Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
1.
J Virol ; 93(6)2019 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-30626671

RESUMEN

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.


Asunto(s)
Proteínas de la Cápside/metabolismo , Infecciones por Herpesviridae/metabolismo , Herpesvirus Bovino 1/metabolismo , Metabolismo de los Lípidos/fisiología , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Virales/metabolismo , Animales , Células COS , Bovinos , Línea Celular , Membrana Celular/metabolismo , Membrana Celular/virología , Núcleo Celular/metabolismo , Núcleo Celular/virología , Chlorocebus aethiops , Citoplasma/metabolismo , Citoplasma/virología , Aparato de Golgi/virología , Infecciones por Herpesviridae/virología , Humanos , Señales de Localización Nuclear/metabolismo , Fosforilación , Virión/metabolismo , Replicación Viral/fisiología
2.
J Gen Virol ; 98(7): 1831-1842, 2017 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-28675355

RESUMEN

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.


Asunto(s)
Enfermedades de los Bovinos/genética , Herpesvirus Bovino 1/fisiología , Factores Reguladores del Interferón/genética , Interferones/genética , Infecciones del Sistema Respiratorio/genética , Animales , Bovinos , Enfermedades de los Bovinos/inmunología , Enfermedades de los Bovinos/virología , Herpesvirus Bovino 1/genética , Factores Reguladores del Interferón/inmunología , Interferones/inmunología , Infecciones del Sistema Respiratorio/inmunología , Infecciones del Sistema Respiratorio/virología , Replicación Viral
3.
J Virol ; 90(10): 4889-4904, 2016 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-26889034

RESUMEN

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.


Asunto(s)
Proteínas de la Cápside/inmunología , Proteínas de la Cápside/metabolismo , Herpesvirus Bovino 1/metabolismo , Interferón beta/metabolismo , Factor de Transcripción STAT1/metabolismo , Transducción de Señal , Animales , Proteínas de la Cápside/química , Proteínas de la Cápside/genética , Chlorocebus aethiops , Citoplasma/metabolismo , Regulación hacia Abajo , Interacciones Huésped-Patógeno , Humanos , Interferón beta/inmunología , Interferón beta/farmacología , Mutación , Señales de Localización Nuclear/metabolismo , Fosforilación , Factor de Transcripción STAT1/inmunología , Homología de Secuencia de Aminoácido , Transducción de Señal/genética , Ubiquitinación , Células Vero , Virión/metabolismo , Replicación Viral
4.
J Virol ; 90(9): 4427-4440, 2016 May.
Artículo en Inglés | MEDLINE | ID: mdl-26889039

RESUMEN

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.


Asunto(s)
Proteínas de la Cápside/metabolismo , ADN Viral , Infecciones por Herpesviridae/virología , Herpesvirus Bovino 1/fisiología , Ensamble de Virus , Replicación Viral , Animales , Proteínas de la Cápside/genética , Bovinos , Línea Celular , Herpesvirus Bovino 1/aislamiento & purificación , Herpesvirus Bovino 1/ultraestructura , Mutación , Fosforilación , Transporte de Proteínas , Recombinación Genética , Liberación del Virus
5.
J Virol ; 89(8): 4598-611, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25673708

RESUMEN

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.


Asunto(s)
Proteínas de la Cápside/metabolismo , Bovinos/virología , Herpesvirus Bovino 1/genética , Animales , Proteínas de la Cápside/genética , Quinasa de la Caseína II/metabolismo , Dominio Catalítico/genética , Cromatografía Liquida , Herpesvirus Bovino 1/metabolismo , Espectrometría de Masas , Mutagénesis Sitio-Dirigida , Fosforilación , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Virión/metabolismo
6.
Virol J ; 13: 57, 2016 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-27036203

RESUMEN

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.


Asunto(s)
Antígenos Virales/inmunología , Infecciones por Coronavirus/veterinaria , Virus de la Diarrea Epidémica Porcina/inmunología , Glicoproteína de la Espiga del Coronavirus/inmunología , Vacunas Virales/inmunología , Adyuvantes Inmunológicos/administración & dosificación , Animales , Anticuerpos Neutralizantes/análisis , Anticuerpos Antivirales/análisis , Antígenos Virales/genética , Calostro/inmunología , Infecciones por Coronavirus/patología , Infecciones por Coronavirus/prevención & control , Ensayo de Inmunoadsorción Enzimática , Femenino , Inyecciones Intramusculares , Pruebas de Neutralización , Virus de la Diarrea Epidémica Porcina/genética , Embarazo , Suero/inmunología , Glicoproteína de la Espiga del Coronavirus/genética , Porcinos , Resultado del Tratamiento , Vacunas de Subunidad/administración & dosificación , Vacunas de Subunidad/genética , Vacunas de Subunidad/inmunología , Vacunas Sintéticas/administración & dosificación , Vacunas Sintéticas/genética , Vacunas Sintéticas/inmunología , Vacunas Virales/administración & dosificación , Vacunas Virales/genética
7.
Biologicals ; 43(6): 444-51, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26460173

RESUMEN

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.


Asunto(s)
Vacunas Bacterianas/inmunología , Enfermedades de los Bovinos/prevención & control , Biología Computacional/métodos , ADN Bacteriano/genética , Genoma Bacteriano , Infecciones por Haemophilus/veterinaria , Haemophilus somnus/inmunología , Animales , Antígenos Bacterianos/genética , Antígenos Bacterianos/inmunología , Proteínas Bacterianas/genética , Proteínas Bacterianas/inmunología , Western Blotting , Bovinos , Enfermedades de los Bovinos/microbiología , Simulación por Computador , ADN Bacteriano/aislamiento & purificación , Biblioteca de Genes , Infecciones por Haemophilus/microbiología , Infecciones por Haemophilus/prevención & control , Haemophilus somnus/genética , Haemophilus somnus/aislamiento & purificación , Haemophilus somnus/patogenicidad , Modelos Genéticos , Sistemas de Lectura Abierta/genética , Proteínas Recombinantes de Fusión/inmunología , Proteínas Recombinantes de Fusión/aislamiento & purificación , Análisis de Secuencia de ADN , Virulencia
8.
Avian Dis ; 56(1): 73-81, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22545531

RESUMEN

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.


Asunto(s)
Infecciones por Adenoviridae/veterinaria , Aviadenovirus/clasificación , Aviadenovirus/aislamiento & purificación , Pollos , Hepatitis Viral Animal/patología , Hígado/patología , Enfermedades de las Aves de Corral/patología , Infecciones por Adenoviridae/epidemiología , Infecciones por Adenoviridae/mortalidad , Infecciones por Adenoviridae/patología , Animales , Aviadenovirus/química , Aviadenovirus/genética , Proteínas de la Cápside/química , Proteínas de la Cápside/genética , Hepatitis Viral Animal/epidemiología , Hepatitis Viral Animal/mortalidad , Cuerpos de Inclusión Viral/patología , Cuerpos de Inclusión Viral/virología , Hígado/citología , Hígado/virología , Microscopía Electrónica de Transmisión/veterinaria , Datos de Secuencia Molecular , Filogenia , Enfermedades de las Aves de Corral/epidemiología , Enfermedades de las Aves de Corral/mortalidad , Saskatchewan/epidemiología , Alineación de Secuencia , Análisis de Secuencia de ADN , Análisis de Secuencia de Proteína
9.
Dev Comp Immunol ; 127: 104271, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34600023

RESUMEN

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.


Asunto(s)
Leucocitos , Norepinefrina , Animales , Bovinos , Epinefrina/metabolismo , Expresión Génica , Leucocitos/metabolismo , Norepinefrina/metabolismo , Receptores Adrenérgicos beta/metabolismo
10.
Cell Tissue Res ; 343(1): 121-30, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-20809414

RESUMEN

Analysis of the genomes of two distantly related bird species, chicken and zebra finch (divergence of about 100 million years), indicate that there are ten avian toll-like receptors and that five of these, TLR2a, 2b, 3, 4, 5 and 7, are clear orthologs to TLRs found in mammals. Duplication of genes has led to TLR1La and 1Lb, TLR2a and 2b, and two TLR7s in the zebra finch. Avian TLR21 may be orthologous to TLR21 found in fish and amphibians, and avian TLR15, which is phylogenetically related to the TLR2 family, appears to be unique to avian species. While TLR2 is conserved between mammalian and avian species, the other TLR2 family members evolved independently. Dimerization between either of the two avian TLR2 species and TLR1La or 1Lb permits recognition of the same broad range of molecules as recognized by mammalian TLR2 dimerized with either TLR1, 6 and 10. Similarly, while TLR9 has been lost from the avian genome, DNA high in unmethylated CpG motifs is immunostimulatory through avian TLR21 which is absent in mammals. Thus, while some TLR members were commonly retained in both mammals and birds, others were separately lost or gained, or diverged independently; but broadly speaking the TLRs of the two classes of vertebrates evolved to recognize very similar spectra of microbial products. Components of downstream TLR signaling are also mostly conserved but with some losses in avian species; notably, TRAM is absent in avian genomes and, hence, the TRIF/TRAM-dependent signaling pathway utilized by mammals in LPS activation appears to be absent in birds.


Asunto(s)
Aves/inmunología , Receptores Toll-Like/inmunología , Animales , Receptores Toll-Like/genética
11.
Vet Immunol Immunopathol ; 237: 110256, 2021 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-33971523

RESUMEN

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.


Asunto(s)
Vacunas Bacterianas/inmunología , Infecciones por Desulfovibrionaceae/prevención & control , Inmunogenicidad Vacunal , Lawsonia (Bacteria)/inmunología , Enfermedades de los Porcinos/prevención & control , Animales , Animales Recién Nacidos , Anticuerpos Antibacterianos/inmunología , Infecciones por Desulfovibrionaceae/inmunología , Femenino , Embarazo , Porcinos , Enfermedades de los Porcinos/microbiología , Vacunas Combinadas/inmunología , Vacunas de Subunidad/inmunología
12.
J Infect Dev Ctries ; 15(5): 653-656, 2021 05 31.
Artículo en Inglés | MEDLINE | ID: mdl-34106888

RESUMEN

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.


Asunto(s)
Vacunas contra la COVID-19/inmunología , COVID-19/inmunología , Inmunidad Humoral/efectos de los fármacos , Anticuerpos Neutralizantes/inmunología , COVID-19/prevención & control , Estudios de Casos y Controles , Femenino , Humanos , Masculino , SARS-CoV-2
13.
J Gen Virol ; 91(Pt 6): 1388-95, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20130133

RESUMEN

Hepatitis C virus genotype-3a (HCV-3a) is directly linked to the development of steatosis. We previously showed that, through sterol regulatory element binding protein-1 (SREBP-1), HCV-3a core protein upregulates the promoter activity of fatty acid synthase, a major enzyme involved in de novo lipid synthesis. In this study, we investigated whether HCV-3a core can activate SREBP-1 and studied the role of phosphoinositide 3-kinase (PI3K)-Akt-2 pathway in modulating SREBP-1 activity by HCV-3a core. To determine whether HCV-3a core could activate SREBP-1, the level of mature SREBP-1 was analysed by Western blotting. Our results showed that the level of mature SREBP-1 was enhanced by HCV-3a core protein after transient expression and in the chimeric HCV-3a core/1b replicon cells in comparison to controls. To investigate the role of the PI3K-Akt-2 pathway in SREBP-1 activation by HCV-3a core, PI3K and Akt-2 activity was inhibited by using the chemical inhibitor LY294002, a dominant-negative Akt-2 plasmid, or knockdown of Akt-2 by small hairpin RNA. Our results showed that inhibition of PI3K and Akt-2 was associated with reduced SREBP-1 activation by HCV-3a core. These results indicate a role for PI3K and Akt-2 in increasing SREBP-1 activity by HCV-3a core protein and provide a mechanism of steatosis caused by HCV.


Asunto(s)
Hepacivirus/patogenicidad , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteína 1 de Unión a los Elementos Reguladores de Esteroles/biosíntesis , Proteínas del Núcleo Viral/fisiología , Western Blotting , Línea Celular , Cromonas/farmacología , Inhibidores Enzimáticos/farmacología , Técnicas de Silenciamiento del Gen , Genotipo , Hepacivirus/clasificación , Hepacivirus/genética , Hepatocitos/virología , Humanos , Morfolinas/farmacología , Inhibidores de las Quinasa Fosfoinosítidos-3 , Proteínas Proto-Oncogénicas c-akt/antagonistas & inhibidores
14.
Mol Immunol ; 46(5): 978-90, 2009 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-18995910

RESUMEN

Toll-like receptors (TLRs) are a family of highly conserved germline-encoded pattern-recognition receptors (PRRs) and are essential for host immune response. Little is known regarding the activation mechanism of TLRs especially of the TLR7/8/9 subfamily. Here we cloned and characterized bovine TLR8 (bTLR8) and found that it is highly responsive to two TLR7 ligands, imiquimod and gardiquimod, in transfected cell lines. Using the transfected cell lines as model systems, we analyzed by mutagenesis the roles of potentially important regions of bTLR8 in receptor signaling: 5 insertions in leucine rich repeats (LRRs) of the ectodomain (ECD), 9 N-glycosylation sites, all the cysteines, an aspartate conserved between TLRs, the transmembrane (TM) domain and different cytoplasmic regions. All 5 insertions, 2 N-glycosylation sites, most of the cysteines, the conserved aspartate, the TM and each of the cytoplasmic regions are essential for TLR8 signaling. We also showed that bTLR8 undergoes dimerization/self-association which was not affected by imidazoquinoline stimulation. This observation together with kinetics of activation suggested that a ligand-induced dimer conformational switch is mainly responsible for TLR8 activation. All the TLR8 signaling essential sites were examined for their requirement in dimerization; no single mutation or group of mutations affected the dimerization. However, among the impaired TLR8 mutants, all those containing mutations in the transmembrane or cytoplasmic regions and only two within the ECD (N515D and D536A) showed dominant negative inhibition to wild type receptor, whereas the others, all within the ECD, did not compete with wild type TLR8. A model for activation of bTLR8 was described based on these data.


Asunto(s)
Bovinos/inmunología , Receptor Toll-Like 8/inmunología , Animales , Células COS , Bovinos/genética , Chlorocebus aethiops , Clonación Molecular , Dimerización , Glicosilación , Humanos , Cinética , Ligandos , Mutagénesis , Mutación , Estructura Terciaria de Proteína/fisiología , Receptor Toll-Like 7/genética , Receptor Toll-Like 7/inmunología , Receptor Toll-Like 8/genética , Receptor Toll-Like 9/genética , Receptor Toll-Like 9/inmunología
15.
Mol Immunol ; 46(5): 884-92, 2009 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19019441

RESUMEN

Toll-like receptors (TLRs), a family of highly conserved germline-encoded pattern-recognition receptors (PRRs), are essential for the host immune response. The cellular localization of TLR proteins determines the access to certain sources of ligands and thus the triggering of downstream cellular signaling. The TLR7/8/9 subfamily proteins are localized intracellularly but the molecular elements determining the cellular localization of these proteins are not fully understood. Here we demonstrated that the bovine TLR8 (bTLR8) protein is localized in the ER cellular compartment of transfected cells before and after cell activation. Using chimeric constructs, we showed that the bTLR8 transmembrane (TM) and cytoplasmic (CP) regions could direct the bovine herpesvirus 1 (BHV-1) glycoprotein D (gD) extracellular domain (ECD) to an intracellular localization. Furthermore, the bTLR8 TM, the linker region between the TM and TIR domains, and the TIR-tail region all partially contributed to the intracellular localization. However, truncation of the bTLR8 with the TM and CP regions removed did not alter its intracellular localization, suggesting that ectodomain (ECD) itself contains intracellular information. Indeed, the bTLR8 ECD also targeted the gD ECD to the intracellular localization. Our results suggest that multiple regions, including ECD, TM, linker and TIR-tail regions of bTLR8, are involved in determining the localization of cellular ER compartment.


Asunto(s)
Bovinos/metabolismo , Retículo Endoplásmico/metabolismo , Transducción de Señal/fisiología , Receptor Toll-Like 8/metabolismo , Animales , Células COS , Bovinos/genética , Bovinos/inmunología , Chlorocebus aethiops , Retículo Endoplásmico/genética , Retículo Endoplásmico/inmunología , Mapeo Peptídico/métodos , Estructura Terciaria de Proteína/fisiología , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/inmunología , Proteínas Recombinantes de Fusión/metabolismo , Receptor Toll-Like 8/genética , Receptor Toll-Like 8/inmunología , Proteínas Virales/genética , Proteínas Virales/inmunología , Proteínas Virales/metabolismo
16.
Vaccine ; 38(10): 2387-2395, 2020 02 28.
Artículo en Inglés | MEDLINE | ID: mdl-32014270

RESUMEN

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.


Asunto(s)
Adyuvantes Inmunológicos/administración & dosificación , Glicoproteínas Hemaglutininas del Virus de la Influenza/inmunología , Vacunas contra la Influenza/inmunología , Infecciones por Orthomyxoviridae , Animales , Anticuerpos Antivirales/sangre , Glicoproteínas Hemaglutininas del Virus de la Influenza/administración & dosificación , Subtipo H5N1 del Virus de la Influenza A , Ratones , Ratones Endogámicos BALB C , Infecciones por Orthomyxoviridae/prevención & control , Células TH1/inmunología , Células Th2/inmunología
17.
Virus Res ; 261: 37-49, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30550895

RESUMEN

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.


Asunto(s)
Proteínas de la Cápside/metabolismo , Chaperonina 60/metabolismo , Herpesvirus Bovino 1/fisiología , Interacciones Huésped-Patógeno , Mitocondrias/patología , Mapeo de Interacción de Proteínas , Adenosina Trifosfato/análisis , Animales , Proteínas de la Cápside/análisis , Bovinos , Línea Celular , Chaperonina 60/análisis , Células Epiteliales/virología , Humanos , Inmunoprecipitación , Potenciales de la Membrana , Mitocondrias/química , Membranas Mitocondriales/fisiología , Unión Proteica
18.
Vet Microbiol ; 235: 270-279, 2019 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-31383312

RESUMEN

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.


Asunto(s)
Anticuerpos Antibacterianos/inmunología , Anticuerpos Neutralizantes/inmunología , Proteínas Bacterianas/inmunología , Vacunas Bacterianas/inmunología , Infecciones por Desulfovibrionaceae/veterinaria , Lawsonia (Bacteria)/inmunología , Animales , Proteínas Bacterianas/genética , Western Blotting , Línea Celular , Biología Computacional , Infecciones por Desulfovibrionaceae/inmunología , Infecciones por Desulfovibrionaceae/prevención & control , Femenino , Intestinos/citología , Intestinos/microbiología , Espectrometría de Masas , Proteómica , Conejos , Proteínas Recombinantes/inmunología , Porcinos , Enfermedades de los Porcinos/inmunología , Enfermedades de los Porcinos/microbiología , Enfermedades de los Porcinos/prevención & control , Vacunas de Subunidad/inmunología
19.
Nat Commun ; 9(1): 3199, 2018 08 10.
Artículo en Inglés | MEDLINE | ID: mdl-30097581

RESUMEN

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.


Asunto(s)
Antivirales/farmacología , Núcleo Celular/metabolismo , Proteína 58 DEAD Box/metabolismo , Inmunidad Innata , Replicación Viral/fisiología , Células A549 , Animales , Compartimento Celular/efectos de los fármacos , Núcleo Celular/efectos de los fármacos , Perros , Células HEK293 , Humanos , Virus de la Influenza A/efectos de los fármacos , Células de Riñón Canino Madin Darby , Unión Proteica/efectos de los fármacos , ARN Viral/metabolismo , Receptores Inmunológicos , Ribonucleoproteínas/metabolismo , Transducción de Señal/efectos de los fármacos
20.
Vaccine ; 35(27): 3466-3473, 2017 06 14.
Artículo en Inglés | MEDLINE | ID: mdl-28527686

RESUMEN

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.


Asunto(s)
Adyuvantes Inmunológicos/administración & dosificación , Diarrea Mucosa Bovina Viral/prevención & control , Virus de la Diarrea Viral Bovina Tipo 1/inmunología , Inmunidad Innata , Proteínas del Envoltorio Viral/inmunología , Vacunas Virales/inmunología , Animales , Anticuerpos Neutralizantes/sangre , Anticuerpos Antivirales/sangre , Peso Corporal , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Bovinos , Fiebre/prevención & control , Inyecciones Intradérmicas , Leucopenia/prevención & control , Vacunas Virales/administración & dosificación , Esparcimiento de Virus
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA