24.12.2014 Views

The Animal Health Research Center - University of Georgia College ...

The Animal Health Research Center - University of Georgia College ...

The Animal Health Research Center - University of Georgia College ...

SHOW MORE
SHOW LESS

You also want an ePaper? Increase the reach of your titles

YUMPU automatically turns print PDFs into web optimized ePapers that Google loves.

VScience in Service to <strong>Animal</strong>s SM<br />

30th Annual Report<br />

<strong>The</strong> <strong>Animal</strong> <strong>Health</strong> <strong>Research</strong> <strong>Center</strong>


Contents<br />

VMES Objectives 1<br />

Report <strong>of</strong> the Director 2<br />

VMES Financial Table 2<br />

<strong>Animal</strong> <strong>Health</strong> <strong>Research</strong> <strong>Center</strong> 3<br />

Bacterial and Parasitic Diseases 6<br />

Diagnostic Science 12<br />

Biomedical Science 14<br />

<strong>The</strong>rapeutics and Immunology 16<br />

Virology 18<br />

<strong>Research</strong> Contracts and Grants 20<br />

Administrators and Advisors 22<br />

VMES Faculty & <strong>Research</strong>ers 23<br />

Selected Publications 24<br />

Director: Dr. Harry W. Dickerson<br />

Managing Editor: Dr. Michael Mispagel<br />

Associate Editor: Dr. Lari M. Cowgill<br />

Designer: Brad Gilleland<br />

Illustrators: Kip Carter, Brad Gilleland, <strong>The</strong>l Melton<br />

Photographer: Christopher Herron<br />

Copyright © 2006 Veterinary Experiment Station,<br />

<strong>College</strong> <strong>of</strong> Veterinary Medicine, <strong>The</strong> <strong>University</strong> <strong>of</strong><br />

<strong>Georgia</strong>. No part <strong>of</strong> this publication may be reproduced<br />

without the permission <strong>of</strong> the publisher.<br />

www.vet.uga.edu/research/vmes/


V<br />

Objectives<br />

Science in Service to <strong>Animal</strong>s SM<br />

30th Annual Report<br />

July 1, 2005 to June 30, 2006<br />

<strong>The</strong> Veterinary Medical Experiment Station supports<br />

a wide range <strong>of</strong> research that impacts on many<br />

aspects <strong>of</strong> our lives; the food we eat and the clothes<br />

we wear, our physical, emotional, and economic<br />

health, and the quality <strong>of</strong> our environment. VMES<br />

research includes efforts to improve the productivity<br />

and health <strong>of</strong> poultry and livestock, to better the<br />

quality <strong>of</strong> life for companion animals, and to improve<br />

public health through disease surveillance.<br />

This year’s research is pr<strong>of</strong>iled in our 2005 - 2006<br />

VMES annual report.<br />

VMES funds help support short-term applied research<br />

that directly benefits the health <strong>of</strong> animals<br />

and livestock in <strong>Georgia</strong> and are used to develop<br />

extramurally funded research programs at the <strong>College</strong><br />

<strong>of</strong> Veterinary Medicine. Projects supported by<br />

VMES funds are evaluated for scientific merit, importance<br />

to animal health, consideration for experimental<br />

animal welfare, and their roles in meeting the<br />

research objectives <strong>of</strong> the VMES.<br />

Our objectives are as follows:<br />

• To improve the health and productivity <strong>of</strong> domestic<br />

livestock, poultry, fish, and other incomeproducing<br />

animals and wildlife through research;<br />

• To assist in preventing disease epidemics by providing<br />

laboratory resources and highly skilled scientific<br />

personnel;<br />

• To assist in protecting human health through the<br />

control <strong>of</strong> animal diseases transmissable to man;<br />

• To improve the health <strong>of</strong> companion animals,<br />

which serve to enrich the lives <strong>of</strong> humankind;<br />

• To train new scientists in animal health research<br />

in order to provide continuity and growth in this<br />

vital area <strong>of</strong> veterinary medicine.<br />

All programs and activities <strong>of</strong> the Veterinary Medical Experiment Station are conducted<br />

without regard to race, color, national origin, age, sex, or handicap.<br />

Enhancing animal production, pr<strong>of</strong>itability, and well-being by improving animal health.<br />

Published by the Veterinary Medical Experiment Station, <strong>The</strong> <strong>University</strong> <strong>of</strong> <strong>Georgia</strong>.


Report <strong>of</strong><br />

the Director<br />

In this, the 30th Annual Report <strong>of</strong> the Veterinary Medical Experiment Station (VMES), I am pleased to introduce the <strong>Animal</strong> <strong>Health</strong><br />

<strong>Research</strong> <strong>Center</strong>, which was opened for occupancy in 2006. As you will read in the accompanying article, the road from conception to<br />

construction and finally to commissioning and occupancy has been long. It was well worth the wait and effort, however, as the AHRC<br />

provides the <strong>College</strong> <strong>of</strong> Veterinary Medicine and the <strong>University</strong> <strong>of</strong> <strong>Georgia</strong> with an unprecedented capacity to conduct research on<br />

infectious pathogens <strong>of</strong> humans and animals in state-<strong>of</strong>-the-art biocontainment facilities. <strong>The</strong> AHRC now houses the research laboratories<br />

<strong>of</strong> Dr. Ralph Tripp, <strong>Georgia</strong> <strong>Research</strong> Alliance Eminent Scholar in <strong>Animal</strong> <strong>Health</strong> Vaccine Development, as well as the laboratories<br />

<strong>of</strong> the infectious disease researchers Drs. Mark Tompkins, Jeff Hogan, and Thomas Hodge. This team <strong>of</strong> researchers is developing<br />

vaccines and therapeutics for a number <strong>of</strong> important viral diseases including influenza, severe acute respiratory syndrome (SARS)<br />

corona virus, and AIDS. <strong>The</strong> containment facilities for animals on the first floor are scheduled for completion and commissioning<br />

in 2007.<br />

In addition to funding basic and applied research projects, the VMES budget continues to support research and training related to both<br />

animal and human health. Veterinary research has an impact on many biomedical fields, and major support for research on animal<br />

models <strong>of</strong> human disease is available from federal agencies such as the National Institutes <strong>of</strong> Medicine. In addition, the National Science<br />

Foundation, and the United States Department <strong>of</strong> Agriculture provide funding for basic and applied animal research, respectively.<br />

Although competitive research dollars are available from some non-federal sources such as the Morris <strong>Animal</strong> Foundation, research<br />

funding that targets companion animals, including horses, is very limited. Thus, the continued commitment from the State <strong>of</strong> <strong>Georgia</strong><br />

to support research on animal health is a critically important investment. <strong>The</strong> companion and food animal industries <strong>of</strong> the State <strong>of</strong><br />

<strong>Georgia</strong> are a major component <strong>of</strong> the State’s economy. For example, sales <strong>of</strong> livestock, poultry and their products account for more<br />

than half <strong>of</strong> <strong>Georgia</strong>’s annual farm income. Protection <strong>of</strong> these resources is paramount to our State’s continued good economic health.<br />

<strong>The</strong> 30th VMES Annual Report provides an overview <strong>of</strong> peer-reviewed, competitive VMES-funded projects conducted during fiscal<br />

year 2006 (July 1, 2005 – June 30, 2006). Additional information on any <strong>of</strong> these projects can be requested by contacting the VMES<br />

<strong>of</strong>fice by phone, email or website, or directly from the investigators themselves. A list <strong>of</strong> publications is provided as well. <strong>The</strong>se<br />

peer-reviewed papers represent a selection <strong>of</strong> VMES supported work and other scholarly research originating at the <strong>College</strong> <strong>of</strong> Veterinary<br />

Medicine.<br />

A summary <strong>of</strong> the <strong>College</strong>’s research funding is provided below. Over the past year approximately four research dollars were leveraged<br />

for each VMES dollar invested.<br />

Harry W. Dickerson<br />

Funding Source FY 2002 FY 2003 FY 2004 FY 2005 FY2006 FY2007 FY2008<br />

VMES/VMAR Expenditures $3,927,297 $3,672,210 $3,380,261 $3,094,649 $3,148,784 $3,249,577 $3,304,117<br />

Federal Grants and Contracts 6,962,300 4,768,808 5,624,962 5,746,363 5,850,096<br />

State Grants and Contracts 4,563,272 4,434,171 3,872,763 4,688,817 4,482,741<br />

Private Grants and Contracts 1,446,110 715,974 1,677,282 2,221,052 2,075,430<br />

2 www.vet.uga.edu/research/vmes/


<strong>Animal</strong> <strong>Health</strong><br />

<strong>Research</strong> <strong>Center</strong><br />

<strong>The</strong> <strong>Animal</strong> <strong>Health</strong> <strong>Research</strong> <strong>Center</strong> (AHRC) at the <strong>College</strong> <strong>of</strong> Veterinary Medicine is a 75,000 sq. ft. free-standing building designed<br />

for the study <strong>of</strong> highly infectious diseases <strong>of</strong> animals, animal models <strong>of</strong> human disease, and hazardous chemical problems. <strong>The</strong><br />

laboratories and animal holding capacities will accomodate interdisciplinary work among colleges. <strong>The</strong> completion <strong>of</strong> a high containment<br />

facility in the academic environment is very timely and long overdue considering the recent spate <strong>of</strong> emerging diseases and<br />

looming threats <strong>of</strong> bioterrorism.<br />

As early as 1978, the faculty <strong>of</strong> the <strong>College</strong> <strong>of</strong> Veterinary Medicine<br />

recognized this need for a biocontainment laboratory and multispecies<br />

animal holding facility for research and training in the investigation<br />

<strong>of</strong> emerging and re-emerging biological threats to human<br />

and animal health. Dr. John Bowen, Associate Dean for <strong>Research</strong><br />

at the college, planned and secured initial funding for the building<br />

through the 1980’s and early 1990’s. Construction took place<br />

from 1996 through 1998 with $16 million secured from state and<br />

federal sources. Nevertheless, due to funding shortfalls and design<br />

and construction deficiencies, the building remained unfinished and<br />

unoccupied. No progress toward completion <strong>of</strong> the facility was made<br />

until 2000, at which time the <strong>Georgia</strong> State Finance and Investment<br />

Commission (GSFIC) contracted with the consulting firm Boyken<br />

International, Inc. to do a cost analysis for completing and commissioning<br />

the facility. An end-user steering committee comprised <strong>of</strong><br />

UGA researchers, staff, and administrators was assembled to critically<br />

review renovation and construction concepts. In December, 2002,<br />

GSFIC hired a design/build team to validate the plan and to carry<br />

out the required design and construction. <strong>The</strong> design-build team, Gilbane Inc. and CUH2A-Smith Carter, with extensive experience<br />

in complex biomedical research laboratory design and construction, including high-level containment facilities, was hired to construct<br />

the facility with a stated cost-limitation <strong>of</strong> $35 million from various state and <strong>University</strong> sources. <strong>The</strong> new design addressed current<br />

containment requirements, original deficiencies, and new programmatic requirements <strong>of</strong> the <strong>College</strong>. Most <strong>of</strong> the original mechanical<br />

systems were removed and replaced with up-to-date technology.<br />

This structure is a high-containment laboratory and animal holding facility<br />

consisting <strong>of</strong> 8 animal rooms (2 large and 6 smaller) capable <strong>of</strong> holding multiple<br />

species <strong>of</strong> animals including rodents, poultry, and livestock. On a scale <strong>of</strong> 1 to<br />

4 recognized biosafety levels, these spaces are categorized as Biosafety Level<br />

3 Agriculture (BSL-3 Ag), one step below the highest containment category <strong>of</strong><br />

BSL-4. An additional 6 animal rooms for rodents, caged animals and penned<br />

animals are rated at a lesser classification <strong>of</strong> <strong>Animal</strong> BSL-3. <strong>The</strong>re is complete<br />

support space for the animal facility such as cage and rack washing and sterilization<br />

equipment, and a pathology suite designed for large and small animals.<br />

Carcass disposal is handled with a 500 lb/hr pathological waste incinerator and a<br />

tissue digester whose effluent is recycled via composting. All liquid waste effluent<br />

is sterilized on-site in a waste water treatment plant before being discharged<br />

to the sanitary sewer.<br />

VMES 2006 3


<strong>Animal</strong> <strong>Health</strong><br />

<strong>Research</strong> <strong>Center</strong><br />

Containment <strong>of</strong> infectious organisms is ensured by a series <strong>of</strong> engineering<br />

designs including sterilizing high efficiency particulate air<br />

filters (HEPA) for the supply air and double HEPA-filtered exhaust air;<br />

directional air flows; differential pressures within containment zones;<br />

special elastomeric wall coatings that are 2.25 mm thick (vs. 0.18 mm<br />

for typical epoxy paint) ensuring virtually air-tight construction; showerout<br />

facilities for each animal room; decontamination and sterilization<br />

procedures for equipment and solid/liquid waste streams. Extensive<br />

commissioning and validation <strong>of</strong> systems are undertaken to assure the<br />

operational capabilities <strong>of</strong> the facility. <strong>The</strong> building is equipped with<br />

multiple security devices throughout.<br />

<strong>The</strong> facility also boasts a BSL-3 enhanced laboratory suite composed <strong>of</strong><br />

five laboratories (1 large, 4 small) for work with highly infectious animal<br />

pathogens. In addition, the second floor has seven BSL-3 laboratories to<br />

study infectious diseases in addition to the five standard BSL-2 laboratories<br />

located there. <strong>The</strong> second floor <strong>of</strong> the building also houses a vaccine<br />

development laboratory for small scale development <strong>of</strong> animal vaccines.<br />

When completely commissioned, the <strong>Animal</strong> <strong>Health</strong> <strong>Research</strong> <strong>Center</strong> at <strong>The</strong> <strong>University</strong> <strong>of</strong> <strong>Georgia</strong> will rank as a world class facility<br />

meeting or exceeding the biocontainment guidelines promulgated by NIH and the USDA/ARS.<br />

<strong>Research</strong> in the <strong>Animal</strong> <strong>Health</strong> <strong>Research</strong> <strong>Center</strong><br />

<strong>The</strong> majority <strong>of</strong> emerging infectious diseases (EID) are <strong>of</strong><br />

zoonotic origin, i.e. transmissible between humans and animals<br />

causing infection in both species. Many <strong>of</strong> the most dangerous and<br />

easily transmitted <strong>of</strong> these agents are viruses and bacteria. <strong>The</strong>se<br />

agents impact global security by affecting food for an increasing<br />

world population, access to international trade and economic<br />

growth, and raise concerns for potential use as pathogens in<br />

bioterrorism. Biodefense is at the extreme end <strong>of</strong> the spectrum <strong>of</strong><br />

serious disease threats. However, the emergence <strong>of</strong> new agents,<br />

the resurgence <strong>of</strong> old diseases, the appearance <strong>of</strong> resistant forms,<br />

and the recognition <strong>of</strong> viral and bacterial agents in the etiology <strong>of</strong><br />

chronic diseases all support the need for an integrated program<br />

that can address both short- and long-term needs. <strong>The</strong>se needs will<br />

involve basic and applied public health research, will be multidisciplinary<br />

in nature, and will utilize modern and robust molecular<br />

and quantitative tools and facilities.<br />

Some progress has been made in identifying disease-causing<br />

agents <strong>of</strong> zoonotic origin, such as rodent-borne viruses like the<br />

hantaviruses and arenaviruses. Nevertheless, there is an acute need for a comprehensive approach to identifying, preventing and controlling<br />

viral and bacterial zoonotic EID. In the past few years the world has had to respond to SARS-associated coronavirus identified<br />

in domestic and wildlife species, influenza viruses from birds, the West Nile virus from birds via mosquitoes, E. coli O157:H7<br />

from spinach contaminated with animal or human feces and a variety <strong>of</strong> multi-drug resistant bacteria acquired from food animals and<br />

wildlife. It is imperative that those in human, animal, agricultural and environmental sciences work together to address EID threats<br />

that will undoubtedly continue to pose a significant threat to human and animal health in the future. Thus, it is an important time to<br />

open the AHRC to initiate studies.<br />

4 www.vet.uga.edu/research/vmes/


<strong>The</strong> AHRC will serve as a multi-disciplinary platform to facilitate<br />

viral and bacterial EID, select agent research, and product development<br />

among investigators at UGA, from outside academic<br />

and public health institutions, as well as by private companies.<br />

Personnel affiliated with the AHRC will consist <strong>of</strong> veterinarians,<br />

virologists, bacteriologists, immunologists, cell biologists,<br />

biochemists, pharmacists, physicists, chemists, epidemiologists,<br />

statisticians, graduate students and laboratory technologists from<br />

UGA, as well as adjunct faculty and collaborators from neighboring<br />

organizations. <strong>The</strong> purpose <strong>of</strong> the AHRC will be to draw on<br />

UGA’s strengths, commitments and resources to create a worldclass<br />

research organization for disease intervention strategies,<br />

provide a means to capture extramural funding opportunities,<br />

facilitate collaborations that lead to new program projects, grants,<br />

and contracts, and provide a conduit for development <strong>of</strong> small<br />

businesses from the research findings. Furthermore, development<br />

<strong>of</strong> AHRC research programs will provide unparalleled expertise<br />

and facilities, and position UGA to take a leadership role in this<br />

critically important public health area.<br />

A Plethora <strong>of</strong> Opportunities<br />

• <strong>The</strong> AHRC will assist investigators working on the national public health agenda which is to expand research on the biology,<br />

diagnostics, therapeutics and prevention <strong>of</strong> diseases caused by infectious organisms. <strong>The</strong> emergence and reemergence <strong>of</strong> infec<br />

tious diseases in animals and man (zoonoses) indicates this agenda will continue into the foreseeable future.<br />

• Increases are expected in extramural research support for all academic units that use this facility.<br />

• <strong>The</strong> AHRC will expand capabilities for ongoing work at the <strong>University</strong> <strong>of</strong> <strong>Georgia</strong> including research on SARS, avian influenza,<br />

avian Newcastle, West Nile virus, tuberculosis, glanders, tularemia, botulism, encephalitic viruses, and other pathogens <strong>of</strong> public<br />

health importance.<br />

• <strong>The</strong> prospect <strong>of</strong> working in this facility is attracting infectious disease researchers <strong>of</strong> the highest quality.<br />

• <strong>The</strong> AHRC has unique capabilities to support research on agricultural diseases and to provide emergency response capabilities for<br />

foreign animal disease outbreaks such as exotic Newcastle disease <strong>of</strong> poultry and foot and mouth disease <strong>of</strong> ho<strong>of</strong>ed animals.<br />

• <strong>The</strong> extramural dollar flow generated by research in the AHRC will augment <strong>Georgia</strong>’s economy at local and state levels.<br />

• <strong>The</strong> research in this facility will result in new, commercialized biological products such as diagnostic reagents, vaccines, and<br />

drugs. For instance, one <strong>of</strong> the largest generators <strong>of</strong> royalty income to the <strong>University</strong> is a drug for the treatment <strong>of</strong> dry-eye<br />

disease that was created through research at the <strong>College</strong> <strong>of</strong> Veterinary Medicine. We expect these sorts <strong>of</strong> contributions to expand<br />

through research conducted at the AHRC.<br />

• A primary focus <strong>of</strong> the programs in the AHRC will be the training <strong>of</strong> scientists and forging <strong>of</strong> industrial collaborations for the dissemination<br />

<strong>of</strong> laboratory research. <strong>The</strong> program already has in place agreements for research and exchange with 19 industrial<br />

partners that include Merck-Merial, Numico <strong>Research</strong>, Pty., Dharmacon, Nomadics, Protiva, Trellis Bio, Ventria, Virax, MedIm<br />

mune, Aerovectrx, Sporian, Smithfield Foods, Protiva, nGimat, Millennium, Lentigen,, BD Biosciences, Creare, and<br />

Alnylam Pharmaceuticals.<br />

• <strong>The</strong> AHRC will facilitate interdisciplinary collaborations among regional biomedical research institutions. As a direct result <strong>of</strong><br />

the AHRC, the following relationships have evolved: the <strong>College</strong> is one <strong>of</strong> the initiating partners in the Southeast <strong>Center</strong><br />

for Emerging Biological Threats; research ties with the <strong>Center</strong>s for Disease Control and Prevention are increasing;<br />

UGA is affiliated with the Southeast Regional <strong>Center</strong> <strong>of</strong> Excellence for Biodefense and Emerging Infectious Disease <strong>Research</strong>;<br />

and collaborations are increasing with the USDA/ARS Southeast Poultry <strong>Research</strong> Laboratory.<br />

• <strong>The</strong> AHRC will be a critical resource for a direct response against bioterrorism and other emergencies involving<br />

infectious organisms.<br />

• <strong>Research</strong>ers at the <strong>College</strong> are involved in the development <strong>of</strong> novel vaccines and therapies against diseases <strong>of</strong> state and national<br />

importance, namely tuberculosis, SARS, avian influenza, RSV and glanders. <strong>The</strong> AHRC is essential for this to occur.<br />

VMES 2006 5


Bacterial and<br />

Parasitic Diseases<br />

Clinical Investigation <strong>of</strong> Poultry Diseases<br />

Two studies were completed under this project. <strong>The</strong> first was to determine is there is a potential interaction between turkey coronavirus<br />

and reticuloendotheliosis virus. <strong>The</strong> objective <strong>of</strong> this experiment was to co-infect SPF turkeys with turkey coronavirus and reticuloendotheliosis<br />

virus obtained from field cases to determine if there is a potential interaction between the two. Body weights <strong>of</strong> the<br />

experimental groups were compared to controls to determine weight reduction after infection. <strong>The</strong> digestive tract and immune system<br />

tissues were examined microscopically for lesions. RT- PCR was performed on intestinal contents <strong>of</strong> individual turkeys to detect the<br />

presence <strong>of</strong> TCV RNA and to assess virus persistence in the gut. Virus isolation was performed from plasma to demonstrate viremia<br />

induced by REV. <strong>The</strong> combination REV-infected and TCoV-infected birds suffered more severe weight depression than the control<br />

birds or the birds infected with REV or TCoV alone. Viral persistence in the gut was variable.<br />

<strong>The</strong> second study involved the investigation <strong>of</strong> alternative routes (oral, eye drop or wing web) <strong>of</strong> vaccination in poultry. Broiler breeder<br />

pullet vaccination for fowl cholera via wing web route is costly due to labor and can result in stress and injuries to vaccinated birds.<br />

Pullets vaccinated with live vaccine by wing web route also risk acquiring the disease contributing to joint problems, morbidity, and<br />

death. <strong>The</strong>refore in 2004, in an effort to investigate alternative routes <strong>of</strong> vaccination, Bruzual et. al, evaluated the use <strong>of</strong> a commercial<br />

Pasteurella multocida PM-1 vaccine administered by the drinking water in broiler breeder pullets versus administration by wing web.<br />

It was concluded that there was no protection by administration <strong>of</strong> a 6X dose <strong>of</strong> PM-1 orally in pullets whereas those given the vaccine<br />

by the w.w. route were protected. In this investigation, oral, eye drop, and wing web vaccination will be evaluated using the less attenuated<br />

CU (Clemson <strong>University</strong>) strain <strong>of</strong> Pasteurella multocida. Both post-vaccination mortality and post-challenge mortality after<br />

inoculation with a highly virulent serotype 1 Pasteurella multocida will be compared between treatment groups vaccinated orally, by<br />

eye drop, and via wing web with the CU strain and a group vaccinated with a commercial PM-1 strain via wing web application.


During the first trial, it was concluded that CU vaccine at commercial titer levels would not provide protection in birds vaccinated<br />

orally or by eyedrop. In the second trial, it was concluded that there was an improvement in the amount <strong>of</strong> protection from eyedrop<br />

administration <strong>of</strong> CU vaccine (54% protection), however, the vaccine administered was contaminated and therefore, the exact titer<br />

level that provided that protection could not be determined.<br />

PI: Dr. Charles H<strong>of</strong>acre (ch<strong>of</strong>acre@uga.edu)<br />

CO-PI: Dr. Guillermo Zavala and Dr. Stephen Collett<br />

A molecular ecology approach for the design <strong>of</strong><br />

effective probiotic formulations<br />

<strong>The</strong> goal <strong>of</strong> this project was to use an ecological approach to<br />

create a strategy for identifying and utilizing putative probiotic<br />

strains. <strong>The</strong> central hypothesis is that by assessing the development<br />

<strong>of</strong> the bacterial community structure <strong>of</strong> the small intestine<br />

we can identify relevant symbiotic bacteria that may improve<br />

intestinal health and feed conversion.<br />

Objective 1. Identify and isolate candidate probiotic strains <strong>of</strong><br />

Lactobacillus and avian-specific anaerobes from the bacterial<br />

community <strong>of</strong> healthy broiler chickens.<br />

Objective 2. Characterize the development <strong>of</strong> the small intestinal<br />

bacterial community resulting from oral administration <strong>of</strong><br />

candidate probiotic formulations.<br />

We performed several culture-based isolations <strong>of</strong> avian anaerobes from the ileum <strong>of</strong> broiler chickens in order to isolate clostridial<br />

species related to clusters IV, XI and XIV. We had detected the 16S rDNA sequences <strong>of</strong> these species from birds fed growth-promotants<br />

and in a commercial probiotic and had hypothesized that they may function as potential intestinal symbionts. <strong>The</strong> bacteria were<br />

cultured on 3 different media (blood agar, reinforced clostridium medium, and Wilkins-Chalgren agar) and incubated under several<br />

different atmospheres (80% nitrogen/20% CO2 or 100%CO2). Individual colonies were subcultured and characterized by 16S rDNA<br />

sequencing in order to determine their identity.<br />

We detected bacterial species such as lactobacilli, enterococci, and cluster I clostridia. We also cultured an isolate with high similarity<br />

to Clostridium bifermentens, cluster XI. We did not detect isolates with similarity to clusters IV and XIV although we DNA-sequenced<br />

nearly 100 isolates.<br />

During these studies we identified several significant hurdles to screening the bacterial isolates that would also affect the distribution<br />

<strong>of</strong> bacteria detected from the cultures. <strong>The</strong> most significant hurdle was the logistical challenge <strong>of</strong> performing the cultures and subcultures<br />

in a timely manner. We couldn’t always access the anaerobe chambers in Dr. Whitman’s lab when we needed them because <strong>of</strong><br />

the number <strong>of</strong> people performing studies at the same time. We found that some cultures did not grow upon subculture, probably due<br />

to the extended time <strong>of</strong> incubation. We tended to isolate sporeforming clostridia (such as Clostridium sporogenes cluster I) because<br />

these organisms are very hardy. We attempted to perform cultivations at night and on weekends, when the chambers were more available,<br />

and discovered that the growth conditions were not as optimal as we had hoped.<br />

PI: Dr. Margie Lee ( leem@vet.uga.edu )<br />

CO-PI: Youngjae Cho, William Whitman<br />

VMES 2006 7


Development <strong>of</strong> a Culture-Independent Tool for Reporting Bacterial Growth Rate<br />

Bacterial growth rate is monitored by traditional techniques, and these methods have their limitations. As an alternative we can use<br />

the growth-rate dependent promoter, rRNA fused to the jellyfish green fluorescent protein (GFP) to monitor bacterial cell growth.<br />

Our hypothesis is based on the growth-rate dependent nature <strong>of</strong> this promoter and direct correlation between bacteria growth rate<br />

and rRNA levels within bacterial cell. We developed a plasmid-borne GFP reporter fused to rRNA-promoter and demonstrated<br />

growth-rate dependence <strong>of</strong> our GFP reporter. However, the plasmid was not stably maintained without antibiotic pressure. We created<br />

a second GFP-reporter, using group II intron to specifically direct insertion <strong>of</strong> our GFP reporter into a universally conserved<br />

region <strong>of</strong> rRNA operon, and introduced it into Salmonella enterica Typhimurium. Our GFP reporter was stably maintained in the<br />

absence <strong>of</strong> antibiotic selection pressure. <strong>The</strong>re was an inverse relationship between fluorescence and growth rate for our GFP-marked<br />

S. Typhimurium grown in complex media at 25C, 30C and 37C. Similar results were observed when bacterial cells were grown in<br />

different nutritional conditions, including egg white, defined minimal media with glucose or glycerol as carbon source, and nutrient<br />

rich, complex media. Fluorescence for cells grown in egg whites was low, comparable to signal produced in a nutrient-poor medium.<br />

Our new growth rate reporter can be theoretically introduced into any bacterial species, stably maintained, and report back<br />

the organism’s growth rate in vitro and in vivo. This is especially important in assessing the potential <strong>of</strong> foods, food processes and<br />

cooking/refrigeration temperatures to support microbial growth.<br />

Group II intron<br />

GFP<br />

rRNA<br />

Plasmid<br />

Intron inserts itself and GFP<br />

into targeted gene, rRNA<br />

chromosome<br />

Intron integration site<br />

GFP<br />

Slow growth rate, weak transcription<br />

Fast growth rate, strong transcription<br />

Jellyfish green fluorescent protein (GFP) for monitoring growth <strong>of</strong> pathogens like Salmonella in foods and animals. <strong>The</strong> promoterless<br />

GFP gene was cloned into a group II, self-splicing intron. This intron was re-engineered to identify sequence unique to rRNA<br />

gene in bacteria. <strong>The</strong> intron, borne on a plasmid, inserted itself and the GFP into the bacterial chromosome <strong>of</strong> Salmonella. When<br />

the bacteria are grown in nutrient poor media, they have a slow growth rate and this is manifested in weak transcription AND low<br />

fluorescence. Salmonella grown in rich media have a “fast” growth rate, which translates into strong transcription AND<br />

high fluorescence.<br />

PI: Dr. John J. Maurer (jmaurer@vet.uga.edu)<br />

Co-PI: Adriana A. Pedroso, Andrea P. Oliveira, Margie D. Lee, Marie Maier, Paulina Cruz-Venegas, Dana<br />

Cole, Susan Sanchez, and Charles H<strong>of</strong>acre<br />

8 www.vet.uga.edu/research/vmes/


P. falciparum cytoadherence: Modulation <strong>of</strong> host cell function.<br />

Malaria is an enormous, global public health problem. A staggering 40% <strong>of</strong> the world’s population lives in areas where malaria<br />

transmission occurs, resulting annually in an estimated 500-600 million cases with 2.7 to 3.5 million deaths. <strong>The</strong> most severely<br />

affected are young children, pregnant women, and their unborn infants. Symptoms <strong>of</strong> severe malaria commonly include anemia, cerebral<br />

malaria (malaria-induced coma) and placental malaria (a condition where numerous parasite infected red blood cells are found<br />

in the placenta). It is significant that complications in the lung are commonly seen in association with these severe symptoms, and are<br />

considered an important contributor to malaria mortality. <strong>The</strong> malaria parasite is thought to induce tissue damage in organs through<br />

the ability <strong>of</strong> parasite-infected red blood cells to bind to endothelial cells lining the circulatory system. While the parasite proteins<br />

and host cell receptors that mediate this binding have been well-studied, relatively little is known about how this binding impacts the<br />

function <strong>of</strong> the bound endothelial cell. Our proposed investigation <strong>of</strong> the effects <strong>of</strong> this binding to endothelial cells has important<br />

implications for the disease process in severe syndromes such as cerebral malaria and acute respiratory distress. We believe that a<br />

detailed understanding <strong>of</strong> the effects <strong>of</strong> this binding will lead to novel drugs and vaccines that would disrupt the disease process.<br />

<strong>The</strong> long-range goal <strong>of</strong> our research is to understand how the binding <strong>of</strong> infected red blood cells to host endothelial cells contributes<br />

to severe disease. <strong>The</strong> objective <strong>of</strong> this application is to determine the responses <strong>of</strong> lung microvascular endothelial cells to the<br />

binding <strong>of</strong> malaria infected red blood cells. <strong>The</strong> central hypothesis for the proposed research is that signaling induced by binding<br />

<strong>of</strong> malaria-infected red blood cells will have a pr<strong>of</strong>ound and specific effect on these microvascular endothelial cells. Preliminary<br />

work in an in vitro system that we have developed has demonstrated that binding <strong>of</strong> infected red blood cells to placental cells leads<br />

to modification <strong>of</strong> several proteins in the host cells. <strong>The</strong> expression <strong>of</strong> genes which encode immune factors is also increased, and a<br />

specific immune factor, macrophage migration inhibitory factor, is specifically secreted in a time-dependent manner. <strong>The</strong> rationale<br />

for the proposed research is that several <strong>of</strong> the endothelial cell receptors bound by malaria binding proteins function in initiating<br />

cell signaling events. Thus, it is likely that binding <strong>of</strong> infected red blood cells activates signaling pathways within these endothelial<br />

cells resulting in changes in gene expression. This could then serve to favorably modify the local environment for parasite survival,<br />

manipulate the host immune response, and in the case <strong>of</strong> lung endothelium directly affect barrier function.<br />

PI: David Peterson (Peterson@vet.uga.edu)<br />

VMES 2006 9


Isolation <strong>of</strong> mycobacteriophage that target Mycobacterium aviun ssp. paratuberculosis<br />

Mycobacterium avium subsp. paratuberculosis (Map), the causative agent <strong>of</strong> Johne’s disease in cattle, is an emerging pathogen. Johne’s<br />

disease is a progressive disease that becomes evident 2-5 years after a Map infection <strong>of</strong> the intestinal ileum primarily in calves in<br />

the first year <strong>of</strong> life. Following infection, Map spreads throughout the infected organ causing chronic inflammation and scarring <strong>of</strong> the<br />

tissue. <strong>The</strong> disease manifests itself in the form <strong>of</strong> progressively more severe diarrhea and wasting until the death <strong>of</strong> the animal. Map is<br />

shed in the manure and contaminates the udders <strong>of</strong> cattle, which can lead to infection <strong>of</strong> suckling calves or contamination <strong>of</strong> milk fed<br />

to calves. As pathogenic mycobacterial infections require administration <strong>of</strong> multiple antibiotics for prolonged periods (years), this is<br />

not an economically viable option for the cattle industry. Various vaccines have been produced; however, each <strong>of</strong>fers only partial protection.<br />

Moreover, vaccination <strong>of</strong>ten results in positive antibody tests for Map or M. bovis. <strong>The</strong>refore, a critical need exists to develop<br />

alternate approaches to stem the transmission <strong>of</strong> Map.<br />

<strong>The</strong> goal <strong>of</strong> this proposal was to isolate phages (viruses that only infect bacteria) that target mycobacteria (mycobacteriophages),<br />

specifically Map. <strong>The</strong> long-term goal <strong>of</strong> this research is to develop the mycobacteriophages into an intervention strategy to prevent<br />

transmission <strong>of</strong> Johne’s disease. Soil and manure samples from various Johne’s positive farms in the state <strong>of</strong> <strong>Georgia</strong> were examined.<br />

<strong>The</strong>se included both beef and dairy operations.<br />

A screen for identification <strong>of</strong> mycobacteriophages was developed. Basically, samples are suspended in buffer and subjected to a series<br />

<strong>of</strong> filtration steps to sequentially remove large debris, intermediate-sized debris, and finally bacteria, leaving submicroscopic particles<br />

that include any mycobacteriophages present. <strong>The</strong> resulting filtrates are spotted onto Petri dishes containing rapidly-growing, nonpathogenic,<br />

Mycobacterium smegmatis in s<strong>of</strong>t agar. This enables the mycobacteriophages to infect the bacteria, replicate inside and<br />

lyse the bacteria, and spread to adjacent bacteria. Bacteriophages produce small zones <strong>of</strong> clearing (plaques) among the smooth bacterial<br />

“lawn”.<br />

Based on differences in plaque morphologies, at least three different mycobacteriophages were isolated. <strong>The</strong> mycobacteriophages<br />

were tested for the ability to form plaques on the slow-growing, pathogenic Johne’s bacillus. One <strong>of</strong> the mycobacteriophages does<br />

indeed generate plaques on Map. Efforts are underway to study the Map-infecting mycobacteriophage. By deleting the region <strong>of</strong> the<br />

mycobacteriophage DNA that under some conditions prevents the phage genes from being expressed inside the host, we anticipate<br />

that the modified phage will only grow uncontrollably upon entering Map resulting in bacterial lysis every time. Farm soil and manure<br />

samples are continuing to be screened for additional mycobacteriophages that target Map. By combining different mycobacteriophages<br />

to treat Map contamination, it is more likely to produce sterilizing effects due to reduced likelihood that the bacteria will be able to<br />

mutate multiple surfaces to evade attack by mycobacteriophages that attach and infect through different receptors on the Map.<br />

This research demonstrates that mycobacteriophages can be isolated from <strong>Georgia</strong> Johne’s positive farms. <strong>The</strong>se mycobacteriophages<br />

have potential to be developed into an antibiotic-free intervention strategy to be used to help halt the spread <strong>of</strong> Johne’s disease.<br />

PI: Russell Karls, PhD (rkarls@vet.uga.edu)<br />

Co PI: Mel Pence, DVM MS PAS Diplomate<br />

10 www.vet.uga.edu/research/vmes/


Staphylococci in companion animals and characterization <strong>of</strong> virulence factors<br />

<strong>The</strong> global rise in antibiotic resistance in both human and animal bacterial pathogens is reflected in the epidemic spread <strong>of</strong> clonal<br />

clusters <strong>of</strong> S. aureus possessing multidrug-resistance, methicillin resistance, and more recently, vancomycin resistance. <strong>The</strong>re is some<br />

evidence that the same phenomenon may be occurring in animal-associated Staphylococcus species, and this prompts concern regarding<br />

treatment failure <strong>of</strong> the severe bacterial infections that both human-associated and animal-associated Staphylococcus species cause<br />

in companion animals. In a recent review <strong>of</strong> the literature, the evolutionary development <strong>of</strong> human Methicillin Resistant S. aureus<br />

(MRSA) strains was hypothesized to have occurred via frequent horizontal transfers <strong>of</strong> virulence gene clusters, resulting in five global<br />

clonal clusters <strong>of</strong> MRSA that arose independently. It is not known how animal-associated MRSA and Methicillin susceptible S. aureus<br />

(MSSA) may be related to these human epidemic clonal clusters, or if the prevalence and types <strong>of</strong> toxin and super-antigen genes is<br />

similar. Additionally, it is not known if the frequency <strong>of</strong> carriage and types <strong>of</strong> toxin and super-antigen genes in other animal-associated<br />

Staphylococcus species is similar to those Staphylococci commonly causing disease in humans. <strong>The</strong> objectives <strong>of</strong> this proposal<br />

was to examine the genetics <strong>of</strong> Staphylococci species affecting companion animals in the state <strong>of</strong> <strong>Georgia</strong>, and determine the carriage<br />

<strong>of</strong> selected virulence genes (toxins, superantigens) which are known to contribute to the severity <strong>of</strong> disease caused by these organisms.<br />

As well as, to examine the genetic relatedness and carriage <strong>of</strong> virulence genes <strong>of</strong> S. aureus isolates from companion animals with<br />

those causing disease in humans. <strong>The</strong> central hypothesis <strong>of</strong> this project is that the S. aureus isolates from animal disease are <strong>of</strong> human<br />

origin (reverse zoonosis), and carriage <strong>of</strong> certain virulence genes contributes to severity <strong>of</strong> disease seen in companion animals. Thirty<br />

nine MSSA as well as 10 MRSA canine isolates were included in the study. Rep-PCR was use to create a fingerprint for each isolate<br />

and generate a dendrogram. Standard format PCR was used to determine the presence <strong>of</strong> eight different super-antigen genes eta, etb,<br />

sea, seb, sec, sed and see. Twenty four bacterial isolates were also tested for the presence <strong>of</strong> seg, she, sei and sej. Our studies show that<br />

S. aureus isolated from dogs are a genetically very diverse group and not related to the most common MRSA clones isolated in the<br />

US. <strong>The</strong> super-antigen gene distribution in MSSA was as follows 0% sea, 8.3% seb, 4.2% sec, 0% sed, 0% see, 4.8% seg, 23.8% seh,<br />

17.4% sei, 13% sej, 16.7% tsst-1, 0% eta, and 0% etb. <strong>The</strong> distribution <strong>of</strong> super-antigens in MRSA is as follows 0% sea, 10% seb, 0%<br />

sec, 70% sed, 0% see, 0% eta, and 0% etb. Super-antigen gene carriage seems to be different between MSSA and MRSA with a high<br />

prevalence <strong>of</strong> sed and no sec in MRSA. Further work needs to be carried out to understand the genetic link between mecA and some<br />

super antigens i.e.: sed. <strong>The</strong> undergraduate student working on this project will be presenting this work at the next CURO symposium<br />

and ASM.<br />

PI: S. Sanchez (ssanchez@vet.uga.edu)<br />

CoI: Mimi Healy and Brooke Wheeler<br />

Abortifacient potential <strong>of</strong> eastern tent caterpillar<br />

in domestic goats<br />

Late instar Eastern tent caterpillar (ETC, Malacosoma americanum),<br />

were administered at the dose <strong>of</strong> 50g/day by oral gavage<br />

to six late term pregnant goats until they aborted or for 10 days.<br />

Late instar autoclaved Eastern tent caterpillar were administered<br />

to six late term pregnant goats which served as negative controls.<br />

No abortions were observed in the test and control groups.<br />

All animals were euthanized at 5-10 days post administration <strong>of</strong><br />

the last dose. No significant gross findings were observed during<br />

necropsy. No significant alterations in blood chemistry and<br />

complete blood counts were observed except for few animals<br />

having mild eosinophilia. Histopathological examination <strong>of</strong> the<br />

tissues showed multiple granulomas in the abomasum and intestinal<br />

tract <strong>of</strong> both control and test group animals with setae (hair<br />

<strong>of</strong> ETC) in the center <strong>of</strong> these granulomas. Similar granulomas<br />

with setae were observed in rat GI tract and no abortions were<br />

observed in rats. <strong>The</strong> experiment indicate that ETC or its setae<br />

as such cannot induce abortions in pregnant goats and abortifacient<br />

potential <strong>of</strong> ETC is species specific, similar to the findings<br />

in pregnant mice and rats in which no abortions were observed.<br />

PI: M. M. Sebastian (msebasti@uga.edu)<br />

CoI: ME. Pence, ME. Hines II, and C. Watson<br />

VMES 2006 11


Diagnostic<br />

Science<br />

Identification <strong>of</strong> immunodominant proteins in Mycobacterium<br />

avium ssp. paratuberculosis using proteomics.<br />

Johne’s disease (JD) caused by Mycobacterium avium subsp. paratuberculosis<br />

(MAP) is an economically significant animal health problem that results<br />

in an annual economic loss <strong>of</strong> ~220 million dollars per year to the dairy cattle<br />

industry in USA. This agent is also implicated as an etiology <strong>of</strong> Crohn’s Disease<br />

in human beings. Control and eradication <strong>of</strong> JD has been hampered by<br />

the lack <strong>of</strong> rapid and unambiguous diagnostic tests. Currently used diagnostic<br />

methods such as ELISA and culture have variable levels <strong>of</strong> sensitivity and<br />

specificity and do not allow early diagnosis <strong>of</strong> this disease. <strong>The</strong> Scientists at<br />

Veterinary Diagnostic and Investigational Laboratory, Tifton are constantly<br />

striving to advance the methods for diagnosis and prevention <strong>of</strong> JD. Currently we are approved by National Veterinary Services Laboratory<br />

for MAP culture and serology. We actively participate in <strong>Georgia</strong>’s JD demonstration herd programs and voluntary JD control<br />

programs. We use an automated broth culture system (ESP para JEM system by Trek Diagnostics) to culture MAP and confirmation<br />

is done by Polymerase Chain Reaction. This method <strong>of</strong>fers faster (6 weeks vs. 16 weeks with the conventional methodology) and accurate<br />

detection <strong>of</strong> MAP from clinical samples. <strong>The</strong> VDIL is equipped to culture 6700 samples per year. We are constantly striving to<br />

develop faster, cost effective and sensitive method to diagnose true infection status <strong>of</strong> an animal. We are concentrating on identifying<br />

immunodominant antigens present in MAP by proteomic technology. Identified antigens will be used for developing antibody based<br />

diagnostic assays such as ELISA, Dot blot, and Western blot. We have made considerable improvements in optimizing the detection <strong>of</strong><br />

immunodominant antigens in MAP. Other projects at VDIL include development <strong>of</strong> vaccines to prevent JD, development <strong>of</strong> improved<br />

procedures for organism and antibody detection. Our long-term goal is to develop rapid, sensitive and specific diagnostic tests and vaccines<br />

which will facilitate the control <strong>of</strong> this chronic and problematic disease in cattle.<br />

PI: Sreekumari Rajeev (srajeev@uga.edu)<br />

Diagnostic Detection, Isolation, and<br />

Characterization <strong>of</strong> Avian Viruses<br />

<strong>The</strong> mission <strong>of</strong> the diagnostic virology laboratory is to provide accurate and timely diagnostic virology services for the U.S. poultry<br />

industry, conduct applied research on current avian disease isolates from the field, and improve detection and isolation methods for<br />

monitoring avian viruses. <strong>The</strong> diagnostic virology laboratory provides a vital function in the overall services <strong>of</strong>fered by the diagnostic<br />

facility at the PDRC. During 2005-2006, the diagnostic virology isolated 175 viruses. No virus was isolated from 81 samples. Two<br />

hundred and seventy whole blood samples were submitted for leukosis virus isolation with subsequent LL antigen capture ELISA.<br />

Seventy six serum samples were submitted for virus neutralization assays.<br />

PI: Dr. Holly Sellers (hsellers@uga.edu)<br />

Co-PI: E. Linneman<br />

Investigation <strong>of</strong> Natural Disease Outbreaks<br />

<strong>The</strong> major activity <strong>of</strong> this project is to provide clinical diagnostic support for the commercial poultry industry <strong>of</strong> <strong>Georgia</strong>. This is<br />

accomplished through the application <strong>of</strong> field investigation acquisition <strong>of</strong> flock and farm histories, application <strong>of</strong> analytical, microbiological,<br />

histopathological testing using classical and molecular methods.<br />

Activity is represented by farm visits by Faculty and students. Clinical investigations may include disease outbreaks, farm management<br />

problems, hatchery and feed mill issues, or problems associated with condemnations at processing. Extensive serology test,<br />

bacterial, virus, or Mycoplasma isolation and PCR test <strong>of</strong>ten reveals a particular disease agent as a causative agent mismanagement,<br />

12 www.vet.uga.edu/research/vmes/


equipment failures, vaccination failures or other issues. Modification <strong>of</strong> current practices <strong>of</strong>ten brings the problem under control. <strong>The</strong><br />

pr<strong>of</strong>essional staff and students <strong>of</strong>ten investigate more long term problems on farms within the region. <strong>The</strong>se are typically multi-faceted<br />

problems. Many times these are assigned as student projects under direct supervision <strong>of</strong> a clinician. <strong>The</strong>se investigations bring recommendations<br />

and changes in vaccination programs and management practices which allow that grower to become competitive again.<br />

<strong>The</strong> polymerase chain reaction (PCR) technique continues to permit generation <strong>of</strong> more useful and timely information <strong>of</strong>ten in hours<br />

rather than days or weeks using classical diagnostic techniques for many viral and bacterial agents as well as the Mycoplasmas. Specifically<br />

avian leukosis virus-J, infectious laryngotracheitis virus, infectious bursal disease virus, avian adenovirus, Newcastle disease<br />

virus, avian pneumovirus, avian influenza virus, avian astrovirus, chicken anemia virus and turkey coronavirus PCRs are available and<br />

provide useful and very timely diagnostic information. PCR testing capability now includes Salmonella serotyping for the more common<br />

serotypes <strong>of</strong> Salmonella. <strong>Research</strong> continues and new PCR tests will be applied to diagnostics as applications are developed.<br />

<strong>The</strong> Diagnostic Services/Teaching Lab has completed conversion <strong>of</strong> the lab and accounting system to MS SQL compliance. <strong>The</strong> integrated<br />

accounting system (AccPac) has been upgraded to a SQL version and a web site for delivery <strong>of</strong> lab reports and for data analysis<br />

<strong>of</strong> client serological data is underway. We will be including an e-business capability on the web site for accessioning cases, scheduling<br />

delivery service pickup and accepting credit card payment for services. In addition clients will be able to check case and account via a<br />

secure web site. We are about to allow client access for testing purposes.<br />

Diagnostic Services/Teaching Laboratory activity is represented by 9,154 accessions, 33,170 bacterial procedures, 91 antimicrobial<br />

susceptibilities, 143,564 ELISA tests , 46,569 IBV-HI tests, 196, 459 total serological tests, 2,687 diagnostic PCR tests, 6,439 histopathology<br />

slides, and 1,267 necropsies.<br />

PIs: Stephan G. Thayer (sthayer@uga.edu)<br />

Co-PIs: J.R. Glisson, S.H. Kleven, C. L. H<strong>of</strong>acre, S. Collett, G. Zavala, P. Villegas, M.W. Jackwood, M.D.<br />

Lee, J.J. Maurer, M. Garcia, and H.S. Sellers, and S.M. Williams<br />

Diagnostic Avian Mycoplasmosis<br />

This project consists <strong>of</strong> diagnostic activity for avian mycoplasmas, including mycoplasma isolation and identification, HI tests, and<br />

fingerprinting <strong>of</strong> isolates or specimens by Random Amplified Polymorphic DNA (RAPD) analysis or by sequencing DNA PCR products<br />

from the mgc2 PCR for Mycoplasma gallisepticum (MG) or the vlhA PCR for Mycoplasma synoviae (MS). We also conduct<br />

PCR tests for Mycoplasma iowae. In addition, we provided MG and MS sera for test kits distributed among various laboratories in<br />

the U.S. Our lab is the de facto reference laboratory for avian mycoplasma diagnosis in the U.S.A. We also consult by person and by<br />

telephone regarding field problems with mycoplasma diagnosis or control.<br />

In 2005 we conducted 3148 diagnostic cultures for mycoplasmas, 4693 HI tests for MG, 5488 HI tests for MS, and 645 HI tests for<br />

MM, in addition to RAPD analysis from 151 cases and mgc2 and vlhA.<br />

PI: Dr. S. H. Kleven (skleven@uga.edu )<br />

Co-PI: V. Leiting<br />

Development <strong>of</strong> Monoclonal Antibodies as Diagnostic Reagent for Reticuloendotheliosis Virus<br />

<strong>The</strong> primary objective <strong>of</strong> this research was to develop antibodies against reticuloendotheliosis virus (REV) with the intention <strong>of</strong> using<br />

them as a diagnostic reagent. Our laboratory isolated multiple field REVs, sequenced several <strong>of</strong> them partially, and also resolved the<br />

full genome sequence <strong>of</strong> one such field isolate. <strong>The</strong> entire gag gene was PCR-amplified and cloned into a suitable vector but expression<br />

was unsuccessful. As an alternative method, the gag region coding for the CA protein known as p30 was also cloned into an<br />

expression vector. This protein was successfully expressed but efficiency <strong>of</strong> expression was extremely poor and inconsistent as determined<br />

in Western blots. Meanwhile, polyclonal antibodies against p30 were used successfully in indirect immun<strong>of</strong>luorescence assays<br />

(IFA) in infected cells in culture. IFA was useful, and relatively efficient and practical, albeit not very sensitive. We are still attempting<br />

expression <strong>of</strong> gag and p30 specifically, but have considered changing our strategy to use a baculovirus expression system, still with the<br />

ultimate goal <strong>of</strong> developing monoclonal antibodies vs. p30 in REV.<br />

PI: Guillermo Zavala (gzavala@uga.edu)<br />

Co-PI: Sunny Cheng and Taylor Barbosa<br />

VMES 2006 13


Biomedical<br />

Science<br />

Altering semen extender and glycerol concentration to optimize results after cryopreservation <strong>of</strong><br />

equine spermatozoa<br />

After cryopreservation, spermatozoa from many stallions may have a lower capacity to fertilize an oocyte than fresh or cooled semen.<br />

<strong>The</strong> aim <strong>of</strong> this study was to evaluate a standard panel <strong>of</strong> semen extenders and varied concentrations <strong>of</strong> the cryoprotective<br />

agent (glycerol) to optimize sperm survival rates after cryopreservation. Semen was collected from Quarter Horse stallions (n=3)<br />

from March to May 2006 (6 collections per stallion). Semen was filtered immediately after collection and sample volume and sperm<br />

concentration were measured. A drop <strong>of</strong> raw semen was placed on two prewarmed slides to estimate the percentage <strong>of</strong> progressively<br />

motile sperm. <strong>The</strong> semen sample was then diluted to 100 x 106 spermatozoa/ml with a dried skim milk glucose extender (EZ Mixin<br />

Original Formula, ARS, Chino, CA) or a chemically defined, milk-free diluent (INRA 96, IMV, Maple Grove, MN). After 1 h slow<br />

cooling and equilibration to 4°C, semen samples were centrifuged for 10 min at 400xg. A defined volume <strong>of</strong> supernatant was removed,<br />

so that a concentration <strong>of</strong> 1000 x 106 spermatozoa/ml was obtained after resuspension <strong>of</strong> the sperm pellet. 150 µl <strong>of</strong> semen was then<br />

added to the same semen extender used after semen collection and cryopreservation medium (Cryoguard, Minitube, Verona, WI) to<br />

obtain a final glycerol concentration <strong>of</strong> 2, 3 and 4%, respectively. This also gave a concentration <strong>of</strong> 100 x 106 spermatozoa/ml. After<br />

equilibrating samples for 1 h at 4°C, spermatozoa were loaded into 0.5 ml straws and frozen in liquid nitrogen vapor. After 10 min,<br />

straws were plunged into liquid nitrogen. Semen was thawed at 37°C for 30 sec and evaluated as prior to cryopreservation. Mean total<br />

semen volumes were 56, 11, and 60 ml in the 3 stallions. <strong>The</strong>ir respective mean sperm concentrations were 124 x 106, 505 x 106 and<br />

161 x 106 sperm/ml. Mean percentages <strong>of</strong> progressively motile sperm prior to cryopreservation were 64, 89 and 72%. With the paired<br />

Students t-test, percentages <strong>of</strong> progressively motile sperm after cryopreservation were evaluated with respect to semen extender and<br />

concentration <strong>of</strong> glycerol used. Mean overall progressive motility <strong>of</strong> spermatozoa after cryopreservation differed significantly between<br />

the two extenders and was 46% for INRA 96 and 35% for EZ Mixin OF (p < 0.001). Using EZ Mixin OF as semen extender, best<br />

mean post thaw progressive motility was achieved with 4% glycerol (39%) and differed significantly from 2% glycerol (32%; p


Wound management <strong>of</strong> the distal limb in the horse using topically applied esterified hyaluronic acid<br />

Wounds to the distal limb are commonly encountered in equine practice. <strong>The</strong> severity <strong>of</strong> the wound and paucity <strong>of</strong> available skin<br />

most <strong>of</strong>ten preclude primary suture closure <strong>of</strong> these wounds, leaving healing by second intention as the only available treatment.<br />

Unfortunately second intention healing <strong>of</strong> wounds <strong>of</strong> the distal limbs <strong>of</strong> horses is almost always complicated by the development <strong>of</strong><br />

excessive granulation tissue, leading to prolonged convalescence, loss <strong>of</strong> athletic ability, inferior cosmesis, increased morbidity, and<br />

increased expense and frustration to the horse owner.<br />

Wound healing is largely regulated through a multitude <strong>of</strong> transforming growth factors (TGF-β 1 and 3) released from cells in response<br />

to injury. <strong>The</strong>se factors in turn alter the expression <strong>of</strong> enzymes such as matrix metalloproteinase (MMPs) and tissue inhibitor<br />

<strong>of</strong> metalloproteinase (TIMP) which regulate collagen synthesis and degradation.<br />

Exogenous hyaluronan (HA), a non-sulfated glycosaminoglycan has been used extensively for intra-articular treatment <strong>of</strong> synovitis<br />

and degenerative joint disease, adhesiolysis and adhesion prevention, and various intra-ocular conditions. More recently, hyaluronan<br />

has been shown to significantly increase the rate and quality <strong>of</strong> wound healing in people. <strong>The</strong> exact mechanism by which hyaluronan<br />

enhances wound healing is not clear, but likely involves the regulation <strong>of</strong> cellular proliferation and signaling<br />

Samples taken, at regular intervals from the equine lower limb, following topical<br />

application <strong>of</strong> an esterified hyaluronic acid (Hyal<strong>of</strong>ill) are currently being<br />

processed. Preliminary results assessing the effect that topical hyaluronic acid<br />

might have on the rate <strong>of</strong> closure <strong>of</strong> a wound through contraction and epithelialisation<br />

do not show an appreciable difference to the control situation.<br />

Histological evaluation <strong>of</strong> the influence that topical hyaluronic acid has on<br />

rate <strong>of</strong> blood vessel ingrowth (angiogensesis) and degree <strong>of</strong> inflammatory<br />

response is underway. Immunocytochemical determination <strong>of</strong> the concentrations<br />

<strong>of</strong> the growth factor TNFα and in situ hybridization techniques assessing<br />

TGF-β 3 and Collagen III levels is also being performed. Looking at these<br />

factors will give some indication as to how the topical product influences the<br />

wound environment. In collaboration with the Department <strong>of</strong> Pharmacology<br />

and Toxicology at the School <strong>of</strong> Veterinary Medicine, in Hannover, Germany,<br />

frozen tissue samples are also being run in order to determine levels <strong>of</strong> the<br />

aforementioned MMPs.<br />

While it appears that, under the conditions <strong>of</strong> this study and using the esterified<br />

hyaluronic acid product, there might be no immediate indication for its<br />

use alone, in promoting wound healing, the results <strong>of</strong> the study will provide<br />

valuable baseline information on the safety <strong>of</strong> this product for use in horses.<br />

This product has been used as a vehicle in a plethora <strong>of</strong> recent studies in the<br />

human field. It has proven valuable in providing a matrix for deposition <strong>of</strong><br />

keratinocytes, osteblasts, chondrocytes, mesenchymal stem cells and hepatocytes<br />

at specified target locations. Similar indications may warrant its future<br />

use in equine medicine.<br />

PI: PO Eric Mueller (emueller@vet.uga.edu)<br />

CoI: 1. Stefan Witte, 2. Randy Eggleston, 3. Jaroslava Halper<br />

VMES 2006 15


<strong>The</strong>rapeutics<br />

and Immunology<br />

Development <strong>of</strong> novel antiviral drugs: inhibition <strong>of</strong> avian influenza by RNAi<br />

Highly pathogenic avian influenza (HPAI) is a major threat to the United States poultry industry. In 1983, a major outbreak <strong>of</strong> HPAI<br />

in the U.S. caused the destruction <strong>of</strong> more than 17 million birds at a cost <strong>of</strong> $65 million. More than 16 outbreaks <strong>of</strong> potential HPAI<br />

strains have occurred in the U.S. since 1997. Quarantine and depopulation are the preferred means for control <strong>of</strong> outbreaks and new<br />

methods for protection <strong>of</strong> poultry flocks are needed. Moreover, several instances <strong>of</strong> human infection with HPAI have been reported,<br />

most likely caused by contact with infected poultry. If HPAI were to emerge as even a moderate pandemic strain, more that 200,000<br />

persons could die in the United States alone and the economic cost could exceed $100 billion. Currently, there is not a vaccine available<br />

and antiviral drugs are partially effective.<br />

RNA interference (RNAi) is an emerging technology that can specifically inhibit gene expression both in vitro and in vivo. Gene<br />

silencing is the result <strong>of</strong> sequence-specific RNA degradation and is mediated by short, 21-26 nucleotide interfering RNAs (siRNAs). A<br />

number <strong>of</strong> studies have demonstrated inhibition <strong>of</strong> replication <strong>of</strong> viruses in cell culture by RNAi. Using an established murine model<br />

<strong>of</strong> influenza virus infection, we have demonstrated that in vivo treatment with virus-specific siRNAs can effectively suppress influenza<br />

virus replication and protect animals from an otherwise lethal infection.<br />

We have screened more than 100 candidate siRNAs for efficacy in inhibiting influenza virus replication and have identified a handful<br />

<strong>of</strong> candidates that silence a variety <strong>of</strong> gene targets, including the nucleoprotein, matrix, and polymerase genes <strong>of</strong> influenza. <strong>The</strong>se<br />

siRNAs target highly conserved regions <strong>of</strong> the genome and should inhibit most influenza viruses, including low pathogenic and highly<br />

pathogenic avian influenza viruses. We found that the candidate siRNAs identified in the primary screen inhibited all influenza viruses<br />

tested, to date. Future studies focus on testing lead candidates for efficacy against HPAI in culture and testing for efficacy in animal<br />

challenge models. Candidate siRNAs from these screens could be developed as a novel therapeutics and prophylactics for poultry<br />

flocks and workers to protect against HPAI infection.<br />

PI: S. Mark Tompkins<br />

<strong>The</strong> Effect <strong>of</strong> Mycoplasma synoviae (MS) challenge at the onset <strong>of</strong> lay, on performance <strong>of</strong> table egg layers<br />

and typing <strong>of</strong> MS and M. Gallisepticum (MG) isolates from field cases.<br />

This project was initiated because <strong>of</strong> concerns that in many cases seroconversion for MS appeared to be delayed, thereby complicating<br />

the early detection <strong>of</strong> outbreaks. <strong>The</strong> objectives were as follows:<br />

1. To determine if delayed or negative serological responses to MS by agglutination, HI, or ELISA can be reproduced by upper respiratory<br />

tract challenge with very low numbers <strong>of</strong> organisms, or if there is something unique about MS strains which have been isolated<br />

from flocks exhibiting poor seroconversion.<br />

2. To determine, in such cases, whether ELISA or HI testing can be relied upon as screening serological tests to substitute for agglutination,<br />

or if such monitoring will need to be conducted by PCR and/or culture.<br />

3. To fully characterize, by DNA fingerprinting, virulence and antigenic differences, such field isolates found to be atypical or unique<br />

with regard to poor serological responses.<br />

Chickens were challenged by intratracheal administration <strong>of</strong> 10-fold dilutions <strong>of</strong> two challenge organisms (F10-2AS, a commonly<br />

used challenge strain, and K5664, an isolate from a field case with atypical serological results) to determine if low challenge doses<br />

readily infect chickens and if they seroconvert normally. <strong>The</strong>re were 10 principals and 2 contact controls per group, and there was<br />

one group <strong>of</strong> unchallenged controls. Chickens were sampled weekly until 8 weeks post challenge and tested by culture, serum plate<br />

agglutination, HI (with standard WVU 1853 antigen, and antigens prepared from F10-2AS, and K5664), and IDEXX and Synbiotics<br />

ELISA. <strong>The</strong> results can be summarized as follows.<br />

16 www.vet.uga.edu/research/vmes/


1. Challenge with as few as 76 color change<br />

units (ccu) <strong>of</strong> K5664 and 24 ccu <strong>of</strong> F10-2AS<br />

readily infected chickens – the majority were<br />

culture positive at 7 days post-challenge.<br />

2. Infection spread readily to the in-contact<br />

controls. With F10-2AS, contact controls were<br />

positive by culture by 1 week post-challenge, and<br />

with K5664, they were positive by 2 weeks<br />

post-challenge.<br />

3. <strong>The</strong> unchallenged controls remained negative<br />

by culture throughout the study, even though<br />

they were maintained in floor pens in the same<br />

house as the challenged groups. <strong>The</strong>re were 2<br />

empty pens between the controls and an<br />

infected group.<br />

4. All groups <strong>of</strong> challenged chickens responded<br />

normally by all serological tests. Groups with<br />

the lowest challenge groups seroconverted about<br />

1 week behind all <strong>of</strong> the other groups, but then<br />

developed a normal serological response.<br />

5. Antigenic differences among strains were clearly detected by using homologous and heterologous HI antigens. <strong>The</strong> homologous<br />

antigen generally detected an earlier and stronger antibody response than did the heterologous antigens. Nevertheless, all HI antigens<br />

detected a positive antibody response.<br />

6. All serological tests exhibited adequate sensitivity and specificity. <strong>The</strong> groups challenged with the lowest numbers <strong>of</strong> organisms<br />

generally had a weaker antibody response with the IDEXX ELISA kits than with the Synbiotics kits with the lowest challenge levels<br />

with both strains.<br />

7. <strong>The</strong>re was no evidence that strain K5664 induces an atypical antibody response.<br />

To summarize, chickens were readily infected with very low challenge doses <strong>of</strong> MS, and seroconversion was delayed only about a<br />

week in those groups. We do not consider this to be abnormally slow. Infection spread readily to contact controls, but unchallenged<br />

controls in the same house remained uninfected, suggesting rapid transmission by contact, but no evidence <strong>of</strong> infection by aerosol.<br />

Only the IDEXX ELISA kits were insensitive in detecting antibody with the lowest challenge doses. <strong>The</strong>re was evidence <strong>of</strong> serological<br />

variability among strains with the HI test, but not to the extent that HI antigens made by any <strong>of</strong> the strains tested would have been<br />

inadequate in detecting seroconversion.<br />

PI: Dr. S.H. Kleven (skleven@uga.edu)<br />

C0-PI: Dr. Ziv Raviv and Naola Ferguson<br />

VMES 2006 17


Virology<br />

Cellular activation induced by FIV vaccine strain viruses<br />

Feline immunodeficiency virus (FIV) naturally infects cats causing AIDS similar to that<br />

induced by HIV in humans. Critical to development <strong>of</strong> an effective commercial FIV vaccine<br />

was the identification <strong>of</strong> which FIV viruses to include in the vaccine; however, the mechanisms<br />

underlying why certain FIV isolates could be used to make a protective vaccine while<br />

others could not are not well understood. <strong>The</strong> central hypothesis for these studies was that<br />

strains <strong>of</strong> FIV that are effective vaccine immunogens induce characteristic patterns <strong>of</strong> cellular<br />

activation in feline T-cells.<br />

<strong>The</strong> specific aim was to identify unique cellular transcription factor activation patterns for FIV<br />

vaccine isolates. <strong>The</strong> experimental approach characterized virus-induced changes in transcription<br />

factor activation pr<strong>of</strong>iles, using a feline T-lymphocyte cell line inoculated with different<br />

FIV isolates, including those used in the FIV vaccine. In addition, the transcription factor<br />

activation pr<strong>of</strong>iles <strong>of</strong> FIV-infected cells treated with a protein known to enhance FIV replication<br />

were also determined. Transcription factor activation pr<strong>of</strong>iles were detected using protein/<br />

DNA arrays that can simultaneously assess activation <strong>of</strong> multiple transcription factors.<br />

Our results indicate that variations in pathogenicity and ability to infect and replicate in<br />

cultured feline T-cells between FIV isolates is associated with differences in transcription<br />

factor activation. Further studies are currently underway to confirm and compare these differences. <strong>The</strong> results <strong>of</strong> these studies were<br />

used as preliminary data for several grant applications, including one that was submitted to the National Institute <strong>of</strong> <strong>Health</strong>, and will<br />

be included in a scientific paper that is currently in preparation. Future studies will build on these results in order to provide a better<br />

understanding <strong>of</strong> protective immunity against FIV. A better understanding <strong>of</strong> these mechanisms will improve our knowledge <strong>of</strong> AIDS<br />

pathogenesis and provide information important for development <strong>of</strong> diagnostic tests that can distinguish between vaccinated cats and<br />

those naturally infected with FIV- a major problem with the current antibody-based diagnostic FIV tests.<br />

PI: Elizabeth W. Uhl (euhl@vet.uga.edu)<br />

siRNA Molecules for Disease Intervention Against Poxviruses<br />

Poxviruses have been studied for more than 200 years. From the time <strong>of</strong> Edward Jenner’s pioneering vaccination experiments in 1796<br />

to the present, poxviruses that infect nearly every vertebrate animal on the planet, from crocodiles to humans, have been identified. In<br />

addition, smallpox was the first disease to be eradicated by man.<br />

While nearly 30 years have passed since smallpox was declared eradicated, poxviruses continue to plague both animals and man as a<br />

zoonotic pathogen. For example, in 2003, 71 people in the United States became infected with monkeypox virus after interaction with<br />

prairie dogs. Based on analyses <strong>of</strong> sera for poxvirus-specific antibodies, a wide variety <strong>of</strong> animals including rats, rabbits, prairie dogs,<br />

and squirrels may also be potential reservoirs <strong>of</strong> poxviruses. Given the potential impact on both animals <strong>of</strong> agricultural importance and<br />

human health, new treatments for acute poxvirus disease are needed.<br />

As a result <strong>of</strong> the termination <strong>of</strong> the smallpox vaccination program in 1972, approximately 42% <strong>of</strong> the current U.S. population has no<br />

immunity to poxviruses. Furthermore, there are no antiviral drugs approved by the F.D.A. or U.S.D.A. for the treatment <strong>of</strong> poxvirus<br />

infection in humans or animal. To bridge this unmet need, cutting-edge technology based on RNA interference (RNAi) was used as a<br />

therapeutic approach to inhibit poxvirus replication. RNAi is an evolutionarily conserved, gene-silencing mechanism in which small<br />

(19-23 nt) double-stranded RNA molecules, or small interfering RNAs (siRNA), target RNAs for destruction with exquisite potency<br />

and selectivity, causing gene silencing at the post-transcriptional stage before proteins can be made.<br />

Using DNA sequence information from a variety <strong>of</strong> different poxvirus genomes, 88 candidate siRNA molecules specific for 30 different<br />

poxvirus genes were designed and synthesized. <strong>The</strong> resulting siRNAs were tested for their abilities to inhibit the replication <strong>of</strong> both<br />

18 www.vet.uga.edu/research/vmes/


Vaccinia virus and Cowpox virus in vitro. siRNA molecules specific for 9 different viral genes were able to efficiently block poxvirus<br />

replication in vitro. <strong>The</strong>se studies revealed that: 1) RNAi can be used to inhibit the replication <strong>of</strong> poxviruses, and 2) transfection <strong>of</strong><br />

cells with siRNAs can dramatically reduce the ability <strong>of</strong> both Vaccinia virus and Cowpox virus to replicate. <strong>The</strong>se data suggest that<br />

poxvirus-specific siRNAs may be useful for the treatment <strong>of</strong> animal and human infection, and can be used to identify viral genes that<br />

may be targets for other potentially useful therapies such as small molecule inhibitors.<br />

PI: Jeff Hogan (jhogan@vet.uga.edu)<br />

Production <strong>of</strong> polyclonal antibodies against ILTV glycoprotein E using Synthetic Peptides<br />

Infectious laryngotracheitis virus (ILTV) is an alphaherpesvirus worldwide distributed and causes acute respiratory disease in chickens<br />

with high morbidity, decreased growth and egg production and causes moderate mortality. <strong>The</strong> herpesviruses have glycoproteins<br />

located on the viral envelop, which play essential roles in viral attachment, penetration, assembly and egress, and also in viral replication<br />

and cell-to-cell. Polyclonal antibodies against ILTV glycoproteins are an important tool to study ILTV the glycoproteins functions,<br />

as well as to detect viral replication and protein expression. This study describes the use <strong>of</strong> synthetic peptides to produce specific ILTV<br />

glycoprotein (gE) antibodies. A peptide <strong>of</strong> 18 amino acids was selected to immunize mice against generated polyclonal serum. <strong>The</strong><br />

antibodies generated reacted with ILTV infected cells and gE transfected cells by immun<strong>of</strong>luorescent assay (IFA) and immunoblot assay.<br />

<strong>The</strong> gE peptide sequence showed to be useful to produce specific ILTV gE antibodies, which can be used to study the expression<br />

<strong>of</strong> this ILTV essential protein during in vitro propagation <strong>of</strong> the virus, and in vivo during different stages <strong>of</strong> infection.<br />

PIs: Ivomar Oldoni, Sylva Riblet and Maricarmen García (mcgarcia@uga.edu)<br />

Runting and Stunting (AV-111)<br />

In late 2004-present, broiler companies throughout the United States had increasing cases <strong>of</strong> a runting and stunting syndrome (RSS),<br />

particularly in the winter and spring. Currently, the few broiler companies in the Southeast that were spared encounters with RSS have<br />

reported severe outbreaks in several <strong>of</strong> their complexes. RSS is a transmissible disease affecting young broilers between 1-2 weeks <strong>of</strong><br />

age. Most notably, RSS causes severe weight suppression during the first few weeks <strong>of</strong> age, lack <strong>of</strong> flock uniformity, diarrhea, distention<br />

<strong>of</strong> the intestines, and a significant increase in feed conversion. Cystic enteropathy is consistently observed by histopathological<br />

examination <strong>of</strong> small intestine samples from affected flocks. <strong>The</strong> economic effect <strong>of</strong> this disease hits both the broiler company and<br />

contract growers. Although descriptions <strong>of</strong> RSS date back to the 1970s, the etiologic agent(s) has yet to be identified. All evidence<br />

suggests this is a multifactorial disease.<br />

Virological studies conducted on RSS at PDRC over the past year have been aimed at isolating potential pathogens. <strong>The</strong> disease is<br />

reliably reproduced using filtered intestinal homogenates from clinically affected broilers, thus implying viral etiologic agents. In<br />

addition, our in vivo studies indicate that heat treatment does mitigate some <strong>of</strong> the affects <strong>of</strong> RSS. We have isolated and characterized<br />

many viruses from field cases <strong>of</strong> RSS. As expected, numerous virus isolates were <strong>of</strong> vaccine origin or not considered to play a<br />

role. However, novel reoviruses and chicken astroviruses were isolated and characterized. Molecular characterization <strong>of</strong> the S1 gene<br />

revealed novel reoviruses that are


<strong>Research</strong><br />

Contracts & Grants<br />

Allen, Sheila. Core <strong>Animal</strong> Diagnostic Laboratory: NAHLN: GA. USDA-CSREES. $300,834<br />

Allen, Sheila. Section 1433 <strong>Animal</strong> <strong>Health</strong> and Disease <strong>Research</strong> Funds FY2006. USDA-CSREES. $102,880<br />

Allen, Sheila. Promoting cultural diversity in the veterinary workforce. USDA-CSREES. $120,000<br />

Baldwin, Charles. Diagnostic services relative to the control, diagnosis, treatment prevention, and eradication <strong>of</strong> livestock diseases 2004. Ga. Dept.<br />

<strong>of</strong> Agriculture. $2,160,738<br />

Barton, Michelle. Relative adrenal insufficiency in critically ill neonatal foals. American <strong>College</strong> <strong>of</strong> Veterinary Internal Medicine Foundation.<br />

$9,485<br />

Blas-Machado, Uriel. Pathogenesis and virulence <strong>of</strong> a bovine enterovirus-1 isolate in cattle - year 2. USDA-CSREES. $15,000<br />

Brown, Corrie. Delivering training on necropsy procedues and coordinating national planning for animal health Afghanistan. USDA-FAS Foreign<br />

Ag. Serv. $13,202<br />

Brown, Corrie. USDA <strong>Research</strong> Support. USDA. $3,850<br />

Brown, Corrie. USDA <strong>Research</strong> Support. USDA. $15,450<br />

Brown, Corrie. Technical consultant animal health infrastructure Afghanistan, Standard Cooperative Agreement. USDA-APHIS. $7,784<br />

Brown, Corrie. Detection <strong>of</strong> key factors in the development <strong>of</strong> Fibropapillomas in sea turtles; Puerto Rico. Morris <strong>Animal</strong> Foundation. $28,620<br />

Brown, Corrie. Development <strong>of</strong> a histopath research platform at the research foundation in tropical diseases - Cameroon. Mectizan Donation<br />

Program. $37,180<br />

Budsberg, Steven. A placebo-controlled study to investigate the efficacy <strong>of</strong> a tachykinin receptor antagonist in a urate induced arthritis model.<br />

Boehringer Ingleheim. $198,929<br />

Budsberg, Steven. In vivo protocol for testing the effects <strong>of</strong> Firocoxib on whole blood, gastric mucosal and osteoarthritic synovial fluid prostaglandin<br />

and leukotriene. Merial Limited. $126,000<br />

Budsberg, Steven. <strong>The</strong> analgesic effects <strong>of</strong> EAA-129 (WAY-209129) and PLA-902 (WAY-195902) in a canine model <strong>of</strong> osteoarthritis. Fort Dodge<br />

<strong>Animal</strong> <strong>Health</strong>. $54,158<br />

Budsberg, Steven. Clinical efficacy and safety <strong>of</strong> an extended release formulation <strong>of</strong> tramadol HCl in dogs: a pilot trial. Farnam Companies, Inc.<br />

$71,492<br />

Budsberg, Steven. Cyclo-oxygnease selectivity <strong>of</strong> nosteroidal anti-inflammatory drugs in feline blood. Merial Limited. $18,397<br />

Carmichael, Paige. Training in clinical ophthalmology and pathology. Graduate Assistantship support for Dr. Shannon Boveland. Tuskegee Univ.<br />

$28,675<br />

C<strong>of</strong>field, Julie. Neuromuscular targets <strong>of</strong> botulinum toxin. NIH-National Institutes <strong>of</strong> <strong>Health</strong>. $349,600<br />

Corn, Joseph. <strong>The</strong> third reservoir <strong>of</strong> infection: M. paratuberculosis in the environment and protozoal amplification. USDA-APHIS. $75,183<br />

Corn, Joseph. Distribution <strong>of</strong> Pseudorabies virus and Brucella suis in feral swine. USDA-APHIS. $87,500<br />

Corn, Joseph. Exotic tick surveillance in the southeastern United States and Puerto Rico FY2005. USDA. $200,000<br />

Corn, Joseph. Exotic arthropod surveillance in the Southeastern U.S. and Puerto Rico FY2006. USDA. $200,000<br />

Corn, Joseph. Pseudorabies virus and Brucella suis in transitional and feral swine. USDA-Veterinary Services. $25,000<br />

Dickerson, Harry. Clinical vaccine trail to test Tetrahymena as a protective vaccine against Ichthyophthirius in Koi: I. Vaccination. Tetragenetics,<br />

Inc. $10,000<br />

Dickerson, Harry. Clinical vaccine trail to test Tetrahymena as a protective vaccine against Ichthyophthirius in Koi: II. Immobilization. Tetragenetics,<br />

Inc. $7,500<br />

Dickerson, Harry. UGA 2006 Veterinary Scholars Program: A research training experience for veterinary students. Merck Merial <strong>Animal</strong> <strong>Health</strong><br />

Grants Program. $20,400<br />

Dickerson, Harry. Emerging and re-emerging infectious disease residency/PhD program. Merial Limited. $240,000<br />

Dietrich, Ursula. Identification <strong>of</strong> ocular tissue MMPs within the aqueous humor outflow pathway in the canine eye. ACVO Vision for <strong>Animal</strong><br />

Foundation. $3,000<br />

Ferguson, Naola M. Evaluation <strong>of</strong> Mycoplasma gallisepticum vaccines in chickens. Fort Dodge <strong>Animal</strong> <strong>Health</strong>. $17,093<br />

Fischer, John. Wildlife services disease training. USDA-APHIS. $150,000<br />

Fischer, John. Cooperative agreement for development and evaluation <strong>of</strong> data relative to disease relationships that may simultaneously involve wildlife,<br />

domestic livestock and poultry. USDA-APHIS. $350,000<br />

Fischer, John. Southeastern Cooperative Wildlife Disease Study. Fish and Wildlife Agencies. $182,280<br />

Fu, Zhen. Personnel training. Changchun Changsheng Life Sciences Ltd. $6,000<br />

Garcia, Maricarmen. Developing avian influenza serological tests for differentiating infected from vaccinated animals (DIVA). Univ. <strong>of</strong> Maryland.<br />

$126,584<br />

Garcia, Maricarmen. Avian influenza virus H9N2: Characterization and control strategies. BARD - Binational Agric. <strong>Research</strong> and Dev. Fund.<br />

$50,000<br />

Halper, Jaroslava. Degenerative suspensory ligament desmitis in horses. U.S. Equestrian Federation, Inc. $25,000<br />

Hernandez-Divers, Stephen. Investigation and characterization <strong>of</strong> shell lesions in free-ranging tortoises and freshwater turtles from National Wildlife<br />

Refuges. Association <strong>of</strong> Reptilian and Amphibian Vets. $3,000<br />

Hodge, Thomas. Identification <strong>of</strong> host genes required for viral pathogenesis, II. Hudson-Alpha Institute for Biotechnology. $287,500<br />

Hoenig, Margarethe. Nuclear magnetic resonance, a noninvasive method to evaluate glucose and fat metabolism in the cat. Nestle Purina, Inc.<br />

$30,125<br />

20 www.vet.uga.edu/research/vmes/


H<strong>of</strong>acre, Charles. Anti-inflammtory effects <strong>of</strong> muscadine grape products on gastrointestinal diseases in chickens and rats. Paulk Vineyards/USDA.<br />

$96,500<br />

H<strong>of</strong>acre, Charles. Reducing Salmonella prevalence in the processing plant by breaking vertical transmission through vaccination <strong>of</strong> affected broilerbreeder<br />

flocks. U.S. Poultry and Egg Assoc. $64,576<br />

Hogan, Robert J. Identification <strong>of</strong> Francisella tularensis-specivid human T cell epitopes. SECEBT. $49,997<br />

Hogan, Robert J. <strong>The</strong> influences <strong>of</strong> N- and O-linked glycosylation on the immunogenicity <strong>of</strong> the Ebola virus glycoprotein. Southern <strong>Research</strong> Institute.<br />

$311,322<br />

Hondalus, Mary K. Virulence <strong>of</strong> the opportunistic pathogen Rhodococcus equi. NIH-National Institutes <strong>of</strong> <strong>Health</strong>. $331,200<br />

Hondalus, Mary K. Needle-free vaccination via nanoparticle aerosols. Harvard <strong>University</strong>. $119,779<br />

Hurley, David. In vitro models <strong>of</strong> cellular level processes in the early pathogenesis <strong>of</strong> Black Walnut induced laminitis. American Quarter Horse Assoc.<br />

$62,814<br />

Jaso-Friedman, Liliana. A novel teleost pattern recognition receptor and its role in antimicrobial innate immunity. USDA-NRI. $300,000<br />

Kaplan, Ray. Sustainable control <strong>of</strong> gastrointestinal nematodes in small ruminants. Fort Valley State <strong>University</strong>. $22,482<br />

Kaplan, Ray. Use <strong>of</strong> condensed tannin-containing plants to control gastrointestinal nematodes in sheep and goats in the southern USA. Fort Valley<br />

State <strong>University</strong>. $50,000<br />

Kaplan, Ray. Maintenance <strong>of</strong> gastrointestinal nematodes for in vitro drug efficacy testing. Divergence, Inc. $2,996<br />

Keel, Kevin. Epizoology <strong>of</strong> latent and active duck plague virus infection in free-flying waterfowl. Morris <strong>Animal</strong> Foundation. $98,660<br />

King, Christopher. VCA: A monoclonal antibody toolkit for functional genomics <strong>of</strong> plant cell walls. NSF-National Science Foundation. $92,736<br />

Kleven, Stanley. Evaluation <strong>of</strong> potential live Mycoplasma gallisepticum vaccines in chickens. Ghen Corporation. $83,588<br />

LeRoy, Bruce. Effect <strong>of</strong> deracoxib on the growth and metastasis <strong>of</strong> a canine prostate carcinoma xenograft. Novartis <strong>Animal</strong> <strong>Health</strong>. $60,852<br />

Maurer, John. A molecular approach for creating Haemophilus paragrallinarum vaccine strain thru site-directed mutagenesis. Fort Dodge <strong>Animal</strong><br />

<strong>Health</strong>. $53,750<br />

McCall, John. Furnish Brugia malayi adult worms and/or Brugia malayi infective larvae. NIH-National Institutes <strong>of</strong> <strong>Health</strong>. $139,499<br />

McCall, John. Filariasis research reagent resource center. NIH-National Institutes <strong>of</strong> <strong>Health</strong>. $422,240<br />

McCall, John. Transfection and promoter analysis <strong>of</strong> Brugia malayi. Univ. <strong>of</strong> Alabama. $20,000<br />

Miller, Doris. BSE Surveillance. USDA-APHIS. $372,264<br />

Miller, Doris. Classical swine fever surveillance. USDA. $208,793<br />

Miller, Doris. Athens Diagnostic Laboratory. GA Dept. <strong>of</strong> Agriculture. $1,324,323<br />

Moore, James. Leukocyte activation in the developmental period <strong>of</strong> equine laminitis. Ohio State <strong>University</strong>. $133,820<br />

Murray, Thomas. Neurotoxins from marine algae and cyanobacteria. Scripps Institute <strong>of</strong> Oceanography. $150,970<br />

Murray, Thomas. Cellular activation induced by multivalent ligands. NIH-National Institutes <strong>of</strong> <strong>Health</strong>. $24,211<br />

Murray, Thomas. Inhibition <strong>of</strong> endotoxin with adenosine receptor agonists. Grayson-Jockey Club <strong>Research</strong> Foundation. $40,792<br />

Pence, Melvin. <strong>Georgia</strong> Johne’s Disease Demonstration Herd Project. USDA. $60,664<br />

Pence, Melvin. <strong>Georgia</strong> Johne’s work plan for GJP FY05. GA Dept <strong>of</strong> Agriculture. $54,414<br />

Quinn, Fred. Crohn’s disease: Potential acquisition through pasteurized milk. Emory Univ. - SECEBT. $47,696<br />

Rawlings, Clarence. Comparison <strong>of</strong> carbon dioxide laser and 4 MHz radi<strong>of</strong>requency for feline onychectomy: Histologic comparison. Ellman International,<br />

Inc. $23,017<br />

Reeves, David. Manage Rogers state prison swine farm. GA Dept. <strong>of</strong> Corrections. $76,061<br />

Reeves, David. Manage Rogers state prison dairy farm. GA Dept. <strong>of</strong> Corrections. $311,883<br />

Ritchie, Branson. <strong>Research</strong> Associate In Exotic/Zoo Infectious Disease And Pathology Postgraduate program. Zoo Atlanta/Riverbanks Zoo.<br />

$13,000<br />

Robertson, Thomas. Atypical regulation <strong>of</strong> vascular tone by protein kinase C. NIH-National Institutes <strong>of</strong> <strong>Health</strong>. $184,000<br />

Robertson, Thomas. Signal transduction pathways underlying laminar microvascular dysfunction in the early stages <strong>of</strong> acute laminitis. USDA-<br />

NRI. $324,024<br />

Saliki, Jeremiah T. Editorial support for the Journal <strong>of</strong> Veterinary Diagnostic Investigation. American Assoc. <strong>of</strong> Veterinary Laboratory Diagnosticians.<br />

$31,250<br />

Sanderson, Sherry. Dirlotapide customer satisfaction study. Pfizer <strong>Animal</strong> <strong>Health</strong>. $52,140<br />

Sellers, Holly. Reovirus progeny challenge study with novel avian reoviruses isolated from flocks with runting stunting syndrome. Fort Dodge <strong>Animal</strong><br />

<strong>Health</strong>. $10,556<br />

Stallknecht, David. Determine infectious rate and distribution <strong>of</strong> avian pathogens in wild birds <strong>of</strong> midwestern and southeastern US for Homeland<br />

Security Surveillance. USDA-ARS. $50,000<br />

Stallknecht, David. Mosquito host preference for peridomestic avian species involved in West Nile virus epidemiology. Morris <strong>Animal</strong> Foundation.<br />

$35,000<br />

Stallknecht, David. HPAI wild birds: Potential for a new wildlife disease or dead end Morris <strong>Animal</strong> Foundation. $11,700<br />

Stallknecht, David. Vector-borne disease surveillance - Wild bird, mosquito, and tick laboratory support. GA Dept. <strong>of</strong> Human Resources. $280,965<br />

Stallknecht, David. West nile virus surveillance in wild birds. GA Dept. <strong>of</strong> Human Resources. $81,197<br />

Sum, Steffen. Determination <strong>of</strong> the role <strong>of</strong> rickettsial pathogens in naturally infected dogs. IDEXX, Inc. $8,000<br />

Torres-Velez, Fernando. Pathogenesis <strong>of</strong> Nipah virus in guinea pigs. NIH-National Institutes <strong>of</strong> <strong>Health</strong>. $108,220<br />

Tripp, Ralph. Phase I: Calf study. Numico <strong>Research</strong> Australia Pty. $16,065<br />

Vandenplas, Michel. Use <strong>of</strong> microarrays to characterize endotoxemia in vivo. Grayson-Jockey Club <strong>Research</strong> Foundation. $22,745<br />

Varela-Stokes, Andrea. Infection dynamics <strong>of</strong> Ehrlichia chaffeensis. NIH-National Institutes <strong>of</strong> <strong>Health</strong>. $105,939<br />

Wagner, John J. Cocaine-induced metaplasticity in the hippocampus. NIDA-National Inst. Drug Abuse. $143,741<br />

Ward, Cynthia R. Environmental influences on signal transduction abnormalities in feline hyperthyroidism. Morris <strong>Animal</strong> Foundation. $14,976<br />

Ward, Cynthia R. Nutritional influences on the development <strong>of</strong> feline hyperthyroidism. Winn Feline Foundation. $13,800<br />

Yabsley, Michael. Molecular and biological characterization <strong>of</strong> Trypanosoma cruzi from United States. NIH-National Institutes <strong>of</strong> <strong>Health</strong>.<br />

$221,250<br />

VMES 2006 21


Administrators<br />

& Advisors<br />

<strong>The</strong> <strong>University</strong> System <strong>of</strong> <strong>Georgia</strong><br />

Board <strong>of</strong> Regents<br />

Allan Vigil, Morrow - CHAIR<br />

Thirteenth District (2010)<br />

William H. Cleveland, Atlanta – VICE CHAIR<br />

State-at-Large (2009)<br />

Hugh A. Carter, Atlanta<br />

State-at-Large (2009)<br />

Michael J. Coles, Kennesaw<br />

Sixth District (2008)<br />

Robert Hatcher, Macon<br />

Eight District (2013)<br />

Julie Ewing Hunt, Tifton<br />

Second District (2013)<br />

Felton Jenkins, Madison<br />

At-large (2013)<br />

W. Mansfield Jennings, Jr., Hawkinsville<br />

First District (2013)<br />

James R. Jolly, Dalton<br />

Tenth District (2008)<br />

Donald M. Leebern, Jr., McDonough<br />

State-at-Large (2012)<br />

Elridge W. McMillan, Atlanta<br />

Fifth District (2010)<br />

Patrick S. Pittard, Atlanta<br />

Ninth District (2008)<br />

Doreen Stiles Poitevint, Bainbridge<br />

State-at-Large (2011)<br />

Willis J. Potts, Jr., Rome<br />

Eleventh District (2013)<br />

Wanda Yancey Rodwell, Stone Mountain<br />

Fourth District (2012)<br />

J. Timothy Shelnut, Augusta<br />

Twelfth District (2007)<br />

Benjamin Tarbutton III, Sandersville<br />

Third District (2013)<br />

Richard L. Tucker, Lawrenceville<br />

Seventh District (2012)<br />

Officers and Staff<br />

Errol B. Davis, Jr.<br />

Chancellor<br />

Ms. Corlis P. Cummings<br />

Chief Operating Officer &<br />

Executive Vice Chancellor<br />

Office <strong>of</strong> Administrative & Fiscal Affairs<br />

Dr. Beheruz N. Sethna<br />

Interim Chief Academic Officer &<br />

Executive Vice Chancellor<br />

Office <strong>of</strong> Academic Affairs<br />

Frank A. Butler<br />

Vice Chancellor<br />

Academics, Faculty and Student Affairs<br />

Dr. Daniel W. Rahn, M.D.<br />

Sr. Vice Chancellor, <strong>Health</strong> and<br />

Medical Programs & President<br />

Medical <strong>College</strong> <strong>of</strong> <strong>Georgia</strong><br />

William Bowes<br />

Vice Chancellor<br />

Office <strong>of</strong> Fiscal Affairs<br />

Thomas E. Daniel<br />

Senior Vice Chancellor<br />

Office <strong>of</strong> External Affairs<br />

Linda M. Daniels<br />

Vice Chancellor<br />

Facilities<br />

Dr. Cathie M. Hudson<br />

Associate Vice Chancellor<br />

Strategic <strong>Research</strong> & Analysis<br />

Dr. Tom Maier<br />

Interim Vice Chancellor<br />

Information & Instructional Technology/CIO<br />

<strong>The</strong> <strong>University</strong> <strong>of</strong> <strong>Georgia</strong><br />

<strong>University</strong> & <strong>College</strong> Administrators<br />

Michael F. Adams<br />

President<br />

<strong>The</strong> <strong>University</strong> <strong>of</strong> <strong>Georgia</strong><br />

Arnett C. Mace, Jr.<br />

Senior Vice President for Academic Affairs and<br />

Provost<br />

<strong>The</strong> <strong>University</strong> <strong>of</strong> <strong>Georgia</strong><br />

David Lee<br />

Vice President for <strong>Research</strong> and Associate Provost<br />

<strong>The</strong> <strong>University</strong> <strong>of</strong> <strong>Georgia</strong><br />

Sheila Allen<br />

Dean<br />

<strong>College</strong> <strong>of</strong> Veterinary Medicine<br />

Harry W. Dickerson<br />

Director<br />

Veterinary Medical Experiment Station<br />

Veterinary Advisory Board<br />

John Callaway, President<br />

<strong>Georgia</strong> Cattlemen’s Association<br />

Lee Myers, State Veterinarian<br />

<strong>Georgia</strong> Department <strong>of</strong> Agriculture<br />

Bill Taff, Chairman<br />

Equine Advisory Board<br />

Tom Thompson, President<br />

<strong>Georgia</strong> Milk Producers<br />

Steve Healy, President<br />

<strong>Georgia</strong> Pork Producers Association<br />

John Bruno, President<br />

<strong>Georgia</strong> Poultry Federation<br />

D. West Hamryka, President<br />

<strong>Georgia</strong> Veterinary Medical Association<br />

Lee Izen, Past President<br />

<strong>Georgia</strong> Veterinary Medical Association<br />

Council to the Advisory Board<br />

Jim Collins<br />

Executive Vice President<br />

<strong>Georgia</strong> Cattlemen’s Association<br />

Melinda Dennis<br />

Director, Equine Division<br />

<strong>Georgia</strong> Equine Advisory Board<br />

Wayne Dollar<br />

President<br />

<strong>Georgia</strong> Farm Bureau<br />

Charles Griffin<br />

Executive Secretary<br />

<strong>Georgia</strong> Pork Producers Association<br />

Abit Massey<br />

Executive Director<br />

<strong>Georgia</strong> Poultry Association<br />

James Scroggs<br />

Executive Director<br />

<strong>Georgia</strong> Poultry Lab Improvement<br />

Association, Inc.<br />

M. Randy Clayton<br />

Director<br />

<strong>Georgia</strong> Sheep and Wool<br />

22 www.vet.uga.edu/research/vmes/


VMES Faculty<br />

& <strong>Research</strong>ers<br />

Allen, Sheila, Pr<strong>of</strong>essor and Dean, Small <strong>Animal</strong><br />

Medicine, (706) 542-5717<br />

Alvarez, Rene, Asst. <strong>Research</strong> Scientist, Infectious<br />

Diseases, (706) 542-9807<br />

Austel, Michaela, Asst. Pr<strong>of</strong>essor, Small <strong>Animal</strong><br />

Medicine, (706) 542-6386<br />

Baldwin, Charles, Director, Tifton Diagnostic Lab,<br />

(229) 386-3340<br />

Blas-Machado, Uriel, Asst. Pr<strong>of</strong>essor, Athens Diagnostic<br />

Lab, (706) 542-5349<br />

Brown, Cathy, Pathologist, Athens Diagnostic Lab,<br />

(706) 542-5917<br />

Brown, Corrie, Pr<strong>of</strong>essor, Pathology, (706) 542-5842<br />

Brown, Scott, Pr<strong>of</strong>essor and Assoc. Dean for Academic<br />

Affairs, Physiology and Pharmacology,<br />

(706) 542-5857<br />

Budsberg, Steven, Pr<strong>of</strong>essor, Small <strong>Animal</strong> Medicine,<br />

(706) 542-6314<br />

Calvert, Clay, Pr<strong>of</strong>essor, Small <strong>Animal</strong> Medicine,<br />

(706) 542-6375<br />

Chambers, Jonathan, Pr<strong>of</strong>essor, Small <strong>Animal</strong> Medicine,<br />

(706) 542-6313<br />

Ciembor, Paula, Asst. Res. Scientist, Small <strong>Animal</strong><br />

Medicine, (706) 542-1267<br />

C<strong>of</strong>field, Julie, Asst. Pr<strong>of</strong>essor, Physiology and Pharmacology<br />

(706) 542-5979<br />

Corn, Joseph, Public Serv. Asst. Wildlife,<br />

(706) 542-5707<br />

Cornell, Karen, Assoc. Pr<strong>of</strong>essor, Small <strong>Animal</strong><br />

Medicine, (706) 542-6379<br />

Crochik, Sonia, Asst. Pr<strong>of</strong>essor, Anatomy and Radiology,<br />

(706) 542-6368<br />

Crowell-Davis, Sharon, Pr<strong>of</strong>essor<br />

Anatomy and Radiology, (706) 542-8343<br />

Dickerson, Harry, Pr<strong>of</strong>essor and Assoc. Dean for<br />

<strong>Research</strong>, Infectious Diseases, (706) 542-5794<br />

Dietrich, Ursula, Asst. Pr<strong>of</strong>essor, Small <strong>Animal</strong><br />

Medicine, (706) 542-6432<br />

Dookwah, Hugh, Instructor, Anatomy and Radiology,<br />

(706) 542-5332<br />

Edwards, Gaylen, Pr<strong>of</strong>essor, Physiology and Pharmacology,<br />

(706) 542-5854<br />

Evans, Donald, Pr<strong>of</strong>essor, Infectious Diseases,<br />

(706) 542-4796<br />

Ferguson, Duncan, Pr<strong>of</strong>essor, Physiology and Pharmacology,<br />

(706) 542-5864<br />

Fu, Zhen, Assoc. Pr<strong>of</strong>essor, Pathology,<br />

(706) 542-7021<br />

Garcia, Maricarmen, Assoc. Pr<strong>of</strong>essor, Population<br />

<strong>Health</strong>, (706) 542-5656<br />

Glisson, John, Department Head, Population <strong>Health</strong>,<br />

(706) 542-5657<br />

Halper, Jaroslava, Pr<strong>of</strong>essor, Pathology,<br />

(706) 542-5830<br />

Harmon, Barry, Pr<strong>of</strong>essor, Pathology, (706) 542-5831<br />

Hensel, Patrick, Instructor, Small <strong>Animal</strong> Medicine,<br />

(706) 542-9566<br />

Hernandez-Divers, Stephen, Asst. Pr<strong>of</strong>essor, Small<br />

<strong>Animal</strong> Medicine, (706) 542-6472<br />

Hodge, Thomas, Sr. Res. Scientist, Infectious Diseases,<br />

(706) 542-5790<br />

Hoenig, Margarethe, Pr<strong>of</strong>essor, Physiology and<br />

Pharmacology, (706) 542-5869<br />

H<strong>of</strong>acre, Charles, Pr<strong>of</strong>essor, Population <strong>Health</strong>,<br />

(706) 542-5653<br />

H<strong>of</strong>meister, Eric, Resident, Small <strong>Animal</strong> Medicine,<br />

(706) 583-0252<br />

Hogan, Robert J., Asst. Pr<strong>of</strong>essor, Anatomy and<br />

Radiology, (706) 542-6487<br />

Hollett, Bruce, Assoc. Pr<strong>of</strong>essor, Large <strong>Animal</strong> Medicine,<br />

(706) 542-5508<br />

Hondalus, Mary K., Asst. Pr<strong>of</strong>essor, Infectious Diseases,<br />

(706) 542-5778<br />

Howerth, Elizabeth, Pr<strong>of</strong>essor, Pathology,<br />

(706) 542-5833<br />

Jackwood, Mark, Pr<strong>of</strong>essor, Population <strong>Health</strong>,<br />

(706) 542-5475<br />

Jain, Anant, Pr<strong>of</strong>essor, Athens Diagnostic Lab,<br />

(706) 542-5919<br />

Karls, Russell, Senior Res. Scientist, Infectious Diseases,<br />

(706) 542-5791<br />

Keel, Kevin, Asst. Res. Scientist, Wildlife,<br />

(706) 542-5697<br />

Kent, Marc, Asst. Pr<strong>of</strong>essor, Small <strong>Animal</strong> Medicine,<br />

(706) 542-2752<br />

Kleven, Stanley, Pr<strong>of</strong>essor and Department Head,<br />

Population <strong>Health</strong>, (706) 542-5644<br />

Krunkosky, Thomas, Assoc. Pr<strong>of</strong>essor, Anatomy and<br />

Radiology, (706) 583-0543<br />

Latimer, Kenneth, Pr<strong>of</strong>essor, Pathology,<br />

(706) 542-5844<br />

Lee, Margie, Pr<strong>of</strong>essor, Population <strong>Health</strong>,<br />

(706) 542-5778<br />

LeRoy, Bruce, Asst. Pr<strong>of</strong>essor, Pathology,<br />

(706) 542-5847<br />

Lewis, Stephen, Asst. Pr<strong>of</strong>essor, Physiology and Pharmacology,<br />

(706) 542-5862<br />

Lowder, Michael, Assoc. Pr<strong>of</strong>essor, Large <strong>Animal</strong><br />

Medicine, (706) 542-6431<br />

Maurer, John, Assoc. Pr<strong>of</strong>essor, Population <strong>Health</strong>,<br />

(706) 542-5071<br />

McCall, John, Pr<strong>of</strong>essor, Infectious Diseases,<br />

(706) 542-8449<br />

McCombs, Candace, Adjunct Faculty, Infectious<br />

Diseases, (706) 542-6516<br />

Mead, Daniel, Asst. Rsch Scientist, Wildlife,<br />

(706) 542-1741<br />

Miller, Debra Lee, Pathologist, Tifton Diagnostic Lab,<br />

(229) 386-3340<br />

Miller, Doris, Pr<strong>of</strong>essor and Director, Athens Diagnostic<br />

Lab, (706) 542-5915<br />

Mispagel, Michael, CVM Grants Officer, Dean’s Office,<br />

(706) 542-5729<br />

Murray, James, <strong>Research</strong> Scientist, Infectious Diseases,<br />

(706) 542-9883<br />

Murray, Thomas, Pr<strong>of</strong>essor and Department Head,<br />

Physiology and Pharmacology, (706) 542-3014<br />

Mysore, Jagannatha, Asst. Pr<strong>of</strong>essor, Pathology,<br />

(706) 542-5850<br />

Northrup, Nicole, Asst. Pr<strong>of</strong>essor, Small <strong>Animal</strong><br />

Medicine, (706) 542-7415<br />

Okinaga, Tatsuyuki, Asst. Rsch Scientist, Population<br />

<strong>Health</strong> and Large <strong>Animal</strong> Medicine, (706) 542-6340<br />

Parks, Andrew, Pr<strong>of</strong>essor and Department Head,<br />

Large <strong>Animal</strong> Medicine, (706) 542-6372<br />

Pence, Melvin, Assoc. Pr<strong>of</strong>essor, Population <strong>Health</strong>,<br />

(229) 386-3340<br />

Peroni, John, Asst. Pr<strong>of</strong>essor, Large <strong>Animal</strong> Medicine,<br />

(706) 542-9321<br />

Rajeev, Sreekumari, Asst. Pr<strong>of</strong>essor, Tifton Diagnostic<br />

Lab, (229) 386-3340<br />

Rakich, Pauline, Pathologist, Athens Diagnostic<br />

Lab, (706) 542-5903<br />

Rawlings, Clarence, Pr<strong>of</strong>essor, Small <strong>Animal</strong> Medicine,<br />

(706) 542-6317<br />

Ritchie, Branson, Pr<strong>of</strong>essor, Small <strong>Animal</strong> Medicine,<br />

(706) 542-6316<br />

Roberts, Royce, Pr<strong>of</strong>essor and Department Head,<br />

Anatomy and Radiology, (706) 542-8309<br />

Robertson, Thomas, Asst. Rsch Scientist, Physiology<br />

and Pharmacology, (706) 583-0979<br />

Sanchez, Susan, Microbiologist, Athens Diagnostic<br />

Lab, (706) 583-0518<br />

Sanderson, Sherry, Asst. Pr<strong>of</strong>essor, Physiology and<br />

Pharmacology, (706) 542-6378<br />

Scherzer, Jakob, Asst. Pr<strong>of</strong>essor, Large <strong>Animal</strong><br />

Medicine, (706) 542-6319<br />

Sebastian, Manu M., Asst. Pr<strong>of</strong>essor, Tifton Diagnostic<br />

Lab, (229) 386-3340<br />

Selcer, Barbara, Pr<strong>of</strong>essor, Anatomy and Radiology,<br />

(706) 542-5550<br />

Sellers, Holly, Asst. Pr<strong>of</strong>essor, Population <strong>Health</strong>,<br />

(706) 542-5647<br />

Sharma, Raghubir, Fred C. Davison Pr<strong>of</strong>essor,<br />

Physiology and Pharmacology, (706) 542-2788<br />

Stallknecht, David, Assoc. Pr<strong>of</strong>essor, Infectious<br />

Diseases, (706) 542-7952<br />

Sum, Steffen O., Instructor, Small <strong>Animal</strong> Medicine,<br />

(706) 542-6432<br />

Supakorndej, Prasit, Asst. Rsch Scientist, Infectious<br />

Diseases, (706) 542-5683<br />

Thompson, Larry, Asst. Pr<strong>of</strong>essor, Tifton Diagnostic<br />

Lab, (229) 386-3340<br />

Tompkins, Mark, Asst. Pr<strong>of</strong>essor, Infectious Diseases,<br />

(706) 542-4716<br />

Trim, Cynthia, Pr<strong>of</strong>essor, Large <strong>Animal</strong> Medicine,<br />

(706) 542-6318<br />

Tripp, Ralph, Pr<strong>of</strong>essor and Eminent Scholar, Infectious<br />

Diseases, (706) 542-5778<br />

Uhl, Elizabeth, Asst. Pr<strong>of</strong>essor, Pathology,<br />

(706) 583-0475<br />

Vandenplas, Michel, Asst. Rsch Scientist, Large<br />

<strong>Animal</strong> Medicine, (706) 542-6389<br />

Varela-Stokes, Andrea, Asst. Res. Scientist, Infectious<br />

Diseases, (706) 542-0195<br />

Villegas, Pedro, Pr<strong>of</strong>essor, Population <strong>Health</strong>, (706)<br />

542-5085<br />

Ward, Cynthia R., Assoc. Pr<strong>of</strong>essor, Small <strong>Animal</strong><br />

Medicine, (706) 542-6380<br />

White, Susan, Pr<strong>of</strong>essor and Acting Assoc. Dean<br />

for Public Services and Outreach Programs, Large<br />

<strong>Animal</strong> Medicine, (706) 542-6319<br />

Williams, Jamie, Asst. Pr<strong>of</strong>essor, Anatomy and<br />

Radiology, (706) 542-8342<br />

Williamson, Lisa, Assoc. Pr<strong>of</strong>esor, Large <strong>Animal</strong><br />

Medicine, (706) 542-9323<br />

Wilson, Heather, Asst. Pr<strong>of</strong>essor, Small <strong>Animal</strong><br />

Medicine, (706) 542-6328<br />

Woolums, Amelia, Asst. Pr<strong>of</strong>essor, Large <strong>Animal</strong><br />

Medicine, (706) 542-9329<br />

Yabsley, Michael, Asst. Res. Scientist, Wildlife,<br />

(706) 542-1741<br />

Zavala, Guillermo, Asst. Pr<strong>of</strong>essor, Population<br />

<strong>Health</strong>, (706) 542-5058<br />

VMES 2006 23


Selected<br />

Publications<br />

Aaronson, P.I., Robertson, T.P., Knock, G.A., Becker, S., Lewis, T.H., Snetkov, V.A., Ward, J.P.T. Hypoxic pulmonary vasoconstriction: Mechanisms and controversies.<br />

J. Physiol. 570:53-58, 2006.<br />

Agnello, K.A., Reynolds, L.R., Budsberg, S.C. In vivo effects <strong>of</strong> tepoxalin, a dual COX/LOX inhibitor, on prostanoid and leukotriene production in dogs with osteoarthritis.<br />

Am. J. Vet. Res. 66:966-972, 2005.<br />

Agnello, K.A., Trumble, T.N., Chambers, J.N., Seewald, W., Budsberg, S.C. <strong>The</strong> effects <strong>of</strong> zoledronate on markers <strong>of</strong> bone metabolism and subchondral bone mineral<br />

density in an experimental model <strong>of</strong> canine osteoarthritis. Am. J. Vet. Res. 66:1487-1495, 2005.<br />

Agrawal, A., Tripp, R.A., Anderson, L.J., Nie, S., Real-time detection <strong>of</strong> virus particles and viral protein expression with two-color nanoparticle probes. J. Virol.<br />

79(13):8625-8628, 2005.<br />

Agrawal, A., Zhang, C., Byassee, T., Tripp, R.A., Nie, S. Counting single native biomolecules and intact viruses with color-coded nanoparticles. Anal. Chem.<br />

78(4):1061-1070, 2006.<br />

Alvarado, I. R., P. Villegas, N. Mossos, and M. W. Jackwood. Molecular Characterization <strong>of</strong> Avian Infectious Bronchitis Virus Strains Isolated in Colombia During<br />

2003. Avian Dis. 494-499, 2005.<br />

Alvarado, I., P. Villegas, J. El-Attrache, M. W. Jackwood. Detection <strong>of</strong> Massachusetts and Arkansas Serotypes <strong>of</strong> Infectious Bronchitis Virus in Broilers. Avian Dis. Dec.<br />

2005.<br />

Alwood, A.J., Brainard, B.M. , Lafond, E., Drobatz, K.J., and King, L.G. Postoperative Pulmonary Complications. In Dogs Undergoing Laparotomy: Frequency,<br />

Characterization And Disease-Related Risk Factors. J. Vet. Emerg. Crit. Care, (in press), 2006.<br />

Aragon, C.L., Budsberg, S.C. Applications <strong>of</strong> Evidence-Based Medicine: Cranial cruciate ligament injury repair in the dog. Vet. Surg. 34:93-98, 2005.<br />

Austel, M., Hensel, P., Jackson, D., Vidyashankar, A., Zhao, Y., and Medleau, L. Evaluation <strong>of</strong> three different histamine concentrations in intradermal testing <strong>of</strong> normal<br />

cats and attempted determination <strong>of</strong> “irritant” threshold concentrations for 48 allergens. Vet. Derm. 17:189-194, 2006.<br />

Bergh, M.S., and Budsberg, S.C. <strong>The</strong> Coxib NSAIDs: Potential clinical and pharmacological importance in veterinary medicine. J. Vet. Int. Med. 19:633-643, 2005.<br />

Biffa, D. and Woldemeskel, M. Causes and Factors Associated With Occurrence <strong>of</strong> External Injuries in Working Equines in Ethiopia. Intern. J. Appl. Res. Vet. Med..<br />

4(1):1-7, 2006.<br />

Biffa, D., Debela, E., Beyene, F., and Woldemeskel, M Factors associated with bovine udder infections in small holder dairy farms in Ethiopia. Bull. Anim. Hlth. Prod.<br />

Afr. 53:258-265, 2005.<br />

Brainard, B.M., Alwood, A.J., Kushner, L.I., Drobatz, K.J., and King, L.G. Postoperative Pulmonary Complications In Dogs Undergoing Laparotomy: Anesthetic and<br />

perioperative factors. J. Vet. Emerg. Crit. Care, (in press), 2006<br />

Brainard, B.M., Campbell, V.L., Drobatz, K.J., and Perkowski, S.Z. Effects <strong>of</strong> surgery and anesthesia on serum ionized magnesium and ionized calcium concentrations<br />

in the healthy canine patient. Vet. Anaes. and Analg., (in press), 2006.<br />

Brainard, B.M., Meredith, C.P., Callan, M.B., Budsberg, S.C., and Otto, C.M. Evidence <strong>of</strong> hypercoagulability in dogs treated with acetylsalicylic acid. Abstracts from<br />

the 11th International Veterinary Emergency and Critical Care Symposium. J. Vet. Emerg. Crit. Care 15(s1):S1-S13. 2005<br />

Brainard, B.M., and Mandell, D.C. Pheochromocytoma. In Manual <strong>of</strong> Critical Care, Silverstein, D.C. and Hopper, K.A., eds. Elsevier publishing, (in press).<br />

Brainard, B.M., Mandell, D.C. Pulmonary artery catheters. In Manual <strong>of</strong> Critical Care, Silverstein, D.C. and Hopper, K.A., eds. Elsevier publishing, (in press).<br />

Brainard, B.M., King, L.G. Feline lower airway disease. In Manual <strong>of</strong> Feline Emergency and Critical Care. Costello, M.C and Drobatz, K.J., eds. Blackwell Publishing,<br />

(in press).<br />

Brown, C.A., Munday, J.S., Mathur, S., Brown, S.A. Hypertensive encephalopathy in cats with reduced renal function. Vet Pathol. 42(5):642-649, 2005.<br />

Buchanan, M.F., Carter, W.C., Cowgill, L.M., Hurley, D.J., Lewis, S.J., MacLeod, J.N., Melton, T.R., Moore, J.N., Pessah, I., Roberson, M., Robertson, T.P., Smith,<br />

M.L., Vandenplas, M.L. Using 3-D Animation to teach intracellular signal transduction mechanisms:taking the arrows out <strong>of</strong> cells. J. Vet. Med. Edu. 32:72-78, 2005.<br />

Budsberg, S.C., Lenz, M.E., Thona,r E.J.-M.A. Cruciate ligament transection in the canine knee induces both local and distant changes in the metabolism <strong>of</strong> aggrecan<br />

and hyaluronan. Am. J. Vet. Res. 76:429-432, 2006.<br />

Burek, K.A., Gulland, F.M., Sheffield, G., Beckmen, K.B., Keyes, E., Spraker, T.R., Smith, A.W., Skilling, D.E., Evermann, J.F., Stott, J.L., Saliki, J.T., Trites, A.W.<br />

Infectious disease and the decline <strong>of</strong> steller sea lions (Eumetopias jubatus) in Alaska, USA: insights from serologic data. J. Wildl. Dis. 41, 512-524, 2005.<br />

24 www.vet.uga.edu/research/vmes/


Callison, S. A., D. A. Hilt, and M. W. Jackwood. Using DNA shuffling to create novel infectious bronchitis virus S1 genes: Implications for S1 gene recombination.<br />

Virus Genes 31:5-11, 2005.<br />

Callison, S. A., D. A. Hilt, and M. W. Jackwood. Design, Synthesis and in vitro analysis <strong>of</strong> a hammerhead ribozyme targeted to infectious bronchitis virus nucleocapsid<br />

mRNA. Avian Dis. 49:159-163, 2005.<br />

Calvert, C.A., Rawlings, C.A. Heartworm disease: Dogs. In <strong>The</strong> 5 Minute Veterinary Consult. Clinical Companion. Canine and Feline Infectious Diseases and Parasitology.<br />

Ed Barr SC, Bowman DD, Blackwell, Ames, IA, 275-281, 2006<br />

Chambers, J.N., McBride, M.P. and Hernandez-Divers, S.J. Dynamic crossed-pin fixation <strong>of</strong> a distal femoral growth plate fracture in a domestic rabbit (Oryctolagus<br />

cuniculus). J. Exotic Mammal Med. Surg. 3:4-5, 2005.<br />

Cistola, A.C. and Ward, C.R. Hypoparathyroid Disease. Standards <strong>of</strong> Care. Accepted for publication, 2006.<br />

Collett, S. R., D K Thomson D York and S P R Bisschop. A Floor Pen Study to Evaluate the Serological Response <strong>of</strong> Broiler Breeders After Vaccination with TS-11<br />

Strain Mycoplasma gallisepticum Vaccine. Avian Dis., (in press), 2005.<br />

Corn, J.L., E.J.B. Manning, S. Sreevatsan, and J.R. Fischer. Isolation <strong>of</strong> Mycobacterium avium subsp. paratuberculosis from free-ranging birds and mammals on<br />

livestock premises. App. Environ. Microbiology 71(11):6963-6967, 2005.<br />

Cox, N.A., C.L. H<strong>of</strong>acre, R.J. Buhr, J.L. Wilson, J.S. Bailey, L.J. Richardson, D.E. Cosby, M.T. Musgrove, K.L. Hiett, and S.M. Russell. Attempts to isolate naturally<br />

occurring Campylobacter, Salmonella, and Clostridium perfringens from the ductus deferens, testes, and cecal <strong>of</strong> commercial broiler breeder roosters. J. Appl. Poult.<br />

Res. 14:1-4, 2005.<br />

Cox, N.A., C.L. H<strong>of</strong>acre, J.S. Bailey, R.J. Buhr, J.L. Wilson, K.L. Hiett, L.J. Richardson, M.T. Muscrove, D.E. Cosby, J.D. Tankson, Y.L. Vizzier, P.F. Cray, L.E.<br />

Vaughn, P.S. Holt, and D.V. Bourassa. Presence <strong>of</strong> Campylobacter jejuni in various organs one hour, one day, and one week following oral or intracloacal inoculations<br />

<strong>of</strong> broiler chicks. Avian Dis. 49:155-158, 2005.<br />

Crowell-Davis, S.L. Diagnosis <strong>of</strong> Behavior Problems. Journal <strong>of</strong> Veterinary Clinical Behavior. 1, 2006.<br />

Crowell-Davis, S.L. Selecting psychoactive medications for behavior problems. Compendium on Continuing Education for the Practicing Veterinarian, 28, 2006<br />

Crowell-Davis, S.L. Behavior Problems in Pet Rabbits. J. Exotic Pet Medicine (accepted)<br />

Crowell-Davis, S.L. and Murray, T.F. Veterinary Psychopharmacology. Blackwell Publishing, Ames, Iowa, 2006.<br />

Crowell-Davis, S.L. Social Organization and Communication. In <strong>The</strong> Welfare <strong>of</strong> Cats. Ed. I. Rochlitz. Springer Publishing, 2005.<br />

Crowell-Davis, S.L. Introduction to Behavioral Diagnosis and Treatment. In Handbook <strong>of</strong> Small <strong>Animal</strong> Practice. W.B. Saunders, Philadelphia, 5th edition (in press)<br />

Crowell-Davis, S.L. Feline Behavioral Disorders. In Handbook <strong>of</strong> Small <strong>Animal</strong> Practice. W.B. Saunders, Philadelphia, 5th edition (in press)<br />

Crowell-Davis, S.L. Canine Behavioral Disorders In Handbook <strong>of</strong> Small <strong>Animal</strong> Practice. W.B. Saunders, Philadelphia, 5th edition (in press)<br />

Debnam, A.L., Jackson, C.R., Avellaneda, G.E., Barrett, J.B., and H<strong>of</strong>acre, C.L. Effect <strong>of</strong> growth promotant usage on enterococci species on a poultry farm. Avian Dis.<br />

49:361-365, 2005.<br />

de Lahunta, A., Glass, E.N., Kent, M. Classifying involuntary muscle contractions. Comp. Cont. Ed. Pract. Vet. 28(7):516-530, 2006.<br />

Driessen, B., Brainard, B.M. Fluid <strong>The</strong>rapy for the traumatized patient. J. Vet. Emerg. Crit. Care, (in press), 2006.<br />

DuLaney, D.B., Purinton, T., Dookwah, H., Budsberg, S.C. Effects <strong>of</strong> starting distance on vertical ground reaction forces in the normal dog. Vet. Comp. Ortho. Trauma<br />

3:183-185, 2005.<br />

Dubay, S.A., Rosenstock, S.S., Stallknecht, D.E. and deVos, Jr., J.C. Determining prevalence <strong>of</strong> bluetongue and epizootic hemorrhagic disease viruses in mule deer in<br />

Arizona (USA) using whole blood dried on paper strips compared to serum analyses. J. Wildlife Diseases 42(1):159-163, 2006.<br />

Dugan, V.G., Yabsley, M.J., Tate, C.M., Mead, D.G., Munderloh, U.G., Herron, M.J., Stallknecht, D.E., Little, S.E., and Davidson, W.R. Evaluation <strong>of</strong> White-Tailed<br />

Deer (Odocoileus virginianus) as Natural Sentinels for Anaplasma phagocytophilum. Vector-Borne Zoonotic Diseases 6:192-207, 2006.<br />

Ellis, A.E., Mead, D.G., Allison, A.B., Gibbs, S.E.J., Gottdenker, N.L., Stallknecht, D.E., and Howerth, E.W. Comparison <strong>of</strong> immunohistochemistry and virus isolation<br />

for diagnosis <strong>of</strong> West Nile virus. J. Clin. Microbiol. 43:2904-2908, 2005.<br />

Estevez, C., and Villegas, P. Sequence Analysis, Viral Rescue from Infectious Clones, and Generation <strong>of</strong> Recombinant Virions <strong>of</strong> the Avian Adeno-Asssociated Virus.<br />

Virus Res. 105:195-208, 2004.<br />

Estevez, C., and Villegas, P. Recombinant Avian Adeno-Associated Virus: Transgene Expression in vivo and Enhancement <strong>of</strong> Expression in vitro. Avian Dis., (in<br />

press), 2006.<br />

Evans, D.L., Kaur, H., Leary, J., Praveen, K., and Jaso-Friedmann, L. Molecular characterization <strong>of</strong> a novel pattern recognition protein from nonspecific cytotoxic<br />

cells: sequence analysis, phylogenetic comparisons and antimicrobial activity <strong>of</strong> a recombinant homologue. Dev. Comp. Immunol. 29(12):1049-1064, 2005..<br />

Fadly, A.M., Witter, R. L. and Zavala, G. Reticuloendotheliosis. In Diseases <strong>of</strong> Poultry. 12th Ed. Iowa State Press. Ames, IA.<br />

Fairchild, A.S., Smith, J.L., Idris, U., Lu, J., Sanchez, S., Purvis, L.B., H<strong>of</strong>acre, C.L., Lee, M.D. Effects <strong>of</strong> orally administered tetracycline on the intestinal community<br />

<strong>of</strong> chickens and tet determinant carriage by commensal bacteria and Campylobacter jejuni. Appl. Environ. Microbiology 71(10): 5865-5872, 2005.<br />

VMES 2006 25


Fairchild, A., Lee, M.D. and Maurer, J.J. PCR basics. In PCR Methods in Foods, John Maurer, ed., Springer, New York, N.Y., pp. 1-25, 2006.<br />

Fairchild, A.S., J.L. Smith, U. Idis, J. Lu, S. Sanchez, L.B. Purvis, C. H<strong>of</strong>acre, and M.D. Lee. Oral tetracycline administration in chickens and its effect on the intestinal<br />

community structure and the tet determinant carriage <strong>of</strong> commensal bacteria and Campylobacter jejuni. Appl. Environ. Microbiol, 71(10):5865-5872.<br />

Farrar, M.D., Miller, D.L., Baldwin, C. A., Stiver, S.L. and Hall, C. L. Eastern equine encephalitis in dogs. J. Veterinary Diagnostic Investigation 17:606-609, 2005.<br />

Ferguson, N. M., Hepp, D., Sun, S., Ikuta, N., Levisohn, S., Kleven, S.H. and García, M. Use <strong>of</strong> molecular diversity <strong>of</strong> Mycoplasma gallisepticum by gene-targeted<br />

sequencing (GTS) and random amplified polymorphic DNA (RAPD) analysis for epidemiological studies. Microbiology 151:1883–1893, 2005.<br />

Fikru, R., Bonnet, P., Moges, W. Prevalence <strong>of</strong> bovine tuberculosis in indigenous Zebu cattle under extensive farming system in western Ethiopia. Bull. Anim. Hlth.<br />

Prod. Afr. 53:85-88, 2005.<br />

Fisher, J.W., Campbell, J., Muralidhara, S., Bruckner, J.V., Ferguson, D.C., Mumtaz, M., H El-Masri, H., Harmon, B., Hedge, J., Cr<strong>of</strong>ton, K., Almekinder T.L., Effect <strong>of</strong><br />

PCB 126 on hepatic metabolism <strong>of</strong> thyroxine and pertubations in the hypothalamic- pituitary-thyroid axis in the rat. Toxicological Sciences 90(1):87-95, 2006.<br />

Flaminio, J.B.F., Nydam, D.V., Sebastian, M., Bernard, W.V., Tobin, T., Hillegas, J.M., and Matychak, M.B. <strong>The</strong> Mare Reproductive Loss Syndrome (MRLS) and the<br />

Eastern Tent Caterpillar: Immunological Testing <strong>of</strong> Aborting Mares. Inter. J. Appl. Res. Vet. Med. 3(3):207-216, 2005.<br />

Fleming, S A., Craig, T., Kaplan, R.M., Miller, J. E., Navarre, C., and Rings, M. Consensus Statement on: Anthelmintic resistance <strong>of</strong> gastrointestinal parasites in small<br />

ruminants. J. Vet. Internal Med. 20:435–444, 2006.<br />

Fulton, R.W., Hessman, B., Johnson, B.J., Ridpath, J.F., Saliki, J.T., Burge, L.J., Sjeklocha, D., Confer, A.W., Funk, R.A., and Payton, M.E. Evaluation <strong>of</strong> diagnostic<br />

tests used for detection <strong>of</strong> bovine viral diarrhea virus and prevalence <strong>of</strong> subtypes 1a, 1b, and 2a in persistently infected cattle entering a feedlot. J. Am. Vet. Med. Assoc.<br />

228:578-584, 2006.<br />

García, M., Ikuta, N., Levisohn, S. and Kleven, S.H. Evaluation and Comparison <strong>of</strong> Various PCR Methods for Detection <strong>of</strong> Mycoplasma gallisepticum. Infection in<br />

Chickens 49:125-132, 2005.<br />

Giri, D.K., Seyed, M.A., Li, L., Lee, D.F., Pin, L., Bartholomeusz, G., Wang, S C and Hung, M.C. Endosomal transport <strong>of</strong> HER2: A mechanism for nuclear entry <strong>of</strong><br />

cell surface receptor. Mol. Cell Biol. 25:11005-11018, 2005.<br />

Gopee, N.V., Johnson, V.J. and Sharma, R.P. Sodium selenite-induced apoptosis in murine B-lymphoma cells is associated with inhibition <strong>of</strong> protein kinase C-d, nuclear<br />

factor kB and inhibitor <strong>of</strong> apoptosis protein. Toxicol. Sci. 78:204-214, 2004.<br />

Gopee, N.V., and Sharma, R.P. <strong>The</strong> mycotoxin fumonisin B1 transiently activates nuclear factor-kB, tumor necrosis factor a and caspase 3 via protein kinase Ca-dependent<br />

pathway in porcine renal epithelial cells. Cell Biol. Toxicol. 20:197-212, 2004.<br />

Gorgi, A.A., H<strong>of</strong>meister, E.H., Higginbotham, M.J., Kent, M. Effect <strong>of</strong> body position on cranial migration <strong>of</strong> epidurally injected methylene blue in recumbent dogs.<br />

Am. J. Vet. Res. 67:219-221, 2006.<br />

Graves, J.E., Lewis, S.J., Kooy, N.W. Loss <strong>of</strong> K+ATP-channel-mediated vasodilation after induction <strong>of</strong> tachyphylaxis to peroxynitrite. J. Cardiovasc. Pharmacol.<br />

46:646-652, 2005.<br />

Graves, J.E., Kooy, N.W., Lewis, S.J. L-b,b-dimethylcysteine attenuates the hemodynamic responses elicited by systemic injections <strong>of</strong> peroxynitrite in anesthetized rats.<br />

Brit. J. Pharmacol. 148:7-15, 2006.<br />

Graves, J.E., Lewis, S.J., Kooy, N.W. Role <strong>of</strong> ATP-sensitive K+-channels in the hemodynamic effects <strong>of</strong> peroxynitrite in anesthetized rats. J. Cardiovasc Pharmacol.<br />

46:653-659, 2005.<br />

Graves, J.E., Bates, J.N., Kooy, N.W., Lewis, S.J. Vasodilator actions <strong>of</strong> the endothelium-derived relaxing factor L-S-nitrosocysteine in anesthetized rats are markedly<br />

diminished by peroxynitrite. Clin Exp Physiol Pharmacol. 32:1137-1141, 2005.<br />

Gray, M.J., Miller, D.L., and Smith, L.M. Coelomic response and signal range <strong>of</strong> implant transmitters in Bufo cognatus. Herpetological Rev. 36:285-288, 2005.<br />

Gibbs, S.E.J., Allison, A.B., Yabsley, M.J., Mead, D.G., Wilcox, B.R. and Stallknecht, D.E. West Nile virus antibodies in avian species <strong>of</strong> <strong>Georgia</strong>, USA: 2000-2004.<br />

Vector-Borne and Zoonotic Diseases 6(1):57-72, 2006.<br />

Gibbs, S.E.J., Ellis, A.E., Mead, D.G., Allison, A.B., Moulton, J. K., Howerth, E.W. and Stallknecht, D.E.<br />

blue jays (Cyanocitta cristata). J. Wildlife Diseases 41(2):354-362, 2005.<br />

West Nile virus detection in the organs <strong>of</strong> naturally infected<br />

Factors affecting the geographic distribution <strong>of</strong> West Nile virus in Geor-<br />

Gibbs, S.E.J., Wimberly, M.C., Madden, M., Masour, J., Yabsley, M.J. and Stallknecht, D.E.<br />

gia, USA: 2002-2004. Vector-Borne and Zoonotic Diseases 6(1):73-82, 2006.<br />

Grewal, S.K., Rajeev, S., Sreevatsan, S., and Michel, F.C. Persistence <strong>of</strong> Mycobacterium avium subsp. paratuberculosis and other zoonotic pathogens during simulated<br />

composting manure packing and liquid storage <strong>of</strong> dairy manure. Appl. Environ. Microbiol. 72:565-574, 2006.<br />

Halper, J., Kim, B., Khan, A., Yoon, J.H., Mueller, P.O.E. Degenerative suspensory ligament desmitis as a systemic disorder characterized by proteoglycan accumulation.<br />

BMC Veterinary <strong>Research</strong> 2:12, 2006.<br />

Hanson, B.A., Swayne, D.E., Senne, D.A., Lobpries, D.S., Hurst, J. and Stallknecht, D.E. Avian influenza viruses and paramyxoviruses in wintering and resident ducks<br />

in Texas. J. Wildlife Diseases 41(3):624-628, 2005.<br />

Harcourt, J., Alvarez, R., Jones, L.P., Henderson, C., Anderson, L.J., and Tripp, R.A. Respiratory syncytial virus G protein and G protein CX3C motif adversely affect<br />

CX3CR1+ T cell responses. J. Immunol. 176(3):1600-8, 2006.<br />

26 www.vet.uga.edu/research/vmes/


Havig, M.E., Cornell, K.K., Hawthorne, J.C., McDonnell, J.J., and Selcer, B.A. Evaluation <strong>of</strong> Nonsurgical Treatment <strong>of</strong> Atlantoaxial Subluxation in Dogs: 19 cases<br />

(1992 - 2001). J. Am. Vet. Med. 227:257-262, 2005.<br />

He, Q., Johnson, V.J., Osuchowski, M.F., and Sharma, R.P. Inhibition <strong>of</strong> serine palmitoyl-transferase by myriocin, a natural mycotoxin, causes induction <strong>of</strong> c-myc in<br />

mouse liver. Mycopathologia 157:339-347, 2004.<br />

He, Q., Kim, J-Y, and Sharma, R.P. Fumonisin B1 hepatotoxicity in mice is attenuated by depletion <strong>of</strong> Kupffer cells by gadolinium chloride. Toxicology 207:137-147,<br />

2005.<br />

He, Q., Riley, R.T., and Sharma R.P. Myriocin prevents fumonisin B1-induced sphingoid base accumulation in mice liver without ameliorating hepatotoxicity. Food<br />

Chem. Toxicol. 43:969-979, 2005.<br />

He, Q., Kim, J-Y., and Sharma R. P. Silymarin protects against liver damage in BALB/c mice exposed to fumonisin B1 despite increasing accumulation <strong>of</strong> free sphingoid<br />

bases. Toxicol. Sci., 80:335-342, 2004.<br />

Hernandez-Divers, S.J. Reptile viral diseases – summary table, Appendix A. In Reptile Medicine and Surgery, Second edition. Mader, D.R., ed. WB Saunders Co,<br />

Philadelphia, PA. pp. 1147-1158, 2006.<br />

Hernandez-Divers, S.J. and Innis, C. Renal disease in reptiles: diagnosis and clinical management. In Reptile Medicine and Surgery, Second edition. Mader, D.R., ed.<br />

WB Saunders Co, Philadelphia, PA. pp. 878-892, 2006.<br />

Hernandez-Divers, S.M., Schumacher, J., Stahl, S. and Hernandez-Divers, S.J. Comparison <strong>of</strong> is<strong>of</strong>lurane and sev<strong>of</strong>lurane following premedication with butorphanol in<br />

the green iguana (Iguana iguana). J. Zoo & Wildlife Medicine 36:169-175, 2005.<br />

Hernandez-Divers, S.J., Stahl, S.J., Stedman, N.L., Hernandez-Divers, S.M., Schumacher, J., Hanley, C.S., Wilson, G.H., Vidyashankar, A.N., Zhao, Y. and Rumbeiha,<br />

W.K. Renal Evaluation in the green iguana (Iguana iguana): Assessment <strong>of</strong> plasma biochemistry, glomerular filtration rate, and endoscopic biopsy. J. Zoo & Wildlife<br />

Medicine 36:155-168, 2005.<br />

Hernandez-Divers, S.J. Reptile parasites – summary table, Appendix B. In Reptile Medicine and Surgery, Second edition. Mader, D.R., ed. WB Saunders Co, Philadelphia,<br />

PA. pp. 1159-1170, 2006.<br />

Hernandez-Divers, S.J. Diagnostic techniques. In Reptile Medicine and Surgery, Second edition. Mader, D.R., ed. WB Saunders Co, Philadelphia, PA. pp. 490-532,<br />

2006.<br />

Hernandez-Divers, S.J. and Cooper, J.E. Hepatic lipidosis. In Reptile Medicine and Surgery, Second edition. Mader, D.R., ed. WB Saunders Co, Philadelphia, PA. pp<br />

806-813, 2006.<br />

Hoenig, M., McGoldrick, DeBeer, M., Demacker, P.N.M., Ferguson, D.C. Activity and tissue-specific expression <strong>of</strong> lipases and tumor-necrosis factor a in lean and<br />

obese cats. Domest. Anim. Endocrinol. 30:333-344, 2006.<br />

Hoenig, M., Caffall, Z.F., McGraw, R.A., and Ferguson, D.C. Cloning, expression and purification <strong>of</strong> feline proinsulin. Domest. Anim.Endocrinol. 30:28-37, 2006.<br />

Hoenig, M., McGoldrick, J.B., deBeer, M., Demacker, P.N.M., Ferguson, D.C. Activity and tissue-specific expression <strong>of</strong> lipases and tumor-necrosis factor a in lean and<br />

obese cats. Domest. Anim. Endocrinol.30:333-344, 2006.<br />

Hoenig, M., Thomaseth, K., Brandao, J., Waldron, M., Ferguson, D.C. Assessment and mathematical modeling <strong>of</strong> glucose turnover and insulin sensitivity in lean and<br />

obese cats. Domest. Anim. Endocrinol., (in press), available online as <strong>of</strong> May, 2006.<br />

H<strong>of</strong>meister, E.H., Herrington, J.L., Mazzaferro, E.M. Opioid dysphoria in three dogs. J. Vet. Emerg. Crit. Care 16:44-49, 2006.<br />

H<strong>of</strong>meister, E.H., Hernandez-Divers, S.J. Anesthesia case <strong>of</strong> the month. J. Am. Vet. Med. Assoc. 227(5):718-720, 2005.<br />

H<strong>of</strong>meister, E.H., Read, M.R., Brainard, B.M. Evaluating veterinarians’ and veterinary students’ knowledge and clinical use <strong>of</strong> pulse oximetry. J. Vet. Med. Educ.<br />

32(2):272-277, 2005.<br />

Hong, Y., García, M., Levisohn, S., Lysnyansky, I., Leiting, V., Savelkoul, P.H.M. and Kleven, S.H. Evaluation <strong>of</strong> Amplified Fragment Length Polymorphism for Differentiation<br />

<strong>of</strong> Avian Mycoplasma Species. J. Clin. Microbiol. 43:909–912, 2005.<br />

Hong, Y., García, M., Levisohn, S., Savelkoul, P., Leiting, V., Lysnyansky, I., Ley, D. and Kleven, S.H. Differentiation <strong>of</strong> Mycoplasma gallispeticum Strains Using<br />

Amplified Fragment Length Polymorphism and other DNA Based Typing Methods. Avian Dis. 49:43–49, 2005.<br />

Hoque, A., Owen, J.R., Bates, J.N., Lewis, S.J. Effects <strong>of</strong> thiol chelation on a1-adrenoceptor-induced vasoconstriction in vivo. J. Cardiovasc. Pharmacol. 46:627-636,<br />

2005.<br />

Howerth, E.W., Mead, D.G., Mueller, P.O.E., Duncan, L., Murphy, M.D., and Stallknecht, D.E. Experimental vesicular stomatitis virus in horses: Effect <strong>of</strong> route <strong>of</strong><br />

inoculation and virus serotype. Vet Path, (in press), 2006.<br />

Idris, U., Lu, J., Maier, M., Sanchez, S., H<strong>of</strong>acre, C.L., Harmon, B.G., Maurer, J.J. and Lee, M.D. Dissemination <strong>of</strong> fluoroquinolone-resistant Campylobacter within an<br />

integrated commercial poultry production system. Accepted by Appl. Environ. Microbiol., March, 2006.<br />

Jackwood, M.W., Hilt, D.A., Lee, C-W., Kwon, H.M., Callison, S.A., Moore, K.M., Moscoso, H., Sellers, H. and Thayer, S. Data from 11 years <strong>of</strong> molecular typing<br />

infectious bronchitis virus field isolates. Avian Dis 49:614-618, 2005.<br />

Jewell, D.E., Toll, P.W., Azain, M.J., Lewis, R.D., Edwards, G.L. Fiber but not conjugated linoleic Acid influences adiposity in dogs. Vet. <strong>The</strong>r. 7(2):78-85, 2006.<br />

Johnson, V.J., He, Q., Osuchowski, M.F. and Sharma, R. P. Disruption <strong>of</strong> sphingolipid homeostasis by myriocin, a mycotoxin, reduces thymic and splenic T-lymphocyte<br />

populations. Toxicology 201:67-75, 2004.<br />

VMES 2006 27


Johnson, V.J., Kim, S.-H., and Sharma R.P. Aluminum-maltolate induces apoptosis and necrosis in neuro-2a cells: potential role for p53 signaling. Toxicol. Sci.,<br />

83:329-339, 2005.<br />

Johnson, V.J., Tsunoda, M., Murray, T.F. and Sharma, R.P. Decreased membrane fluidity and hyperpolarization in aluminum-treated PC-12 cells correlates with increased<br />

production <strong>of</strong> cellular oxidants. Environ. Toxicol. Pharmacol. 19:221-230, 2005.<br />

Kaplan, R.M. Drug resistance in helminth parasites: concepts, perspectives and lessons learned. In Proceedings <strong>of</strong> the 142nd Annual Convention <strong>of</strong> the American Veterinary<br />

Medical Association, Minneapolis, MN July 16-20, 2005.<br />

Karls, R.K., Guarner, J., McMurray, D.N., Birkness, K.A. and Quinn, F.D. Examination <strong>of</strong> Mycobacterium tuberculosis sigma factor mutants using low-dose aerosol<br />

infection <strong>of</strong> guinea pigs suggests a role for SigC in pathogenesis. Microbiol. 152:1591-1600, 2006.<br />

Kaur, H., Jaso-Friedmann, L, Leary J.H. III, Praveen, K., Brahmi, Z., and Evans, D.L. Activation <strong>of</strong> NK-like YT-INDY cells by oligodeoxynucleotides and binding by<br />

homologous pattern recognition proteins. Scand. J. Immunol. 62:361-370, 2005.<br />

Kim, J-Y., and Sharma, R.P. Calcium-mediated activation <strong>of</strong> c-jun NH2-terminal kinase (JNK) and apoptosis in response to cadmium in murine macrophages. Toxicol.<br />

Sci. 81:518-527, 2004.<br />

Kim S.-H. and Sharma R.P. Mercury alters endotoxin-induced inflammatory cytokine expression in liver:differential roles <strong>of</strong> p38 and extracellular signal-regulated<br />

mitogen-activated protein kinases. Immunopharmacol. Immunotoxicol. 27:123-135, 2005.<br />

Kim, S-H., Kim, J., and Sharma, R.P. Inhibition <strong>of</strong> p38 and ERK MAP kinases blocks endotoxin-induced nitric oxide production and differentially modulates cytokine<br />

expression. Pharmacol. Res. 49:433-439, 2004.<br />

Kim, S-H., and Sharma, R.P. Mercury-induced apoptosis and necrosis in murine macrophages:Role <strong>of</strong> calcium-induced reactive oxygen species and p38 mitogen-activated<br />

protein kinase signaling. Toxicol. Appl. Pharmacol. 196:47-57, 2004.<br />

Kleven, S.H. Mycoplasmosis. In Merck Veterinary Manual, 9th Edition. C. M. Kahn ed., Merck & Co., Inc., Whitehouse Station, N.J. pp. 2242-2246, 2005.<br />

Krunkosky, T.M., Mauro, K., Potempta, J., Jarrett, C.L., Travis, J. Inhibition <strong>of</strong> tumor necrosis factor-alpha-induced RANTES secretion by alkaline protease in A549<br />

cells. Am. J. Respir. Cell. Mol. Biol. 33(5):483-489, 2005.<br />

Krunkosky, T.M., and Jarrett, C.L. Selective regulation <strong>of</strong> MAP kinases and chemokine expression after ligation <strong>of</strong> ICAM-1 on human airway epithelial cells. Respir<br />

Res. 7:12, 2006.<br />

Kuppinger, O., Fischer, E., Sum, S., Hirschberger, J., Rausch, W. P., Keene, B. W., Tipett, F., Schulz, R. Lack <strong>of</strong> GNB3 exon 9 polymorphism in primary hypertensive<br />

and normotensive dogs. Vet Rec<br />

Lacolley, P., Sandock, K., Lewis, T.H., Bates, J.N., Robertson, T.P., Lewis, S.J. Occipital artery injections <strong>of</strong> 5-hydroxytryptamine directly activate the cell bodies <strong>of</strong><br />

vagal and glossopharyngeal afferent cell bodies in the rat:Role <strong>of</strong> 5-HT2 and 5-HT3 receptors. Neuroscience, (in press), 2006.<br />

Lacolley, P., Owen, J.R., Bates, J.N., Johnson, A.K., Lewis, S.J. Tachyphylaxis to 5-HT3 receptor-mediated activation <strong>of</strong> vagal afferents is prevented by co-activation <strong>of</strong><br />

5-HT2 receptors. Brain Res. 1093:105-115, 2006.<br />

Lankester, F. and Hernandez-Divers, S.J. Paraphimosis and amputation in a Nile Crocodile (Crocodylus niloticus). J. Zoo Wildl. Med. 36:698-701, 2006.<br />

Lappin, M.R, Jensen, W.A., Basaraba, T.D., Brown, R.J., Radecki, C.A., Hawley, Jr., S.V. Investigation <strong>of</strong> the induction <strong>of</strong> antibodies against Crandell-Rees feline<br />

kidney cell lysates and feline renal cell lysates after parenteral administration <strong>of</strong> vaccines against feline viral rhinotracheitis, calicivirus, and panleukopenia in cats. Am.<br />

J. Vet. Res. 66(3):506-511, 2005.<br />

Lascelles, B.D.X., Roe, S.C., Smith, E., Reynolds, L., Markham, J., Marcellin-Little, D., Bergh, M.S., Budsberg, S.C. Evaluation <strong>of</strong> a pressure walkway system for<br />

measurement <strong>of</strong> vertical limb forces in normal dogs. Am. J. Vet. Res. 76:277-282, 2006.<br />

Lee, M.D., Lu, J., Harmon, B., H<strong>of</strong>acre, C.L. and Maurer, J.J. Molecular basis for AGP effects in poultry. In: Antimicrobial growth promoters Where do we go from<br />

here D. Barug, J. de Jong, A.K. Kies, and M.W.A. Verstegne, eds., Wageningen Academic Publishers, <strong>The</strong> Netherlands. pp. 149-164, 2006.<br />

Lee, M.D., and Maurer, J.J. Colibacillosis. In <strong>The</strong> Merck Veterinary Manual, C. M. Kahn, ed., 9th Edition, National Publishing Inc., Philadelphia, PA. pp. 2221-2222,<br />

2005.<br />

Lee, M.D. and Newell, D.G. Campylobacter in Poultry: filling an ecological niche. Avian Diseases. (in press).<br />

Lee, M.D. Avian Campylobacter Infection. In Merck Veterinary Manual, 9th edition, S. E. Aiello, Editor. Merck & Co., Inc. Whitehouse Station, NJ, pp. 2214-2216,<br />

2005.<br />

LeRoy, B.E., Knight, M.C., Eggleston, R., Torres-Velez, F., and Harmon, B.G. What is your diagnosis Tail-base mass from a “horse <strong>of</strong> a different color. Vet. Clin. Path.<br />

34:69-71, 2005.<br />

Lewis, S.J., Hashmi-Hill, M.P., Owen, J.R., Sandock, K., Robertson, T.P., Bates, J.N. <strong>The</strong> vasodilator potency <strong>of</strong> the endothelium-derived relaxing factor, L S-nitrosocysteine,<br />

is impaired in Spontaneously Hypertensive rats. Relevance to the identity <strong>of</strong> EDRF. Vasc. Pharmacol. 44(6):476-490, 2006.<br />

Lewis, S.J., Hashmi-Hill, M.P., Owen, J.R., Sandock, K., Robertson, T.P., Bates, J.N. ACE inhibition restores the vasodilator potency <strong>of</strong> the endothelium-derived relaxing<br />

factor, L-S-nitrosocysteine, in spontaneously hypertensive rats. Vasc Pharmacol. 44:491-507, 2006.<br />

Lewis, S.J., Travis, M.D., Hashmi-Hill, M.P., Sandock, K., Robertson, T.P., Bates, J.N. Differential effects <strong>of</strong> ouabain on the vasodilator actions <strong>of</strong> nitric oxide and S-<br />

nitrosothiols in vivo. Vasc. Pharmacol., (in press), 2006.<br />

Lewis, S.J., Hoque, A., Sandock, K., Robertson, T.P., Bates, J.N., Kooy, N.W. Differential effects <strong>of</strong> peroxynitrite on the function <strong>of</strong> arginine vasopressin V1a receptors<br />

and alpha1-adrenoceptors in vivo. Vasc. Pharmacol., (in press), 2006.<br />

28 www.vet.uga.edu/research/vmes/


Lewis, S.J., Bhopatkar, M.Y., Walton, T.M., Bates, J.N. Role <strong>of</strong> voltage-sensitive calcium-channels in nitric oxide-mediated vasodilation in spontaneously hypertensive<br />

rats. Eur J Pharmacol. 528:144-149, 2005.<br />

Lewis, S.J., Owen, J.R., Bates, J.N. S-nitrosocysteine elicits hemodynamic responses similar to those <strong>of</strong> the Bezold-Jarisch reflex via activation <strong>of</strong> stereoselective recognition<br />

sites. Eur. J. Pharmacol. 531:254-258, 2006.<br />

Lewis, S.J., Hoque, A., Bates, J.N. Differentiation <strong>of</strong> L- and D-S-nitrosothiol recognition sites. J Cardiovasc. Pharmacol. 46:660-671, 2005.<br />

Lewis, S.J., Graves, J.E., Bates, J.N., Kooy, N.W. Peroxynitrite elicits dysfunction <strong>of</strong> stereoselective S-nitrosocysteine recognition sites. J. Cardiovasc. Pharmacol.<br />

46:637-645, 2005.<br />

Lewis, S.J., Hoque, A., Walton, T.M., Kooy, N.W. Potential role <strong>of</strong> nitration and oxidation reactions in the effects <strong>of</strong> peroxynitrite on the function <strong>of</strong> b-adrenoceptor subtypes<br />

in the rat. Eur. J. Pharmacol. 518:187-194, 2005.<br />

Li, X.Q., Fu, Z.F., Alvarez, R., Henderson, C., Tripp, R.A. Respiratory syncytial virus (RSV) infects neuronal cells and processes that innervate the lung by a process<br />

involving RSV G protein. J. Virol. 80(1):537-540, 2006.<br />

Liljebjelke, K.A., H<strong>of</strong>acre, C.L., Liu, T., White, D.G., Ayers, S., Young, S. and Maurer, J.J. Vertical and horizontal transmission <strong>of</strong> Salmonella within integrated broiler<br />

production system. Foodborne Pathogens and Disease 2(1):90-102, 2005.<br />

Loughin, C.A., Kerwin, S.C., Hosgood, G., Ringwood, P.B., Williams, J., Stefanacci, J.D., and McCarthy, R.J. Clinical signs and results <strong>of</strong> treatment in cats with patellar<br />

luxation: 42 cases (1992-2002). JAVMA 228(9):1370-1375, 2006.<br />

Loughin, C.A., Dewey, C.W., Ringwood, P.B., Pettigrew, R.W., Kent, M., Budsberg, S.C. Effect <strong>of</strong> durotomy on functional outcome <strong>of</strong> dogs with type I thoracolumbar<br />

disc extrusion and absent deep pain perception. Vet. Comp. Ortho. Trauma 3:141-146, 2005.<br />

Lu, J., H<strong>of</strong>acre, C.L., and Lee, M.D. Emerging Technologies in Microbial Ecology Aid in Understand the Complex Disease Necrotic Enteritis. Avian Pathol. (in press).<br />

Lucchi, N. and Moore, J.M. LPS induces secretion <strong>of</strong> chemokines by human syncytiotrophoblast cells in a MAPK-dependent manner. J. Reprod. Immunol., 2006.<br />

Lucchi, N., Koopman, R., Peterson, D.S., and Moore, J.M. Plasmodium falciparum-infected red blood cells selected for binding to cultured syncytiotrophoblast bind to<br />

chondroitin sulfate A and induce tyrosine phosphorylation in the syncytiotrophoblast. Placenta 27(4-5):384-394, 2006.<br />

Lysnyansky, I., García, M. and Levisohn, S. Use <strong>of</strong> mgc2-RFLP-PCR for Rapid Differentiation between Field Isolates and Vaccine Strains <strong>of</strong> Mycoplasma gallisepticum<br />

in Israel. Avian Dis. 49:238–245, 2005.<br />

Mader, D.M., Bennett, A., Funk, R.S., Fitzgerald, K.T., Vera, R., and Hernandez-Divers, S.J. Surgery. In Reptile Medicine and Surgery, Second edition. Mader, D.R.,<br />

ed. WB Saunders Co, Philadelphia, PA. pp 581-630, 2006.<br />

Mahalingam, S., Schwarze, J., Zaid, A., Nissen, M., Sloots, T., Tauro, S., Storer, J., Alvarez, R., and Tripp, R.A. Perspective on the host response to human metapneumovirus<br />

infection: what can we learn from respiratory syncytial virus infections Microbes Infect. 8(1):285-293, 2006.<br />

Mauel, M.J., Miller, D.L., Styer, E., Pouder, D.B., Yanong, R., Goodman, A., and Schwedler, T. Occurrence <strong>of</strong> Piscirickettsiosis-like syndrome in tilapia in the continental<br />

United States. J. Vet. Diag. Invest. 17:601-605, 2005.<br />

Maurer, J.J. <strong>The</strong> mythology <strong>of</strong> PCR: a warning to the wise. PCR Methods in Foods, John Maurer, Ed., Springer, New York, N.Y., pp. 27-40, 2006.<br />

Maurer, J.J. and Lee, M.D. Salmonella: virulence, stress response, and resistance. In Understanding Pathogen Behavior: Virulence, Stress Response and Resistance,<br />

Mansel Griffiths Ed., CRC Press, Boca Raton, FL, and Woodhead Publishing Limited, Cambridge, United Kingdom. pp.215-239, 2005.<br />

McGraw, R.A., Carmichael, K.P. Molecular basis <strong>of</strong> globoid cell leukodystrophy in Irish setters. Vet. J. 171(2):370-372, 2006.<br />

Miller, J.E., Bishop, S.C., Cockett, N.E., McGraw, R.A. Segregation <strong>of</strong> natural and experimental gastrointestinal nematode infection in F(2) progeny <strong>of</strong> susceptible Suffolk<br />

and resistant Gulf Coast Native sheep and its usefulness in assessment <strong>of</strong> genetic variation. Vet. Parasitol. Apr 16; 2006, [Epub ahead <strong>of</strong> print]<br />

Miller, D.L., Mauel, M.J., Liggett, A., Hines, M.E. II, Frazier, K.S., Pence, M., Whittington, L., and Baldwin, C.A. Molecular characterization and phylogenetic analysis<br />

<strong>of</strong> Cryptosporidium spp. by soil provinces in <strong>Georgia</strong>. Vet. J. (in press)<br />

Miller, D.L., Taylor, S.K., Rotstein, D.S., Pough M.B., Barr, M.C., Baldwin, C.A., Cunningham, M., Roelke, M. and Ingram, D. Feline immunodeficiency virus and<br />

puma lentivirus in Florida panthers (Puma concolor coryi): epidemiology and diagnostic issues. Vet. Res. Comm. 30:307-317, 2006.<br />

Miller, D.L., Radi, Z.A., Baldwin, C., and Ingram, D. A Case <strong>of</strong> Fatal West Nile virus in a white-tailed deer (Odocoileus virginianus). J. Wildlife Dis. 41:246-249,<br />

2005<br />

Moore, J.N. Five decades <strong>of</strong> colic: A view thirty-five years on. Equine Vet. J. 37(4), 2005.<br />

Moscoso, H., Raybon, E.O., Thayer, S.G. and H<strong>of</strong>acre, C.L. Molecular detection and serotyping <strong>of</strong> infectious bronchitis virus from FTA® filter paper. Avian Dis.<br />

49:24-29, 2005.<br />

Munday, J.S., Richey, L.J., Brown, C.A., Rodriguez, N.A., and Kiupel, M. Extramedullary plasmacytoma <strong>of</strong> the salivary gland in two Syrian hamsters (Mesocricetus<br />

auratus). Vet. Pathol. 42(6):819-823, 2005.<br />

Murphy, M.D., Howerth, E.W., MacLachlan, N.J. and Stallknecht, D.E. Genetic variation among epizootic hemorrhagic disease viruses in the southeastern United<br />

States: 1978-2001. Infection, Genetics and Evolution 5:157-165, 2005.<br />

Northrup, N.C., Howerth, E.W., Harmon, B.G., Brown, C.A., Carmichael, K.P., Garcia, A.P., Latimer, K.S., Munday, J.S., Rakich, P.M., Richey, L.J., Stedman, N.L., and<br />

Gieger, T.L. Variation among pathologists in the histologic grading <strong>of</strong> canine cutaneous mast cell tumors with uniform use <strong>of</strong> a single grading reference. J. Vet. Diagn.<br />

Invest. 17(6):561-564, 2005.<br />

VMES 2006 29


Okano, S., Hurley, D.J., Bergh, M.S., Vandenplas, M.L., Budsberg, S.C., Moore, J.N. Optimization <strong>of</strong> conditions for in vitro production <strong>of</strong> radical oxygen species and<br />

expression <strong>of</strong> tissue factor by canine mononuclear and granulocytes for use in high-throughput assays. Vet. Immun. and Immunopth. 112:234-242, 2006.<br />

Osuchowski, M.F., Johnson, V.J., He, Q., and Sharma, R.P. Alterations in regional brain neurotransmitters by silymarin, a natural antioxidant flavonoid mixture, in<br />

BALB/c mice. Pharmaceutical Biology 42:384-389, 2004.<br />

Osuchowski, M.F., Edwards, G.L., and Sharma, R.P. Fumonisin B1-induced neurodegeneration in mice after intracerebroventricular infusion is concurrent with disruption<br />

<strong>of</strong> sphingolipid metabolism and activation <strong>of</strong> proinflammatory signaling. NeuroToxicol., 26:211-221, 2005.<br />

Osuchowski, M.F., He, Q. and Sharma, R.P. Endotoxin Exposure Alters Brain and Liver Effects <strong>of</strong> Fumonisin B1 in BALB/c Mice: Implication <strong>of</strong> Blood Brain Barrier.<br />

Food Chem. Toxicol. 43:1389-1397, 2005.<br />

Osuchowski, M.F., Johnson, V.J., He, Q., and Sharma, R. P. Myriocin, a serine palmitoyltransferase inhibitor, alters regional brain neurotransmitter levels without concurrent<br />

inhibition <strong>of</strong> the brain sphingolipid biosynthesis in mice. Toxicol. Lett. 147:87-94, 2004.<br />

Othoro, C., Moore, J.M., Wannemuehler, K., Nahlen, B.L., Otieno, J., Slutsker, L., Lal, A.A., and Shi, Y.P. 2006. Evaluation <strong>of</strong> various methods <strong>of</strong> maternal placental<br />

blood collection for immunology studies. Clin. Vaccine Immunol. 13(5):568-574, 2006.<br />

Overall, K., Rodan, I., Beaver, B., Carney, H., Crowell-Davis, S., Hird, N., Kudrak, S. and Wexler-Mitchell, E. Feline Behavior Guidelines. JAVMA 227(1):70-84,<br />

2005 .<br />

Owen, J.R., Bates, J.N., Lewis, S.J. Differential effects <strong>of</strong> nitroblue tetrazolium on the hemodynamic responses elicited by activation <strong>of</strong> a1-adrenoceptors and 5-HT2<br />

receptors in conscious rats. Eur. J. Pharmacol. 535:248-252, 2006.<br />

Owen, J.R., Bates, J.N., Lewis, S.J. Endogenous nitrosyl factors may inhibit desensitization <strong>of</strong> 5-HT3 receptors on vagal cardiopulmonary afferents. Brain Res.<br />

1059:167-172, 2005.<br />

Parthasarathy, V. and Crowell-Davis, S.L. Relationship between attachment to owners and separation anxiety in pet dogs. J. Vet. Clin. Behavior (in press) 1: 2006.<br />

Peroni, J.F., Moore, J.N., Noschka, E., Grafton, M.E., Aceves-Avila, M., Lewis, S.J., Robertson, T.P. Predisposition for venoconstriction in the equine laminar dermis:<br />

implications in equine laminitis. J. Appl. Physiol. 100:759-763, 2006.<br />

Peroni, J.F., Harrison, W.E., Moore, J.N., Graves, J.E., Lewis, S.J., Krunkosky, T.M.<br />

and Robertson, T.P. Black walnut extract-induced laminitis in horses is associated with heterogeneous dysfunction <strong>of</strong> the laminar microvasculature. Eq. Vet. J. 37:546-<br />

551, 2005.<br />

Perry, N.D., Hanson, B.A., Hobgood, W., Lopez, R.L., Okraska, C.R., Karem, K., Damon, I.K. and Carroll, D.S. New invasive species in southern Florida: Gambian<br />

rat (Cricetomys gambianus). J. Mammalogy 87(2):262-264, 2006.<br />

Peterson, A.T., Pape, M., Reynolds, M.G., Perry, N.D., Hanson, B.A., Regnery, R.L., Hutson, C.L., Muizniek, B., Damon, I.K. and Carroll, D.S. Native-range ecology<br />

and invasive potential <strong>of</strong> cricetomys in North America. J. Mammalogy 87(3):427-432, 2006.<br />

Petkov, D.I., Linnemann, E., Kapczynski, D.R. and Sellers, H.S. Full length sequence analysis <strong>of</strong> four Infectious Bursal Disease Virus isolates with different pathogenicities.<br />

Virus Genes. February, 2006. In Press.<br />

Poovassery J, and Moore, J.M. Murine malaria infection induces fetal loss associated with accumulation <strong>of</strong> Plasmodium chabaudi AS-infected erythrocytes in the<br />

placenta. Infect. Immun. 74(5):2839-2848, 2006.<br />

Posey, J.E., Shinnick, T.M. and Quinn, F.D. Characterization <strong>of</strong> the twin arginine translocase (TAT) secretion system <strong>of</strong> Mycobacterium smegmatis. J. Bacteriol.<br />

188:1332-1340, 2006.<br />

Possas, O., Johnson, A.K., Lewis, S.J. Role <strong>of</strong> nitrosyl factors in the hindlimb vasodilation produced by baroreceptor afferent nerve stimulation. Am. J. Physiol. 290:<br />

R741-R748, 2006.<br />

Praveen, K., Leary, J.H. 3rd, Evans, D.L., Jaso-Friedmann, L. Molecular characterization and expression <strong>of</strong> a granzyme <strong>of</strong> an ectothermic vertebrate with chymase-like<br />

activity expressed in the cytotoxic cells <strong>of</strong> Nile tilapia (Oreochromis niloticus). Immunogenetics 58(1):41-55, 2006.<br />

Praveen, K., Leary, J.H. 3rd, Evans, D.L., Jaso-Friedmann, L. Nonspecific cytotoxic cells <strong>of</strong> teleosts are armed with multiple granzymes and other components <strong>of</strong> the<br />

granule exocytosis pathway. Mol. Immunol. 43(8):1152-1162, 2006.<br />

Praveen, K., Evans, D.L. and Jaso-Friedmann, L. Constitutive expression <strong>of</strong> tumor necrosis factor-alpha in cytotoxic cells <strong>of</strong> teleosts and its role in regulation <strong>of</strong> cellmediated<br />

cytotoxicity. Mol. Immunol. 43:279-291, 2006.<br />

Praveen, K., Evans, D.L., and Jaso-Freidmann, L. Molecular cloning <strong>of</strong> cellular apoptosis susceptibility (CAS) gene in Oreochromis niloticus and its proposed role in<br />

regulation <strong>of</strong> nonspecific cytotoxic cell (NCC) functions. Fish and Shellfish Immunol. 20:647-655, 2006.<br />

Radi, Z.A. and Miller, D.L. Immunohistochemical expression <strong>of</strong> calretinin in canine testicular tumours. Res. Vet. Sci. 79:125-129, 2005.<br />

Rajeev, S., Shulaw, W., Berghaus, R., Zhang, Y., and Byrum, B. A testing scheme for the detection <strong>of</strong> Mycobacterium avium subsp. paratuberculosis in bovine feces<br />

utilizing the ESP para-JEM broth culture system. 2006. Accepted in JVDI<br />

Rayalam, S., Eizenstat, L.D., Hoenig, M., and Ferguson, D.C. Cloning and sequencing <strong>of</strong> feline thyrotropin (fTSH):heterodimeric and yoked constructs. Dom. Anim.<br />

Endocrin. 30:203-217, 2006.<br />

Rayalam, S., Eizenstat, L.D., Davis, R.R., Hoenig, M., Ferguson, D.C. Expression and purification <strong>of</strong> feline thyrotropin (fTSH):immunological detection and bioactivity<br />

<strong>of</strong> heterodimeric and yoked glycoproteins. Dom. Anim. Endocrin. 30:185-202, 2006.<br />

30 www.vet.uga.edu/research/vmes/


Robertson, T.P., Peroni, J.F., Lewis, S.J., and Moore, J.N. Capacitative calcium entry elicits selective venoconstriction in equine laminar blood vessels. Am. J. Vet. Res.<br />

66:1877-1880, 2005.<br />

Saliki, J.T., Cooper, E.J., Rotstein, D.S., Caseltine, S.L., Pabst, D.A., McLellan, W.A., Govett, P., Harms, C., Smolarek, K.A., and Romero, C.H. 2006. A novel gammaherpesvirus<br />

associated with genital lesions in a Blainville’s beaked whale (Mesoplodon densirostris). J. Wildlife Dis., 42: (in press), 2006.<br />

Santangelo, P., Nitin, N., LaConte, L., Woolums, A.R., and Bao, G. Live-cell characterization and analysis <strong>of</strong> a clinical isolate <strong>of</strong> bovine respiratory syncytial virus by<br />

using molecular beacons. J. Virol. 80:682-688, 2006.<br />

Schat, K. and Sellers, H.S. Cell Culture Methods In A Laboratory Manual for the Isolation and Identification <strong>of</strong> Avian Pathogens, 5th edition. Amer. Assoc. Avian<br />

Path. (in press).<br />

Schulz, T.W., Yarbrough, J.Y., and Woldemeskel, M. Toxicity to Tetrahymena and abiotic thiol reactivity <strong>of</strong> aromatic Isothiocyanates. Cell Biol. Toxic. 21:181-189,<br />

2005.<br />

Sebastian, M.M., Bernard, W., and Fitzgerald, T. Mare reproductive loss syndrome. Comp. Cont. Educ. for the Pract. Vet. 1(1):20-33, 2006.<br />

Selleri, P. and Hernandez-Divers, S.J. Renal diseases <strong>of</strong> reptiles. Veterinary Clinics <strong>of</strong> North America Exotic Pet Practice 9:161-174, 2006.<br />

Shaik, S.A., Terrill, T.H., Miller, J.E., Kouakou, B., Kannan, G., Kaplan, R.M., Burke, J. and Mosjidis, J. Use <strong>of</strong> Sericea lespedeza hay as a natural deworming agent in<br />

goats infected with Haemonchus contortus. Vet. Parasit., 139:150–157, 2006.<br />

Shaik, S.A., Terrill, T.H., Miller, J.E., Kouakou, B., Kannan, G., Kaplan, R.M., Burke, J.M. and Mosjidis, J.A. Anthelmintic effects <strong>of</strong> Sericea lespedeza hay fed to<br />

goats infected with Haemonchus contortus. Proceedings <strong>of</strong> the XX International Grassland Congress, Dublin, Ireland, 26 June – 6 July, 2005.<br />

Shanmukh, S., Jones, L., Zhao, Y., Dluhy, R., and Tripp, R.A. Rapid and sensitive detection <strong>of</strong> respiratory virus molecular signatures using an Ag nanorod array SERS<br />

substrate. J. Nanolett., accepted, 2006.<br />

Sharma, N., He. Q., and Sharma, R.P. Augmented fumonisin B1 toxicity in co-cultures:evidence for crosstalk between macrophages and nonparenchymatous liver<br />

epithelial cells involving proinflammatory cytokines. Toxicology, 203:239-251, 2004.<br />

Sharma, N., He. Q., and Sharma, R.P. Sphingosine kinase activity confers resistance to apoptosis by fumonisin B1 in human embryonic kidney (HEK-293) cells.<br />

Chem-Biol. Interactions 151:33-42, 2004.<br />

Sledge, D.G., Miller, D.L., Styer, E.L., Hydrick, H.A. and Baldwin, C.A. Equine herpesvirus 2-associated granulomatous dermatitis in a horse. Vet. Path. (in press)<br />

Spackman, E., Pantin-Jackwood, M., Day, J.M. and Sellers, H. <strong>The</strong> Pathogenesis <strong>of</strong> turkey origin reoviruses in turkeys and chickens. Avian Pathol. 34:291-296, 2005.<br />

Stewart, J.N., Rivera, H.N., Harten, J.B., Karls, R.K., Quinn, F.D., Roman, J. and Rivera-Marrero, C.A. Increased pathology and expression <strong>of</strong> lysosomal cathepsin<br />

proteases in lungs <strong>of</strong> mice after infection with an alpha-crystallin mutant <strong>of</strong> M. tuberculosis. Microbiol. 152:233-244, 2006.<br />

Stiffler, K.S., McCrackin Stevenson, M.A., Sanchez, S., Barsanti, J.A., H<strong>of</strong>meister, E., Budsberg, S.C. Prevalence and charactization <strong>of</strong> urinary tract infections in dogs<br />

with surgically treated type 1 thoracolumbar intervertebral disc extrusion. Vet. Surg .35:330-336, 2006.<br />

Sung, W. and Crowell-Davis, S.L. Elimination behavior patterns <strong>of</strong> domestic cats (Felis catus) with and without elimination behavior problems. Amer. J. Vet. Res. (in<br />

press), 2006.<br />

Swords, W.E., Guenthner, P.C., Birkness, K.A., Lal, R.B., Dezzutti, C.S. and Quinn, F.D. Mycobacterium xenopi multiplies within human macrophages and enhances<br />

HIV replication during co-infection in human PBMC. Microb. Path. 40:41-47, 2006.<br />

Tandon, R., Lyons, E.T., Tolliver, S.C., and Kaplan, R.M. Effect <strong>of</strong> moxidectin selection on the genetic variation within Cylicocyclus nassatus based on amplified fragment<br />

length polymorphism (AFLP). Intern. J. Parasitol. 35(7):813-819, 2005.<br />

Tate, C.M., Mead, D.G., Luttrell, M.P., Howerth, E.W., Dugan, V.G., Munderloh, U.G. and Davidson, W.R. Experimental infection <strong>of</strong> white-tailed deer with Anaplasma<br />

phagocytophilum, etiologic agent <strong>of</strong> human granulocytic anaplasmosis. J. Clin. Microbiol. 43(8):3595-3601, 2005.<br />

Tatsufumi, U., Konnai, S., Ohashi, K.and Onuma, M. Expression <strong>of</strong> tumor necrosis factor-α in IgM+ B-cells from bovine leukemia virus-infected lymphocytotic sheep.<br />

Vet. Immunol. Immunopathol. 112(3-4):296-301, 2006<br />

Thayer, S.G. and Beard, C.W. Serological Procedures. In Isolation and Identification <strong>of</strong> Avian Pathogens. 5th edition, American Assoc. <strong>of</strong> Avian Pathologists. Accepted.<br />

2006.<br />

Thayer, S.G., Waltman, W.D. and Jensen. M.M. Staphylococcus. In Isolation and Identification <strong>of</strong> Avian Pathogens. 5th edition, American Assoc. <strong>of</strong> Avian Pathologists.<br />

Accepted, 2006.<br />

Thayer, S.G., Waltman, W.D. and Wages, D.P. Streptococcus and Enterococcus. In Isolation and Identification <strong>of</strong> Avian Pathogens. 5th edition, American Assoc. <strong>of</strong><br />

Avian Pathologists. Accepted, 2006.<br />

Thompson, A.M., Swant, J., Gosnell, B.A., Wagner, J.J. Modulation <strong>of</strong> long-term potentiation in the rat hippocampus following cocaine self-administration. Neuroscience<br />

127(1):177-185, 2004.<br />

Touzot-Jourde, G., Hernandez-Divers, S.J., and Trim, C.M. Cardiopulmonary effects <strong>of</strong> controlled versus spontaneous ventilation in pigeons anesthetized for coelioscopy.<br />

J. Am. Vet. Med. Assoc. 227:1424-1428, 2005.<br />

Touzot-Jourde, G., Stedman, N.L., and Trim, C.M. Endotracheal intubation in horses: a study <strong>of</strong> 2 cuff inflation pressures, correlation with liquid aspiration and tracheal<br />

wall damage. Vet Anaesth Analg, 32:23-29, 2005.<br />

VMES 2006 31


Tripp, R.A., Alvarez, R., Anderson, B., Jones, L., Weeks, C., and Chen,W. Bioconjugated nanoparticle detection <strong>of</strong> respiratory syncytial virus infection. Int. J. Nanomed,<br />

accepted, 2006.<br />

Tripp, R.A., Oshansky, C., Alvarez, R. Cytokines and respiratory syncytial virus infection. Proc. Am. Thorac. Soc. 2(2):147-149, 2005.<br />

Tripp, R.A., Haynes, L.M., Moore, D., Anderson, B., Tamin, A., Harcourt, B.H., Jones, L.P., Yilla, M., Babcock, G.J., Greenough, T., Ambrosino, D.M., Alvarez, R.,<br />

Callaway, J., Cavitt, S., Kamrud, K., Alterson, H., Smith, J., Harcourt, J.L., Miao, C., Razdan, R., Comer, J.A., Rollin, P.E., Ksiazek, T.G., Sanchez, A., Rota, P.A.,<br />

Bellini, W.J., and Anderson, L.J. Monoclonal antibodies to SARS-associated coronavirus (SARS-CoV): identification <strong>of</strong> neutralizing and antibodies reactive to S, N, M<br />

and E viral proteins. J. Virol. Methods 128(1-2):21-28, 2005.<br />

Tripathy, D.N., and García, M. Laryngotracheitis. In Isolation and identification <strong>of</strong> avian pathogens. D. E. Swayne, J. R. Glisson, M. W. Jackwood, J. E. Pearson, and<br />

W. M. Reed., eds., 5th edition. American Association <strong>of</strong> Avian Pathologist, <strong>University</strong> <strong>of</strong> Pennsylvania, New Bolton <strong>Center</strong>, PA. (in press).<br />

Vaden, S.L. and Brown, C.A. Renal biopsy. In Manual <strong>of</strong> Canine and Feline Nephrology and Urology. Elliot, J. and Grauer, G., eds. Second ed. Brit. Small <strong>Animal</strong><br />

Vet. Assoc. (in press).<br />

Vandenplas, M.L., Moore, J.N., Barton, M.H., Roussel, A.J., and Cohen, N.D. Concentrations <strong>of</strong> serum amyloid A and lipoplysaccharide-binding protein in horses with<br />

colic. Am. J. Vet. Res. 66(9):1509-1516, 2005<br />

van Wyk, J.A., Hoste, H., Kaplan, R.M., Besier, R.B. Targeted selective treatment for worm management - how do we sell rational programs to farmers Vet. Parasitol.<br />

139:269-396, 2006.<br />

Villegas, Pedro. Inclusion Body Hepatitis – Hydropericardium Syndrome. In <strong>The</strong> Merck Veterinary Manual, 9th edition. Pp. 2198-2199, 2005.<br />

Villegas, Pedro. Egg drop Syndrome. In <strong>The</strong> Merck Veterinary Manual, 9th edition. Pp. 2294-2296, 2005.<br />

Visser ’t Ho<strong>of</strong>t, K., Drobatz, K.J., Ward, C.R. Hypophosphatemia: an overview. Comp. Cont. Ed. 27:900-911, 2006.<br />

Wall, M., Platt, S., Selcer, B.A., Brockus, C. Multifocal spinal papillary meningioma in a dog. Vet. Radiol. Ultrasound 46:209-312, 2005<br />

Ward, C.R. Thyroid Storm In Saunder’s Manual <strong>of</strong> Critical Care Medicine for Dogs and Cats, D. Silverstein and K. Hopper, eds, Elsevier, St. Louis, MO., 2006.<br />

Ward, C.R. Hyperparathyroid Disease. Standards <strong>of</strong> Care. 8:8-12, 2006.<br />

Ward, J.P.T., Robertson, T.P., Aaronson, P.I. Capacitative calcium entry: A central role in hypoxic pulmonary vasoconstriction Am. J. Physiol. 289:L2-4, 2005.<br />

Ward, M.R., Stallknecht, D.E., Willis, J., Conroy, M.J. and Davidson, W.R. Wild bird mortality and West Nile virus surveillance: Biases associated with detection,<br />

reporting, and carcass persistence. J. Wildlife Dis. 42(1):92-106, 2006.<br />

Whalen, E.J., Bates, J.N., Johnson, A.K, Lewis, S.J. Down-regulation <strong>of</strong> propranolol-sensitive b-adrenoceptor signaling after inhibition <strong>of</strong> nitric oxide synthesis. Brit. J.<br />

Pharmacol. 147:755-764, 2006.<br />

Whalen, E.J., Saurer, T.B., Johnson, A.K., Lewis, S.J. Intracellular cGMP may promote Ca2+-dependent and Ca2+-independent release <strong>of</strong> catecholamines from sympathetic<br />

nerve terminals. Vasc. Pharmacol., (in press). 2006.<br />

Witte, S., Rodgerson, D.H., Hunt, R.J., Spirito, M., Mueller, P.O.E. Extraluminal uterine adhesions in peripartum mares. Compend. Contin. Educ. Pract. Vet., Equine<br />

Edition, 2006,<br />

Wohlsein, .P and Saliki, J.T. Rinderpest and peste des petits ruminants – the diseases: clinical signs and pathology. In Rinderpest and Peste des Petits Ruminants. Barrett<br />

T, Pastoret P.-P. & Taylor WP, eds. Pp. 68-85. Academic Press New York, 2006.<br />

Evidence <strong>of</strong> tick-borne organisms in mule deer (Odocoi-<br />

Yabsley, M.J., Davidson, W.R., Stallknecht, D.E., Varela, A.S. Swift, ,P.K., deVos, Jr., J.C. and Dubay, S.A.<br />

leus hemionus) from the western United States. Vector-Borne and Zoonotic Dis. 5(4):351-362, 2005.<br />

Yabsley, M.J. and Gibbs, S.E.J. Description and phylogeny <strong>of</strong> a new species <strong>of</strong> Eimeria from double-crested cormorants (Phalacrocorax auritus) near Fort Gaines,<br />

<strong>Georgia</strong>. J. Parasitol. 92:385-388, 2006.<br />

Yabsley, M.J., Quick, T.C. and Little, S.E. <strong>The</strong>ileriosis in a white-tailed deer (Odocoileus virginianus) fawn. J. Wildlife Dis. 41(4):806-809, 2005.<br />

Yabsley, M.J., Romines, J. and Nettles, V.F. Detection <strong>of</strong> Babesia and Anaplasma species in rabbits from Texas and <strong>Georgia</strong>, USA. Vector-Borne and Zoonotic Dis.<br />

6:7-13, 2006.<br />

Yabsley, M.J., Work, T.M. and Rameyer, R.A. Molecular phylogeny <strong>of</strong> Babesia poelea from brown boobies (Sula leucogaster) from Johnston Atoll, Central Pacific. J.<br />

Parasitol. 92:423-5, 2006.<br />

Zarnke, R.L., Saliki, J.T., MacMillan, A.P., Brew, S.D., Dawson, C.E., Ver Hoef, J.M. and Small, R.J. Serologic survey for Brucella spp bacteria, phocid herpesvirus-1,<br />

phocid herpesvirus-2, and phocine distemper virus in harbor seals (Phoca vitulina richardsi) from Alaska. J. Wildlife Dis., 42: (in press). 2006.<br />

Zavala, G. Rétroviroses de la poule/leucoses aviaires. In , 2nd édition. Ecole nationale vétérinaire d’Alfort, Cedex, France.<br />

Zavala, G., Lucio-Martinez, B., Cheng, S. and Barbosa, T. Sarcomas and myelocytomas induced by avian leukosis virus in white Leghorn egg layer chickens. Avian<br />

Dis. (in press). 2006.<br />

Zavala, G. and Cheng, S. Detection and characterization <strong>of</strong> avian leukosis virus in Marek’s disease vaccines. Avian Dis. (in press), 2006.<br />

Zavala, G. and Cheng, S. Experimental infection with avian leukosis virus isolated from Marek’s disease vaccines. Avian Dis. (in press), 2006.<br />

32 www.vet.uga.edu/research/vmes/


Working for <strong>Georgia</strong><br />

Cover Illustrations and Lead Articles<br />

2002-2005<br />

Food <strong>Animal</strong> <strong>Health</strong> and<br />

Management Program<br />

2002<br />

Agroterrorism<br />

2003<br />

Vaccinology<br />

2004<br />

RNA interference<br />

2005


Veterinary Medical Experiment Station<br />

<strong>College</strong> <strong>of</strong> Veterinary Medicine<br />

<strong>The</strong> <strong>University</strong> <strong>of</strong> <strong>Georgia</strong><br />

Athens, <strong>Georgia</strong> 30602

Hooray! Your file is uploaded and ready to be published.

Saved successfully!

Ooh no, something went wrong!