Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 57
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
J Virol ; 97(7): e0013523, 2023 07 27.
Artigo em Inglês | MEDLINE | ID: mdl-37338377

RESUMO

The development of effective and flexible vaccine platforms is a major public health challenge, especially in the context of influenza vaccines that have to be renewed every year. Adenoviruses (AdVs) are easy to produce and have a good safety and efficacy profile when administered orally, as demonstrated by the long-term use of oral AdV-4 and -7 vaccines in the U.S. military. These viruses therefore appear to be the ideal backbone for the development of oral replicating vector vaccines. However, research into these vaccines is limited by the ineffectiveness of human AdV replication in laboratory animals. The use of mouse AdV type 1 (MAV-1) in its natural host allows infection to be studied under replicating conditions. Here, we orally vaccinated mice with a MAV-1 vector expressing influenza hemagglutinin (HA) to assess the protection conferred against an intranasal challenge of influenza. We showed that a single oral immunization with this vaccine generates influenza-specific and -neutralizing antibodies and completely protects mice against clinical signs and viral replication, similar to traditional inactivated vaccines. IMPORTANCE Given the constant threat of pandemics and the need for annual vaccination against influenza and possibly emerging agents such as SARS-CoV-2, new types of vaccines that are easier to administer and therefore more widely accepted are a critical public health need. Here, using a relevant animal model, we have shown that replicative oral AdV vaccine vectors can help make vaccination against major respiratory diseases more available, better accepted, and therefore more effective. These results could be of major importance in the coming years in the fight against seasonal or emerging respiratory diseases such as COVID-19.


Assuntos
Infecções por Adenoviridae , Vacinas contra Adenovirus , COVID-19 , Vacinas contra Influenza , Influenza Humana , Humanos , Camundongos , Animais , Adenoviridae/genética , Influenza Humana/prevenção & controle , Anticorpos Antivirais , SARS-CoV-2 , Imunização , Vacinação/métodos , Glicoproteínas de Hemaglutininação de Vírus da Influenza/genética
2.
Front Immunol ; 14: 1149015, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37081878

RESUMO

Respiratory infections and especially viral infections, along with other extrinsic environmental factors, have been shown to profoundly affect macrophage populations in the lung. In particular, alveolar macrophages (AMs) are important sentinels during respiratory infections and their disappearance opens a niche for recruited monocytes (MOs) to differentiate into resident macrophages. Although this topic is still the focus of intense debate, the phenotype and function of AMs that recolonize the niche after an inflammatory insult, such as an infection, appear to be dictated in part by their origin, but also by local and/or systemic changes that may be imprinted at the epigenetic level. Phenotypic alterations following respiratory infections have the potential to shape lung immunity for the long-term, leading to beneficial responses such as protection against allergic airway inflammation or against other infections, but also to detrimental responses when associated with the development of immunopathologies. This review reports the persistence of virus-induced functional alterations in lung macrophages, and discusses the importance of this imprinting in explaining inter-individual and lifetime immune variation.


Assuntos
Pulmão , Infecções Respiratórias , Humanos , Macrófagos Alveolares , Inflamação , Macrófagos
3.
Sci Immunol ; 8(80): eabl9041, 2023 02 24.
Artigo em Inglês | MEDLINE | ID: mdl-36827420

RESUMO

Immunological dysregulation in asthma is associated with changes in exposure to microorganisms early in life. Gammaherpesviruses (γHVs), such as Epstein-Barr virus, are widespread human viruses that establish lifelong infection and profoundly shape host immunity. Using murid herpesvirus 4 (MuHV-4), a mouse γHV, we show that after infection, lung-resident and recruited group 2 innate lymphoid cells (ILC2s) exhibit a reduced ability to expand and produce type 2 cytokines in response to house dust mites, thereby contributing to protection against asthma. In contrast, MuHV-4 infection triggers GM-CSF production by those lung ILC2s, which orders the differentiation of monocytes (Mos) into alveolar macrophages (AMs) without promoting their type 2 functions. In the context of γHV infection, ILC2s are therefore essential cells within the pulmonary niche that imprint the tissue-specific identity of Mo-derived AMs and shape their function well beyond the initial acute infection.


Assuntos
Asma , Infecções por Vírus Epstein-Barr , Rhadinovirus , Humanos , Camundongos , Animais , Macrófagos Alveolares , Imunidade Inata , Linfócitos , Herpesvirus Humano 4 , Rhadinovirus/fisiologia
4.
Viruses ; 14(9)2022 08 23.
Artigo em Inglês | MEDLINE | ID: mdl-36146652

RESUMO

Canine Distemper Virus (CDV) is a fatal and highly contagious pathogen of multiple carnivores. While injectable vaccines are very effective in protecting domestic animals, their use in the wild is unrealistic. Alternative vaccines are therefore needed. Adenovirus (AdV) vectors are popular vaccine vectors due to their capacity to elicit potent humoral and cellular immune responses against the antigens they carry. In parallel, vaccines based on live human AdV-4 and -7 have been used in U.S. army for several decades as replicative oral vaccines against respiratory infection with the same viruses. Based on these observations, the use of oral administration of replication competent AdV-vectored vaccines has emerged as a promising tool especially for wildlife vaccination. Developing this type of vaccine is not easy, however, given the high host specificity of AdVs and their very low replication in non-target species. To overcome this problem, the feasibility of this approach was tested using mouse adenovirus 1 (MAV-1) in mice as vaccine vectors. First, different vaccine vectors expressing the entire or part H or F proteins of CDV were constructed. These different strains were then used as oral vaccines in BALB/c mice and the immune response to CDV was evaluated. Only the strain expressing the full length CDV H protein generated a detectable and neutralizing immune response to CDV. Secondly, using this strain, we were able to show that although this type of vaccine is sensitive to pre-existing immunity to the vector, a second oral administration of the same vaccine is able to boost the immune response against CDV. Overall, this study demonstrates the feasibility of using replicating AdVs as oral vaccine vectors to immunize against CDV in wildlife carnivores.


Assuntos
Infecções por Adenoviridae , Vacinas contra Adenovirus , Vírus da Cinomose Canina , Cinomose , Vacinas Virais , Adenoviridae/genética , Animais , Anticorpos Neutralizantes , Anticorpos Antivirais , Cinomose/prevenção & controle , Vírus da Cinomose Canina/fisiologia , Cães , Humanos , Imunização , Camundongos , Camundongos Endogâmicos BALB C , Vacinação
5.
Transbound Emerg Dis ; 69(4): e194-e203, 2022 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-34357691

RESUMO

Nursing home (NH) residents and staff have been severely affected by the COVID-19 pandemic. The aim of this study was to examine the use of weekly saliva RT-qPCR testing for SARS-CoV-2 detection among NH workers as a strategy to control disease transmission within NHs in Belgium. From 16 November to 27 December 2020, a voluntary and anonymous weekly screening was implemented in a cohort of 50,000 workers across 572 NHs in the Walloon region of Belgium to detect asymptomatic cases of SARS-CoV-2 via saliva RT-qPCR testing and using the Diagenode saliva sample collection device. Positive workers were isolated to avoid subsequent infections in residents and other staff. RT-qPCR testing was based on pooled saliva sampling techniques from three workers, followed by individual testing of each positive or inconclusive pool. The majority of NHs (85%) and 55% of their workers participated. Pooling did not affect sensitivity as it only induced a very decrease in sensitivity estimated as 0.33%. Significant decreases in the prevalence (34.4-13.4%) and incidence of NHs with either single (13.8-2%) or multiple positive workers (3.7-0%) were observed over time. In addition, deaths among NH residents and NH worker absences decreased significantly over time. Weekly saliva RT-qPCR testing for SARS-CoV-2 demonstrated large-scale feasibility and efficacy in disrupting the chain of transmission. Implementation of this testing strategy in NHs could also be extended to other settings with the aim to control viral transmission for maintaining essential activities.


Assuntos
COVID-19 , SARS-CoV-2 , Animais , COVID-19/diagnóstico , COVID-19/epidemiologia , COVID-19/veterinária , Teste para COVID-19/veterinária , Técnicas de Laboratório Clínico/métodos , Técnicas de Laboratório Clínico/veterinária , Humanos , Programas de Rastreamento/veterinária , Casas de Saúde , Pandemias/prevenção & controle , Saliva
6.
Biomedicines ; 9(12)2021 Dec 05.
Artigo em Inglês | MEDLINE | ID: mdl-34944654

RESUMO

(1) Background: viral infections are a frequent cause of chronic obstructive pulmonary disease (COPD) exacerbations, which are responsible for disease progression and mortality. Previous reports showed that IL-20 cytokines facilitate bacterial lung infection, but their production and their role in COPD and viral infection has not yet been investigated. (2) Methods: C57BL/6 WT and IL-20 Rb KO mice were chronically exposed to air or cigarette smoke (CS) to mimic COPD. Cytokine production, antiviral response, inflammation and tissue damages were analyzed after PVM infection. (3) Results: CS exposure was associated with an increase in viral burden and antiviral response. PVM infection in CS mice enhanced IFN-γ, inflammation and tissue damage compared to Air mice. PVM infection and CS exposure induced, in an additive manner, IL-20 cytokines expression and the deletion of IL-20 Rb subunit decreased the expression of interferon-stimulated genes and the production of IFN-λ2/3, without an impact on PVM replication. Epithelial cell damages and inflammation were also reduced in IL-20 Rb-/- mice, and this was associated with reduced lung permeability and the maintenance of intercellular junctions. (4) Conclusions: PVM infection and CS exposure additively upregulates the IL-20 pathway, leading to the promotion of epithelial damages. Our data in our model of viral exacerbation of COPD identify IL-20 cytokine as a potential therapeutic target.

7.
Nano Lett ; 21(1): 847-853, 2021 01 13.
Artigo em Inglês | MEDLINE | ID: mdl-33373240

RESUMO

Viruses are one of the most efficient pathogenic entities on earth, resulting from millions of years of evolution. Each virus particle carries the minimum number of genes and proteins to ensure their reproduction within host cells, hijacking some host replication machinery. However, the role of some viral proteins is not yet unraveled, with some appearing even redundant. For example, murid herpesvirus 4, the current model for human gammaherpesvirus infection, can bind to cell surface glycosaminoglycans using both glycoproteins gp70 and gH/gL. Here, using atomic force microscopy, we discriminate their relative contribution during virus binding to cell surface glycosaminoglycans. Single-virus force spectroscopy experiments demonstrate that gH/gL is the main actor in glycosaminoglycan binding, engaging more numerous and more stable interactions. We also demonstrated that Fab antibody fragments targeting gH/gL or gp70 appear to be a promising treatment to prevent the attachment of virions to cell surfaces.


Assuntos
Proteínas do Envelope Viral , Vírus , Linhagem Celular , Glicoproteínas , Humanos , Análise Espectral
8.
Mucosal Immunol ; 13(5): 799-813, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32424182

RESUMO

Human respiratory syncytial virus (RSV) is a pneumovirus that causes severe infections in infants worldwide. Despite intensive research, safe and effective vaccines against RSV have remained elusive. The main reason is that RSV infection of children previously immunized with formalin-inactivated-RSV vaccines has been associated with exacerbated pathology, a phenomenon called RSV vaccine-enhanced respiratory disease. In parallel, despite the high RSV prevalence, only a minor proportion of children develop severe diseases. Interestingly, variation in the immune responses against RSV or following RSV vaccination could be linked with differences of exposure to microbes during childhood. Gammaherpesviruses (γHVs), such as the Epstein-Barr virus, are persistent viruses that deeply influence the immune system of their host and could therefore affect the development of pneumovirus-induced immunopathologies for the long term. Here, we showed that a previous ɣHV infection protects against both pneumovirus vaccine-enhanced disease and pneumovirus primary infection and that CD8 T cells are essential for this protection. These observations shed a new light on the understanding of pneumovirus-induced diseases and open new perspectives for the development of vaccine strategies.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/metabolismo , Suscetibilidade a Doenças , Gammaherpesvirinae/imunologia , Interações Hospedeiro-Patógeno/imunologia , Infecções por Pneumovirus/etiologia , Infecções por Pneumovirus/metabolismo , Pneumovirus/imunologia , Animais , Anticorpos Neutralizantes/imunologia , Anticorpos Antivirais/imunologia , Modelos Animais de Doenças , Humanos , Imunofenotipagem , Leucócitos/imunologia , Leucócitos/metabolismo , Leucócitos/patologia , Pulmão/imunologia , Pulmão/metabolismo , Pulmão/patologia , Camundongos , Interações Microbianas , Infecções por Pneumovirus/patologia , Infecções por Vírus Respiratório Sincicial/etiologia , Infecções por Vírus Respiratório Sincicial/metabolismo , Vírus Sincicial Respiratório Humano/imunologia , Subpopulações de Linfócitos T/imunologia , Subpopulações de Linfócitos T/metabolismo , Vacinação , Vacinas Virais/imunologia
9.
J Virol ; 93(13)2019 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-30996103

RESUMO

Since the 1970s, replication-competent human adenoviruses 4 and 7 have been used as oral vaccines to protect U.S. soldiers against the severe respiratory diseases caused by these viruses. These vaccines are thought to establish a digestive tract infection conferring protection against respiratory challenge through antibodies. The success of these vaccines makes replication-competent adenoviruses attractive candidates for use as oral vaccine vectors. However, the inability of human adenoviruses to replicate efficiently in laboratory animals has hampered the study of such vectors. Here, we used mouse adenovirus type 1 (MAV-1) in mice to study oral replication-competent adenovirus-based vaccines. We show that MAV-1 oral administration provides protection that recapitulates the protection against homologous respiratory challenge observed with adenovirus 4 and 7 vaccines. Moreover, live oral MAV-1 vaccine better protected against a respiratory challenge than inactivated vaccines. This protection was linked not only with the presence of MAV-1-specific antibodies but also with a better recruitment of effector CD8 T cells. However, unexpectedly, we found that such oral replication-competent vaccine systemically spread all over the body. Our results therefore support the use of MAV-1 to study replication-competent oral adenovirus-based vaccines but also highlight the fact that those vaccines can disseminate widely in the body.IMPORTANCE Replication-competent adenoviruses appear to be promising vectors for the development of oral vaccines in humans. However, the study and development of these vaccines suffer from the lack of any reliable animal model. In this study, mouse adenovirus type 1 was used to develop a small-animal model for oral replication-competent adenovirus vaccines. While this model reproduced in mice what is observed with human adenovirus oral vaccines, it also highlighted that oral immunization with such a replication-competent vaccine is associated with the systemic spread of the virus. This study is therefore of major importance for the future development of such vaccine platforms and their use in large human populations.


Assuntos
Infecções por Adenoviridae/prevenção & controle , Vacinas contra Adenovirus/imunologia , Administração Oral , Trato Gastrointestinal/imunologia , Vacinação , Adenoviridae/imunologia , Infecções por Adenoviridae/imunologia , Adenovírus Humanos , Animais , Anticorpos Antivirais/imunologia , Modelos Animais de Doenças , Feminino , Humanos , Imunização , Pulmão/patologia , Camundongos , Camundongos Endogâmicos BALB C , Vacinas Atenuadas/imunologia , Vacinas Sintéticas/imunologia
10.
Eur J Immunol ; 49(7): 1067-1081, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-30919955

RESUMO

Alternatively activated Mφs (AAMφ) accumulate in hepatic granulomas during schistosomiasis and have been suggested to originate in the bone marrow. What is less understood is how these Mφ responses are regulated after S. mansoni infection. Here, we investigated the role of IL-4 receptor α-chain (IL-4Rα)-signalling in the dynamics of liver Mφ responses. We observed that IL-4Rα signalling was dispensable for the recruitment of Ly6Chi monocytes and for their conversion into F4/80hi CD64hi CD11bhi Mφ. Moreover, while IL-4Rα provided an AAMφ phenotype to liver F4/80hi CD64hi CD11bhi Mφ that was associated with regulation of granuloma formation, it was dispensable for host survival. Resident F4/80hi CD64hi CD11blo Mφ did not upregulate the AAMφ signature gene Ym1. Rather, resident Mφ nearly disappeared by week 8 after infection and artificial ablation of resident Mφ in CD169DTR mice did not affect the response to S. mansoni infection. Interestingly, ablation of CD169+ cells in naive mice resulted in the accumulation of F4/80hi CD64hi CD11bhi Mφ, which was amplified when ablation occurred during schistosomiasis. Altogether, our results suggest the ablation of resident KCs after S. mansoni infection to be associated with the recruitment and accumulation of F4/80hi CD64hi CD11bhi Mφ with lyz2-dependent IL-4Rα contributing to the regulation of granuloma inflammation but being dispensable for host survival.


Assuntos
Granuloma/imunologia , Células de Kupffer/imunologia , Fígado/patologia , Macrófagos/imunologia , Receptores de Superfície Celular/metabolismo , Schistosoma mansoni/fisiologia , Esquistossomose/imunologia , Técnicas de Ablação , Animais , Modelos Animais de Doenças , Feminino , Humanos , Ativação de Macrófagos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout , Receptores de Superfície Celular/genética , Transdução de Sinais
11.
J Invest Dermatol ; 139(3): 626-637, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30227138

RESUMO

Dermatophytoses are among the most common fungal infections worldwide, but little is known about the immune response in them. By comparing Trichophyton benhamiae acute superficial dermatophytosis in wild-type and Rag2-/- mice, we showed that TCR-mediated immunity is critical for fungal clearance and clinical recovery. In WT mice, CD4+ T cells isolated from the skin-draining lymph nodes exhibit both T helper type (Th) 1 and Th17 differentiation during infection, with regard to produced cytokines or mRNA levels of transcription factors. Using IL-17A- and IFN-γ-deficient mice, we showed that IL-17A and IFN-γ are individually dispensable but together contribute to the optimal resolution of dermatophytosis. Furthermore, we generated and infected IL-17A and IFN-γ double-deficient mice and showed that both fungal clearance and clinical recovery were much lower in these mice than in single-deficient mice, suggesting the complementary roles of the two cytokines in dermatophytosis resolution. Thus, our data suggest that TCR-mediated immunity is critical for the optimal control of superficial dermatophytosis and that adaptive immunity is polarized to both Th1 and Th17 responses, with the Th17 antifungal response acting on dermatophyte clearance and the Th1 response being involved in both fungal clearance and Th17-inflammation down-modulation.


Assuntos
Imunidade Adaptativa/fisiologia , Linfócitos T CD4-Positivos/imunologia , Células Th1/imunologia , Células Th17/imunologia , Tinha/imunologia , Animais , Biópsia por Agulha , Proteínas do Sistema Complemento/imunologia , Citocinas/imunologia , Modelos Animais de Doenças , Feminino , Humanos , Imuno-Histoquímica , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Distribuição Aleatória , Índice de Gravidade de Doença , Tinha/patologia
13.
Nat Commun ; 9(1): 4516, 2018 10 30.
Artigo em Inglês | MEDLINE | ID: mdl-30375396

RESUMO

Infection with parasitic helminths can imprint the immune system to modulate bystander inflammatory processes. Bystander or virtual memory CD8+ T cells (TVM) are non-conventional T cells displaying memory properties that can be generated through responsiveness to interleukin (IL)-4. However, it is not clear if helminth-induced type 2 immunity functionally affects the TVM compartment. Here, we show that helminths expand CD44hiCD62LhiCXCR3hiCD49dlo TVM cells through direct IL-4 signaling in CD8+ T cells. Importantly, helminth-mediated conditioning of TVM cells provided enhanced control of acute respiratory infection with the murid gammaherpesvirus 4 (MuHV-4). This enhanced control of MuHV-4 infection could further be explained by an increase in antigen-specific CD8+ T cell effector responses in the lung and was directly dependent on IL-4 signaling. These results demonstrate that IL-4 during helminth infection can non-specifically condition CD8+ T cells, leading to a subsequently raised antigen-specific CD8+ T cell activation that enhances control of viral infection.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Infecções por Herpesviridae/imunologia , Memória Imunológica/imunologia , Interleucina-4/imunologia , Infecções Respiratórias/imunologia , Esquistossomose mansoni/imunologia , Infecções Tumorais por Vírus/imunologia , Animais , Linhagem Celular , Cricetinae , Camundongos , Rhadinovirus , Schistosoma mansoni
14.
J Virol ; 91(13)2017 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-28424280

RESUMO

Gammaherpesviruses are important human and animal pathogens. Infection control has proven difficult because the key process of transmission is ill understood. Murid herpesvirus 4 (MuHV-4), a gammaherpesvirus of mice, is transmitted sexually. We show that this depends on the major virion envelope glycoprotein gp150. gp150 is redundant for host entry, and in vitro, it regulates rather than promotes cell binding. We show that gp150-deficient MuHV-4 reaches and replicates normally in the female genital tract after nasal infection but is poorly released from vaginal epithelial cells and fails to pass from the female to the male genital tract during sexual contact. Thus, we show that the regulation of virion binding is a key component of spontaneous gammaherpesvirus transmission.IMPORTANCE Gammaherpesviruses are responsible for many important diseases in both animals and humans. Some important aspects of their life cycle are still poorly understood. Key among these is viral transmission. Here we show that the major envelope glycoprotein of murid herpesvirus 4 functions not in entry or dissemination but in virion release to allow sexual transmission to new hosts.


Assuntos
Glicoproteínas/metabolismo , Infecções por Herpesviridae/veterinária , Rhadinovirus/fisiologia , Doenças Virais Sexualmente Transmissíveis/veterinária , Proteínas do Envelope Viral/metabolismo , Liberação de Vírus , Animais , Transmissão de Doença Infecciosa , Glicoproteínas/genética , Infecções por Herpesviridae/transmissão , Infecções por Herpesviridae/virologia , Doenças Virais Sexualmente Transmissíveis/transmissão , Doenças Virais Sexualmente Transmissíveis/virologia , Proteínas do Envelope Viral/genética , Ligação Viral , Internalização do Vírus
15.
Immunity ; 46(3): 457-473, 2017 03 21.
Artigo em Inglês | MEDLINE | ID: mdl-28329706

RESUMO

Living in a microbe-rich environment reduces the risk of developing asthma. Exposure of humans or mice to unmethylated CpG DNA (CpG) from bacteria reproduces these protective effects, suggesting a major contribution of CpG to microbe-induced asthma resistance. However, how CpG confers protection remains elusive. We found that exposure to CpG expanded regulatory lung interstitial macrophages (IMs) from monocytes infiltrating the lung or mobilized from the spleen. Trafficking of IM precursors to the lung was independent of CCR2, a chemokine receptor required for monocyte mobilization from the bone marrow. Using a mouse model of allergic airway inflammation, we found that adoptive transfer of IMs isolated from CpG-treated mice recapitulated the protective effects of CpG when administered before allergen sensitization or challenge. IM-mediated protection was dependent on IL-10, given that Il10-/- CpG-induced IMs lacked regulatory effects. Thus, the expansion of regulatory lung IMs upon exposure to CpG might underlie the reduced risk of asthma development associated with a microbe-rich environment.


Assuntos
Quimiotaxia de Leucócito/imunologia , DNA Bacteriano/imunologia , Hipersensibilidade/imunologia , Macrófagos Alveolares/imunologia , Hipersensibilidade Respiratória/imunologia , Animais , Modelos Animais de Doenças , Citometria de Fluxo , Ativação de Macrófagos/imunologia , Macrófagos/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Oligodesoxirribonucleotídeos/imunologia , Baço/imunologia
16.
J Virol ; 90(5): 2455-72, 2015 Dec 16.
Artigo em Inglês | MEDLINE | ID: mdl-26676769

RESUMO

UNLABELLED: Gammaherpesviruses are important human and animal pathogens. Despite the fact that they display the classical architecture of herpesviruses, the function of most of their structural proteins is still poorly defined. This is especially true for tegument proteins. Interestingly, a potential role in immune evasion has recently been proposed for the tegument protein encoded by Kaposi's sarcoma-associated herpesvirus open reading frame 63 (ORF63). To gain insight about the roles of ORF63 in the life cycle of a gammaherpesvirus, we generated null mutations in the ORF63 gene of murid herpesvirus 4 (MuHV-4). We showed that disruption of ORF63 was associated with a severe MuHV-4 growth deficit both in vitro and in vivo. The latter deficit was mainly associated with a defect of replication in the lung but did not affect the establishment of latency in the spleen. From a functional point of view, inhibition of caspase-1 or the inflammasome did not restore the growth of the ORF63-deficient mutant, suggesting that the observed deficit was not associated with the immune evasion mechanism identified previously. Moreover, this growth deficit was also not associated with a defect in virion egress from the infected cells. In contrast, it appeared that MuHV-4 ORF63-deficient mutants failed to address most of their capsids to the nucleus during entry into the host cell, suggesting that ORF63 plays a role in capsid movement. In the future, ORF63 could therefore be considered a target to block gammaherpesvirus infection at a very early stage of the infection. IMPORTANCE: The important diseases caused by gammaherpesviruses in human and animal populations justify a better understanding of their life cycle. In particular, the role of most of their tegument proteins is still largely unknown. In this study, we used murid herpesvirus 4, a gammaherpesvirus infecting mice, to decipher the role of the protein encoded by the viral ORF63 gene. We showed that the absence of this protein is associated with a severe growth deficit both in vitro and in vivo that was mainly due to impaired migration of viral capsids toward the nucleus during entry. Together, our results provide new insights about the life cycle of gammaherpesviruses and could allow the development of new antiviral strategies aimed at blocking gammaherpesvirus infection at the very early stages.


Assuntos
Transporte Biológico , Capsídeo/metabolismo , Rhadinovirus/fisiologia , Proteínas Virais/metabolismo , Internalização do Vírus , Animais , Linhagem Celular , Cricetinae , Feminino , Deleção de Genes , Infecções por Herpesviridae/patologia , Infecções por Herpesviridae/virologia , Histocitoquímica , Pulmão/patologia , Pulmão/virologia , Camundongos Endogâmicos BALB C , Rhadinovirus/genética , Rhadinovirus/crescimento & desenvolvimento , Proteínas Virais/genética
17.
Curr Opin Virol ; 15: 34-40, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26246389

RESUMO

The oncogenicity of gamma-herpesviruses (γHVs) motivates efforts to control them and their persistence makes early events key targets for intervention. Human γHVs are often assumed to enter naive hosts orally and infect B cells directly. However, neither assumption is supported by direct evidence, and vaccination with the Epstein-Barr virus (EBV) gp350, to block virion binding to B cells, failed to reduce infection rates. Thus, there is a need to re-evaluate assumptions about γHV host entry. Given the difficulty of analysing early human infections, potentially much can be learned from animal models. Genomic comparisons argue that γHVs colonized mammals long before humans speciation, and so that human γHVs are unlikely to differ dramatically in behaviour from those of other mammals. Murid Herpesvirus-4 (MuHV-4), which like EBV and the Kaposi's Sarcoma-associated Herpesvirus (KSHV) persists in memory B cells, enters new hosts via olfactory neurons and exploits myeloid cells to spread. Integrating these data with existing knowledge of human and veterinary γHVs suggests a new model of host entry, with potentially important implications for infection control.


Assuntos
Gammaherpesvirinae/fisiologia , Infecções por Herpesviridae/virologia , Internalização do Vírus , Vírus/patogenicidade , Animais , Linfócitos B/imunologia , Linfócitos B/virologia , Infecções por Herpesviridae/imunologia , Infecções por Herpesviridae/transmissão , Interações Hospedeiro-Parasita , Humanos , Células Mieloides/virologia , Mucosa Olfatória/virologia
18.
PLoS One ; 10(2): e0118806, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25705900

RESUMO

Myxoma virus (MYXV) induces a lethal disease called Myxomatosis in European rabbits. MYXV is one of the rare viruses that encodes an α2,3-sialyltransferase through its M138L gene. In this study, we showed that although the absence of the enzyme was not associated with any in vitro deficit, the M138L deficient strains are highly attenuated in vivo. Indeed, while all rabbits infected with the parental and the revertant strains died within 9 days post-infection from severe myxomatosis, all but one rabbit inoculated with the M138L deficient strains survived the infection. In primary lesions, this resistance to the infection was associated with an increased ability of innate immune cells, mostly neutrophils, to migrate to the site of virus replication at 4 days post-infection. This was followed by the development of a better specific immune response against MYXV. Indeed, at day 9 post-infection, we observed an important proliferation of lymphocytes and an intense congestion of blood vessels in lymph nodes after M138L knockouts infection. Accordingly, in these rabbits, we observed an intense mononuclear cell infiltration throughout the dermis in primary lesions and higher titers of neutralizing antibodies. Finally, this adaptive immune response provided protection to these surviving rabbits against a challenge with the MYXV WT strain. Altogether, these results show that expression of the M138L gene contributes directly or indirectly to immune evasion by MYXV. In the future, these results could help us to better understand the pathogenesis of myxomatosis but also the importance of glycans in regulation of immune responses.


Assuntos
Tolerância Imunológica/imunologia , Myxoma virus/imunologia , Mixomatose Infecciosa/imunologia , Sialiltransferases/imunologia , Proteínas Virais/imunologia , Imunidade Adaptativa/imunologia , Animais , Anticorpos Antivirais/sangue , Anticorpos Antivirais/imunologia , DNA Viral/sangue , DNA Viral/genética , DNA Viral/imunologia , Técnicas de Inativação de Genes , Interações Hospedeiro-Patógeno/imunologia , Leucócitos Mononucleares/imunologia , Leucócitos Mononucleares/virologia , Masculino , Myxoma virus/patogenicidade , Myxoma virus/fisiologia , Mixomatose Infecciosa/sangue , Mixomatose Infecciosa/virologia , Coelhos , Sialiltransferases/genética , Sialiltransferases/metabolismo , Análise de Sobrevida , Fatores de Tempo , Proteínas Virais/genética , Proteínas Virais/metabolismo , Virulência/genética , Virulência/imunologia , Fatores de Virulência/genética , Fatores de Virulência/imunologia , Fatores de Virulência/metabolismo
19.
PLoS Pathog ; 9(10): e1003753, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-24204281

RESUMO

Human gammaherpesviruses are associated with the development of lymphomas and epithelial malignancies. The heterogeneity of these tumors reflects the ability of these viruses to route infection to different cell types at various stages of their lifecycle. While the Epstein Barr virus uses gp42--human leukocyte antigen class II interaction as a switch of cell tropism, the molecular mechanism that orientates tropism of rhadinoviruses is still poorly defined. Here, we used bovine herpesvirus 4 (BoHV-4) to further elucidate how rhadinoviruses regulate their infectivity. In the absence of any gp42 homolog, BoHV-4 exploits the alternative splicing of its Bo10 gene to produce distinct viral populations that behave differently based on the originating cell. While epithelial cells produce virions with high levels of the accessory envelope protein gp180, encoded by a Bo10 spliced product, myeloid cells express reduced levels of gp180. As a consequence, virions grown in epithelial cells are hardly infectious for CD14+ circulating cells, but are relatively resistant to antibody neutralization due to the shielding property of gp180 for vulnerable entry epitopes. In contrast, myeloid virions readily infect CD14+ circulating cells but are easily neutralized. This molecular switch could therefore allow BoHV-4 to promote either, on the one hand, its dissemination into the organism, or, on the other hand, its transmission between hosts.


Assuntos
Processamento Alternativo/imunologia , Anticorpos Neutralizantes/imunologia , Anticorpos Antivirais/imunologia , Herpesvirus Bovino 4/imunologia , Proteínas Virais/imunologia , Processamento Alternativo/genética , Animais , Bovinos , Linhagem Celular , Herpesvirus Bovino 4/genética , Humanos , Receptores de Lipopolissacarídeos/genética , Receptores de Lipopolissacarídeos/imunologia , Coelhos
20.
J Virol ; 87(19): 10828-42, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23903840

RESUMO

Glycoprotein B (gB) is a conserved herpesvirus virion component implicated in membrane fusion. As with many-but not all-herpesviruses, the gB of murid herpesvirus 4 (MuHV-4) is cleaved into disulfide-linked subunits, apparently by furin. Preventing gB cleavage for some herpesviruses causes minor infection deficits in vitro, but what the cleavage contributes to host colonization has been unclear. To address this, we mutated the furin cleavage site (R-R-K-R) of the MuHV-4 gB. Abolishing gB cleavage did not affect its expression levels, glycosylation, or antigenic conformation. In vitro, mutant viruses entered fibroblasts and epithelial cells normally but had a significant entry deficit in myeloid cells such as macrophages and bone marrow-derived dendritic cells. The deficit in myeloid cells was not due to reduced virion binding or endocytosis, suggesting that gB cleavage promotes infection at a postendocytic entry step, presumably viral membrane fusion. In vivo, viruses lacking gB cleavage showed reduced lytic spread in the lungs. Alveolar epithelial cell infection was normal, but alveolar macrophage infection was significantly reduced. Normal long-term latency in lymphoid tissue was established nonetheless.


Assuntos
Glicoproteínas/metabolismo , Pulmão/virologia , Células Mieloides/virologia , Rhadinovirus/fisiologia , Proteínas do Envelope Viral/metabolismo , Animais , Anticorpos Neutralizantes/imunologia , Sequência de Bases , Western Blotting , Células Cultivadas , Células Dendríticas/metabolismo , Células Dendríticas/virologia , Ensaio de Imunoadsorção Enzimática , Células Epiteliais/metabolismo , Células Epiteliais/virologia , Feminino , Citometria de Fluxo , Imunofluorescência , Furina/metabolismo , Glicoproteínas/genética , Pulmão/metabolismo , Macrófagos Alveolares/metabolismo , Macrófagos Alveolares/virologia , Camundongos , Camundongos Endogâmicos BALB C , Dados de Sequência Molecular , Mutação/genética , RNA Mensageiro/genética , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Homologia de Sequência do Ácido Nucleico , Proteínas do Envelope Viral/genética , Vírion , Replicação Viral
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA