary of Minutes

Accomplishments

Publications

Impact Statements

Back to top
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.