Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 3.141
Filtrar
1.
J Exp Med ; 221(7)2024 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-38661718

RESUMO

Chemokines guide immune cells during their response against pathogens and tumors. Various techniques exist to determine chemokine production, but none to identify cells that directly sense chemokines in vivo. We have generated CCL3-EASER (ErAse, SEnd, Receive) mice that simultaneously report for Ccl3 transcription and translation, allow identifying Ccl3-sensing cells, and permit inducible deletion of Ccl3-producing cells. We infected these mice with murine cytomegalovirus (mCMV), where Ccl3 and NK cells are critical defense mediators. We found that NK cells transcribed Ccl3 already in homeostasis, but Ccl3 translation required type I interferon signaling in infected organs during early infection. NK cells were both the principal Ccl3 producers and sensors of Ccl3, indicating auto/paracrine communication that amplified NK cell response, and this was essential for the early defense against mCMV. CCL3-EASER mice represent the prototype of a new class of dual fluorescence reporter mice for analyzing cellular communication via chemokines, which may be applied also to other chemokines and disease models.


Assuntos
Comunicação Celular , Quimiocina CCL3 , Células Matadoras Naturais , Muromegalovirus , Biossíntese de Proteínas , Transcrição Gênica , Animais , Camundongos , Muromegalovirus/fisiologia , Quimiocina CCL3/metabolismo , Quimiocina CCL3/genética , Células Matadoras Naturais/imunologia , Células Matadoras Naturais/metabolismo , Genes Reporter , Camundongos Endogâmicos C57BL , Infecções por Herpesviridae/imunologia , Infecções por Herpesviridae/genética , Camundongos Transgênicos , Interferon Tipo I/metabolismo , Transdução de Sinais
2.
J Neurovirol ; 30(1): 86-99, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-38453879

RESUMO

Simian varicella virus (SVV) produces peripheral inflammatory responses during varicella (primary infection) and zoster (reactivation) in rhesus macaques (RM). However, it is unclear if peripheral measures are accurate proxies for central nervous system (CNS) responses. Thus, we analyzed cytokine and Aß42/Aß40 changes in paired serum and cerebrospinal fluid (CSF) during the course of infection. During varicella and zoster, every RM had variable changes in serum and CSF cytokine and Aß42/Aß40 levels compared to pre-inoculation levels. Overall, peripheral infection appears to affect CNS cytokine and Aß42/Aß40 levels independent of serum responses, suggesting that peripheral disease may contribute to CNS disease.


Assuntos
Peptídeos beta-Amiloides , Citocinas , Macaca mulatta , Animais , Peptídeos beta-Amiloides/líquido cefalorraquidiano , Peptídeos beta-Amiloides/sangue , Citocinas/líquido cefalorraquidiano , Citocinas/sangue , Ativação Viral , Fragmentos de Peptídeos/líquido cefalorraquidiano , Fragmentos de Peptídeos/sangue , Varicellovirus/genética , Varicellovirus/imunologia , Herpesvirus Humano 3/patogenicidade , Herpesvirus Humano 3/imunologia , Infecções por Herpesviridae/líquido cefalorraquidiano , Infecções por Herpesviridae/virologia , Infecções por Herpesviridae/sangue , Infecções por Herpesviridae/imunologia , Masculino , Herpes Zoster/líquido cefalorraquidiano , Herpes Zoster/virologia , Herpes Zoster/sangue , Herpes Zoster/imunologia , Doenças dos Macacos/virologia , Doenças dos Macacos/líquido cefalorraquidiano , Doenças dos Macacos/sangue
3.
J Virol ; 97(11): e0132223, 2023 Nov 30.
Artigo em Inglês | MEDLINE | ID: mdl-37882519

RESUMO

IMPORTANCE: Chickens immunized with the infectious laryngotracheitis chicken embryo origin (CEO) vaccine (Medivac, PT Medion Farma Jaya) experience adverse reactions, hindering its safety and effective use in poultry flocks. To improve the effect of the vaccine, we sought to find a strategy to alleviate the respiratory reactions associated with the vaccine. Here, we confirmed that co-administering the CEO vaccine with chIL-2 by oral delivery led to significant alleviation of the vaccine reactions in chickens after immunization. Furthermore, we found that the co-administration of chIL-2 with the CEO vaccine reduced the clinical signs of the CEO vaccine while enhancing natural killer cells and cytotoxic T lymphocyte response to decrease viral loads in their tissues, particularly in the trachea and conjunctiva. Importantly, we demonstrated that the chIL-2 treatment can ameliorate the replication of the CEO vaccine without compromising its effectiveness. This study provides new insights into further applications of chIL-2 and a promising strategy for alleviating the adverse reaction of vaccines.


Assuntos
Galinhas , Infecções por Herpesviridae , Herpesvirus Galináceo 1 , Interleucina-2 , Células Matadoras Naturais , Linfócitos T Citotóxicos , Vacinas Virais , Animais , Administração Oral , Galinhas/imunologia , Galinhas/virologia , Túnica Conjuntiva/virologia , Infecções por Herpesviridae/imunologia , Infecções por Herpesviridae/prevenção & controle , Infecções por Herpesviridae/veterinária , Infecções por Herpesviridae/virologia , Herpesvirus Galináceo 1/imunologia , Interleucina-2/administração & dosagem , Interleucina-2/imunologia , Células Matadoras Naturais/imunologia , Doenças das Aves Domésticas/imunologia , Doenças das Aves Domésticas/prevenção & controle , Doenças das Aves Domésticas/virologia , Doenças Respiratórias/imunologia , Doenças Respiratórias/prevenção & controle , Doenças Respiratórias/veterinária , Doenças Respiratórias/virologia , Linfócitos T Citotóxicos/imunologia , Traqueia/virologia , Carga Viral , Vacinas Virais/administração & dosagem , Vacinas Virais/efeitos adversos , Vacinas Virais/biossíntese , Vacinas Virais/imunologia
4.
Front Immunol ; 14: 1235590, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37600809

RESUMO

The incidence of human herpesvirus (HHVs) is gradually increasing and has affected a wide range of population. HHVs can result in serious consequences such as tumors, neonatal malformations, sexually transmitted diseases, as well as pose an immense threat to the human health. The cGAS-STING pathway is one of the innate immune pattern-recognition receptors discovered recently. This article discusses the role of the cGAS-STING pathway in human diseases, especially in human herpesvirus infections, as well as highlights how these viruses act on this pathway to evade the host immunity. Moreover, the author provides a comprehensive overview of modulators of the cGAS-STING pathway. By focusing on the small molecule compounds based on the cGAS-STING pathway, novel targets and concepts have been proposed for the development of antiviral drugs and vaccines, while also providing a reference for the investigation of disease models related to the cGAS-STING pathway. HHV is a double-stranded DNA virus that can trigger the activation of intracellular DNA sensor cGAS, after which the host cells initiate a cascade of reactions that culminate in the secretion of type I interferon to restrict the viral replication. Meanwhile, the viral protein can interact with various molecules in the cGAS-STING pathway. Viruses can evade immune surveillance and maintain their replication by inhibiting the enzyme activity of cGAS and reducing the phosphorylation levels of STING, TBK1 and IRF3 and suppressing the interferon gene activation. Activators and inhibitors of the cGAS-STING pathway have yielded numerous promising research findings in vitro and in vivo pertaining to cGAS/STING-related disease models. However, there remains a dearth of small molecule modulators that have been successfully translated into clinical applications, which serves as a hurdle to be overcome in the future.


Assuntos
Infecções por Herpesviridae , Imunidade Inata , Proteínas de Membrana , Nucleotidiltransferases , Transdução de Sinais , Humanos , Cromogranina A , Infecções por Herpesviridae/imunologia , Fosforilação
5.
Virology ; 576: 134-140, 2022 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-36244319

RESUMO

Gammaherpesviruses establish life-long infection in over 95% of adults and are associated with several cancers, including B cell lymphomas. Using the murine gammaherpesvirus 68 (MHV68) animal model, we previously showed a pro-viral role of Interleukin-1 (IL-1) signaling that supported viral reactivation during the establishment of chronic infection. Unexpectedly, in this study we found that the proviral effects of IL-1 signaling originally observed during the establishment of chronic gammaherpesvirus infection convert to antiviral effects during the long-term stage of infection. Specifically, IL-1 signaling promoted expansion of antiviral CD8+ T cells and control of viral reactivation in the peritoneal cavity of a long-term infected host. Using a novel mouse model of T cell-specific IL-1 signaling deficiency, we found that the antiviral effects of IL-1 signaling were T cell extrinsic. Our study highlights a dynamic nature of host factors that shape the parameters of chronic gammaherpesvirus infection.


Assuntos
Gammaherpesvirinae , Infecções por Herpesviridae , Interleucina-1 , Animais , Camundongos , Antivirais , Linfócitos B , Linfócitos T CD8-Positivos/patologia , Infecções por Herpesviridae/imunologia , Infecções por Herpesviridae/metabolismo , Interleucina-1/imunologia , Interleucina-1/metabolismo , Camundongos Endogâmicos C57BL , Latência Viral
6.
Viruses ; 14(10)2022 10 19.
Artigo em Inglês | MEDLINE | ID: mdl-36298850

RESUMO

Kaposi's sarcoma-associated herpesvirus (KSHV) is associated with vascular endothelial cell tumor, Kaposi's sarcoma (KS) and lymphoproliferative disorder, multicentric Castleman's disease (MCD), primary effusion lymphoma (PEL) and KSHV inflammatory cytokine syndrome (KICS). Dysregulation of proinflammatory cytokines is found in most KSHV associated diseases. However, little is known about the role of host microenvironment in the regulation of KSHV establishment in B cells. In the present study, we demonstrated that IFN-γ has a strong inhibitory effect on KSHV infection but only in a subset of tonsil-derived lymphocyte samples that are intrinsically more susceptible to infection, contain higher proportions of naïve B cells, and display increased levels of IRF1 and STAT1-pY701. The effect of IFN-γ in responsive samples was associated with increased frequencies of germinal center B cells (GCB) and decreased infection of plasma cells, suggesting that IFN-γ-mediated modulation of viral dynamics in GC can inhibit the establishment of KSHV infection.


Assuntos
Linfócitos B , Infecções por Herpesviridae , Herpesvirus Humano 8 , Interferon gama , Humanos , Síndrome de Imunodeficiência Adquirida/imunologia , Linfócitos B/imunologia , Hiperplasia do Linfonodo Gigante/imunologia , Citocinas/imunologia , Infecções por Herpesviridae/genética , Infecções por Herpesviridae/imunologia , Herpesvirus Humano 8/genética , Herpesvirus Humano 8/imunologia , Fator Regulador 1 de Interferon/genética , Fator Regulador 1 de Interferon/imunologia , Interferon gama/imunologia , Sarcoma de Kaposi/imunologia , Microambiente Tumoral/genética , Microambiente Tumoral/imunologia , Suscetibilidade a Doenças/imunologia
7.
Proc Natl Acad Sci U S A ; 119(32): e2123362119, 2022 08 09.
Artigo em Inglês | MEDLINE | ID: mdl-35921433

RESUMO

The germinal center (GC) plays a central role in the generation of antigen-specific B cells and antibodies. Tight regulation of the GC is essential due to the inherent risks of tumorigenesis and autoimmunity posed by inappropriate GC B cell processes. Gammaherpesviruses such as Epstein-Barr virus (EBV) and murine gammaherpesvirus 68 (MHV68) utilize numerous armaments to drive infected naïve B cells, independent of antigen, through GC reactions to expand the latently infected B cell population and establish a stable latency reservoir. We previously demonstrated that the MHV68 microRNA (miRNA) mghv-miR-M1-7-5p represses host EWSR1 (Ewing sarcoma breakpoint region 1) to promote B cell infection. EWSR1 is a transcription and splicing regulator that is recognized for its involvement as a fusion protein in Ewing sarcoma. A function for EWSR1 in B cell responses has not been previously reported. Here, we demonstrate that 1) B cell-specific deletion of EWSR1 had no effect on generation of mature B cell subsets or basal immunoglobulin levels in naïve mice, 2) repression or ablation of EWSR1 in B cells promoted expansion of MHV68 latently infected GC B cells, and 3) B cell-specific deletion of EWSR1 during a normal immune response to nonviral antigen resulted in significantly elevated numbers of antigen-specific GC B cells, plasma cells, and circulating antibodies. Notably, EWSR1 deficiency did not affect the proliferation or survival of GC B cells but instead resulted in the generation of increased numbers of precursor GC B cells. Cumulatively, these findings demonstrate that EWSR1 is a negative regulator of B cell responses.


Assuntos
Linfócitos B , Gammaherpesvirinae , Centro Germinativo , Infecções por Herpesviridae , MicroRNAs , Proteína EWS de Ligação a RNA , Infecções Tumorais por Vírus , Animais , Linfócitos B/imunologia , Linfócitos B/virologia , Gammaherpesvirinae/genética , Gammaherpesvirinae/fisiologia , Deleção de Genes , Centro Germinativo/imunologia , Centro Germinativo/virologia , Infecções por Herpesviridae/genética , Infecções por Herpesviridae/imunologia , Infecções por Herpesviridae/virologia , Camundongos , MicroRNAs/genética , MicroRNAs/metabolismo , Proteína EWS de Ligação a RNA/genética , Proteína EWS de Ligação a RNA/metabolismo , Infecções Tumorais por Vírus/genética , Infecções Tumorais por Vírus/imunologia , Infecções Tumorais por Vírus/virologia , Latência Viral
8.
Front Immunol ; 13: 787021, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35173716

RESUMO

Vaccination is the best form of protecting fish against viral diseases when the pathogen cannot be contained by biosecurity measures. Vaccines based on live attenuated viruses seem to be most effective for vaccination against challenging pathogens like Cyprinid herpesvirus 3. However, there are still knowledge gaps how these vaccines effectively protect fish from the deadly disease caused by the epitheliotropic CyHV-3, and which aspects of non-direct protection of skin or gill integrity and function are important in the aquatic environment. To elucidate some elements of protection, common carp were vaccinated against CyHV-3 using a double deletion vaccine virus KHV-T ΔDUT/TK in the absence or presence of a mix of common carp beta-defensins 1, 2 and 3 as adjuvants. Vaccination induced marginal clinical signs, low virus load and a minor upregulation of cd4, cd8 and igm gene expression in vaccinated fish, while neutralisation activity of blood serum rose from 14 days post vaccination (dpv). A challenge infection with CyHV-3 induced a severe disease with 80-100% mortality in non-vaccinated carp, while in vaccinated carp, no mortality was recorded and the virus load was >1,000-fold lower in the skin, gill and kidney. Histological analysis showed strongest pathological changes in the skin, with a complete destruction of the epidermis in non-vaccinated carp. In the skin of non-vaccinated fish, T and B cell responses were severely downregulated, inflammation and stress responses were increased upon challenge, whereas vaccinated fish had boosted neutrophil, T and B cell responses. A disruption of skin barrier elements (tight and adherence junction, desmosomes, mucins) led to an uncontrolled increase in skin bacteria load which most likely exacerbated the inflammation and the pathology. Using a live attenuated virus vaccine, we were able to show that increased neutrophil, T and B cell responses provide protection from CyHV-3 infection and lead to preservation of skin integrity, which supports successful protection against additional pathogens in the aquatic environment which foster disease development in non-vaccinated carp.


Assuntos
Doenças dos Peixes/imunologia , Doenças dos Peixes/prevenção & controle , Infecções por Herpesviridae/veterinária , Herpesviridae/imunologia , Vacinas Virais/imunologia , Animais , Carpas , Herpesviridae/genética , Infecções por Herpesviridae/imunologia , Vacinação , Vacinas Atenuadas/imunologia , Vacinas Virais/genética
9.
Viruses ; 14(1)2022 01 06.
Artigo em Inglês | MEDLINE | ID: mdl-35062301

RESUMO

Human respiratory syncytial virus (hRSV) infection brings a wide spectrum of clinical outcomes, from a mild cold to severe bronchiolitis or even acute interstitial pneumonia. Among the known factors influencing this clinical diversity, genetic background has often been mentioned. In parallel, recent evidence has also pointed out that an early infectious experience affects heterologous infections severity. Here, we analyzed the importance of these two host-related factors in shaping the immune response in pneumoviral disease. We show that a prior gammaherpesvirus infection improves, in a genetic background-dependent manner, the immune system response against a subsequent lethal dose of pneumovirus primary infection notably by inducing a systematic expansion of the CD8+ bystander cell pool and by modifying the resident alveolar macrophages (AMs) phenotype to induce immediate cyto/chemokinic responses upon pneumovirus exposure, thereby drastically attenuating the host inflammatory response without affecting viral replication. Moreover, we show that these AMs present similar rapid and increased production of neutrophil chemokines both in front of pneumoviral or bacterial challenge, confirming recent studies attributing a critical antibacterial role of primed AMs. These results corroborate other recent studies suggesting that the innate immunity cells are themselves capable of memory, a capacity hitherto reserved for acquired immunity.


Assuntos
Patrimônio Genético , Infecções por Herpesviridae/imunologia , Macrófagos Alveolares/imunologia , Infecções por Pneumovirus/imunologia , Pneumovirus/imunologia , Rhadinovirus/imunologia , Animais , Linfócitos T CD8-Positivos/imunologia , Citocinas/metabolismo , Feminino , Infecções por Herpesviridae/genética , Infecções por Herpesviridae/patologia , Infecções por Herpesviridae/virologia , Imunidade Inata , Inflamação/imunologia , Pulmão/imunologia , Pulmão/patologia , Pulmão/virologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Monócitos/imunologia , Infecções Pneumocócicas/imunologia , Pneumovirus/fisiologia , Infecções por Pneumovirus/genética , Infecções por Pneumovirus/patologia , Infecções por Pneumovirus/virologia , Rhadinovirus/fisiologia
10.
Immunohorizons ; 6(1): 8-15, 2022 01 14.
Artigo em Inglês | MEDLINE | ID: mdl-35031582

RESUMO

NK cells are important mediators of viral control with the capacity to form adaptive immune features following infection. However, studies of infection-induced adaptive NK cells require adoptive cell transfer to lower the precursor frequency of "Ag-specific" NK cells, potentially limiting the diversity of the NK cell response. In seeking an unmanipulated model to probe the adaptive NK cells, we interrogated a wide range of Collaborative Cross (CC) inbred mice, inbred mouse strains that exhibit broad genetic diversity across strains. Our assessment identified and validated a putative "ideal" CC strain, CC006, which does not require manipulation to generate and maintain adaptive NK cells. Critically, CC006 mice, in contrast to C57BL/6 mice, are capable of developing enhanced NK cell-mediated protective responses to murine CMV infection following m157-mediated vaccination. This work both furthers our understanding of adaptive NK cells and demonstrates the utility of CC mice in the development and interrogation of immunologic models.


Assuntos
Modelos Animais de Doenças , Infecções por Herpesviridae/imunologia , Células Matadoras Naturais/imunologia , Muromegalovirus/imunologia , Transferência Adotiva , Animais , Feminino , Infecções por Herpesviridae/patologia , Células Matadoras Naturais/patologia , Ativação Linfocitária/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL
11.
J Mol Biol ; 434(6): 167214, 2022 03 30.
Artigo em Inglês | MEDLINE | ID: mdl-34437888

RESUMO

Gammaherpesviruses are ubiquitous pathogens that establish lifelong infections in the vast majority of adults worldwide. Importantly, these viruses are associated with numerous malignancies and are responsible for significant human cancer burden. These virus-associated cancers are due, in part, to the ability of gammaherpesviruses to successfully evade the innate immune response throughout the course of infection. In this review, we will summarize the current understanding of how gammaherpesviruses are detected by innate immune sensors, how these viruses evade recognition by host cells, and how this knowledge can inform novel therapeutic approaches for these viruses and their associated diseases.


Assuntos
Gammaherpesvirinae , Infecções por Herpesviridae , Evasão da Resposta Imune , Imunidade Inata , Gammaherpesvirinae/imunologia , Infecções por Herpesviridae/imunologia , Infecções por Herpesviridae/virologia , Humanos , Latência Viral
12.
Nat Commun ; 12(1): 7323, 2021 12 16.
Artigo em Inglês | MEDLINE | ID: mdl-34916520

RESUMO

T cell responses to Kaposi's sarcoma-associated herpesvirus (KSHV) are likely essential in the control of KSHV infection and protection from associated disease, but remain poorly characterised. KSHV prevalence in rural Uganda is high at >90%. Here we investigate IFN- γ T cell responses to the KSHV proteome in HIV-negative individuals from a rural Ugandan population. We use an ex-vivo IFN- γ ELISpot assay with overlapping peptide pools spanning 83 KSHV open reading frames (ORF) on peripheral blood mononuclear cells (PBMC) from 116 individuals. KSHV-specific T cell IFN- γ responses are of low intensity and heterogeneous, with no evidence of immune dominance; by contrast, IFN- γ responses to Epstein-Barr virus, Cytomegalovirus and influenza peptides are frequent and intense. Individuals with KSHV DNA in PBMC have higher IFN- γ responses to ORF73 (p = 0.02) and lower responses to K8.1 (p = 0.004) when compared with those without KSHV DNA. In summary, we demonstrate low intensity, heterogeneous T cell responses to KSHV in immune-competent individuals.


Assuntos
Infecções por Herpesviridae/imunologia , Herpesvirus Humano 8/imunologia , Linfócitos T/imunologia , Adolescente , Adulto , Idoso , Idoso de 80 Anos ou mais , Criança , Estudos de Coortes , Feminino , Anticorpos Anti-HIV/sangue , Infecções por HIV/sangue , Soronegatividade para HIV , Infecções por Herpesviridae/sangue , Infecções por Herpesviridae/virologia , Herpesvirus Humano 8/genética , Humanos , Interferon gama/imunologia , Leucócitos Mononucleares/imunologia , Masculino , Pessoa de Meia-Idade , Uganda , Adulto Jovem
13.
mBio ; 12(6): e0211321, 2021 12 21.
Artigo em Inglês | MEDLINE | ID: mdl-34933450

RESUMO

The interferon-induced transmembrane proteins (IFITMs) are broad-spectrum antiviral proteins that inhibit the entry of enveloped viruses. We analyzed the effect of IFITMs on the gamma-2 herpesviruses Kaposi's sarcoma-associated herpesvirus (KSHV) and the closely related rhesus monkey rhadinovirus (RRV). We used CRISPR/Cas9-mediated gene knockout to generate A549 cells, human foreskin fibroblasts (HFF), and human umbilical vein endothelial cells (HUVEC) with combined IFITM1/2/3 knockout and identified IFITMs as cell-dependent inhibitors of KSHV and RRV infection in A549 cells and HFF but not HUVEC. IFITM overexpression revealed IFITM1 as the relevant IFITM that inhibits KSHV and RRV infection. Fluorescent KSHV particles did not pronouncedly colocalize with IFITM-positive compartments. However, we found that KSHV and RRV glycoprotein-mediated cell-cell fusion is enhanced upon IFITM1/2/3 knockout. Taken together, we identified IFITM1 as a cell-dependent restriction factor of KSHV and RRV that acts at the level of membrane fusion. Of note, our results indicate that recombinant IFITM overexpression may lead to results that are not representative for the situation at endogenous levels. Strikingly, we observed that the endotheliotropic KSHV circumvents IFITM-mediated restriction in HUVEC despite high IFITM expression, while influenza A virus (IAV) glycoprotein-driven entry into HUVEC is potently restricted by IFITMs even in the absence of interferon. Mechanistically, we found that KSHV colocalizes less with IFITM1 and IFITM2 in HUVEC than in A549 cells immediately after attachment, potentially contributing to the observed difference in restriction. IMPORTANCE IFITM proteins are the first line of defense against infection by many pathogens and may also have therapeutic importance, as they, among other effectors, mediate the antiviral effect of interferons. Neither their function against herpesviruses nor their mechanism of action is well understood. We report here that in some cells but not in, for example, primary umbilical vein endothelial cells, IFITM1 restricts KSHV and RRV and that, mechanistically, this is likely effected by reducing the fusogenicity of the cell membrane. Further, we demonstrate potent inhibition of IAV glycoprotein-driven infection of cells of extrapulmonary origin by high constitutive IFITM expression.


Assuntos
Antígenos de Diferenciação/imunologia , Infecções por Herpesviridae/imunologia , Herpesvirus Humano 8/fisiologia , Proteínas de Membrana/imunologia , Proteínas de Ligação a RNA/imunologia , Rhadinovirus/fisiologia , Animais , Antígenos de Diferenciação/genética , Coinfecção/genética , Coinfecção/imunologia , Coinfecção/virologia , Fibroblastos/imunologia , Fibroblastos/virologia , Infecções por Herpesviridae/genética , Infecções por Herpesviridae/virologia , Herpesvirus Humano 8/genética , Interações Hospedeiro-Patógeno , Células Endoteliais da Veia Umbilical Humana/imunologia , Células Endoteliais da Veia Umbilical Humana/virologia , Humanos , Proteínas de Membrana/genética , Proteínas de Ligação a RNA/genética , Rhadinovirus/genética , Especificidade da Espécie , Internalização do Vírus , Replicação Viral
14.
Viruses ; 13(12)2021 11 26.
Artigo em Inglês | MEDLINE | ID: mdl-34960641

RESUMO

γδ T cells are activated in viral, bacterial and parasitic infections. Among viruses that promote γδ T cell mobilisation in humans, herpes viruses (HHVs) occupy a particular place since they infect the majority of the human population and persist indefinitely in the organism in a latent state. Thus, other infections should, in most instances, be considered co-infections, and the reactivation of HHV is a serious confounding factor in attributing γδ T cell alterations to a particular pathogen in human diseases. We review here the literature data on γδ T cell mobilisation in HHV infections and co-infections, and discuss the possible contribution of HHVs to γδ alterations observed in various infectious settings. As multiple infections seemingly mobilise overlapping γδ subsets, we also address the concept of possible cross-protection.


Assuntos
Coinfecção , Infecções por Herpesviridae/imunologia , Herpesviridae/imunologia , Linfócitos Intraepiteliais/imunologia , Malária/complicações , Infecções por Mycobacterium/complicações , Viroses/complicações , Imunidade Adaptativa , Infecções por Herpesviridae/complicações , Infecções por Herpesviridae/virologia , Humanos , Imunidade Inata , Linfócitos Intraepiteliais/virologia , Linfócitos/imunologia , Viroses/virologia , Latência Viral
15.
Front Immunol ; 12: 757302, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34790199

RESUMO

Roles for viral infections and aberrant immune responses in driving localized neuroinflammation and neurodegeneration in multiple sclerosis (MS) are the focus of intense research. Epstein-Barr virus (EBV), as a persistent and frequently reactivating virus with major immunogenic influences and a near 100% epidemiological association with MS, is considered to play a leading role in MS pathogenesis, triggering localized inflammation near or within the central nervous system (CNS). This triggering may occur directly via viral products (RNA and protein) and/or indirectly via antigenic mimicry involving B-cells, T-cells and cytokine-activated astrocytes and microglia cells damaging the myelin sheath of neurons. The genetic MS-risk factor HLA-DR2b (DRB1*1501ß, DRA1*0101α) may contribute to aberrant EBV antigen-presentation and anti-EBV reactivity but also to mimicry-induced autoimmune responses characteristic of MS. A central role is proposed for inflammatory EBER1, EBV-miRNA and LMP1 containing exosomes secreted by viable reactivating EBV+ B-cells and repetitive release of EBNA1-DNA complexes from apoptotic EBV+ B-cells, forming reactive immune complexes with EBNA1-IgG and complement. This may be accompanied by cytokine- or EBV-induced expression of human endogenous retrovirus-W/-K (HERV-W/-K) elements and possibly by activation of human herpesvirus-6A (HHV-6A) in early-stage CNS lesions, each contributing to an inflammatory cascade causing the relapsing-remitting neuro-inflammatory and/or progressive features characteristic of MS. Elimination of EBV-carrying B-cells by antibody- and EBV-specific T-cell therapy may hold the promise of reducing EBV activity in the CNS, thereby limiting CNS inflammation, MS symptoms and possibly reversing disease. Other approaches targeting HHV-6 and HERV-W and limiting inflammatory kinase-signaling to treat MS are also being tested with promising results. This article presents an overview of the evidence that EBV, HHV-6, and HERV-W may have a pathogenic role in initiating and promoting MS and possible approaches to mitigate development of the disease.


Assuntos
Retrovirus Endógenos/patogenicidade , Herpesvirus Humano 4/patogenicidade , Herpesvirus Humano 6/patogenicidade , Esclerose Múltipla/etiologia , Doenças Neuroinflamatórias/virologia , Anticorpos Antivirais/imunologia , Complexo Antígeno-Anticorpo/imunologia , Autoimunidade , Linfócitos B/imunologia , Barreira Hematoencefálica , Encéfalo/virologia , Coinfecção , DNA Viral/imunologia , Retrovirus Endógenos/fisiologia , Antígenos Nucleares do Vírus Epstein-Barr/genética , Antígenos Nucleares do Vírus Epstein-Barr/imunologia , Produtos do Gene env/fisiologia , Predisposição Genética para Doença , Infecções por Herpesviridae/complicações , Infecções por Herpesviridae/imunologia , Infecções por Herpesviridae/virologia , Herpesvirus Humano 4/imunologia , Herpesvirus Humano 6/imunologia , Humanos , Linfonodos/virologia , Modelos Imunológicos , Mimetismo Molecular , Esclerose Múltipla/imunologia , Esclerose Múltipla/terapia , Esclerose Múltipla/virologia , Bainha de Mielina/imunologia , Bainha de Mielina/patologia , Doenças Neuroinflamatórias/etiologia , Proteínas da Gravidez/fisiologia , Ativação Transcricional , Ativação Viral , Latência Viral
16.
Front Immunol ; 12: 702971, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34531856

RESUMO

Polyploidy and subsequent diploidization provide genomic opportunities for evolutionary innovations and adaptation. The researches on duplicated gene evolutionary fates in recurrent polyploids have seriously lagged behind that in paleopolyploids with diploidized genomes. Moreover, the antiviral mechanisms of Viperin remain largely unclear in fish. Here, we elaborate the distinct antiviral mechanisms of two viperin homeologs (Cgviperin-A and Cgviperin-B) in auto-allo-hexaploid gibel carp (Carassius gibelio). First, Cgviperin-A and Cgviperin-B showed differential and biased expression patterns in gibel carp adult tissues. Subsequently, using co-immunoprecipitation (Co-IP) screening analysis, both CgViperin-A and CgViperin-B were found to interact with crucian carp (C. auratus) herpesvirus (CaHV) open reading frame 46 right (ORF46R) protein, a negative herpesvirus regulator of host interferon (IFN) production, and to promote the proteasomal degradation of ORF46R via decreasing K63-linked ubiquitination. Additionally, CgViperin-B also mediated ORF46R degradation through autophagosome pathway, which was absent in CgViperin-A. Moreover, we found that the N-terminal α-helix domain was necessary for the localization of CgViperin-A and CgViperin-B at the endoplasmic reticulum (ER), and the C-terminal domain of CgViperin-A and CgViperin-B was indispensable for the interaction with degradation of ORF46R. Therefore, the current findings clarify the divergent antiviral mechanisms of the duplicated viperin homeologs in a recurrent polyploid fish, which will shed light on the evolution of teleost duplicated genes.


Assuntos
Carpas , Doenças dos Peixes , Proteínas de Peixes , Infecções por Herpesviridae , Herpesviridae/imunologia , Poliploidia , Proteína Viperina , Animais , Carpas/genética , Carpas/imunologia , Carpas/virologia , Linhagem Celular , Doenças dos Peixes/genética , Doenças dos Peixes/imunologia , Proteínas de Peixes/genética , Proteínas de Peixes/imunologia , Infecções por Herpesviridae/genética , Infecções por Herpesviridae/imunologia , Infecções por Herpesviridae/veterinária , Proteína Viperina/genética , Proteína Viperina/imunologia
17.
Int J Mol Sci ; 22(17)2021 Sep 06.
Artigo em Inglês | MEDLINE | ID: mdl-34502533

RESUMO

Various intrinsic and extrinsic factors can interfere with the process of protein folding, resulting in protein aggregates. Usually, cells prevent the formation of aggregates or degrade them to prevent the cytotoxic effects they may cause. However, during viral infection, the formation of aggregates may serve as a cellular defense mechanism. On the other hand, some viruses are able to exploit the process of aggregate formation and removal to promote their replication or evade the immune response. This review article summarizes the process of cellular protein aggregation and gives examples of how different viruses exploit it. Particular emphasis is placed on the ribonucleotide reductases of herpesviruses and how their additional non-canonical functions in viral immune evasion are closely linked to protein aggregation.


Assuntos
Evasão da Resposta Imune/imunologia , Agregados Proteicos , Agregação Patológica de Proteínas/imunologia , Viroses/imunologia , Vírus/imunologia , Herpesviridae/imunologia , Herpesviridae/fisiologia , Infecções por Herpesviridae/imunologia , Infecções por Herpesviridae/metabolismo , Infecções por Herpesviridae/virologia , Interações Hospedeiro-Patógeno/imunologia , Humanos , Agregação Patológica de Proteínas/metabolismo , Agregação Patológica de Proteínas/virologia , Ribonucleotídeo Redutases/imunologia , Ribonucleotídeo Redutases/metabolismo , Viroses/metabolismo , Viroses/virologia
18.
Viral Immunol ; 34(8): 552-558, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-34463142

RESUMO

Infectious laryngotracheitis virus (ILTV) is a cause of main respiratory disease of chickens controlled through live attenuated vaccines. To reduce the risk of adverse effects associated with live vaccines, a recombinant vaccine expressing PH-1 domain of viral glycoprotein B was constructed using the pET expression system under isopropylthiogalactoside (IPTG) induction. The potential immunogenicity of recombinant PH-1 (rPH-1) was evaluated in chickens. Eight-week-old specific-pathogen-free chickens were intramuscularly administered two doses of rPH-1, 25 and 50 µg, alone or with a combination of ISA70 adjuvant. The humoral immune responses were determined up to 3 months postvaccination at 2 weeks apart. The T cell proliferation response was determined on day 28 after primary immunization. The vaccinated birds with rPH-1/ISA70 developed higher and constant-specific anti-ILTV enzyme-linked immunosorbent assay (ELISA) antibodies than in those vaccinated with rPH-1 alone. Coinjection of rPH-1 and adjuvant significantly (p < 0.01) increased the T cell proliferation responses. There were no significant differences in eliciting the immune responses in chickens immunized with the higher dose of the antigen than that with the lower dose. The data indicate the immunogenic efficiency of rPH-1 against ILTV. Vaccination with recombinant proteins offers a preventing option to control the ILTV infection and could be a candidate to replace current live vaccines.


Assuntos
Infecções por Herpesviridae , Herpesvirus Galináceo 1 , Doenças das Aves Domésticas , Proteínas do Envelope Viral , Vacinas Virais , Animais , Anticorpos Antivirais , Galinhas , Infecções por Herpesviridae/imunologia , Herpesvirus Galináceo 1/imunologia , Imunidade , Vacinas Atenuadas , Proteínas do Envelope Viral/imunologia
19.
Immunity ; 54(10): 2288-2304.e7, 2021 10 12.
Artigo em Inglês | MEDLINE | ID: mdl-34437840

RESUMO

Upon viral infection, natural killer (NK) cells expressing certain germline-encoded receptors are selected, expanded, and maintained in an adaptive-like manner. Currently, these are thought to differentiate along a common pathway. However, by fate mapping of single NK cells upon murine cytomegalovirus (MCMV) infection, we identified two distinct NK cell lineages that contributed to adaptive-like responses. One was equivalent to conventional NK (cNK) cells while the other was transcriptionally similar to type 1 innate lymphoid cells (ILC1s). ILC1-like NK cells showed splenic residency and strong cytokine production but also recognized and killed MCMV-infected cells, guided by activating receptor Ly49H. Moreover, they induced clustering of conventional type 1 dendritic cells and facilitated antigen-specific T cell priming early during MCMV infection, which depended on Ly49H and the NK cell-intrinsic expression of transcription factor Batf3. Thereby, ILC1-like NK cells bridge innate and adaptive viral recognition and unite critical features of cNK cells and ILC1s.


Assuntos
Imunidade Adaptativa/imunologia , Linhagem da Célula/imunologia , Infecções por Herpesviridae/imunologia , Imunidade Inata/imunologia , Células Matadoras Naturais/imunologia , Animais , Feminino , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Muromegalovirus
20.
Cells ; 10(8)2021 08 18.
Artigo em Inglês | MEDLINE | ID: mdl-34440891

RESUMO

Infection of a host cell by an invading viral pathogen triggers a multifaceted antiviral response. One of the most potent defense mechanisms host cells possess is the interferon (IFN) system, which initiates a targeted, coordinated attack against various stages of viral infection. This immediate innate immune response provides the most proximal defense and includes the accumulation of antiviral proteins, such as IFN-stimulated genes (ISGs), as well as a variety of protective cytokines. However, viruses have co-evolved with their hosts, and as such, have devised distinct mechanisms to undermine host innate responses. As large, double-stranded DNA viruses, herpesviruses rely on a multitude of means by which to counter the antiviral attack. Herein, we review the various approaches the human herpesviruses employ as countermeasures to the host innate immune response.


Assuntos
Infecções por Herpesviridae/imunologia , Imunidade Inata/fisiologia , Animais , Infecções por Herpesviridae/metabolismo , Humanos , Imunidade Inata/genética , Transdução de Sinais/fisiologia , Replicação Viral/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...