Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 394
Filtrar
1.
Intervirology ; 65(3): 144-150, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35158367

RESUMEN

INTRODUCTION: Epstein-Barr virus (EBV/HHV-4) has been implicated in the pathogenesis of multiple sclerosis (MS). This study was conducted to investigate the levels of pro-inflammatory cytokines IL-1ß and IL-6 in healthy EBV carriers and MS patients with prior EBV infection in response to treatment with EBV nuclear antigen 1 (EBNA-1) and replication and transcription activator (BRLF-1/Rta) peptide antigens in whole blood cell culture to assess the cytokine expression across all cells in the peripheral blood. METHODS: Isolated whole blood cells from the included participants were incubated at a concentration of 106 cells/mL with BRLF-1 or EBNA-1. The amount of IL-1ß and IL-6 transcripts were measured with quantitative RT-PCR at day 3 after incubation. MTT assay was conducted to examine cytotoxicity of the peptides and their effect on cell viability. Changes in cytokine expression and cell viability were analyzed using one-way and two-way ANOVA, respectively. RESULTS: Ten MS patients and ten healthy donors were enrolled in the study. Treatment with the peptide antigens resulted in increased cytokines expression in both MS patients and healthy subjects. Furthermore, IL-1ß levels were higher in MS patients compared to healthy EBV carriers. MTT assay revealed no significant difference in cell viability between the two groups. DISCUSSION: The higher levels of IL-1ß in response to EBV antigens in MS patients may reflect the host neuroinflammatory environment and support the notion that immune response against EBV has a role as an aggravating factor in the progression of MS by contributing to the neuroinflammatory cascade.


Asunto(s)
Infecciones por Virus de Epstein-Barr , Antígenos Nucleares del Virus de Epstein-Barr , Proteínas Inmediatas-Precoces , Esclerosis Múltiple , Transactivadores , Citocinas/metabolismo , Infecciones por Virus de Epstein-Barr/complicaciones , Antígenos Nucleares del Virus de Epstein-Barr/inmunología , Herpesvirus Humano 4 , Humanos , Proteínas Inmediatas-Precoces/inmunología , Interleucina-1beta/metabolismo , Interleucina-6/metabolismo , Esclerosis Múltiple/tratamiento farmacológico , Transactivadores/inmunología
2.
Front Immunol ; 12: 743466, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34925320

RESUMEN

In the process of infecting the host, alphaherpesviruses have derived a series of adaptation and survival strategies, such as latent infection, autophagy and immune evasion, to survive in the host environment. Infected cell protein 22 (ICP22) or its homologue immediate early protein 63 (IE63) is a posttranslationally modified multifunctional viral regulatory protein encoded by all alphaherpesviruses. In addition to playing an important role in the efficient use of host cell RNA polymerase II, it also plays an important role in the defense process of the virus overcoming the host immune system. These two effects of ICP22/IE63 are important survival strategies for alphaherpesviruses. In this review, we summarize the complex mechanism by which the ICP22 protein regulates the transcription of alphaherpesviruses and their host genes and the mechanism by which ICP22/IE63 participates in immune escape. Reviewing these mechanisms will also help us understand the pathogenesis of alphaherpesvirus infections and provide new strategies to combat these viral infections.


Asunto(s)
Alphaherpesvirinae/fisiología , Regulación Viral de la Expresión Génica/fisiología , Proteínas Inmediatas-Precoces/inmunología , Evasión Inmune/fisiología , Animales , Infecciones por Herpesviridae , Humanos , Proteínas Inmediatas-Precoces/metabolismo
3.
Viruses ; 13(11)2021 11 17.
Artículo en Inglés | MEDLINE | ID: mdl-34835102

RESUMEN

Following acute infection, herpes simplex virus 1 (HSV-1) establishes lifelong latency in neurons, including sensory neurons within trigeminal ganglia. During latency, lytic cycle viral gene expression is silenced. However, stressful stimuli can trigger reactivation from latency. The viral tegument protein, VP-16, transactivates all immediate early (IE) promoters during productive infection. Conversely, cellular factors are expected to trigger viral gene expression during early stages of reactivation from latency and in non-neuronal cells that do not support high levels of productive infection. The glucocorticoid receptor (GR), synthetic corticosteroid dexamethasone, and certain stress-induced transcription factors cooperatively transactivate infected cell protein 0 (ICP0) and ICP4 promoters. Since ICP27 protein expression is required for productive infection, we hypothesized that the ICP27 promoter is transactivated by stress-induced transcription factors. New studies have demonstrated that ICP27 enhancer sequences were transactivated by GR and Krüppel-like factor 15 (KLF15). Mutation of a consensus Sp1 binding site within ICP27 enhancer sequences impaired transactivation by GR and KLF15. Chromatin immunoprecipitation studies have demonstrated that GR and KLF15 occupy ICP27 promoter sequences during productive infection. Cells transfected with an ICP27 enhancer fragment revealed the GR and KLF15 occupancy of ICP27 enhancer sequences required the intact Sp1 binding site. Notably, GR and KLF15 form a feed-forward transcription loop in response to stress, suggesting these cellular factors promote viral replication following stressful stimuli.


Asunto(s)
Herpes Simple/inmunología , Herpes Simple/virología , Herpesvirus Humano 1/fisiología , Proteínas Inmediatas-Precoces/inmunología , Receptores de Glucocorticoides/inmunología , Latencia del Virus , Animales , Línea Celular Tumoral , Chlorocebus aethiops , Inmunoprecipitación de Cromatina , Regulación Viral de la Expresión Génica , Factores de Transcripción de Tipo Kruppel/inmunología , Ratones , Factores de Transcripción , Células Vero , Activación Viral
4.
Nature ; 600(7887): 138-142, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34759314

RESUMEN

Pathogens use virulence factors to inhibit the immune system1. The guard hypothesis2,3 postulates that hosts monitor (or 'guard') critical innate immune pathways such that their disruption by virulence factors provokes a secondary immune response1. Here we describe a 'self-guarded' immune pathway in human monocytes, in which guarding and guarded functions are combined in one protein. We find that this pathway is triggered by ICP0, a key virulence factor of herpes simplex virus type 1, resulting in robust induction of anti-viral type I interferon (IFN). Notably, induction of IFN by ICP0 is independent of canonical immune pathways and the IRF3 and IRF7 transcription factors. A CRISPR screen identified the ICP0 target MORC34 as an essential negative regulator of IFN. Loss of MORC3 recapitulates the IRF3- and IRF7-independent IFN response induced by ICP0. Mechanistically, ICP0 degrades MORC3, which leads to de-repression of a MORC3-regulated DNA element (MRE) adjacent to the IFNB1 locus. The MRE is required in cis for IFNB1 induction by the MORC3 pathway, but is not required for canonical IFN-inducing pathways. As well as repressing the MRE to regulate IFNB1, MORC3 is also a direct restriction factor of HSV-15. Our results thus suggest a model in which the primary anti-viral function of MORC3 is self-guarded by its secondary IFN-repressing function-thus, a virus that degrades MORC3 to avoid its primary anti-viral function will unleash the secondary anti-viral IFN response.


Asunto(s)
Adenosina Trifosfatasas/inmunología , Proteínas de Unión al ADN/inmunología , Modelos Inmunológicos , Factores de Virulencia/inmunología , Adenosina Trifosfatasas/deficiencia , Adenosina Trifosfatasas/metabolismo , Sistemas CRISPR-Cas , Línea Celular , Proteínas de Unión al ADN/deficiencia , Proteínas de Unión al ADN/metabolismo , Edición Génica , Herpesvirus Humano 1/inmunología , Herpesvirus Humano 1/patogenicidad , Humanos , Proteínas Inmediatas-Precoces/inmunología , Inmunidad Innata , Factor 3 Regulador del Interferón/metabolismo , Factor 7 Regulador del Interferón/metabolismo , Interferón Tipo I/antagonistas & inhibidores , Interferón Tipo I/genética , Interferón Tipo I/inmunología , Monocitos/inmunología , Receptor de Interferón alfa y beta , Proteínas Represoras/deficiencia , Proteínas Represoras/inmunología , Proteínas Represoras/metabolismo , Elementos de Respuesta/genética , Ubiquitina-Proteína Ligasas/inmunología
5.
J Gen Virol ; 102(11)2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34726593

RESUMEN

Kaposi's sarcoma-associated herpesvirus (KSHV) is an oncogenic etiological factor for Kaposi's sarcoma and primary effusion lymphoma in immunocompromised patients. KSHV utilizes two immune evasion E3 ubiquitin ligases, namely K3 and K5, to downregulate the expression of antigen-presenting molecules and ligands of natural killer (NK) cells in the host cells through an ubiquitin-dependent endocytic mechanism. This allows the infected cells to evade surveillance and elimination by cytotoxic lymphocytes and NK cells. The number of host cell molecular substrates reported for these ubiquitin ligases is limited. The identification of novel substrates for these ligases will aid in elucidating the mechanism underlying immune evasion of KSHV. This study demonstrated that K5 downregulated the cell surface expression of l-selectin, a C-type lectin-like adhesion receptor expressed in the lymphocytes. Tryptophan residue located at the centre of the E2-binding site in the K5 RINGv domain was essential to downregulate l-selectin expression. Additionally, the lysine residues located at the cytoplasmic tail of l-selectin were required for the K5-mediated downregulation of l-selectin. K5 promoted the degradation of l-selectin through polyubiquitination. These results suggest that K5 downregulates l-selectin expression on the cell surface by promoting polyubiquitination and ubiquitin-dependent endocytosis, which indicated that l-selectin is a novel substrate for K5. Additionally, K3 downregulated l-selectin expression. The findings of this study will aid in the elucidation of a novel immune evasion mechanism in KSHV.


Asunto(s)
Herpesvirus Humano 8/enzimología , Proteínas Inmediatas-Precoces/inmunología , Selectina L/genética , Sarcoma de Kaposi/genética , Sarcoma de Kaposi/virología , Ubiquitina-Proteína Ligasas/inmunología , Proteínas Virales/inmunología , Regulación hacia Abajo , Herpesvirus Humano 8/genética , Herpesvirus Humano 8/inmunología , Interacciones Huésped-Patógeno , Humanos , Proteínas Inmediatas-Precoces/genética , Evasión Inmune , Células Asesinas Naturales/inmunología , Selectina L/inmunología , Sarcoma de Kaposi/inmunología , Ubiquitina-Proteína Ligasas/genética , Proteínas Virales/genética
6.
J Immunol ; 207(1): 268-280, 2021 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-34162726

RESUMEN

Expression and activity of serum- and glucocorticoid-inducible kinase 1 (SGK1) are associated with many metabolic and inflammatory diseases. In this study, we report that SGK1 promotes alternative macrophage polarization and restrains inflammation in the infectious milieu of the gingiva. Inhibition of SGK1 expression or activity enhances characteristics of classically activated (M1) macrophages by directly activating the transcription of genes encoding iNOS, IL-12P40, TNF-α, and IL-6 and repressing IL-10 at message and protein levels. Moreover, SGK1 inhibition robustly reduces the expression of alternatively activated (M2) macrophage molecular markers, including arginase-1, Ym-1, Fizz1, and Mgl-1. These results were confirmed by multiple gain- and loss-of-function approaches, including small interfering RNA, a plasmid encoding SGK1, and LysM-Cre-mediated sgk1 gene knockout. Further mechanistic analysis showed that SGK1 deficiency decreases STAT3 but increases FoxO1 expression in macrophages under M2 or M1 macrophage-priming conditions, respectively. Combined with decreased FoxO1 phosphorylation and the subsequent suppressed cytoplasmic translocation observed, SGK1 deficiency robustly enhances FoxO1 activity and drives macrophage to preferential M1 phenotypes. Furthermore, FoxO1 inhibition abrogates M1 phenotypes, and STAT3 overexpression results in a significant increase of M2 phenotypes, indicating that both FoxO1 and STAT3 are involved in SGK1-mediated macrophage polarization. Additionally, SGK1 differentially regulates the expression of M1 and M2 molecular markers, including CD68 and F4/F80 and CD163 and CD206, respectively, and protects against Porphyromonas gingivalis-induced alveolar bone loss in a mouse model. Taken together, these results have demonstrated that SGK1 is critical for macrophage polarization and periodontal bone loss, and for the first time, to our knowledge, we elucidated a bifurcated signaling circuit by which SGK1 promotes alternative, while suppressing inflammatory, macrophage polarization.


Asunto(s)
Proteína Forkhead Box O1/inmunología , Proteínas Inmediatas-Precoces/inmunología , Inflamación/inmunología , Macrófagos/inmunología , Proteínas Serina-Treonina Quinasas/inmunología , Factor de Transcripción STAT3/inmunología , Animales , Activación de Macrófagos/inmunología , Ratones , Transducción de Señal/inmunología
7.
Int J Biol Macromol ; 178: 514-526, 2021 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-33662419

RESUMEN

The binding mode to TAP (i.e., the peptide transporter associated with antigen processing) from a viral peptide thus far has been unknown in the field of antiviral immunity, but an interfering mode from a virus-encoded TAP inhibitor has been well documented with respect to blocking the TAP function. In the current study, we predicted the structure of the pig TAP transporter and its inhibition complex by the small viral protein ICP47 of the herpes simplex virus (HSV) encoded by the TAP inhibitor to exploit inhibition of the TAP transporter as the host's immune evasion strategy. We found that the hot spots (residues Leu5, Tyr22, and Leu51) on the ICP47 inhibitor interface tended to prevail over the favored Leu and Tyr, which contributed to significant functional binding at the C-termini recognition principle of the TAP. We further characterized the specificity determinants of the peptide transporter from the pig TAP by the ICP47 inhibitor effects and multidrug TmrAB transporter from the Thermus thermophillus and its immunity regarding its structural homolog of the pig TAP. The specialized structure-function relationship from the pig TAP exporter could provide insight into substrate specificity of the unique immunological properties from the host organism. The TAP disarming capacity from all five viral inhibitors (i.e., the five virus-encoded TAP inhibitors of ICP47, UL49.5, U6, BNLF2a, and CPXV012 proteins) was linked to the infiltration of the TAP functional structure in an unstable conformation and the mounting susceptibility caused by the host's TAP polymorphism. It is anticipated that the functional characterization of the pig TAP transporter based on the pig genomic variants will lead to additional insights into the genotype and single nucleotide polymorphism (SNP) in relation to antiviral resistance and disease susceptibility.


Asunto(s)
Transportadoras de Casetes de Unión a ATP/antagonistas & inhibidores , Transportadoras de Casetes de Unión a ATP/química , Proteínas Inmediatas-Precoces/química , Evasión Inmune , Simplexvirus/química , Transportadoras de Casetes de Unión a ATP/inmunología , Animales , Humanos , Proteínas Inmediatas-Precoces/inmunología , Simplexvirus/inmunología , Relación Estructura-Actividad , Porcinos
8.
J Virol ; 95(4)2021 01 28.
Artículo en Inglés | MEDLINE | ID: mdl-33208447

RESUMEN

Following acute infection, herpes simplex virus 1 (HSV-1) lytic cycle viral gene expression is silenced; consequently, lifelong latency in neurons is established. Certain external stimuli that trigger reactivation from latency also activate the glucocorticoid receptor (GR). The synthetic corticosteroid dexamethasone, but not a GR-specific antagonist, increases the frequency of explant-induced reactivation from latency and stimulates productive infection. Furthermore, dexamethasone increases expression of cellular transcription factors in trigeminal ganglionic neurons: for example, SLUG and three Krüppel-like transcription factor (KLF) family members, KLF4, KLF15, and promyelocytic leukemia zinc finger protein (PLZF). Consequently, we hypothesized that stress-induced transcription factors stimulate expression of ICP4, a viral transcriptional regulator required for productive infection. New studies demonstrated that GR and KLF4, PLZF, or SLUG cooperatively transactivate the ICP4 enhancer upstream of a minimal promoter in monkey kidney cells (Vero) and mouse neuroblastoma cells (Neuro-2A). Strikingly, mutagenesis of two KLF4/Sp1 binding sites reduced GR- plus KLF4-, PLZF-, or SLUG-mediated transactivation to basal levels. A consensus enhancer (E)-Box adjacent to a KLF4/Sp1 binding site was also required for GR- and SLUG-, but not KLF family member-, mediated transactivation of the ICP4 promoter. Chromatin immunoprecipitation studies (ChIP) revealed GR and stress-induced transcription factors occupy ICP4 enhancer sequences. Conversely, specific binding was generally reduced in the KLF4/Sp1 mutant. Furthermore, GR and SLUG occupancy of ICP4 enhancer sequences was reduced in the E-Box mutant. Based on these studies, we suggest stressful stimuli can trigger productive infection because GR and specific stress-induced transcription factors activate ICP4 expression.IMPORTANCE Certain stressful stimuli activate the glucocorticoid receptor (GR) and increase the incidence of herpes simplex virus 1 (HSV-1) reactivation from latency. For example, a corticosteroid antagonist impairs productive infection and virus shedding following explant of trigeminal ganglia from latently infected mice. Infected cell protein 4 (ICP4) is the only immediate early viral transcriptional regulator required for productive infection, suggesting stressful stimuli stimulate ICP4 expression. New studies revealed GR and stress-induced transcription factors identified during reactivation from latency, SLUG and three Krüppel-like transcription factor family members (KLF4, KLF15, and promyelocytic leukemia zinc finger protein), cooperatively transactivate the ICP4 enhancer. Two KLF4 consensus binding sites were crucial for cooperative transactivation of the ICP4 enhancer. A consensus enhancer-box also mediated cooperative transactivation of the ICP4 enhancer by GR and SLUG. The ability of GR and stress-induced transcription factors to transactivate ICP4 enhancer activity is predicted to trigger productive infection following stressful stimuli.


Asunto(s)
Herpes Simple , Herpesvirus Humano 1/fisiología , Proteínas Inmediatas-Precoces/inmunología , Receptores de Glucocorticoides/inmunología , Activación Viral , Latencia del Virus , Animales , Línea Celular Tumoral , Chlorocebus aethiops , Regulación Viral de la Expresión Génica , Herpes Simple/inmunología , Herpes Simple/virología , Humanos , Factor 4 Similar a Kruppel , Factores de Transcripción de Tipo Kruppel/inmunología , Ratones , Proteína de la Leucemia Promielocítica con Dedos de Zinc/inmunología , Factores de Transcripción de la Familia Snail/inmunología , Activación Transcripcional , Células Vero
9.
J Virol ; 94(21)2020 10 14.
Artículo en Inglés | MEDLINE | ID: mdl-32796067

RESUMEN

Neurotropic Alphaherpesvirinae subfamily members such as bovine herpesvirus 1 (BoHV-1) and herpes simplex virus 1 (HSV-1) establish and maintain lifelong latent infections in neurons. Following infection of ocular, oral, or nasal cavities, sensory neurons within trigeminal ganglia (TG) are an important site for latency. Certain external stressors can trigger reactivation from latency, in part because activation of the glucocorticoid receptor (GR) stimulates productive infection and promoters that drive expression of key viral transcriptional regulators. The Akt serine/threonine protein kinase family is linked to maintaining latency. For example, Akt3 is detected in more TG neurons during BoHV-1 latency than in reactivation and uninfected calves. Furthermore, Akt signaling correlates with maintaining HSV-1 latency in certain neuronal models of latency. Finally, an active Akt protein kinase is crucial for the ability of the HSV-1 latency-associated transcript (LAT) to inhibit apoptosis in neuronal cell lines. Consequently, we hypothesized that viral and/or cellular factors impair stress-induced transcription and reduce the incidence of reactivation triggered by low levels of stress. New studies demonstrate that Akt1 and Akt2, but not Akt3, significantly reduced GR-mediated transactivation of the BoHV-1 immediate early transcription unit 1 (IEtu1) promoter, the HSV-1 infected cell protein 0 (ICP0) promoter, and the mouse mammary tumor virus long terminal repeat (MMTV-LTR). Akt3, but not Akt1 or Akt2, significantly enhanced neurite formation in mouse neuroblastoma cells, which correlates with repairing damaged neurons. These studies suggest that unique biological properties of the three Akt family members promote the maintenance of latency in differentiated neurons.IMPORTANCE External stressful stimuli are known to increase the incidence of reactivation of Alphaherpesvirinae subfamily members. Activation of the glucocorticoid receptor (GR) by the synthetic corticosteroid dexamethasone (DEX) stimulates bovine herpesvirus 1 (BoHV-1) and herpes simplex virus 1 (HSV-1) reactivation. Furthermore, GR and dexamethasone stimulate productive infection and promoters that drive expression of viral transcriptional regulators. These observations lead us to predict that stress-induced transcription is impaired by factors abundantly expressed during latency. Interestingly, activation of the Akt family of serine/threonine protein kinases is linked to maintenance of latency. New studies reveal that Akt1 and Ak2, but not Akt3, impaired GR- and dexamethasone-mediated transactivation of the BoHV-1 immediate early transcription unit 1 and HSV-1 ICP0 promoters. Strikingly, Akt3, but not Akt1 or Akt2, stimulated neurite formation in mouse neuroblastoma cells, a requirement for neurogenesis. These studies provide insight into how Akt family members may promote the maintenance of lifelong latency.


Asunto(s)
Herpes Simple/inmunología , Infecciones por Herpesviridae/inmunología , Interacciones Huésped-Patógeno/inmunología , Proteínas Proto-Oncogénicas c-akt/inmunología , Células Receptoras Sensoriales/virología , Animales , Bovinos , Diferenciación Celular , Línea Celular Tumoral , Herpes Simple/genética , Herpes Simple/patología , Herpes Simple/virología , Infecciones por Herpesviridae/genética , Infecciones por Herpesviridae/patología , Infecciones por Herpesviridae/virología , Herpesvirus Bovino 1/genética , Herpesvirus Bovino 1/inmunología , Herpesvirus Humano 1/genética , Herpesvirus Humano 1/inmunología , Interacciones Huésped-Patógeno/genética , Humanos , Proteínas Inmediatas-Precoces/genética , Proteínas Inmediatas-Precoces/inmunología , Ratones , Neuritas/inmunología , Neuritas/ultraestructura , Neuritas/virología , Regiones Promotoras Genéticas , Proteínas Proto-Oncogénicas c-akt/genética , Receptores de Glucocorticoides/genética , Receptores de Glucocorticoides/inmunología , Células Receptoras Sensoriales/inmunología , Células Receptoras Sensoriales/patología , Transducción de Señal , Activación Transcripcional/inmunología , Ganglio del Trigémino/inmunología , Ganglio del Trigémino/patología , Ganglio del Trigémino/virología , Ubiquitina-Proteína Ligasas/genética , Ubiquitina-Proteína Ligasas/inmunología
10.
J Neurovirol ; 26(5): 687-695, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32671812

RESUMEN

Pseudorabies virus (PRV) establishes a lifelong latent infection in swine trigeminal ganglion (TG) following acute infection. Increased corticosteroid levels, due to stress, increases the incidence of reactivation from latency. Muscle injection combined with intravenous deliver of the synthetic corticosteroid dexamethasone (DEX) consistently induces reactivation from latency in pigs. In this study, PRV-free piglets were infected with PRV. Viral shedding in nasal and ocular swabs demonstrated that PRV infection entered the latent period. The anti-PRV antibody was detected by enzyme-linked immunosorbent assay and the serum neutralization test, which suggested that the PRV could establish latent infection in the presence of humoral immunity. Immunohistochemistry and viral genome detection of TG neurons suggested that PRV was reactivated from latency. Viral gene expressions of IE180, EP0, VP16, and LLT-intron were readily detected at 3-h post-DEX treatment, but gB, a γ1 gene, was not detectable. The differentially expressed phosphorylated proteins of TG neurons were analyzed by ITRAQ coupled with LC-MS/MS, and p-EIF2S2 differentially expression was confirmed by western blot assay. Taken together, our study provides the evidence that typical gene expression in PRV reactivation from latency in TG is disordered compared with known lytic infection in epithelial cells.


Asunto(s)
Dexametasona/farmacología , Regulación Viral de la Expresión Génica/efectos de los fármacos , Herpesvirus Suido 1/efectos de los fármacos , Seudorrabia/virología , Enfermedades de los Porcinos/virología , Ganglio del Trigémino/efectos de los fármacos , Activación Viral/efectos de los fármacos , Animales , Anticuerpos Antivirales/sangre , Ojo/virología , Glucocorticoides/farmacología , Proteína Vmw65 de Virus del Herpes Simple/genética , Proteína Vmw65 de Virus del Herpes Simple/inmunología , Herpesvirus Suido 1/genética , Herpesvirus Suido 1/inmunología , Herpesvirus Suido 1/patogenicidad , Proteínas Inmediatas-Precoces/genética , Proteínas Inmediatas-Precoces/inmunología , Inmunidad Humoral/efectos de los fármacos , Cavidad Nasal/virología , Neuronas/efectos de los fármacos , Neuronas/inmunología , Neuronas/virología , Seudorrabia/inmunología , Seudorrabia/patología , Porcinos , Enfermedades de los Porcinos/inmunología , Enfermedades de los Porcinos/patología , Ganglio del Trigémino/inmunología , Ganglio del Trigémino/virología , Latencia del Virus/efectos de los fármacos , Esparcimiento de Virus/efectos de los fármacos
11.
J Korean Med Sci ; 35(27): e218, 2020 Jul 13.
Artículo en Inglés | MEDLINE | ID: mdl-32657085

RESUMEN

BACKGROUND: Human cytomegalovirus (HCMV) infection in glioblastoma multiforme (GBM) is associated with a poor prognosis and may affect the pathogenesis of GBM. In this study, we investigated the role of HCMV-infected astrocytoma cells in impairing the activity of cytotoxic T lymphocytes (CTLs) specific to the HCMV protein. METHODS: CTLs specific to HCMV immediate early (IE)-1 were expanded from peripheral blood mononuclear cells of healthy donors by stimulating CD8+ T lymphocytes with U373MG cells (ATCC HTB-17: male) expressing HCMV IE-1. The death rate of the target and the effector cells was determined by the total count of the remaining respective cells after the interaction of them. RESULTS: The death rate of the target cells by CTLs increased depending on HLA restriction and the effector:target (E:T) ratio. The death rate of effector cells in the HCMV-infected U373MG cell culture was 37.1% on day 4 post-infection. The removal of the culture supernatant from HCMV-infected U373MG cells prior to adding the effector cells increased target cell death from 8.4% to 40.8% at E:T = 1:1, but not at E:T = 3:1. The transfer of cells from a 24-hour co-culture of the HCMV-infected U373MG cells and CTLs to HCMV IE-1-expressing target cells resulted in decreasing the cell death rate of the target cells from 31.1% to 13.0% at E:T = 1:1, but not at E:T = 3:1. HCMV infection of U373MG cells decreases the activity of CTLs specific to HCMV when the number of CTLs is low. CONCLUSION: These results suggest that HCMV could impair CTL activity and facilitate glioblastoma growth unchecked by CTLs.


Asunto(s)
Astrocitoma/patología , Linfocitos T CD8-positivos/inmunología , Citomegalovirus/fisiología , Apoptosis , Astrocitoma/metabolismo , Linfocitos T CD8-positivos/citología , Linfocitos T CD8-positivos/metabolismo , Linfocitos T CD8-positivos/virología , Células Cultivadas , Técnicas de Cocultivo , Citomegalovirus/metabolismo , Antígenos HLA-A/metabolismo , Humanos , Proteínas Inmediatas-Precoces/inmunología , Proteínas Inmediatas-Precoces/metabolismo , Interferón gamma/metabolismo , Leucocitos Mononucleares/citología , Leucocitos Mononucleares/metabolismo , Masculino
12.
J Immunol ; 205(5): 1281-1292, 2020 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-32699158

RESUMEN

Type I IFNs play an important role in innate immunity against viral infections by inducing the expression of IFN-stimulated genes (ISGs), which encode effectors with various antiviral functions. We and others previously reported that HSV type 2 (HSV-2) inhibits the synthesis of type I IFNs, but how HSV-2 suppresses IFN-mediated signaling is less understood. In the current study, after the demonstration of HSV-2 replication resistance to IFN-ß treatment in human epithelial cells, we reveal that HSV-2 and the viral protein ICP22 significantly decrease the expression of ISG54 at both mRNA and protein levels. Likewise, us1 del HSV-2 (ICP22-deficient HSV-2) replication is more sensitive to IFN-ß treatment, indicating that ICP22 is a vital viral protein responsible for the inhibition of type I IFN-mediated signaling. In addition, overexpression of HSV-2 ICP22 inhibits the expression of STAT1, STAT2, and IFN regulatory factor 9 (IRF9), resulting in the blockade of ISG factor 3 (ISGF3) nuclear translocation, and mechanistically, this is due to ICP22-induced ubiquitination of STAT1, STAT2, and IRF9. HSV-2 ICP22 appears to interact with STAT1, STAT2, IRF9, and several other ubiquitinated proteins. Following further biochemical study, we show that HSV-2 ICP22 functions as an E3 ubiquitin protein ligase to induce the formation of polyubiquitin chains. Taken together, we demonstrate that HSV-2 interferes with type I IFN-mediated signaling by degrading the proteins of ISGF3, and we identify HSV-2 ICP22 as a novel E3 ubiquitin protein ligase to induce the degradation of ISGF3. Findings in this study highlight a new mechanism by which HSV-2 circumvents the host antiviral responses through a viral E3 ubiquitin protein ligase.


Asunto(s)
Herpes Genital/inmunología , Herpesvirus Humano 2/inmunología , Proteínas Inmediatas-Precoces/inmunología , Interferón beta/inmunología , Transducción de Señal/inmunología , Ubiquitina-Proteína Ligasas/inmunología , Proteínas Virales/inmunología , Antivirales/inmunología , Línea Celular , Línea Celular Tumoral , Células HEK293 , Células HeLa , Herpesvirus Humano 1/inmunología , Humanos , Inmunidad Innata/inmunología , Factor de Transcripción STAT1/inmunología , Factor de Transcripción STAT2/inmunología , Ubiquitinación/inmunología
13.
PLoS Pathog ; 16(5): e1008537, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32365141

RESUMEN

Promyelocytic leukemia (PML) bodies are nuclear organelles implicated in intrinsic and innate antiviral defense. The eponymous PML proteins, central to the self-organization of PML bodies, and other restriction factors found in these organelles are common targets of viral antagonism. The 72-kDa immediate-early protein 1 (IE1) is the principal antagonist of PML bodies encoded by the human cytomegalovirus (hCMV). IE1 is believed to disrupt PML bodies by inhibiting PML SUMOylation, while PML was proposed to act as an E3 ligase for IE1 SUMOylation. PML targeting by IE1 is considered to be crucial for hCMV replication at low multiplicities of infection, in part via counteracting antiviral gene induction linked to the cellular interferon (IFN) response. However, current concepts of IE1-PML interaction are largely derived from mutant IE1 proteins known or predicted to be metabolically unstable and globally misfolded. We performed systematic clustered charge-to-alanine scanning mutagenesis and identified a stable IE1 mutant protein (IE1cc172-176) with wild-type characteristics except for neither interacting with PML proteins nor inhibiting PML SUMOylation. Consequently, IE1cc172-176 does not associate with PML bodies and is selectively impaired for disrupting these organelles. Surprisingly, functional analysis of IE1cc172-176 revealed that the protein is hypermodified by mixed SUMO chains and that IE1 SUMOylation depends on nucleosome rather than PML binding. Furthermore, a mutant hCMV expressing IE1cc172-176 was only slightly attenuated compared to an IE1-null virus even at low multiplicities of infection. Finally, hCMV-induced expression of cytokine and IFN-stimulated genes turned out to be reduced rather than increased in the presence of IE1cc172-176 relative to wild-type IE1. Our findings challenge present views on the relationship of IE1 with PML and the role of PML in hCMV replication. This study also provides initial evidence for the idea that disruption of PML bodies upon viral infection is linked to activation rather than inhibition of innate immunity.


Asunto(s)
Infecciones por Citomegalovirus , Citomegalovirus/fisiología , Proteínas Inmediatas-Precoces , Inmunidad Innata , Proteína de la Leucemia Promielocítica , Replicación Viral , Línea Celular , Infecciones por Citomegalovirus/genética , Infecciones por Citomegalovirus/inmunología , Infecciones por Citomegalovirus/patología , Regulación Viral de la Expresión Génica/inmunología , Humanos , Proteínas Inmediatas-Precoces/genética , Proteínas Inmediatas-Precoces/inmunología , Mutación , Proteína de la Leucemia Promielocítica/genética , Proteína de la Leucemia Promielocítica/inmunología , Sumoilación/inmunología , Replicación Viral/genética , Replicación Viral/inmunología
14.
J Virol ; 94(12)2020 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-32295906

RESUMEN

ND10 nuclear bodies, as part of the intrinsic defenses, impose repression on incoming DNA. Infected cell protein 0 (ICP0), an E3 ubiquitin ligase of herpes simplex virus 1 (HSV-1), can derepress viral genes by degrading ND10 organizers to disrupt ND10. These events are part of the initial tug of war between HSV-1 and host, which determines the ultimate outcome of infection. Previously, we reported that ICP0 differentially recognizes promyelocytic leukemia (PML) isoforms. ICP0 depends on a SUMO-interaction motif located at residues 362 to 364 (SIM362-364) to trigger the degradation of PML isoforms II, IV, and VI, while using a bipartite sequence flanking the RING domain to degrade PML I. In this study, we investigated how the SUMO-SIM interaction regulates the degradation of PML II and PML II-associated proteins in ND10. We found that (i) the same regulatory mechanism for PML II degradation was detected in cells permissive or nonpermissive to the ICP0-null virus; (ii) the loss of a single SIM362-364 motif was restored by the presence of four consecutive SIMs from RNF4, but was not rescued by only two of the RNF4 SIMs; (iii) the loss of three C-terminal SIMs of ICP0 was fully restored by four RNF4 SIMs and also partially rescued by two RNF4 SIMs; and (iv) a PML II mutant lacking both lysine SUMOylation and SIM was not recognized by ICP0 for degradation, but was localized to ND10 and mitigated the degradation of other ND10 components, leading to delayed viral production. Taken together, SUMO regulates ICP0 substrate recognition via multiple fine-tuned mechanisms in HSV-1 infection.IMPORTANCE HSV-1 ICP0 is a multifunctional immediate early protein key to effective replication in the HSV-1 lytic cycle and reactivation in the latent cycle. ICP0 transactivates gene expression by orchestrating an overall mitigation in host intrinsic/innate restrictions. How ICP0 coordinates its multiple active domains and its diverse protein-protein interactions is a key question in understanding the HSV-1 life cycle and pathogenesis. The present study focuses on delineating the regulatory effects of the SUMO-SIM interaction on ICP0 E3 ubiquitin ligase activity regarding PML II degradation. For the first time, we discovered the importance of multivalency in the PML II-ICP0 interaction network and report the involvement of different regulatory mechanisms in PML II recognition by ICP0 in HSV-1 infection.


Asunto(s)
Herpesvirus Humano 1/inmunología , Interacciones Huésped-Patógeno/inmunología , Proteínas Inmediatas-Precoces/inmunología , Proteínas Nucleares/inmunología , Proteína de la Leucemia Promielocítica/inmunología , Procesamiento Proteico-Postraduccional , Factores de Transcripción/inmunología , Ubiquitina-Proteína Ligasas/inmunología , Línea Celular Tumoral , Células Epiteliales/inmunología , Células Epiteliales/virología , Regulación de la Expresión Génica , Herpesvirus Humano 1/genética , Interacciones Huésped-Patógeno/genética , Humanos , Proteínas Inmediatas-Precoces/genética , Mutación , Proteínas Nucleares/genética , Proteína de la Leucemia Promielocítica/genética , Unión Proteica , Isoformas de Proteínas/genética , Isoformas de Proteínas/inmunología , Proteolisis , Transducción de Señal , Sumoilación , Factores de Transcripción/genética , Ubiquitina-Proteína Ligasas/genética
15.
Viral Immunol ; 33(5): 378-383, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32181702

RESUMEN

Human cytomegalovirus (HCMV) is a paradigm for pathogen-mediated immune evasion. The immune response to HCMV has been intensively studied for many years and still remains the focus of attention for numerous research groups. UL23 is an early gene of HCMV, belonging to the US22 gene family, encoding protein UL23. However, no monoclonal antibodies against to HCMV UL23 protein have been reported to prepare for the research. In this study, we prepared a highly specific monoclonal antibody against UL23 protein by alternately immunizing BALB/C mice with both UL23 recombinant protein and HCMV Towne. Recombinant protein UL23 was used as a detection antigen to screen 305 strains of hybridoma cells. One of them was identified to secrete IgG1 mAb named as 26C5. Western blotting results showed that not only the overexpressed UL23 protein in 293T cells but also the viral UL23 protein in HCMV-infected human foreskin fibroblast cells specifically were recognized by 26C5 mAb. Notably, we found that UL23 protein were enriched by 26C5 mAb in coimmunoprecipitation experiment with high potency and the native form of UL23 protein localizing primarily in the cytoplasm were recognized by 26C5 mAb in immunofluorescence assay with high specificity. The monoclonal antibody obtained in this study lays the foundation for further study of HCMV UL23 protein.


Asunto(s)
Anticuerpos Monoclonales/inmunología , Antígenos Virales/inmunología , Citomegalovirus/inmunología , Hibridomas/inmunología , Proteínas Inmediatas-Precoces/inmunología , Animales , Anticuerpos Monoclonales/biosíntesis , Antígenos Virales/genética , Femenino , Fibroblastos/inmunología , Fibroblastos/virología , Técnica del Anticuerpo Fluorescente Directa , Prepucio/citología , Células HEK293 , Humanos , Proteínas Inmediatas-Precoces/genética , Inmunoglobulina G/biosíntesis , Inmunoglobulina G/inmunología , Masculino , Ratones , Ratones Endogámicos BALB C , Proteínas Recombinantes/biosíntesis , Proteínas Recombinantes/inmunología
16.
J Neurovirol ; 26(3): 330-337, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32125664

RESUMEN

Varicella-zoster virus (VZV) is a pathogenic human herpesvirus that causes varicella (chickenpox) as a primary infection following which it becomes latent in ganglionic neurons. Following viral reactivation many years later VZV causes herpes zoster (shingles) as well as a variety of other neurological syndromes. The molecular mechanisms of the conversion of the virus from a lytic to a latent state in ganglia are not well understood. In order to gain insights into the neuron-virus interaction, we studied virus-induced apoptosis in cultures of both highly pure terminally differentiated human neurons and human fetal lung fibroblasts (HFL). It was found that (a) VZV DNA did not accumulate in infected human neurons; (b) VZV transcripts were present at lower levels at all days studied post-infection in neurons; (c) Western blot analysis showed less VZV IE 63 and very little detectable VZV gE proteins in infected neurons compared with HFL; (d) lower levels of the apoptotic marker cleaved Caspase-3 protein were detected in VZV-infected neurons compared with HFL, and higher levels of the known anti-apoptotic proteins Bcl2, Bcl-XL and also the mitochondrial MT-CO2 protein were found in VZV-infected neurons compared with uninfected cells; and (e) both the MT-CO2 protein and VZV IE 63-encoded protein were detected in infected neurons by dual immunofluorescence. These findings showed that neurons are resistant to VZV-induced apoptosis, which may have relevance to the switching of VZV from a lytic to latent ganglionic neuronal infection.


Asunto(s)
Apoptosis/genética , ADN Viral/genética , Herpesvirus Humano 3/genética , Interacciones Huésped-Patógeno/genética , Neuronas/virología , Latencia del Virus/genética , Apoptosis/inmunología , Caspasa 3/genética , Caspasa 3/inmunología , Línea Celular , ADN Viral/inmunología , Complejo IV de Transporte de Electrones/genética , Complejo IV de Transporte de Electrones/inmunología , Feto , Fibroblastos/inmunología , Fibroblastos/virología , Regulación de la Expresión Génica , Herpesvirus Humano 3/crecimiento & desarrollo , Herpesvirus Humano 3/inmunología , Interacciones Huésped-Patógeno/inmunología , Humanos , Proteínas Inmediatas-Precoces/genética , Proteínas Inmediatas-Precoces/inmunología , Neuronas/inmunología , Especificidad de Órganos , Proteínas Proto-Oncogénicas c-bcl-2/genética , Proteínas Proto-Oncogénicas c-bcl-2/inmunología , ARN Mensajero/genética , ARN Mensajero/inmunología , Transducción de Señal , Proteínas del Envoltorio Viral/genética , Proteínas del Envoltorio Viral/inmunología , Latencia del Virus/inmunología , Proteína bcl-X/genética , Proteína bcl-X/inmunología
17.
Sci Immunol ; 5(43)2020 01 24.
Artículo en Inglés | MEDLINE | ID: mdl-31980485

RESUMEN

Detection of intracellular DNA by the cGAS-STING pathway activates a type I interferon-mediated innate immune response that protects from virus infection. Whether there are additional DNA sensing pathways, and how such pathways might function, remains controversial. We show here that humans-but not laboratory mice-have a second, potent, STING-independent DNA sensing pathway (SIDSP). We identify human DNA-dependent protein kinase (DNA-PK) as the sensor of this pathway and demonstrate that DNA-PK activity drives a robust and broad antiviral response. We show that the E1A oncoprotein of human adenovirus 5 and the ICP0 protein of herpes simplex virus 1 block this response. We found heat shock protein HSPA8/HSC70 as a target for inducible phosphorylation in the DNA-PK antiviral pathway. Last, we demonstrate that DNA damage and detection of foreign DNA trigger distinct modalities of DNA-PK activity. These findings reveal the existence, sensor, a specific downstream target, and viral antagonists of a SIDSP in human cells.


Asunto(s)
Proteína Quinasa Activada por ADN/inmunología , Adenoviridae , Proteínas E1A de Adenovirus/inmunología , Animales , Línea Celular , Herpes Simple/inmunología , Herpesvirus Humano 1 , Humanos , Proteínas Inmediatas-Precoces/inmunología , Proteínas de la Membrana/genética , Proteínas de la Membrana/inmunología , Ratones , Ubiquitina-Proteína Ligasas/inmunología
18.
J Microbiol ; 58(1): 54-60, 2020 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-31898253

RESUMEN

We previously reported that human cytomegalovirus (HCMV) 86 kDa immediate-early 2 gene product (IE86) promotes proteasome-dependent degradation of STING. In the present study, we determined the specific residues of IE86 responsible for STING degradation using a STING-firefly luciferase fusion protein expression system for quantitative meas-urement of STING protein levels. IE86 amino acids (aa) 136-289 were sufficient to promote STING degradation and further induced down-regulation of 2'3'-cyclic GMP-AMP (cGAMP)-mediated IFN-ß promoter activation. Interestingly, transactivation domains (TAD) of the IE86 protein located at the N- and C-termini were required for down-regulation of Toll/interleukin-1 receptor (TIR) domain-containing adaptor-inducing interferon ß (IFN-ß) (TRIF)-mediated IFN-ß-and p65/RelA-induced NF-κB-dependent promoter activation while amino acids (aa) 136-289 had no significant effects. Our collective data suggest that the IE86 protein utilizes the aa 136-289 region to promote STING degradation and inhibit the cGAS-STING pathway.


Asunto(s)
Infecciones por Citomegalovirus/inmunología , Citomegalovirus/inmunología , Proteínas Inmediatas-Precoces , Proteínas de la Membrana/inmunología , Nucleotidiltransferasas/inmunología , Transactivadores , Proteínas Virales , Células HEK293 , Humanos , Proteínas Inmediatas-Precoces/inmunología , Proteínas Inmediatas-Precoces/metabolismo , Proteínas de la Membrana/metabolismo , Nucleotidiltransferasas/metabolismo , Unión Proteica , Transactivadores/inmunología , Transactivadores/metabolismo , Proteínas Virales/inmunología , Proteínas Virales/metabolismo
19.
Mult Scler Relat Disord ; 38: 101859, 2020 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-31855843

RESUMEN

BACKGROUND: Fingolimod (FTY) applied as treatment regimen of relapsing-remitting multiple sclerosis (RRMS) induces downregulation of sphingosine-1-phosphate receptors on the lymphocytes. As a result CC chemokine receptor type 7 (CCR7) expressing lymphocytes are retained within the peripheral lymph nodes thus suppressing their accumulation into the cerebrospinal fluid of multiple sclerosis (MS) patients and hampering disease progress. Unfortunately, MS patients treated with FTY suffer from an increased incidence of varicella-zoster virus (VZV) infections which has been associated with a decrease of VZV immediate early 63 (IE63)-specific T-cell immunity. To elucidate VZV-specific T-cell immunity over the course of FTY-treatment, we analyzed T-cell immunity for immediate early, early and late VZV-antigens. METHODS: T-cell immune responses were detected via intracellular IFN-γ staining after stimulation with VZV-specific peptide mixes for IE62 and IE63 and recombinant proteins for open reading frame 26 (ORF26), ORF9 and glycoprotein E (gE) using flow cytometry. Analyzed samples comprised of different groups including 18 patients with RRMS at baseline (BL), 6 and 12 months after FTY-treatment start, 12 patients with long-term (LT) FTY-treatment, one FTY-treated patient, before and after VZV-reactivation. In addition, VZV-specific IgG and IgM titers were assessed by ELISA. RESULTS: After FTY-treatment start, absolute numbers of CCR7 expressing CD4+ T cells and CD8+T cells dropped rapidly. However, VZV-specific immunity could be detected in the majority of RRMS patients throughout FTY-treatment with increasing prevalence after 6 months of treatment. We found an increase in the prevalence of VZV-specific IFN-γ+CD8+ T-cell immunity in FTY-treated patients after six months of therapy, while in parallel VZV-specific IFN-γ+CD4+ T cells declined dramatically. Additionally, a strong correlation between VZV-specific IgG serum titers and the percentage of RRMS patients with detectable VZV-specific T cells was observed (r = 0.985). Most remarkably, FTY-treated RRMS patients presented a shift in the predominant CD8+ T cell-mediated antigen-response from immediate early (IE62) to early virus antigens (ORF26) six months after treatment in parallel to a decrease of VZV-specific CD4+ T-cell immunity. ORF26-specific CD8+ T cells still dominated the VZV-specific cellular immunity at month 12 after FTY-treatment start and in LT FTY-treated MS patients. In a RRMS patient an increase of VZV-specific CD4+ T cells at VZV-reactivation accompanied with a four-fold increase of a VZV-specific IgG titer was detected which might indicate an important role in cellular immune control of VZV-infections. CONCLUSION: Monitoring VZV-specific T-cell immunity might provide a valuable tool to RRMS patient risk management during FTY-treatment.


Asunto(s)
Antígenos Virales/inmunología , Clorhidrato de Fingolimod/farmacología , Herpesvirus Humano 3/inmunología , Inmunosupresores/farmacología , Esclerosis Múltiple Recurrente-Remitente/tratamiento farmacológico , Esclerosis Múltiple Recurrente-Remitente/inmunología , Linfocitos T/inmunología , Adulto , Antígenos CD8 , Femenino , Humanos , Proteínas Inmediatas-Precoces/inmunología , Masculino , Receptores CCR7 , Transactivadores/inmunología , Proteínas del Envoltorio Viral/inmunología
20.
Sci Rep ; 9(1): 16660, 2019 11 13.
Artículo en Inglés | MEDLINE | ID: mdl-31723204

RESUMEN

Peptides presented by Human leukocyte antigen (HLA) class-I molecules are generally 8-10 amino acids in length. However, the predominant pool of peptide fragments generated by proteasomes is less than 8 amino acids in length. Using the Epstein - Barr virus (EBV) Rta-epitope (ATIGTAMYK, residues 134-142) restricted by HLA-A*11:01 which generates a strong immunodominant response, we investigated the minimum length of a viral peptide that can constitute a viral epitope recognition by CD8 T cells. The results showed that Peripheral blood mononuclear cells (PBMCs) from healthy donors can be stimulated by a viral peptide fragment as short as 4-mer (AMYK), together with a 5-mer (ATIGT) to recapitulate the full length EBV Rta epitope. This was confirmed by generating crystals of the tetra-complex (2 peptides, HLA and ß2-microglobulin). The solved crystal structure of HLA-A*11:01 in complex with these two short peptides revealed that they can bind in the same orientation similar to parental peptide (9-mer) and the free ends of two short peptides acquires a bulged conformation that is directed towards the T cell receptor. Our data shows that suboptimal length of 4-mer and 5-mer peptides can complement each other to form a stable peptide-MHC (pMHC) complex.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Infecciones por Virus de Epstein-Barr/inmunología , Antígenos HLA-A/química , Herpesvirus Humano 4/inmunología , Proteínas Inmediatas-Precoces/química , Leucocitos Mononucleares/inmunología , Fragmentos de Péptidos/química , Transactivadores/química , Epítopos de Linfocito T/química , Epítopos de Linfocito T/inmunología , Infecciones por Virus de Epstein-Barr/virología , Antígenos HLA-A/inmunología , Humanos , Proteínas Inmediatas-Precoces/inmunología , Leucocitos Mononucleares/virología , Fragmentos de Péptidos/inmunología , Conformación Proteica , Linfocitos T Citotóxicos/inmunología , Transactivadores/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA