SAES-422 Multistate Research Activity Accomplishments Report

Status: Approved

Basic Information

Participants

See meeting minutes for list of participants.

See attached meeting minutes.

Accomplishments

Objective 1: To evaluate the prevalence of viral and bacterial agents of respiratory disease by developing, validating and disseminating new state-of-the-art molecular diagnostics for rapid identification of these agents. Short term outcomes: Commercially available assays for human respiratory syncytial virus (RSV) were shown to be effective for use in identification of bovine RSV (BRSV). The infectious agents associated with fatal bovine respiratory disease (BRD) in feedlots were characterized, providing a much-needed updating of our understanding of the pathogens involved in fatal feedlot BRD, and the relationship between specific infectious agents and specific pathologic lesions. The prevalence of different genotypes of bovine viral diarrhea virus (BVDV) in U.S. and Australian diagnostic lab isolates was determined; it was found that there is a difference in the genotypes most prevalent in the two countries. Annual summaries of the number of major pathogens isolated from BRD cases presented to regional diagnostic laboratories were disseminated to the membership. Outputs: An annual summary of the frequency of isolation of major pathogens from BRD cases presented to regional diagnostic laboratories was made available to the membership. Research abstracts were presented that demonstrated that a commercial assay for human RSV can be used to identify BRSV and which described the prevalence of various genotypes of BVDV. Research papers were published which described the prevalence of various BVDV genotypes in U.S. cattle populations. Activities: Project members worked together to share prevalence data on pathogens isolated in BRD cases presented to regional diagnostic laboratories, and to characterize the distribution of BVDV genotypes in various U.S. cattle populations. Objective 2: To investigate the basic biology, molecular pathogenesis, and immunopathogenesis of polymicrobial infections including important viral and bacterial agents. Short term outcomes: An in vitro system was developed which allows assessment of the interaction of infected bovine respiratory epithelial with cells of the immune response, providing a method of studying airway epithelial cells, which because of restrictions of anatomy and physiology cannot be studied in the living animal. Immune cells from cattle infected with Mannheimia haemolytica (M. haemolytica) were shown to produce increased amounts of substance P, which likely contributes to increased vascular leakage and edema in the lungs of infected cattle. Proteogenomic mapping was developed to better characterize the relationship between genes, the proteins they encode, and the function of those proteins for M. haemolytica. Proteomic analysis was used to evaluate the mechanism by which BVDV interferes with function of dendritic cells, which initiate the immune response to the virus; acute phase protein signaling was most commonly modified. A molecule which may be among the most important proteins regulating bovine herpesvirus-1 (BHV-1) replication was evaluated extensively and was shown to interfere with host immune function; the means by which this molecule interacts with host cell proteins to enhance BHV-1 replication was also deduced. The methods by which BHV-1 establishes latency and reactivates from latency (which is among the most potent of the virus disease inducing strategies) were characterized extensively. Methods of typing Pasteurella multocida (P. multocida) strains in order to distinguish between commensal and virulent isolates were developed. A model of BVDV and M. haemolytica co-infection utilizing exposure to naturally PI cattle was validated, and co-infection was shown to impact metabolism and productivity of feedlot cattle. A method of evaluating the effect of BVDV on development of immunity in the fetal calf was developed which utilizes a strategy to isolate immune cells from the fetal liver. The impact of BVDV on the cellular structure of macrophages, a critical immune cell, was determined. Research was undertaken which demonstrated the interaction of M. haemolytica leukotoxin with mitochondria, revealing a new pathogenic mechanism by which the leukotoxin causes disease. The role of neutrophil DNA traps (or nets) in the control of M. haemolytica was determined. Outputs: Presentations were made to veterinarians at annual veterinary continuing education conferences on the latest information regarding BRSV, Mycoplasma bovis (M. bovis), and on vaccination to control respiratory disease in cattle. Research abstracts were presented that demonstrated the interaction between infected bovine respiratory epithelial cells and immune cells, which demonstrated how molecular transport systems are related to pathogenicity of M. bovis; which demonstrated the relationship between quantitative and qualitative load of bacterial pathogens and respiratory disease; that provided information on proteogenomic mapping of the M. haemolytica genome; that showed that cytopathic BVDV and noncytopathic BVDV differentially modulate induction of the host immune response; that described the effect of BHV-1 infection of airway epithelial cells to increase activation of the host immune response; and that described the region of the M. haemolytica leukotoxin which specifically interacts with mitochondria. Research papers were published which demonstrated the effect of M. bovis to suppress leukocyte antibacterial responses; which demonstrated the effect of M. bovis on growth and health in feedlot cattle, which demonstrated that BVDV interferes with the initiation of the host immune response by impairing the function of professional antigen presenting cells; which showed how a protein encoded by BHV-1 impairs the host interferon response and thus counteracts host immunity; which demonstrated how BHV-1 undergoes latency and reactivates from latency; which reported the current relationship between infectious agents and various pathologic agents in feedlot cattle; which reported the knowledge gaps impacting the development of BVDV control programs; which outlined methods for identifying pathogenic vs. commensal P. multocida isolates; which outlined the current evidence regarding risk factors and effective control measures in BRD; and which presented the results of immunoproteomic analysis of M. haemolytica outer membrane proteins. Activities: Project members worked to test the effect of various vaccination strategies on productivity and health of feedlot cattle, and to disseminate this information to veterinarians and producers. Work on the Genetics of Feedlot Health Project was continued, which evaluated the role of genetics, nutrition, behavior, and infection and immunity on the occurrence of feedlot respiratory disease and carcass quality. Objective 3: To develop management and prevention strategies that incorporate new vaccines and treatment protocols to combat bovine respiratory disease and reduce economic loss. Short term outcomes: A research trial was completed which evaluated the response of young calves to intranasal or subcutaneous vaccination to improve immunity to respiratory infections; the data indicated that one commercial vaccine induced better immunity when given subcutaneously. A gene deleted M. bovis vaccine was developed which could be given intranasally and did not disseminate to other sites in the body, indicating that it was stable and safe. Seroconversion to M. bovis was found to be associated with decreased growth of feedlot cattle. It was found that subjective and laboratory indicators of disease were not good measures of disease progression, but measures of activity (by pedometers) showed promise in providing accurate assessment of disease progression. A research trial was carried out which confirmed that vaccination of calves before weaning improved their health and productivity when they were exposed to cattle persistently infected (PI) with BVDV in the feedlot. Immunoproteomic analysis was used to identify outer membrane proteins of M. haemolytica which should form the basis for improved vaccines. A research trial was completed which showed that vaccination once was as effective as vaccination twice to prevent respiratory disease in feedlot cattle; however, cattle vaccinated twice had improved feed efficiency. Outputs: Presentations were made to veterinarians and producers at continuing education meetings on the accurate prediction of respiratory disease risk based on daily morbidity and mortality counts, and on the approach to eradicating BVDV in a region (Upper Peninsula of Michigan). Research abstracts were presented that compared the results of vaccinating calves intranasally or subcutaneously to improve immunity to respiratory infection; that demonstrated how genetic modifications can be applied to the development of M. bovis vaccines; that demonstrated that vaccination mitigates the adverse health effects of exposure to BVDV PIs in feedlots; that demonstrated the feasibility of BVDV eradication on a regional level; that described the degree to which whitetail deer infected with BVDV can spread disease to cattle; and that described a method for isolating fetal bovine immune cells from the liver. Research papers were published that presented the impact of various vaccine-parasiticide combinations on health and productivity in beef cattle, that demonstrated the use of rumen temperature reporting boluses to monitor fever in cattle co-infected with BVDV and M. haemolytica; and which demonstrated the efficacy of a new intranasal M. haemolytica vaccine containing choleratoxin as an adjuvant. Activities: Project members worked to assess how vaccination strategies and evaluation of physiologic and behavioral indicators could be used to improve health and productivity of cattle in field settings. A project to eradicate BVDV in the UP of MI continued to progress; producers were engaged to work with project researchers to identify and remove persistently infected cattle and to carry out surveillance and management practices to advance the project. Plans for the coming year: This is the final year of the current project; major plans are to continue to undertake surveillance of the rate of isolation of major BRD pathogens by regional diagnostic laboratories, to cooperate in research that provides foundational knowledge regarding the basic mechanisms by which infectious BRD agents interact with the bovine immune response, and to test and disseminate to veterinarians and producers practices that can be translated to the field to decrease the impact of BRD in U.S. cattle. NC-1027 will also hold its annual meeting in conjunction with the American Association of Bovine Practitioners (AABP) Convention in St. Louis on September 21-22. NC-1027 has met in conjunction with AABP for several years, and this has provided an excellent opportunity for the Project to inform veterinarians of new developments in the science of BRD control, and to learn from veterinarians the current problems on which research should be focused. In 2010 NC-1027 met in conjunction with AFRI awardees conducting research on BRD; this also provided a new opportunity for improved collaboration between scientists working on BRD from different perspectives. In 2011 NC-1027 is again planning to meet in conjunction with AFRI awardees and to offer the meeting as a pre-conference seminar billed as Cutting Edge Science Related to BRD. This will allow veterinarians and other AABP attendees the opportunity to hear the latest developments in BRD prevention and control, and will allow NC-1027 members and AFRI awardees the opportunity to discuss questions and receive input from veterinarians working in the field. Members of NC-1027 have also been in contact with the investigators leading the new BRD CAP; plans are underway to coordinate efforts between NC-1027 and the CAP to develop impactful outreach efforts that will deliver information from CAP activities to veterinarians, scientists in industry and academia, and producers. Cooperation between NC-1027 and the CAP should lead to a high level of impact, as the CAP scientists will benefit from the historical perspective and experience of NC-1027, and the established links NC-1027 has developed with AABP, the Academy of Veterinary Consultants, the National Cattlemens Beef Association, and the American Association of Veterinary Laboratory Diagnosticians.

Impacts

  1. Ongoing work by the Committee to track the rate of isolation of major pathogens from BRD cases presented to regional diagnostic laboratories provides year-to-year surveillance on changes in the impact of various agents, and on the development of new or emerging pathogens. For example, increased surveillance of respiratory coronavirus which has occurred in recent years will allow scientists to determine the relative importance of this agent as an emerging cause of BRD.
  2. Characterization of the prevalence of different BVDV genotypes in U.S. cattle populations will allow scientists to determine the degree to which BVDV is evolving over time, and to determine whether current vaccines are formulated with appropriate genotypes to optimally control disease in U.S. cattle.
  3. The validation of a patient-side test for human RSV in the diagnosis of BRSV provides the basis for more rapid diagnosis of respiratory pathogens; this effort represents a direct response to stakeholders, who have repeatedly expressed a need for more rapid tests to diagnose BRD.
  4. The in vitro system developed to allow assessment of the interaction of infected bovine respiratory epithelial with cells of the immune response provides a method of studying the responses of living airway epithelial cells, which because of restrictions of anatomy and physiology cannot be studied in the living animal
  5. The effort to develop proteogenomic mapping of M. haemolytica will provide a valuable new tool for researchers attempting to better understand how the genome of a major pathogen translates to its disease-causing phenotype.
  6. The proteomic analysis of immune cells infected with either cytopathic or noncytopathic BVDV provides foundational knowledge regarding a major bovine pathogen. The data obtained will thus provide the basis for future studies to determine targets for vaccines and therapeutics to minimize the negative effects of BVDV on bovine health and productivity.
  7. The research to determine the molecular mechanisms by which BHV-1 impairs the host response and undergo latency are providing important foundational knowledge that is improving the ability of scientists to elucidate basic functions of an important viral pathogen. The information on latency will help improve understanding not only of BHV-1 but also of other herpesviral diseases which impair the health and productivity of agricultural animals, because all herpesviruses undergo latency and latency is currently poorly understood.
  8. The new model of experimental BRD which utilizes field exposure to naturally BVDV PI cattle followed by M. haemolytica challenge will provide a real-life model for testing vaccines and management strategies designed to minimize the negative impacts of BVDV and BRD.
  9. Work to determine the impact of BVDV on the development of immunity in the fetus will not only provide foundational knowledge regarding the interaction of infectious agents with the developing bovine immune system, but the model will also be useful for evaluating the pathogenesis of other diseases that impact bovine health and productivity by causing fetal disease and death
  10. The research demonstrating that M. haemolytica leukotoxin causes cell damage in part through interaction with mitochondria provides foundational knowledge regarding a new mechanism of pathogenesis by a major BRD pathogen; it also provides a basis for the development of methods which could mitigate the damaging effect of leukotoxin on bovine immune cells.
  11. The research describing the role of neutrophil traps (nets) in host resistance to M. haemolytica provides foundational knowledge regarding the immune response to bacterial pathogens; this could form the basis for future studies to develop therapies based on molecules which could bind to and inactivate M. haemolytica.
  12. Early diagnosis of BRD remains a challenge for producers, particularly feedlot producers; thus, the research evaluating the use of clinical signs, laboratory tests, and measures of activity to identify BRD will provide valuable new information to veterinarians and producers which should improve early diagnosis of BRD and thus improve response to treatment.
  13. The gene deleted strain developed by project researchers provides a new platform for the development of safe and effective vaccines for M. bovis.
  14. The information gained from the trial evaluating the effect of respiratory vaccines given to young calves by either the intranasal or subcutaneous routes will help veterinarians make decisions about when to most effectively administer vaccines, and to choose the best route of administration.
  15. The BVDV eradication project being undertaken in the UP of Michigan provides an invaluable test case on the feasibility of eradicating BVDV from the national herd, an effort which has been advocated by the Academy of Veterinary Consultants and other stakeholder groups. This project provides a unique opportunity to educate producers and to develop partnerships with various entities to meet a common goal of improving cattle health.
  16. The research proving that the impact of exposure to BVDV can be mitigated by vaccination prior to feedlot entry provides producers with a valuable tool to counteract the impact of BVDV while it remains present in the U.S. herd.
  17. The characterization, cloning, and production of subunit components of M. haemolytica and P. multocida offer opportunity for the development of improved vaccines based on the most current concepts in bacterial pathogenesis to control BRD.
  18. Research testing the effect of single vaccination versus vaccination plus booster on health and performance of feedlot cattle will help veterinarians and producers decide whether two doses of vaccine are warranted in managing high risk cattle.
  19. Improved understanding of the interplay of genetics, nutrition, and infection in feedlot respiratory disease will provide the possibility of selecting cattle resistant to BRD, or tailoring management strategies to the phenotype of different cattle groups to improve BRD control.

Publications

Ammari, M. G., McCarthy, F. M., Nanduri, B., Pinchuk, L.M. Analysis of Bovine Viral Diarrhea Viruses-infected monocytes: identification of cytopathic and non-cytopathic biotype differences. BMC Bioinformatics. 11 SUPPL, 2010, MCBIOS, in press. Atapattu DN, Aulik NA, McCaslin DR, Czuprynski CJ. 2009. Brief heat treatment increases cytotoxicity of Mannheimia haemolytica leukotoxin in an LFA-1 independent manner. Microb Pathog. 46:159-165. Ayalew S, Step DL, Montelongo M, Confer AW. Intranasal vaccination of calves with Mannheimia haemolytica chimeric protein containing the major surface epitope of outer membrane lipoprotein PlpE, the neutralizing epitope of leukotoxin, and cholera toxin subunit B. Vet Immun & Immunopathol 132: 295-302, 2009 Ayalew S, Confer AW, Hartson SD, Shrestha B. Immunoproteomic Analysis of Outer Membrane Proteins of Mannheimia haemolytica and Identification of Potential Vaccine Candidates. Proteomics 10: 2151-2164, 2010 Brum M, Coats C., Sangena B.R, Doster.A., Jones, C. and Chowdhury.S.I. (2009). Role of Envelope proteins gE in the anterograde transport of BHV-1 following reactivation in the Trigeminal Ganglia. J. Neuro virology. 15 (2): 196-201. Brum. M.C.S., C. Coats, B.R. Sangena, A. Doster, C. Jones, and S.I. Chowdhury. 2009. Bovine herpesvirus type 1 (BoHV-1) anterograde neuronal transport from trigeminal ganglia to nose and eye requires glycoprotein E. J. Neurovirology, 15:1-6. Burciaga-Robles, L.O., Step, D.L., Krehbiel, C.R., Holland, B.P., Richards, C.J., Montelongo, M.A., Confer, A.W., Fulton, R.W.: Exposure by Persistently Infected Calves with Bovine Viral Diarrhea Virus Type 1b and Subsequent Infection with Mannheimia haemolytica Demonstrating Effects on Clinical Signs and Immune Parameters: Model for Bovine Respiratory Disease Via Viral and Bacterial Interaction. Journal of Animal Science.88: 2166-2178, 2010. Burciaga-Robles, L.O., Krehbiel, C.R., Step, D.L., Holland, B. P., Richards, C.J., Montelongo, M.A., Confer, A.W., Fulton, R.W.: Effects of Exposure to Calves Persistently Infected with Bovine Viral Diarrhea Virus Type 1b and Mannheimia haemolytica Challenge on Animal Performance, N Balance, and Visceral Organ Mass in Beef Steers. Journal of Animal Science. 88:2179-2188, 2010. Burge, L.J., Welsh, R.D., Johnson, B.J., Reck. A.: Lung Pathology and Infectious Agents in Fatal Feedlot Pneumonias and Relationship with Mortality, Disease Onset, and Treatments. Journal of Veterinary Diagnostic Investigation 21: 464-477, 2009. Chowdhury, S. and C. Jones. Bovine herpesvirus type 1 (BHV-1) is an important cofactor in the bovine respiratory disease complex. Veterinary Clinics of North America: IN PRESS, Food Animal Practice. Confer AW. Update on bacterial pathogenesis in BRD. Animal Health Res Rev 10: 145-149: 2009. Confer AW, Ayalew S, Step D L, Trojan B, Montelongo M. Intranasal vaccination of young Holstein calves with Mannheimia haemolytica chimeric protein PlpE-LKT (SAC89) and cholera toxin Vet Immun & Immunopathol 132: 232-236, 2009 Czuprynski, C.J. 2009. Host response to bovine respiratory pathogens. Anim Health Res Rev. 2009 10:141-143. Ellis, J., S. Gow, N. Goji, C. Jones, A. Workman, G. Henderson, G. Alaniz, and T. Meinert. 2009. Efficacy of a combination viral vaccine in protecting cattle from experimental infection with bovine herpesviruses-1 isolated from recent vaccine breaks. J of American Veterinary Medical Association, 235:563-572. Fulton, R.W., Whitley, E.M., Johnson, B.J., Ridpath, J.F., Kapil, S., Burge, L.J., Cook, B.J., Conf er, A.W.: Prevalence of Bovine Viral Diarrhea Virus (BVDV) in Persistently Infected Cattle and BVDV Subtypes in Affected Cattle in Beef Herds in South Central United States. Canadian Journal for Veterinary Research 73: 283-291, 2009. Fulton, R.W.: Bovine Respiratory Disease Research: 1983-2009. Animal Health Research Reviews. 10: 131- 140, 2009. Hanzlicek G, White B, Anderson D, Mosier D, Renter D. Pathological and physiological changes following induced Mannheimia pneumonia in beef feeder calves. Amer J Vet Res. Mar 2010. 71(3):359-369. Hanzlicek G, White B, Renter D, Blasi D. A field study evaluating health, performance, and behavior differences in crossbred beef calves administered different vaccine-parasiticide product combinations. Vaccine. In press. Henningson, J.L., C.L.Topliff, L.H.Gil, R.O.Donis, D.J.Steffen, K.M.Eskridge, C.L.Kelling. 2009. Influence of Npro on BVDV virulence and interferon type induction in calves. American Journal of Veterinary Research 70: 1117-1123. Jaber, T., A. Workman, and C. Jones. Small non-coding RNAs encoded within the bovine herpesvirus 1 latency related gene can reduce steady state levels of infected cell protein 0 (bICP0). 2010. J Virology, 84: 62976307 Jones, C. 2009. Regulation of Innate Immune Responses by Bovine Herpesvirus 1 and Infected Cell Protein 0 (bICP0). Viruses 1:255-275. Jones C, Chowdhury S (2010). Bovine herpesvirus type 1 (BHV-1) is an important cofactor in the bovine respiratory disease complex. Vet. Clin. Food Anim. Practice. (In Press). Kisiela DI, Czuprynski CJ.2009. Identification of Mannheimia haemolytica adhesins involved in binding to bovine bronchial epithelial cells. Infect Immun. 2009 77:446-455. Kisiela DI, Aulik NA, Atapattu DN, Czuprynski ,CJ. 2010. N-terminal region of Mannheimia haemolytica leukotoxin serves as a mitochondrial targeting signal in mammalian cells. Cell. Microbiol. 12:976-987. Lee, S.-R., Nanduri, B., Pharr, G. T., Stokes, J. V., Pinchuk, L. M. 2009. Bovine Viral Diarrhea Virus infection affects the expression of proteins related to professional antigen presentation in bovine monocytes. Biochim. Biophys. Acta. 1794:14-22. Leyh, R.D., Fulton, R.W., Stegner, J.E., Goodyear, M., Witte, S., Taylor, L.P., Johnson, B.J., Step, D.L., Ridpath, J.F., Holland, B.P.: Fetal Protection in Heifers Vaccinated with Modified Live Virus Vaccine Containing Bovine Viral Diarrhea Virus Subtypes BVDV1a and BVDV2a after Challenge with BVDV1b Persistently Infected Cattle. Accepted for publication, 2010. American Journal for Veterinary Research. Liang, D., L. Chen, I.H. Ansari, L. H. Gil, C.L. Topliff, C.L. Kelling, R.O.Donis. 2009. A replicon trans-packing system reveals the requirement of nonstructural proteins for the assembly of bovine viral diarrhea virus (BVDV) virion. Virology 387:331-340. McClenahan D, Hillenbrand K, Kapur A, Carlton D, Czuprynski C. 2009. Effects of extracellular ATP on bovine lung endothelial and epithelial cell monolayer morphologies, apoptoses, and permeabilities. Clin Vaccine Immunol. 16:43-48. Meyer, F. and C. Jones. 2009. C/EBP-alpha cooperates with bTIF to activate the bovine herpesvirus 1 immediate early transcription unit 1 promoter. J. Neurovirology 15:123-130. Panciera RJ, Confer AW. Pathogenesis and pathology of bovine pneumonia. Vet Clin North Amer Food Anim Prac 26: 191-214, 2010 Ridpath, J.F., Fulton, R.W.: Knowledge Gaps Impacting the Development of Bovine Viral Diarrhea Virus Control Programs in the United States. Journal of American Veterinary Medical Association 235: 1171-1179, 2009. Ridpath, J.F., Fulton, R.W., Kirkland, P.D., Neil, J.: Prevalence and Antigenic Differences Observed Between Bovine Viral Diarrhea Virus Subgenotypes Isolated From Cattle in Australia and Feedlots in the Southwestern United States. Journal of Veterinary Diagnostic Investigation 22; 184-191, 2010. Rivera-Rivas JJ, Kisiela D, Czuprynski CJ. 2009. Bovine herpesvirus type 1 infection of bovine bronchial epithelial cells increases neutrophil adhesion and activation. Vet Immunol Immunopathol. 131:167-176. Rose-Dye, T.K., Burciaga-Robles, L.O., Krehbiel, C.R., Step, D.L., Fulton, R.W., Confer. A.W., Richards, C.J.: Rumen Temperature Change Monitored with Remote Reporting Rumen Temperature Boluses Following Challenge with Bovine Viral Diarrhea Virus and Mannheimia haemolytica. Journal of Animal Science, Accepted for publication, 2010. Rosenbusch R, Woolums A, Grooms D, Larson R. Summary of breakout sessions of 2009 Bovine Respiratory Disease Symposium. An Health Res Rev 2009;10:169-171. Saira, K., Y. Zhou, and C. Jones. 2009. The infected cell protein 0 encoded by bovine herpesvirus 1 (bICP0) associates with interferon regulatory factor 7 (IRF7), and consequently inhibits beta interferon promoter activity. J. Virology. 83:3977-3981. Shanthalingham, S., C.L.Topliff, C.L.Kelling, S.Srikumaran. 2010. Bighorn sheep fetal lung cell line for detection of respiratory viruses. Canadian Journal of Veterinary Research 74:75-77. Singh K, Ritchey JW, Confer AW. Mannheimia haemolytica: Bacterial-host interactions in bovine pneumonia. Vet Pathol 2010, in press Singh K, Confer AW, Hope JC, Rizzi T, Wyckoff JW, Weng HY, Ritchey JW. Cytotoxicity and cytokine production by bovine alveolar macrophages challenged with wild type and leukotoxindeficient Mannheimia haemolytica. The Vet J 2010, Epub ahead of print. Step, D.L., Krehbiel, C.R.., Burciaga-Robles, L.O., Holland, B.P., Fulton, R.W., Confer, A.W., Bechtol, D.T., Brister, D.L., Hutcheson, J.P., Newcomb, H.L.: Comparison of Single Vaccination Versus Revaccination with a Modified Live Virus Vaccine Containing Bovine Herpesvirus-1, Bovine Viral Diarrhea Virus (Types 1a and 2a), Parainfluenza Type 3 Virus, and Bovine Respiratory Syncytial Virus in the Prevention of Bovine Respiratory Disease. Journal of American Veterinary Medical Association 235: 580-587, 2009. Stevens ET, AD Zimmerman , RE Buterbaugh, K Barling, D Scholz, J Rhoades , CCL Chase. 2009. The Induction of a Cell-Mediated Immune Response to Bovine Viral Diarrhea Virus with an Adjuvanted Inactivated Vaccine. Vet Therapeutics 10(4):E1-8. Taylor, J.D., Fulton, R.W., Dabo, S.M., Lehenbauer, T.W., Confer, A.W.: Comparison of Genotypic and Phenotypic Characterization Methods for Pasteurella multocida Isolates from Fatal Cases of Bovine Respiratory Disease. Journal of Veterinary Diagnostic Investigation 22; 366-375, 2010. Taylor, J.D., Fulton, R.W., Lehenbauer, T.W., Step, D.L., Confer, A.W.: The Epidemiology of Bovine Respiratory Disease: What is The Evidence for Predisposing Factors? In press, 2010. Canadian Veterinary Journal. Taylor, J.D., Fulton R.W., Lehenbauer T.W., Step D.L, Confer A.W.: The Epidemiology of Bovine Respiratory Disease: What is the Evidence for Preventive Measures? In press, 2010. Canadian Veterinary Journal. Tiwari R, Sullivan J, Czuprynski CJ. 2009. PECAM-1 is involved in neutrophil transmigration across Histophilus somni treated bovine brain endothelial cells. Microb Pathog. 47:164-170. Topliff C.L., D.R. Smith, S.L.Clowser,D.J. Steffen, J.N. Henningson, B.W. Brodersen, D. Bedenice, R. J. Callan, C.Reggiardo, K.L. Kurth, C.L. Kelling. 2009. Prevalence of bovine viral diarrhea virus infections in alpacas in the United States. Journal of American Veterinary Medical Association 234:519-529. White B, Hanzlicek G, Sanderson M, Anderson D, Larson R. Mollicutes species and Mycoplasma bovis prevalence and association with health outcomes in beef feeder calves at arrival and initial treatment for bovine respiratory disease. Can Vet J. 2010;51: In Press. Wiggins MC, Woolums AR, Hurley DJ, Sanchez S, Ensley DT, Donovan D. The effect of various Mycoplasma bovis isolates on bovine leukocyte responses. Comp Immunol Microbiol Infect Dis Epub ahead of print. 2010. doi:10.1016/j.cimid.2010.02.001 Woolums A, Chase C, Fulton R, Rosenbusch R, Tremblay R. Introduction to the Proceedings of the 2009 Bovine Respiratory Disease Symposium. An Health Res Rev 2009;10:99. Workman, A., S. Perez, A. Doster, and C. Jones. 2009. Dexamethasone treatment of calves latently infected with bovine herpesvirus 1 (BHV-1) leads to activation of the bICP0 early promoter, in part by the cellular transcription factor C/EBP-alpha. J. Virology, 83:8800-8809. Workman, A. and C. Jones. Bovine herpesvirus 1 (BoHV1) productive infection and bICP0 early promoter activity are stimulated by E2F1. 2010. J Virology, 84: 63086317
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.