Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Dev Comp Immunol ; 127: 104271, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-34600023

RESUMO

The α- and ß-adrenergic receptors (ARs) bind the stress hormones epinephrine (E), norepinephrine (NE), and dopamine and activate diverse physiological responses. A lack of information on AR gene expression in leukocytes limits our understanding of how stress alters immune function. Quantitative analyses of AR gene expression was completed for bovine leukocytes. Individual leukocyte lineages and subpopulations within lineages were isolated with high-speed cell sorting to facilitate a targeted analysis of AR gene expression. These analyses confirmed all 9 AR genes were expressed in bovine leukocytes with marked differences in AR gene expression when comparing among leukocyte lineages. Furthermore, separation of polymorphonuclear cells into neutrophils and eosinophils revealed these key innate immune cells also differ significantly in AR gene expression. This study provides the first comprehensive survey of AR gene expression in immune cells of any mammalian species and provides insight into conflicting reports that stress can either activate or suppress immune function.


Assuntos
Leucócitos , Norepinefrina , Animais , Bovinos , Epinefrina/metabolismo , Expressão Gênica , Leucócitos/metabolismo , Norepinefrina/metabolismo , Receptores Adrenérgicos beta/metabolismo
2.
J Infect Dev Ctries ; 15(5): 653-656, 2021 05 31.
Artigo em Inglês | MEDLINE | ID: mdl-34106888

RESUMO

Understanding the efficacy and durability of heterologous immunization schedules against SARS-CoV-2 is critical, as supply demands and vaccine choices become significant issues in the global vaccination strategy. Here we characterize the neutralizing antibodies produced in two subjects who received combination immunizations against SARS-CoV-2, first with Covishield (Oxford-AstraZeneca) vaccine, followed 33 days later with a second dose (booster) shot of the Pfizer-BioNTech vaccine. Serum samples were collected 25 days following the primary vaccination and 13 days after the secondary Pfizer vaccination. Both subjects exhibited increased levels of isotype IgG and IgM antibodies directed against the entire spike protein following immunizations. These antibodies also exhibited increased reactivity with the receptor binding domain (RBD) in the spike protein and neutralized the infectivity of replicating vesicular stomatitis virus (VSV) that contains the COVID-19 coronavirus S protein gene in place of its normal G glycoprotein. This VSV pseudovirus also contains the reporter gene for enhanced green fluorescent protein (eGFP). Antibody titers against the spike protein and serum neutralization titers against the reporter virus are reported for the 2 heterologous vaccinated individuals and compared to a positive control derived from a convalescent patient and a negative control from an unexposed individual. The Pfizer-BioNTech vaccine increased antibody binding to the spike protein and RBD, and approached levels found in the convalescent positive control. Neutralizing antibodies against the VSV-S pseudovirus in the 2 subjects also approached levels in the convalescent sera. These results firmly validate the value of the Pfizer-BioNTech vaccine in boosting immunity following initial Covishield inoculation.


Assuntos
Vacinas contra COVID-19/imunologia , COVID-19/imunologia , Imunidade Humoral/efeitos dos fármacos , Anticorpos Neutralizantes/imunologia , COVID-19/prevenção & controle , Estudos de Casos e Controles , Feminino , Humanos , Masculino , SARS-CoV-2
3.
Vet Immunol Immunopathol ; 237: 110256, 2021 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-33971523

RESUMO

Lawsonia intracellularis is an economically important bacterium that causes ileitis in pigs. Current vaccines for L. intracellularis do not allow for differentiation between infected and vaccinated animals (DIVA), which is beneficial for disease tracking and surveillance. Previously, we identified five putative surface L. intracellularis proteins that were targeted by antibodies from pigs infected with L. intracellularis which could serve as antigens in a subunit vaccine. We conducted two trials to determine whether these antigens were immunogenic and provided protection against infectious challenge and whether truncated glycoprotein D could be used as a DIVA antigen. For Trial 1, 5 week-old piglets were administered intramuscular monovalent vaccines comprised of a recombinant (r) flagella subunit protein (rFliC,) and DIVA antigen (truncated glycoprotein D (TgD), a herpes virus antigen) both formulated with a combination adjuvant consisting of polyinosinic:polycytidylic acid(poly I:C), host defense peptide 1002 and polyphosphazene, referred to as Triple Adjuvant (TriAdj). Relative to control animals, animals vaccinated with rFliC and rTgD had significantly elevated antigen-specific humoral immunity in sera suggesting that rFliC and TgD are immunogenic. Control animals had negligible anti-TgD titres suggesting that TgD may be a suitable DIVA antigen for pigs. For Trial 2, piglets were immunized with a trivalent vaccine (FOG vaccine consisting of rFLiC, rOppA protein (a ABC Type dipeptide transport system) and rGroEL (a stress response protein)) and a divalent vaccine (CM vaccine consisting of rClpP (an ATP-dependent Clp protease proteolytic subunit) and rMetK (a S-adenosyl methionine synthase)) formulated with Emulsigen®. Relative to the control pigs, pigs immunized with the FOG vaccine produced robust and significantly higher serum IgG antibodies against rFliC and rGroEL, and significantly higher anti-FliC and anti-GroEL IgA antibodies in jejunal (GroEL only) and ileal intestinal mucosa. Pigs immunized with CM vaccine produced significantly higher serum antibodies against rClpP and rMetK and significantly higher anti-rClpP IgA antibodies in the ileum relative to the control pigs. Quantitative polymerase chain reaction (qPCR) analysis showed that 18 days after challenge with infectious L. intracellularis, challenged/control pigs and pigs that received the CM vaccine, but not the pigs vaccinated with the FOG vaccine, shed significantly more bacteria in feces than the unchallenged controls pigs. These data suggest that the FOG vaccinated pigs showed limited protection. While promising, more work is needed to enhance the efficiency of the intramuscular vaccine to show significant disease protection.


Assuntos
Vacinas Bacterianas/imunologia , Infecções por Desulfovibrionaceae/prevenção & controle , Imunogenicidade da Vacina , Lawsonia (Bactéria)/imunologia , Doenças dos Suínos/prevenção & controle , Animais , Animais Recém-Nascidos , Anticorpos Antibacterianos/imunologia , Infecções por Desulfovibrionaceae/imunologia , Feminino , Gravidez , Suínos , Doenças dos Suínos/microbiologia , Vacinas Combinadas/imunologia , Vacinas de Subunidades Antigênicas/imunologia
4.
Vaccine ; 38(10): 2387-2395, 2020 02 28.
Artigo em Inglês | MEDLINE | ID: mdl-32014270

RESUMO

The highly pathogenic avian influenza (HPAI) H5N1 viruses and their spillover into the human population pose substantial economic and public health threats. Although antiviral drugs have some effect in treating influenza infection, vaccination is still the most effective intervention to prevent possible pandemic outbreaks. We have developed a novel H5 influenza vaccine to improve the world's pandemic preparedness. We produced a hemagglutinin (HA) of HPAI H5N1 virus A/Alberta/01/2014 (AB14) using both mammalian (m) and bacterial (b) expression systems. The purified recombinant proteins were formulated with a proprietary adjuvant (TriAdj) and their efficacy as vaccine candidates was evaluated in mice. Intramuscular delivery of two doses of TriAdj formulated mammalian expressed HA (m-HA/TriAdj) was shown to provide full protection against a lethal challenge of AB14 in mice. In contrast, bacterially expressed HA with TriAdj (b-HA/TriAdj), b-HA without adjuvant, and m-HA without adjuvant resulted in no protection in immunized mice. Furthermore, m-HA/TriAdj elicited significantly higher levels of balanced Th1 and Th2 responses and neutralizing antibody titres. All the mice in the m-HA/TriAdj group survived a lethal AB14 H5N1 challenge and showed no signs of disease or infection as demonstrated by no loss of body weight or detectable virus in the lungs. Our results suggest that m-HA formulated with TriAdj has potential to protect against pandemic H5N1 in the event of its cross over to the human host.


Assuntos
Adjuvantes Imunológicos/administração & dosagem , Glicoproteínas de Hemaglutininação de Vírus da Influenza/imunologia , Vacinas contra Influenza/imunologia , Infecções por Orthomyxoviridae , Animais , Anticorpos Antivirais/sangue , Glicoproteínas de Hemaglutininação de Vírus da Influenza/administração & dosagem , Virus da Influenza A Subtipo H5N1 , Camundongos , Camundongos Endogâmicos BALB C , Infecções por Orthomyxoviridae/prevenção & controle , Células Th1/imunologia , Células Th2/imunologia
5.
Vet Microbiol ; 235: 270-279, 2019 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-31383312

RESUMO

Lawsonia intracellularis is an obligate intracellular microorganism and the causative agent of porcine proliferative enteropathy. Due to its obligate intracellular nature, characterization of antigens and proteins involved in host-pathogen interaction and immune recognition have been difficult to achieve using conventional microbiological techniques. In this work, we used 2-dimensional gel electrophoresis coupled with Western-immunoblotting, mass spectrometry and bioinformatics to identify bacterial proteins that interact in vitro with pig intestinal cells (IPEC-1), have immunogenic properties and the potential to be used as subunit vaccine antigens. We detected eleven immunogenic bacterial proteins from which fliC (LI0710), LI1153 (annotated by NCBI as Putative protein N), and LI0649 (annotated as autotransporter) were predicted to be expressed on the outer membrane while LI0169 (oppA; annotated as ABC dipeptide transport system) was predicted to be periplasmic with a transmembrane domain forming a central pore through the plasma membrane. Genes coding for these four proteins were cloned and expressed in Escherichia coli and the corresponding recombinant proteins were purified using affinity chromatography. Porcine hyperimmune serum against whole Lawsonia lysate established that all four recombinant proteins were immunogenic. Further, rabbit hyperimmune sera generated against the vaccine strain of L. intracellularis and rabbit serum specific for each recombinant protein showed an inhibitory effect on the attachment and penetration of live, avirulent L. intracellularis, thus indicating that each protein is a potential neutralizing antibody target and a candidate for subunit vaccine formulation.


Assuntos
Anticorpos Antibacterianos/imunologia , Anticorpos Neutralizantes/imunologia , Proteínas de Bactérias/imunologia , Vacinas Bacterianas/imunologia , Infecções por Desulfovibrionaceae/veterinária , Lawsonia (Bactéria)/imunologia , Animais , Proteínas de Bactérias/genética , Western Blotting , Linhagem Celular , Biologia Computacional , Infecções por Desulfovibrionaceae/imunologia , Infecções por Desulfovibrionaceae/prevenção & controle , Feminino , Intestinos/citologia , Intestinos/microbiologia , Espectrometria de Massas , Proteômica , Coelhos , Proteínas Recombinantes/imunologia , Suínos , Doenças dos Suínos/imunologia , Doenças dos Suínos/microbiologia , Doenças dos Suínos/prevenção & controle , Vacinas de Subunidades Antigênicas/imunologia
6.
J Virol ; 93(6)2019 03 15.
Artigo em Inglês | MEDLINE | ID: mdl-30626671

RESUMO

Bovine herpesvirus 1 (BoHV-1) infects bovine species, causing respiratory infections, genital disorders and abortions. VP8 is the most abundant tegument protein of BoHV-1 and is critical for virus replication in cattle. In this study, the cellular transport of VP8 in BoHV-1-infected cells and its ability to alter the cellular lipid metabolism were investigated. A viral kinase, US3, was found to be involved in regulating these processes. In the early stages of infection VP8 was localized in the nucleus. Subsequently, presumably after completion of its role in the nucleus, VP8 was translocated to the cytoplasm. When US3 was deleted or the essential US3 phosphorylation site of VP8 was mutated in BoHV-1, the majority of VP8 was localized in the nuclei of infected cells. This suggests that phosphorylation by US3 may be critical for cytoplasmic localization of VP8. Eventually, the cytoplasmic VP8 was accumulated in the cis-Golgi apparatus but not in the trans-Golgi network, implying that VP8 was not involved in virion transport toward and budding from the cell membrane. VP8 caused lipid droplet (LD) formation in the nuclei of transfected cells and increased cellular cholesterol levels. Lipid droplets were not found in the nuclei of BoHV-1-infected cells when VP8 was cytoplasmic in the presence of US3. However, when US3 was deleted or phosphorylation residues in VP8 were mutated, nuclear VP8 and LDs appeared in BoHV-1-infected cells. The total cholesterol level was increased in BoHV-1-infected cells but not in ΔUL47-BoHV-1-infected cells, further supporting a role for VP8 in altering the cellular lipid metabolism during infection.IMPORTANCE Nuclear localization signals (NLSs) and nuclear export signals (NESs) are important elements directing VP8 to the desired locations in the BoHV-1-infected cell. In this study, a critical regulator that switches the nuclear and cytoplasmic localization of VP8 in BoHV-1-infected cells was identified. BoHV-1 used viral kinase US3 to regulate the cellular localization of VP8. Early during BoHV-1 infection VP8 was localized in the nucleus, where it performs various functions; once US3 was expressed, phosphorylated VP8 was cytoplasmic and ultimately accumulated in the cis-Golgi apparatus, presumably to be incorporated into virions. The Golgi localization of VP8 was only observed in virus-infected cells and not in US3-cotransfected cells, suggesting that this is mediated by other viral factors. Interestingly, VP8 was shown to cause increased cholesterol levels, which is a novel function for VP8 and a potential strategy to supply lipid for viral replication.


Assuntos
Proteínas do Capsídeo/metabolismo , Infecções por Herpesviridae/metabolismo , Herpesvirus Bovino 1/metabolismo , Metabolismo dos Lipídeos/fisiologia , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Virais/metabolismo , Animais , Células COS , Bovinos , Linhagem Celular , Membrana Celular/metabolismo , Membrana Celular/virologia , Núcleo Celular/metabolismo , Núcleo Celular/virologia , Chlorocebus aethiops , Citoplasma/metabolismo , Citoplasma/virologia , Complexo de Golgi/virologia , Infecções por Herpesviridae/virologia , Humanos , Sinais de Localização Nuclear/metabolismo , Fosforilação , Vírion/metabolismo , Replicação Viral/fisiologia
7.
Virus Res ; 261: 37-49, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30550895

RESUMO

The UL47 gene product, VP8, is a major tegument protein of BoHV-1. While VP8 is not essential for virus replication in cell culture, a UL47-deleted virus exhibits a smaller tegument structure and is avirulent in cattle. To obtain pure VP8 protein for structural analysis, we expressed a N-terminally truncated version of VP8 in Eschericia coli. However, the recombinant VP8 was consistently co-purified with a tightly associated bacterial protein; this protein was identified by mass spectrometry as GroEL, which has considerable homology with mammalian heat shock protein-60 (HSP60), thus suggesting a new role for VP8 in virus-host interaction. A physical interaction of HSP60 and VP8 in both VP8-transfected and BoHV-1-infected cells was demonstrated by immunoprecipitation. Analysis of different truncated VP8 constructs revealed that amino acids 259-482 and 632-741 are involved in binding to HSP60. Full-length VP8 and VP8 219-741 (containing both interacting domains, 259-482 and 632-741) co-localized with HSP60 and mitochondria. VP8 was localized in the mitochondria from 2 to 14 h post infection in BoHV-1-infected cells. The mitochondrial membrane potential was reduced in both VP8-transfected and BoHV-1-infected cells and was further diminished by overexpression of HSP60 in the presence of VP8. In addition, VP8 expression decreased the ATP concentration during transfection, as well as BoHV-1 infection. Thus, VP8 may play a role in the deregulation of mitochondrial function through interaction with HSP60. This is consistent with the fact that BoHV-1 infection is known to promote mitochondrial dysfunction.


Assuntos
Proteínas do Capsídeo/metabolismo , Chaperonina 60/metabolismo , Herpesvirus Bovino 1/fisiologia , Interações Hospedeiro-Patógeno , Mitocôndrias/patologia , Mapeamento de Interação de Proteínas , Trifosfato de Adenosina/análise , Animais , Proteínas do Capsídeo/análise , Bovinos , Linhagem Celular , Chaperonina 60/análise , Células Epiteliais/virologia , Humanos , Imunoprecipitação , Potenciais da Membrana , Mitocôndrias/química , Membranas Mitocondriais/fisiologia , Ligação Proteica
8.
Nat Commun ; 9(1): 3199, 2018 08 10.
Artigo em Inglês | MEDLINE | ID: mdl-30097581

RESUMO

The nucleus represents a cellular compartment where the discrimination of self from non-self nucleic acids is vital. While emerging evidence establishes a nuclear non-self DNA sensing paradigm, the nuclear sensing of non-self RNA, such as that from nuclear-replicating RNA viruses, remains unexplored. Here, we report the identification of nuclear-resident RIG-I actively involved in nuclear viral RNA sensing. The nuclear RIG-I, along with its cytoplasmic counterpart, senses influenza A virus (IAV) nuclear replication leading to a cooperative induction of type I interferon response. Its activation signals through the canonical signaling axis and establishes an effective antiviral state restricting IAV replication. The exclusive signaling specificity conferred by nuclear RIG-I is reinforced by its inability to sense cytoplasmic-replicating Sendai virus and appreciable sensing of hepatitis B virus pregenomic RNA in the nucleus. These results refine the RNA sensing paradigm for nuclear-replicating viruses and reveal a previously unrecognized subcellular milieu for RIG-I-like receptor sensing.


Assuntos
Antivirais/farmacologia , Núcleo Celular/metabolismo , Proteína DEAD-box 58/metabolismo , Imunidade Inata , Replicação Viral/fisiologia , Células A549 , Animais , Compartimento Celular/efeitos dos fármacos , Núcleo Celular/efeitos dos fármacos , Cães , Células HEK293 , Humanos , Vírus da Influenza A/efeitos dos fármacos , Células Madin Darby de Rim Canino , Ligação Proteica/efeitos dos fármacos , RNA Viral/metabolismo , Receptores Imunológicos , Ribonucleoproteínas/metabolismo , Transdução de Sinais/efeitos dos fármacos
9.
J Gen Virol ; 98(7): 1831-1842, 2017 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-28675355

RESUMO

Invitro investigations have identified a variety of mechanisms by which herpesviruses evade interferon-stimulated antiviral effector mechanisms. However, these immune evasion mechanisms have not been evaluated during a bovine herpesvirus-1 (BHV-1) infection. This study investigated the transcription and secretion of type I and II interferons (IFNs) and the transcription of IFN-stimulated genes (ISGs) during a primary BHV-1 infection of the upper respiratory tract (URT) in naïve calves. IFN-α, -ß and -γ transcription in nasal turbinates and protein levels in nasal secretions increased following infection. Increased IFN type I and II secretion was detected 3 days post-infection (p.i.) and IFN production increased in parallel with virus shedding. Expression of ISGs, including Mx1, OAS and BST-2, also increased significantly (P<0.05) in nasal turbinates on day 3 p.i. and elevated ISG expression persisted throughout the period of viral shedding. In contrast, RNAase L gene expression was not induced during the BHV-1 infection in the nasal turbinates, but was induced on day 10 p.i. in the trachea. In vitro studies confirmed that recombinant bovine (rBo)IFN-α, -ß and -γ induced expression of Mx1, OAS and BST-2, but decreased RNAse L transcript in bovine epithelial cells. Relative to vesicular stomatitisvirus (VSV), BHV-1 was resistant to the antiviral activity of rBoIFN-α and -γ, but treatment of epithelial cells with 10 ng rBoIFN-ß ml-1 effected an 80 % inhibition of BHV-1 replication and complete inhibition of VSV replication. These observations confirm that the transcription and translation of type I and II IFNs increase during BHV-1 infection, while the transcription of some ISGs is not inhibited.


Assuntos
Doenças dos Bovinos/genética , Herpesvirus Bovino 1/fisiologia , Fatores Reguladores de Interferon/genética , Interferons/genética , Infecções Respiratórias/genética , Animais , Bovinos , Doenças dos Bovinos/imunologia , Doenças dos Bovinos/virologia , Herpesvirus Bovino 1/genética , Fatores Reguladores de Interferon/imunologia , Interferons/imunologia , Infecções Respiratórias/imunologia , Infecções Respiratórias/virologia , Replicação Viral
10.
Vaccine ; 35(27): 3466-3473, 2017 06 14.
Artigo em Inglês | MEDLINE | ID: mdl-28527686

RESUMO

Bovine viral diarrhea virus (BVDV) is one of the most serious pathogens in cattle. Recently, we developed a novel adjuvant platform (TriAdj) that includes a toll-like receptor 3 agonist, poly (I:C); an innate defense regulatory peptide; and water-soluble polymer, poly[di(sodiumcarboxylatoethylphenoxy)]-phosphazene (PCEP). To develop a needle-free intradermal (ID) subunit vaccine, the BVDV type-2 E2 protein was formulated with TriAdj, and immune protection was evaluated in calves against a BVDV-2 strain. Intradermal delivery of E2/TriAdj elicited robust virus neutralizing antibodies and cell-mediated immune responses including CD4+ and CD8+ T-cell responses. The development of CD8+ T-cell responses in vaccinated calves indicates that TriAdj promotes cross-presentation. Upon challenge with virulent BVDV-2, the vaccinated calves showed no weight loss, leukopenia or virus shedding, and almost no temperature increase, in contrast to the control animals, which had severe clinical disease and shed virus for three to six days in nasal fluids and white blood cells. Intradermal vaccination was shown to attract various immune cell populations including dendritic cells, the most important antigen presenting cells. These data demonstrate that ID delivery is suitable as an administration route in cattle and that ID delivered, TriAdj-formulated E2 can protect cattle from BVDV-2.


Assuntos
Adjuvantes Imunológicos/administração & dosagem , Doença das Mucosas por Vírus da Diarreia Viral Bovina/prevenção & controle , Vírus da Diarreia Viral Bovina Tipo 1/imunologia , Imunidade Inata , Proteínas do Envelope Viral/imunologia , Vacinas Virais/imunologia , Animais , Anticorpos Neutralizantes/sangue , Anticorpos Antivirais/sangue , Peso Corporal , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/imunologia , Bovinos , Febre/prevenção & controle , Injeções Intradérmicas , Leucopenia/prevenção & controle , Vacinas Virais/administração & dosagem , Eliminação de Partículas Virais
11.
Virol J ; 13: 57, 2016 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-27036203

RESUMO

BACKGROUND: Porcine epidemic diarrhea virus (PEDV) is a highly contagious virus infecting pigs of all ages with high morbidity and mortality among newborn piglets. Currently, there is no effective vaccine available to protect the pigs from PEDV. The N-terminal subunit of spike protein (S1) is responsible for virus binding to the cellular receptor and contains a number of neutralizing antibody epitopes. Thus, we expressed and produced recombinant S1 protein to protect newborn piglets by immunization of sows. METHODS: Affinity tagged PEDV S1 protein was expressed in a secretory form in yeast, insect and mammalian cells to identify the most suitable production system. Purified recombinant protein was analysed by SDS-PAGE, Western blot and deglycosylation assay. A pregnant sow was intramuscularly immunized three times with adjuvanted recombinant protein prior to farrowing. PEDV-specific immune responses in sera and colostrum of the sow and piglets were assayed by ELISA and virus neutralization assays. Piglets were challenged orally with PEDV, and clinical parameters were monitored for 6 days post-challenge. RESULTS AND CONCLUSION: Of three eukaryotic expression systems tested (yeast, insect-cell, and mammalian), expression by HEK-293 T cells gave the highest yield of protein that was N-glycosylated and was the most appropriate candidate for vaccination. Administration of the subunit vaccine in a sow resulted in induction of S1-specific IgG and IgA that were passively transferred to the suckling piglets. Also, high virus neutralization titres were observed in the serum of the vaccinated sow and its piglets. After PEDV challenge, piglets born to the vaccinated sow exhibited less severe signs of disease and significantly lower mortality compared to the piglets of a control sow. However, there were no significant differences in diarrhea, body weight and virus shedding. Thus, vaccination with S1 subunit vaccine failed to provide complete protection to suckling piglets after challenge exposure, and further improvements are needed for the development of a subunit vaccine that fully protects against PEDV infection.


Assuntos
Antígenos Virais/imunologia , Infecções por Coronavirus/veterinária , Vírus da Diarreia Epidêmica Suína/imunologia , Glicoproteína da Espícula de Coronavírus/imunologia , Vacinas Virais/imunologia , Adjuvantes Imunológicos/administração & dosagem , Animais , Anticorpos Neutralizantes/análise , Anticorpos Antivirais/análise , Antígenos Virais/genética , Colostro/imunologia , Infecções por Coronavirus/patologia , Infecções por Coronavirus/prevenção & controle , Ensaio de Imunoadsorção Enzimática , Feminino , Injeções Intramusculares , Testes de Neutralização , Vírus da Diarreia Epidêmica Suína/genética , Gravidez , Soro/imunologia , Glicoproteína da Espícula de Coronavírus/genética , Suínos , Resultado do Tratamento , Vacinas de Subunidades Antigênicas/administração & dosagem , Vacinas de Subunidades Antigênicas/genética , Vacinas de Subunidades Antigênicas/imunologia , Vacinas Sintéticas/administração & dosagem , Vacinas Sintéticas/genética , Vacinas Sintéticas/imunologia , Vacinas Virais/administração & dosagem , Vacinas Virais/genética
12.
J Virol ; 90(10): 4889-4904, 2016 05 15.
Artigo em Inglês | MEDLINE | ID: mdl-26889034

RESUMO

UNLABELLED: The UL47 gene product, VP8, is the most abundant tegument protein of bovine herpesvirus 1 (BoHV-1). Previously, we demonstrated that a UL47-deleted BoHV-1 mutant (BoHV1-ΔUL47) exhibits 100-fold-reduced virulence in vitro and is avirulent in vivo In this study, we demonstrated that VP8 expression or BoHV-1 infection inhibits interferon beta (IFN-ß) signaling by using an IFN-α/ß-responsive plasmid in a luciferase assay. As transducer and activator of transcription (STAT) is an essential component in the IFN-signaling pathways, the effect of VP8 on STAT was investigated. An interaction between VP8 and STAT1 was established by coimmunoprecipitation assays in both VP8-transfected and BoHV-1-infected cells. Two domains of VP8, amino acids 259 to 482 and 632 to 686, were found to be responsible for its interaction with STAT1. The expression of VP8 did not induce STAT1 ubiquitination or degradation. Moreover, VP8 did not reduce STAT1 tyrosine phosphorylation to downregulate IFN-ß signaling. However, the expression of VP8 or a version of VP8 (amino acids 219 to 741) that contains the STAT1-interacting domains but not the nuclear localization signal prevented nuclear accumulation of STAT1. Inhibition of nuclear accumulation of STAT1 also occurred during BoHV-1 infection, while nuclear translocation of STAT1 was observed in BoHV1-ΔUL47-infected cells. During BoHV-1 infection, VP8 was detected in the cytoplasm at 2 h postinfection without any de novo protein synthesis, at which time STAT1 was already retained in the cytoplasm. These results suggest that viral VP8 downregulates IFN-ß signaling early during infection, thus playing a role in overcoming the antiviral response of BoHV-1-infected cells. IMPORTANCE: Since VP8 is the most abundant protein in BoHV-1 virions and thus may be released in large amounts into the host cell immediately upon infection, we proposed that it might have a function in the establishment of conditions suitable for viral replication. Indeed, while nonessential in vitro, it is critical for BoHV-1 replication in vivo In this study, we determined that VP8 plays a role in downregulation of the antiviral host response by inhibiting IFN-ß signaling. VP8 interacted with and prevented nuclear accumulation of STAT1 at 2 h postinfection in the absence of de novo viral protein synthesis. Two domains of VP8, amino acids 259 to 482 and 632 to 686, were found to be responsible for this interaction. These results provide a new functional role for VP8 in BoHV-1 infection and a potential explanation for the lack of viral replication of the UL47 deletion mutant in cattle.


Assuntos
Proteínas do Capsídeo/imunologia , Proteínas do Capsídeo/metabolismo , Herpesvirus Bovino 1/metabolismo , Interferon beta/metabolismo , Fator de Transcrição STAT1/metabolismo , Transdução de Sinais , Animais , Proteínas do Capsídeo/química , Proteínas do Capsídeo/genética , Chlorocebus aethiops , Citoplasma/metabolismo , Regulação para Baixo , Interações Hospedeiro-Patógeno , Humanos , Interferon beta/imunologia , Interferon beta/farmacologia , Mutação , Sinais de Localização Nuclear/metabolismo , Fosforilação , Fator de Transcrição STAT1/imunologia , Homologia de Sequência de Aminoácidos , Transdução de Sinais/genética , Ubiquitinação , Células Vero , Vírion/metabolismo , Replicação Viral
13.
J Virol ; 90(9): 4427-4440, 2016 May.
Artigo em Inglês | MEDLINE | ID: mdl-26889039

RESUMO

UNLABELLED: VP8 is a major tegument protein of bovine herpesvirus 1 (BoHV-1) and is essential for viral replication in cattle. The protein undergoes phosphorylation after transcription through cellular casein kinase 2 (CK2) and a viral kinase, US3. In this study, a virus containing a mutated VP8 protein that is not phosphorylated by CK2 and US3 (BoHV-1-YmVP8) was constructed by homologous recombination in mammalian cells. When BoHV-1-YmVP8-infected cells were observed by transmission electron microscopy, blocking phosphorylation of VP8 was found to impair viral DNA encapsidation, resulting in release of incomplete viral particles to the extracellular environment. Consequently, less infectious virus was produced by the mutant virus than by wild-type (WT) virus. A comparison of mutant and WT VP8 by confocal microscopy revealed that mutant VP8 is nuclear throughout infection while WT VP8 is nuclear early during infection and is associated with the Golgi apparatus at later stages. This, together with the observation that mutant VP8 is present in virions, albeit in smaller amounts, suggests that the incorporation of VP8 may occur at two stages. The first takes place without the need for phosphorylation and before or during nuclear egress of capsids, whereas the second occurs in the Golgi apparatus and requires phosphorylation of VP8. The results indicate that phosphorylated VP8 plays a role in viral DNA encapsidation and in the secondary virion incorporation of VP8. To perform these functions, the cellular localization of VP8 is adjusted based on the phosphorylation status. IMPORTANCE: In this study, phosphorylation of VP8 was shown to have a function in BoHV-1 replication. A virus containing a mutated VP8 protein that is not phosphorylated by CK2 and US3 (BoHV-1-YmVP8) produced smaller numbers of infectious virions than wild-type (WT) virus. The maturation and egress of WT and mutant BoHV-1 were studied, showing a process similar to that reported for other alphaherpesviruses. Interestingly, lack of phosphorylation of VP8 by CK2 and US3 resulted in reduced incorporation of viral DNA into capsids during mutant BoHV-1 infection, as well as lower numbers of extracellular virions. Furthermore, mutant VP8 remained nuclear throughout infection, in contrast to WT VP8, which is nuclear at early stages and Golgi apparatus associated late during infection. This correlates with smaller amounts of mutant VP8 in virions and suggests for the first time that VP8 may be assembled into the virions at two stages, with the latter dependent on phosphorylation.


Assuntos
Proteínas do Capsídeo/metabolismo , DNA Viral , Infecções por Herpesviridae/virologia , Herpesvirus Bovino 1/fisiologia , Montagem de Vírus , Replicação Viral , Animais , Proteínas do Capsídeo/genética , Bovinos , Linhagem Celular , Herpesvirus Bovino 1/isolamento & purificação , Herpesvirus Bovino 1/ultraestrutura , Mutação , Fosforilação , Transporte Proteico , Recombinação Genética , Liberação de Vírus
14.
Biologicals ; 43(6): 444-51, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26460173

RESUMO

Histophilosis of cattle is caused by the Gram negative bacterial pathogen Histophilus somni (H. somni) which is also associated with the bovine respiratory disease (BRD) complex. Existing vaccines for H. somni include either killed cells or bacteria-free outer membrane proteins from the organism which have proven to be moderately successful. In this study, reverse vaccinology was used to predict potential H. somni vaccine candidates from genome sequences. In turn, these may protect animals against new strains circulating in the field. Whole genome sequencing of six recent clinical H. somni isolates was performed using an Illumina MiSeq and compared to six genomes from the 1980's. De novo assembly of crude whole genomes was completed using Geneious 6.1.7. Protein coding regions was predicted using Glimmer3. Scores from multiple web-based programs were utilized to evaluate the antigenicity of these predicted proteins which were finally ranked based on their surface exposure scores. A single new strain was selected for future vaccine development based on conservation of the protein candidates among all 12 isolates. A positive signal with convalescent serum for these antigens in western blots indicates in vivo recognition. In order to test the protective capacity of these antigens bovine animal trials are ongoing.


Assuntos
Vacinas Bacterianas/imunologia , Doenças dos Bovinos/prevenção & controle , Biologia Computacional/métodos , DNA Bacteriano/genética , Genoma Bacteriano , Infecções por Haemophilus/veterinária , Haemophilus somnus/imunologia , Animais , Antígenos de Bactérias/genética , Antígenos de Bactérias/imunologia , Proteínas de Bactérias/genética , Proteínas de Bactérias/imunologia , Western Blotting , Bovinos , Doenças dos Bovinos/microbiologia , Simulação por Computador , DNA Bacteriano/isolamento & purificação , Biblioteca Gênica , Infecções por Haemophilus/microbiologia , Infecções por Haemophilus/prevenção & controle , Haemophilus somnus/genética , Haemophilus somnus/isolamento & purificação , Haemophilus somnus/patogenicidade , Modelos Genéticos , Fases de Leitura Aberta/genética , Proteínas Recombinantes de Fusão/imunologia , Proteínas Recombinantes de Fusão/isolamento & purificação , Análise de Sequência de DNA , Virulência
15.
J Virol ; 89(8): 4598-611, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25673708

RESUMO

UNLABELLED: The major tegument protein of bovine herpesvirus 1 (BoHV-1), VP8, is essential for virus replication in cattle. VP8 is phosphorylated in vitro by casein kinase 2 (CK2) and BoHV-1 unique short protein 3 (US3). In this study, VP8 was found to be phosphorylated in both transfected and infected cells but was detected as a nonphosphorylated form in mature virions. This suggests that phosphorylation of VP8 is strictly controlled during different stages of the viral life cycle. The regulation and function of VP8 phosphorylation by US3 and CK2 were further analyzed. An in vitro kinase assay, site-directed mutagenesis, and liquid chromatography-mass spectrometry were used to identify the active sites for US3 and CK2. The two kinases phosphorylate VP8 at different sites, resulting in distinct phosphopeptide patterns. S(16) is a primary phosphoreceptor for US3, and it subsequently triggers phosphorylation at S(32). CK2 has multiple active sites, among which T(107) appears to be the preferred residue. Additionally, CK2 consensus motifs in the N terminus of VP8 are essential for phosphorylation. Based on these results, a nonphosphorylated VP8 mutant was constructed and used for further studies. In transfected cells phosphorylation was not required for nuclear localization of VP8. Phosphorylated VP8 appeared to recruit promyelocytic leukemia (PML) protein and to remodel the distribution of PML in the nucleus; however, PML protein did not show an association with nonphosphorylated VP8. This suggests that VP8 plays a role in resisting PML-related host antiviral defenses by redistributing PML protein and that this function depends on the phosphorylation of VP8. IMPORTANCE: The progression of VP8 phosphorylation over time and its function in BoHV-1 replication have not been characterized. This study demonstrates that activation of S(16) initiates further phosphorylation at S(32) by US3. Additionally, VP8 is phosphorylated by CK2 at several residues, with T(107) having the highest level of phosphorylation. Evidence for a difference in the phosphorylation status of VP8 in host cells and mature virus is presented for the first time. Phosphorylation was found to be a critical modification, which enables VP8 to attract and to redistribute PML protein in the nucleus. This might promote viral replication through interference with a PML-mediated antiviral defense. This study provides new insights into the regulation of VP8 phosphorylation and suggests a novel, phosphorylation-dependent function for VP8 in the life cycle of BoHV-1, which is important in view of the fact that VP8 is essential for virus replication in vivo.


Assuntos
Proteínas do Capsídeo/metabolismo , Bovinos/virologia , Herpesvirus Bovino 1/genética , Animais , Proteínas do Capsídeo/genética , Caseína Quinase II/metabolismo , Domínio Catalítico/genética , Cromatografia Líquida , Herpesvirus Bovino 1/metabolismo , Espectrometria de Massas , Mutagênese Sítio-Dirigida , Fosforilação , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Supressoras de Tumor/metabolismo , Vírion/metabolismo
16.
Mol Biotechnol ; 57(1): 58-64, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25173687

RESUMO

One of the impediments in the development of safe and cost effective vaccines for veterinary use has been the availability of appropriate delivery vehicle. We have chosen to develop and use bovine adenovirus (BAdV)-3 as vaccine delivery vector in cattle. Here, we describe the construction of recombinant E3 deleted BAdV-3 vectors expressing single vaccine antigen (BAV360; bovine respiratory syncytial virus G) or two vaccine antigens (BAV851; bovine herpesvirus-1gDt and bovine respiratory syncytial virus G). Recombinant proteins expressed by BAV360 or BAV851 were recognized by protein-specific monoclonal antibodies. Moreover, intranasal immunization of cotton rats with BAV360 (expressing a single vaccine antigen) or BAV851 (expressing two vaccine antigens) induced strong antigen-specific immune responses. These results suggest that single replication-competent BAdV-3 expressing vaccine antigens of two economically important respiratory pathogens of calves has potential to act as a feasible approach in the development of economically effective veterinary vaccines for cattle.


Assuntos
Herpesvirus Bovino 1/metabolismo , Imunidade , Proteínas Recombinantes/metabolismo , Vírus Sincicial Respiratório Bovino/metabolismo , Sigmodontinae/imunologia , Proteínas Virais de Fusão/metabolismo , Proteínas Virais/metabolismo , Animais , Formação de Anticorpos/imunologia , Bovinos , Linhagem Celular , Vacinas Virais/imunologia
17.
Vaccine ; 32(50): 6758-64, 2014 Nov 28.
Artigo em Inglês | MEDLINE | ID: mdl-25454860

RESUMO

Bovine viral diarrhea virus (BVDV) is still one of the most serious pathogens in cattle, meriting the development of improved vaccines. Recently, we developed a new adjuvant consisting of poly[di(sodium carboxylatoethylphenoxy)]-phosphazene (PCEP), either CpG ODN or poly(I:C), and an immune defense regulator (IDR) peptide. As this adjuvant has been shown to mediate the induction of robust, balanced immune responses, it was evaluated in an E2 subunit vaccine against BVDV in lambs and calves. The BVDV type 2 E2 protein was produced at high levels in a mammalian expression system and purified. When formulated with either CpG ODN or poly(I:C), together with IDR and PCEP, the E2 protein elicited high antibody titers and production of IFN-γ secreting cells in lambs. As the immune responses were stronger when poly(I:C) was used, the E2 protein with poly(I:C), IDR and PCEP was subsequently tested in cattle. Robust virus neutralizing antibodies as well as cell-mediated immune responses, including CD8(+) cytotoxic T cell (CTL) responses, were induced. The fact that CTL responses were demonstrated in calves vaccinated with an E2 protein subunit vaccine indicates that this adjuvant formulation promotes cross-presentation. Furthermore, upon challenge with a high dose of virulent BVDV-2, the vaccinated calves showed almost no temperature response, weight loss, leukopenia or virus replication, in contrast to the control animals, which had severe clinical disease. These data suggest that this E2 subunit formulation induces significant protection from BVDV-2 challenge, and thus is a promising BVDV vaccine candidate; in addition, the adjuvant platform has applications in bovine vaccines in general.


Assuntos
Adjuvantes Imunológicos/administração & dosagem , Doenças dos Bovinos/prevenção & controle , Vírus da Diarreia Viral Bovina Tipo 2/imunologia , Infecções por Pestivirus/veterinária , Doenças dos Ovinos/prevenção & controle , Proteínas do Envelope Viral/imunologia , Vacinas Virais/imunologia , Animais , Anticorpos Neutralizantes/sangue , Anticorpos Antivirais/sangue , Bovinos , Doenças dos Bovinos/patologia , Interferon gama/metabolismo , Leucócitos Mononucleares/imunologia , Infecções por Pestivirus/patologia , Infecções por Pestivirus/prevenção & controle , Ovinos , Doenças dos Ovinos/patologia , Linfócitos T Citotóxicos/imunologia , Vacinas de Subunidades Antigênicas/administração & dosagem , Vacinas de Subunidades Antigênicas/imunologia , Vacinas Virais/administração & dosagem
18.
Can J Vet Res ; 77(3): 161-9, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24101791

RESUMO

We evaluated the immunogenic and protective potential of a recombinant VapA/CpG oligodeoxynucleotide (ODN) 2395 vaccine in neonatal foals undergoing experimental Rhodococcus equi challenge. Foals (n = 8) were vaccinated by intramuscular injection on days 1 and 15 of the study; control foals (n = 7) received a phosphate-buffered saline (PBS) solution. All foals were challenged by intrabronchial administration of 5 × 106 R. equi 103⁺ on day 29. Bronchoalveolar lavages were done on days 15, 29, and 36 and total cell count, differential cell count, rVapA-stimulated cell proliferation and interferon (IFN)-γ mRNA expression determined. Clinical examination, complete blood (cell) counts, serology for VapA-specific antibodies, and culture of nasal and fecal swabs were done on days 1, 15, 29, 36, 43, and 50. Foals were humanely euthanized on day 50 and severity of pneumonia scored on a 4-point scale. Vaccination resulted in a significant increase in VapA-specific immunoglobulin (Ig) production, with total IgG and IgG(T) being increased by day 15. Expression of VapA-specific IFN-γ mRNA by BAL cells was increased in the vaccinated foals following challenge. Postmortem lung severity scores did not differ between groups. Two foals shed virulent R. equi in feces; however, real-time polymerase chain reaction (PCR) revealed the isolates to be different from the challenge strain.


Nous avons évalué le potentiel immunogène et protecteur d'un vaccin recombinant VapA/oligodéoxynucléotide CpG (ODN) 2395 chez des poulains nouveau-nés soumis à une infection défi par Rhodococcus equi. Les poulains (n = 8) étaient vaccinés par voie intramusculaire aux jours 1 et 15 de l'étude; les poulains témoins (n = 7) ont reçu une injection d'une solution de saline tamponnée (PBS). Tous les poulains ont été challengés par administration intra-bronchique de 5 × 106R. equi 103+ au jour 29. Des lavages broncho-alvéolaires (LBA) ont été effectués aux jours 15, 29 et 36 et on détermina le nombre total de cellules, un dénombrement cellulaire différentiel, la prolifération des cellules rVapA stimulées et l'expression d'ARNm de l'interféron (IFN)-γ. Un examen clinique, des comptages cellulaires sanguins complets, une analyse sérologique pour détecter les anticorps spécifiques contre VapA, et une culture d'écouvillons nasal et fécal ont été effectués aux jours 1, 15, 29, 36, 43 et 50. Les poulains ont été euthanasiés au jour 50 et la sévérité de la pneumonie notée sur une échelle de 4 points. La vaccination a causé une augmentation significative de la production d'immunoglobulines (Ig) spécifiquement dirigées contre VapA, les quantités totales d'IgG et d'IgG(T) ayant augmentées au jour 15. L'expression d'ARNm de l'IFN-γ spécifique au VapA par les cellules des LBA était augmentée chez les poulains vaccinés suite au challenge. Aucune différence ne fut notée dans les pointages de sévérité des lésions pulmonaires lors des examens post-mortem. Deux poulains excrétaient du R. equi virulent dans leurs fèces; toutefois, l'analyse par réaction d'amplification en chaîne par la polymérase (PCR) a démontré que ces isolats étaient différents de la souche utilisée pour le challenge.(Traduit par Docteur Serge Messier).


Assuntos
Infecções por Actinomycetales/veterinária , Vacinas Bacterianas/imunologia , Doenças dos Cavalos/microbiologia , Pneumonia/veterinária , Rhodococcus equi/imunologia , Vacinação/veterinária , Infecções por Actinomycetales/imunologia , Infecções por Actinomycetales/microbiologia , Infecções por Actinomycetales/prevenção & controle , Animais , Animais Recém-Nascidos , Proteínas de Bactérias/genética , Proteínas de Bactérias/imunologia , Vacinas Bacterianas/genética , Vacinas Bacterianas/normas , Líquido da Lavagem Broncoalveolar/citologia , Líquido da Lavagem Broncoalveolar/imunologia , Doenças dos Cavalos/imunologia , Doenças dos Cavalos/prevenção & controle , Cavalos , Interferon gama/genética , Interferon gama/imunologia , Modelos Lineares , Masculino , Oligodesoxirribonucleotídeos/genética , Oligodesoxirribonucleotídeos/imunologia , Pneumonia/imunologia , Pneumonia/microbiologia , Pneumonia/prevenção & controle , RNA Mensageiro/química , RNA Mensageiro/genética , Distribuição Aleatória , Reação em Cadeia da Polimerase em Tempo Real/veterinária , Rhodococcus equi/genética , Vacinação/normas , Vacinas Sintéticas/genética , Vacinas Sintéticas/imunologia , Vacinas Sintéticas/normas
19.
Prion ; 7(4): 301-11, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23787697

RESUMO

Transmissible spongiform encephalopathies (TSEs) are fatal neurodegenerative diseases that are based on the misfolding of a cellular prion protein (PrP(C)) into an infectious, pathological conformation (PrP(Sc)). There is proof-of-principle evidence that a prion vaccine is possible but this is tempered with concerns of the potential dangers associated with induction of immune responses to a widely-expressed self-protein. By targeting epitopes that are specifically exposed upon protein misfolding, our group developed a vaccine that induces PrP(Sc)-specific antibody responses. Here we consider the ability of this polyclonal antibody (SN6b) to bind to a mutant of PrP(C) associated with spontaneous prion disease. Polyclonal antibodies were selected to mimic the vaccination outcome and also explore all possible protein conformations of the recombinant bovine prion protein with mutation T194A [bPrP(T194A)]. This mutant is a homolog of the human T183A mutation of PrP(C) that is associated with early onset of familial dementia. With nanopore analysis, under non-denaturing conditions, we observed binding of the SN6b antibody to bPrP(T194A). This interaction was confirmed through ELISAs as well as immunoprecipitation of the recombinant and cellularly expressed forms of bPrP(T194A). This interaction did not promote formation of a protease resistant conformation of PrP in vitro. Collectively, these findings support the disease-specific approach for immunotherapy of prion diseases but also suggest that the concept of conformation-specific immunotherapy may be complicated in individuals who are genetically predisposed to PrP(C) misfolding.


Assuntos
Anticorpos/química , Mutação de Sentido Incorreto , Proteínas PrPC/química , Proteínas PrPSc/química , Substituição de Aminoácidos , Animais , Anticorpos/genética , Anticorpos/imunologia , Especificidade de Anticorpos , Bovinos , Demência/genética , Demência/imunologia , Demência/terapia , Células HEK293 , Humanos , Imunoterapia , Proteínas PrPC/genética , Proteínas PrPC/metabolismo , Proteínas PrPSc/genética , Proteínas PrPSc/imunologia , Dobramento de Proteína , Vacinas/química , Vacinas/genética , Vacinas/imunologia
20.
Avian Dis ; 56(1): 73-81, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-22545531

RESUMO

Inclusion body hepatitis (IBH) is one of the major global disease problems, causing significant economic losses to poultry industry of the United States and Canada. The disease is characterized by its sudden onset and high mortalities. Amongst different serotypes of fowl adenoviruses (FAdVs) associated with IBH, serotype 8 of group I FAdV has been isolated from majority of IBH cases. In present studies, we isolated a FAdV from morbid liver of a 17-day-old broiler from a Saskatchewan broiler farm. This newly isolated virus was designated as IBHV(SK). However, based on the sequence analysis of the L1 region of the hexon gene, the IBHV(SK) may be classified as FAdV 8b strain 764. These studies describe for the first time the complete hexon gene sequence of FAdV serotype 8b. Experimental infection of 2-day-old (n = 48) and 2-wk-old (n = 56) chicks caused 83% and 43% mortalities, respectively. Determination of the complete hexon gene sequence of IBHV(SK) with establishment of a disease model in chickens will facilitate the development of type-specific diagnostic reagents and assays for the evaluation of potential experimental vaccines against pathogenic FAdV infections.


Assuntos
Infecções por Adenoviridae/veterinária , Aviadenovirus/classificação , Aviadenovirus/isolamento & purificação , Galinhas , Hepatite Viral Animal/patologia , Fígado/patologia , Doenças das Aves Domésticas/patologia , Infecções por Adenoviridae/epidemiologia , Infecções por Adenoviridae/mortalidade , Infecções por Adenoviridae/patologia , Animais , Aviadenovirus/química , Aviadenovirus/genética , Proteínas do Capsídeo/química , Proteínas do Capsídeo/genética , Hepatite Viral Animal/epidemiologia , Hepatite Viral Animal/mortalidade , Corpos de Inclusão Viral/patologia , Corpos de Inclusão Viral/virologia , Fígado/citologia , Fígado/virologia , Microscopia Eletrônica de Transmissão/veterinária , Dados de Sequência Molecular , Filogenia , Doenças das Aves Domésticas/epidemiologia , Doenças das Aves Domésticas/mortalidade , Saskatchewan/epidemiologia , Alinhamento de Sequência , Análise de Sequência de DNA , Análise de Sequência de Proteína
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA