Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 347
Filtrar
1.
Vaccine ; 42(23): 126227, 2024 Oct 03.
Artigo em Inglês | MEDLINE | ID: mdl-39180978

RESUMO

Elephant endotheliotropic herpesvirus (EEHV) causes lethal hemorrhagic disease (HD) in Asian and African elephants. Although rapid detection of viremia and supportive treatments may improve survival rates, an effective vaccine would mitigate the devastating effects of this virus. In elephants, chronic infection with EEHV leads to adaptive immunity against glycoproteins gB and gH/gL, the core entry machinery for most herpesviruses. We previously evaluated two EEHV gB vaccines in mice but not a gH/gL vaccine. Here, we found that inoculation of mice with an adjuvanted EEHV gH/gL subunit vaccine induced a significant antibody response that was similar to the response observed in elephants chronically infected with EEHV. Moreover, the gH/gL heterodimer elicited polyfunctional T cells with a Th1 phenotype but no detectable Th2 response. These results suggest that gH/gL, possibly in combination with gB, may be suitable immunogens for a vaccine comprising herpesvirus glycoproteins that are known to mediate cell entry and infection.


Assuntos
Infecções por Herpesviridae , Imunidade Celular , Imunidade Humoral , Vacinas de Subunidades Antigênicas , Animais , Feminino , Camundongos , Anticorpos Antivirais/imunologia , Anticorpos Antivirais/sangue , Infecções por Herpesviridae/imunologia , Infecções por Herpesviridae/prevenção & controle , Infecções por Herpesviridae/veterinária , Herpesvirus Equídeo 1/imunologia , Vacinas contra Herpesvirus/imunologia , Imunidade Celular/imunologia , Imunidade Humoral/imunologia , Camundongos Endogâmicos BALB C , Vacinas de Subunidades Antigênicas/imunologia , Proteínas do Envelope Viral/imunologia
2.
Front Immunol ; 15: 1408510, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39021566

RESUMO

Equid alphaherpesvirus 1 (EqAHV1) is a viral pathogen known to cause respiratory disease, neurologic syndromes, and abortion storms in horses. Currently, there are no vaccines that provide complete protection against EqAHV1. Marker vaccines and the differentiation of infected and vaccinated animals (DIVA) strategy are effective for preventing and controlling outbreaks but have not been used for the prevention of EqAHV1 infection. Glycoprotein 2 (gp2), located on the envelope of viruses (EqAHV1), exhibits high antigenicity and functions as a molecular marker for DIVA. In this study, a series of EqAHV1 mutants with deletion of gp2 along with other virulence genes (TK, UL24/TK, gI/gE) were engineered. The mutant viruses were studied in vitro and then in an in vivo experiment using Golden Syrian hamsters to assess the extent of viral attenuation and the immune response elicited by the mutant viruses in comparison to the wild-type (WT) virus. Compared with the WT strain, the YM2019 Δgp2, ΔTK/gp2, and ΔUL24/TK/gp2 strains exhibited reduced growth in RK-13 cells, while the ΔgI/gE/gp2 strain exhibited significantly impaired proliferation. The YM2019 Δgp2 strain induced clinical signs and mortality in hamsters. In contrast, the YM2019 ΔTK/gp2 and ΔUL24/TK/gp2 variants displayed diminished pathogenicity, causing no observable clinical signs or fatalities. Immunization with nasal vaccines containing YM2019 ΔTK/gp2 and ΔUL24/TK/gp2 elicited a robust immune response in hamsters. In particular, compared with the vaccine containing the ΔTK/gp2 strain, the vaccine containing the ΔUL24/TK/gp2 strain demonstrated enhanced immune protection upon challenge with the WT virus. Furthermore, an ELISA for gp2 was established and refined to accurately differentiate between infected and vaccinated animals. These results confirm that the ΔUL24/TK/gp2 strain is a safe and effective live attenuated vaccine candidate for controlling EqAHV1 infection.


Assuntos
Infecções por Herpesviridae , Herpesvirus Equídeo 1 , Vacinas Atenuadas , Animais , Vacinas Atenuadas/imunologia , Infecções por Herpesviridae/prevenção & controle , Infecções por Herpesviridae/imunologia , Infecções por Herpesviridae/virologia , Infecções por Herpesviridae/veterinária , Herpesvirus Equídeo 1/imunologia , Herpesvirus Equídeo 1/genética , Cavalos , Mesocricetus , Anticorpos Antivirais/sangue , Anticorpos Antivirais/imunologia , Proteínas do Envelope Viral/imunologia , Proteínas do Envelope Viral/genética , Cricetinae , Doenças dos Cavalos/prevenção & controle , Doenças dos Cavalos/imunologia , Doenças dos Cavalos/virologia , Vacinas Virais/imunologia , Vacinas Virais/genética , Linhagem Celular , Mutação
3.
PLoS One ; 19(7): e0301987, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38995916

RESUMO

Equid alphaherpesviruses 1 (EHV-1) and 4 (EHV-4) are closely related and both endemic in horses worldwide. Both viruses replicate in the upper respiratory tract, but EHV-1 may additionally lead to abortion and equine herpesvirus myeloencephalopathy (EHM). We focused on antibody responses in horses against the receptor-binding glycoprotein D of EHV-1 (gD1), which shares a 77% amino acid identity with its counterpart in EHV-4 (gD4). Both antigens give rise to cross-reacting antibodies, including neutralizing antibodies. However, immunity against EHV-4 is not considered protective against EHM. While a diagnostic ELISA to discriminate between EHV-1 and EHV-4 infections is available based on type-specific fragments of glycoprotein G (gG1 and gG4, respectively), the type-specific antibody reaction against gD1 has not yet been sufficiently addressed. Starting from the N-terminus of gD1, we developed luciferase immunoprecipitation system (LIPS) assays, using gD1-fragments of increasing size as antigens, i.e. gD1_83 (comprising the first 83 amino acids), gD1_160, gD1_180, and gD1_402 (the full-length molecule). These assays were then used to analyse panels of horse sera from Switzerland (n = 60) and Iceland (n = 50), the latter of which is considered EHV-1 free. We detected only one true negative horse serum from Iceland, whereas all other sera in both panels were seropositive for both gG4 (ELISA) and gD1 (LIPS against gD1_402). In contrast, seropositivity against gG1 was rather rare (35% Swiss sera; 14% Icelandic sera). Therefore, a high percentage of antibodies against gD1 could be attributed to cross-reaction and due to EHV-4 infections. In contrast, the gD1_83 fragment was able to identify sera with type-specific antibodies against gD1. Interestingly, those sera stemmed almost exclusively from vaccinated horses. Although it is uncertain that the N-terminal epitopes of gD1 addressed in this communication are linked to better protection, we suggest that in future vaccine developments, type-common antigens should be avoided, while a broad range of type-specific antigens should be favored.


Assuntos
Anticorpos Antivirais , Herpesvirus Equídeo 1 , Doenças dos Cavalos , Proteínas do Envelope Viral , Animais , Cavalos/imunologia , Herpesvirus Equídeo 1/imunologia , Anticorpos Antivirais/imunologia , Anticorpos Antivirais/sangue , Proteínas do Envelope Viral/imunologia , Doenças dos Cavalos/virologia , Doenças dos Cavalos/imunologia , Doenças dos Cavalos/prevenção & controle , Herpesvirus Equídeo 4/imunologia , Infecções por Herpesviridae/veterinária , Infecções por Herpesviridae/imunologia , Infecções por Herpesviridae/virologia , Reações Cruzadas/imunologia , Ensaio de Imunoadsorção Enzimática , Anticorpos Neutralizantes/imunologia , Anticorpos Neutralizantes/sangue , Domínios Proteicos/imunologia
4.
J Virol ; 98(6): e0025024, 2024 Jun 13.
Artigo em Inglês | MEDLINE | ID: mdl-38742875

RESUMO

Equine herpesvirus type 1 (EHV-1) is a contagious respiratory pathogen that infects the mucosa of the upper respiratory tract (URT). Mucosal immune responses at the URT provide the first line of defense against EHV-1 and are crucial for orchestrating immunity. To define host-pathogen interactions, we characterized B-cell responses, antibody isotype functions, and EHV-1 replication of susceptible (non-immune) and clinically protected (immune) horses after experimental EHV-1 infection. Nasal secretion and nasal wash samples were collected and used for the isolation of DNA, RNA, and mucosal antibodies. Shedding of infectious virus, EHV-1 copy numbers, viral RNA expression, and host B-cell activation in the URT were compared based on host immune status. Mucosal EHV-1-specific antibody responses were associated with EHV-1 shedding and viral RNA transcription. Finally, mucosal immunoglobulin G (IgG) and IgA isotypes were purified and tested for neutralizing capabilities. IgG1 and IgG4/7 neutralized EHV-1, while IgG3/5, IgG6, and IgA did not. Immune horses secreted high amounts of mucosal EHV-1-specific IgG4/7 antibodies and quickly upregulated B-cell pathway genes, while EHV-1 was undetected by virus isolation and PCR. RNA transcription analysis reinforced incomplete viral replication in immune horses. In contrast, complete viral replication with high viral copy numbers and shedding of infectious viruses was characteristic for non-immune horses, together with low or absent EHV-1-specific neutralizing antibodies during viral replication. These data confirm that pre-existing mucosal IgG1 and IgG4/7 and rapid B-cell activation upon EHV-1 infection are essential for virus neutralization, regulation of viral replication, and mucosal immunity against EHV-1.IMPORTANCEEquine herpesvirus type 1 (EHV-1) causes respiratory disease, abortion storms, and neurologic outbreaks known as equine herpes myeloencephalopathy (EHM). EHV-1 is transmitted with respiratory secretions by nose-to-nose contact or via fomites. The virus initially infects the epithelium of the upper respiratory tract (URT). Host-pathogen interactions and mucosal immunity at the viral entry site provide the first line of defense against the EHV-1. Robust mucosal immunity can be essential in protecting against EHV-1 and to reduce EHM outbreaks. It has previously been shown that immune horses do not establish cell-associated viremia, the prerequisite for EHM. Here, we demonstrate how mucosal antibodies can prevent the replication of EHV-1 at the epithelium of the URT and, thereby, the progression of the virus to the peripheral blood. The findings improve the mechanistic understanding of mucosal immunity against EHV-1 and can support the development of enhanced diagnostic tools, vaccines against EHM, and the management of EHV-1 outbreaks.


Assuntos
Anticorpos Neutralizantes , Anticorpos Antivirais , Infecções por Herpesviridae , Herpesvirus Equídeo 1 , Doenças dos Cavalos , Imunoglobulina G , Replicação Viral , Animais , Herpesvirus Equídeo 1/imunologia , Cavalos , Infecções por Herpesviridae/imunologia , Infecções por Herpesviridae/veterinária , Infecções por Herpesviridae/virologia , Anticorpos Antivirais/imunologia , Anticorpos Neutralizantes/imunologia , Doenças dos Cavalos/virologia , Doenças dos Cavalos/imunologia , Imunoglobulina G/imunologia , Imunidade nas Mucosas , Eliminação de Partículas Virais/imunologia , Linfócitos B/imunologia , Linfócitos B/virologia , Interações Hospedeiro-Patógeno/imunologia
5.
J Gen Virol ; 105(5)2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38767608

RESUMO

Herpesviruses establish a well-adapted balance with their host's immune system. Despite this co-evolutionary balance, infections can lead to severe disease including neurological disorders in their natural host. In horses, equine herpesvirus 1 (EHV-1) causes respiratory disease, abortions, neonatal foal death and myeloencephalopathy (EHM) in ~10 % of acute infections worldwide. Many aspects of EHM pathogenesis and protection from EHM are still poorly understood. However, it has been shown that the incidence of EHM increases to >70 % in female horses >20 years of age. In this study we used old mares as an experimental equine EHV-1 model of EHM to identify host-specific factors contributing to EHM. Following experimental infection with the neuropathogenic strain EHV-1 Ab4, old mares and yearling horses were studied for 21 days post-infection. Nasal viral shedding and cell-associated viremia were assessed by quantitative PCR. Cytokine/chemokine responses were evaluated in nasal secretions and cerebrospinal fluid (CSF) by Luminex assay and in whole blood by quantitative real-time PCR. EHV-1-specific IgG sub-isotype responses were measured by ELISA. All young horses developed respiratory disease and a bi-phasic fever post-infection, but only 1/9 horses exhibited ataxia. In contrast, respiratory disease was absent in old mares, but all old mares developed EHM that resulted in euthanasia in 6/9 old mares. Old mares also presented significantly decreased nasal viral shedding but higher viremia coinciding with a single fever peak at the onset of viremia. According to clinical disease manifestation, horses were sorted into an EHM group (nine old horses and one young horse) and a non-EHM group (eight young horses) for assessment of host immune responses. Non-EHM horses showed an early upregulation of IFN-α (nasal secretions), IRF7/IRF9, IL-1ß, CXCL10 and TBET (blood) in addition to an IFN-γ upregulation during viremia (blood). In contrast, IFN-α levels in nasal secretions of EHM horses were low and peak levels of IRF7, IRF9, CXCL10 and TGF-ß (blood) coincided with viremia. Moreover, EHM horses showed significantly higher IL-10 levels in nasal secretions, peripheral blood mononuclear cells and CSF and higher serum IgG3/5 antibody titres compared to non-EHM horses. These results suggest that protection from EHM depends on timely induction of type 1 IFN and upregulation cytokines and chemokines that are representative of cellular immunity. In contrast, induction of regulatory or TH-2 type immunity appeared to correlate with an increased risk for EHM. It is likely that future vaccine development for protection from EHM must target shifting this 'at-risk' immunophenotype.


Assuntos
Citocinas , Infecções por Herpesviridae , Herpesvirus Equídeo 1 , Doenças dos Cavalos , Animais , Cavalos , Herpesvirus Equídeo 1/imunologia , Feminino , Doenças dos Cavalos/virologia , Doenças dos Cavalos/imunologia , Infecções por Herpesviridae/veterinária , Infecções por Herpesviridae/imunologia , Infecções por Herpesviridae/virologia , Citocinas/sangue , Citocinas/imunologia , Anticorpos Antivirais/sangue , Eliminação de Partículas Virais , Viremia/imunologia , Viremia/veterinária , Imunoglobulina G/sangue
6.
Mol Immunol ; 135: 329-341, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-33975251

RESUMO

Horses have many naturally occurring diseases that mimic similar conditions in humans. The ability to conduct environmentally controlled experiments and induced disease studies in a genetically diverse host makes the horse a valuable intermediate model between mouse studies and human clinical trials. This review highlights important similarities in the immune landscape between horses and humans using current research on two equine diseases as examples. First, equine herpesvirus type 1 (EHV-1) infection initiates a series of innate inflammatory signals at its mucosal entry site in the upper respiratory tract. These inflammatory markers are highly synchronized and predictable between individuals during viral respiratory infection and ultimately lead to adaptive immune induction and protection. The timing of early inflammatory signals, followed by specific adaptive immune markers correlating with immunity and protection, allow accurate outbreak tracking and also provide a foundation for understanding the importance of local mucosal immunity during other viral respiratory infections. Second, rare peripheral blood immune cells that promote allergic inflammation can be analyzed during Culicoides hypersensitivity, a naturally occurring type I IgE-mediated allergic disease of horses. Rare immune cells, such as IgE-binding monocytes or basophils, can be studied repeatedly in the horse model to unravel their larger mechanistic role in inflammation during allergic and other inflammatory diseases. We conclude with a survey of all other common equine inflammatory conditions. Together, this review serves as a reference and rationale for the horse as a non-rodent model for immunological research.


Assuntos
Ceratopogonidae/imunologia , Infecções por Herpesviridae/veterinária , Herpesvirus Equídeo 1/imunologia , Doenças dos Cavalos/imunologia , Hipersensibilidade/veterinária , Imunidade nas Mucosas/imunologia , Imunidade Adaptativa/imunologia , Animais , Modelos Animais de Doenças , Infecções por Herpesviridae/imunologia , Infecções por Herpesviridae/patologia , Doenças dos Cavalos/virologia , Cavalos , Humanos , Hipersensibilidade/imunologia , Hipersensibilidade/patologia , Imunoglobulina E/imunologia
8.
Viruses ; 13(3)2021 02 24.
Artigo em Inglês | MEDLINE | ID: mdl-33668216

RESUMO

Equine herpesvirus-1 is the cause of respiratory disease, abortion, and equine herpesvirus myeloencephalopathy (EHM) in horses worldwide. EHM affects as many as 14% of infected horses and a cell-associated viremia is thought to be central for EHM pathogenesis. While EHM is infrequent in younger horses, up to 70% of aged horses develop EHM. The aging immune system likely contributes to EHM pathogenesis; however, little is known about the host factors associated with clinical EHM. Here, we used the "old mare model" to induce EHM following EHV-1 infection. Peripheral blood mononuclear cells (PBMCs) of horses prior to infection and during viremia were collected and RNA sequencing with differential gene expression was used to compare the transcriptome of horses that did (EHM group) and did not (non-EHM group) develop clinical EHM. Interestingly, horses exhibiting EHM did not show respiratory disease, while non-EHM horses showed significant respiratory disease starting on day 2 post infection. Multiple immune pathways differed in EHM horses in response to EHV-1. These included an upregulation of IL-6 gene expression, a dysregulation of T-cell activation through AP-1 and responses skewed towards a T-helper 2 phenotype. Further, a dysregulation of coagulation and an upregulation of elements in the progesterone response were observed in EHM horses.


Assuntos
Infecções por Herpesviridae/virologia , Herpesvirus Equídeo 1/imunologia , Doenças dos Cavalos/imunologia , Doenças dos Cavalos/virologia , Leucócitos Mononucleares/virologia , Transcriptoma/genética , Animais , Feminino , Expressão Gênica/genética , Expressão Gênica/imunologia , Perfilação da Expressão Gênica/métodos , Infecções por Herpesviridae/imunologia , Cavalos , Interleucina-6/genética , Interleucina-6/imunologia , Leucócitos Mononucleares/imunologia , Ativação Linfocitária/genética , Ativação Linfocitária/imunologia , Masculino , Doenças Respiratórias/genética , Doenças Respiratórias/imunologia , Doenças Respiratórias/virologia , Linfócitos T/imunologia , Linfócitos T/virologia , Linfócitos T Auxiliares-Indutores/imunologia , Linfócitos T Auxiliares-Indutores/virologia , Transcriptoma/imunologia , Regulação para Cima/genética , Regulação para Cima/imunologia
9.
J Gen Virol ; 102(3)2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33528354

RESUMO

Equine herpesvirus type 1 (EHV-1) is an emerging pathogen that causes encephalomyelitis in horses and non-equid species. Several aspects of the immune response in the central nervous system (CNS), mainly regarding the role of inflammatory mediators during EHV-1 encephalitis, remain unknown. Moreover, understanding the mechanisms underlying extensive neuropathology induced by viruses would be helpful to establish therapeutic strategies. Therefore, we aimed to evaluate some aspects of the innate immune response during highly neurovirulent EHV-1 infection. C57BL/6 mice infected intranasally with A4/72 and A9/92 EHV-1 strains developed a fulminant neurological disease at 3 days post-inoculation with high viral titres in the brain. These mice developed severe encephalitis with infiltration of monocytes and CD8+ T cells to the brain. The inflammatory infiltrate followed the detection of the chemokines CCL2, CCL3, CCL4, CCL5, CXCL2, CXCL9 and CXCL-10 in the brain. Notably, the levels of CCL3, CCL4, CCL5 and CXCL9 were higher in A4/72-infected mice, which presented higher numbers of inflammatory cells within the CNS. Pro-inflammatory cytokines, such as interleukins (ILs) IL-1α, IL-1ß, IL-6, IL-12ß, and tumour necrosis factor (TNF), were also detected in the CNS, and Toll-like receptor (TLR) TLR2, TLR3 and TLR9 genes were also upregulated within the brain of EHV-1-infected mice. However, no expression of interferon-γ (IFN-γ) and IL-12α, which are important for controlling the replication of other herpesviruses, was detected in EHV-1-infected mice. The results show that the activated innate immune mechanisms could not prevent EHV-1 replication within the CNS, but most likely contributed to the extensive neuropathology. The mouse model of viral encephalitis proposed here will also be useful to study the mechanisms underlying extensive neuropathology.


Assuntos
Encéfalo/imunologia , Encefalite Viral/imunologia , Infecções por Herpesviridae/imunologia , Herpesvirus Equídeo 1/imunologia , Herpesvirus Equídeo 1/patogenicidade , Animais , Encéfalo/metabolismo , Encéfalo/virologia , Quimiocinas/genética , Quimiocinas/metabolismo , Citocinas/genética , Citocinas/metabolismo , Modelos Animais de Doenças , Encefalite Viral/virologia , Infecções por Herpesviridae/virologia , Imunidade Inata , Leucócitos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Receptores Toll-Like/genética , Regulação para Cima , Carga Viral
10.
Can Vet J ; 61(5): 517-520, 2020 05.
Artigo em Inglês | MEDLINE | ID: mdl-32355351

RESUMO

This study aimed to determine if the administration of a modified live equine influenza virus vaccine (FluAvert) to foals would positively impact their health and reduce colonization of their upper airways with equine herpesviruses (EHV) during the weaning period. A single dose of FluAvert was given to 20 healthy foals 7 days prior to being weaned; 20 healthy foals served as unvaccinated controls. Nasal secretions and blood were collected before vaccination, the day of weaning, and weekly thereafter for 3 weeks. Nasal secretions were tested by quantitative polymerase chain reaction (qPCR) for EHV-1, -2, -4 and -5. Whole blood was analyzed for a complete blood cell count and fibrinogen concentration. Physical assessments were made daily. The use of FluAvert was associated with a better clinical outcome. However, the equine influenza virus (EIV) vaccine did not influence selected hematological parameters and kinetics of herpesviruses. The clinical benefit observed in vaccinates may explain the perception that the EIV vaccine induces cross-protection against respiratory agents.


Prévention des infections respiratoires causées par les alpha- et gamma-herpesvirus chez les poulains au sevrage en utilisant un vaccin vivant modifié intra-nasal contre l'influenza. La présente étude visait à déterminer si l'administration d'un vaccin vivant modifié du virus de l'influenza (FluAvert) à des poulains affecterait positivement leur santé et réduirait la colonisation de leurs voies respiratoires supérieures par les herpesvirus équins (EHV) durant la période de sevrage. Une dose unique de FluAvert fut administrée à 20 poulains en santé 7 jours avant le sevrage; 20 poulains en santé ont servi de témoins non-vaccinés. Des sécrétions nasales et du sang furent prélevés avant la vaccination, le jour du sevrage, et de manière hebdomadaire pour les trois semaines suivantes. Les sécrétions nasales furent testées par réaction d'amplification en chaîne par la polymérase quantitative (qPCR) pour EHV-1, -2, -4 et -5. Le sang entier fut analysé pour un dénombrement complet des cellules sanguines et la concentration de fibrinogène. Des examens physiques étaient réalisés quotidiennement. L'utilisation de FluAvert fut associée avec une meilleure issue clinique. Toutefois, le vaccin contre le virus de l'influenza équin (EIV) n'influença pas des paramètres hématologiques sélectionnés et la cinétique des herpesvirus. Les bienfaits cliniques observés chez les chevaux vaccinés pourraient expliquer la perception que le vaccin EIV induit une protection croisée contre des agents infectieux respiratoires.(Traduit par Dr Serge Messier).


Assuntos
Infecções por Herpesviridae/prevenção & controle , Infecções por Herpesviridae/veterinária , Herpesvirus Equídeo 1/imunologia , Doenças dos Cavalos/prevenção & controle , Vacinas contra Influenza , Infecções Respiratórias/prevenção & controle , Infecções Respiratórias/veterinária , Animais , Anticorpos Antivirais , Cavalos
11.
Vaccine ; 38(2): 388-398, 2020 01 10.
Artigo em Inglês | MEDLINE | ID: mdl-31629571

RESUMO

Equid herpesvirus-1 (EHV-1) causes respiratory and neurological disease and late gestation abortion in pregnant mares. Current vaccines contain either inactivated or live EHV-1, but fail to provide complete clinical or virological protection, namely prevention of nasopharyngeal shedding and cell-associated viraemia. Thus, the development of novel products, such as modified live virus (MLV) vaccines which stimulate virus-specific, humoral and cell mediated immune responses more effectively remains a priority. Two groups of weaned foals (n = 6 each group) were used in a longitudinal, prospective, experimental study to evaluate immune responses elicited by two vaccinations with a glycoprotein M (gM) deletion mutant of EHV-1 (RacHdeltagM). Following two concurrent intranasal and intramuscular inoculations six weeks apart, vaccinated (8.4 ±â€¯0.2 months old) and control foals (6.2 ±â€¯0.4 months) were challenge infected intranasally with EHV-1 Ab4/8 four weeks after the second vaccination and clinical signs and virological replication measured. Vaccination caused no adverse events, but did stimulate significantly higher complement fixing and virus neutralizing antibodies in serum compared with control foals at either equivalent or pre-vaccination time points. Virus-specific nasopharyngeal antibody levels and cytotoxic T lymphocyte responses were not significantly different between the groups. Following challenge infection, these immune responses were associated with a reduction in clinical signs and virological replication in the vaccinated foals, including a reduction in duration and magnitude of pyrexia, nasopharyngeal shedding and cell-associated viraemia. We conclude that the RacHΔgM MLV primed EHV-1-specific humoral immune responses in weaned foals. However, complete virological protection by vaccination against EHV-1 requires further research.


Assuntos
Infecções por Herpesviridae/prevenção & controle , Herpesvirus Equídeo 1/imunologia , Doenças dos Cavalos/prevenção & controle , Vacinas Virais/administração & dosagem , Animais , Anticorpos Neutralizantes/imunologia , Feminino , Deleção de Genes , Infecções por Herpesviridae/imunologia , Infecções por Herpesviridae/veterinária , Doenças dos Cavalos/imunologia , Doenças dos Cavalos/virologia , Cavalos , Imunidade Humoral , Estudos Longitudinais , Masculino , Nasofaringe/virologia , Estudos Prospectivos , Vacinas Virais/imunologia , Replicação Viral/imunologia , Eliminação de Partículas Virais
12.
J Gen Virol ; 100(11): 1567-1579, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-31490114

RESUMO

The ancestral equine herpesvirus 1 (EHV1), closely related to human herpes viruses, exploits leukocytes to reach its target organs, accordingly evading the immune surveillance system. Circulating EHV1 strains can be divided into abortigenic/neurovirulent, causing reproductive/neurological disorders. Neurovirulent EHV1 more efficiently recruits monocytic CD172a+ cells to the upper respiratory tract (URT), while abortigenic EHV1 tempers monocyte migration. Whether similar results could be expected for T lymphocytes is not known. Therefore, we questioned whether differences in T cell recruitment could be associated with variations in cell tropism between both EHV1 phenotypes, and which viral proteins might be involved. The expression of CXCL9 and CXCL10 was evaluated in abortigenic/neurovirulent EHV1-inoculated primary respiratory epithelial cells (ERECs). The bioactivity of chemokines was tested with a functional migration assay. Replication of neurovirulent EHV1 in the URT resulted in an enhanced expression/bioactivity of CXCL9 and CXCL10, compared to abortigenic EHV1. Interestingly, deletion of glycoprotein 2 resulted in an increased recruitment of both monocytic CD172a+ cells and T lymphocytes to the corresponding EREC supernatants. Our data reveal a novel function of EHV1-gp2, tempering leukocyte migration to the URT, further indicating a sophisticated virus-mediated orchestration of leukocyte recruitment to the URT.


Assuntos
Quimiocina CXCL10/metabolismo , Quimiocina CXCL9/metabolismo , Células Epiteliais/imunologia , Células Epiteliais/virologia , Herpesvirus Equídeo 1/imunologia , Fatores Imunológicos/metabolismo , Animais , Movimento Celular , Células Cultivadas , Genótipo , Cavalos , Monócitos/imunologia , Monócitos/virologia , Linfócitos T/imunologia , Linfócitos T/virologia , Regulação para Cima , Proteínas do Envelope Viral/genética , Proteínas do Envelope Viral/metabolismo , Replicação Viral
13.
J Virol ; 93(23)2019 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-31511388

RESUMO

Equid herpesvirus 1 (EHV-1) is a viral pathogen of horse populations worldwide spread by the respiratory route and is known for causing outbreaks of neurologic syndromes and abortion storms. Previously, we demonstrated that an EHV-1 strain of the neuropathogenic genotype, T953, downregulates the beta interferon (IFN-ß) response in vitro in equine endothelial cells (EECs) at 12 h postinfection (hpi). In the present study, we explored the molecular correlates of this inhibition as clues toward an understanding of the mechanism. Data from our study revealed that EHV-1 infection of EECs significantly reduced both Toll-like receptor 3 (TLR3) and TLR4 mRNA expression at 6 hpi and 12 hpi. While EHV-1 was able to significantly reduce IRF9 mRNA at both 6 hpi and 12 hpi, the virus significantly reduced IFN regulatory factor 7 (IRF7) mRNA only at 12 hpi. EHV-1 did not alter the cellular level of Janus-activated kinase 1 (JAK1) at any time point. However, EHV-1 reduced the cellular level of expression of tyrosine kinase 2 (TYK2) at 12 hpi. Downstream of JAK1-TYK2 signaling, EHV-1 blocked the phosphorylation and activation of signal transducer and activator of transcription 2 (STAT2) when coincubated with exogenous IFN, at 12 hpi, although not at 3 or 6 hpi. Immunofluorescence staining revealed that the virus prevented the nuclear translocation of STAT2 molecules, confirming the virus-mediated inhibition of STAT2 activation. The pattern of suppression of phosphorylation of STAT2 by EHV-1 implicated viral late gene expression. These data help illuminate how EHV-1 strategically inhibits the host innate immune defense by limiting steps required for type I IFN sensitization and induction.IMPORTANCE To date, no commercial vaccine label has a claim to be fully protective against the diseases caused by equid herpesvirus 1 (EHV-1), especially the neurologic form. The interferon (IFN) system, of which type I IFN is of great importance, still remains a viable immunotherapeutic option against EHV-1 infection. The type I IFN system has been exploited successfully to treat other viral infections, such as chronic hepatitis B and C in humans. The current state of research on how EHV-1 interferes with the protective effect of type I IFN has indicated transient induction of type I IFN production followed by a rapid shutdown in vitro in equine endothelial cells (EECs). The significance of our study is the identification of certain steps in the type I IFN signaling pathway targeted for inhibition by EHV-1. Understanding this pathogen-host relationship is essential for the long-term goal of developing effective immunotherapy against EHV-1.


Assuntos
Células Endoteliais/metabolismo , Células Endoteliais/virologia , Infecções por Herpesviridae/imunologia , Infecções por Herpesviridae/metabolismo , Herpesvirus Equídeo 1/imunologia , Interferon Tipo I/metabolismo , Animais , Regulação da Expressão Gênica , Hepatite B Crônica , Infecções por Herpesviridae/virologia , Herpesvirus Equídeo 1/genética , Doenças dos Cavalos/virologia , Cavalos , Interações Hospedeiro-Patógeno , Humanos , Imunidade Inata , Janus Quinase 1/metabolismo , RNA Mensageiro/metabolismo , Fator de Transcrição STAT2/metabolismo , Transdução de Sinais , TYK2 Quinase/metabolismo , Receptor 3 Toll-Like/metabolismo , Receptor 4 Toll-Like/metabolismo
14.
J Virol ; 93(22)2019 11 15.
Artigo em Inglês | MEDLINE | ID: mdl-31462575

RESUMO

Equine herpesvirus type 1 (EHV-1) outbreaks continue to occur despite widely used vaccination. Therefore, development of EHV-1 vaccines providing improved immunity and protection is ongoing. Here, an open reading frame 2 deletion mutant of the neuropathogenic EHV-1 strain Ab4 (Ab4ΔORF2) was tested as a vaccine candidate. Three groups of horses (n = 8 each) were infected intranasally with Ab4ΔORF2 or the parent Ab4 virus or were kept as noninfected controls. Horses infected with Ab4ΔORF2 had reduced fever and nasal virus shedding compared to those infected with Ab4 but mounted similar adaptive immunity dominated by antibody responses. Nine months after the initial infection, all horses were challenged intranasally with Ab4. Previously noninfected horses (control/Ab4) displayed clinical signs, shed large amounts of virus, and developed cell-associated viremia. In contrast, 5/8 or 3/8 horses previously infected with Ab4ΔORF2 or Ab4, respectively, were fully protected from challenge infection as indicated by the absence of fever, clinical disease, nasal virus shedding, and viremia. All of these outcomes were significantly reduced in the remaining, partially protected 3/8 (Ab4ΔORF2/Ab4) and 5/8 (Ab4/Ab4) horses. Protected horses had EHV-1-specific IgG4/7 antibodies prior to challenge infection, and intranasal antibodies increased rapidly postchallenge. Intranasal inflammatory markers were not detectable in protected horses but quickly increased in control/Ab4 horses during the first week after infection. Overall, our data suggest that preexisting nasal IgG4/7 antibodies neutralize EHV-1, prevent viral entry, and thereby protect from disease, viral shedding, and cell-associated viremia. In conclusion, improved protection from challenge infection emphasizes further evaluation of Ab4ΔORF2 as a vaccine candidate.IMPORTANCE Nasal equine herpesvirus type 1 (EHV-1) shedding is essential for virus transmission during outbreaks. Cell-associated viremia is a prerequisite for the most severe disease outcomes, abortion and equine herpesvirus myeloencephalopathy (EHM). Thus, protection from viremia is considered essential for preventing EHM. Ab4ΔORF2 vaccination prevented EHV-1 challenge virus replication in the upper respiratory tract in fully protected horses. Consequently, these neither shed virus nor developed cell-associated viremia. Protection from virus shedding and viremia during challenge infection in combination with reduced virulence at the time of vaccination emphasizes ORF2 deletion as a promising modification for generating an improved EHV-1 vaccine. During this challenge infection, full protection was linked to preexisting local and systemic EHV-1-specific antibodies combined with rapidly increasing intranasal IgG4/7 antibodies and lack of nasal type I interferon and chemokine induction. These host immune parameters may constitute markers of protection against EHV-1 and be utilized as indicators for improved vaccine development and informed vaccination strategies.


Assuntos
Herpesvirus Equídeo 1/genética , Herpesvirus Equídeo 1/imunologia , Vacinas contra Herpesvirus/imunologia , Doenças dos Cavalos/virologia , Administração Intranasal/métodos , Animais , Anticorpos Antivirais , Feminino , Infecções por Herpesviridae/virologia , Herpesvirus Equídeo 1/metabolismo , Cavalos , Masculino , Mucosa Nasal/virologia , Fases de Leitura Aberta , Rhadinovirus/imunologia , Vacinação/veterinária , Viremia/imunologia , Virulência , Eliminação de Partículas Virais/imunologia
15.
Virology ; 531: 219-232, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-30928700

RESUMO

Equid herpesvirus-1 (EHV-1) outbreaks continue despite widely used vaccination. We demonstrated previously that an ORF1/ORF71 gene deletion mutant of the EHV-1 strain Ab4 (Ab4ΔORF1/71) is less virulent than its parent Ab4 virus. Here, we describe the Ab4 challenge infection evaluating protection induced by the Ab4ΔORF1/71 vaccine candidate. Susceptible control horses developed respiratory disease, fever, nasal shedding, and viremia. Full protection after challenge infection was observed in 5/5 previously Ab4 infected horses and 3/5 Ab4ΔORF1/71 horses. Two Ab4ΔORF1/71 horses developed short-lasting viremia and/or virus shedding. Protective immunity in the respiratory tract was characterized by pre-existing EHV-1-specific IgG4/7 antibodies, the absence of IFN-α secretion and rapidly increasing IgG4/7 upon challenge infection. Pre-existing systemic EHV-1-specific IgG4/7 highly correlated with protection. T-cell immunity was overall low. In conclusion, protective immunity against EHV-1 infection including prevention of viremia was associated with robust systemic and intranasal IgG4/7 antibodies suggesting immediate virus neutralization at the local site.


Assuntos
Anticorpos Antivirais/imunologia , Infecções por Herpesviridae/veterinária , Herpesvirus Equídeo 1/imunologia , Vacinas contra Herpesvirus/administração & dosagem , Doenças dos Cavalos/prevenção & controle , Imunoglobulina G/imunologia , Viremia/veterinária , Administração Intranasal , Animais , Feminino , Infecções por Herpesviridae/imunologia , Infecções por Herpesviridae/prevenção & controle , Infecções por Herpesviridae/virologia , Herpesvirus Equídeo 1/efeitos dos fármacos , Herpesvirus Equídeo 1/genética , Herpesvirus Equídeo 1/fisiologia , Vacinas contra Herpesvirus/imunologia , Doenças dos Cavalos/imunologia , Doenças dos Cavalos/virologia , Cavalos , Masculino , Mucosa Nasal/imunologia , Mucosa Nasal/virologia , Vacinação , Viremia/imunologia , Viremia/prevenção & controle , Viremia/virologia , Eliminação de Partículas Virais
16.
Arch Virol ; 164(5): 1371-1382, 2019 May.
Artigo em Inglês | MEDLINE | ID: mdl-30888564

RESUMO

Equine herpesvirus 1 (EHV-1) induces serious respiratory infections, viral abortion, neurological signs, and neonatal mortality in horses. Despite the use of vaccines, EHV-1 infection also causes a high annual economic burden to the equine industry. The poor immunogenicity of and protection conferred by EHV-1 vaccines are the major factors responsible for the spread of EHV-1 infection. The present study examined the immunogenicity of a novel DNA vaccine co-expressing FliC, a flagellin protein, in Salmonella abortus equi and the gD protein of EHV-1. Mice and horses were immunized intramuscularly with the vaccine, and mice were challenged with EHV-1. Immunofluorescence and western blotting revealed that FliC and gD can be efficiently expressed in cells. This novel vaccine significantly increased gD-specific antibody and interferon gamma (IFN-γ) levels in immunized mice and horses. Compared with controls, the viral load and morbidity were markedly reduced in FliC-gD-immunized mice after they were challenged with EHV-1. Furthermore, the immunogenicity of FliC-gD in a natural host was tested. Our results indicate that vaccinated mice and horses exhibit increased humoral and improved cellular immune responses.


Assuntos
Anticorpos Antivirais/sangue , Flagelina/imunologia , Infecções por Herpesviridae/prevenção & controle , Infecções por Herpesviridae/veterinária , Herpesvirus Equídeo 1/imunologia , Vacinas de DNA/imunologia , Proteínas do Envelope Viral/imunologia , Vacinas Virais/imunologia , Animais , Linhagem Celular , Feminino , Flagelina/genética , Células HEK293 , Infecções por Herpesviridae/imunologia , Cavalos , Humanos , Imunoglobulina G/sangue , Interferon gama/sangue , Camundongos , Camundongos Endogâmicos C57BL , Testes de Neutralização , Salmonella/imunologia , Receptor 5 Toll-Like/metabolismo , Proteínas do Envelope Viral/genética , Carga Viral
17.
J Virol ; 93(7)2019 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-30651370

RESUMO

Equine herpesvirus 1 (EHV1) replicates in the respiratory epithelium and disseminates through the body via a cell-associated viremia in leukocytes, despite the presence of neutralizing antibodies. "Hijacked" leukocytes, previously identified as monocytic cells and T lymphocytes, transmit EHV1 to endothelial cells of the endometrium or central nervous system, causing reproductive (abortigenic variants) or neurological (neurological variants) disorders. In the present study, we questioned the potential route of EHV1 infection of T lymphocytes and how EHV1 misuses T lymphocytes as a vehicle to reach the endothelium of the target organs in the absence or presence of immune surveillance. Viral replication was evaluated in activated and quiescent primary T lymphocytes, and the results demonstrated increased infection of activated versus quiescent, CD4+ versus CD8+, and blood- versus lymph node-derived T cells. Moreover, primarily infected respiratory epithelial cells and circulating monocytic cells efficiently transferred virions to T lymphocytes in the presence of neutralizing antibodies. Albeit T-lymphocytes express all classes of viral proteins early in infection, the expression of viral glycoproteins on their cell surface was restricted. In addition, the release of viral progeny was hampered, resulting in the accumulation of viral nucleocapsids in the T cell nucleus. During contact of infected T lymphocytes with endothelial cells, a late viral protein(s) orchestrates T cell polarization and synapse formation, followed by anterograde dynein-mediated transport and transfer of viral progeny to the engaged cell. This represents a sophisticated but efficient immune evasion strategy to allow transfer of progeny virus from T lymphocytes to adjacent target cells. These results demonstrate that T lymphocytes are susceptible to EHV1 infection and that cell-cell contact transmits infectious virus to and from T lymphocytes.IMPORTANCE Equine herpesvirus 1 (EHV1) is an ancestral alphaherpesvirus that is related to herpes simplex virus 1 and causes respiratory, reproductive, and neurological disorders in Equidae. EHV1 is indisputably a master at exploiting leukocytes to reach its target organs, accordingly evading the host immunity. However, the role of T lymphocytes in cell-associated viremia remains poorly understood. Here we show that activated T lymphocytes efficiently become infected and support viral replication despite the presence of protective immunity. We demonstrate a restricted expression of viral proteins on the surfaces of infected T cells, which prevents immune recognition. In addition, we indicate a hampered release of progeny, which results in the accumulation of nucleocapsids in the T cell nucleus. Upon engagement with the target endothelium, late viral proteins orchestrate viral synapse formation and viral transfer to the contact cell. Our findings have significant implications for the understanding of EHV1 pathogenesis, which is essential for developing innovative therapies to prevent the devastating clinical symptoms of infection.


Assuntos
Infecções por Herpesviridae/imunologia , Herpesvirus Equídeo 1/imunologia , Doenças dos Cavalos/imunologia , Cavalos/imunologia , Linfócitos T/imunologia , Animais , Células Cultivadas , Células Endoteliais/imunologia , Células Endoteliais/virologia , Células Epiteliais/imunologia , Células Epiteliais/virologia , Infecções por Herpesviridae/virologia , Doenças dos Cavalos/virologia , Cavalos/virologia , Evasão da Resposta Imune/imunologia , Monócitos/imunologia , Monócitos/virologia , Mucosa Respiratória/imunologia , Mucosa Respiratória/virologia , Linfócitos T/virologia , Proteínas Virais/imunologia , Viremia/imunologia , Viremia/virologia , Replicação Viral/imunologia
18.
Artigo em Inglês | MEDLINE | ID: mdl-30258819

RESUMO

Equine herpesvirus 1 (EHV1) is considered as a major pathogen of Equidae, causing symptoms from mild respiratory disease to late-term abortion and neurological disorders. Different EHV1 strains circulating in the field have been characterized to be of abortigenic or neurovirulent phenotype. Both variants replicate in a plaque-wise manner in the epithelium of the upper respiratory tract (URT), where the abortigenic strains induce more prominent viral plaques, compared to the neurovirulent strains. Considering the differences in replication at the URT, we hypothesized that abortigenic strains may show an increased ability to modulate the type I IFN secretion/signaling pathway, compared to strains that display the neurovirulent phenotype. Here, we analyze IFN levels induced by abortigenic and neurovirulent EHV1 using primary respiratory epithelial cells (EREC) and respiratory mucosa ex vivo explants. Similar levels of IFNα (~70 U/ml) were detected in explants inoculated with both types of EHV1 strains from 48 to 72 hpi. Second, EREC and mucosa explants were treated with recombinant equine IFNα (rEqIFNα) or Ruxolitinib (Rux), an IFN signaling inhibitor, prior to and during inoculation with abortigenic or neurovirulent EHV1. Replication of both EHV1 variants was suppressed by rEqIFNα. Further, addition of Rux increased replication in a concentration-dependent manner, indicating an IFN-susceptibility for both variants. However, in two out of three horses, at a physiological concentration of 100 U/ml of rEqIFNα, an increase in abortigenic EHV1 replication was observed compared to 10 U/ml of rEqIFNα, which was not observed for the neurovirulent strains. Moreover, in the presence of Rux, the plaque size of the abortigenic variants remained unaltered, whereas the typically smaller viral plaques induced by the neurovirulent variants became larger. Overall, our results demonstrate the importance of IFNα in the control of EHV1 replication in the URT for both abortigenic and neurovirulent variants. In addition, our findings support the speculation that abortigenic variants of EHV1 may have developed anti-IFN mechanisms that appear to be absent or less pronounced in neurovirulent EHV1 strains.


Assuntos
Herpesvirus Equídeo 1/crescimento & desenvolvimento , Herpesvirus Equídeo 1/imunologia , Interações Hospedeiro-Patógeno , Evasão da Resposta Imune , Fatores Imunológicos/análise , Interferon-alfa/análise , Animais , Células Cultivadas , Células Epiteliais/imunologia , Células Epiteliais/virologia , Herpesvirus Equídeo 1/classificação , Cavalos , Modelos Biológicos , Técnicas de Cultura de Órgãos , Mucosa Respiratória/imunologia , Mucosa Respiratória/virologia , Ensaio de Placa Viral , Replicação Viral
19.
Vet Microbiol ; 222: 18-24, 2018 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-30080668

RESUMO

Immune responses were compared after intranasal (IN) and intramuscular (IM) vaccination of horses with a modified live equine herpesvirus type-1 (EHV-1) vaccine, and the protective effect after EHV-1 challenge was evaluated. IN- and IM-vaccinated groups (n = 5 each) showed significant rises in serum virus-neutralizing titers with increased levels of IgGa and IgGb antibodies after the first vaccination (P < 0.05). In nasal secretions, the IN group had significantly increased levels of IgA antibodies after vaccination (P < 0.05), whereas the response of the IM group was dominated by IgGa and IgGb subclasses. After challenge infection, the numbers of pyretic horses from 1 to 4 days post-inoculation were 3/5 in the placebo (PBO) group (n = 5), 0/5 in the IN group, and 1/5 in the IM group. The IN and IM groups had significantly lower levels of virus shedding than the PBO group (P < 0.05). There were no significant between-group differences in the numbers of viremic horses each day. Notably, two horses in the IM group had no virus shedding or viremia, whereas all horses in the other group did. Both IN and IM vaccination induced systemic humoral immunity and mucosal immunity, suppressing virus replication in the nasal mucosa, and partially protected horses from pyrexia, especially early in infection. This study showed a mucosal antibody response was induced, not only by IN vaccination but also by IM vaccination.


Assuntos
Administração Intranasal/métodos , Infecções por Herpesviridae/veterinária , Herpesvirus Equídeo 1/imunologia , Vacinas contra Herpesvirus/imunologia , Injeções Intramusculares/métodos , Vacinação/veterinária , Animais , Anticorpos Antivirais/sangue , Infecções por Herpesviridae/imunologia , Infecções por Herpesviridae/prevenção & controle , Infecções por Herpesviridae/virologia , Vacinas contra Herpesvirus/administração & dosagem , Doenças dos Cavalos/imunologia , Doenças dos Cavalos/prevenção & controle , Doenças dos Cavalos/virologia , Cavalos , Imunidade nas Mucosas , Imunoglobulina A/sangue , Imunoglobulina G/sangue , Testes de Neutralização , Vacinação/métodos , Vacinas Atenuadas/administração & dosagem , Eliminação de Partículas Virais/imunologia
20.
BMC Vet Res ; 14(1): 245, 2018 Aug 22.
Artigo em Inglês | MEDLINE | ID: mdl-30134896

RESUMO

BACKGROUND: Equine herpesvirus type 1 (EHV-1) induces respiratory infection, abortion, and neurologic disease with significant impact. Virulence factors contributing to infection and immune evasion are of particular interest. A potential virulence factor of the neuropathogenic EHV-1 strain Ab4 is ORF2. This study on 24 Icelandic horses, 2 to 4 years of age, describes the infection with EHV-1 Ab4, or its deletion mutant devoid of ORF2 (Ab4ΔORF2) compared to non-infected controls (each group n = 8). The horses' clinical presentation, virus shedding, viremia, antibody and cellular immune responses were monitored over 260 days after experimental infection. RESULTS: Infection with Ab4ΔORF2 reduced fever and minimized nasal virus shedding after infection compared to the parent virus strain Ab4, while Ab4ΔORF2 established viremia similar to Ab4. Concurrently with virus shedding, intranasal cytokine and interferon α (IFN-α) production increased in the Ab4 group, while horses infected with Ab4ΔORF2 expressed less IFN-α. The antibody response to EHV-1 was evaluated by a bead-based multiplex assay and was similar in both infected groups, Ab4 and Ab4ΔORF2. EHV-1 specific immunoglobulin (Ig) G1 was induced 8 days after infection (d8 pi) with a peak on d10-12 pi. EHV-1 specific IgG4/7 increased starting on d10 pi, and remained elevated in serum until the end of the study. The intranasal antibody response to EHV-1 was dominated by the same IgG isotypes and remained elevated in both infected groups until d130 pi. In contrast to the distinct antibody response, no induction of EHV-1 specific T-cells was detectable by flow cytometry after ex vivo re-stimulation of peripheral blood mononuclear cells (PBMC) with EHV-1 in any group. The cellular immune response was characterized by increased secretion of IFN-γ and interleukin10 in response to ex vivo re-stimulation of PBMC with EHV-1. This response was present during the time of viremia (d5-10 pi) and was similar in both infected groups, Ab4 and Ab4ΔORF2. CONCLUSIONS: ORF2 is a virulence factor of EHV-1 Ab4 with impact on pyrexia and virus shedding from the nasal mucosa. In contrast, ORF2 does not influence viremia. The immunogenicity of the Ab4ΔORF2 and parent Ab4 viruses are identical. Graphical abstract - Deletion of ORF2 reduces virulence of EHV-1 Ab4. Graphical summary of the main findings of this study: ORF2 is a virulence factor of EHV-1 Ab4 with impact on pyrexia and virus shedding from the nasal mucosa.


Assuntos
Infecções por Herpesviridae/veterinária , Herpesvirus Equídeo 1/genética , Herpesvirus Equídeo 1/patogenicidade , Doenças dos Cavalos/virologia , Proteínas Virais/genética , Fatores de Virulência/genética , Virulência/genética , Animais , Citocinas/metabolismo , Feminino , Herpesvirus Equídeo 1/imunologia , Doenças dos Cavalos/imunologia , Cavalos , Leucócitos Mononucleares/virologia , Masculino , Mucosa Nasal/virologia , Deleção de Sequência , Viremia/veterinária , Eliminação de Partículas Virais/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...