NC1019: Control of Emerging and Re-emerging Poultry Respiratory Diseases in the United States (was NC-228)

(Multistate Research Project)

Status: Inactive/Terminating

SAES-422 Reports

Annual/Termination Reports:

[02/28/2005] [11/27/2006] [02/27/2007] [11/30/2007] [02/04/2008] [03/06/2009]

Date of Annual Report: 02/28/2005

Report Information

Annual Meeting Dates: 11/12/2004 - 11/13/2004
Period the Report Covers: 10/01/2004 - 11/01/2004

Participants

CT AES -M. Khan (Secretary);
DE AES -J. E. Dohms and J. Gelb, Jr. (Chair);
IA AES -D. L. Reynolds;
IL AES -D. N. Tripathy;
IN AES -C. C. Wu;
MN AES -M. K. Njenga;
OH AES -Y. M. Saif;
USDA ARS SEPRL -D. Suarez;

New member for the new/renewal project NC-1019:
B. Buckles (New York);

Administrative Advisor: Jeffrey Klausner (University of Minnesota);
USDA CSREES Representative: Peter Johnson

Brief Summary of Minutes

The NC-228 annual Technical Committee meeting was held on Friday, November 12, 2004 at Grant Park Room in the Congress Hotel, Chicago, IL. Jack Gelb, Chair of NC-228, opened the meeting at 2 PM. Welcomes and introductions followed.


New Project NC-1019 and the Annual and Termination Reports for Current Project NC-228
Dr. Klausner congratulated the Committee for submitting an excellent proposal for the renewal project NC-1019. He and Dr. Gelb also indicated that the NCRA Multistate Research Committee that had reviewed the project had made two recommendations: to develop more stakeholder interaction with representatives of the animal health industry and practitioners; and to make additional efforts to leverage financial support.


Dr. Gelb agreed to coordinate the preparation of the NC-228 2003-2004 annual report and will prepare a draft and circulate it to the participants for review. After making all changes, Dr. Gelb will submit the final version of the NC-228 2003-2004 annual report by January 13, 2005, the 60-day deadline from the date of the 2004 annual meeting.


Dr. Klausner indicated that this years 2003-2004 annual report, the last report for the NC-228 project, could serve as the termination report with the addition of the impact and accomplishments sections. The Committee agreed to the best way to prepare the NC-228 termination report was to take all the highlighted bullets from the annual reports and assemble them, as well as the publications and a summary of the impact statements into a draft for circulation to the participants for review. Dr. Gelb, the out-going chair of NC-228, agreed to assist the newly elected chair of the NC-1019 project with the preparation of the NC-228 termination report. The deadline for the NC-228 termination report is March 15, 2005.


Given that the new project, NC-1019 had just commenced on October 1, 2004, the first annual report will be prepared after next years 2005 annual meeting.


Update from Dr. Peter Johnson (USDA CREES)

Dr. Johnson (USDA, CREES) also congratulated the Committee for their the efforts in successfully developing a new proposal on respiratory diseases of poultry. He reiterated his strong support for multi-state regional projects. Dr. Johnson distributed a handout entitled, USDA Cooperative State Research, Education, and Extension Service (CREES) Report, Chicago IL  November 2004. He reported on the following.
1.CREES personnel changes.
2.A new CSREES website (www.csrees.usda.gov).
3.Competitive Programs (www.csrees.usda.gov/fo/funding.cfm). Updated FY2005 competitive grant program with emphasis on the ones of interest to our regional project. Discussed the FY2004 NRI program dates and the funding within various programs. Noted the Postdoctoral Fellowship program and the restriction to US citizens and the need to make these awards. Noted the Animal Well-Being Assessment and Improvement and Veterinary Immunological Reagents programs. Noted the identification of Avian Coccidia, Mareks Disease, and Poult Enteritis Mortality Syndrome as diseases receiving high priority species-specific status in FY 2005. Some concern was expressed on the part of the Committee as to how these diseases were identified. Indicated that proposals on exotic ND and AI would be considered under non-species specific high priority areas.
4.The projected Presidents non-defense budget from 2004-2009 detailing anticipated reductions in USDAs as well as other agencys budgets.


Election of officers for 2004-2005

A Nomination Subcommittee consisting of Drs. Wu, Saif and Gelb recommended Dr. David Suarez (USDA, ARS, SEPRL) for Chair and Dr. Ching-Ching Wu (Indiana) for Secretary for two-year terms (2005-2006). Both were unanimously elected and thanked by the Committee for taking on these important responsibilities.


Stakeholders participation in NC-1019

Dr. Gelb initiated a discussion about bringing in stakeholders from the poultry industry to seek their input for the research direction and support for our regional project. It was proposed that one or two persons could be invited from the industry. The group discussed several names of stakeholders and agreed that Dr. Bruce Stewart-Brown of Perdue Farms, Inc. should be invited to our next meeting. We will ask Dr. Stewart-Brown to report on the status of current poultry diseases in general and the specific nature and impact of respiratory diseases on production in layers, broilers and turkeys.


Potential new location for the NC-1019 annual meeting

The Committee discussed the possibility of moving the NC-1019 annual meeting to another location in conjunction with another meeting to encourage better attendance. Members polled at this years meeting indicated a preference in the following order: Southern Conference on Avian Diseases meeting and the US Poultry and Egg Assn. trade show in Atlanta, Georgia in January; US Animal Health Assn. meeting in various locations in October, Western Poultry Disease Conference in various locations in March, and lastly, the current Chicago location in conjunction with the Conference of Research Workers in Animal Diseases in November. Given that several members were not in attendance, Dr. Gelb agreed to poll all participants via e-mail so that this matter could be discussed further before any decision was made.


Discussion of the annual station reports

The Committee discussed research activities during the period from October 1, 2003 - September 30, 2004.


Work planned for the coming year

AVIAN INFLUENZA VIRUS
Avian influenza reservoirs will be identified in the New England states (CONNECTICUT). Develop improved diagnostic capabilities including real time PCR as well as other rapid on-farm tests for economically important respiratory diseases (CONNECTICUT). Develop and optimize of real time multiplex-PCR tests for avian influenza (CONNECTICUT).


Characterize AIV isolates from the 2004 Delmarva outbreak (DELAWARE).

Continue surveillance for AIV. Studies will be initiated on the molecular changes in influenza viruses associated with crossing the species barrier (OHIO)


AVIAN METAPNEUMOVIRUS
Develop a continuous cell line capable of growing higher titers of APV (MINNESOTA).


Continue surveillance for avian pneumoviruses (OHIO).


ESCHERICHIA COLI
Expand studies to evaluate the effect of various strains of avian pathogenic E. coli (APEC) on avian macrophage gene expression and to compare these responses to those observed when macrophage are exposed to viral pathogens (DELAWARE).


Continue sequencing the genome of an avian clone of E. coli (MINNESOTA).


INFECTIOUS BRONCHITIS VIRUS
IBV S gene specific recombinant DNA vaccine and its application in-ovo using interferon Type 1 as an adjuvant will continue. Develop and optimize of real time multiplex-PCR tests for infectious bronchitis for differentiation of serotypes (CONNECTICUT).


Characterize Delmarva isolates from 2004 (DELAWARE).


INFECTIOUS BURSAL DISEASE VIRUS
Examine the pathogenesis of IBDV using reverse genetically engineered strains for the purpose of understanding the molecular events and mechanisms by which the virus interacts with bursa of Fabricius. Study effect of chicken cytokine genes on immunomodulation and protection of chickens against IBD by DNA vaccination. Study the effect of boosting with transgenic algae expressing IBDV VP2 on DNA vaccination of chickens against IBD (INDIANA).


Continue studies on the genetic drift observed in IBDV and its relationship to antigenic variations in these viruses. Produce and characterize monoclonal antibodies to the vvIBDV (OHIO).


ORNITHOBACTERIUM RHINOTRACHEALE (ORT)
Investigate the role of ORT on peritonitis in laying chickens (MINNESOTA)


NEWCASTLE DISEASE VIRUS
Characterize Delmarva isolates from 2004 (DELAWARE).


MYCOPLASMAS
Additional work with the unusual M. synoviae strain will continue (DELAWARE).


The meeting was adjourned at 4:30 pm on November 13, 2004.


Minutes submitted by M. Khan.

Accomplishments

Publications

Impact Statements

Back to top

Date of Annual Report: 11/27/2006

Report Information

Annual Meeting Dates: 12/02/2005 - 12/03/2005
Period the Report Covers: 10/01/2004 - 09/01/2005

Participants

The annual meeting was adjourned at 5:00 pm December 3 2005.
1. MEETING PARTICIPANTS
Drs. Glisson (Georgia), Giambrone (Alabama), Johnson (USDA), Keeler and Dohm (Delaware), Khan (Connecticut), Klausner (Advisor), Saif and Lee (Ohio), Sharma (Minnesota), Tripathy (Illinois), Suarez (USDA), and Wu (Indiana).

Administrative Advisor: Jeffrey Klausner (University of Minnesota)
USDA CSREES Representative: Peter Johnson

Brief Summary of Minutes

The annual meeting was adjourned at 5:00 pm December 3 2005.

BRIEF SUMMARY OF MINUTES OF ANNUAL MEETING

The annual NC 1019 business meeting was held on Friday December 2 2005 at Davos room in the Sheraton Westport Plaza Hotel, St. Louis, MO. Dr. David Suarez, Chair of NC 1019 opened the meeting at 2:00 pm. He welcomed the committee advisor Dr. Jeff Klausner, new member Dr. Joe Jiambrone from Alabama, and guests Dr. John Glisson from Georgia and Dr. W Lee from Ohio, Dr. J. Sharma who sat in for Dr. M. Njenga from Minnesota, and Dr. C. Keeler from Delaware. Dr. Peter Johnson, USDA/CSREES representative, joined the group around 4 pm.

Dr. Klausner commended the group for a successful submission and approval of the new project and gave a special thank to the past Chair Dr. Jack Gelb for the time and effort he spent during the process. Dr. Klausner also reminded the Chair and Secretary that the minutes of the meeting should be submitted to his office within 30 days after the meeting is held.

Dr. Peter Johnson briefed the group with the CSREES budget appropriation process. Most categories in the budgets, including the NRI, stayed the same with the exception that a new appropriation of $500,000 for Veterinary Medical Service Act was added.
Dr. Johnson shared with the group the research priority list for 2006 NRI. He indicated that CSREES continued their process in soliciting input from federal, state and local partners to guide competitive programs as they increase focus. He requested NC 1019 multi-state committee to also provide consensus feedback on the research priority again for 2007.

In response to Dr. Johnsons request, the group discussed and prioritized diseases of importance. While there are many important diseases, three came to the top for this year: ILT>IBDV>APV. Since AI has been in many lists for funding (including CSREES and HHS), the group did not recommend AI. Drs. David Suarez and Ching Ching Wu will prepare a feedback letter to Dr. Johnson reflecting our consensus. In addition, the letter will recommend CSREES to add Emerging and reemerging diseases to b. Foreign Animal Diseases under the Priority for Research 2) Non-species specific, high priority areas. This will allow the inclusion of emerging diseases such as broiler and poult enteritis to the example lists given in this category.

The meeting venue for the next year was discussed and it was voted to have our next meeting held at the end of January 2007 in Atlanta in conjunction with the International Poultry Exposition and the AI CAP annual meeting. Drs. Suarez will finalize the detail on meeting place, date, and time. He will inform the members of the specifics as soon as they are available. The members will make a decision in Atlanta as to the future meeting plans. The meeting will remain closed but the Chair and Secretary can invite guests per members suggestion or as needed. The business meeting end at 5:00 pm December 2 2005.

Station progress report resumed at 8:00 am Saturday December 3 2005. We had excellent discussion on the clinical aspects, pathogenesis, diagnostics, and vaccine/immunology of various poultry disease research among the stations. Since vvIBDV could pose serious threat to the poultry industry, upon hearing the station report from Indiana on new and sensitive vvIBDV detection assay, Dr. Sharma suggested that Drs. Saif and Wu look into the availability and validation of current available vvIBDV detection assays and recommended the committee to urge NAHLN and HHS to provide funds to support what is needed for the inclusion of this assay to their current panel. The committee will provide a letter in support of this request.

At the end of the meeting, Dr. Suarez requested that each station submit the annual report and the collaboration records electronically to him ASAP.

Minutes Submitted by C.C. Wu

Accomplishments

Objective 1. Determine the pathogenesis and interactions of specific agents.<br /> <br /> Avian Influenza Virus <br /> Quail have previously been suggested as host that can support replication of a number of avian influenza viruses. Quail and other species were examined for the ability of viruses to attach to different tissue types. Quail trachea and intestine appeared capable of binding both avian and human lineage viruses. Human trachea/bronchial epithelial (HTBE) cells were also evaluated for the ability to support influenza replication. These cultured cells included a variety of cell types that were polarized. This cell line allowed avian viruses with human virus receptor specificity to replicate, but avian viruses with normal receptor specificity did not. This appears to be a useful model to examine virus replication and host specificity.(U Maryland)<br /> <br /> We characterized a highly pathogenic outbreak of H5N1 from poultry from South Korea for its relationship to other outbreaks in the region and its potential for crossing the species barrier. We performed sequence analysis and animal studies of the outbreak in conjunction with the Centers for Disease Control in Atlanta and the Veterinary Research and Quarantine Service in South Korea. The results suggested a multiple point source of introduction which helped shape the regulatory response to the outbreaks.<br /> Reporting of low pathogenicity avian influenza outbreaks in poultry in the USA has resulted in trade embargos on chicken meat.<br /> <br /> Avian Metapneumovirus (AMV)<br /> Studies were initiated to examine the ability of Avian metapneumovirus (AMV) to induce mucosal cellular and humoral immunity in the upper respiratory tract (URT). Avian metapneumovirus in turkeys revealed that the attenuated and virulent strains of the virus induced IgA+ cells in the respiratory mucosa of the upper respiratory tract (URT). Turkey macrophages were also shown to be susceptible to in vitro infection and activation by AMV. (U Minnesota)<br /> The sequence of the Colorado 96 AMV isolate was completed. The virus was similar to a human metapneumovirus and to other type C viruses from Minnesota. The virus was more divergent with the types A and B AMV isolates found in other countries. The g protein was highly variable with up to 19 % sequence divergence with the Minnesota viruses.<br /> Using the cloned genes in a reverse genetic system, a complete genome was rescued of the type C AMV virus. The rescued virus biological was the same as the parent virus. (U Maryland)<br /> <br /> Newcastle Disease Virus<br /> Newcastle disease virus is a negative sense single stranded virus infection of poultry. The virus encodes for multiple proteins that are produced as viral constructs separated by short intergenic sequences. In an effort to evaluate the role these intergenic sequences have on viral virulence, reverse genetics was used to modify the length of the sequences. The viruses were rescued and little difference was seen in growth in cell culture, but in general a decrease in virulence was seen when chickens were challenged with the virus.<br /> The use of reverse genetics was also evaluated for the examination of the virulence role of the polymerase gene, which is important for virus replication. The polymerase gene was swapped from a virulent virus to a non-virulent virus. These studies suggest a role for the polymerase gene as a possible virulence factor.(U Maryland)<br /> <br /> Characterization of infection and disease of turkeys with NDV strains of low to high virulence. In general, disease among NDV infected turkeys was found to be less severe than in similarly infected chickens, and turkeys infected with virulent strains shed virus for a longer time and appeared to be subclinical carriers for some of the isolates. (SEPRL)<br /> <br /> Infectious Bursal Disease Virus<br /> Positive sense RNA transcripts of Infectious bursal disease virus (IBDV) genome segments A and B have previously been shown to be infectious. We demonstrated that recovery of IBDV from the transfection of Vero cells with positive sense RNA transcripts of genome segments A and B was enhanced by expression of the viral structural proteins VP2 with VP3 or by expression of viral polyprotein VP243 from DNA plasmids in trans. Expression of individual viral proteins VP2, VP3, or VP4 alone from DNA plasmids did not enhance IBDV recovery. Earliest virus recovery from transfection of positive sense RNA transcripts of genomic segments A and B was at 36 h and mean titers were 101.8 pfu/ml. IBDV was recovered 6 hours after transfection in cells concurrently expressing either VP2 with VP3 or VP243 and mean titers were 108.5 pfu/ml or 109.2 pfu/ml, respectively. Likewise, expression of the viral polyprotein from DNA plasmid increased the permissiveness of Vero cells for infection with non-culture adapted IBDV. The titer of recovered non-culture adapted virus from 103.3pfu/ml to 1010.3pfu/ml with expression of the viral polyprotein.<br /> (Purdue U)<br /> <br /> Objective 2. Surveillance, occurrence and consequences of agents and host <br /> variation on disease susceptibility.<br /> <br /> Avian Influenza<br /> Over 5000 poultry samples were tested by real-time RT-PCR (RRT-PCR) and over 3000 blood samples by AGID were tested and found to be negative for avian influenza from live bird markets and backyard flocks in the New England States. (U Connecticut)<br /> <br /> 60 wild bird samples were collected from Alabama and tested by virus isolation for avian influenza. All the samples were negative. (Auburn U)<br /> <br /> H3N2 viruses have been isolated from outbreaks in several states from turkey breeder flocks experiencing drops in egg production. The antigenic relatedness of four isolates, the IL isolate, OH isolate, an NC recent turkey isolate, and the swine vaccine used in Illinois, were compared and a high antigenic relatedness was observed between the turkey isolates, but only a 10% relatedness of the swine vaccine to the turkey isolates. The variability of circulating H3N2 viruses may affect how vaccination is used to control this problem. (Ohio State U)<br /> <br /> Infectious Bursal Disease Virus (IBDV)<br /> A comparison of the detection of antibodies against serotypes 1 and 2 IBDV by commercial ELISA kits indicated that currently available commercial ELISA kits detected antibodies elicited by the two serotypes of IBDV. Hence, the prevalence of serotype 2 antibodies in the flocks should be considered while determining antibody profiles of the flocks against serotype 1 viruses. <br /> (Ohio State U)<br /> <br /> Objective 3. Develop new and improved methods for the diagnosis, prevention, and control of avian respiratory diseases.<br /> <br /> Avian Influenza <br /> An Adenovirus-vectored vaccine, originally developed for humans against H5N1, was given to SPF leghorns in ovo and was shown to produce measurable antibody titers. The birds were challenged with a highly pathogenic avian influenza virus and most birds were clinically protected from disease. This vector system is replication restricted virus and therefore it has the safety of a killed vaccine, but the immune response of a live vaccine. The use of in ovo vaccination offers the possibility of mass vaccination of poultry during an avian influenza outbreak. (Auburn U and SEPRL).<br /> <br /> The use of DNA vaccines for use in the production of reference diagnostic reagents were improved with the addition of the cytokine adjuvants, IL-2 and interferon. DNA vaccines can produce high quality antibodies to specific proteins that are extremely valuable for diagnostic reagents, but the response to DNA vaccines are variable. Alternative methods for adjuvanting or increasing the immune response were conducted for both the H5 and H7 hemagglutinin protein of avian influenza, and it was found that both the inclusion of plasmids with the cytokines IL-2 and type 1 interferon improved the response for H7 vaccines. This work will allow the improved production of reference antibodies in the future that are safer and easier to produce than the current methods.(SEPRL)<br /> <br /> Avian Metapneumovirus (AMV)<br /> Turkeys were immunized with adjuvanted rNP and recombinant M protein (rMP) administered intramuscularly and immunized and unimmunized controls were challenged with virulent AMV by the respiratory route. At a dose of 40 ug/bird, rNP protected eight of nine birds. rMP at the same dosage level protected three out of seven birds. At a dose of 80 ug of each rNP and rMP, 100% protection was achieved. This recombinant vaccine shows promise as an improved control measure for AMV. <br /> Another study was designed to determine if in vivo passages of AMV would increase virus virulence leading to consistent clinical signs in turkeys. The results of this preliminary study indicate that in vivo passage of AMV in birds may increase virus virulence and the resulting virus could serve as a suitable challenge inoculum for use in vaccination trials. <br /> Currently Vero cells are commonly used for the isolation of AMV from clinical samples, but because Vero cells are a mammalian cell line, concern that the virus is changed by passage in this cell line is a concern. Sequence analysis supports this idea, since 11 amino acid differences in the F gene of AMV propagated in turkey cells and that propagated in Vero cells. In an attempt to find a better avian origin cell line for isolation of AMV, alternatives were tested. A non-tumorigenic immortal turkey turbinate cell line was developed that is susceptible to AMV, and may provide a valuable alternative to Vero cells. (U Minnesota)<br /> <br /> Infectious Bursal Disease Virus (IBDV)<br /> IBDV is an immunosuppressive disease found in chickens. A mild form is found in the U.S., but very virulent forms are found in many other countries. A sequence comparison of the virulent and non-virulent forms showed sequence difference that were used as a differential RT-PCR test. A diagnostic assay was developed that reliably differentiated very virulent infectious bursal disease virus (vvIBDV) from non-vvIBDV strains. The availability of of rapid diagnostic tests facilitate identification of field isolates, and allow a rapid response for control of highly virulent IBDV viruses are introduced into the U.S.<br /> <br /> IBDV also exists as two antigenically different groups, and a RT-PCR test was developed that could rapidly differentiate serotype 1 viruses, serotype 2 viruses, and the vv strains of IBDV. These tools allow the rapid differentiation of the different serotypes to provide rapid characterization of the viruses. (Ohio State U)<br /> <br /> A real-time RT-PCR assay was developed utilizing dual-labeled fluorescent probes binding to VP4 sequence that are specific to the classical, variant and very virulent strains of Infectious Bursal Disease Virus (IBDV). The assay was highly sensitive and could detect as little as 3 ´ 102 to 3 ´ 103 copies of viral template. The variant sequence-specific probe was found to be highly specific in detecting isolates classified as variant A, D, E, G and GLS-5, and did not react with classical strains. The classical sequence-specific probe also demonstrated high sensitivity and specificity and differentiate between isolates that were variant and classical strains. The very virulent sequence-specific probe positively detected the Holland vvIBDV isolate and did not react with classical or variant strains. (Purdue U)<br /> <br /> WORK PLANNED FOR THE COMING YEAR<br /> University of Connecticut- Surveillance of wild birds for avian influenza collected from tracheas and cloacal samples from Connecticut will be tested. <br /> Multiplex PCR (MPCR) for avian influenza and subtypes H5, H7 and H9 is being developed at the Guangxi Veterinary Research Institute Nanning, China. MPCR will be tested on the North American AI isolates to confirm its sensitivity and specificity.<br /> <br /> Auburn University- The wild bird surveillance for avian influenza will be increased in the next year. Additional studies of antibody response in wild birds will be conducted.<br /> The Mx gene will be evaluated for protection against avian influenza in different poultry lines.<br /> Comparison of different procedures for poultry litter management will be evaluated to determine if it adequately inactivates Infectious Laryngotracheitis Virus (ILTV) and AIV.<br /> <br /> The Ohio State University- Surveillance for influenza and pneumoviruses will continue using for serologic and molecular diagnostic tests. Studies will be initiated on the molecular changes in influenza viruses associated with crossing the species barrier. Production and characterization of monoclonal antibodies to the vvIBDV will continue.<br /> <br /> Purdue University- We will examine the pathogenesis of IBDV using reverse genetically engineered strains for the purpose of understanding the molecular events and mechanisms by which the virus interacts with bursa of Fabricius. We will also study additional chicken cytokine genes on immune response and protection of chickens against IBD by DNA vaccination. We will also study the effect of boosting with transgenic algae expressing IBDV VP2 on DNA vaccination of chickens against IBD.<br />

Publications

Bennett, R.S., , J. Nezworski, B.T. Velayudhan, K.V. Nagaraja, D.H. Zeman, Dyer, T. Graham, D.C. Lauer, M.K. Njenga and D.A. Halvorson: Evidence of Avian Pnuemovirus spread beyond Minnesota among wild and Domestic birds in Central North America. Avian Diseases 48:902-908.2004<br /> <br /> Bennett, R.S., R. LaRue, K.V. Nagaraja, D. Shaw, Q. Yu, D.A. Halvorson, and M.K. Njenga. A Wild Goose Avian metapneumovirus Containing a Large Attachment (G) Glycoprotein is Avirulent but Immunoprotective to Commercial Turkeys. Accepted. J. Virology. 2004.<br /> <br /> Chary, P, M.K. Njenga and J.M. Sharma. Protection by recombinant viral proteins against a respiratory challenge with virulent avian metapneumovirus. Veterinary Immunology and Immunopathology. 108:427-432. 2005<br /> <br /> Christman S.A., B.-W. Kong, M.M. Landry, H. Kim, and D.N. Foster. 2005 Modulation of p53 expression and its role in the conversion to a fully immortalized chicken embryo fibroblast line. FEBS Letters. In Press.<br /> <br /> Christman S.A., B.-W. Kong, M.M. Landry, and D.N. Foster. 2005. Chicken Embryo Extract Mitigates Growth and Morphological Changes in a Spontaneously Immortalized Chicken Embryo Fibroblast Cell Line. Poultry Sci. 84:1423-1431.<br /> <br /> Govindarajan, D. and Samal, S.K. (2004). Sequence analysis of the large polymerase (L) protein of the US strain of avian metapneumovirus indicates a close resemblance to that of the human metapneumovirus. Virus Res. 105: 59-66.<br /> <br /> Govindarajan, D., Yunus, A.S. and Samal, S.K. (2004). Complete sequence of the G glycoprotein gene of avian metapneumovirus subgroup C and identification of a divergent domain in the predicted protein. J Gen Virol 85: 3671-3675.<br /> <br /> Govindarajan, D. and Samal, S.K. (2005). Analysis of the complete genome sequence of avian metapneumovirus subgroup C indicates that it possesses the longest genome among metapneumoviruses. Virus Genes 30(3): 329-331.<br /> <br /> Hongquan Wan and Daniel R. Perez. (2006). Quail carry sialic acid receptors compatible with binding of avian and human influenza viruses. Virology: In Press<br /> <br /> Huang, Z., A. Panda, S. Elankumaran, D. D. Rockemann and S. K. Samal (2004). The Hemagglutinin-Neuraminidase protein of Newcastle Disease Virus determines tropism and virulence. J Virol 78:4176-4184.<br /> <br /> Jackwood, D. J. and S. E. Sommer. Molecular studies on suspect very virulent infectious bursal disease virus genomic RNA samples. Avian Dis. 49:246-251. 2005.<br /> <br /> Jackwood, D. J. and S. E. Sommer. Molecular epidemiology of infectious bursal disease viruses: Distribution and genetic analysis of new variants in the United States. Avian Dis. 49:220-226. 2005.<br /> <br /> Jirjis, F., S. Noll, F. Martin, D.A. Halvorson, K.V. Nagaraja and D.P. Shaw: The effects of bacterial co-infection on the Pathogenesis of Avian pneumovirus infection in turkeys.Avian Diseases, 48: 34-49.2004.<br /> <br /> Keeler, C.L., Schnitzlein, W.M., Shaffer, A.E. and Tripathy, D.N. Characterization of a Glycoprotein C Mutant of Infectious Laryngotracheitis. Abst. American Veterinary Medical Association, Minneapolis, MN., 2005<br /> <br /> King, D. J. Newcastle disease. In: Merck Veterinary Manual, 9th edition. Kahn, C.M. and Line, S. (editors). 2005, pp. 2255-2257.<br /> <br /> Kong B.-W., L.K. Foster, and D.N. Foster. 2005. Comparison of Avian Cell Substrates for Propagating Subtype C Avian Metapneumovirus Virus Res. In Press.<br /> <br /> Lee, C.W., D.A. Senne, and D.L. Suarez. 2004. Effect of Vaccine Use in the Evolution of Mexican-lineage H5N2 Avian Influenza Virus. Journal of Virology 78:8372-8381.<br /> <br /> Lee, C.-W., D. L. Suarez, T. M. Tumpey, H.-W. Sung, Y.-K. Kwon, Y.-J. Lee, J.-G. Choi, S.-J. Joh, M.-C. Kim, E.-K. Lee, J.-M. Park, X. L., J. M. Katz, E. Spackman, D. E. Swayne, J.-H. Kim. 2005. Characterization of Highly Pathogenic H5N1 Avian Influenza A Viruses Isolated from Korean Poultry. Journal of Virology. 79:3692-3702.<br /> <br /> Lee, C.-W. and D. L. Suarez. 2005. Avian influenza: prospect for prevention and control by vaccination against antigenic drift of the virus. Animal Health Research Reviews. 6:1-15.<br /> <br /> Maherchandani, S., Munoz-Zanzi, Patnayak, D.P., Malik, Y.S. and Goyal, S.M. 2004. The effect of pooling sera on the detection of avian pneumovirus antibodies using an enzyme-linked immunosorbent assay. J. Vet. Diagn. Invest. 16:497-502.<br /> <br /> Maherchandani, S., Patnayak, D.P., Lauer, D., and Goyal, S.M. 2005. Evaluation of five different antigens in enzyme-linked immunosorbent assay for the detection of avian pneumovirus antibodies. J. Vet. Diagn. Invest. 17:16-22.<br /> <br /> Malik, Y.S., Patnayak, D.P., and Goyal, S.M. 2004. Detection of three avian respiratory viruses by single-tube multiplex reverse transcription-polymerase chain reaction assay. J.Vet. Diagn. Invest. 16:244-248.<br /> <br /> Mickael, C. S. and D. J. Jackwood. Real-Time RT-PCR analysis of two epitope regions encoded by the VP2 gene of infectious bursal disease viruses. J. Virol. Methods. 128:37-46. 2005.<br /> <br /> <br /> Munir, S., J.M. Sharma and V. Kapur. Transcropitional response of avian cells to infection with Newcastle disease virus. Virus Research 107:103-8. 2004<br /> <br /> Panda, A., S. Elankumaran, S. Krishnamurthy, Z. Huang, and S. K. Samal (2004). Loss of N-linked glycosylation from the Hemagglutinin-Neuraminidase protein alters virulence of Newcastle Disease Virus. J Virol 78: 4965-4975.<br /> <br /> Patnayak, D.P., Sheikh, A.M., and Goyal, S.M. 2004. Stability of attenuation in live avian pneumovirus vaccines. J. Appl. Poultry Res. 13:253-257.<br /> <br /> Patnayak, D.P., and Goyal, S.M. 2004. Duration of immunity produced by a live attenuated vaccine against avian pneumovirus type C. Avian Pathol. 33:465-469.<br /> <br /> Patnayak, D.P., and Goyal, S.M. 2004. Cold-adapted strain of avian pneumovirus as a vaccine in one-day-old turkeys and the effect of inoculation routes. Avian Dis. 48:155-159.<br /> <br /> Patnayak, D.P., Tiwari, A., and Goyal, S.M. 2005. Growth of vaccine strains of avian pneumovirus in different cell lines. Avian Pathol. 34:123-126.<br /> <br /> Patnayak, D.P., and Goyal, S.M. 2005. Duration of immunity engendered by a single dose of cold adapted strain of avian pneumovirus. Can. J. Vet. Res.<br /> <br /> Peters, M, Lin, T.L., and Wu, C.C. 2004. Infectious bursal disease virus polyprotein expression arrests growth and mitogenic Stimulation of B lymphocytes. Archives of Virology, 149(12): 2413-2426.<br /> <br /> Peters, M.A., Lin, T.L., and Wu, C.C. 2005. Real-time PCR differentiation and quantitation of infectious bursal disease virus strains. Journal of Virological Methods, 127(1): 87-95.<br /> <br /> Peters, M.A., Lin, T.L., and Wu, C.C. 2005. Infectious bursal disease virus recovery from Vero cells transfected with RNA transcripts is enhanced by expression of the structural proteins in trans. Archives of Virology, 150 (11): 2183-2194.<br /> <br /> Saif, Y.M. 2005. Control of infectious bursal disease virus by vaccination. Control of Infectious Animal Diseases by Vaccination (A. Schudel and M. Lombard, eds), Developments in Biologicals. Karger, New York, NY, Vol. 119:143.<br /> <br /> Seal, B.S.: Nucleotide and predicted amino acid sequence analysis of the fusion protein and hemagglutinin-neuraminidase protein genes among Newcastle disease virus isolates. Phylogenetic relationships among the Paramyxovirinae based on attachment glycoprotein sequences. Funct. Integr. Genomics. 2004. 4(4):246-257<br /> <br /> Seal BS, Wise MG, Pedersen JC, Senne DA, Alvarez R, Scott MS, King DJ, Yu Q, Kapczynski DR. Genomic sequences of low-virulence avian paramyxovirus-1 (Newcastle disease virus) isolates obtained from live-bird markets in North America not related to commonly utilized commercial vaccine strains. Vet Microbiol. 2005.106(1-2):7-16.<br /> <br /> Singh, P, Schnitzlein, W.M. and Tripathy, DN. Construction and characterization of a Fowlpox Virus Isolate whose genome lacks reticuloendotheliosis provirus nucleotide sequences. Avian Diseases, 49: 401-408, 2005<br /> <br /> Singh, P., Schnitzlein, W.M and Tripathy, D.N. The Genome of Reticuloendotheliosis Virus integrated in the Genome of Fowlpox Virus. Poster presentation # 37. American Veterinary Medical Association, Minneapolis, MN. 2005.<br /> <br /> Srinivasan, V and Tripathy, D.N. The DNA repair enzyme, CPD-photolyase restores the infectivity of UV- Damaged fowlpox virus isolated from infected scabs of chickens. Veterinary Microbiology 108: 215-223, 2005.<br /> <br /> Tang, Y., C.W. Lee, Y. Zhang, D.A. Senne, R. Dearth, B. Byrum, D.R. Perez, D.O. Suarez, and Y.M. Saif: Isolation and characterization of H3N2 influenza A virus from turkeys. Avian Dis. 49:207-213, 2005.<br /> <br /> Tripathy, D.N. and Kim, T.J. Evaluation of Pathogenicity of Avianpox Viruses from Endangered Hawaiian Forest Birds. Poster presentation # 39. American Veterinary Medical Association, Minneapolis, MN. 2005.<br /> <br /> Tripathy, D.N. The impact of vaccines and future of genetically modified poxvirus vaccines for poultry. Animal Health Research Reviews, 5: 263-266, 2004<br /> <br /> Tumpey, T.M., and R. Alvarez, D. E. Swayne and D. L. Suarez. 2005. Diagnostic Approach for Differentiating Infected from Vaccinated Poultry on the Basis of Antibodies to NS1, the Nonstructural Protein of Influenza A Virus. Journal of Clinical Microbiology. 43:676-683.<br /> <br /> Velayudhan, B.T., B. McComb, V.C. Lopes, D.A. Halvorson and K.V. Nagaraja: Bird-Proof netting over barns can prevent the introduction of Avian pneumovirus (APV) to turkeys. Submitted to Journal of Wild life Diseases, 2004.<br /> <br /> Velayudhan, B.T., R. C. Bennett., B. McComb, V.C. Lopes, D. Shaw., D.A.Halvorson and K. V. Nagaraja. Pathogenesis of avian pneumovirus Infection in two-week-old turkeys. submitted for publication in Am. J. Vet Research. 2004 <br />

Impact Statements

  1. In a series of studies, two low pathogenicity avian influenza viruses given intranasally to chickens grew only in the respiratory and intestinal tracts, and no virus was found in the blood, meat, bone or bone marrow. By contrast, two high pathogenicity avian influenza viruses grew not only in respiratory and intestinal tracts but spread systemically with virus being found in blood, meat, bone or bone marrow. Killed vaccines or recombinant fowl pox-avian influenza vaccines prevented the high pathogenicity avian influenza viruses from being in the meat.
  2. Avian metapneumovirus - a reverse genetics system allows for detailed experiments on pathogenesis to be conducted as well as the potential to make improved vaccines.
  3. Diagnosis of Newcastle disease in turkeys was dependent on virus isolation to detect infected birds, a factor that must be considered for ND control programs.
  4. The development of a reverse genetics model for infectious bursal disease virus with high efficiency of virus recovery will help delineate the pathogenesis of IBDV and that of polymicrobial interactions of IBDV and poultry respiratory diseases.
  5. A DNA vaccine was developed using the infectious bronchitis virus-S gene in a plasmid expression system for vaccination of chickens.
  6. A diagnostic test was developed to differentiate avian poxviruses using seven sets of primers from fowlpox virus genome (39K, EGF, REV envelope, REV LTR, homolog of HA, A-type inclusion and TK). This new test provides a rapid way to characterize field strains of avian pox virus.
Back to top

Date of Annual Report: 02/27/2007

Report Information

Annual Meeting Dates: 01/23/2007 - 01/24/2007
Period the Report Covers: 10/01/2005 - 09/01/2006

Participants

Glisson (Georgia); Giambrone (Alabama); Kong (Arkansas); Johnson (USDA/CSREES); Keeler and Dohm (Delaware); Khan (Connecticut); Klausner (Advisor); Saif (Ohio); Sharma (Minnesota); Suarez and Zsak (USDA/SEPRL); Wu and Lin (Indiana)

Brief Summary of Minutes

BRIEF SUMMARY OF MINUTES OF ANNUAL MEETING.
The annual NC 1019 business meeting was held on Tuesday, January 24, 2007, in room C211 in the Georgia World Congress Center, Atlanta, GA. Dr. David Suarez, Chair of NC 1019 opened the meeting at 12:30 pm. He welcomed the committee advisor Dr. Jeff Klausner, guests Dr. John Glisson from Georgia, Dr. B. Kong from Arkansas, Dr. L. Zsak from USDA/ SEPRL, and Dr. T. L. Lin from Indiana. Dr. Peter Johnson, USDA/CSREES representative, joined the group around 3 pm.


Dr. Klausner reminded the Chair and Secretary that the minutes of the meeting should be submitted to his office within 30 days after the meeting is held.


Dr. Peter Johnson briefed the group with the CSREES budget. The budget review panel has not met yet but some cuts will occur. Dr. Johnson also discussed with the group on the research priority list for 2006 NRI. He indicated one out of the three agents recommended by NC 1019 was included in the published priority list and that CSREES will continue to solicit input from federal, state and local partners to guide competitive programs. He requested the NC 1019 multi-state committee to again provide consensus feedback on the research priorities for 2007.


In response to Dr. Johnsons request, the group discussed and prioritized diseases of importance. While there are many important diseases, three came to the top for this year: ILT>IBDV>APV. Both avian influenza virus and Newcastle disease virus are typically covered in the foreign animal disease priority, they were not included in the recommendations for the priority list. Drs. David Suarez and Ching Ching Wu will prepare a feedback letter to Dr. Johnson reflecting our consensus.


NC 1019 members reached the consensus on the importance to bring industry guests and renowned speakers to the committee meeting periodically and as needed. Possible funds and revenues to support this activity were discussed.


The meeting venue for the next year was discussed and it was agreed to meet in Atlanta next year in association with the International Poultry Exposition, but we will likely hold it at a hotel to provide more flexibility on times to meet. The exact date has not been set, but hopefully that will be determined soon. Drs. Giambrone and Wu will finalize the detail. Dr. Wu will check out the cost of renting meeting room at local hotels and Drs. Glisson and Giambrone will find funds to cover the room. Dr. Giambrone will inform the members of the specifics as soon as they are available. The members will reassess the meeting place in 2008.


The meeting will remain closed but the Chair and Secretary can invite guests per members suggestion or as needed. The group will continue to actively recruit other stations to participate.


Dr. Suarez, after completing his two year term of office, decided not to seek renewal of his Chair position. After polling the membership by email and at the meeting, Dr. Joseph Giambrone was nominated for the chair position and Dr. Ching-Ching Wu for secretary position. No additional nominations were offered from the floor, and Drs Giambrone and Wu were elected by unanimous consent. The members expressed their appreciation for the leadership that Dr. Suarez provided during his term. The business meeting end at 4:00 pm Jan 23, 2007. Station reports followed and stopped by 5:30pm.


Station progress report resumed at 7:30 am Wed., Jan. 24, 2007. We had excellent discussion on the clinical aspects, pathogenesis, diagnostics, and vaccine/immunology of various poultry disease research among the stations.


At the end of the meeting, Dr. Suarez requested that each station submit their annual report and the collaboration records electronically to him ASAP.


The annual meeting was adjourned at 11am, Jan. 24, 2007.


WORK PLANNED FOR THE COMING YEAR

University of Connecticut:
Development of Avian Influenza vaccine using reverse genetic system. Reverse genetics methods, i.e., methods that allow the generation of an influenza virus entirely from cloned cDNAs, have provided us with one means to address these issues. This technique allows customized construction of influenza vaccine by assembling genes that code for the desired features of the particular virus strain. We plan to use reverse genetic in order to develop a vaccine for the H7N2 strain of Avian Influenza and expect that this vaccine will be able to protect chickens against this specific low path virus infection.
Infectious Bronchitis: Efficiency of recombinant IBV DNA vaccine developed earlier in our laboratory is in progress.


University of Delaware:
1)We plan to continue work on AIV, IBV and ILTV .
2)Mycoplasma gallisepticum will involve confocal microscopy to more accurately evaluate the intracellular invasion in Chick Embryo Fibroblasts using MG-685 and additional strains of MG.


Auburn University:
The objective is to develop transgenic vaccines for poultry against AIV. There are a number of commercially available, efficacious vaccines against HP H5 AIV for use in poultry. However, they must be injected. The injection system is suitable for smaller breeder and layer flocks, which are routinely injected with killed vaccines, but is too costly for larger broiler flocks. In contrast, vaccines in plants or yeast could be propagated and given in mass to poultry. Yeast are routinely given in the drinking water as a probiotic, in place of antibiotics to kill bacteria, and plants can be given in feed to commercial poultry.


The Ohio State University:
1. Completing the antigenic and genetic relatedness work using the newly obtained human and duck viruses H5 AIVS.
2. Sequencing and comparing the HA and NA genes of the different isolates of TK/OH/03 virus from the transmission studies, to see how the virus changes upon replicating in different host systems.
3. Start cloning all the genes of: TK/IL/04, TK/OH/03, SW/NC/03 and HUM/OH/06 for the use in reverse genetics studies.
4. Studies will be conducted on the molecular evolution of IBDV in the United States.
6. The molecular basis for antigenicity in IBDV strains will be further studied using a reverse genetics system.
7. More intensive animal study using a large number of eggs and chickens will be conducted to examine the potential of each NS variant as live attenuated vaccine candidates for AIV.
8. Microsphere-based influenza diagnostic assay will be validated by comparing side by side with those obtained using traditional virus isolation and RRT-PCR.


Purdue University:
We will examine the pathogenesis of IBDV using reverse genetically engineered strains for the purpose of understanding the molecular events and mechanisms by which the virus interacts with bursa of Fabricius. We will also continue to study the effect of prime-boost on protection of chicken by DNA vaccination against IBD, such as boosting with transgenic algae expressing IBDV VP2. Additional studies on chicken cytokine genes (other than chicken interferon-r gene and chicken IL-2 gene) on immune response and protection of chickens against IBD by DNA vaccination.


University of Minnesota:
1)Examine the feasibility of inducing protective immunity by respiratory exposure to inactivated virus or viral subunits.
2)Define the functional role of the G gene in aMPV propagation by modifying important amino acid differences by site-directed mutagenesis.
3)Identify the virulence gene of aMPV and create a safe by reverse genetics.
4)Define the role of macrophages in the immunopathogenesis of aMPV and IBDV.


Minutes Submitted by C.C. Wu

Accomplishments

Objective 1. Determine the pathogenesis and interactions of specific agents.<br /> <br /> Avian Influenza<br /> Research to determine the resistance and susceptibility of various poultry lines against AIV infection was conducted with an emphasis on the MX gene. Mice that express high levels of the MX gene are resistant to AIV and VSV challenge. Chickens have an MX gene, but it is unknown the gene provides similar resistance to chickens. We determined that there is polymorphism in the expression of the Mx gene in various chicken breeds. The amino acid residue 631, at which, Asn determines antiviral activity, and Ser renders the MX protein inactive. We've spent most of the year developing reagents (an expression vector for chicken interferon-alpha from another scientist, transfecting it, and characterizing the titer of the interferon). Also we've have refined our assays for CEF preparation, developing faster methods of Mx typing, developing quantitative PCR for Mx expression, and evaluating replication by ELISA. Now we have most all our tools and are ready to do animal experiments, and plan to correlate MX expression of various breeds with susceptibility of CEFs from their eggs to AIV replication. (Auburn U)<br /> <br /> The interspecies transmission of different H3N2 influenza viruses between turkeys and swine were examined in an aerosol transmission experimental model. Of the viruses tested viruses, TK/IL/04 (H3N2); TK/OH/03 (H3N2); TK/NC/03 (H3N2); SW/NC/03 (H3N2); A/TK/OH/88 (H1N1) and A/SW/OH/06 (H1N1), only TK/OH/03 efficiently transmitted from pigs to turkeys (replicated for two days or more in both species). All pigs seroconverted with mean HI titer of 1:360 and 50% of turkeys seroconverted with mean HI titer of 1:80. The TK/IL/04 and TK/NC/03 replicated well in pigs, but were detected in turkeys for only one day. Only pigs seroconverted. A/TK/OH/88 and A/SW/OH/06 replicated efficiently in pigs but were not detected at all in turkeys. Only pigs seroconverted. In the reverse experiment of transmission from experimentally infected turkeys to pigs four viruses were compared, TK/IL/04 (H3N2); TK/OH/03 (H3N2); TK/NC/03 (H3N2) and SW/NC/03 (H3N2). In this experiment, the TK/OH/03 was also the only virus that efficiently transmitted from turkeys to pigs. Both species seroconverted at HI titers of 1:344 and 1:320 respectively. None of the other viruses were transmitted. The TK/OH/03 influenza virus was also inoculated into chickens, and ducks. The virus replicated in the inoculated chicken for 5 days but was not detected in the contact chicken. Infected chickens seroconverted but the contact ones did not. The virus could not be detected in either inoculated nor contact ducks. (OhioStateU)<br /> <br /> The first comprehensive biological characterization of H5N1 high pathogenicity avian influenza (HPAI) virus from wild birds were completed on viruses that came from Mongolia. H5N1 HPAI virus has caused outbreaks of disease in poultry and wild birds of 50 countries in Asia, Europe and Africa. With field assistance of Wildlife Conservation Society, Food and Agricultural Organization and Government of Mongolia, H5N1 HPAI viruses were isolated from a dead Whooper Swan in Mongolia. In experimental studies, this virus expressed high lethality in chickens and young domestic ducks, and was easily passed between ducks by causal contact. The virus grew in many internal tissues including the brain and heart. Because the outbreak occurred where no poultry exist, this indicates the H5N1 HPAI virus spread into Mongolia by migrating wild birds, but is a virus that can infect poultry and cause severe disease. (SEPRL)<br /> <br /> We provided an in-depth analysis, including sequencing and animal studies, of the first highly pathogenic avian influenza virus (HPAIV) in the U.S. in the last 20 years. An outbreak of avian influenza occurred in Texas in 2004 that had the sequence of a highly pathogenic avian influenza virus, but it was not highly pathogenic when it was used to infect chickens. Our laboratory in collaboration with the National Veterinary Services Laboratories, APHIS provided in depth sequence analysis, animal studies, and mutational studies to try and understand why the sequence and animal studies did not match together. The data helped to provide evidence that the sequence definition of highly pathogenic avian influenza needs to be reconsidered based on this and other exceptions to the O.I.E. rules. It remains critical, because of the sever affects in trade, to accurately diagnosis and report correctly any HPAI outbreaks. (SEPRL)<br /> <br /> A pathogenesis experiment was performed to examine the interactions of different immunosuppressive agents and infection with a low pathogenic avian influenza virus. Two experiments were performed to evaluate the effects of exposure of chickens with and without maternal antibodies to chicken anemia virus (CAV) and infectious bursal disease virus (IBDV) to low path AIV challenge. In the first experiment, the infection of commercial broiler chickens at different ages with CAV and IBDV did not affect (enhance or decrease) the pattern of low path H7N2 AIV (A/chicken/Maryland/Minh Ma/2004) detection (virus isolation and real time RT-PCR) in tracheal swabs following challenge on day 21 days of age or in the serum antibody responses (AGID. ELISA, and HI). The second experiment was set up similarly to the first experiment except that specific pathogen free leghorn type chickens were used instead of commercial broilers. Because of the lack of maternal antibody, a 60% mortality occurred in the SPF leghorns concurrently infected with IBD and CAV prior to the planned 21 day challenge with the LP H7N2 AIV, so this treatment group was not used in the trial. Remaining CAV only and IBDV infected only birds and controls were challenged intraocularly with the H7N2 virus at 21 days of age. No affect (enhance or decrease) on the pattern of low path H7N2 AIV detection (VI or RRT-PCR)) in tracheal swab was observed between treatment groups. Compared to AI challenge controls, SPF leghorns infected with IBDV had diminished AIV antibody titers determined by HI and ELISA, as well as fewer positive AGID responders. Infection with CAV reduced HI antibody titers but had no effect on ELISA and AGID antibody responses. The results of this experiment show that early infection with CAV or IBDV reduces AIV serological responses in response to a later challenge, but not likely enough to prevent detection by standard test. (U Delaware)<br /> <br /> Avian Metapneumovirus (AMV)<br /> Turkeys exposed to aMPV showed extensive lymphoid cell infiltrations in the upper respiratory tract (URT). The cellular infiltration occurred after the first virus exposure but not after reexposure. Quantitation of the relative proportions of mucosal IgA+, IgG+ and IgM+ cells in controls and virus-exposed turkeys revealed that at 7 days following the first virus exposure, when mucosal infiltration was well pronounced, there was a significant increase (P<0.05) in the numbers of infiltrating IgA+ but not of IgG+ and IgM+ cells. Following the second virus exposure, although the overall numbers of mucosal lymphoid cells were similar in the virus-exposed and control turkeys, the relative proportions of IgA+ and IgG+ cells were significantly higher in the virus-exposed turkeys (P<0.05) than in controls. Further, elevated levels of aMPV-specific IgA were detected in the nasal secretions and the bile of virus-exposed birds after the second but not after the first virus exposure. This result suggested, for the first time, the possible involvement of local mucosal immunoglobulins in the pathogenesis of aMPV in turkeys. (U Minnesota)<br /> <br /> Two respiratory adjuvants at three dose levels (10¼g, 20 ¼g , 40 ¼g or 60¼g) were tested: poly(I:C) and holotoxin-containing cholera toxin B (hCTB). One-day-old turkeys were given daily intranasal injections of each adjuvant for 6 days. Neither of the adjuvants caused detectable gross or microscopic lesions in the URT or lasting loss in body weight gain. This result indicated that two of the most commonly used respiratory adjuvants were safe for turkeys. Poly(I:C) and hCTB were given intranasally or oculonasally alone or in combination with inactivated aMPV. IgA cells were enumerated in the turbinate tissue of treated and untreated turkeys. Poly(I:C)+inactivated aMPV group had higher numbers of IgA+ cells than the untreated group or the groups given Poly(I:C) or inactivated aMPV alone. (U Minnesota)<br /> <br /> The availability of the complete genome information is essential for development of a reverse genetics system to study the molecular biology and rescue infectious ampv from cloned cdna. Therefore, we determined the nucleotide (nt) sequence of the complete genome of ampv-c colorado strain (ampv-c-co) propagated in vero cells in our laboratory (here designated as seprl variant). The full-length genome is comprised of 13,136 nt encoding eight genes, a 40 nt leader at its 3 end and a 45 nt trailer at its 5 end. It is two nt longer than the ampv-c-co strain propagated in the university of minnesota (umn variant, lwamba et al., 2005), and 1,014 nt shorter than the same strain of virus propagated in the university of maryland (umd variant, govindarajan and samal, 2005). The significant difference in length between these variants was found in the coding region of the g gene, where the seprl and umn variants were 1,015 nt or 333 amino acids (aa) shorter when compared with the umd variant. In addition, there were 23 nt differences scattered along the genome of the variants. Nine of them resulted in eight aa coding changes in five genes, three of which were located in the l gene. Based on the genomic sequence of the seprl variant, we developed a reverse genetics minireplicon system using a green fluorescence protein (gfp) gene as a reporter, which allowed us to assess the effects of coding differences in the l gene on viral gene expression. It was found that one of the coding differences (position 1371 leu vs phe) in the rna-dependent polymerase l gene was critical for the polymerase functionality. (SEPRL)<br /> <br /> Adherent cells from spleen, bone marrow and the circulation of normal turkeys were cultured in vitro. After 7 days, the cells were inoculated with the 63rd Vero cell passage of subtype C aMPV. At 96 hours following exposure, viral genome was detected by RT-PCR in the RNA extracts of virus exposed cells. Immunohistochemistry staining of the cells revealed the presence of intracellular viral proteins. Virus-exposed adherent cells had upregulation of nitric oxide production, iNOS gene and genes of several proinflammatory cytokines and chemokines. These results indicated that turkey macrophages were susceptible to infection and activation by aMPV63. (U Minnesota)<br /> <br /> The aMPV G protein is a major determinant for distinguishing virus subtypes and different lengths (and resulting changes in the predicted amino acid and nucleotide sequences) have been reported. Sequence analysis revealed that the complete 1.8kb G gene was found when aMPV was propagated in our immortalized turkey turbinate (TT-1) cells. In contrast, Vero cell propagated aMPV revealed an essentially deleted G gene in the viral genome, resulting in no G gene mRNA expression. The lack of expression was confirmed by Northern blot hybridization. As expected, viral G gene mRNA was not detected in the Vero-aMPV at any time post infection (p.i.), while the TT-1-aMPV showed increased levels of G gene mRNA in a time-dependent manner post infection. Both the TT-1-aMPV and Vero-aMPV templates were examined for the existence of splicing mRNA variants containing any partial G gene fragments using an RNase protection assay. After RNase digestion, the TT-1-aMPV showed a single mRNA transcript of approximately 1.8kb, while the Vero-aMPV did not show any detectable G gene fragments. The functional role of viral genes may be different depending on the species of the cellular host substrate. While the G protein may function as a key attachment protein in TT-1 cells, it appears not to be required for Vero cell infection. (U Minnesota)<br /> <br /> Infectious Bronchitis Virus<br /> From 1997 to 2002, the ARK type of IBV was the most dominant strain isolated from birds with respiratory disease submitted to the AL State Lab in Auburn. S1 gene analysis and challenge studies of these isolates indicated that they were closely related to vaccine strains. Experimental IBV vaccine and challenge studies with CAV and IBDV infections indicated that these 2 pathogens compromised IBV vaccine efficacy. (Auburn U)<br /> <br /> Newcastle Disease Virus<br /> A system to make Newcastle disease viruses with specific genetic changes was established to study Newcastle disease virus (NDV) infection in chickens. Different NDV strains cause variable clinical disease in chickens that ranges from severe to mild or in some cases inapparent infections. We have constructed a full-length copy of the genome of the NDV anhinga strain by combining each of the virus genes in an artificial system that allows virus replication to generate a virus that can be propagated and infect chickens just like the original field isolate. This system is being utilized to replace NDV anhinga genes with genes from other NDV strains that cause different clinical outcomes to identify which genes are important in controlling the severity and form of the clinical disease resulting from an NDV infection. The findings from application of this system will impact vaccine development and the identification of the role of the genes controlling the different clinical disease forms may impact other control strategies. (SEPRL)<br /> <br /> Domestic pigeons and other hobby birds can be infected with exotic Newcastle disease (END) virus, and this presents a risk of spread of the virus to poultry. The potential role of racing pigeons in that dissemination was examined by evaluating their susceptibility to infection and disease. Susceptible and Newcastle disease (ND) vaccinated pigeons were infected by eye drop and intranasally with an END virus isolate recovered during the 2002-03 END outbreak in the Southwestern U. S. Pigeons were readily infected and shed virus from both the respiratory and intestinal tract, but they were more resistant to disease with a virus dosage that would cause high mortality in chickens. Vaccination reduced the virus shed from infected pigeons and thereby reduced but didnt eliminate the risk of transmitting virus to other birds. The results provide a basis for establishing regulations concerning the vaccination as well as the movement and flying of racing pigeons in a quarantine zone during an END outbreak. (SEPRL)<br /> <br /> <br /> Infectious Bursal Disease Virus<br /> SPF chickens were exposed to virulent IBDV and bursal adherent cells were examined by immunohistochemisrty and RT-PCR for virus infection and by real-time quantitative RT-PCR (qRT-PCR) for mRNA transcripts of proinflammatory cytokines and iNOS. Viral genome was detected in bursal macrophages at 3, 5 and 7 days post-infection (dpi). Immunohistochemical staining revealed double positive cells for KUL01 (macrophage marker) and intracellular viral proteins, showing viral replication in bursal macrophages of infected chickens. We noted a significant decrease in the total number of bursal macrophages in infected chickens, probably due to the lysis of infected cells Inflammatory cytokines (IL-6, IL-1b and IL-18) were upregulated. These data suggested that B cells may not be the sole targets for the virus; macrophages and possibly other cells may serve as host for IBDV. (U Minnesota) <br /> <br /> Infection with infectious bursal disease virus (IBDV) causes activation of macrophages, the key cells involved in inflammatory and immune-regulatory functions. Exposure of cells of avian macrophage line, NCSU and cultured spleen macrophages (SM) from SPF chickens to IBDV resulted in the production of nitric oxide (NO). In addition, there was upregulation of gene expression of inducible nitric oxide synthase (iNOS), IL-8 and cyclooxygenase-2 (COX-2). The signal transduction pathways involved in macrophage activation were examined. The role of mitogen- activated protein kinases (MAPKs) and nuclear factor-ºB (NF-ºB) was tested by using specific pharmacological inhibitors. Addition of p38 MAPK inhibitor, SB-203580, and NF-ºB inhibitor Bay 11-7082, suppressed IBDV-induced NO production and mRNA expression of iNOS, IL-8 and COX-2. The results suggest that IBDV uses cellular signal transduction machinery, in particular the p38 MAPK and NF-ºB pathways, to elicit macrophage activation. The increased production of NO, IL-8 and COX-2 by macrophages may contribute to bursa inflammatory responses commonly seen during the acute IBDV infection. (U Minnesota) <br /> <br /> <br /> Objective 2. Surveillance, occurrence and consequences of agents and host <br /> variation on disease susceptibility.<br /> <br /> Avian Influenza<br /> The surveillance of commercial poultry, backyard poultry, wild birds and live poultry markets in New England for avian influenza continues as part of a USDA initiated program. A summary of results for the sampling period of Jan 1, 2006 to Dec 31, 2005 are as follows: 1632 samples were tested by real-time RT-PCR (RRT-PCR) from birds from live bird markets (LBMs) and backyard flocks; 1723 wild bird samples were tested by RRT-PCR; 3,779 blood samples from birds from LBMs were tested serologically. All samples were negative for avian influenza. Additional samples from LBMs were also tested by virus isolation in embryonating chicken eggs. (UConn)<br /> <br /> Surveillance of 200 samples from non-migrating and migrating wild water fowl for AIVs from Alabama, Georgia and Florida were conducted in 2006 using both net caught ducks and hunter killed ducks. Samples were from non-migrating wood ducks, and migrating hooded mergansers, blue-winged teal, gadwall, and ring-necked ducks. Twenty samples were from net caught adults ducks, whereas the rest were hunter killed ducks. Six of the samples produced HA positive results. However, only one sample was positive for AIV using real time RT-PCR and antigen capture ELISA. This sample was sent to the NVSL in Ames, Iowa and found to be a low pathogenic H10N7. We isolated and amplified the H10 gene and it is being sequenced. (Auburn U)<br /> <br /> Four viruses, three from turkeys and one from swine, were tested for their antigenic relatedness using Hemagglutinin Inhibition (HI) test and Virus Neutralization (VN) test in cell culture. The viruses are: TK/IL/04 (H3N2); TK/OH/03 (H3N2); TK/NC/03 (H3N2) and SW/NC/03 (H3N2).<br /> The formula of Archetti and Horsfall was employed to express the antigenic relatedness of the different isolates. Results showed that turkey isolates are highly related (71-100 % similar), however the swine isolate was distantly related from the others (< 30% similar to the turkey isolates). The genetic analysis revealed a high degree of similarity between the turkey virus isolates which were less similar to the swine isolate. All eight genes were more than 99% similar between the three different turkey isolates, however, genes from swine isolate were 94-96% similar to the turkey isolates genes. (Ohio State U)<br /> <br /> We conducted monitoring for avian influenza viruses in poultry and wild birds from samples from the U.S. and around the world. Avian influenza viruses are present in various wild birds and poultry throughout the world. Southeast Poultry Research Laboratory worked with several laboratories to monitor and study avian influenza viruses. No viruses were identified by molecular tests or were isolated from wild birds in Tunisia and Canada Geese in the USA. Some avian influenza (AI) viruses and Newcastle disease viruses (NDV) were obtained from samples from Iraq, Yemen and Nigeria. The Iraqi viruses included both virulent and non-virulent NDV, but antibodies to low pathogenic AI H9N2 viruses were detected. The Nigerian viruses were H5N1 high pathogenicity AI viruses and virulent Newcastle disease viruses. A virulent Newcastle disease virus was isolated from the Yemeni samples. These studies emphasize the need to continue to monitor poultry and wild birds worldwide for AI virus and NDV. (SEPRL)<br /> <br /> Avian influenza virus (AIV) surveillance in poultry (commercial and backyard) and wild birds is ongoing at University of Delaware's Lasher Laboratory and Allen Laboratory, respectively. No AIV activity using USDA NAHLN approved agent detection (real time RT-PCR and antigen capture on oropharyngeal swabs) or antibody detection assays was observed in over 15,000 active (pre-slaughter) or passive (clinical disease cases) surveillance samples in commercial broilers. One backyard duck flock was found to be positive by real time RT PCR. <br /> Wild bird surveillance was initiated at the Allen Lab in Newark in October 2006 in cooperation with the Delaware Department of Natural Resources & Environmental Control. Testing has yielded many real time RT-PCR positive cloacal samples. Only one H5 sample was identified but was determined by NVSL to be a non-Nl neuraminidase subtype. Virus isolation attempts on these samples are now being performed. Our ongoing collaboration with Dr. Richard Slemons (Ohio State University) and his research group yielded several AIV isolates from waterfowl and shorebirds from the Delmarva Peninsula region. The isolates will be characterized in poultry in laboratory trials. (U Delaware)<br /> <br /> <br /> Infectious Bursal Disease Virus (IBDV)<br /> Infectious bursal disease virus (IBDV) exists in several different antigenic and pathogenic forms. The immune suppression caused by this virus in young chickens is not always associated with clinical signs of disease. The antigenic Variant viruses originally described in the United States, typically do not cause clinical signs of disease but can cause a marked immune suppression via the destruction of B lymphocytes. Using a reverse-transcriptase polymerase chain reaction (RT-PCR) assay we conducted a survey of asymptomatic chicken flocks in Europe for IBDV. Restriction fragment length polymorphisms in the VP2 gene of four viruses from Spain and four viruses from France indicated they may be different from the Classic and very virulent (vv) IBDV strains found throughout Europe. Nucleotide sequence and phylogenetic analysis of the hypervariable region of the VP2 gene indicated that all eight viruses were more similar to U.S. Variant viruses than Classic viruses. In two viruses, one from France and one from Spain, Threonine was observed at amino acid position 222 and Serine was found at position 254. These two substitution mutations are characteristic of the Delaware Variant viruses. In addition, all eight viruses had mutated amino acid position 318 from Glycine to Aspartic acid; another substitution mutation commonly found in U.S. Variant viruses. Although importation restrictions prevented us from directly testing the antigenicity of these viruses, their nucleotide and predicted amino acid sequences strongly suggest they may be antigenically unique compared to Classic and vvIBDV commonly found in Europe. (Ohio State U)<br /> <br /> Infectious Bronchitis Virus<br /> Routine virus isolation attempts from respiratory disease accessions from Delmarva commercial broiler chickens yielded four isolates of a variant of IBV based on S1 gene sequencing. This variant (Fig. 1) is similar to a 2004 cecal tonsil isolate (K0401737 ct) from commercial broilers in California, recovered by Dr. Peter Woolcock's laboratory and sequenced by Dr. Mark Jackwood. The potential role of this variant to cause disease in vaccinated or unvaccinated chickens under laboratory conditions has not been established. Other field isolates from broilers were Arkansas, Massachusetts or Connecticut S 1 genotypes. (U Delaware)<br /> <br /> Objective 3. Develop new and improved methods for the diagnosis, prevention, and <br /> control of avian respiratory diseases.<br /> <br /> Avian Influenza <br /> Real time multiplex RT-PCR for avian influenza for subtypes H5, H7, and H9 and multiplex PCR or RT-PCR tests for Mycoplasmas and Infectious bronchitis infections were developed at the Guangxi Veterinary Research Institute Nanning, China. Plans are to continue to optimize the test and collaborate with the National Veterinary Services Laboratory (Ames, IA) and other experiment stations to test the new protocols. (UConn)<br /> <br /> A replication-defective adenovirus recombinant vaccine to protect chickens against avian influenza virus was developed that encoded the hemagglutinin gene from the low pathogenic avian influenza virus Turkey/Wisconsi/68 H5N9. The vaccine was given to SPF leghorns in ovo or at day of age by SQ route. The AdTW68.H5 vectored vaccine induced measurable HI (log29) titers against the LP turkey H5N9, but no titers using IDEXX ELISA, when given in ovo at 18 days or SQ at day of age. Thirty one day old vaccinated birds were challenged at the USDA SEPRL lab in Athens, GA with either the Mongolian HP H5N1 (89% hemagglutinin sequence homology) or Mexican H5N2 (94% hemagglutinin sequence homology) AIVs. The vaccine induced 68 % protection against the Asian and 100% against the Mexican virus. (Auburn U) <br /> <br /> Live-virus vaccines have distinct advantages over inactivated vaccines such as triggering mucosal immune responses and inducing a cell-mediated immunity, which may give the animal a more cross-protective and longer-lasting immunity. From the TK/OR/71-del (H7N3) virus, we previously found that several variants with different sizes of the NS gene can be generated by serial passage of the virus in embryonating chicken eggs. To create a H5 vaccine strain (since the selected variants are H7 subtype) that contains the selected NS gene, we utilized a traditional reassortment method. Briefly, D-del var1 and TK/WI/68 (H5N9) viruses were co-infected into 10-day-old embryonating eggs for reassortment. After 48 hrs of co-infection, infectious allantoic fluid was harvested, followed by intensive plaque purification of derivatives in CEF cells. Individual clones were examined for their gene composition by RT-PCR and sequencing. We obtained a H5-D-del-v1 variant which has the NS gene of D-del var1 and other remaining genes of TK/WI/68 virus. (Ohio State U)<br /> <br /> We are developing a microsphere-based multiplex assays as an alternative to RRT-PCR for the detection and subtyping of H5 and H7 subtype avian influenza virus. To accomplish this, we utilized branched DNA (bDNA) signal amplification technology (a sandwich nucleic acid hybridization assay) and microsphere-based assay for the detection of influenza viral RNA. The microshpere-based array system is a newly emerging technology that provides the multiplexing of up to 100 different assays within a single sample. In this study, we utilized this system coupled with branched DNA (bDNA) signal amplification technology (a sandwich nucleic acid hybridization assay) to detect and subtype H5 and H7 influenza virus. In our 3-plex assay, we were able to detect different HA subtype of influenza virus and differentiate H5 and H7 HA subtype at the same time based on capture probes specific for the M, H5, and H7 gene. In addition to multiplex capacity, this system does not require an RNA extraction step and samples can simply be treated with lysis buffer for the assay. (Ohio State U)<br /> <br /> In the last two years our laboratories have developed and successfully evaluated an avian influenza DNA microarray. This array contains 21 elements representing various avian influenza hemagglutinin (HA) and neuraminidase (NA) subtypes, as well as a pan-influenza probe, based on the matrix (M) gene sequence. These 21 elements are spotted in duplicate (42 spots) creating a "subarray". As a result of the subarray being spotted four times on each slide, each element is represented by 8 individual spots. Each subarray consists of a number of hermagglutinin, matrix, and neuraminidase genes ). The three matrix elements were derived from AIV strains containing three different HA subtypes. Six elements on the array represent three neuraminidase subtypes ( N1,N2, and N3). A DNA product of the Newcastle disease virus (NDV)fusion (F) gene is also included as a negative control. The majority of the array elements (9) correspond to hemagglutinin subtypes (HAS, HA7, HA9). The microarray was evaluated with a panel of 10 coded samples provided by Dr. Suarez. The results of the unknown panel test indicated 80% of the HA and NA subtypes were correctly identified, and all of the isolates were correctly identified as type A influenza. All of the neuraminidase subtypes were correctly identified with the exception of a N7. No N7 gene elements are present on the array. The H1 strain (A) was also incorrectly identified, and was also not represented on the array. (U Delaware)<br /> Fowlpox<br /> We continue to use polymerase chain reaction (PCR) and immunoblotting for differentiation of avianpox virus strains isolated from domestic poultry or wild birds. The presence of A-type inclusion (ATI) gene was detected in genomes of all strains. Common as well as different antigens were detected among various strains during immunoblotting analysis. (U Illinois)<br /> <br /> Avian Metapneumovirus (AMV)<br /> A sequencing project is almost complete to try and identify the genes related to virulence in aPMV. Once the genes are identified, a virus will be created by reverse genetics to remove that gene and create an improved vaccine based on reverse genetics. Infectious clones for aMPV have been developed by our collaborator Dr. Siba Samal at the University of Maryland. The viruses sequenced are: 1. MN-1a 9p: This virus was isolated from an outbreak of respiratory illness in turkeys in Minnesota in 1997. The virus was passaged in CEF for seven times and then twice in Vero cells; 2. MN-1a 41p: The virus MN-1a was passaged seven times in CEF cells and then 34 times in Vero cells; 3. MN-1a 63p: The virus MN-1a was passaged seven times in CEF cells and then 56 times in Vero cells; 4. MN-1a 65/Cp: The aMPV MN-1a after 41 passages in Vero cells was adapted to grow at cold temperature. It was passaged eight times each at 35ºC, 33ºC and 31ºC; and 5. MN-2a 7p: This virus was isolated from an outbreak of respiratory illness in turkeys in Minnesota in 1997 from a farm different from where MN-1a was isolated. This was passaged in CEF for seven times. (U Minnesota)<br /> <br /> Infectious Laryngotracheitis Virus<br /> <br /> ILTV is highly contagious pathogen of poultry that is often controlled by vaccination. For broilers mass vaccination techniques results in environmental contamination that can result in persistence in the house leading to back passage (bird to bird transfer) of the virus resulting in increased virulence. With reduced down time between lots, 5 days or less, and the use of built up litter, this condition is causing serious losses in the broiler belt in SE USA. In addition, there is not sufficient vaccine produced in the US on a yearly basis to vaccinate all the broilers in affected areas. Therefore, management practices to reduce the ILTV concentrations in chicken houses are needed. We developed a natural challenge method, using sentinel chickens reared in isolation units on reused litter contaminated with ILT back passed vaccine virus and a nested polymerase chain reaction (PCR) to determine the presence of ILT vaccine virus in the feces and tracheas of the chickens. Using these methods, we determined that several commercially available poultry litter treatments (Poultry GuardTM, Al+Clear TM, PLTTM), heating the litter to 38C0 (1000 F) for 24 h, and in house composting for 5 days inactivated ILT vaccine virus. This information is of immediate use to the poultry industry for controlling ILT vaccine virus induced disease in broilers and may reduce other important viral pathogens as well. (Auburn U)<br /> <br /> Infectious Bursal Disease Virus (IBDV)<br /> The effect of prime-boost on protection of chickens against infectious bursal disease by DNA vaccination was examined. Multiple intramuscular injections with a large dose of DNA carrying a large segment gene of the infectious bursal disease virus (IBDV) have been shown to provide effective protection to chickens against infectious bursal disease (IBD). The present study was conducted to determine if priming with DNA carrying a large segment gene of the IBDV and boosting with killed IBD vaccine could adequately confer protection of specific pathogen free (SPF) chickens against IBD. One-day-old chickens were intramuscularly injected with DNA plasmid coding for a large segment gene of the IBDV strain variant E (VE) (P/VP243/E) followed by an intramuscular injection of killed IBD vaccine containing both standard and variant IBDV at 1 or 2 weeks of age. Chickens were orally challenged with IBDV strain VE or standard challenge strain (STC) at 3 weeks of age and observed for 10 days. Bursal lesion scores, bursa weight/body weight (B/B) ratios, protection efficacy, IBDV antigen in bursae, enzyme-linked immunosorbent assay (ELISA) titers to IBDV, and virus neutralization (VN) titers to IBDV were determined. Chickens primed with 50, 100, 200, or 400 mg of P/VP243/E at 1 day of age and boosted with 0.5 ml of killed IBD vaccine at 1 or 2 weeks of age had 80 to 100% protection against challenge by IBDV strain VE or 71 to 100% protection against challenge by IBDV strain STC. Chickens in the groups primed with P/VP243/E and boosted with killed vaccine had significantly higher (P<0.05) B/B ratios and significantly lower (P<0.05) bursal lesion scores than chickens in the challenge control (CC) groups and groups primed with vector plasmid and boosted with killed IBD vaccine or only primed with P/VP243/E. No IBDV antigen was detected by immunofluorescent antibody assay (IFA) in bursae of chickens protected by the DNA vaccine prime and killed vaccine boost vaccination. Prior to challenge, chickens (21 days of age) in the groups primed with P/VP243/E and boosted with killed IBD vaccine had significantly higher (P<0.05) ELISA and VN titers to IBDV. These results indicate that SPF chickens at 1 day of age primed with a DNA vaccine and boosted with killed IBD vaccine can be adequately protected against challenge by homologous variant or heterologous classical IBDV. A prime-boost strategy may be useful in enhancing immunity and protection of chickens against IBD by DNA vaccination. (PurdueU)<br /> <br /> Mycoplasma<br /> Last year we reported the unusual Mycoplasma gallisepticum -685 highly attenuated vaccine strain, in which MG-685 percent invasion rate was 18.8 percent in to Chick embryo Fibroblast cells. In contrast MG-S6, MG-PG31, MG-IOIO and MG-r strain ranged from 0.45-5.6%. In order to more carefully document these phenomena we will use confocal microscopy using propidium iodide and cytodye 119 to gain more accurate information on M. gallisepticum 685 invasion of Chick embryo fibroblast cells. (U Delaware)<br />

Publications

1) Bennett, R.S., R. Larue, D. Shaw, Q. Yu, D.A. Halvorson, M. Kariuki, and M.K. Njenga. 2005 A Wild goose avian metapneumovirus containg a large attachment glycoproteins is avirulent but immunoprotective to domestic turkeys. J. Virol 79:14834-14842.<br /> <br /> <br /> 2) Boettger, C., and J. E. Dahms. Separating Mycoplasma gallisepticum Field Strains from Nonpathogenic Avian Mycoplasmas. Avian Diseases 50:605-607, 2006.<br /> <br /> <br /> 3) Chary, P, M.K. Njenga and J.M. Sharma. 2005. Protection by recombinant viral proteins against a respiratory challenge with virulent avian metapneumovirus. Veterinary Immunology and Immunopathology. 108:427-432.<br /> <br /> <br /> 4) Christman S.A., B.-W. Kong, M.M. Landry, H. Kim, and D.N. Foster. 2006 Contributions of differential p53 expression in the spontaneous immortalization of a chicken embryo fibroblast cell line. BMC Cell Biology, 7:27.<br /> <br /> <br /> 5) Donis, R., D.L. Suarez, D.E. Swayne. C.W. Lee., E. Spackman, et al. 2005. Evolution of H5N1 avian influenza viruses in Asia: antigenicity, antiviral drug sensitivity and vaccine development. Emerging Infectious Diseases. 10:1515-1521.<br /> <br /> 6) Gao, W., Soloff, A.C., Lu, X., Montecalvo, A., Matsuoka, Y., Robbins, P.D., Swayne, D.E., Donis, R.O., Katz, J.M., Barratt-Boyes, S.M., Gambotto, A., 2006. Protective vaccine for the rapid response to lethal Avian Influenza outbreaks. Journal of Virology. 80:1959-1964.<br /> <br /> 7) Gelb, J., Jr., B. S. Ladman, and C. Pope. Impact of respiratory virus vaccination on detection of avian influenza virus infection in broiler chickens. Proc. 143rd American Veterinary Medical Assn.lAmerican Assn. Avian Pathologist Ann. Mtg. Honolulu, Hawaii. July 15-19, 2006.<br /> <br /> 8) Hawkins, M.G. B. M. Crossley, A. Osofsky, R. J. Webby, C.W. Lee, D. L. Suarez, S. K. Hietala. 2006. H5N2 Avian Influenza A in a Red-lored Amazon parrot (Amazona autumnalis autumnalis) Journal of the American Veterinary Medical Association. 228: 236-241.<br /> <br /> 9) Hsieh, M.K., Wu, C.C., and Lin, T.L. 2006. The effect of co-administration of DNA carrying chicken interferon-g gene on protection of chickens against infectious bursal disease by DNA-mediated vaccination. Vaccine, 24: 6955-6965.<br /> <br /> 10) Hunt, H.D., R.M. Goto, D.N. Foster, L.D. Bacon, and M.M. Miller. 2006. At least one YMHCI molecule in the chicken is alloimmunogenic and dynamically expressed on spleen cells during development. Immunogenetics 58:297-307.<br /> <br /> 11) Khatri, M and J.M. Sharma. 2006. Infectious bursal disease virus infection induces macrophage activation via p38 MAPK and NF-kB Pathways. Virus Res. 118:70-77.<br /> <br /> 12) Khatri, M and J.M. Sharma. In Press. 2006. Modulation of macrophages by infectious bursal disease virus. Special Edition, CGR.<br /> <br /> 13) Khatri, M and J.M. Sharma. Submitted 2006. Activation of neonatal lymphoid cells following in ovo exposure to infectious bursal disease virus..<br /> <br /> 14) Khatri, M and J.M. Sharma. Submitted. 2006. Infectious Bursal Disease Virus Grown in Chicken Macrophage Cell line has Altered Tropism for Non-permissive Chicken Embryo Fibroblast Cells..<br /> <br /> 15) Khatri, M, J.M. Palmquist, Ra Mi Cha, and J.M. Sharma. 2005. Infection and activation of bursal macrophages by virulent infectious bursal disease virus. Virus Res 113:44-50.<br /> <br /> 16) Kim, S-H., J. Rowe, H. Fujii. R. Jones, B. Schmierer, B-W Kong, K. Kuchler, D. Foster, D. Ish-Horowicz, and G. Peters. 2006. Upregulation of chicken p15INK4b at senescence and in the developing brain. J. Cell Sci. 119:2435-2443.<br /> <br /> 17) Kong B.-W.,L.K. Foster, and D.N. Foster. 2006. Comparison of Avian Cell Substrates for Propagating Subtype C Avian Metapneumovirus Virus Res. 116:58-68.<br /> <br /> 18) Ladman, B. S., A. B. Loupos, and J. Gelb, JI. Infectious bronchitis virus S 1 gene sequence comparison is a better predictor of challenge of immunity in chickens than serotyping by virus neutralization. Avian Pathology 35:127-33. 2006.<br /> <br /> <br /> 19) Lee, C.W. D. A. Senne, and D. L. Suarez. 2006. Development and Application of Reference Antisera against 15 Hemagglutinin Subtypes of Influenza Virus by DNA Vaccination of Chickens. Clinical and Vaccine Immunology. 13:395-402.<br /> <br /> 20) Lee, C.W., D. E. Swayne, J.A. Linares, D.A. Senne, and D. L. Suarez. 2005. H5N2 Avian Influenza Outbreak in Texas in 2004: the First Highly Pathogenic Strain in the United States in 20 Years? Journal of Virology. 79:11412-11421.<br /> <br /> 21) Palmquist, J.M., M. Khatri, Ra Mi Cha, B. Goddeeris, B. Walcheck and J.M. Sharma. 2006. In vivo infection of chicken macrophages by virulent infectious bursal disease virus: Effects of infection on macrophage function. Viral Immunol. 2006 19:305-15.<br /> <br /> <br /> 22) Park, M., Steel, J., Garcia-Sastre, A., Swayne, D.E., Palase, P. 2006 Engineered viral vaccine constructs with dual specificity: Avian Influenza and Newcastle disease. Proceedings of the National Academy of Sciences. 103:8203-8206.<br /> <br /> 23) Patnayak, D.P., and Goyal, S.M. 2006. Duration of immunity engendered by a single dose of cold adapted strain of avian pneumovirus. Can. J. Vet. Res. 70:65-67.<br /> <br /> 24) Perdue, M.L., Swayne, D.E. Public Health Risk from Avian Influenza Viruses. Avian Diseases. 49(3):317-327, 2005.<br /> <br /> 25) Poxvirus Isolated from an Endangered Hawaiian Goose (Banta sandvisdcensis). Avian Diseases, 50:15-21 2006<br /> <br /> 26) Sapats, S. L., L. Trinidad, G. Gould, H. G. Heine, T. P. van den Berg, N. Eterradossi, D. Jackwood, L. Parede, D. Toquin and J. Ignjatovic. Chicken recombinant antibodies specific for very virulent infectious bursal disease virus. Arch. Virology 151:1551-1566. 2006.<br /> <br /> 27) Senne, D.A., D.L. Suarez, D.E. Stallnecht, J.C. Pedersen, B. Panigrahy. 2006 Ecology and Epidemiology of Avian Influenza in North and South America. Developments in Biologicals. 124:37-44.<br /> <br /> 28) Spackman, E., Stallknecht, D.E., Slemons, R.D., Winker, K., Suarez, D.L., Scott, M.A., Swayne, D.E. 2005. Phylogenetic Analyses Of Type A Influenza Genes In Natural Reservoir Species In North America Reveals Genetic Variation. Virus Research. 114:89-100.<br /> <br /> 29) Srinivasan, V., Schnitzlein, W.M. and Tripathy, D.N. Genetic manipulation of two fowlpox virus late transcriptional regulatory elements influences their ability to direct expression of foreign genes. Virus Research, 116:85-90, 2006.<br /> <br /> 30) Suarez, D.L. C.W. Lee, and D. E. Swayne. 2006 Avian Influenza Vaccination in North America: Strategies and Difficulties. Developments in Biologicals. 124:117-124.<br /> <br /> 31) Suarez, D.L. 2005. Overview of Avian Influenza DIVA Test Strategies. Biologicals 33:221-226.<br /> <br /> 32) Subler, K. A., C. S. Mickael and D. J. Jackwood. Infectious bursal disease virus-induced immunosuppression exacerbates C. jejuni colonization and shedding in chickens. Avian Dis. 50:179-184. 2006.<br /> <br /> 33) Swayne, D.E. Occupational and Consumer Risks from Avian Influenza Viruses. Developments in Biologics (Basel) 124:85-90, 2005<br /> <br /> 34) Swayne, D.E., Beck, J.R. Microassay for Measuring Thermal Inactivation of H5N1 High Pathogenicity Avian Influenza Virus in Naturally-Infected Chicken Meat. International Journal of Food Microbiology 108(2):268-271, 2006<br /> <br /> 35) Swayne, D.E., Pantin-Jackwood, M. Pathogenicity of Avian Influenza Viruses in Poultry. Developments in Biologics (Basel) 124:61-67, 2005.<br /> <br /> 36) Swayne. D.E., Lee, C.W., Spackman, E. 2006. Inactivated North American and European H5N2 avian influenza virus vaccines protect chickens from Asian H5N1 high pathogenicity avian influenza virus. Avian Pathology. 35:141-146.<br /> <br /> 37) Tiwari, A., Patanayak, D.P. and Goyal, S.M. 2006. Survival of two avian respiratory viruses on porous and nonporous surfaces. Avian Dis. 50:284-287.<br /> <br /> 38) Tiwari, A., Patnayak, D.P., and Goyal, S.M. 2006. Attempts to improve on a challenge model for subtype C avian pneumovirus. Avian Pathol. 35:117-121.<br /> <br /> 39) Xie, Z., Y. Pang, J. Liu, X. Deng, X. Tang, J. Sun and M. I. Khan. A multiplex RT-PCR for detection of type A influenza virus and differentiation of avian H5, H7 and H9 subtypes. Molecular and Cellular Probes. 20: 245-249. 2006.<br />

Impact Statements

  1. The first comprehensive biological characterization of H5N1 high pathogenicity avian influenza (HPAI) virus from wild birds were completed on viruses that came from Mongolia. Because the outbreak occurred where no poultry exist, this indicates the H5N1 HPAI virus spread into Mongolia by migrating wild birds, but is a virus that can infect poultry and cause severe disease. (SEPRL)
  2. Evaluation of the potential role of domestic pigeons and other hobby birds in the dissemination of Newcastle Disease has provided a basis for establishing regulations concerning the vaccination as well as the movement and flying of racing pigeons in a quarantine zone during an END outbreak. (SEPRL)
  3. The development of a reverse genetics model for Infectious Bursal Disease Virus with high efficiency of virus recovery will help delineate the pathogenesis of IBDV and that of polymicrobial interactions of IBDV and poultry respiratory diseases. (Purdue U)
  4. Using a nested polymerase chain reaction (PCR) to determine the presence of Infectious Laryngotracheitis vaccine virus in the feces and tracheas of the chickens, we determined that several commercially available poultry litter treatments (Poultry GuardTM, Al+Clear TM, PLTTM), heating the litter to 38C0 (1000 F) for 24 h, and in house composting for 5 days inactivated ILT vaccine virus. This information is of immediate use to the poultry industry for controlling ILT vaccine virus induced disease in broilers and may reduce other important viral pathogens as well. (Auburn U)
  5. These results indicate that SPF chickens at 1 day of age primed with a DNA vaccine and boosted with killed Infectious Bursal Disease vaccine can be adequately protected against challenge by homologous variant or heterologous classical IBDV. A prime-boost strategy may be useful in enhancing immunity and protection of chickens against IBD by DNA vaccination. (PurdueU) Gamma irradiation is not a practical intervention to reduce the risk of IBDV introduction via processed poultry. (Ohio State U)
Back to top

Date of Annual Report: 11/30/2007

Report Information

Annual Meeting Dates: 01/22/2008 - 01/23/2008
Period the Report Covers: 10/01/2006 - 09/01/2007

Participants

The annual meeting is scheduled for January 22 - 23, 2008. This section will be updated subsequent to the meeting.

Brief Summary of Minutes

The annual meeting is scheduled for January 22 - 23, 2008. This section will be updated subsequent to the meeting. See attached.

Accomplishments

Objective 1. Determine the pathogenesis and interactions of specific agents<br /> <br /> Improved understanding of Avian metapneumovirus (aMPV), an important viral disease of turkeys, epidemiology:<br /> SUMMARY: We are characterizing avian pneumovirus (APV) isolates to extend the capabilities for molecular epidemiology of the most important avian paramyxoviruses (APMV) infections. IMPACT: This work is important for determining APMV pathogenesis for improved control. <br /> <br /> SUMMARY: To examine public health implications of a MPV, similar to the aMPV, which was isolated from humans. It is important to examine if the human virus and aMPV cross species barriers. IMPACT: Turkeys developed clinical disease when exposed to the human virus.<br /> <br /> Improved control and eradication of avian metapneumovirus (aMPV):<br /> SUMMARY: We have shown that the virus causes a local immune response in the respiratory tract and that a non-infectious respiratory vaccine can be used to control the disease. IMPACT: This vaccine will reduce virus contamination of the environment and facilitate eradication. <br /> <br /> Characterization of the G gene variation among North American aMPV-C isolates:<br /> SUMMARY: Serial passage of aMPV-C in cell cultures and natural passage in turkeys led to truncation of the G gene. IMPACT: This may be a mechanism of virus evolution for survival in a new host or environment. <br /> <br /> Innate immunity influences host susceptibility to avian influenza viruses (AIV), an important worldwide cause of disease in birds, reptiles, and mammals:<br /> SUMMARY: Comparison of the innate immune response in chickens and ducks to H5N1 avian influenza showed a markedly different response between species. IMPACT: These studies emphasize the importance of innate immunity in birds and correlate increased pathogenicity of recent H5N1 viruses for wild waterfowl with an enhanced suppression of the host immune response.<br /> <br /> Intraspecies transmission of triple reassortant influenza viruses in turkeys:<br /> SUMMARY: We examined the intraspecies transmissibility of the A/turkey/OH/ 313053 in turkeys. The virus replicated in 90% of the inoculated turkeys and transmitted to more than 70% of the contact turkeys. The A/turkey/OH/313053 virus is highly infectious and transmissible in turkeys. IMPACT: Producers should be aware of the possible impact of this virus in their flocks.<br /> <br /> Reverse genetic studies of AIVs for improved vaccines:<br /> SUMMARY: We rescued one strain of Hemagglutinin (H) 3 Neuraminidase (N) 2 turkey viruses using reverse genetics techniques. All the genes of two other strains were cloned into the transcriptional vector PHH21 and the rescue experiments are underway. Six genes of the A/turkey/IL/04 were cloned into PHH21 and the work is continuing to clone 2 more genes. IMPACT: Using reverse genetics, a better understanding of the genetic basis for the virus and important biological activities will be gained and development of vaccines will be facilitated.<br /> <br /> Infection and transmission studies of the low pathogenic H5 subtype AIVs:<br /> SUMMARY: The wild bird H5 subtypes were found to replicate and transmit among poultry without clinical disease. IMPACT: Wild bird isolates will be useful in determining the molecular basis of interspecies transmission.<br /> <br /> SUMMARY: Low path avian influenza virus isolates representing H5, H7, H6, and H3 subtypes recovered from wild waterfowl and shorebirds on the Delmarva peninsula were not pathogenic for two-week-old meat type turkeys and broiler chickens based on clinical signs and microscopic lesions. IMPACT: However, the viruses were recovered from the trachea or cloaca. <br /> <br /> Improved control of Infectious Laryngotracheitis Virus (ILTV), and important pathogen of commercial chickens:<br /> SUMMARY: Live ILTV vaccine virus, which causes silent ILTV outbreaks, was present in drinking water lines and darkling beetles in commercial poultry houses. IMPACT: Poultry producers need to improved beetle control and remove biofilm in their drinker lines for improved ILTV control.<br /> <br /> SUMMARY and IMPACT: Infectious laryngotracheitis virus isolates from commercial broilers raised in Delmarva, North Carolina and Arkansas produced conjunctivitis, microscopic lesions (trachea and eyelid), and weight depression and in broiler chickens in laboratory trials.<br /> <br /> Infectious bronchitis virus (IBV) pathogenesis:<br /> SUMMARY and IMPACT: Infectious bronchitis virus field isolate, DMV/5642/06, obtained from Delmarva broilers, produced respiratory disease but not renal lesions in commercial broiler type chickens.<br /> <br /> Study of recent E. coli isolates from Delmarva poultry flocks: <br /> SUMMARY: Three hundred fifty E. coli isolates were characterized. E. coli are an important cause of disease in poultry. Specific virulence factors include tsh, iss, iucC, Intll and TraT. Tsh encodes for an autotransporter protein adhesin. Iss encodes for increased serum survival and is commonly seen in chickens with colibacillosis. This gene allows the bacteria to evade the host complement system by preventing the deposition of host proteins on the bacterial surface. Trat and Iss proteins prevent the formation of the membrane attack complex of the complement system that may contribute to serum resistance. iucC resides on the aerobactin operon and is involved in the iron transport system and lntll encodes for a class 1 integrase which may link with antibiotic resistance genes. IMPACTS: One hundred forty isolates were Ciprofloxacin resistant whereas 2 were intermediate. Nalidixic acid showed 101 susceptible while 40 were resistant. <br /> <br /> Objective 2. Surveillance, occurrence and consequenses of agents and host variation on disease susceptibility<br /> <br /> AIV surveillance in New England: <br /> SUMMARY: Through out NewEngland, clinical samples from commercial poultry, back yard poultry, and livebird markets were tested at the Connecticut Veterinary Medical Diagnostic Laboratory by serological and real time PCR specific for H5 and H7. IMPACT: All were negative for H5 and 7 using these tests. <br /> <br /> Multiplex real time reverse transcriptase (RT)-polymerase chain reaction (PCR) for improved AIV detection:<br /> SUMMARY: A Multiplex real time RT-PCR for avian influenza and subtypes H5, H7 and H9 was developed at the Guangxi Veterinary Research Institute Nanning. IMPACT: Multiplex real time RT-PCR is being tested on the North American AI isolates to confirm its sensitivity and specificity for improved AIV detection.. <br /> Real time RT multiplex PCR for improved detection of AIV subtypes and infectious bronchitis virus (IBV), a important cause of respiratory disease in chickens:<br /> SUMMARY: PCR inhibitors are a problem in some types of samples and cause false negative results. IMPACTS: Improving the specificity of rapid diagnostic tests and an internal control provides greater test assurance particularly for fecal or tissue samples.<br /> Indentifying disease resistance to AIVs<br /> SUMMARY: Lines of chickens were selected for high and low frequency of Mx gene expression. Chickens from each line were sent to the South Eastern Poultry Research Laboratory (SEPRL) for challenge with a highly pathogenic (HP) H5 isolate. Birds from Mx Asn 631 allele line showed significantly delayed mortality compared to birds from the M x Ser611 allele line. IMPACT: This study paves the way for selection of lines of chickens resistant to AIV. <br /> <br /> Comparing various AIV detection methods:<br /> SUMMARY: Cloacal swabs were collected from hunter killed or trap-nested wild ducks in the South Eastern USA. Three tested positive by Directigen antigen capture (AC) enzyme linked immunosorbent (ELISA). Five real time RT-PCR matrix-positive samples were negative for Newcastle Disease Virus (NDV) by the H inhibition test (HI) test. Five samples were submitted to National Veterinary Service Laboratory (NVSL) for subtyping. All were H1N1. The % identity of the H1 genes from the 5 isolates ranged from 85 to 98. IMPACT: Knowledge gained from these findings will be used for developing a more effective intervention program for AIVs. <br /> <br /> Isolation and Characterization of H1N1 triple reassortants influenza virus:<br /> SUMMARY: Search continues for unique influenza viruses in different species. Recently we isolated the first H1N1 triple reassortant virus from swine and are currently comparing it with recent H1N1 swine and human isolates. IMPACT: Viruses circulating in swine are a possible threat to commercial turkeys and it is important to recognize and characterize these isolates.<br /> <br /> Isolation of H3 AIVs from commercial turkeys:<br /> SUMMARY: In the past few years, we isolated H3N2 viruses from commercial turkey breeder flocks. Lately, we isolated these viruses from young commercial turkeys experiencing respiratory disease. IMPACT: This is the first report of the isolation of H3N2 viruses from commercial turkeys.<br /> <br /> Studies of Infectious Bursal Disease Virus (IBDV), an important cause of immunosuppression in commercial chickens, using viruses from four continents: <br /> SUMMARY: Some IBDVs associated with high mortality did not have the typical molecular characteristics of very virulent (vv) IBDV. IMPACT: In vivo studies are required to identify vv viruses.<br /> <br /> Characterization of IBDVs from four layer flocks in the United States:<br /> SUMMARY: Infectious bursal disease was observed in layer flocks and four viruses were isolates from these flocks. These viruses were genetically similar to classic vaccine viruses, but these viruses were pathogenic in SPF birds. IMPACT: In vivo studies are required to accurately assess pathogenicity of IBDV isolates since the genetic basis for virulence is not known.<br /> <br /> Comparison of VP4 sequence of different IBDVs:<br /> SUMMARY: Amino acid (aa) mutations in the VP4 region of the attenuated and pathogenic strains were not consistently associated with virulence. Four aa were observed to be consistent with the vv viruses. IMPACT: Further studies are needed on the molecular basis for IBDV biologic activities<br /> <br /> ILTV surveillance:<br /> SUMMARY and IMPACT: Surveillance activities on the Delmarva Peninsula have yielded infectious laryngotracheitis virus and infectious bronchitis virus isolates from commercial broiler chickens and avian influenza virus isolates from wild birds<br /> <br /> Objective 3. Develop new and improve methods for the diagnosis, prevention, and control of avian respiratory diseases <br /> <br /> Improved aMPV Vaccines:<br /> SUMMARY: We developed reverse genetics technology for aMPV-C research. IMPACT: This new technique provides us a powerful tool to genetically manipulate the virus for proper attenuation as a vaccine candidate as well as for performing pathogenesis studies. <br /> <br /> Improved method for IBDV detection:<br /> SUMMARY: Real-time RT-PCR detection and sequence analysis of the VP2 hyper variable region of Indian vvIBDV isolates indicated the value of real time RT-PCR in screening field samples for the presence of vvIBDV strains. IMPACT: The use of real-time RT-PCR for screening field strains is highly recommended.<br /> <br /> New IBDV Vaccines:<br /> SUMMARY: Effort has been devoted to study the co-administration of molecular adjuvants on immunomodulation and protection of chickens against IBD virus (IBDV) infection by DNA vaccination. . IMPACT: The vaccination strategy by priming the chickens with DNA vaccine encoding IBDV VP243 gene linked to chicken CRT gene has the potential to enhance immunity and protection of chickens against IBD for practical prevention and control of IBD in the field. <br /> <br /> SUMMARY: We examined the susceptibility of stem cells to infectious bursal disease virus (IBDV) and developed a non-pathogenic in ovo vaccine that stimulates humoral and cell-mediated immunity: <br /> IMPACT: A non-pathogenic protective in ovo vaccine was developed which induced strong humoral and cell-mediated immunity. Bone marrow mesenchymal stem cells were identified as new targets for IBDV replication. Susceptibility of these cells may contribute to immunosuppression caused by the virus<br /> <br /> New ILTV vaccine:<br /> SUMMARY: We intend to develop a new recombinant ILT vaccine based on tissue culture. Deleting mutant ITLV will allow for the vaccine virus to eliminate threat from vaccinal ILT outbreak. IMPACT: This vaccine could lead to better control of ILTV.<br /> <br /> SUMMARY and IMPACT: Three commercial chicken embryo origin vaccines provided protection against recent infectious laryngotracheitis virus field isolates representing RFLP genotypes 5 and 6. <br /> <br /> More rapid detection of ILTV:<br /> SUMMARY: A PCR-restriction fragment length polymorphism (RFLP) test determined that most of the ILTVs isolated from commercial poultry outbreaks in Alabama and Georgia were of vaccine origin. IMPACT: This is important because a diagnosis of live virulent ILTV could result in the loss of export market. This would cost millions of dollars in losses for commercial US poultry companies<br /> <br /> New AIV and IBV vaccines:<br /> SUMMARY: We developed and optimized the reverse genetic technology for avian influenza subtype H7 and H5 viruses for vaccine development and pathogenicity studies. IMPACT: In-ovo vaccination of 18 days old chicken embryos with a recombinant DNA specific for IBV S gene along with interferon as an adjuvant suggest a good protection against IBV infection. <br /> <br /> SUMMARY and IMPACT: Commercial vaccine containing modified live infectious bronchitis virus strains Massachusetts and Arkansas provided protection against a novel genotype, DMV/5642/06, recovered from broilers raised on the Delmarva Peninsula.<br /> <br /> SUMMARY: An adenovirus recombinant vaccine against avian influenza virus was used to immunize poultry by in ovo route. IMPACT: This is the first efficacious vaccine developed for the in ovo route. <br /> <br /> SUMMARY: A transgenic vaccine in edible yeast for poultry against AIV was developed. A vaccine in yeast could be propagated and given in mass by drinking water to poultry of all ages. IMPACT: Yeast are routinely given in the drinking water as a probiotic, in place of antibiotics to kill bacteria in commercial poultry<br /> <br /> SUMMARY: A commercial H3 vaccine strain of influenza virus was found to be antigenically and genetically very different from H3N2 viruses currently circulating in commercial turkeys. Field evidence has indicated lack of protection from egg production drops in flocks vaccinated with that vaccine when the birds were naturally exposed to field challenge with the H3N2 virus. IMPACT: Results point to the importance of using live virus strains that are antigenically similar to circulating field strains.<br /> <br /> NDV Vaccines<br /> <br /> Study of the antigenic and genetic differences among NDV vaccine strains: <br /> SUMMARY: NDV is a common cause of respiratory disease in commercial poultry. The current ND vaccines are formulated with virus strains isolated several years ago. However, the vaccine that was homologous with the challenge virus reduced virus shedding significantly more than the other vaccines. IMPACT: Results demonstrated that NDV vaccines formulated to be more closely related to potential outbreak viruses may provide better ND control by reducing virus transmission from infected birds.<br /> <br /> Improved AIV and NDV control: <br /> SUMMARY: Common cooking methods kill highly pathogenic avian influenza (HPAI) and Newcastle disease viruses in poultry. HPAI viruses can be present in the meat of infected poultry and a prior study demonstrated cooking was effective in killing an H5N1 HPAI virus. IMPACT: Proper cooking of poultry using the Food Safety Inspection Services salmonella standards would be effective in killing both AIVs and NDVs.<br /> <br /> For a summary of previous accomplishments and impacts please see: http://www.wisc.edu/ncra/impstate-NC1019.doc<br />

Publications

University of Minnesota, St. Paul, MN 55108. Submitted by J. M. Sharma<br /> 1. Antharaman, S., Chander, Y., Dhanasekaran, G., Samal, S., and Goyal, S.M. (2007) Comparativegenome analysis of virulent and cell culture adapted avian metapneumovirus (aMPV). Gobbles. 64: 4-6. <br /> 2. Cha, R., M. Khatri and J.M. Sharma. B Cell Infiltration in the Respiratory Mucosa of <br /> Turkeys Exposed to Avian Metapneumovirus. Avian Dis. 51:764-770. 2007.<br /> 3. Clark. K.J., D.F. Carlson, L. Foster, A. Geurts, B-W. Kong, D.N. Foster, and S.C. Fahrenkrug. 2007. Enzymatic engineering of the porcine genome with transposons and recombinases. BMC Biotechnology. 7:42.<br /> 4. Khatri, M and J.M. Sharma. Modulation of macrophages by infectious bursal disease virus. Review. Cytogenetic Genome Res. 117:388-393. 2007. <br /> 5. Khatri, M and J.M. Sharma. Replication of infectious bursal disease virus in macrophages and altered tropism of progeny virus. Veterinary Immunology and Immunopathology. 15: 106-115. 2007. <br /> 6. Khatri, M and J.M. Sharma. Activation of neonatal lymphoid cells following in ovo exposure to infectious bursal disease virus. Submitted 2007.<br /> 7. Khatri, M and J.M. Sharma. Susceptibility of chicken mesenchymal stem cells to infectious bursal disease virus. Submitted 2007. Khatri, M and J.M. Sharma. Infectious bursal disease virus of chickens upregulates the expression of Toll like receptors and MDA 5. Submitted 2007. <br /> 8. Kong B.-W., L.K. Foster, and D.N. Foster. 2007. Establishment of an immortal turkey turbinate cell line suitable for avian metapneumovirus propagation. Virus Research. 127:106-115.<br /> 9. Kong B.-W., L.K. Foster, and D.N. Foster. 2007. A method for the rapid isolation of virus from cultured cells. BioTechniques. In Press. <br /> <br /> Agricultural Research Program and School of Veterinary Medicine, Purdue University, Indiana. Submitted by Drs. C. C. Wu* and T. L. Lin<br /> 1. Wu, C.C., Rubinelli, P., Lin, T.L. 2007. Invited Minireview: Molecular detection and differentiation of infectious bursal disease virus. Avian Diseases, 51:515-526. <br /> 2. Hsieh, M.K., Wu, C.C., and Lin, T.L. 2007. Priming with DNA vaccine and boosting with killed vaccine conferring protection of chickens against infectious bursal disease. Vaccine, 25:5417-5427. <br /> 3. Hsieh, M.K., Wu, C.C., and Lin, T.L. 2007. A prime-boost approach to enhance DNA vaccination mediated protection against infectious bursal disease. The Proceedings of the 58th North Central Avian Disease Conference, Minneapolis, Minnesota (Page 6).<br /> <br /> University of Arkansas. Fayetteville, Ark. Submitted by Byung-Whi Kong<br /> 1. Kong B.-W., L.K. Foster, and D.N. Foster. 2007. Establishment of an immortal turkey turbinate cell line suitable for avian metapneumovirus propagation. Virus Research. 127:106-115.<br /> 2. Clark. K.J., D.F. Carlson, L. Foster, A. Geurts, B-W. Kong, D.N. Foster, and S.C. Fahrenkrug. 2007.<br /> 3. Enzymatic engineering of the porcine genome with transposons and recombinases. BMC Biotechnology. 7:42. <br /> 4. Kong B.-W., L.K. Foster, and D.N. Foster. 2007. A method for the rapid isolation of virus from cultured cells. BioTechniques. In Press. <br /> 5. Kong B.-W., L.K. Foster, and D.N. Foster. 2007. Species-specific deletion of the viral <br /> attachment glycoprotein of avian metapneumovirus. In Press. <br /> 6. Kong B.-W., S.C. Fahrenkrug and D.N. Foster. 2007. Application of the sleeping beauty transposon system to avian cells. Accepted with revisions to Animal Genetics.<br /> Scientific meetings:<br /> 1. Kong BW, Foster LK and Foster DN. Application of the Sleeping Beauty transposon system to avian cells. 2007. Annual Meeting of Poultry Science Association, San Antonio, TX . July. 16-18.<br /> <br /> Southeast Poultry Research Laboratory, Athens, Ga. Submitted by Laszlo Zsak<br /> 1. Yu, Q., Estevez, C.N., Kapczynski, D.R. 2006. Production and characterization of monoclonal antibodies produced against avian Metapneumovirus subtype C which react to the nucleocapsid protein. Avian Diseases. 50(3):419-424.<br /> 2. Yu, Q., Estevez, C.N. 2006. Development of a reverse genetics system for avian metapneumovirus subgroup C virus. In: Proceedings of the 5th International Symposium on Avian Corona- and Pneumoviruses and Complicating Pathogens, May 14-16, 2006, Rauischholzhausen, Germany. p. 6-15.<br /> <br /> University of Connecticut, Storrs, CT. Submitted by Mazhar I. Khan<br /> 1. Khan, M. I. Avian influenza surveillancs and testing program. The veterinarian place at: Tuft University MA. May 19, 2007.<br /> 2. Khan, M. I. Assessing avian influenza plans. North Atlantic Poultry Biosecurity and Pest Management Workshop, University of Connecticut. June 2, 2007.<br /> 3. Khan, M. I., Zhixun Xie, Jianbao Dong, Xiaofei Tang, Jiabo Liu, Yaoshan Pang, Xianwen Deng, and Zhiqin Xie. Sequence analysis and phylogenetic study of the entire genome of three avian influenza H9N2 subtypes from south China. 144th AVMA, Washington D.C., July19-23, 2007. <br /> 4. Khan, M. I., Sankhiros Babapoor, Jarslaw Fabis and Zhiqin Xie. Embryo vaccination against infectious bronchitis virus (IBV) challenge using recombinant DNA containing IBV-spike gene along with interferon type 1 as an adjuvant. 15th World Veterinary Congress, September 10-15, 2007. Beijing.<br /> 5. Babapoor, S., Zeinab Helal, Dipu M. Kumar and M. I. Khan. Rescue of an Avian Influenza subtype H7 vaccine candidate modified virus using reverse genetic technique. 79th Northeastern Conference <br /> on Avian Diseases, Lancaster, PA. September 19-20, 2007. <br /> <br /> Southeastern Poultry Disease Research Laboratory, Athens, Ga. Submitted by David Suarez,<br /> 1. Lee, C.W., Y.J. Lee, D.A. Senne, D., and D.L. Suarez. 2006. Pathogenic potential of North American H7N2 avian influenza virus: a mutagenesis study using reverse genetics. Virology. 353:388-395.<br /> 2. Desheva, J.A., X.H. Lu, A.R. Rekstin, L.G. Rudenko, D.E. Swayne, N.J. Cox, J.M. Katz, and A.I. Klimov. 2006. Characterization of an influenza A H5N2 reassortant as a candidate for live-attenuated and inactivated vaccines against highly pathogenic H5N1 viruses with pandemic potential. Vaccine 24(47-48):6859-6866.<br /> 3. Das, A. E. Spackman, D. Senne, J. Pedersen, and D. L. Suarez. 2006. Development of an Internal Positive Control for Rapid Diagnosis of Avian Influenza Viral Infections by Real-Time RT-PCR with Lyophilized Reagents. Journal of Clinical Microbiology. 44:3065-3073.<br /> 4. Scott, A., Zepeda, C., Garber, L., Smith, J., Swayne, D., Rhorer, A., Kellar, J., Shimshony, A., Batho, H. Caporale, V., Giovannini, A. The concept of compartmentalization. Office of International Epizootics Scientific and Technical Review 25(3):873-879, 2006.<br /> 5. Winker, K., K. G. McCracken, D. D. Gibson, C. L. Pruett, R. Meier, F. Huettmann , M. Wege , I. V. Kulikova, Y. N. Zhuravlev, M. L. Perdue, E. Spackman, D. L. Suarez and D. E. Swayne. 2007. Movement of Birds and Avian Influenza from Asia into Alaska. Emerging Infectious Diseases. 13:547-552. <br /> 6. Brown, J.D., Swayne, D.E., Cooper, R.J., Burns, R.E., Stallknecht, D.E. Persistence of H5 and H7 avian influenza viruses in water. Avian Diseases 51(Supplement):285-289, 2007. <br /> 7. Brown, J.D., D.E. Stallknecht, J.R. Beck, D.L. Suarez, and D.E. Swayne. 2006. Old Reservoirs, New Viruses: the Susceptibility of North American Ducks and Gulls to H5N1 Highly Pathogenic Avian Influenza Viruses. Emerging Infectious Diseases. 12:1663-1670.<br /> 8. Bublot, M., Le Gros, F.X., Nieddu, D., Pritchard, N., Mickle, T.R., Swayne, D.E. Efficacy of two H5N9-inactivated vaccines against challenge with a recent H5N1 highly pathogenic avian influenza isolate from a chicken in Thailand. Avian Diseases 51(Supplement):332-337, 2007.<br /> 9. Bublot, M., Pritchard, N., Cruz, J.S., Mickle, T.R., Selleck, P., Swayne, D.E. Efficacy of a fowlpox-vectored avian influenza H5 vaccine against Asian H5N1 highly pathogenic avian influenza virus challenge. Avian Diseases 51(Supplement):498-500, 2007.<br /> 10. Lee,C.W., Y.J. Lee, D. Swayne, D. Senne, J. Linares, D.L. Suarez. 2007. Assessing potential pathogenicity of avian influenza virus: current and experimental system. Avian Diseases. 51:260-263.<br /> The Ohio State University. Submitted by Dr. Mo Saif<br /> Refereed journals<br /> 1. Spackman, E., D.E. Swayne, D.L. Suarez, D.A. Senne, J.C. Pedersen, M.L. Killian, J. Pasick, <br /> K. Handel, S.P. Somanathan Pillai, C.W. Lee, D. Stallknecht, R. Slemons, H.S. p, and T. <br /> Deliberto. 2007. Characterization of low pathogenicity H5N1 avian influenza viruses from <br /> North America. J. Virol. 81:11612-19.<br /> 3. Jackwood, D. J. and S. E. Sommer-Wagner. Genetic characteristics of infectious bursal disease <br /> viruses from four continents. Virology 365:369-375. 2007.<br /> 4. Sreedevi, B. and D. J. Jackwood. Real-time reverse transcriptase-polymerase chain reaction <br /> detection and sequence analysis of the VP2 hypervariable region of Indian very virulent infectious bursal disease isolates. Avian Dis. 51:750-757. 2007.<br /> Abstracts<br /> 1. Pillai, S.P.S., H. Yassine, S. Jadhao, D.L. Suarez, Y.M. Saif, and C.W. Lee. Pathogenicity <br /> and antigenicity of different lineage of H3N2 viruses in turkeys. Midwest Poultry Consortium Research Summit/Annual Meeting, March 13, 2007, St. Paul, MN.<br /> 2. Pillai, S.P.S., H. Yassine, S. Jadhao, D.L. Suarez, M. Pantin-Jackwood, Y.M. Saif, and C.W. Lee. Do we need better vaccine for Ohio turkeys? OARDC Conference, April 19, 2007, Columbus, OH.<br /> 3. Pillai, S.P.S., D.L. Suarez, and C.W. Lee. Genetic and biological characterization of the H5N2 virus isolated from parrot. Proc. 144th AVMA Annual Convention, July 14-18, 2007, Washington, DC.<br /> 4. Lee, C.W. AI vaccine  A new approach. North Central Avian Disease Conference. March, 11-13, 2007, St. Paul, MN.<br /> 5. Lee, C.W., Y.M. Saif, M. Pantin-Jackwood, and D.L. Suarez. Development of live attenuated vaccine against avian influenza. March 26-29, 2007, Las Vegas, NV.<br /> 6. Lee, C.W. Influenza: Diagnostics, vaccine and interspecies transmission. Infectious Disease Interest Group, The Ohio State University, May 24, 2007, Columbus, OH.<br /> 7. Wang, L., S.P.S. Pillai, M. Strother, K. Hong, Y.M. Saif, M. Pantin-Jackwood, D.L. Suarez, and C.W. Lee. New way to develop live influenza vaccine candidate strains. Proc. 144th AVMA Annual Convention, July 14-18, 2007, Washington, DC.<br /> <br /> Auburn University, Auburn, Alabama. Submitted by Dr. J. J. Giambrone<br /> Published data: Scientific Meetings<br /> 1) Dormitorio, T. V., J.J. Giambrone, K. Guo and G. Hepp. Evaluation of methods for detecting influenza viruses in wild aquatic birds. PSA Annual Meeting. San Antonio, TX. July. 16-18. <br /> 2) Toro, Haroldo. RCA-free recombinant adenovirus vectored vaccine for mass administration against avian influenza. Annual AVMA meeting. Washington, DC. July 14-18. <br /> 3) Giambrone, J. J., Fagbohun, S., and K. Macklin. 2007. A natural challenge model for ILTV laboratory studies. Southern Conference on Avian Diseases. Atlanta, Ga. Jan. 23. <br /> Trade Journals<br /> 1) Giambrone, J. J. 2007. AI Keeps Threatening South East Asia. World Poultry. 23: 40-41.<br /> 2) Giambrone, J.J. 2007. Avian Influenza Research at Auburn University. Alabama Poultry. January/February.16.<br /> 3) Macklin, K., J. Hess, and J. J. Giambrone. 2007. Windrow composting as a disease preventative method. Alabama Poultry. January/February.23. <br /> 4) Macklin, K., and J. J. Giambrone. 2007. Eliminating LT from the farm. Alabama Poultry. Spring.21. <br /> <br /> University of Delaware, Newark, Del. Submitted by Jack Gelb<br /> <br /> Gelb, Jr., Jack, B. S. Ladman, M. J. Licata, M. H. Shapiro and L. R. Campion. Evaluating viral interference between infectious bronchitis virus and Newcastle disease virus vaccine strains using quantitative reverse transcription polymerase chain reaction. Avian Diseases. 51(4):2007 In press.<br /> <br /> Gelb, Jack, Jr., Brian S. Ladman, Conrad R. Pope, and Michelle K. Wood. Characteristics of a Novel Infectious Bronchitis Virus Isolate from Delmarva Broiler Chickens. Proc. 144th American Veterinary Medical Assn./American Assn. Avian Pathologist Ann. Mtg. Washington, D.C. July 14-18, 2007.<br /> <br /> Gelb, J., Jr., M. Ruano, B. Ladman, M. Troeber, L. Preskenis, C. Pope, and D. Bautista. Laryngotracheitis-pathogenicity and vaccine protection. Proc. 42nd National Meeting on Poultry Health and Processing. Ocean City, Maryland. p. 27-28. October 8-10, 2006.<br /> <br />

Impact Statements

  1. Grant obtained by project member(s): Wu, C.C. and Lin, T.L. Microalgal-based oral delivery system for poultry vaccines. Indiana 21st Century Research and Technology Grant. $150,000. 2007-2008.
  2. Grant obtained by project member(s): Wu, C.C. and Lin, T.L. Infectious bursal disease virus vector. Purdue University School of Veterinary Medicine Internal Competitive Grant. $8,000. 2007.
  3. Grant obtained by project member(s): US Poultry & Egg Association, Development of an immortalized chicken cell substrate for infectious laryngotracheitis virus (ILTV) propagation for vaccine production. B-W. Kong (PI). $110,000. 7/01/2007-6/30/2009
  4. Grant obtained by project member(s): Two years (2007-2008) AU Biogrant, $48,000, Development of Transgenic Vaccines against Influenza Virus (AIV) in Poultry. J.J. Giambrone, H. Wu, and N. Singh.
  5. Grant obtained by project member(s): One year. Alabama Agriculture Experiment Station Initiative Grant (2008). $61,985. The effectiveness of in-house composting in controlling coccidiosis and destroying avian influenza virus in poultry litter. K. Macklin and J. J. Giambrone
  6. Grant obtained by project member(s): One year (2007). USPE+A grant. $25,300. Management practices to minimize infectious laryngotracheitis in poultry. K. Macklin and J. J. Giambrone
  7. Grant obtained by project member(s): Effects of Concurrent Respiratory and Immunosuppressive Viral Infections on the Pathogenesis, Diagnosis and Potential Viral Mutations of Low-Path Avian Influenza Virus (H7N2) in Chickens and Turkeys. USDA NRI Avian Influenza CAP proposal, J. Gelb, Jr. Principal investigator . Co-investigators: B. S. Ladman, S. S. Cloud, C. R. Pope, J. K. Rosenberger, and D. Suarez. 2005-2008. $172,980.
  8. Grant obtained by project member(s): Generation of Infectious Bronchitis Virus from Cloned cDNA: Potential for Basic Studies and Vaccine Development. USDA, NRI. Principal investigator. V. Vakharia, U. Maryland. Co-investigator, J. Gelb, Jr. $360, 000 total for 3 years, 2004-2007; U. Delaware subcontract $110,040.
  9. For a summary of previous impacts please see: http://www.wisc.edu/ncra/impstate-NC1019.doc
Back to top

Date of Annual Report: 02/04/2008

Report Information

Annual Meeting Dates: 01/27/2009 - 01/28/2009
Period the Report Covers: 10/01/2007 - 09/01/2008

Participants

Brief Summary of Minutes

Please see attached minutes file.

Accomplishments

Publications

Impact Statements

Back to top

Date of Annual Report: 03/06/2009

Report Information

Annual Meeting Dates: 03/01/2009 - 03/01/2009
Period the Report Covers: 10/01/2004 - 09/01/2009

Participants

Brief Summary of Minutes

NC1019 termination report is attached as the minutes file.

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.