Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.753
Filtrar
Mais filtros











Intervalo de ano de publicação
1.
J Clin Invest ; 134(9)2024 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-38690731

RESUMO

Herpesviruses establish latent infections, and most reactivate frequently, resulting in symptoms and virus shedding in healthy individuals. In immunocompromised patients, reactivating virus can cause severe disease. Persistent EBV has been associated with several malignancies in both immunocompromised and nonimmunocompromised persons. Reactivation and shedding occur with most herpesviruses, despite potent virus-specific antibodies and T cell immunity as measured in the blood. The licensure of therapeutic vaccines to reduce zoster indicates that effective therapeutic vaccines for other herpesviruses should be feasible. However, varicella-zoster virus is different from other human herpesviruses in that it is generally only shed during varicella and zoster. Unlike prophylactic vaccines, in which the correlate of immunity is antibody function, T cell immunity is the correlate of immunity for the only effective therapeutic herpesvirus vaccine-zoster vaccine. While most studies of therapeutic vaccines have measured immunity in the blood, cellular immunity at the site of reactivation is likely critical for an effective therapeutic vaccine for certain viruses. This Review summarizes the status of therapeutic vaccines for herpes simplex virus, cytomegalovirus, and Epstein-Barr virus and proposes approaches for future development.


Assuntos
Vacinas contra Herpesvirus , Humanos , Vacinas contra Herpesvirus/imunologia , Vacinas contra Herpesvirus/uso terapêutico , Infecções por Herpesviridae/imunologia , Infecções por Herpesviridae/prevenção & controle , Infecções por Herpesviridae/virologia , Herpesvirus Humano 4/imunologia , Animais , Herpesviridae/imunologia , Ativação Viral/imunologia , Citomegalovirus/imunologia
2.
Viruses ; 16(4)2024 04 16.
Artigo em Inglês | MEDLINE | ID: mdl-38675960

RESUMO

Reactivation and infection with cytomegalovirus (CMV) are frequently observed in recipients of solid organ transplants, bone marrow transplants, and individuals with HIV infection. This presents an increasing risk of allograft rejection, opportunistic infection, graft failure, and patient mortality. Among immunocompromised hosts, interstitial pneumonia is the most critical clinical manifestation of CMV infection. Recent studies have demonstrated the potential therapeutic benefits of exosomes derived from mesenchymal stem cells (MSC-exos) in preclinical models of acute lung injury, including pneumonia, ARDS, and sepsis. However, the role of MSC-exos in the pathogenesis of infectious viral diseases, such as CMV pneumonia, remains unclear. In a mouse model of murine CMV-induced pneumonia, we observed that intravenous administration of mouse MSC (mMSC)-exos reduced lung damage, decreased the hyperinflammatory response, and shifted macrophage polarization from the M1 to the M2 phenotype. Treatment with mMSC-exos also significantly reduced the infiltration of inflammatory cells and pulmonary fibrosis. Furthermore, in vitro studies revealed that mMSC-exos reversed the hyperinflammatory phenotype of bone marrow-derived macrophages infected with murine CMV. Mechanistically, mMSC-exos treatment decreased activation of the NF-κB/NLRP3 signaling pathway both in vivo and in vitro. In summary, our findings indicate that mMSC-exo treatment is effective in severe CMV pneumonia by reducing lung inflammation and fibrosis through the NF-κB/NLRP3 signaling pathway, thus providing promising therapeutic potential for clinical CMV infection.


Assuntos
Modelos Animais de Doenças , Exossomos , Células-Tronco Mesenquimais , Muromegalovirus , NF-kappa B , Proteína 3 que Contém Domínio de Pirina da Família NLR , Transdução de Sinais , Animais , Exossomos/metabolismo , Células-Tronco Mesenquimais/metabolismo , Camundongos , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , NF-kappa B/metabolismo , Muromegalovirus/fisiologia , Camundongos Endogâmicos C57BL , Macrófagos/imunologia , Infecções por Citomegalovirus/terapia , Infecções por Citomegalovirus/virologia , Pulmão/virologia , Pulmão/patologia , Pneumonia Viral/terapia , Pneumonia Viral/virologia , Infecções por Herpesviridae/terapia , Infecções por Herpesviridae/virologia , Infecções por Herpesviridae/imunologia , Pneumonia/terapia , Pneumonia/virologia
3.
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 , Modelos Animais , Biossíntese de Proteínas , Transcrição Gênica , Animais , Camundongos , Comunicação Celular/imunologia , Quimiocina CCL3/genética , Quimiocina CCL3/imunologia , Técnicas de Introdução de Genes , Camundongos Transgênicos , Muromegalovirus , Biossíntese de Proteínas/efeitos dos fármacos , Biossíntese de Proteínas/imunologia , Transcrição Gênica/imunologia , Células Matadoras Naturais/imunologia , Interferon beta/farmacologia , Infecções por Herpesviridae/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 da 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.
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
9.
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
10.
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
11.
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
12.
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
13.
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
14.
Front Immunol ; 12: 687151, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34290708

RESUMO

Anti-disease breeding is becoming the most promising solution to cyprinid herpesvirus-3 (CyHV-3) infection, the major threat to common carp aquaculture. Virus challenging studies suggested that a breeding strain of common carp developed resistance to CyHV-3 infection. This study illustrates the immune mechanisms involved in both sensitivity and anti-virus ability for CyHV3 infection in fish. An integrative analysis of the protein-coding genes and long non-coding RNAs (lncRNAs) using transcriptomic data was performed. Tissues from the head kidney of common carp were extracted at days 0 (the healthy control) and 7 after CyHV-3 infection (the survivors) and used to analyze the transcriptome through both Illumina and PacBio sequencing. Following analysis of the GO terms and KEGG pathways involved, the immune-related terms and pathways were merged. To dig out details on the immune aspect, the DEGs were filtered using the current common carp immune gene library. Immune gene categories and their corresponding genes in different comparison groups were revealed. Also, the immunological Gene Ontology terms for lncRNA modulation were retained. The weighted gene co-expression network analysis was used to reveal the regulation of immune genes by lncRNA. The results demonstrated that the breeding carp strain develops a marked resistance to CyHV-3 infection through a specific innate immune mechanism. The featured biological processes were autophagy, phagocytosis, cytotoxicity, and virus blockage by lectins and MUC3. Moreover, the immune-suppressive signals, such as suppression of IL21R on STAT3, PI3K mediated inhibition of inflammation by dopamine upon infection, as well as the inhibition of NLRC3 on STING during a steady state. Possible susceptible factors for CyHV-3, such as ITGB1, TLR18, and CCL4, were also revealed from the non-breeding strain. The results of this study also suggested that Nramp and PAI regulated by LncRNA could facilitate virus infection and proliferation for infected cells respectively, while T cell leukemia homeobox 3 (TLX3), as well as galectin 3 function by lncRNA, may play a role in the resistance mechanism. Therefore, immune factors that are immunogenetically insensitive or susceptible to CyHV-3 infection have been revealed.


Assuntos
Carpas/genética , Carpas/imunologia , Doenças dos Peixes/genética , Doenças dos Peixes/imunologia , Infecções por Herpesviridae/veterinária , Imunidade Inata/genética , Animais , Carpas/virologia , Suscetibilidade a Doenças , Doenças dos Peixes/virologia , Perfilação da Expressão Gênica , Rim Cefálico/patologia , Herpesviridae/imunologia , Herpesviridae/fisiologia , Infecções por Herpesviridae/imunologia , Infecções por Herpesviridae/virologia , Sequenciamento de Nucleotídeos em Larga Escala
15.
Front Immunol ; 12: 681380, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34168650

RESUMO

Viral vectors have emerged as a promising alternative to classical vaccines due to their great potential for induction of a potent cellular and humoral immunity. Cytomegalovirus (CMV) is an attractive vaccine vector due to its large genome with many non-essential immunoregulatory genes that can be easily manipulated to modify the immune response. CMV generates a strong antigen-specific CD8 T cell response with a gradual accumulation of these cells in the process called memory inflation. In our previous work, we have constructed a mouse CMV vector expressing NKG2D ligand RAE-1γ in place of its viral inhibitor m152 (RAE-1γMCMV), which proved to be highly attenuated in vivo. Despite attenuation, RAE-1γMCMV induced a substantially stronger CD8 T cell response to vectored antigen than the control vector and provided superior protection against bacterial and tumor challenge. In the present study, we confirmed the enhanced protective capacity of RAE-1γMCMV as a tumor vaccine vector and determined the phenotypical and functional characteristics of memory CD8 T cells induced by the RAE-1γ expressing MCMV. RNAseq data revealed higher transcription of numerous genes associated with effector-like CD8 T cell phenotype in RAE-1γMCMV immunized mice. CD8 T cells primed with RAE-1γMCMV were enriched in TCF1 negative population, with higher expression of KLRG1 and lower expression of CD127, CD27, and Eomes. These phenotypical differences were associated with distinct functional features as cells primed with RAE-1γMCMV showed inferior cytokine-producing abilities but comparable cytotoxic potential. After adoptive transfer into naive hosts, OT-1 cells induced with both RAE-1γMCMV and the control vector were equally efficient in rejecting established tumors, suggesting the context of latent infection and cell numbers as important determinants of enhanced anti-tumor response following RAE-1γMCMV vaccination. Overall, our results shed new light on the phenotypical and functional distinctness of memory CD8 T cells induced with CMV vector expressing cellular ligand for the NKG2D receptor.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/metabolismo , Vacinas contra Citomegalovirus/imunologia , Memória Imunológica , Subfamília K de Receptores Semelhantes a Lectina de Células NK/genética , Fenótipo , Animais , Vacinas Anticâncer/imunologia , Biologia Computacional/métodos , Citomegalovirus/imunologia , Citotoxicidade Imunológica , Perfilação da Expressão Gênica , Infecções por Herpesviridae/imunologia , Infecções por Herpesviridae/prevenção & controle , Imunofenotipagem , Ativação Linfocitária/imunologia , Camundongos , Muromegalovirus/imunologia , Subfamília K de Receptores Semelhantes a Lectina de Células NK/metabolismo , Transcriptoma
16.
Mol Cell ; 81(13): 2823-2837.e9, 2021 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-34015248

RESUMO

DNA-induced liquid-liquid phase separation of cyclic GMP-AMP synthase (cGAS) triggers a potent response to detect pathogen infection and promote innate immune signaling. Whether and how pathogens manipulate cGAS-DNA condensation to mediate immune evasion is unknown. We report the identification of a structurally related viral tegument protein family, represented by ORF52 and VP22 from gamma- and alpha-herpesvirinae, respectively, that employs a conserved mechanism to restrict cGAS-DNA phase separation. ORF52/VP22 proteins accumulate into, and effectively disrupt, the pre-formed cGAS-DNA condensation both in vitro and in cells. The inhibition process is dependent on DNA-induced liquid-liquid phase separation of the viral protein rather than a direct interaction with cGAS. Moreover, highly abundant ORF52 proteins carried within viral particles are able to target cGAS-DNA phase separation in early infection stage. Our results define ORF52/VP22-type tegument proteins as a family of inhibitors targeting cGAS-DNA phase separation and demonstrate a mechanism for how viruses overcome innate immunity.


Assuntos
Alphaherpesvirinae , Betaherpesvirinae , DNA , Infecções por Herpesviridae , Evasão da Resposta Imune , Nucleotidiltransferases , Proteínas Estruturais Virais , Alphaherpesvirinae/química , Alphaherpesvirinae/genética , Alphaherpesvirinae/imunologia , Betaherpesvirinae/química , Betaherpesvirinae/genética , Betaherpesvirinae/imunologia , DNA/química , DNA/genética , DNA/imunologia , Células HEK293 , Células HeLa , Infecções por Herpesviridae/genética , Infecções por Herpesviridae/imunologia , Humanos , Imunidade Inata , Nucleotidiltransferases/química , Nucleotidiltransferases/genética , Nucleotidiltransferases/imunologia , Proteínas Estruturais Virais/química , Proteínas Estruturais Virais/genética , Proteínas Estruturais Virais/imunologia
17.
BMC Cancer ; 21(1): 468, 2021 Apr 27.
Artigo em Inglês | MEDLINE | ID: mdl-33906629

RESUMO

BACKGROUND: Primary effusion lymphoma is a rare, aggressive large B-cell lymphoma strictly linked to infection by Human Herpes virus 8/Kaposi sarcoma-associated herpes virus. In its classic form, it is characterized by body cavities neoplastic effusions without detectable tumor masses. It often occurs in immunocompromised patients, such as HIV-positive individuals. Primary effusion lymphoma may affect HIV-negative elderly patients from Human Herpes virus 8 endemic regions. So far, rare cases have been reported in transplanted patients. The purpose of our systematic review is to improve our understanding of this type of aggressive lymphoma in the setting of transplantation, focusing on epidemiology, clinical presentation, pathological features, differential diagnosis, treatment and outcome. The role of assessing the viral serological status in donors and recipients is also discussed. METHODS: We performed a systematic review adhering to the PRISMA guidelines. The literature search was conducted on PubMed/MEDLINE, Web of Science, Scopus, EMBASE and Cochrane Library, using the search terms "primary effusion lymphoma" and "post-transplant". RESULTS: Our search identified 13 cases of post-transplant primary effusion lymphoma, predominantly in solid organ transplant recipients (6 kidney, 3 heart, 2 liver and 1 intestine), with only one case after allogenic bone marrow transplantation. Long-term immunosuppression is important in post-transplant primary effusion lymphoma commonly developing several years after transplantation. Kaposi Sarcoma occurred in association with lymphoma in 4 cases of solid organ recipients. The lymphoma showed the classical presentation with body cavity effusions in absence of tumor masses in 10 cases; 2 cases presented as solid masses, lacking effusions and one case as effusions associated with multiple organ involvement. Primary effusion lymphoma occurring in the setting of transplantation was more often Epstein Barr-virus negative. The prognosis was poor. In addition to chemotherapy, reduction of immunosuppressive treatment, was generally attempted. CONCLUSIONS: Primary effusion lymphoma is a rare, but often fatal post-transplant complication. Its rarity and the difficulty in achieving the diagnosis may lead to miss this complication. Clinicians should suspect primary effusion lymphoma in transplanted patients, presenting generally with unexplained body cavity effusions, although rare cases with solid masses are described.


Assuntos
Transplantados , Transplante de Medula Óssea , Diagnóstico Diferencial , Transplante de Coração , Infecções por Herpesviridae/imunologia , Herpesvirus Humano 8/imunologia , Humanos , Hospedeiro Imunocomprometido , Intestinos/transplante , Transplante de Rim , Transplante de Fígado , Linfoma de Efusão Primária/epidemiologia , Linfoma de Efusão Primária/patologia , Linfoma de Efusão Primária/virologia , Complicações Pós-Operatórias/epidemiologia , Complicações Pós-Operatórias/patologia , Complicações Pós-Operatórias/virologia , Doenças Raras/epidemiologia , Doenças Raras/patologia , Doenças Raras/virologia , Sarcoma de Kaposi
18.
Dev Comp Immunol ; 121: 104102, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-33862099

RESUMO

CD63 is a member of the four-transmembrane-domain protein superfamily and is the first characterized tetraspanin protein. In the present study, we cloned the common carp (Cyprinus Carpio) CD63 (ccCD63) sequence and found that the ccCD63 ORF contained 711 bp and encoded a protein of 236 amino acids. Homology analysis revealed that the complete ccCD63 sequence had 84.08% amino acid similarity to CD63 of Sinocyclocheilus anshuiensis. Subcellular localization analysis revealed that ccCD63 was localized in the cytoplasm. Quantitative real-time PCR (qRT-PCR) analysis indicated that ccCD63 was expressed in the gill, intestine, liver, spleen, brain and kidney, with higher expression in spleen and brain tissues than in the other examined tissues. After koi herpesvirus (KHV) infection, these tissues exhibited various expression levels of ccCD63. The expression level was the lowest in the liver and highest in the brain; the expression level in the brain was 8.7-fold higher than that in the liver. Furthermore, knockdown of ccCD63 promoted KHV infection. Moreover, ccCD63 was correlated with the regulation of RIG-I/MAVS/TRAF3/TBK1/IRF3 and may be involved in the antiviral response through the RIG-I viral recognition signalling pathway in a TRAF3/TBK1-dependent manner. Taken together, our results suggested that ccCD63 upregulated the interaction of KHV with the host immune system and suppressed the dissemination of KHV.


Assuntos
Carpas/imunologia , Doenças dos Peixes/imunologia , Proteínas de Peixes/metabolismo , Infecções por Herpesviridae/veterinária , Tetraspanina 30/metabolismo , Animais , Carpas/genética , Carpas/virologia , Clonagem Molecular , DNA Viral , Doenças dos Peixes/virologia , Proteínas de Peixes/genética , Técnicas de Silenciamento de Genes , Brânquias , Herpesviridae/imunologia , Infecções por Herpesviridae/imunologia , Infecções por Herpesviridae/virologia , Interações Hospedeiro-Patógeno/imunologia , Transdução de Sinais/imunologia , Tetraspanina 30/genética
19.
mBio ; 12(2)2021 04 06.
Artigo em Inglês | MEDLINE | ID: mdl-33824206

RESUMO

Gammaherpesviruses establish lifelong infection and are associated with a variety of cancers, including B cell lymphomas. These viruses manipulate the B cell differentiation process to establish lifelong infection in memory B cells. Specifically, gammaherpesviruses infect naive B cells and promote entry of both infected and uninfected naive B cells into germinal centers, where the virus usurps rapid proliferation of germinal center B cells to exponentially increase its cellular latent reservoir. In addition to facilitating the establishment of latent infection, germinal center B cells are thought to be the target of viral transformation. In this study, we have uncovered a novel proviral role of host interleukin 17A (IL-17A), a well-established antibacterial and antifungal factor. Loss of IL-17A signaling attenuated the establishment of chronic gammaherpesvirus infection and gammaherpesvirus-driven germinal center response in a route of inoculation-dependent manner. Further, IL-17A treatment directly supported gammaherpesvirus reactivation and de novo lytic infection. This study is the first demonstration of a multifaceted proviral role of IL-17 signaling.IMPORTANCE Gammaherpesviruses establish lifelong infections in a majority of humans and are associated with B cell lymphomas. IL-17A is a host cytokine that plays a well-established role in the clearance of bacterial and fungal infections; however, the role of IL-17A in viral infections is poorly understood. In this study, we show that IL-17A signaling promoted the establishment of chronic gammaherpesvirus infection following the mucosal route of infection, viral lytic replication, and reactivation from latency. Thus, our study unveils a novel proviral role of IL-17A signaling in gammaherpesvirus infection.


Assuntos
Gammaherpesvirinae/imunologia , Infecções por Herpesviridae/imunologia , Interações Hospedeiro-Patógeno/imunologia , Interleucina-17/imunologia , Transdução de Sinais/imunologia , Animais , Células Cultivadas , Doença Crônica , Feminino , Interações Hospedeiro-Patógeno/genética , Interleucina-17/genética , Macrófagos/imunologia , Macrófagos/virologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL
20.
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
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA