Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 42
Filtrar
1.
J Virol ; 96(23): e0101022, 2022 12 14.
Artigo em Inglês | MEDLINE | ID: mdl-36416585

RESUMO

Bovine herpesvirus 1 (BoHV-1), an important pathogen of cattle, establishes lifelong latency in sensory neurons within trigeminal ganglia (TG) after acute infection. The BoHV-1 latency-reactivation cycle, like other alphaherpesvirinae subfamily members, is essential for viral persistence and transmission. Notably, cells within pharyngeal tonsil (PT) also support a quiescent or latent BoHV-1 infection. The synthetic corticosteroid dexamethasone, which mimics the effects of stress, consistently induces BoHV-1 reactivation from latency allowing early stages of viral reactivation to be examined in the natural host. Based on previous studies, we hypothesized that stress-induced cellular factors trigger expression of key viral transcriptional regulatory genes. To explore this hypothesis, RNA-sequencing studies compared viral gene expression in PT during early stages of dexamethasone-induced reactivation from latency. Strikingly, RNA encoding infected cell protein 4 (bICP4), which is translated into an essential viral transcriptional regulatory protein, was detected 30 min after dexamethasone treatment. Ninety minutes after dexamethasone treatment bICP4 and, to a lesser extent, bICP0 RNA were detected in PT. All lytic cycle viral transcripts were detected within 3 h after dexamethasone treatment. Surprisingly, the latency related (LR) gene, the only viral gene abundantly expressed in latently infected TG neurons, was not detected in PT during latency. In TG neurons, bICP0 and the viral tegument protein VP16 are expressed before bICP4 during reactivation, suggesting distinct viral regulatory genes mediate reactivation from latency in PT versus TG neurons. Finally, these studies confirm PT is a biologically relevant site for BoHV-1 latency, reactivation from latency, and virus transmission. IMPORTANCE BoHV-1, a neurotropic herpesvirus, establishes, maintains, and reactivates from latency in neurons. BoHV-1 DNA is also detected in pharyngeal tonsil (PT) from latently infected calves. RNA-sequencing studies revealed the viral infected cell protein 4 (bICP4) RNA was expressed in PT of latently infected calves within 30 min after dexamethasone was used to initiate reactivation. As expected, bICP4 RNA was not detected during latency. All lytic cycle viral genes were expressed within 3 h after dexamethasone treatment. Conversely, bICP0 and the viral tegument protein VP16 are expressed prior to bICP4 in trigeminal ganglionic neurons during reactivation. The viral latency related gene, which is abundantly expressed in latently infected neurons, was not abundantly expressed in PT during latency. These studies provide new evidence PT is a biologically relevant site for BoHV-1 latency and reactivation. Finally, we predict other alphaherpesvirinae subfamily members utilize PT as a site for latency and reactivation.


Assuntos
Tonsila Faríngea , Infecções por Herpesviridae , Herpesvirus Bovino 1 , Proteínas do Envelope Viral , Ativação Viral , Animais , Bovinos , Tonsila Faríngea/virologia , Dexametasona/farmacologia , Etoposídeo/farmacologia , Herpesvirus Bovino 1/fisiologia , RNA/metabolismo , Gânglio Trigeminal , Proteínas Virais/genética , Proteínas Virais/metabolismo , Latência Viral , Proteínas do Envelope Viral/metabolismo
2.
Int J Mol Sci ; 23(23)2022 Nov 29.
Artigo em Inglês | MEDLINE | ID: mdl-36499292

RESUMO

Polypeptide variation encoded by the ovine transmembrane protein 154 gene (TMEM154) is associated with susceptibility to ovine lentivirus, the causative agent of Ovine Progressive Pneumonia (OPP) and Visna/Maedi. Our aim was to compare the four most prevalent TMEM154 haplotypes on the incidence of infection and ewe productivity during natural multiyear virus exposure. Prospective cohort studies were designed to test gene action and estimate effects of TMEM154 haplotypes encoding distinctive variant residues: K35 ("1"), I70 ("2"), ancestral ("3"), and A4del/M44 ("4"). Exposure consisted of co-mingling infected ewes at a rate greater than 30% with serological status evaluated every four months. For ewes with one or two copies of the highly susceptible haplotypes "2" and "3", the infection prevalence steadily increased to nearly 100% at 55 months. Haplotypes "2" and "3" were equally susceptible and dominant to haplotype "1". A difference was not detected (p < 0.53) in the magnitude of effect with haplotype combinations of "1" and "4". The ewe infection prevalence with "1,1"; "1,4"; and "4,4" was 10% to 40% at 55 months. The latter suggested that two copies of the K35 amino acid substitution ("1") were as effective as a homozygous TMEM154 "knockout" with the frame-shift deletion mutation ("4") in reducing infection susceptibility. When considering ewe reproductive performance, a difference was not detected when comparing haplotypes "2", and "3" to each other, or "1" and "4" to each other. Our study indicated that ewes with two copies of the severely truncated versions of TMEM154 ("4,4") had normal lamb productivity. Without complete understanding of the natural function of TMEM154 our recommendations to producers interested in using TMEM154 selection to reduce their flock's genetic predisposition to OPP are encouraged to increase the frequency of TMEM154 haplotype K35 ("1") since it encodes a full-length protein with minimal difference to the ancestral polypeptide.


Assuntos
Pneumonia Intersticial Progressiva dos Ovinos , Doenças dos Ovinos , Ovinos , Animais , Feminino , Lentivirus/genética , Estudos Prospectivos , Pneumonia Intersticial Progressiva dos Ovinos/genética , Haplótipos , Predisposição Genética para Doença
3.
J Virol ; 92(7)2018 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-29321317

RESUMO

Sensory neurons in trigeminal ganglia (TG) of calves latently infected with bovine herpesvirus 1 (BoHV-1) abundantly express latency-related (LR) gene products, including a protein (ORF2) and two micro-RNAs. Recent studies in mouse neuroblastoma cells (Neuro-2A) demonstrated ORF2 interacts with ß-catenin and a ß-catenin coactivator, high-mobility group AT-hook 1 (HMGA1) protein, which correlates with increased ß-catenin-dependent transcription and cell survival. ß-Catenin and HMGA1 are readily detected in a subset of latently infected TG neurons but not TG neurons from uninfected calves or reactivation from latency. Consequently, we hypothesized that the Wnt/ß-catenin signaling pathway is differentially expressed during the latency and reactivation cycle and an active Wnt pathway promotes latency. RNA-sequencing studies revealed that 102 genes associated with the Wnt/ß-catenin signaling pathway were differentially expressed in TG during the latency-reactivation cycle in calves. Wnt agonists were generally expressed at higher levels during latency, but these levels decreased during dexamethasone-induced reactivation. The Wnt agonist bone morphogenetic protein receptor 2 (BMPR2) was intriguing because it encodes a serine/threonine receptor kinase that promotes neuronal differentiation and inhibits cell death. Another differentially expressed gene encodes a protein kinase (Akt3), which is significant because Akt activity enhances cell survival and is linked to herpes simplex virus 1 latency and neuronal survival. Additional studies demonstrated ORF2 increased Akt3 steady-state protein levels and interacted with Akt3 in transfected Neuro-2A cells, which correlated with Akt3 activation. Conversely, expression of Wnt antagonists increased during reactivation from latency. Collectively, these studies suggest Wnt signaling cooperates with LR gene products, in particular ORF2, to promote latency.IMPORTANCE Lifelong BoHV-1 latency primarily occurs in sensory neurons. The synthetic corticosteroid dexamethasone consistently induces reactivation from latency in calves. RNA sequencing studies revealed 102 genes associated with the Wnt/ß-catenin signaling pathway are differentially regulated during the latency-reactivation cycle. Two protein kinases associated with the Wnt pathway, Akt3 and BMPR2, were expressed at higher levels during latency but were repressed during reactivation. Furthermore, five genes encoding soluble Wnt antagonists and ß-catenin-dependent transcription inhibitors were induced during reactivation from latency. These findings are important because Wnt, BMPR2, and Akt3 promote neurogenesis and cell survival, processes crucial for lifelong viral latency. In transfected neuroblastoma cells, a viral protein expressed during latency (ORF2) interacts with and enhances Akt3 protein kinase activity. These findings provide insight into how cellular factors associated with the Wnt signaling pathway cooperate with LR gene products to regulate the BoHV-1 latency-reactivation cycle.


Assuntos
Receptores de Proteínas Morfogenéticas Ósseas Tipo II/biossíntese , Regulação Enzimológica da Expressão Gênica , Herpesvirus Bovino 1/fisiologia , Proteínas Proto-Oncogênicas c-akt/biossíntese , Células Receptoras Sensoriais/imunologia , Gânglio Trigeminal/enzimologia , Ativação Viral/fisiologia , Latência Viral/fisiologia , Via de Sinalização Wnt , Animais , Receptores de Proteínas Morfogenéticas Ósseas Tipo II/genética , Bovinos , Sobrevivência Celular , Proteínas Proto-Oncogênicas c-akt/genética , Células Receptoras Sensoriais/patologia , Células Receptoras Sensoriais/virologia , Gânglio Trigeminal/patologia , Gânglio Trigeminal/virologia
4.
BMC Vet Res ; 15(1): 161, 2019 May 22.
Artigo em Inglês | MEDLINE | ID: mdl-31118011

RESUMO

BACKGROUND: Bovine coronavirus (BCV) is associated with respiratory infections in cattle of all ages; however, a temporal study to evaluate the effect of BCV immunity on virus shedding and bovine respiratory disease (BRD) incidence in pre-weaned beef calves has not been reported. Thus, we report here a prospective study in three herds of crossbred beef calves (n = 817) with endemic BCV. Serial blood samples for measurement of serum anti-BCV antibody titers and nasal swabs for detection of BCV and other common viral and bacterial BRD pathogens were collected from all calves or subsets of calves at predetermined times from birth through weaning. The calves were monitored for BRD and those that developed signs of respiratory disease were sampled for diagnostic testing. To discover additional risk factors that could have influenced BRD development, sequence analysis of the BCV strain(s) circulating in each herd, and the prevalence of common opportunistic bacterial pathogens in the upper respiratory tract of sick and apparently healthy cattle were also evaluated. RESULTS: Two hundred forty-eight of the 817 study calves (30.4%) were treated for BRD prior to weaning; 246 of those were from a single herd involved in two outbreaks of BRD leading to mass treatment of all calves in that group. Molecular diagnostic testing found BCV and Histophilus somni in nasal swabs taken at the time of BRD treatment. Between herd analyses revealed anti-BCV serum antibody abundance did not associate with the incidence of BRD or BCV shedding, though these measurements may have been hindered by the long periods between sample collections. Analysis of the BCV spike gene hypervariable region revealed four polymorphisms in 15 isolates from the three herds, making strain variation unlikely to account for differences in treatment rates between herds. Persistent or recurrent shedding episodes of BCV occurred in some animals treated for BRD. CONCLUSION: Co-detection of BCV and H. somni at the time of the disease outbreak suggests that these pathogens contributed to disease pathogenesis. Developing appropriate control measures for respiratory BCV infections may help decrease the incidence of pre-weaning BRD. The role of antibodies in protection must still be further defined.


Assuntos
Doenças dos Bovinos/imunologia , Infecções por Coronavirus/veterinária , Coronavirus Bovino/imunologia , Imunidade Humoral/imunologia , Animais , Anticorpos Antivirais/sangue , Bovinos , Doenças dos Bovinos/microbiologia , Coinfecção/veterinária , Infecções por Coronavirus/diagnóstico , Infecções por Coronavirus/imunologia , Infecções por Coronavirus/microbiologia , Coronavirus Bovino/genética , Pasteurellaceae/fisiologia , Polimorfismo Genético , Eliminação de Partículas Virais
5.
J Virol ; 91(5)2017 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-28003484

RESUMO

The latency-related (LR) RNA encoded by bovine herpesvirus 1 (BoHV-1) is abundantly expressed in latently infected sensory neurons. Although the LR gene encodes several products, ORF2 appears to mediate important steps during the latency-reactivation cycle because a mutant virus containing stop codons at the amino terminus of ORF2 does not reactivate from latency in calves. We recently found that the Wnt/ß-catenin signaling pathway is regulated during the BoHV-1 latency-reactivation cycle (Y. Liu, M. Hancock, A. Workman, A. Doster, and C. Jones, J Virol 90:3148-3159, 2016). In the present study, a ß-catenin coactivator, high-mobility group AT-hook 1 protein (HMGA1), was detected in significantly more neurons in the trigeminal ganglia of latently infected calves than in those of uninfected calves. Consequently, we hypothesized that HMGA1 cooperates with ORF2 and ß-catenin to maintain latency. In support of this hypothesis, coimmunoprecipitation studies demonstrated that ORF2 stably interacts with a complex containing ß-catenin and/or HMGA1 in transfected mouse neuroblastoma (Neuro-2A) cells. Confocal microscopy provided evidence that ORF2 was relocalized by HMGA1 and ß-catenin in Neuro-2A cells. ORF2 consistently enhanced the ability of HMGA1 to stimulate ß-catenin-dependent transcription, suggesting that interactions between ORF2 and a complex containing ß-catenin and HMGA1 have functional significance. An ORF2 stop codon mutant, an ORF2 nuclear localization mutant, or a mutant lacking the 5 protein kinase A or C phosphorylation sites interfered with its ability to stimulate ß-catenin-dependent transcription. Since the canonical Wnt/ß-catenin signaling pathway promotes neurogenesis (synapse formation and remodeling) and inhibits neurodegeneration, interactions between ORF2, HMGA1, and ß-catenin may be important for certain aspects of the latency-reactivation cycle.IMPORTANCE The lifelong latency of bovine herpesvirus 1 (BoHV-1) requires that significant numbers of infected sensory neurons survive infection and maintain normal functions. Consequently, we hypothesize that viral products expressed during latency cooperate with neuronal factors to maintain latency. Our studies revealed that a ß-catenin coactivator, high-mobility group AT-hook 1 protein (HMGA1), was readily detected in a subset of trigeminal ganglion neurons in latently infected calves but not in uninfected calves. A viral protein (ORF2) expressed in latently infected neurons interacted with ß-catenin and HMGA1 in transfected cells, which resulted in the nuclear localization of ß-catenin. This interaction correlated with the ability of ORF2 to stimulate the coactivator functions of HMGA1. These findings are significant because the canonical Wnt/ß-catenin signaling pathway promotes neurogenesis and inhibits neurodegeneration.


Assuntos
Doenças dos Bovinos/virologia , Proteínas HMGA/fisiologia , Infecções por Herpesviridae/veterinária , Herpesvirus Bovino 1/fisiologia , Animais , Bovinos , Doenças dos Bovinos/metabolismo , Linhagem Celular Tumoral , Infecções por Herpesviridae/metabolismo , Infecções por Herpesviridae/virologia , Interações Hospedeiro-Patógeno , Camundongos , Latência Viral , beta Catenina/metabolismo
6.
J Virol ; 90(6): 3148-59, 2016 Jan 06.
Artigo em Inglês | MEDLINE | ID: mdl-26739046

RESUMO

UNLABELLED: Like many Alphaherpesvirinae subfamily members, bovine herpesvirus 1 (BoHV-1) expresses an abundant transcript in latently infected sensory neurons, the latency-related (LR)-RNA. LR-RNA encodes a protein (ORF2) that inhibits apoptosis, interacts with Notch family members, interferes with Notch-mediated transcription, and stimulates neurite formation in cells expressing Notch. An LR mutant virus containing stop codons at the amino terminus of ORF2 does not reactivate from latency or replicate efficiently in certain tissues, indicating that LR gene products are important. In this study, ß-catenin, a transcription factor activated by the canonical Wnt signaling pathway, was frequently detected in ORF2-positive trigeminal ganglionic neurons of latently infected, but not mock-infected, calves. Conversely, the lytic cycle regulatory protein (BoHV-1 infected cell protein 0, or bICP0) was not frequently detected in ß-catenin-positive neurons in latently infected calves. During dexamethasone-induced reactivation from latency, mRNA expression levels of two Wnt antagonists, Dickkopf-1 (DKK-1) and secreted Frizzled-related protein 2 (SFRP2), were induced in bovine trigeminal ganglia (TG), which correlated with reduced ß-catenin protein expression in TG neurons 6 h after dexamethasone treatment. ORF2 and a coactivator of ß-catenin, mastermind-like protein 1 (MAML1), stabilized ß-catenin protein levels and stimulated ß-catenin-dependent transcription in mouse neuroblastoma cells more effectively than MAML1 or ORF2 alone. Neuroblastoma cells expressing ORF2, MAML1, and ß-catenin were highly resistant to cell death following serum withdrawal, whereas most cells transfected with only one of these genes died. The Wnt signaling pathway interferes with neurodegeneration but promotes neuronal differentiation, suggesting that stabilization of ß-catenin expression by ORF2 promotes neuronal survival and differentiation. IMPORTANCE: Bovine herpesvirus 1 (BoHV-1) is an important pathogen of cattle, and like many Alphaherpesvirinae subfamily members establishes latency in sensory neurons. Lifelong latency and the ability to reactivate from latency are crucial for virus transmission. Maintaining the survival and normal functions of terminally differentiated neurons is also crucial for lifelong latency. Our studies revealed that BoHV-1 gene products expressed during latency stabilize expression of the transcription factor ß-catenin and perhaps its cofactor, mastermind-like protein 1 (MAML1). In contrast to expression during latency, ß-catenin expression in sensory neurons is not detectable following treatment of latently infected calves with the synthetic corticosteroid dexamethasone to initiate reactivation from latency. A viral protein (ORF2) expressed in a subset of latently infected neurons stabilized ß-catenin and MAML1 in transfected cells. ORF2, ß-catenin, and MAML1 also enhanced cell survival when growth factors were withdrawn, suggesting that these genes enhance survival of latently infected neurons.


Assuntos
Herpesvirus Bovino 1/fisiologia , Interações Hospedeiro-Patógeno , Células Receptoras Sensoriais/virologia , Proteínas Virais/metabolismo , Latência Viral , Via de Sinalização Wnt , beta Catenina/biossíntese , Animais , Bovinos , Doenças dos Bovinos/virologia , Infecções por Herpesviridae/veterinária , Infecções por Herpesviridae/virologia , Gânglio Trigeminal/virologia
7.
Vet Res ; 46: 22, 2015 Mar 05.
Artigo em Inglês | MEDLINE | ID: mdl-25756342

RESUMO

Small ruminant lentivirus (SRLV) infections of sheep are influenced by genetics on both the host and pathogen sides. Genetic variation in the ovine transmembrane 154 (TMEM154) gene associates with infection susceptibility, and distinct SRLV genetic subgroups infect sheep in association with their TMEM154 diplotypes. In this study, a novel SRLV subgroup was identified that naturally infected sheep with various TMEM154 diplotypes, including those homozygous for a rare frameshift mutation (A4 delta53), which is predicted to abolish TMEM154 protein function. Thus, these SRLVs may infect sheep that lack functional TMEM154, and may not be restricted by TMEM154 diplotypes in establishing infections.


Assuntos
Sequência de Bases , Produtos do Gene gag/genética , Infecções por Lentivirus/veterinária , Lentivirus/genética , Deleção de Sequência , Doenças dos Ovinos/virologia , Animais , Produtos do Gene gag/metabolismo , Variação Genética , Haplótipos , Infecções por Lentivirus/virologia , Dados de Sequência Molecular , Filogenia , Ovinos
8.
J Virol ; 87(23): 13042-7, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24027338

RESUMO

Alphaherpesvirinae family members can reactivate from latency following stress. The synthetic corticosteroid dexamethasone induces certain cellular transcription factors in murine and bovine trigeminal ganglionic neurons. Three dexamethasone-induced transcription factors, Krüppel-like factor 15, Slug, and SPDEF, stimulated the herpes simplex virus type 1-infected cell protein 0 (ICP0) promoter more than 150-fold. Conversely, other viral promoters (VP16 and ICP4) were not strongly stimulated, suggesting that the ICP0 promoter is preferentially activated by dexamethasone-simulated stress.


Assuntos
Herpes Simples/metabolismo , Herpesvirus Humano 1/genética , Proteínas Imediatamente Precoces/genética , Fatores de Transcrição Kruppel-Like/metabolismo , Proteínas Nucleares/metabolismo , Regiões Promotoras Genéticas , Proteínas Proto-Oncogênicas c-ets/metabolismo , Fatores de Transcrição/metabolismo , Gânglio Trigeminal/metabolismo , Ubiquitina-Proteína Ligases/genética , Animais , Dexametasona/farmacologia , Feminino , Regulação Viral da Expressão Gênica , Herpes Simples/genética , Herpes Simples/fisiopatologia , Herpes Simples/virologia , Herpesvirus Humano 1/metabolismo , Humanos , Proteínas Imediatamente Precoces/metabolismo , Fatores de Transcrição Kruppel-Like/genética , Camundongos , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Neurônios/virologia , Proteínas Nucleares/genética , Proteínas Proto-Oncogênicas c-ets/genética , Fatores de Transcrição da Família Snail , Estresse Fisiológico , Fatores de Transcrição/genética , Gânglio Trigeminal/efeitos dos fármacos , Gânglio Trigeminal/virologia , Ubiquitina-Proteína Ligases/metabolismo , Regulação para Cima
9.
J Vet Diagn Invest ; 36(2): 274-277, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-38414254

RESUMO

Bovine hepacivirus (BoHV) is closely related to the hepatitis C virus (HCV) in humans and can cause both acute and chronic liver infections in cattle. BoHV was first identified in Ghana and Germany in 2015 and since then it has been detected and characterized in other countries around the world, but no strains have been sequenced from U.S. cattle. To date, BoHV has been classified into 2 genotypes (1 and 2), with genotype 1 being further divided into 11 subtypes (A-K). However, the true genetic diversity of BoHV is likely underestimated given limited surveillance and a lack of published genome sequences. Here, we sequenced 2 nearly complete BoHV genomes from serum samples collected in 2019 from beef cattle in Missouri. Sequence comparisons and phylogenetic analysis showed that isolate MARC/2019/60 had high sequence homology with genotype 1, subtype E isolates from China. In contrast, isolate MARC/2019/50 represented a novel BoHV subtype within genotype 2. Thus, we report the first genomic characterization of BoHV isolates from U.S. cattle, and the second complete BoHV2 genome worldwide. This work increases our knowledge of the global genetic diversity of BoHV and demonstrates the co-circulation of divergent BoHV strains in U.S. cattle.


Assuntos
Doenças dos Bovinos , Infecções por Herpesviridae , Humanos , Bovinos , Animais , Hepacivirus/genética , Genoma Viral , Variação Genética , Filogenia , Genótipo , Infecções por Herpesviridae/veterinária
10.
J Virol ; 86(5): 2459-73, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-22190728

RESUMO

Bovine herpesvirus 1 (BHV-1), an alphaherpesvirinae subfamily member, establishes latency in sensory neurons. Elevated corticosteroid levels, due to stress, reproducibly triggers reactivation from latency in the field. A single intravenous injection of the synthetic corticosteroid dexamethasone (DEX) to latently infected calves consistently induces reactivation from latency. Lytic cycle viral gene expression is detected in sensory neurons within 6 h after DEX treatment of latently infected calves. These observations suggested that DEX stimulated expression of cellular genes leads to lytic cycle viral gene expression and productive infection. In this study, a commercially available assay-Bovine Gene Chip-was used to compare cellular gene expression in the trigeminal ganglia (TG) of calves latently infected with BHV-1 versus DEX-treated animals. Relative to TG prepared from latently infected calves, 11 cellular genes were induced more than 10-fold 3 h after DEX treatment. Pentraxin three, a regulator of innate immunity and neurodegeneration, was stimulated 35- to 63-fold after 3 or 6 h of DEX treatment. Two transcription factors, promyelocytic leukemia zinc finger (PLZF) and Slug were induced more than 15-fold 3 h after DEX treatment. PLZF or Slug stimulated productive infection 20- or 5-fold, respectively, and Slug stimulated the late glycoprotein C promoter more than 10-fold. Additional DEX-induced transcription factors also stimulated productive infection and certain viral promoters. These studies suggest that DEX-inducible cellular transcription factors and/or signaling pathways stimulate lytic cycle viral gene expression, which subsequently leads to successful reactivation from latency in a small subset of latently infected neurons.


Assuntos
Doenças dos Bovinos/genética , Regulação Viral da Expressão Gênica/efeitos dos fármacos , Infecções por Herpesviridae/veterinária , Herpesvirus Bovino 1/fisiologia , Fatores de Transcrição/genética , Gânglio Trigeminal/metabolismo , Ativação Viral/efeitos dos fármacos , Latência Viral , Animais , Bovinos , Doenças dos Bovinos/metabolismo , Doenças dos Bovinos/virologia , Linhagem Celular , Dexametasona/farmacologia , Infecções por Herpesviridae/genética , Infecções por Herpesviridae/metabolismo , Infecções por Herpesviridae/virologia , Herpesvirus Bovino 1/efeitos dos fármacos , Herpesvirus Bovino 1/genética , Camundongos , Regiões Promotoras Genéticas/efeitos dos fármacos , Coelhos , Fatores de Transcrição/metabolismo , Gânglio Trigeminal/virologia , Regulação para Cima , Latência Viral/efeitos dos fármacos
11.
PNAS Nexus ; 2(5): pgad125, 2023 May.
Artigo em Inglês | MEDLINE | ID: mdl-37181049

RESUMO

Bovine viral diarrhea virus (BVDV) is one of the most important viruses affecting the health and well-being of bovine species throughout the world. Here, we used CRISPR-mediated homology-directed repair and somatic cell nuclear transfer to produce a live calf with a six amino acid substitution in the BVDV binding domain of bovine CD46. The result was a gene-edited calf with dramatically reduced susceptibility to infection as measured by reduced clinical signs and the lack of viral infection in white blood cells. The edited calf has no off-target edits and appears normal and healthy at 20 months of age without obvious adverse effects from the on-target edit. This precision bred, proof-of-concept animal provides the first evidence that intentional genome alterations in the CD46 gene may reduce the burden of BVDV-associated diseases in cattle and is consistent with our stepwise, in vitro and ex vivo experiments with cell lines and matched fetal clones.

12.
J Virol ; 85(6): 2536-46, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21191019

RESUMO

Like other Alphaherpesvirinae subfamily members, bovine herpesvirus 1 (BHV-1) establishes latency in sensory neurons. The latency-related RNA (LR-RNA) is abundantly expressed in latently infected sensory neurons. An LR mutant virus with stop codons at the amino terminus of the first open reading frame (ORF) in the LR gene (ORF2) does not reactivate from latency, in part because it induces higher levels of apoptosis in infected neurons. ORF2 is not the only viral product expressed during latency, but it is important for the latency reactivation cycle because it inhibits apoptosis. In this study, a yeast 2-hybrid screen revealed that ORF2 interacted with two cellular transcription factors, Notch1 and Notch3. These interactions were confirmed in mouse neuroblastoma cells by confocal microscopy and in an in vitro "pulldown" assay. During reactivation from latency, Notch3 RNA levels in trigeminal ganglia were higher than those during latency, suggesting that Notch family members promote reactivation from latency or that reactivation promotes Notch expression. A plasmid expressing the Notch1 intercellular domain (ICD) stimulated productive infection and promoters that encode the viral transcription factor bICP0. The Notch3 ICD did not stimulate productive infection as efficiently as the Notch1 ICD and had no effect on bICP0 promoter activity. Plasmids expressing the Notch1 ICD or the Notch3 ICD trans-activated a late promoter encoding glycoprotein C. ORF2 reduced the trans-activation potential of Notch1 and Notch3, suggesting that ORF2 interfered with the trans-activation potential of Notch. These studies provide evidence that ORF2, in addition to inhibiting apoptosis, has the potential to promote establishment and maintenance of latency by sequestering cellular transcription factors.


Assuntos
Herpesvirus Bovino 1/patogenicidade , Interações Hospedeiro-Patógeno , Mapeamento de Interação de Proteínas , Receptores Notch/metabolismo , Proteínas Virais/metabolismo , Ativação Viral , Animais , Bovinos , Linhagem Celular , Camundongos , Técnicas do Sistema de Duplo-Híbrido
13.
Viruses ; 14(10)2022 09 27.
Artigo em Inglês | MEDLINE | ID: mdl-36298681

RESUMO

Bovine coronavirus (BCoV) has spilled over to many species, including humans, where the host range variant coronavirus OC43 is endemic. The balance of the opposing activities of the surface spike (S) and hemagglutinin-esterase (HE) glycoproteins controls BCoV avidity, which is critical for interspecies transmission and host adaptation. Here, 78 genomes were sequenced directly from clinical samples collected between 2013 and 2022 from cattle in 12 states, primarily in the Midwestern U.S. Relatively little genetic diversity was observed, with genomes having >98% nucleotide identity. Eleven isolates collected between 2020 and 2022 from four states (Nebraska, Colorado, California, and Wisconsin) contained a 12 nucleotide insertion in the receptor-binding domain (RBD) of the HE gene similar to one recently reported in China, and a single genome from Nebraska collected in 2020 contained a novel 12 nucleotide deletion in the HE gene RBD. Isogenic HE proteins containing either the insertion or deletion in the HE RBD maintained esterase activity and could bind bovine submaxillary mucin, a substrate enriched in the receptor 9-O-acetylated-sialic acid, despite modeling that predicted structural changes in the HE R3 loop critical for receptor binding. The emergence of BCoV with structural variants in the RBD raises the possibility of further interspecies transmission.


Assuntos
Doenças dos Bovinos , Infecções por Coronavirus , Coronavirus Bovino , Humanos , Bovinos , Animais , Hemaglutininas/metabolismo , Ácido N-Acetilneuramínico/metabolismo , Mutação , Glicoproteínas/genética , Esterases/genética , Esterases/metabolismo , Nucleotídeos/metabolismo , Glicoproteína da Espícula de Coronavírus/genética
14.
Viruses ; 14(12)2022 12 18.
Artigo em Inglês | MEDLINE | ID: mdl-36560826

RESUMO

Porcine reproductive and respiratory syndrome virus (PRRSV) has a restricted tropism for macrophages and CD163 is a key receptor for infection. In this study, the PRRSV strain NCV1 was passaged on MARC-145 cells for 95 passages, and two plaque-clones (C1 and C2) were randomly selected for further analysis. The C1 virus nearly lost the ability to infect porcine alveolar macrophages (PAMs), as well as porcine kidney cells expressing porcine CD163 (PK15-pCD163), while the C2 virus replicates well in these two cell types. Pretreatment of MARC-145 cells with an anti-CD163 antibody nearly blocked C1 virus infection, indicating that the virus still required CD163 to infect cells. The C1 virus carried four unique amino acid substitutions: three in the nonstructural proteins and a K160I in GP2. The introduction of an I160K substitution in GP2 of the C1 virus restored its infectivity in PAMs and PK15-pCD163 cells, while the introduction of a K160I substitution in GP2 of the low-passaged, virulent PRRSV strain NCV13 significantly impaired its infectivity. Importantly, pigs inoculated with the rNCV13-K160I mutant exhibited lower viremia levels and lung lesions than those infected with the parental rNCV13. These results demonstrated that the K160 residue in GP2 is one of the key determinants of PRRSV tropism.


Assuntos
Síndrome Respiratória e Reprodutiva Suína , Vírus da Síndrome Respiratória e Reprodutiva Suína , Suínos , Animais , Vírus da Síndrome Respiratória e Reprodutiva Suína/genética , Linhagem Celular , Substituição de Aminoácidos , Macrófagos , Glicoproteínas
15.
Viruses ; 14(2)2022 02 13.
Artigo em Inglês | MEDLINE | ID: mdl-35215974

RESUMO

The entry of BVDV into bovine cells was studied using CRIB cells (cells resistant to infection with bovine viral diarrhea virus [BVDV]) that have evolved from MDBK cells by a spontaneous loss of susceptibility to BVDV. Recently, larger genetic deletions were reported but no correlation of the affected genes and the resistance to BVDV infection could be established. The metalloprotease ADAM17 was reported as an essential attachment factor for the related classical swine fever virus (CSFV). To assess whether ADAM17 might be involved in the resistance of CRIB-1 cells to pestiviruses, we analyzed its expression in CRIB-1 and MDBK cells. While ADAM17 protein was detectable in MBDK cells, it was absent from CRIB-1 cells. No functional full-length ADAM17 mRNA could be detected in CRIB cells and genetic analysis revealed the presence of two defective alleles. Transcomplementation of functional ADAM17 derived from MDBK cells in CRIB-1 cells resulted in a nearly complete reversion of their resistance to pestiviral infection. Our results demonstrate that ADAM17 is a key cellular factor for the pestivirus resistance of CRIB-1 cells and establishes its essential role for a broader range of pestiviruses.


Assuntos
Proteína ADAM17/metabolismo , Linhagem Celular/virologia , Vírus da Diarreia Viral Bovina/metabolismo , Pestivirus/metabolismo , Animais , Bovinos , Vírus da Diarreia Viral Bovina/fisiologia , Pestivirus/fisiologia , Replicação Viral/fisiologia
16.
F1000Res ; 11: 385, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-38680232

RESUMO

Background: Bovine congestive heart failure (BCHF) has become increasingly prevalent among feedlot cattle in the Western Great Plains of North America with up to 7% mortality in affected herds. BCHF is an untreatable complex condition involving pulmonary hypertension that culminates in right ventricular failure and death. Genes associated with BCHF in feedlot cattle have not been previously identified. Our aim was to search for genomic regions associated with this disease. Methods: A retrospective, matched case-control design with 102 clinical BCHF cases and their unaffected pen mates was used in a genome-wide association study. Paired nominal data from approximately 560,000 filtered single nucleotide polymorphisms (SNPs) were analyzed with McNemar's test. Results: Two independent genomic regions were identified as having the most significant association with BCHF: the arrestin domain-containing protein 3 gene ( ARRDC3), and the nuclear factor IA gene ( NFIA, mid- p-values, 1x10 -8 and 2x10 -7, respectively). Animals with two copies of risk alleles at either gene were approximately eight-fold more likely to have BCHF than their matched pen mates with either one or zero risk alleles at both genes (CI 95 = 3-17). Further, animals with two copies of risk alleles at both genes were 28-fold more likely to have BCHF than all others ( p-value = 1×10 -7, CI 95 = 4-206). A missense variant in ARRDC3 (C182Y) represents a potential functional variant since the C182 codon is conserved among all other jawed vertebrate species observed. A two-SNP test with markers in both genes showed 29% of 273 BCHF cases had homozygous risk genotypes in both genes, compared to 2.5% in 198 similar unaffected feedlot cattle. This and other DNA tests may be useful for identifying feedlot animals with the highest risk for BCHF in the environments described here. Conclusions: Although pathogenic roles for variants in the ARRDC3 and NFIA genes are unknown, their discovery facilitates classifying animals by genetic risk and allows cattle producers to make informed decisions for selective breeding and animal health management.


Assuntos
Arrestinas , Doenças dos Bovinos , Predisposição Genética para Doença , Insuficiência Cardíaca , Fatores de Transcrição NFI , Animais , Bovinos , Arrestinas/genética , Estudos de Casos e Controles , Doenças dos Bovinos/genética , Estudo de Associação Genômica Ampla , Insuficiência Cardíaca/genética , Insuficiência Cardíaca/veterinária , Fatores de Transcrição NFI/genética , Polimorfismo de Nucleotídeo Único , Estudos Retrospectivos
17.
J Virol ; 84(13): 6308-17, 2010 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-20410283

RESUMO

Bovine herpesvirus 1 (BoHV-1) is an important viral pathogen of cattle. Like other members of the subfamily Alphaherpesvirinae, BoHV-1 establishes latency in sensory neurons and has the potential to reactivate from latency. Dexamethasone (DEX) treatment of latently infected calves or rabbits consistently leads to reactivation from latency. The BoHV-1 transcript encoding the infected cell protein 0 (bICP0) is consistently detected during reactivation from latency, in part because the bICP0 early promoter is activated by DEX. During DEX-induced reactivation from latency, cyclin expression is stimulated in infected sensory neurons. Cyclin-dependent kinase activity phosphorylates Rb (retinoblastoma tumor suppressor gene product) family proteins and consequently releases the E2F family of transcription factors, suggesting that E2F family members stimulate productive infection and/or reactivation from latency. In this study, we provide evidence that repression of E2F1 by a specific small interfering RNA (siRNA) reduced productive infection approximately 5-fold. E2F1 or E2F2 stimulated bICP0 early promoter activity at least 100-fold in transient transfection assays. Two E2F-responsive regions (ERR) were identified within the early promoter, with one adjacent to the TATA box (ERR1) and one approximately 600 bp upstream from the TATA box (ERR2). Mobility shift assays suggested that E2F interacts with ERR1 and ERR2. E2F1 protein levels were increased at late times after infection, which correlated with enhanced binding to a consensus E2F binding site, ERR1, or ERR2. Collectively, these studies suggest that E2F1 stimulates productive infection and bICP0 early promoter activity, in part because E2F family members interact with ERR1 and ERR2.


Assuntos
Fator de Transcrição E2F1/metabolismo , Herpesvirus Bovino 1/fisiologia , Regiões Promotoras Genéticas , Transativadores/biossíntese , Ubiquitina-Proteína Ligases/biossíntese , Ativação Viral , Animais , Sítios de Ligação , Bovinos , Linhagem Celular , Fator de Transcrição E2F1/antagonistas & inibidores , Ensaio de Desvio de Mobilidade Eletroforética , Técnicas de Silenciamento de Genes , Camundongos , Ligação Proteica , Mapeamento de Interação de Proteínas , RNA Interferente Pequeno , Coelhos , Receptores de Estrogênio/metabolismo , Receptor ERRalfa Relacionado ao Estrogênio
18.
J Virol ; 84(13): 6297-307, 2010 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-20410286

RESUMO

Following acute infection in mucosal epithelium, bovine herpes virus 1 (BHV-1) establishes lifelong latency in sensory neurons within trigeminal ganglia. The latency-related RNA (LR-RNA) is abundantly expressed in sensory neurons of latently infected calves. Expression of LR proteins is necessary for the latency reactivation cycle because a mutant virus that does not express LR proteins is unable to reactivate from latency after dexamethasone treatment. LR-RNA sequences also inhibit bICP0 expression, productive infection, and cell growth. However, it is unclear how LR-RNA mediates these functions. In this study, we identified a 463-bp region within the LR gene (the XbaI-PstI [XP] fragment) that inhibited bICP0 protein and RNA expression in transiently transfected mouse neuroblastoma cells. Small noncoding RNAs (sncRNAs) encoded within the XP fragment (20 to 90 nucleotides in length) were detected in transiently transfected mouse neuroblastoma cells. Two families of sncRNAs were cloned from this region, and each family was predicted to contain a mature microRNA (miRNA). Both miRNAs were predicted to base pair with bICP0 mRNA sequences, suggesting that they reduce bICP0 levels. To test this prediction, sequences encompassing the respective sncRNAs and mature miRNAs were synthesized and cloned into a small interfering RNA expression vector. Both sncRNA families and their respective miRNAs inhibited bICP0 protein expression in mouse neuroblastoma cells and productive infection in bovine cells. In trigeminal ganglia of latently infected calves, an sncRNA that migrated between nucleotides 20 and 25 hybridized to the XP fragment. During dexamethasone-induced reactivation from latency, XP-specific sncRNA levels were reduced, suggesting that these sncRNAs support the establishment and maintenance of lifelong latency in cattle.


Assuntos
Herpesvirus Bovino 1/fisiologia , RNA Interferente Pequeno/metabolismo , RNA Viral/metabolismo , Transativadores/biossíntese , Ubiquitina-Proteína Ligases/biossíntese , Proteínas Virais/genética , Replicação Viral , Animais , Bovinos , Doenças dos Bovinos/virologia , Linhagem Celular , Infecções por Herpesviridae/veterinária , Camundongos , Neurônios/virologia , Gânglio Trigeminal/virologia , Latência Viral
19.
Viruses ; 13(11)2021 10 25.
Artigo em Inglês | MEDLINE | ID: mdl-34834954

RESUMO

Bovine viral diarrhea virus's (BVDV) entry into bovine cells involves attachment of virions to cellular receptors, internalization, and pH-dependent fusion with endosomal membranes. The primary host receptor for BVDV is CD46; however, the complete set of host factors required for virus entry is unknown. The Madin-Darby bovine kidney (MDBK) cell line is susceptible to BVDV infection, while a derivative cell line (CRIB) is resistant at the level of virus entry. We performed complete genome sequencing of each to identify genomic variation underlying the resistant phenotype with the aim of identifying host factors essential for BVDV entry. Three large compound deletions in the BVDV-resistant CRIB cell line were identified and predicted to disrupt the function or expression of the genes PTPN12, GRID2, and RABGAP1L. However, CRISPR/Cas9 mediated knockout of these genes, individually or in combination, in the parental MDBK cell line did not impact virus entry or replication. Therefore, resistance to BVDV in the CRIB cell line is not due to the apparent spontaneous loss of PTPN12, GRID2, or RABGAP1L gene function. Identifying the functional cause of BVDV resistance in the CRIB cell line may require more detailed comparisons of the genomes and epigenomes.


Assuntos
Linhagem Celular , Vírus da Diarreia Viral Bovina/fisiologia , Deleção de Genes , Animais , Sistemas CRISPR-Cas , Diarreia/virologia , Cães , Proteínas Ativadoras de GTPase/genética , Técnicas de Inativação de Genes , Proteínas do Tecido Nervoso/genética , Proteína Tirosina Fosfatase não Receptora Tipo 12/genética , Receptores de Glutamato/genética , Internalização do Vírus , Replicação Viral , Sequenciamento Completo do Genoma
20.
Front Vet Sci ; 8: 611927, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33816585

RESUMO

Fifty-six head of cattle, 28 animals with bovine respiratory disease complex (BRDC), and 28 healthy animals that were matched by treatment, sale barn of origin, day, and interactions among these variables, were identified from a population of 180 animals (60 each purchased at three sale barns located in Missouri, Tennessee, and Kentucky) enrolled in a study comparing animals receiving metaphylaxis to saline-treated controls. Cattle were transported to a feedlot in KS and assigned to treatment group. Blood samples were collected at Day 0 (at sale barn), Day 1, Day 9, and Day 28 (at KS feedlot), and transported to the US Meat Animal Research Center in Clay Center, NE where plasma was harvested and stored at -80°C until assayed for the cytokines IFN-γ, IL-1ß, IL-6, and TNF-α, and the acute stress protein haptoglobin (HPT). Our objectives were to determine if cytokine and haptoglobin profiles differed between control and metaphylaxis treatment groups over time, and if profiles differed between animals presenting with BRDC and those that remained healthy. There was no difference between the treated animals and their non-treated counterparts for any of the analytes measured. Sale barn of origin tended to affect TNF-α concentration. Differences for all analytes changed over days, and on specific days was associated with state of origin and treatment. The Treatment by Day by Case interaction was significant for HPT. The analyte most associated with BRDC was HPT on D9, possibly indicating that many of the cattle were not exposed to respiratory pathogens prior to entering the feedlot.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA