Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 38
Filter
Add more filters










Publication year range
1.
J Wildl Dis ; 60(2): 362-374, 2024 Apr 01.
Article in English | MEDLINE | ID: mdl-38345467

ABSTRACT

Mass mortality events in wildlife can be indications of an emerging infectious disease. During the spring and summer of 2021, hundreds of dead passerines were reported across the eastern US. Birds exhibited a range of clinical signs including swollen conjunctiva, ocular discharge, ataxia, and nystagmus. As part of the diagnostic investigation, high-throughput metagenomic next-generation sequencing was performed across three molecular laboratories on samples from affected birds. Many potentially pathogenic microbes were detected, with bacteria forming the largest proportion; however, no singular agent was consistently identified, with many of the detected microbes also found in unaffected (control) birds and thus considered to be subclinical infections. Congruent results across laboratories have helped drive further investigation into alternative causes, including environmental contaminants and nutritional deficiencies. This work highlights the utility of metagenomic approaches in investigations of emerging diseases and provides a framework for future wildlife mortality events.


Subject(s)
Communicable Diseases, Emerging , Songbirds , Animals , Animals, Wild , Metagenome , Bacteria/genetics , Communicable Diseases, Emerging/veterinary , Metagenomics/methods
2.
Article in English | MEDLINE | ID: mdl-38415687

ABSTRACT

A novel species of the genus Moraxella was isolated from an ocular swab from a cow with infectious bovine keratoconjunctivitis. 16S rRNA gene sequencing suggested this species was Moraxella bovis (99.59 % nucleotide identity). Average nucleotide identity was calculated using a draft whole genome sequence of this strain compared with type strains of closely related Moraxella species and results established that it represents a new species. The genome size was 2 006 474 nucleotides and the G+C content was 42.51 mol%. The species could not be identified by matrix assisted laser desorption/ionization-time of flight mass spectrometry using a commercial database, confirming the novelty of the strain. We propose the name Moraxella oculi sp. nov. for this new species. The type strain is Tifton1T and has been deposited into the American Type Culture Collection (TSD-373T) and the National Collection of Type Cultures (NCTC), UK Health Security Agency (NCTC 14942T).


Subject(s)
Cattle Diseases , Keratoconjunctivitis, Infectious , Keratoconjunctivitis , Moraxellaceae Infections , Cattle , Animals , Moraxella/genetics , RNA, Ribosomal, 16S/genetics , Phylogeny , DNA, Bacterial/genetics , Bacterial Typing Techniques , Base Composition , Sequence Analysis, DNA , Fatty Acids/chemistry , Moraxellaceae Infections/veterinary , Nucleotides
3.
J Vet Diagn Invest ; 36(1): 120-123, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38018659

ABSTRACT

Infectious bovine keratoconjunctivitis (IBK) is associated with 2 species of Moraxella: M. bovis and M. bovoculi. A third novel Moraxella spp., designated tentatively as M. oculi, has been identified from the eyes of cattle with and without pinkeye. These 3 Moraxella spp. can be found in various combinations within the same clinical sample, making speciation of this genus directly from a sample impossible with Sanger sequencing. Assessing Moraxella diversity found in IBK- and non-IBK-affected cattle eyes, independent of culture, may provide additional information about IBK by avoiding the selectivity bias of culturing. We developed a targeted NGS panel to detect and speciate these 3 Moraxella spp. directly from bovine ocular swabs. Our targeted panel amplifies bacterial essential genes and the 16S-23S ribosomal RNA intergenic spacer region (ITS) of the 3 Moraxella spp. and speciates based on these sequences. Our panel was able to differentiate the 3 species directly from DNA extracted from 13 swabs (6 from healthy animals, 7 from animals with IBK), and every swab except one (clinically healthy eye) had the 3 Moraxella spp. Targeted NGS with sequencing of Moraxella spp. housekeeping genes appears to be a suitable method for speciation of Moraxella directly from ocular swabs.


Subject(s)
Cattle Diseases , Keratoconjunctivitis, Infectious , Moraxellaceae Infections , Mycoplasma Infections , Cattle , Animals , Moraxella/genetics , Keratoconjunctivitis, Infectious/diagnosis , Keratoconjunctivitis, Infectious/microbiology , Mycoplasma Infections/veterinary , Moraxellaceae Infections/diagnosis , Moraxellaceae Infections/veterinary , Moraxellaceae Infections/microbiology , Cattle Diseases/diagnosis , Cattle Diseases/microbiology , High-Throughput Nucleotide Sequencing/veterinary
4.
J Vet Intern Med ; 37(5): 1889-1892, 2023.
Article in English | MEDLINE | ID: mdl-37515307

ABSTRACT

BACKGROUND: Equine granulocytic anaplasmosis (EGA) is a common disease in adult horses, but clinical disease in foals is rarely reported. The relationship between equine maternal and neonatal antibodies to Anaplasma phagocytophilum is unclear. HYPOTHESIS/OBJECTIVES: That mares in an endemic region would be seropositive for A. phagocytophilum and that mare and foal serum IgG concentrations for A. phagocytophilum would correlate. Additionally, we hypothesized that foal IgG concentrations for A. phagocytophilum acquired by passive immunity would decline by 6 months of age. ANIMALS: Twenty-two healthy mare-foal pairs. METHODS: This prospective observational study investigated serum IgG concentrations specific for A. phagocytophilum in mares and foals using an immunofluorescent antibody test (IFA). The association between foal titer (as a binary variable) and age in months was assessed using a mixed-effects logistic regression. RESULTS: A positive correlation between newborn foal antibody titers and mare titers was identified at both the pre-foaling (τa = 0.38, τb = 0.50, P = .009) and foaling timepoints (τa = 0.36, τb = 0.47, P = .01). In A. phagocytophilum seropositive neonates, it was unlikely that a positive titer would be detected by 3 months of age (OR = 0.002, P = .02, 95% CI: 0.00001-0.38). Three out of 20 foals seroconverted between 3 and 6 months of age. CONCLUSIONS AND CLINICAL IMPORTANCE: Transfer of specific passive immunity to A. phagocytophilum occurred in 80% of foals born to seropositive mares and declined by 3 months of age. A. phagocytophilum infection should be considered in foals displaying clinical signs consistent with EGA.


Subject(s)
Anaplasma phagocytophilum , Horse Diseases , Animals , Horses , Female , Maryland , Pennsylvania , Immunity, Maternally-Acquired , Immunoglobulin G , Animals, Newborn
5.
Vet Microbiol ; 282: 109752, 2023 Jul.
Article in English | MEDLINE | ID: mdl-37104939

ABSTRACT

Infectious bovine keratoconjunctivitis (IBK), commonly known as pinkeye, has a marked negative impact on the economy of the cattle industry. Moraxella species, including Mor. bovis and Mor. bovoculi, which have been associated with this disease, colonize clinically healthy eyes as well, suggesting that there are intrinsic changes that may occur to the ocular microbiota or the involvement of additional unrecognized organisms that contribute to IBK. To evaluate this, 104 ocular swabs collected from eyes with IBK or clinically healthy eyes from 16 different cattle herds were subjected to 16 S rRNA gene PCR and next generation sequencing (NGS) analysis. Organisms detected were similar across the herds and there was no difference in the total number of bacterial groups detected among IBK cases and controls. However, the percentages of the different organisms detected varied between the two groups, including Moraxella spp., with more Moraxella spp. in eyes with IBK than controls. Further, using culture and whole genome NGS, a new species of Moraxella (suggested name Mor. oculobovii) was detected from the eyes of cattle from two farms. This strain is non-hemolytic on blood agar, is missing the RTX operon, and is likely a non-pathogenic strain of the bovine ocular microbiome. Alteration of the ocular microbiota composition may have a predisposing role, enhancing bacterial infection and the occurrence of clinical IBK. Future studies are required to evaluate if these changes are permanent or if there is a shift in the microbiome following recovery from the infection and how antibiotics might affect the microbiome.


Subject(s)
Cattle Diseases , Conjunctivitis, Bacterial , Keratoconjunctivitis, Infectious , Keratoconjunctivitis , Moraxellaceae Infections , Mycoplasma Infections , Animals , Cattle , High-Throughput Nucleotide Sequencing/veterinary , Keratoconjunctivitis, Infectious/epidemiology , Keratoconjunctivitis, Infectious/microbiology , Keratoconjunctivitis/epidemiology , Keratoconjunctivitis/veterinary , Keratoconjunctivitis/microbiology , Conjunctivitis, Bacterial/microbiology , Conjunctivitis, Bacterial/veterinary , Moraxella/genetics , Mycoplasma Infections/veterinary , Moraxellaceae Infections/epidemiology , Moraxellaceae Infections/veterinary , Moraxellaceae Infections/microbiology , Cattle Diseases/epidemiology , Cattle Diseases/microbiology
6.
J Zoo Wildl Med ; 53(4): 811-816, 2023 Jan.
Article in English | MEDLINE | ID: mdl-36640084

ABSTRACT

Bat coronaviruses (CoVs) are extremely prevalent throughout the globe and exhibit a wide range of genetic diversity. Currently, little is known about the susceptibility of New World bats to severe acute respiratory syndrome-2 (SARS-CoV-2), the causative agent of COVID-19. Also, there is limited information about the genetic diversity of other CoVs in the New World bats. The determination of genetic diversity of bat CoVs through continuous surveillance is essential to predict and mitigate the emergence of new CoVs and their impacts on the health of both humans and animals. In this study, 491 guano specimens collected from New World bats and 37 specimens collected from Old World bats during July 2020 to July 2021 were tested for SARS-COV-2 and other CoVs using a real-time reverse transcriptase-polymerase chain reaction (RT-PCR) panel and pan-coronavirus PCR that target a highly conserved region of CoVs. No evidence of SARS-CoV-2 was found in the tested specimens. An alpha CoV was detected in a single specimen from a big brown bat (Eptesicus fuscus). This information was used by wildlife agencies and rehabilitation facilities to permit the release of bats during the pandemic while mitigating the risk of spreading SARS-CoV-2 among North American bats and other wild animal populations.


Subject(s)
COVID-19 , Chiroptera , Animals , United States/epidemiology , Humans , SARS-CoV-2/genetics , COVID-19/epidemiology , COVID-19/veterinary , Phylogeny , Genome, Viral , Animals, Wild
7.
mBio ; 13(5): e0210122, 2022 10 26.
Article in English | MEDLINE | ID: mdl-36000731

ABSTRACT

The SARS-CoV-2 pandemic began by viral spillover from animals to humans; today multiple animal species are known to be susceptible to infection. White-tailed deer, Odocoileus virginianus, are infected in North America at substantial levels, and genomic data suggests that a variant in deer may have spilled back to humans. Here, we characterize SARS-CoV-2 in deer from Pennsylvania (PA) sampled during fall and winter 2021. Of 123 nasal swab samples analyzed by RT-qPCR, 20 (16.3%) were positive for SARS-CoV-2. Seven whole genome sequences were obtained, together with six more partial spike gene sequences. These annotated as alpha and delta variants, the first reported observations of these lineages in deer, documenting multiple new jumps from humans to deer. The alpha lineage persisted in deer after its displacement by delta in humans, and deer-derived alpha variants diverged significantly from those in humans, consistent with a distinctive evolutionary trajectory in deer. IMPORTANCE Coronaviruses have been documented to replicate in numerous species of vertebrates, and multiple spillovers of coronaviruses from animals into humans have founded human epidemics. The COVID-19 epidemic likely derived from a spillover of SARS-CoV-2 from bats into humans, possibly via an intermediate host. There are now several examples of SARS-CoV-2 jumping from humans into other mammals, including mink and deer, creating the potential for new animal reservoirs from which spillback into humans could occur. For this reason, data on formation of new animal reservoirs is of great importance for understanding possible sources of future infection. Here, we identify extensive infection in white-tailed deer in Pennsylvania, including what appear to be multiple independent transmissions. Data further suggests possible transmission among deer. These data thus help identify a potential new animal reservoir and provide background information relevant to its management.


Subject(s)
COVID-19 , Deer , Animals , Humans , SARS-CoV-2/genetics , Pennsylvania/epidemiology , COVID-19/epidemiology , COVID-19/veterinary
8.
Immunohorizons ; 6(5): 299-306, 2022 05 20.
Article in English | MEDLINE | ID: mdl-35595326

ABSTRACT

RBCs demonstrate immunomodulatory capabilities through the expression of nucleic acid sensors. However, little is known about bat RBCs, and no studies have examined the immune function of bat erythrocytes. In this study, we show that bat RBCs express the nucleic acid-sensing TLRs TLR7 and TLR9 and bind the nucleic acid ligands, ssRNA, and CpG DNA. Collectively, these data suggest that, like human RBCs, bat erythrocytes possess immune function and may be reservoirs for nucleic acids. These findings provide unique insight into bat immunity and may uncover potential mechanisms by which virulent pathogens of humans are concealed in bats.


Subject(s)
Chiroptera , Nucleic Acids , Animals , Chiroptera/genetics , DNA , Erythrocytes , Humans , RNA
9.
Animals (Basel) ; 11(11)2021 Nov 09.
Article in English | MEDLINE | ID: mdl-34827935

ABSTRACT

Infectious laryngotracheitis (ILT) is a viral disease of chickens' respiratory system that imposes considerable financial burdens on the chicken industry. Rapid, simple, and specific detection of this virus is crucial to enable proper control measures. Polymerase chain reaction (PCR)-based molecular tests require relatively expensive instruments and skilled personnel, confining their application to centralized laboratories. To enable chicken farms to take timely action and contain the spread of infection, we describe a rapid, simple, semi-quantitative benchtop isothermal amplification (LAMP) assay, and a field-deployable microfluidic device for the diagnosis of ILTV infection in chickens. Our assay performance was compared and favorably agreed with quantitative PCR (qPCR). The sensitivity of our real-time LAMP test is 250 genomic copies/reaction. Clinical performance of our microfluidic device using samples from diseased chickens showed 100% specificity and 100% sensitivity in comparison with benchtop LAMP assay and the gold-standard qPCR. Our method facilitates simple, specific, and rapid molecular ILTV detection in low-resource veterinary diagnostic laboratories and can be used for field molecular diagnosis of suspected ILT cases.

10.
Vet Med Sci ; 7(5): 1536-1541, 2021 09.
Article in English | MEDLINE | ID: mdl-34216115

ABSTRACT

Clostridioides difficile is an important enteric pathogen that causes significant morbidity and mortality in humans. With community-acquired infections on the rise, it is important to identify reservoirs of the pathogen. Companion animals can be asymptomatic carriers of C. difficile and may therefore represent a reservoir, but epidemiological studies of C. difficile within the pet-owner unit are needed, along with validated methods to detect C. difficile in both people and animals. The goal of this study was to assess the performance of commercial qPCR assays and a multiplex PCR for C. difficile compared to toxigenic culture. These assays were tested on up to 103 fecal samples from puppies, a population in which the prevalence of C. difficile is the highest. The sensitivities, specificities, positive predictive values and negative predictive values were respectively 84.2%, 87.7%, 61.5%, and 95.9% for the Cepheid GeneXpert; 66.7%, 66.7%, 29.6%, and 90.9% for the DiaSorin Simplexa; and 94.4%, 85.0%, 65.4%, and 98.1%, for the multiplex qPCR. The agreement was highest between the GeneXpert and the multiplex PCR (90.1% agreement, with a kappa statistic of 0.77). For diagnostic purposes, the positive predictive values of the assays were low. However, the high sensitivities of the assays could render them useful for epidemiologic purposes.


Subject(s)
Bacterial Toxins , Clostridioides difficile , Animals , Bacterial Toxins/analysis , Bacterial Toxins/genetics , Clostridioides , Clostridioides difficile/genetics , Dogs , Feces/chemistry , Multiplex Polymerase Chain Reaction/veterinary , Sensitivity and Specificity
11.
PLoS One ; 16(6): e0252804, 2021.
Article in English | MEDLINE | ID: mdl-34125848

ABSTRACT

Streptococcus equi subsp. equi (SEE) is a host-restricted bacterium that causes the common infectious upper respiratory disease known as strangles in horses. Perpetuation of SEE infection appears attributable to inapparent carrier horses because it neither persists long-term in the environment nor infects other host mammals or vectors, and infection results in short-lived immunity. Whether pathogen factors enable SEE to remain in horses without causing clinical signs remains poorly understood. Thus, our objective was to use next-generation sequencing technologies to characterize the genome, methylome, and transcriptome of isolates of SEE from horses with acute clinical strangles and inapparent carrier horses-including isolates recovered from individual horses sampled repeatedly-to assess pathogen-associated changes that might reflect specific adaptions of SEE to the host that contribute to inapparent carriage. The accessory genome elements and methylome of SEE isolates from Sweden and Pennsylvania revealed no significant or consistent differences between acute clinical and inapparent carrier isolates of SEE. RNA sequencing of SEE isolates from Pennsylvania demonstrated no genes that were differentially expressed between acute clinical and inapparent carrier isolates of SEE. The absence of specific, consistent changes in the accessory genomes, methylomes, and transcriptomes of acute clinical and inapparent carrier isolates of SEE indicates that adaptations of SEE to the host are unlikely to explain the carrier state of SEE. Efforts to understand the carrier state of SEE should instead focus on host factors.


Subject(s)
Carrier State/diagnosis , Epigenome/genetics , Genome/genetics , Horse Diseases/diagnosis , Streptococcus/genetics , Transcriptome/genetics , Animals , Carrier State/microbiology , DNA, Bacterial/analysis , DNA, Bacterial/genetics , DNA, Bacterial/isolation & purification , Diagnosis, Differential , Disease Outbreaks , Horse Diseases/epidemiology , Horse Diseases/microbiology , Horses , Pennsylvania/epidemiology , RNA, Bacterial/analysis , RNA, Bacterial/genetics , RNA, Bacterial/isolation & purification , RNA-Seq/methods , Species Specificity , Streptococcus/classification , Streptococcus/physiology , Sweden/epidemiology , Whole Genome Sequencing/methods
12.
J Vet Diagn Invest ; 33(2): 331-335, 2021 Mar.
Article in English | MEDLINE | ID: mdl-33522461

ABSTRACT

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is the causative agent of coronavirus disease 2019 (COVID-19), which is an ongoing global health concern. The exact source of the virus has not been identified, but it is believed that this novel coronavirus originated in animals; bats in particular have been implicated as the primary reservoir of the virus. SARS-CoV-2 can also be transmitted from humans to other animals, including tigers, cats, and mink. Consequently, infected people who work directly with bats could transfer the virus to a wild North American bat, resulting in a new natural reservoir for the virus, and lead to new outbreaks of human disease. We evaluated a reverse-transcription real-time PCR panel for detection of SARS-CoV-2 in bat guano. We found the panel to be highly specific for SARS-CoV-2, and able to detect the virus in bat guano samples spiked with SARS-CoV-2 viral RNA. Our panel could be utilized by wildlife agencies to test bats in rehabilitation facilities prior to their release to the wild, minimizing the risk of spreading this virus to wild bat populations.


Subject(s)
COVID-19/transmission , Chiroptera/virology , SARS-CoV-2/isolation & purification , Animals , Animals, Wild , Feces/virology , Humans , Real-Time Polymerase Chain Reaction/veterinary , Sensitivity and Specificity , Zoonoses
13.
Analyst ; 146(4): 1311-1319, 2021 Feb 21.
Article in English | MEDLINE | ID: mdl-33367323

ABSTRACT

Sensitive, specific and rapid molecular diagnosis of respiratory diseases in animals and humans is critical to facilitate appropriate control measures and treatment. Conventional polymerase chain reaction (PCR)-based molecular diagnostics requires relatively expensive equipment and trained staff, restricting its use to centralized laboratories with significant delays between sample collection and test results. Herein, we report a highly sensitive, rapid, point-of-need, two-stage-molecular test that requires minimal instrumentation and training. Our test, dubbed Penn-RAMP, combines recombinase polymerase amplification (RPA, 38 °C) and loop-mediated isothermal amplification (LAMP, 63 °C) in one tube, enabling nested, two-stage isothermal amplification. We demonstrate Penn-RAMP's efficacy by testing for two common viral respiratory diseases of chickens: infectious laryngotracheitis (ILT) and infectious bronchitis (IB) that impose great economic burden worldwide. Test results of clinical samples with our closed-tube Penn-RAMP assays concord with the gold standard quantitative PCR (qPCR) assay; with 10-fold better limit of detection than LAMP and qPCR. Our closed-tube Penn-RAMP assays have the potential to greatly reduce false negatives while requiring minimal instrumentation and training.


Subject(s)
Chickens , Nucleic Acid Amplification Techniques , Animals , Humans , Molecular Diagnostic Techniques , Sensitivity and Specificity
14.
J Vet Diagn Invest ; 33(2): 227-234, 2021 Mar.
Article in English | MEDLINE | ID: mdl-33305693

ABSTRACT

Equine infectious disease outbreaks may have profound economic impact, resulting in losses of millions of dollars of revenue as a result of horse loss, quarantine, and cancelled events. Early and accurate diagnosis is essential to limit the spread of infectious diseases. However, laboratory detection of infectious agents, especially the simultaneous detection of multiple agents, can be challenging to the clinician and diagnostic laboratory. Next-generation sequencing (NGS), which allows millions of DNA templates to be sequenced simultaneously in a single reaction, is an ideal technology for comprehensive testing. We conducted a proof-of-concept study of targeted NGS to detect 62 common equine bacterial, viral, and parasitic pathogens in clinical samples. We designed 264 primers and constructed a bioinformatics tool for the detection of targeted pathogens. The designed primers were able to specifically detect the intended pathogens. Results of testing 27 clinical samples with our targeted NGS assay compared with results of routine tests (assessed as a group) yielded positive percent agreement of 81% and negative percent agreement of 83%, overall agreement of 81%, and kappa of 0.56 (moderate agreement). This moderate agreement was likely the result of low sensitivity of some primers. However, our NGS assay successfully detected multiple pathogens in the clinical samples, including some pathogens missed by routine techniques.


Subject(s)
High-Throughput Nucleotide Sequencing/veterinary , Horse Diseases/diagnosis , Animals , High-Throughput Nucleotide Sequencing/methods , Horse Diseases/microbiology , Horse Diseases/parasitology , Horse Diseases/virology , Horses
15.
Pathogens ; 9(7)2020 Jul 21.
Article in English | MEDLINE | ID: mdl-32708148

ABSTRACT

One free-ranging Gray fox (Urocyon cinereoargenteus) underwent autopsy following neurologic disease, with findings including morbilliviral inclusions and associated lesions in numerous tissues, adenoviral intranuclear inclusions in bronchial epithelial cells, and septic pleuropneumonia, hepatitis, splenitis, and meningoencephalitis. Molecular diagnostics on fresh lung identified a strain within a distinct clade of canine distemper that is currently unique to wildlife in New England, as well as the emerging multi-host viral pathogen skunk adenovirus-1. Bacterial culture of fresh liver resulted in a pure growth of Listeria monocytogenes, with whole genome sequencing indicating that the isolate had a vast array of antimicrobial resistance and virulence-associated genes. One year later, a second fox was euthanized for inappropriate behavior in a residential area, and diagnostic workup revealed canine distemper and septic L. monocytogenes, with the former closely related to the distemper virus found in the previous fox and the latter divergent from the L. monocytogenes from the previous fox.

16.
J Virol Methods ; 284: 113940, 2020 10.
Article in English | MEDLINE | ID: mdl-32687868

ABSTRACT

Infectious bronchitis (IB) is a viral infection of the chicken respiratory tract that causes substantial economic burden on the industry. Simple, specific and rapid diagnosis of this disease is critical for the initiation of appropriate control measures. Conventional molecular diagnostic methods require a relatively sophisticated equipment and skilled staff. Here we describe a rapid, simple, semi-quantative, closed-tube, single-step, real-time- reverse transcription-loop-mediated isothermal amplification (RT-LAMP) assay for IB and compare our assay with quantative, reverse transcription- polymerase chain reaction (RT-qPCR). The limit of detection (LOD) of our RT-LAMP assay is 1 EID50/ ml. Clinical evaluation of samples from diseased chickens with our RT-LAMP showed a very good concordance with RT-qPCR. Our assay enables simple, specific, rapid molecular detection and semi-quantification of the infectious bronchitis virus (IBV) in veterinary diagnostic laboratories. Furthermore, our RT-LAMP detection is carried out in a sealed tube, eliminating the risk of false-positive results in subsequent tests because of any contamination of the work area as in the case of lateral flow strip or gel electrophoresis-based amplicon detection.


Subject(s)
Coronavirus Infections/veterinary , Infectious bronchitis virus/isolation & purification , Molecular Diagnostic Techniques/veterinary , Nucleic Acid Amplification Techniques/veterinary , Poultry Diseases/diagnosis , Animals , Chickens , Coronavirus Infections/diagnosis , Limit of Detection , RNA, Viral/genetics , Reverse Transcription , Sensitivity and Specificity
17.
Vet Pathol ; 57(2): 311-315, 2020 03.
Article in English | MEDLINE | ID: mdl-32079498

ABSTRACT

An outbreak of canine distemper virus in a private zoo in eastern Tennessee in July 2016 led to fatal clinical disease in 5 adult, wild-caught Linnaeus's 2-toed sloths (Choloepus didactylus). Clinical signs included hyporexia, lethargy, mucopurulent nasal discharge, and oral and facial ulcers. At necropsy, affected animals had crusts and ulcers on the lips, nose, tongue, and oral cavity. Microscopically, all sloths had widespread, random, hepatic necrosis; lymphoid depletion; and bronchointerstitial pneumonia. The central nervous system did not contain gross or histopathologic lesions in any of the 5 sloths, although immunoreactivity for viral antigen was present within vessel walls. Epithelial cells and histiocytes within numerous organs contained intranuclear and intracytoplasmic inclusions and occasional syncytial cells. Canine distemper virus was confirmed with immunohistochemistry and virus isolation. Viral sequencing identified the novel American-4 strain prevalent in eastern Tennessee wildlife. This is the first pathologic characterization of canine distemper virus infection in sloths (family Choloepodidae, order Pilosa) and emphasizes the significant morbidity and mortality in this species.


Subject(s)
Disease Outbreaks/veterinary , Distemper Virus, Canine/isolation & purification , Distemper/diagnosis , Sloths/virology , Animals , Animals, Zoo , Distemper/pathology , Distemper/virology , Distemper Virus, Canine/immunology , Epithelial Cells/pathology , Epithelial Cells/virology , Female , Immunohistochemistry/veterinary , Inclusion Bodies, Viral/pathology , Liver/pathology , Liver/virology , Male , Tongue/pathology , Tongue/virology
18.
J Zoo Wildl Med ; 50(4): 790-797, 2020 Jan 09.
Article in English | MEDLINE | ID: mdl-31926508

ABSTRACT

Canine distemper virus (CDV) is a highly contagious disease of wild and domestic mammals. Maintenance of CDV among wildlife plays an important role in the disease epidemiology. Wild animals, including raccoons (Procyon lotor) and gray foxes (Urocyon cinereoargenteus), serve as reservoirs of CDV and hamper the control of the disease. Recently, we discovered that at least three different CDV lineages (America-3 [Edomex], America-4, and America-5] that are genetically different from the available vaccine strains are circulating in domestic dogs in the United States. Because wildlife serve as a reservoir for the virus, it is important to determine if wildlife play a role in the maintenance and spread of these lineages. To determine the genetic characteristics of circulating strains of CDV in wildlife in various geographic regions in the United States, we studied the nucleotide sequences of the hemagglutinin (H) gene of 25 CDV strains detected in nondomestic species. The species included were free-ranging wildlife: three fishers (Martes pennanti), six foxes, one skunk (Mephitis mephitis), 10 raccoons, two wolves (Canis lupus), and one mink (Neovison vison). Strains from two species in managed care, one sloth (Choloepus didactylus) and one red panda (Ailurus fulgens), were also evaluated. Phylogenetic analysis of the H genes indicated that in addition to America-3, America-4, and America-5 lineages, there are at least two other lineages circulating in US wildlife. One of these includes CDV nucleotide sequences that grouped with that of a single CDV isolate previously detected in a raccoon from Rhode Island in 2012. The other lineage is independent and genetically distinct from other CDV strains included in the analysis. Additional genetically variable strains were detected, mainly in raccoons, suggesting that this species may be the host responsible for the genetic variability of newly detected strains in the domestic dog population.


Subject(s)
Animals, Wild/virology , Carnivora/virology , Distemper Virus, Canine/genetics , Distemper/virology , Animals , Distemper/epidemiology , Phylogeny , United States/epidemiology
19.
J Zoo Wildl Med ; 50(4): 798-802, 2020 Jan 09.
Article in English | MEDLINE | ID: mdl-31926509

ABSTRACT

Two methods for delivering a canarypox-vectored canine distemper vaccine to tigers (Panthera tigris) and domestic cats (Felis catus) were investigated. Eight tigers were divided randomly into two vaccination groups: subcutaneous injection or topical tonsillar application. Each tiger received 2 ml of canine distemper virus (CDV) vaccine (Merial Ferret Distemper Vaccine). Blood was collected from tigers on days 0, 21, 35 or 37, and 112 post-initial vaccination (PIV). Domestic cats were divided randomly into four treatment groups: saline injection (negative controls), low- and high-dose oral, and subcutaneous vaccinates. Blood was collected from domestic cats on days 0, 7, 21, and 28 and 165 or 208 PIV. Sera were tested for CDV antibodies by virus neutralization. All individuals were seronegative at the beginning of the study. One tiger vaccinated subcutaneously developed a titer of 32 by day 35, which reduced to 16 by day 112. Another tiger vaccinated by tonsillar application developed a titer of 8 on day 112. All other tigers remained seronegative. Cats that received saline injection or oral vaccination remained seronegative at each sampling time. Domestic cats vaccinated subcutaneously developed titers ranging from 4 to >128 by day 28, and those re-bled at day 166 had titers of 16 or 64. The disparity in response between domestic cats and tigers may be due to species differences or it may represent a dose-dependent effect. Subcutaneous vaccination with canarypox-vectored Purevax Ferret Distemper® is safe and elicits persistent antibody titers in domestic cats vaccinated parenterally.


Subject(s)
Canarypox virus , Cat Diseases/prevention & control , Distemper Virus, Canine/immunology , Distemper/prevention & control , Tigers/immunology , Viral Vaccines/immunology , Administration, Oral , Animals , Antibodies, Viral/blood , Cat Diseases/immunology , Cat Diseases/virology , Cats , Female , Male , Vaccines, Synthetic
20.
J Zoo Wildl Med ; 50(4): 972-975, 2020 Jan 09.
Article in English | MEDLINE | ID: mdl-31926530

ABSTRACT

This study evaluated the safety of and humoral response to the Merial Recombitek® recombinant canine distemper virus (rCDV) vaccine in maned wolves (n = 9, age 2-9 yr). All maned wolves had prior history of annual vaccination with the Merial Purevax® ferret rCDV vaccine. Serum neutralization (SN) to CDV was measured prior to initial vaccination with the rCDV Recombitek vaccine followed by a booster vaccination at 4-6 wk. Final SN titers were obtained at 13 wk post initial vaccination. The maned wolves developed no observable adverse side effects through the study. Pre-Recombitek vaccination SN titers ranged from negative to 1: 8. Postvaccination CDV titers ranged from negative to 1: 8, and were therefore below the range of that considered protective in domestic dogs.


Subject(s)
Antibodies, Viral/blood , Canidae , Distemper Virus, Canine/immunology , Distemper/prevention & control , Viral Vaccines/immunology , Animals , Female , Male , Viral Vaccines/adverse effects
SELECTION OF CITATIONS
SEARCH DETAIL
...