Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 37
Filtrar
1.
Arch Virol ; 167(8): 1659-1668, 2022 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-35708765

RESUMO

Bovine gammaherpesvirus 4 (BoHV-4) is ubiquitous in cattle worldwide, and it has been detected in animals exhibiting broad clinical presentations. The virus has been detected in the United States since the 1970s; however, its clinical relevance remains unknown. Here, we determined the complete genome sequences of two contemporary BoHV-4 isolates obtained from respiratory (SD16-38) or reproductive (SD16-49) tract specimens and assessed clinical, virological, and pathological outcomes upon intranasal (IN) inoculation of calves with the respiratory BoHV-4 isolate SD16-38. A slight and transient increase in body temperature was observed in BoHV-4-inoculated calves. Additionally, transient viremia and virus shedding in nasal secretions were observed in all inoculated calves. BoHV-4 DNA was detected by nested PCR in the tonsil and regional lymph nodes (LNs) of calves euthanized on day 5 post-inoculation (pi) and in the lungs of calves euthanized on day 10 pi. Calves euthanized on day 35 pi harbored BoHV-4 DNA in the respiratory tract (turbinates, trachea, lungs), regional lymphoid tissues, and trigeminal ganglia. Interestingly, in situ hybridization revealed the presence of BoHV-4 DNA in nerve bundles surrounding the trigeminal ganglia and retropharyngeal lymph nodes (day 35 pi). No histological changes were observed in the respiratory tract (turbinate, trachea, and lung), lymphoid tissues (tonsil, LNs, thymus, and spleen), or central nervous tissues (olfactory bulb and trigeminal ganglia) sampled throughout the animal studies (days 5, 10, and 35 pi). This study contributes to the understanding of the infection dynamics and tissue distribution of BoHV-4 following IN infection in calves. These results suggest that BoHV-4 SD16-38 used in our study has low pathogenicity in calves upon intranasal inoculation.


Assuntos
Doenças dos Bovinos , Infecções por Herpesviridae , Herpesvirus Bovino 1 , Herpesvirus Bovino 4 , Animais , Anticorpos Antivirais , Bovinos , Infecções por Herpesviridae/veterinária , Herpesvirus Bovino 4/genética , Eliminação de Partículas Virais
2.
Arch Virol ; 166(10): 2835-2839, 2021 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-34319454

RESUMO

The bovine adenovirus 7 (BAdV-7) isolate SD18-74 was recovered from lung tissue of calves in South Dakota. The 30,043-nucleotide (nt) genome has the typical organization of Atadenovirus genus members. The sequence shares over 99% nt sequence identity with two Japanese BAdV-7 sequences, followed by 74.9% nt sequence identity with the ovine adenovirus 7 strain OAV287, a member of the species Ovine atadenovirus D. SD18-74 was amplified in both bovine and ovine primary nasal turbinate cells, demonstrating greater fitness in bovine cells. The genomic and biological characteristics of BAdV-7 SD18-74 support the inclusion of the members of the BAdV-7 group in a new species in the genus Atadenovirus.


Assuntos
Infecções por Adenoviridae/veterinária , Atadenovirus/classificação , Atadenovirus/genética , Bovinos/virologia , Infecções por Adenoviridae/virologia , Animais , Atadenovirus/isolamento & purificação , Atadenovirus/fisiologia , Doenças dos Bovinos/virologia , Linhagem Celular , DNA Viral/genética , Genoma Viral/genética , Ovinos , Estados Unidos , Replicação Viral
3.
J Virol ; 93(21)2019 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-31434730

RESUMO

Senecavirus A (SVA) is a picornavirus that causes acute vesicular disease (VD), that is clinically indistinguishable from foot-and-mouth disease (FMD), in pigs. Notably, SVA RNA has been detected in lymphoid tissues of infected animals several weeks following resolution of the clinical disease, suggesting that the virus may persist in select host tissues. Here, we investigated the occurrence of persistent SVA infection and the contribution of stressors (transportation, immunosuppression, or parturition) to acute disease and recrudescence from persistent SVA infection. Our results show that transportation stress leads to a slight increase in disease severity following infection. During persistence, transportation, immunosuppression, and parturition stressors did not lead to overt/recrudescent clinical disease, but intermittent viremia and virus shedding were detected up to day 60 postinfection (p.i.) in all treatment groups following stress stimulation. Notably, real-time PCR and in situ hybridization (ISH) assays confirmed that the tonsil harbors SVA RNA during the persistent phase of infection. Immunofluorescence assays (IFA) specific for double-stranded RNA (dsRNA) demonstrated the presence of double-stranded viral RNA in tonsillar cells. Most importantly, infectious SVA was isolated from the tonsil of two animals on day 60 p.i., confirming the occurrence of carrier animals following SVA infection. These findings were supported by the fact that contact piglets (11/44) born to persistently infected sows were infected by SVA, demonstrating successful transmission of the virus from carrier sows to contact piglets. Results here confirm the establishment of persistent infection by SVA and demonstrate successful transmission of the virus from persistently infected animals.IMPORTANCE Persistent viral infections have significant implications for disease control strategies. Previous studies demonstrated the persistence of SVA RNA in the tonsil of experimentally or naturally infected animals long after resolution of the clinical disease. Here, we showed that SVA establishes persistent infection in SVA-infected animals, with the tonsil serving as one of the sites of virus persistence. Importantly, persistently infected carrier animals shedding SVA in oral and nasal secretions or feces can serve as sources of infection to other susceptible animals, as evidenced by successful transmission of SVA from persistently infected sows to contact piglets. These findings unveil an important aspect of SVA infection biology, suggesting that persistently infected pigs may function as reservoirs for SVA.


Assuntos
Portador Sadio/veterinária , Transmissão Vertical de Doenças Infecciosas/veterinária , Infecções por Picornaviridae/veterinária , Picornaviridae/patogenicidade , Doenças dos Suínos/transmissão , Animais , Portador Sadio/patologia , Portador Sadio/transmissão , Portador Sadio/virologia , Doença Crônica , Feminino , Tonsila Palatina/virologia , Infecções por Picornaviridae/patologia , Infecções por Picornaviridae/transmissão , Infecções por Picornaviridae/virologia , Recidiva , Estresse Fisiológico , Suínos , Doenças dos Suínos/patologia , Doenças dos Suínos/virologia , Viremia/patologia , Viremia/transmissão , Viremia/veterinária , Viremia/virologia , Eliminação de Partículas Virais
4.
J Virol ; 92(3)2018 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-29142122

RESUMO

Senecavirus A (SVA), an emerging picornavirus of swine, causes vesicular disease (VD) that is clinically indistinguishable from foot-and-mouth disease (FMD) in pigs. Many aspects of SVA interactions with the host and the host immune responses to infection, however, remain unknown. In the present study, humoral and cellular immune responses to SVA were evaluated following infection in pigs. We show that SVA infection elicited an early and robust virus-neutralizing (VN) antibody response, which coincided and was strongly correlated with VP2- and VP3-specific IgM responses. Notably, the neutralizing antibody (NA) responses paralleled the reduction of viremia and resolution of the disease. Analysis of the major porcine T-cell subsets revealed that during the acute/clinical phase of SVA infection (14 days postinfection [p.i.]), T-cell responses were characterized by an increased frequency of αß T cells, especially CD4+ T cells, which were first detected by day 7 p.i. and increased in frequency until day 14 p.i. Additionally, the frequency of CD8+ and double-positive CD4+ CD8+ T cells (effector/memory T cells) expressing interferon gamma (IFN-γ) or proliferating in response to SVA antigen stimulation increased after day 10 p.i. Results presented here show that SVA elicits B- and T-cell activation early upon infection, with IgM antibody levels being correlated with early neutralizing activity against the virus and peak B- and T-cell responses paralleling clinical resolution of the disease. The work provides important insights into the immunological events that follow SVA infection in the natural host.IMPORTANCE Senecavirus A (SVA) has recently emerged in swine, causing outbreaks of vesicular disease (VD) in major swine-producing countries around the world, including the United States, Brazil, China, Thailand, and Colombia. Notably, SVA-induced disease is clinically indistinguishable from other high-consequence VDs of swine, such as FMD, swine vesicular disease, vesicular stomatitis, and vesicular exanthema of swine. Despite the clinical relevance of SVA-induced VD, many aspects of the virus infection biology remain unknown. Here, we assessed host immune responses to SVA infection. The results show that SVA infection elicits early B- and T-cell responses, with the levels of VN antibody and CD4+ T-cell responses paralleling the reduction of viremia and resolution of the disease. SVA-specific CD8+ T cells are detected later during infection. A better understanding of SVA interactions with the host immune system may allow the design and implementation of improved control strategies for this important pathogen of swine.


Assuntos
Imunidade Adaptativa , Picornaviridae , Doença Vesicular Suína/patologia , Linfócitos T/imunologia , Animais , Anticorpos Neutralizantes/imunologia , Anticorpos Antivirais/imunologia , Febre Aftosa/patologia , Interações Hospedeiro-Patógeno , Imunidade Celular , Imunidade Humoral , Suínos , Viremia/imunologia , Viremia/veterinária
5.
Arch Virol ; 164(9): 2321-2326, 2019 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-31175435

RESUMO

Here we describe the identification and genetic characterization of a porcine hepe-astrovirus, or bastrovirus, obtained from feces from pigs in the United States. The genome of the new bastrovirus is 5,955 nt long and contains two open reading frames (ORFs). ORF1 encodes a protein containing three domains, viral methyltransferase, RNA helicase and RNA-dependent RNA polymerase (RdRp), and is closely related to the RdRp of hepatitis E virus. The ORF2 protein shares similarities with the astrovirus capsid precursor protein. Although structural features of bastroviruses may resemble those of astroviruses, distinct characteristics of the newly identified bastrovirus include the presence of an RNA helicase domain in ORF1 and the lack of ORF1b. In addition to genetic characterization, screening of 368 porcine samples (oral fluids, oral swabs or fecal swabs) collected in the United States (US) using a porcine-bastrovirus-specific real-time PCR assay revealed that 31% of those samples were positive. These results suggest a broad distribution of bastroviruses in the swine population in the US. This represents the first description of bastrovirus in swine in the US.


Assuntos
Infecções por Astroviridae/veterinária , Astroviridae/genética , Astroviridae/isolamento & purificação , Doenças dos Suínos/virologia , Animais , Astroviridae/classificação , Infecções por Astroviridae/epidemiologia , Infecções por Astroviridae/virologia , Genoma Viral , Fases de Leitura Aberta , Filogenia , RNA Polimerase Dependente de RNA/genética , Suínos , Doenças dos Suínos/epidemiologia , Estados Unidos/epidemiologia , Proteínas Virais/genética
6.
Genet Mol Biol ; 42(2): 374-379, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31259361

RESUMO

Acute infections of bovine viral diarrhea virus (BVDV) lead to a range of clinical presentations. Laboratory tests for detection depend on collection of samples during a short viremia. Acutely infected animals remain largely undiagnosed. Transfer RNA halves (tsRNAs) are hypothesized to function like microRNAs to regulate gene expression during an immune response. The objective of this study was to identify tsRNAs in cattle that had been challenged with a non-cytopathic field strain of BVDV. Colostrum-deprived neonatal Holstein calves were either challenged with BVDV (n=5) or mock challenged (n=4). Sera was collected prior to challenge and days 4, 9, and 16 post challenge. RNA was extracted and read counts of small non-coding RNAs were assessed using next-generation sequencing. A total of 87,838,207 reads identified 41 different tsRNAs. Two 5' tsRNAs, tsRNAProAGG and tsRNAValAAC, differed across time. Two 5' tsRNAs, tsRNAGlyCCC and tsRNAGlyGCC, differed between treatment groups across time. Four days post challenge, 5' tsRNAGlyCCC and tsRNAGlyGCC were significantly lower in the challenged group than the control group. Further studies are needed to identify the importance and function of 5' tsRNAGlyCCC and tsRNAGlyGCC in serum samples of cattle challenged with BVDV.

7.
Arch Virol ; 163(9): 2327-2335, 2018 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-29725899

RESUMO

Passive immunity is critical for protection of neonatal piglets against porcine epidemic diarrhea virus (PEDV). Here, we investigated the immunogenicity of an orf virus (ORFV) vector expressing the full-length spike (S) protein of PEDV (ORFV-PEDV-S) in pregnant gilts and its ability to confer passive immunity and protection in piglets. Three doses of ORFV-PEDV-S were given to two groups of PEDV-negative pregnant gilts, with the last dose being administered two weeks prior to farrowing. One of the two groups immunized with the ORFV-PEDV-S recombinant virus was also exposed to live PEDV orally on day 31 post-immunization (pi). Antibody responses were assessed in serum, colostrum and milk of immunized gilts, and passive transfer of antibodies was evaluated in piglet sera. The protective efficacy of ORFV-PEDV-S was evaluated after challenge of the piglets with PEDV. PEDV-specific IgG, IgA and neutralizing antibody (NA) responses were detected in ORFV-PEDV-S-immunized and ORFV-PEDV-S-immunized/PEDV-exposed gilts. PEDV NA, IgG and IgA were detected in the serum of piglets born to immunized gilts, demonstrating the transfer of antibodies through colostrum and milk. Piglets born to immunized gilts showed reduced morbidity and a marked reduction in mortality after PEDV challenge in comparison to control piglets. Piglets born to gilts that received ORFV-PEDV-S and were exposed to live PEDV showed stronger NA responses and lower clinical scores when compared to piglets born to gilts immunized with ORFV-PEDV-S alone. These results demonstrate the potential of ORFV as a vaccine delivery platform capable of eliciting passive immunity against PEDV.


Assuntos
Anticorpos Antivirais/sangue , Infecções por Coronavirus/prevenção & controle , Imunidade Materno-Adquirida , Vírus do Orf/imunologia , Vírus da Diarreia Epidêmica Suína/imunologia , Glicoproteína da Espícula de Coronavírus/administração & dosagem , Doenças dos Suínos/prevenção & controle , Animais , Animais Recém-Nascidos , Anticorpos Neutralizantes/sangue , Colostro , Infecções por Coronavirus/imunologia , Infecções por Coronavirus/virologia , Feminino , Vetores Genéticos/administração & dosagem , Vetores Genéticos/química , Vetores Genéticos/imunologia , Imunização Passiva/métodos , Imunoglobulina A/sangue , Imunoglobulina G/sangue , Leite , Vírus do Orf/genética , Vírus da Diarreia Epidêmica Suína/patogenicidade , Gravidez , Glicoproteína da Espícula de Coronavírus/genética , Glicoproteína da Espícula de Coronavírus/imunologia , Suínos , Doenças dos Suínos/imunologia , Doenças dos Suínos/virologia
8.
Arch Virol ; 162(11): 3473-3480, 2017 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-28795249

RESUMO

Naïve pregnant cattle exposed to pestiviruses between 40-125 days of gestation can give birth to persistently infected (PI) calves. Clinical presentation and survivability, in PI cattle, is highly variable even with the same pestivirus strain whereas the clinical presentation in acute infections is more uniform with severity of symptoms being primarily a function of virulence of the infecting virus. The aim of this study was to compare thymic depletion, as measured by comparing the area of the thymic cortex to the medulla (corticomedullary ratio), in acute and persistent infections of the same pestivirus isolate. The same general trends were observed with each pestivirus isolate. Thymic depletion was observed in both acutely and persistently infected calves. The average thymic depletion observed in acutely infected calves was greater than that in age matched PI calves. PI calves, regardless of infecting virus, revealed a greater variability in amount of depletion compared to acutely infected calves. A trend was observed between survivability and depletion of the thymus, with PI calves surviving less than 5 weeks having lower corticomedullary ratios and greater depletion. This is the first study to compare PI and acutely infected calves with the same isolates as well as to evaluate PI calves based on survivability. Further, this study identified a quantifiable phenotype associated with potential survivability.


Assuntos
Vírus da Diarreia Viral Bovina Tipo 1 , Vírus da Diarreia Viral Bovina Tipo 2 , Linfócitos/patologia , Infecções por Pestivirus/veterinária , Pestivirus/classificação , Timo/citologia , Animais , Bovinos , Vírus da Diarreia Viral Bovina Tipo 1/patogenicidade , Vírus da Diarreia Viral Bovina Tipo 2/patogenicidade , Pestivirus/patogenicidade , Infecções por Pestivirus/patologia , Infecções por Pestivirus/virologia , Timo/patologia , Timo/virologia , Virulência
9.
Arch Virol ; 162(10): 3225-3228, 2017 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-28616671

RESUMO

Papillomaviruses are a diverse group of viruses that are known to infect a wide range of animal species. Bovine papillomaviruses (BPVs) are divided into at least 21 genotypes (BPV1 to BPV21),  with most BPV isolates/strains described to date belonging to one of four genera, including Deltapapillomavirus, Xipapillomavirus, Epsilonpapillomavirus and Dyoxipapillomavirus. Here, we describe the identification and genetic characterization of a new BPV type in the genus Dyokappapapillomavirus. A farm in the state of New York, USA, reported chronic cases of vulvovaginitis in Holstein cows in 2016. Biopsies and/or swab samples collected from the vaginal mucosa were subjected to diagnostic investigation. Conventional diagnostic assays yielded negative results, and vaginal swab samples were subjected to viral metagenomic sequencing. Notably, BLAST searches revealed a papillomavirus genome with 7480 bp in length (67% nt sequence identity to BPV16). Additionally, phylogenetic analysis of the L1 gene of the papillomavirus identified here (tentatively named BPV22) revealed that it clusters with members of the genus Dyokappapapillomavirus. Interestingly, the recently identified BPV16, which was detected in fibropapilloma lesions in cattle also clusters within the Dyokappapapillomavirus group. Each virus, however, forms a separate branch in the phylogenetic tree. These results indicate that the putative BPV22 represents the second BPV within the genus Dyokappapapillomavirus.


Assuntos
Doenças dos Bovinos/virologia , Papillomaviridae/isolamento & purificação , Infecções por Papillomavirus/veterinária , Animais , Bovinos , Feminino , Papillomaviridae/genética , Infecções por Papillomavirus/virologia , Filogenia , Vulvovaginite/veterinária , Vulvovaginite/virologia
10.
Viruses ; 15(2)2023 02 06.
Artigo em Inglês | MEDLINE | ID: mdl-36851667

RESUMO

Pestiviruses are globally distributed and cause substantial economic losses to the cattle industry. In Brazil, the country with the world's largest cattle population, pestivirus infections are well described in some regions, such as in the south, where a high frequency of BVDV-2 is described and contrasts with the high prevalence of HoBi-like pestivirus (HoBiPeV) in the northeast. However, there is a lack of information about pestiviruses in the Amazon Region, in northern Brazil, with a cattle population estimated at 55.7 million head, which has a significant impact on the international livestock market. Therefore, this study investigated the seroprevalence and genetic variability of ruminant pestiviruses in 944 bovine serum samples from four states in northern Brazil: Pará (PA), Amapá (AP), Roraima (RR), and Amazonas (AM). Our results showed that 45.4% of the samples were seropositive (19.8% for BVDV-1, 14.1% for BVDV-2, and 20.9% for HoBiPeV). All samples were tested by RT-qPCR, and three were positive and classified as HoBiPeV in a phylogenetic analysis. These serological and molecular results contrast with those from other regions of the world, suggesting that the northern Brazilian states have a high prevalence of all bovine pestiviruses including HoBiPeV.


Assuntos
Vírus da Diarreia Viral Bovina Tipo 2 , Vírus da Diarreia Viral Bovina , Pestivirus , Animais , Bovinos , Pestivirus/genética , Brasil/epidemiologia , Filogenia , Estudos Soroepidemiológicos , Vírus da Diarreia Viral Bovina/genética , Vírus da Diarreia Viral Bovina Tipo 2/genética
11.
Front Vet Sci ; 9: 821247, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35372539

RESUMO

The US Department of Agriculture (USDA), Animal Plant Health Inspection Service (APHIS), Cattle Fever Tick Eradication Program (CFTEP) monitor a quarantine zone along the Texas border to prevent the introduction of stray livestock carrying cattle fever ticks entering the United States from Mexico. Stray cattle collected by CFTEP are checked for ticks and several infectious disease-causing pathogens, but not for bovine viral diarrhea virus (BVDV). BVDV is one of the most economically impactful viruses affecting US cattle producers. BVDV is present in all parts of the world, but it has been demonstrated that another distantly related pestivirus, HoBi-like pestivirus (HoBiPev), can also cause BVD. To date, HoBiPev has not been detected in the United States, but is commonly found in Brazil, and sporadically in Europe and Asia. The objective of the current study was to evaluate the seroprevalence of pestiviruses, with a specific focus on HoBiPev, in stray cattle. Virus neutralization (VN) assay was used to determine seroprevalence (or antibody titers) of BVDV-1, BVDV-2, and HoBiPev. Approximately 50% (67 of 134) of the samples were seropositive for pestiviruses; all 67 positive samples were positive (50%) for BVDV-1, 66 samples of the 67 were positive (49.3%) for BVDV-2, and the same 66 samples of the 67 were also positive (49.3%) for HoBiPev. Due to the antigenic cross-reactivity among Pestiviruses, the comparative antibody against each pestivirus was calculated from all VN-positive samples. Titers were clearly higher against BVDV-1, and only one sample had a titer clearly higher against BVDV-2. No sample had an antibody titer higher for HoBiPev, and while this does not prove the absence of HoBiPev, it does provide evidence that the prevalence of HoBiPev is less predominant than BVDV-1. Additionally, data from these samples provide evidence on the susceptibility of animals that may enter into the United States, with ~50% of the animals seronegative for bovine pestiviruses. This cattle population provides a unique opportunity to evaluate and monitor changes in seroprevalence of economically important cattle diseases affecting the cattle industry.

12.
Front Vet Sci ; 8: 693041, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34368280

RESUMO

Along with viruses in the Pestivirus A (Bovine Viral Diarrhea Virus 1, BVDV1) and B species (Bovine Viral Diarrhea Virus 2, BVDV2), members of the Pestivirus H are mainly cattle pathogens. Viruses belonging to the Pestivirus H group are known as HoBi-like pestiviruses (HoBiPev). Genetic and antigenic characterization suggest that HoBiPev are the most divergent pestiviruses identified in cattle to date. The phylogenetic analysis of HoBiPev results in at least five subgroups (a-e). Under natural or experimental conditions, calves infected with HoBiPev strains typically display mild upper respiratory signs, including nasal discharge and cough. Although BVDV1 and BVDV2 are widely distributed and reported in many South American countries, reports of HoBiPev in South America are mostly restricted to Brazil. Despite the endemicity and high prevalence of HoBiPev in Brazil, only HoBiPev-a was identified to date in Brazil. Unquestionably, HoBiPev strains in BVDV vaccine formulations are required to help curb HoBiPev spread in endemic regions. The current situation in Brazil, where at this point only HoBiPev-a seems present, provides a more significant opportunity to control these viruses with the use of a vaccine with a single HoBiPev subtype. Despite the lack of differentiation among bovine pestiviruses by current BVDV tests, the reduced genetic variability of HoBiPev in Brazil may allow reliable identification of cases within the region. On the other hand, introducing foreign ruminants, biologicals, and genetic material to South America, especially if it originated from other HoBiPev-endemic countries, should consider the risk of introducing divergent HoBiPev subtypes.

13.
Virology ; 556: 87-95, 2021 04.
Artigo em Inglês | MEDLINE | ID: mdl-33550118

RESUMO

In the United States, show pigs are raised to compete in agricultural events. These animals are usually raised in small herds with extensive human, domestic, and wild animal contact. Therefore, pathogen monitoring in this animal category is critical for improved disease surveillance and preparedness. This study characterized the virome of healthy show pigs using high-throughput sequencing using pooled serum samples from 2018 or 2019 (200 samples each pool). Results demonstrated the presence of DNA viral families (Parvoviridae, Circoviridae, and Herpesviridae) and RNA families (Arteriviridae, Flaviviridae, and Retroviridae). Twenty-three viral species were identified, including the first detection of porcine bufavirus in the US. Moreover, important swine pathogens identified included porcine reproductive and respiratory syndrome virus, atypical porcine pestivirus, and porcine circovirus (PCV). Additionally, complete coding genomes of 17 viruses from the Parvoviridae, Anelloviridae, and Circoviridae families were retrieved and included the first near full-length genomes of US Ungulate bocaparvovirus 3 species.


Assuntos
Animais Domésticos/virologia , Suínos/virologia , Viroma , Animais , Oklahoma
14.
Viruses ; 13(12)2021 12 03.
Artigo em Inglês | MEDLINE | ID: mdl-34960693

RESUMO

Bovine serum has been widely used as a universal supplement in culture media and other applications, including the manufacture of biological products and the production of synthetic meat. Currently, commercial bovine serum is tested for possible viral contaminants following regional guidelines. Regulatory agencies' established tests focused on detecting selected animal origin viruses and are based on virus isolation, immunofluorescence, and hemadsorption assays. However, these tests may fail to detect new or emerging viruses in biological products. High-throughput sequencing is a powerful option since no prior knowledge of the viral targets is required. In the present study, we evaluate the virome of seven commercial batches of bovine serum from Mexico (one batch), New Zealand (two batches), and the United States (four batches) using a specific preparation and enrichment method for pooled samples and sequencing using an Illumina platform. A variety of circular replicase-encoding single-stranded (CRESS) DNA families (Genomoviridae, Circoviridae, and Smacoviridae) was identified. Additionally, CrAssphage, a recently discovered group of bacteriophage correlated with fecal contamination, was identified in 85% of the tested batches. Furthermore, sequences representing viral families with single-stranded DNA (Parvoviridae), double-stranded DNA (Polyomaviridae and Adenoviridae), single-stranded RNA (Flaviviridae, Picornaviridae, and Retroviridae), and double-stranded RNA (Reoviridae) were identified. These results support that high-throughput sequencing associated with viral enrichment is a robust tool and should be considered an additional layer of safety when testing pooled biologicals to detect viral contaminants overlooked by the current testing protocols.


Assuntos
Bacteriófagos/isolamento & purificação , Produtos Biológicos , Bovinos/sangue , Vírus de DNA/isolamento & purificação , Vírus de RNA/isolamento & purificação , Soro/virologia , Viroma , Animais , Bacteriófagos/genética , Vírus de DNA/genética , Contaminação de Medicamentos , Sequenciamento de Nucleotídeos em Larga Escala , Filogenia , Vírus de RNA/genética
15.
J Vet Diagn Invest ; 32(4): 513-526, 2020 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-32484424

RESUMO

Bovine coronaviruses (BoCVs) have been found in respiratory tissues in cattle and frequently associated with bovine respiratory disease (BRD); however, pathogenesis studies in calves are limited. To characterize the pathogenesis and pathogenicity of BoCV isolates, we used 5 different BoCV strains to inoculate colostrum-deprived calves, ~ 2-5 wk of age. Later, to determine if dual viral infection would potentiate pathogenicity of BoCV, calves were inoculated with BoCV alone, bovine viral diarrhea virus (BVDV) alone, or a series of dual-infection (BVDV-BoCV) schemes. A negative control group was included in all studies. Clinical signs and body temperature were monitored during the study and samples collected for lymphocyte counts, virus isolation, and serology. During autopsy, gross lesions were recorded and fixed tissues collected for histopathology and immunohistochemistry; fresh tissues were collected for virus isolation. Results suggest increased pathogenicity for isolate BoCV OK 1776. Increased body temperature was found in all virus-inoculated groups. Lung lesions were present in calves in all dual-infection groups; however, lesions were most pronounced in calves inoculated with BVDV followed by BoCV inoculation 6 d later. Lung lesions were consistent with mild-to-moderate interstitial pneumonia, and immunohistochemistry confirmed the presence of BoCV antigen. Our studies demonstrated that BVDV-BoCV dual infection may play an important role in BRD pathogenesis, and timing between infections seems critical to the severity of lesions.


Assuntos
Anticorpos Antivirais/sangue , Doença das Mucosas por Vírus da Diarreia Viral Bovina/virologia , Coronavirus Bovino/isolamento & purificação , Vírus da Diarreia Viral Bovina Tipo 1/isolamento & purificação , Doenças Respiratórias/veterinária , Animais , Doença das Mucosas por Vírus da Diarreia Viral Bovina/patologia , Bovinos , Colostro , Diarreia/veterinária , Vírus da Diarreia Viral Bovina/imunologia , Feminino , Gravidez , Doenças Respiratórias/patologia , Doenças Respiratórias/virologia
16.
Transbound Emerg Dis ; 66(3): 1107-1113, 2019 May.
Artigo em Inglês | MEDLINE | ID: mdl-30688036

RESUMO

Porcine reproductive and respiratory syndrome virus (PRRSV) causes significant economic losses to the swine industry worldwide. While PRRSV has been endemic in North America since 1989, it was not until 1999 that the virus was first described in South America. Notably, recently an increased number of PRRSV outbreaks have been reported in South American countries. However, epidemiological information related to these outbreaks is limited and the genetic characteristics of the PRRSV strains circulating in the region are poorly understood. In this study, we describe the genetic analyses of PRRSV strains associated with severe PRRS outbreaks in Peru. Samples originating from 14 farms located in two Departments in Peru (Lima and Arequipa), were subjected to RT-PCR amplification of the PRRSV ORF5 gene and sequencing followed by restriction fragment length polymorphism (RFLP) analysis. Results demonstrated the circulation of PRRSV-2 in Peru. Notably ORF5 RFLP typing revealed that 15 (75%) of the PRRSV strains detected in this study belong to the RFLP 1-7-4 type. Phylogenetic analysis showed that the Peruvian strains are closely related to the highly virulent PRRSV 1-7-4 strains that emerged in the US in 2013-2014. Results here indicate the presence of highly virulent PRRSV 1-7-4 strains in Peru and provide important information on the geographical distribution of PRRSV, confirming the recent geographical expansion of this important swine pathogen towards South America.


Assuntos
Surtos de Doenças/veterinária , Síndrome Respiratória e Reprodutiva Suína/virologia , Vírus da Síndrome Respiratória e Reprodutiva Suína/isolamento & purificação , Animais , Fazendas , Feminino , Geografia , Masculino , Peru/epidemiologia , Filogenia , Polimorfismo de Fragmento de Restrição , Vírus da Síndrome Respiratória e Reprodutiva Suína/genética , Vírus da Síndrome Respiratória e Reprodutiva Suína/patogenicidade , Reação em Cadeia da Polimerase em Tempo Real/veterinária , Suínos , Proteínas do Envelope Viral/genética
17.
Virus Res ; 260: 53-59, 2019 01 15.
Artigo em Inglês | MEDLINE | ID: mdl-30359622

RESUMO

Fowlpox virus (FWPV), the type species of the genus Avipoxvirus family Poxviridae, is a large double-stranded DNA virus that causes fowlpox in chickens and turkeys. Notably, sequences of the avian retrovirus reticuloendotheliosis virus (REV) are frequently found integrated into the genome of FWPV. While some FWPV strains carry remnants of the REV long terminal repeats (LTRs), other strains have been shown to contain insertions of nearly the full-length REV provirus in their genome. In the present study we detected heterogeneous FWPV populations carrying the REV LTR or the near full-length REV provirus genome in a Merriam's wild turkey (Meleagris gallopavo merriami). The bird presented papules distributed throughout the non-feathered areas of the head. Avipoxvirus-like virions were observed in the lesions by transmission electron microscopy and the presence of FWPV was confirmed by DNA sequencing. Metagenomic sequencing performed on nucleic acid extracted from the skin lesions revealed two FWPV genome populations carrying either a 197-nt remnant of the REV LTR or a 7939-nt long fragment corresponding to the full-length REV provirus. Notably, PCR amplification using primers targeting FWPV sequences flanking the REV insertion site, confirmed the natural occurrence of the heterogeneous FWPV genome populations in one additional clinical sample from another turkey affected by fowlpox. Additionally, sequencing of a historical FWPV isolate obtained from chickens in the US in 2000 also revealed the presence of the two FWPV-REV genome populations. Results here demonstrate distinct FWPV populations containing variable segments of REV genome integrated into their genome. These distinct genome populations are likely a result of homologous recombination events that take place during FWPV replication.


Assuntos
Vírus da Varíola das Aves Domésticas/genética , Varíola Aviária/virologia , Vírus da Reticuloendoteliose/genética , Perus/virologia , Animais , Varíola Aviária/patologia , Vírus da Varíola das Aves Domésticas/isolamento & purificação , Genoma Viral , Metagenômica , Microscopia Eletrônica de Transmissão , Reação em Cadeia da Polimerase , Análise de Sequência de DNA , Pele/patologia , Pele/virologia , Sequências Repetidas Terminais , Integração Viral
19.
Virology ; 522: 147-157, 2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-30029014

RESUMO

The goals of this study were to compare the pathogenicity and infection dynamics of a historical and a contemporary SVA strains (SVV 001 and SD15-26) and to assess cross-neutralizing and cross-reactive T cell responses following experimental infection in pigs. Both SVA strains successfully infected all inoculated animals, resulting in viremia and robust antibody and cellular immune responses. SVA SD15-26 infection resulted in characteristic clinical signs and vesicular lesions, however, SVA SVV 001 did not cause overt clinical disease with inoculated animals remaining clinically normal during the experiment. Notably, neutralization- and -recall IFN-γ expression-assays revealed marked cross-neutralizing antibody and cross-reactive T cell responses between the two viral strains. Together these results demonstrate that the historical SVA SVV 001 strain presents low virulence in pigs when compared to the contemporary SVA SD15-26 strain. Additionally, immunological assays indicate that SVA SVV 001 and SD15-26 are antigenically related and share conserved antigenic determinants.


Assuntos
Anticorpos Neutralizantes/imunologia , Anticorpos Antivirais/imunologia , Reações Cruzadas , Infecções por Picornaviridae/veterinária , Picornaviridae/imunologia , Picornaviridae/patogenicidade , Doenças dos Suínos/virologia , Linfócitos T/imunologia , Animais , Anticorpos Neutralizantes/sangue , Anticorpos Antivirais/sangue , Interferon gama/metabolismo , Picornaviridae/isolamento & purificação , Infecções por Picornaviridae/imunologia , Infecções por Picornaviridae/patologia , Infecções por Picornaviridae/virologia , Suínos , Doenças dos Suínos/imunologia , Doenças dos Suínos/patologia , Virulência
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA