Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
1.
J Biol Chem ; 299(9): 105082, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-37495108

RESUMEN

The development and progression of nasopharyngeal carcinoma (NPC) is closely associated with Epstein-Barr virus (EBV) infection. NPC is usually asymptomatic until it spreads to other sites, and more than 70% of cases are classified as locally advanced disease at diagnosis. EBV-positive nasopharyngeal cancer tissues express only limited viral latent proteins, but express high levels of the EBV-encoded BamHI-A rightward transcript (BART) miRNA molecules. Here, we report that EBV-miRNA-BART2-5p (BART2-5p) promotes NPC cell invasion and metastasis in vivo and in vitro but has no effect on NPC cell proliferation and apoptosis. In addition, BART2-5p altered the mRNA and miRNA expression profiles of NPC cells. The development of human tumors has been reported to be associated with altered miRNAs expression, and overall miRNAs expression is reduced in many types of tumors. We found that BART2-5p downregulated the expression of several miRNAs that could exert oncogenic functions. Mechanistically, BART2-5p directly targets the RNase III endonuclease DICER1, inhibiting its function of cleaving double-stranded stem-loop RNA into short double-stranded RNA, which in turn causes altered expression of a series of key epithelial-mesenchymal transition molecules, and reverting DICER1 expression can rescue this phenotype. Furthermore, analysis from clinical samples showed a negative correlation between BART2-5p and DICER1 expression. According to our study, high expression of BART2-5p in tissues and plasma of patients with NPC is associated with poor prognosis. Our results suggest that, BART2-5p can accelerate NPC metastasis through modulating miRNA profiles which are mediated by DICER1, implying a novel role of EBV miRNAs in the pathogenesis of NPC.


Asunto(s)
Infecciones por Virus de Epstein-Barr , MicroARNs , Carcinoma Nasofaríngeo , Neoplasias Nasofaríngeas , Ribonucleasa III , Humanos , Infecciones por Virus de Epstein-Barr/enzimología , Infecciones por Virus de Epstein-Barr/genética , Herpesvirus Humano 4/genética , MicroARNs/genética , MicroARNs/metabolismo , Carcinoma Nasofaríngeo/genética , Carcinoma Nasofaríngeo/patología , Neoplasias Nasofaríngeas/genética , Neoplasias Nasofaríngeas/patología , Ribonucleasa III/genética , Ribonucleasa III/metabolismo , Línea Celular Tumoral , Regulación Neoplásica de la Expresión Génica/genética , Movimiento Celular/genética
2.
Eur J Immunol ; 52(8): 1273-1284, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-35503749

RESUMEN

Endemic Burkitt lymphoma (eBL) is characterized by an oncogenic IGH/c-MYC translocation and Epstein-Barr virus (EBV) positivity, and is epidemiologically linked to Plasmodium falciparum malaria. Both EBV and malaria are thought to contribute to eBL by inducing the expression of activation-induced cytidine deaminase (AID), an enzyme involved in the IGH/c-MYC translocation. AID/apolipoprotein B mRNA editing catalytic polypeptide-like (AID/APOBEC) family enzymes have recently emerged as potent mutagenic sources in a variety of cancers, but apart from AID, their involvement in eBL and their regulation by EBV and P. falciparum is unknown. Here, we show that upon inoculation with EBV, human B cells strongly upregulate the expression of enzymatically active APOBEC3B and APOBEC3G. In addition, we found significantly increased levels of APOBEC3A in B cells of malaria patients, which correlated with parasite load. Interestingly, despite the fact that APOBEC3A, APOBEC3B, and APOBEC3G caused c-MYC mutations when overexpressed in HEK293T cells, a mutational enrichment in eBL tumors was only detected in AID motifs. This suggests that even though the EBV- and P. falciparum-directed immune response triggers the expression and activity of several AID/APOBEC members, only the upregulation of AID has oncogenic consequences, while the induction of the APOBEC3 subfamily may primarily have immunoprotective functions.


Asunto(s)
Desaminasas APOBEC , Linfoma de Burkitt , Citidina Desaminasa , Infecciones por Virus de Epstein-Barr , Malaria Falciparum , Desaminasas APOBEC/genética , Desaminasa APOBEC-3G , Linfoma de Burkitt/enzimología , Linfoma de Burkitt/genética , Citidina Desaminasa/genética , Infecciones por Virus de Epstein-Barr/enzimología , Infecciones por Virus de Epstein-Barr/genética , Células HEK293 , Herpesvirus Humano 4 , Humanos , Malaria Falciparum/enzimología , Malaria Falciparum/genética , Antígenos de Histocompatibilidad Menor , Mutágenos
3.
JCI Insight ; 7(11)2022 06 08.
Artículo en Inglés | MEDLINE | ID: mdl-35482424

RESUMEN

Myalgic encephalomyelitis/chronic fatigue syndrome (ME/CFS) is a chronic, debilitating, multisystem illness of unknown etiology for which no cure and no diagnostic tests are available. Despite increasing evidence implicating EBV and human herpesvirus 6A (HHV-6A) as potential causative infectious agents in a subset of patients with ME/CFS, few mechanistic studies address a causal relationship. In this study we examined a large ME/CFS cohort and controls and demonstrated a significant increase in activin A and IL-21 serum levels, which correlated with seropositivity for antibodies against the EBV and HHV-6 protein deoxyuridine triphosphate nucleotidohydrolase (dUTPases) but no increase in CXCL13. These cytokines are critical for T follicular helper (TFH) cell differentiation and for the generation of high-affinity antibodies and long-lived plasma cells. Notably, ME/CFS serum was sufficient to drive TFH cell differentiation via an activin A-dependent mechanism. The lack of simultaneous CXCL13 increase with IL-21 indicates impaired TFH function in ME/CFS. In vitro studies revealed that virus dUTPases strongly induced activin A secretion while in vivo, EBV dUTPase induced the formation of splenic marginal zone B and invariant NKTFH cells. Together, our data indicate abnormal germinal center (GC) activity in participants with ME/CFS and highlight a mechanism by which EBV and HHV6 dUTPases may alter GC and extrafollicular antibody responses.


Asunto(s)
Síndrome de Fatiga Crónica , Herpesvirus Humano 4 , Herpesvirus Humano 6 , Pirofosfatasas , Linfocitos T Colaboradores-Inductores , Diferenciación Celular , Infecciones por Virus de Epstein-Barr/enzimología , Infecciones por Virus de Epstein-Barr/virología , Síndrome de Fatiga Crónica/diagnóstico , Síndrome de Fatiga Crónica/enzimología , Síndrome de Fatiga Crónica/virología , Herpesvirus Humano 4/enzimología , Herpesvirus Humano 6/enzimología , Humanos , Pirofosfatasas/metabolismo , Infecciones por Roseolovirus/enzimología , Infecciones por Roseolovirus/virología , Linfocitos T Colaboradores-Inductores/enzimología , Linfocitos T Colaboradores-Inductores/patología , Linfocitos T Colaboradores-Inductores/virología
4.
J Virol ; 92(6)2018 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-29263273

RESUMEN

Epstein-Barr virus (EBV) is linked to the development of both lymphoid and epithelial malignancies worldwide. The M81 strain of EBV, isolated from a Chinese patient with nasopharyngeal carcinoma (NPC), demonstrates spontaneous lytic replication and high-titer virus production in comparison to the prototype B95-8 EBV strain. Genetic comparisons of M81 and B95-8 EBVs were previously been performed in order to determine if the hyperlytic property of M81 is associated with sequence differences in essential lytic genes. EBV SM is an RNA-binding protein expressed during early lytic replication that is essential for virus production. We compared the functions of M81 SM and B95-8 SM and demonstrate that polymorphisms in SM do not contribute to the lytic phenotype of M81 EBV. However, the expression level of the EBV DNA polymerase protein was much higher in M81- than in B95-8-infected cells. The relative deficiency in the expression of B95-8 DNA polymerase was related to the B95-8 genome deletion, which truncates the BALF5 3' untranslated region (UTR). Similarly, the insertion of bacmid DNA into the widely used recombinant B95-8 bacmid creates an inefficient BALF5 3' UTR. We further showed that the while SM is required for and facilitates the efficient expression of both M81 and B95-8 mRNAs regardless of the 3' UTR, the BALF5 3' UTR sequence is important for BALF5 protein translation. These data indicate that the enhanced lytic replication and virus production of M81 compared to those of B95-8 are partly due to the robust translation of EBV DNA polymerase required for viral DNA replication due to a more efficient BALF5 3' UTR in M81.IMPORTANCE Epstein-Barr virus (EBV) infects more than 90% of the human population, but the incidence of EBV-associated tumors varies greatly in different parts of the world. Thus, understanding the connection between genetic polymorphisms from patient isolates of EBV, gene expression phenotypes, and disease is important and may help in developing antiviral therapy. This study examines potential causes of the enhanced lytic replicative properties of M81 EBV isolated from a nasopharyngeal carcinoma (NPC) patient and provides new evidence for the role of the BALF5 gene 3' UTR sequence in DNA polymerase protein expression during lytic replication. Variation in the gene structure of the DNA polymerase gene may therefore contribute to lytic virus reactivation and pathogenesis.


Asunto(s)
Replicación del ADN/fisiología , ADN Viral/biosíntesis , Proteínas de Unión al ADN/biosíntesis , ADN Polimerasa Dirigida por ADN/biosíntesis , Infecciones por Virus de Epstein-Barr/enzimología , Genoma Viral , Herpesvirus Humano 4/fisiología , Biosíntesis de Proteínas , Proteínas Virales/biosíntesis , Replicación Viral/fisiología , Regiones no Traducidas 3' , Carcinoma/enzimología , Carcinoma/genética , Carcinoma/patología , Carcinoma/virología , ADN Viral/genética , Proteínas de Unión al ADN/genética , ADN Polimerasa Dirigida por ADN/genética , Infecciones por Virus de Epstein-Barr/genética , Infecciones por Virus de Epstein-Barr/patología , Células HEK293 , Humanos , Carcinoma Nasofaríngeo , Neoplasias Nasofaríngeas/enzimología , Neoplasias Nasofaríngeas/genética , Neoplasias Nasofaríngeas/patología , Neoplasias Nasofaríngeas/virología , Proteínas de Neoplasias/biosíntesis , Proteínas de Neoplasias/genética , Proteínas Virales/genética
5.
J Pathol ; 243(2): 220-229, 2017 10.
Artículo en Inglés | MEDLINE | ID: mdl-28722111

RESUMEN

Monoamine oxidase A (MAOA) is a mitochondrial enzyme that catalyzes oxidative deamination of neurotransmitters and dietary amines and produces H2 O2 . It facilitates the progression of gliomas and prostate cancer, but its expression and functional relevance have not been studied in lymphoma. Here, we evaluated MAOA in 427 cases of Hodgkin and non-Hodgkin lymphoma and in a spectrum of reactive lymphoid tissues by immunohistochemistry on formalin-fixed, paraffin-embedded specimens. MAOA was expressed by Hodgkin Reed-Sternberg (HRS) cells in the majority of classical Hodgkin lymphomas (cHLs) (181/241; 75%), with 34.8% showing strong expression. Weak MAOA was also noted in a minority of primary mediastinal large B-cell lymphomas (8/47; 17%) and in a mediastinal gray-zone lymphoma. In contrast, no MAOA was found in non-neoplastic lymphoid tissues, nodular lymphocyte-predominant Hodgkin lymphoma (NLPHL; 0/8) or any other non-Hodgkin lymphomas studied (0/123). MAOA was more common in Epstein-Barr virus (EBV)-negative compared to EBV-positive cHL (p < 0.0001) and was especially prevalent in the EBV-negative nodular sclerosing subtype. Similar to primary human lymphoma specimens, most cHL-derived cell lines displayed MAOA activity, whereas non-Hodgkin-lymphoma-derived cell lines did not. The MAOA inhibitor clorgyline reduced the growth of L1236 cells and U-HO1 cells, and shRNA knockdown of MAOA reduced the growth of L1236 cells. Conversely, ectopic overexpression of MAOA increased the growth of MAOA-negative HDLM2 cells. Combined treatment with clorgyline and ABVD (doxorubicin, bleomycin, vinblastine, dacarbazine) was more effective in reducing cell growth than either regimen alone. In summary, MAOA is highly expressed in cHL and may reflect the distinct biology of this lymphoma. Further studies on the potential utility of MAOA as a diagnostic marker and therapeutic target are warranted. Copyright © 2017 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.


Asunto(s)
Enfermedad de Hodgkin/enzimología , Monoaminooxidasa/metabolismo , Línea Celular Tumoral , Clorgilina/farmacología , Infecciones por Virus de Epstein-Barr/enzimología , Femenino , Humanos , Masculino , Persona de Mediana Edad , Inhibidores de la Monoaminooxidasa/farmacología , Células de Reed-Sternberg/metabolismo
6.
Virology ; 507: 220-230, 2017 07.
Artículo en Inglés | MEDLINE | ID: mdl-28456021

RESUMEN

The Epstein Barr virus (EBV) genome persists in infected host cells as a chromatinized episome and is subject to chromatin-mediated regulation. Binding of the host insulator protein CTCF to the EBV genome has an established role in maintaining viral latency type, and in other herpesviruses, loss of CTCF binding at specific regions correlates with viral reactivation. Here, we demonstrate that binding of PARP1, an important cofactor of CTCF, at the BZLF1 lytic switch promoter restricts EBV reactivation. Knockdown of PARP1 in the Akata-EBV cell line significantly increases viral copy number and lytic protein expression. Interestingly, CTCF knockdown has no effect on viral reactivation, and CTCF binding across the EBV genome is largely unchanged following reactivation. Moreover, EBV reactivation attenuates PARP activity, and Zta expression alone is sufficient to decrease PARP activity. Here we demonstrate a restrictive function of PARP1 in EBV lytic reactivation.


Asunto(s)
Infecciones por Virus de Epstein-Barr/enzimología , Infecciones por Virus de Epstein-Barr/virología , Herpesvirus Humano 4/fisiología , Poli(ADP-Ribosa) Polimerasa-1/metabolismo , Regiones Promotoras Genéticas , Transactivadores/genética , Activación Viral , Línea Celular , Infecciones por Virus de Epstein-Barr/genética , Regulación Viral de la Expresión Génica , Herpesvirus Humano 4/genética , Interacciones Huésped-Patógeno , Humanos , Poli(ADP-Ribosa) Polimerasa-1/genética , Unión Proteica , Transactivadores/metabolismo , Latencia del Virus
7.
Virology ; 500: 96-102, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-27792904

RESUMEN

Previous data demonstrate that Epstein-Barr Virus Latent Membrane Protein 2A (LMP2A) enhances IL-10 to promote the survival of LMP2A-expressing B cell lymphomas. Since STAT3 is an important regulator of IL-10 production, we hypothesized that LMP2A activates a signal transduction cascade that increases STAT3 phosphorylation to enhance IL-10. Using LMP2A-negative and -positive B cell lines, the data indicate that LMP2A requires the early signaling molecules of the Syk/RAS/PI3K pathway to increase IL-10. Additional studies indicate that the PI3K-regulated kinase, BTK, is responsible for phosphorylating STAT3, which ultimately mediates the LMP2A-dependent increase in IL-10. These data are the first to show that LMP2A signaling results in STAT3 phosphorylation in B cells through a PI3K/BTK-dependent pathway. With the use of BTK and STAT3 inhibitors to treat B cell lymphomas in clinical trials, these findings highlight the possibility of using new pharmaceutical approaches to treat EBV-associated lymphomas that express LMP2A.


Asunto(s)
Infecciones por Virus de Epstein-Barr/metabolismo , Herpesvirus Humano 4/metabolismo , Interleucina-10/metabolismo , Proteínas Tirosina Quinasas/metabolismo , Factor de Transcripción STAT3/metabolismo , Proteínas de la Matriz Viral/metabolismo , Agammaglobulinemia Tirosina Quinasa , Linfocitos B/enzimología , Linfocitos B/metabolismo , Linfocitos B/virología , Línea Celular , Activación Enzimática , Infecciones por Virus de Epstein-Barr/enzimología , Infecciones por Virus de Epstein-Barr/genética , Infecciones por Virus de Epstein-Barr/virología , Herpesvirus Humano 4/genética , Interacciones Huésped-Patógeno , Humanos , Interleucina-10/genética , Fosfatidilinositol 3-Quinasas/metabolismo , Fosforilación , Proteínas Tirosina Quinasas/genética , Factor de Transcripción STAT3/genética , Transducción de Señal , Proteínas de la Matriz Viral/genética
8.
Oncotarget ; 8(4): 6461-6474, 2017 Jan 24.
Artículo en Inglés | MEDLINE | ID: mdl-28031537

RESUMEN

Primary infection of the immunocompromised host with the oncovirus Epstein-Barr virus (EBV) that targets mainly B-cells is associated with an increased risk for EBV-associated tumors. The early events subsequent to primary infection with potential for B-cell transformation are poorly studied. Here, we modeled in vitro the primary infection by using B-cells isolated from tonsils, the portal of entry of EBV, since species specificity of EBV hampers modeling in experimental animals. Increasing evidence indicates that the host DNA damage response (DDR) can influence and be influenced by EBV infection. Thus, we inoculated tonsillar B-cells (TBCs) with EBV-B95.8 and investigated cell proliferation and the DDR during the first 96 hours thereafter. We identified for the first time that EBV infection of TBCs induces a period of hyperproliferation 48-96 hours post infection characterized by the activation of ataxia telangiectasia and Rad3-releated (ATR) and checkpoint kinase-1 (Chk1). Whereas inhibition of Chk1 did not affect B-cell transformation, the specific inhibition of ATR robustly decreased the transformation efficiency of EBV. Our results suggest that activation of ATR is key for EBV-induced B-cell transformation. Thus, targeting the interaction between ATR/Chk1 and EBV could offer new options for the treatment of EBV-associated malignancies.


Asunto(s)
Linfocitos B/virología , Transformación Celular Viral , Quinasa 1 Reguladora del Ciclo Celular (Checkpoint 1)/metabolismo , Infecciones por Virus de Epstein-Barr/virología , Herpesvirus Humano 4/patogenicidad , Tonsila Palatina/enzimología , Tonsila Palatina/virología , Antígenos CD19/metabolismo , Proteínas de la Ataxia Telangiectasia Mutada/análisis , Proteínas de la Ataxia Telangiectasia Mutada/metabolismo , Linfocitos B/efectos de los fármacos , Linfocitos B/enzimología , Linfocitos B/inmunología , Ligando de CD40/metabolismo , Proliferación Celular , Células Cultivadas , Quinasa 1 Reguladora del Ciclo Celular (Checkpoint 1)/análisis , Daño del ADN , Reparación del ADN , Activación Enzimática , Infecciones por Virus de Epstein-Barr/enzimología , Infecciones por Virus de Epstein-Barr/inmunología , Infecciones por Virus de Epstein-Barr/patología , Herpesvirus Humano 4/inmunología , Interacciones Huésped-Patógeno , Humanos , Tonsila Palatina/efectos de los fármacos , Tonsila Palatina/inmunología , Inhibidores de Proteínas Quinasas/farmacología , Transducción de Señal , Factores de Tiempo
9.
Virol J ; 13: 145, 2016 08 26.
Artículo en Inglés | MEDLINE | ID: mdl-27565721

RESUMEN

The Epstein-Barr virus (EBV) is a ubiquitous herpesvirus that transforms B cells and causes several malignancies including Burkitt's lymphoma. EBV differentially expresses at least 49 mature microRNAs (miRNAs) during latency in various infected epithelial and B cells. Recent high-throughput studies and functional assays have begun to reveal the function of the EBV miRNAs suggesting roles in latency, cell cycle control, and apoptosis. In particular, the central executioner of apoptosis, Caspase 3 (CASP3), was proposed as a target of select EBV miRNAs. However, whether CASP3 is truly a target of EBV miRNAs, and if so, which specific miRNAs target CASP3 is still under debate. Based on previously published high-throughput biochemical data and a bioinformatic analysis of the entire CASP3 3'-UTR, we identified 12 EBV miRNAs that have one or more seed binding sites in the CASP3 3'-UTR. We individually tested all 12 miRNAs for repression of CASP3 in luciferase reporter assays, and nine showed statistically significant (P < 0.001) repression of a full-length CASP3 reporter. Further, three EBV miRNAs, including BART22, exhibited repression of endogenous CASP3 protein. These data confirm that CASP3 is a direct target of specific EBV BART miRNAs.


Asunto(s)
Caspasa 3/metabolismo , Infecciones por Virus de Epstein-Barr/enzimología , Herpesvirus Humano 4/metabolismo , MicroARNs/metabolismo , ARN Viral/metabolismo , Apoptosis , Caspasa 3/genética , Infecciones por Virus de Epstein-Barr/genética , Infecciones por Virus de Epstein-Barr/virología , Herpesvirus Humano 4/genética , Interacciones Huésped-Patógeno , Humanos , MicroARNs/genética , ARN Viral/genética , Transcripción Genética , Latencia del Virus
11.
PLoS One ; 10(4): e0123645, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25856387

RESUMEN

The Epstein-Barr virus (EBV) is transmitted from host-to-host via saliva and is associated with epithelial malignancies including nasopharyngeal carcinoma (NPC) and some forms of gastric carcinoma (GC). Nevertheless, EBV does not transform epithelial cells in vitro where it is rapidly lost from infected primary epithelial cells or epithelial tumor cells. Long-term infection by EBV, however, can be established in hTERT-immortalized nasopharyngeal epithelial cells. Here, we hypothesized that increased telomerase activity in epithelial cells enhances their susceptibility to infection by EBV. Using HONE-1, AGS and HEK293 cells we generated epithelial model cell lines with increased or suppressed telomerase activity by stable ectopic expression of hTERT or of a catalytically inactive, dominant negative hTERT mutant. Infection experiments with recombinant prototypic EBV (rB95.8), recombinant NPC EBV (rM81) with increased epithelial cell tropism compared to B95.8, or recombinant B95.8 EBV with BZLF1-knockout that is not able to undergo lytic replication, revealed that infection frequencies positively correlate with telomerase activity in AGS cells but also partly depend on the cellular background. AGS cells with increased telomerase activity showed increased expression mainly of latent EBV genes, suggesting that increased telomerase activity directly acts on the EBV infection of epithelial cells by facilitating latent EBV gene expression early upon virus inoculation. Thus, our results indicate that infection of epithelial cells by EBV is a very selective process involving, among others, telomerase activity and cellular background to allow for optimized host-to-host transmission via saliva.


Asunto(s)
Infecciones por Virus de Epstein-Barr/enzimología , Neoplasias Nasofaríngeas/genética , Nasofaringe/enzimología , Telomerasa/biosíntesis , Carcinoma , Replicación del ADN/genética , Células Epiteliales/enzimología , Células Epiteliales/patología , Células Epiteliales/virología , Infecciones por Virus de Epstein-Barr/genética , Infecciones por Virus de Epstein-Barr/virología , Regulación Enzimológica de la Expresión Génica , Células HEK293 , Herpesvirus Humano 4/genética , Herpesvirus Humano 4/patogenicidad , Humanos , Carcinoma Nasofaríngeo , Neoplasias Nasofaríngeas/enzimología , Neoplasias Nasofaríngeas/patología , Nasofaringe/patología , Telomerasa/genética
12.
PLoS One ; 9(8): e105163, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25126743

RESUMEN

Nasopharyngeal carcinoma (NPC) is a common viral-associated neoplasm in which multiple signaling cascades are interfered with by Epstein-Bar virus (EBV) latent proteins and various genetic alterations. Aside from the previously reported PIK3CA amplification, we examined the role of INPP4B, a negative regulator of the PI3K/AKT signaling pathway in the development of NPC. By RT-PCR and Western blotting, we revealed that the expression of INPP4B was down-regulated in all five established EBV-positive tumor lines. While INPP4B was consistently expressed in normal nasopharyngeal epithelial cells, downregulation of INPP4B was found in 32/65 (49.2%) of primary tumors by immunohistochemistry. Furthermore, our study also demonstrated the hypermethylation of the 5'CpG island of INPP4B in the tumors in which INPP4B transcription was downregulated. Notably, the re-expression of INPP4B was detected in the NPC cells treated with the demethylation agent (5-aza-2'deoxycytidine). Our study showed that promoter hypermethylation was the major mechanism for transcriptional silencing of INPP4B in NPC. Furthermore, restoration of INPP4B expression significantly suppressed PI3K/AKT downstream signals in the NPC C666-1 cells. In vivo growth inhibition was clearly demonstrated in the tumor cells stably expressing INPP4B. The findings indicate that epigenetic inactivation of INPP4B is one of the key mechanisms in activating PI3K/AKT signaling cascade and playing a role in the tumorigenesis of NPC.


Asunto(s)
Infecciones por Virus de Epstein-Barr/enzimología , Neoplasias Nasofaríngeas/enzimología , Monoéster Fosfórico Hidrolasas/genética , Animales , Carcinogénesis/genética , Carcinogénesis/metabolismo , Carcinoma , Línea Celular Tumoral , Proliferación Celular , Metilación de ADN , Epigénesis Genética , Infecciones por Virus de Epstein-Barr/genética , Femenino , Expresión Génica , Regulación Neoplásica de la Expresión Génica , Humanos , Masculino , Ratones Desnudos , Persona de Mediana Edad , Carcinoma Nasofaríngeo , Neoplasias Nasofaríngeas/genética , Neoplasias Nasofaríngeas/virología , Trasplante de Neoplasias , Fosfatidilinositol 3-Quinasas/metabolismo , Monoéster Fosfórico Hidrolasas/metabolismo , Regiones Promotoras Genéticas , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal
13.
J Virol ; 88(12): 6660-71, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24696473

RESUMEN

UNLABELLED: Epstein-Barr virus (EBV) infection has been observed in tumor-infiltrated macrophages, but its infection effects on macrophage immune functions are poorly understood. Here, we showed that some macrophages in the tumor stroma of nasopharyngeal carcinoma (NPC) tissue expressed the immunosuppressive protein indoleamine 2,3-dioxygenase (IDO) more strongly than did tumor cells. EBV infection induced mRNA, protein, and enzymatic activity of IDO in human monocyte-derived macrophages (MDMs). Infection increased the production of tumor necrosis factor alpha (TNF-α) and interleukin-6 (IL-6), whereas the neutralizing antibodies against TNF-α and IL-6 inhibited IDO induction. EBV infection also activated the mitogen-activated protein kinase (MAPK) p38 and NF-κB, and the inhibition of these two pathways with SB202190 and SN50 almost abrogated TNF-α and IL-6 production and inhibited IDO production. Moreover, the activation of IDO in response to EBV infection of MDMs suppressed the proliferation of T cells and impaired the cytotoxic activity of CD8(+) T cells, whereas the inhibition of IDO activity with 1-methyl-l-tryptophan (1-MT) did not affect T cell proliferation and function. These findings indicate that EBV-induced IDO expression in MDMs is substantially mediated by IL-6- and TNF-α-dependent mechanisms via the p38/MAPK and NF-κB pathways, suggesting that a possible role of EBV-mediated IDO expression in tumor stroma of NPC may be to create a microenvironment of suppressed T cell immune responses. IMPORTANCE: CD8(+) cytotoxic T lymphocytes (CTLs) play an important role in the control of viral infections and destroy tumor cells. Activation of the tryptophan-catabolizing enzyme indoleamine 2,3-dioxygenase (IDO) in cancer tissues facilitates immune escape by the impairment of CTL functions. IDO expression was observed in some macrophages of the tumor stroma of nasopharyngeal carcinoma (NPC) tissue, and IDO could be induced in Epstein-Barr virus (EBV)-infected human monocyte-derived macrophages (MDMs). NPC cells and macrophages have been found to produce IDO in a gamma interferon (IFN-γ)-dependent manner. Instead, EBV-induced IDO expression in MDMs is substantially mediated by IL-6- and TNF-α-dependent mechanisms via the p38/MAPK and NF-κB pathways, which suppressed the proliferation of T cells and impaired the cytotoxic activity of CD8(+) T cells. This finding provides a new interpretation of the mechanism of immune escape of EBV and shows the immunosuppressive role of EBV-mediated IDO expression in tumor stroma of NPC.


Asunto(s)
Infecciones por Virus de Epstein-Barr/inmunología , Herpesvirus Humano 4/fisiología , Indolamina-Pirrol 2,3,-Dioxigenasa/inmunología , Macrófagos/enzimología , FN-kappa B/inmunología , Neoplasias Nasofaríngeas/inmunología , Linfocitos T Citotóxicos/inmunología , Proteínas Quinasas p38 Activadas por Mitógenos/inmunología , Adulto , Carcinoma , Células Cultivadas , Infecciones por Virus de Epstein-Barr/enzimología , Infecciones por Virus de Epstein-Barr/genética , Femenino , Humanos , Indolamina-Pirrol 2,3,-Dioxigenasa/genética , Interleucina-6/genética , Interleucina-6/inmunología , Sistema de Señalización de MAP Quinasas , Macrófagos/inmunología , Masculino , Monocitos/enzimología , Monocitos/inmunología , FN-kappa B/genética , Carcinoma Nasofaríngeo , Neoplasias Nasofaríngeas/enzimología , Neoplasias Nasofaríngeas/genética , Proteínas Quinasas p38 Activadas por Mitógenos/genética
14.
Virology ; 448: 293-302, 2014 Jan 05.
Artículo en Inglés | MEDLINE | ID: mdl-24314660

RESUMEN

Ubiquitin C-terminal Hydrolase L1 (UCH-L1) has oncogenic properties and is highly expressed during malignancies. We recently documented that Epstein-Barr virus (EBV) infection induces uch-l1 expression. Here we show that Kaposi's Sarcoma-associated herpesvirus (KSHV) infection induced UCH-L1 expression, via cooperation of KSHV Latency-Associated Nuclear Antigen (LANA) and RBP-Jκ and activation of the uch-l1 promoter. UCH-L1 expression was also increased in Primary Effusion Lymphoma (PEL) cells co-infected with KSHV and EBV compared with PEL cells infected only with KSHV, suggesting EBV augments the effect of LANA on uch-l1. EBV latent membrane protein 1 (LMP1) is one of the few EBV products expressed in PEL cells. Results showed that LMP1 was sufficient to induce uch-l1 expression, and co-expression of LMP1 and LANA had an additive effect on uch-l1 expression. These results indicate that viral latency products of both human γ-herpesviruses contribute to uch-l1 expression, which may contribute to the progression of lymphoid malignancies.


Asunto(s)
Antígenos Virales/metabolismo , Transformación Celular Viral , Infecciones por Virus de Epstein-Barr/enzimología , Infecciones por Herpesviridae/enzimología , Herpesvirus Humano 4/metabolismo , Herpesvirus Humano 8/metabolismo , Proteínas Nucleares/metabolismo , Ubiquitina Tiolesterasa/genética , Proteínas de la Matriz Viral/metabolismo , Antígenos Virales/genética , Línea Celular Transformada , Infecciones por Virus de Epstein-Barr/genética , Infecciones por Virus de Epstein-Barr/virología , Infecciones por Herpesviridae/genética , Infecciones por Herpesviridae/virología , Herpesvirus Humano 4/genética , Herpesvirus Humano 8/genética , Humanos , Proteínas Nucleares/genética , Ubiquitina Tiolesterasa/metabolismo , Regulación hacia Arriba , Proteínas de la Matriz Viral/genética
15.
Ann Oncol ; 25(1): 182-8, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24356628

RESUMEN

BACKGROUND: Oncogenic phosphatidylinositol-3-kinase/serine-threonine kinase (PI3K/AKT) pathway plays a critical role in cell proliferation and growth. Phosphorylated AKT (p-AKT) has been reported to be abnormally overexpressed and to have poor prognostic impact in solid tumors. PATIENTS AND METHODS: To define the clinical implications of p-AKT expression in diffuse large B-cell lymphoma (DLBCL), we calculated arbitrary units (AUs) by multiplying the intensity and the proportion of p-AKT expression and investigated the impact of p-AKT expression on clinical outcomes. We assessed 262 patients with DLBCL. Based on a cutoff value of the upper limit of the third quartile of AUs, 56 patients were classified as high p-AKT and the remaining 206 patients were classified as low p-AKT. RESULTS: The high p-AKT group was closely associated with more advanced stage (stage III-IV, P = 0.02), two or more extranodal involvement (P = 0.03), lactic dehydrogenase elevation (P = 0.03), higher International Prognostic Index risk groups (high intermediate/high, P = 0.02), and the presence of B-symptoms (P = 0.01). The high p-AKT group showed substantially worse overall survival (OS) (median OS, 115.0 months versus not reached, P = 0.004) and progression-free survival (PFS) (median PFS, 25.5 versus 105.8 months, P = 0.019) compared with the low p-AKT group. Multivariate analysis revealed that high p-AKT expression retained its significant poor prognostic impact for OS (hazard ratio 1.7; 95% confidence interval, 1.0-2.7; P = 0.031). The subgroup with high p-AKT expression and concurrent Epstein-Barr virus positivity showed worst prognosis with the median OS and PFS of 15.2 and 7.4 months. CONCLUSION: DLBCL patients with high p-AKT expression showed distinct clinical features and followed a more rapidly deteriorating clinical course with worse OS and PFS. Thus, a more effective treatment option should be developed for this subset of DLBCL patients, and targeting PI3K/AKT pathway may be a promising therapeutic strategy.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Linfoma de Células B Grandes Difuso/enzimología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Supervivencia sin Enfermedad , Infecciones por Virus de Epstein-Barr/tratamiento farmacológico , Infecciones por Virus de Epstein-Barr/enzimología , Infecciones por Virus de Epstein-Barr/mortalidad , Infecciones por Virus de Epstein-Barr/terapia , Femenino , Humanos , Estimación de Kaplan-Meier , Linfoma de Células B Grandes Difuso/tratamiento farmacológico , Linfoma de Células B Grandes Difuso/mortalidad , Linfoma de Células B Grandes Difuso/virología , Masculino , Persona de Mediana Edad , Análisis Multivariante , Fosforilación , Pronóstico , Modelos de Riesgos Proporcionales , Procesamiento Proteico-Postraduccional , Resultado del Tratamiento , Adulto Joven
16.
Scand J Immunol ; 78(2): 130-9, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23672610

RESUMEN

BTK and ITK are cytoplasmic tyrosine kinases of crucial importance for B and T cell development, with loss-of-function mutations causing X-linked agammaglobulinemia and susceptibility to severe, frequently lethal, Epstein-Barr virus infection, respectively. Over the last few years, considerable efforts have been made in order to develop small-molecule inhibitors for these kinases to treat lymphocyte malignancies, autoimmunity or allergy/hypersensitivity. The rationale is that even if complete lack of BTK or ITK during development causes severe immunodeficiency, inactivation after birth may result in a less severe phenotype. Moreover, therapy can be transient or only partially block the activity of BTK or ITK. Furthermore, a drug-induced B cell deficiency is treatable by gamma globulin substitution therapy. The newly developed BTK inhibitor PCI-32765, recently renamed Ibrutinib, has already entered several clinical trials for various forms of non-Hodgkin lymphoma as well as for multiple myeloma. Experimental animal studies have demonstrated highly promising treatment effects also in autoimmunity. ITK inhibitors are still under the early developmental phase, but it can be expected that such drugs will also become very useful. In this study, we present BTK and ITK with their signalling pathways and review the development of the corresponding inhibitors.


Asunto(s)
Agammaglobulinemia/tratamiento farmacológico , Antineoplásicos/uso terapéutico , Infecciones por Virus de Epstein-Barr/tratamiento farmacológico , Enfermedades Genéticas Ligadas al Cromosoma X/tratamiento farmacológico , Linfoma/tratamiento farmacológico , Proteínas Tirosina Quinasas/antagonistas & inhibidores , Adenina/análogos & derivados , Agammaglobulinemia Tirosina Quinasa , Agammaglobulinemia/enzimología , Agammaglobulinemia/genética , Agammaglobulinemia/patología , Autoinmunidad/efectos de los fármacos , Linfocitos B/efectos de los fármacos , Linfocitos B/inmunología , Infecciones por Virus de Epstein-Barr/enzimología , Infecciones por Virus de Epstein-Barr/genética , Infecciones por Virus de Epstein-Barr/patología , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Enfermedades Genéticas Ligadas al Cromosoma X/enzimología , Enfermedades Genéticas Ligadas al Cromosoma X/genética , Enfermedades Genéticas Ligadas al Cromosoma X/patología , Humanos , Inflamación/prevención & control , Linfoma/enzimología , Linfoma/genética , Linfoma/patología , Piperidinas , Inhibidores de Proteínas Quinasas/uso terapéutico , Proteínas Tirosina Quinasas/genética , Proteínas Tirosina Quinasas/metabolismo , Pirazoles/uso terapéutico , Pirimidinas/uso terapéutico , Transducción de Señal/efectos de los fármacos , Bibliotecas de Moléculas Pequeñas/uso terapéutico , Linfocitos T/efectos de los fármacos , Linfocitos T/inmunología
17.
Am J Transplant ; 13(4): 883-890, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23398911

RESUMEN

Posttransplant lymphoproliferative disorder (PTLD)-associated Epstein-Barr virus (EBV)+ B cell lymphomas are serious complications of solid organ and bone marrow transplantation. The EBV protein LMP2a, a B cell receptor (BCR) mimic, provides survival signals to virally infected cells through Syk tyrosine kinase. Therefore, we explored whether Syk inhibition is a viable therapeutic strategy for EBV-associated PTLD. We have shown that R406, the active metabolite of the Syk inhibitor fostamatinib, induces apoptosis and cell cycle arrest while decreasing downstream phosphatidylinositol-3'-kinase (PI3K)/Akt signaling in EBV+ B cell lymphoma PTLD lines in vitro. However, Syk inhibition did not inhibit or delay the in vivo growth of solid tumors established from EBV-infected B cell lines. Instead, we observed tumor growth in adjacent inguinal lymph nodes exclusively in fostamatinib-treated animals. In contrast, direct inhibition of PI3K/Akt significantly reduced tumor burden in a xenogeneic mouse model of PTLD without evidence of tumor growth in adjacent inguinal lymph nodes. Taken together, our data indicate that Syk activates PI3K/Akt signaling which is required for survival of EBV+ B cell lymphomas. PI3K/Akt signaling may be a promising therapeutic target for PTLD, and other EBV-associated malignancies.


Asunto(s)
Infecciones por Virus de Epstein-Barr/enzimología , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Trastornos Linfoproliferativos/enzimología , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Tirosina Quinasas/metabolismo , Aminopiridinas , Animales , Apoptosis , Linfocitos B/metabolismo , Ciclo Celular , Línea Celular Tumoral , Activación Enzimática , Herpesvirus Humano 4 , Humanos , Péptidos y Proteínas de Señalización Intracelular/antagonistas & inhibidores , Ganglios Linfáticos/patología , Linfoma de Células B/enzimología , Linfoma de Células B/virología , Trastornos Linfoproliferativos/virología , Masculino , Ratones , Ratones Endogámicos NOD , Ratones SCID , Morfolinas , Oxazinas/farmacología , Complicaciones Posoperatorias , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Tirosina Quinasas/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-akt/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-akt/metabolismo , Piridinas/farmacología , Pirimidinas , Transducción de Señal , Quinasa Syk , Trasplante Heterólogo
18.
Am J Pathol ; 181(5): 1773-81, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22974584

RESUMEN

Nasopharyngeal carcinoma (NPC) is characteristic for its strong association with Epstein-Barr virus (EBV) and high metastatic rate. Recently, overexpressed recepteur d'origine nantais (RON) (MST1R), receptor tyrosine kinase has been reported in human cancers and tumor metastasis. Therefore, the role of RON in EBV-associated NPC and its metastasis was investigated. Here we show that RON was found in NPC but not in control tissues. A significant correlation of latent membrane protein 1 (LMP1) and RON expression was found in NPC (Pearson's χ(2) test; P = 0.0023). At the molecular level, LMP1 stimulates nuclear factor-κB binding to the RON promoter through its carboxyl-terminal activation region 1 to induce expression of RON. Knockdown of RON in cells expressing LMP1 significantly reverses LMP1-induced epithelial-mesenchymal transition and suppresses LMP1-induced cell migration and invasion. These results suggest an important role of RON in the tumorigenesis and metastasis of NPC and RON may be a novel therapeutic target for EBV-associated NPC.


Asunto(s)
Infecciones por Virus de Epstein-Barr/enzimología , Infecciones por Virus de Epstein-Barr/virología , Herpesvirus Humano 4/fisiología , Neoplasias Nasofaríngeas/enzimología , Neoplasias Nasofaríngeas/virología , Proteínas Tirosina Quinasas Receptoras/metabolismo , Carcinoma , Movimiento Celular , Forma de la Célula , Transición Epitelial-Mesenquimal , Infecciones por Virus de Epstein-Barr/patología , Humanos , Inmunohistoquímica , FN-kappa B/metabolismo , Carcinoma Nasofaríngeo , Neoplasias Nasofaríngeas/patología , Invasividad Neoplásica , Metástasis de la Neoplasia , Fosforilación , Transducción de Señal , Proteínas de la Matriz Viral/metabolismo
19.
Int Arch Allergy Immunol ; 158(4): 418-22, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22487848

RESUMEN

IL-2-inducible T-cell kinase (ITK) deficiency is a rare inherited immunodeficiency disease characterized by homozygous mutations in the ITK gene and the inability to control Epstein-Barr virus (EBV) infection leading to EBV-associated lymphoproliferative disorders of B cell origin. Many aspects of its clinical presentation and immunologic phenotype are still unclear to clinicians. We report on a 14-year-old female patient with complaints of an 8-month history of cough and fever. Imaging studies revealed diffuse pulmonary nodules and mediastinal lymphadenopathy. Transbronchial lung biopsy showed nonmalignant polyclonal B cell proliferation. High titers of EBV DNA were detected by PCR analysis in bronchoalveolar lavage fluid, bone marrow, and blood. Genomic analysis revealed a homozygous single base pair deletion in exon 5 of the ITK gene (c.468delT) in this patient. Treatment with rituximab (anti-CD20 mab) resulted in complete clinical remission with resolution of pulmonary lesions and a negative EBV titer in serum. All patients with EBV-associated lymphoproliferative disorders should be analyzed for mutations in ITK.


Asunto(s)
Infecciones por Virus de Epstein-Barr/enzimología , Neumonía Viral/enzimología , Proteínas Tirosina Quinasas/genética , Adolescente , Anticuerpos Monoclonales de Origen Murino/uso terapéutico , Linfocitos B/efectos de los fármacos , Linfocitos B/patología , Linfocitos B/virología , Líquido del Lavado Bronquioalveolar/virología , Tos/diagnóstico , Tos/tratamiento farmacológico , Tos/enzimología , Tos/patología , Tos/virología , ADN Viral/análisis , Infecciones por Virus de Epstein-Barr/tratamiento farmacológico , Infecciones por Virus de Epstein-Barr/patología , Femenino , Fiebre/diagnóstico , Fiebre/tratamiento farmacológico , Fiebre/enzimología , Fiebre/patología , Fiebre/virología , Humanos , Factores Inmunológicos/uso terapéutico , Pulmón/diagnóstico por imagen , Pulmón/efectos de los fármacos , Pulmón/enzimología , Pulmón/patología , Pulmón/virología , Activación de Linfocitos/efectos de los fármacos , Activación de Linfocitos/genética , Trastornos Linfoproliferativos/diagnóstico por imagen , Trastornos Linfoproliferativos/tratamiento farmacológico , Trastornos Linfoproliferativos/enzimología , Trastornos Linfoproliferativos/patología , Trastornos Linfoproliferativos/virología , Neumonía Viral/tratamiento farmacológico , Neumonía Viral/patología , Mutación Puntual , Rituximab , Tomografía Computarizada por Rayos X
20.
J Virol ; 86(12): 6656-67, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22514348

RESUMEN

Epstein-Barr virus (EBV)-associated nasopharyngeal carcinoma (NPC) is highly metastatic, and this malignant feature may be promoted by an EBV oncoprotein, latent membrane protein 2A (LMP2A). Acting as a signal regulator, LMP2A can enhance invasiveness and motility of epithelial cells. Downstream from the LMP2A-triggered signaling events, it is largely unknown what key effector proteins are induced and essentially promote cell invasion. In the present study, we found that in NPC cells, LMP2A upregulated matrix metalloproteinase 9 (MMP9), a metastasis-associated protease. LMP2A increased MMP9 expression at both the mRNA and protein levels. It also activated the MMP9 promoter, in which two AP-1 elements were required for the promoter activation. Among AP-1 transcription factors, Fra-1 was induced by LMP2A and is essential for LMP2A-triggered MMP9 expression. Induction of Fra-1 was dependent on the LMP2A-activated ERK1/2 pathway, and induction of the ERK1/2-Fra-1-MMP9 axis required PY motifs in the amino-terminal domain of LMP2A. Notably, LMP2A-promoted invasion of NPC cells was blocked when MMP9 expression, Fra-1 induction, or ERK1/2 activation was inhibited. In addition, we found an association of LMP2A with MMP9 expression in NPC tumor biopsy specimens, where Fra-1 was a major mediation factor. This study reveals an underlying mechanism of LMP2A-induced cell invasion, from signal transduction to upregulation of a critical protease. Considering that MMP9 can also be upregulated by another EBV oncoprotein, LMP1, this protease may be a pivotal effector at which the EBV-induced, invasion-promoting mechanisms converge, serving as an attractive therapeutic target for NPC treatment.


Asunto(s)
Infecciones por Virus de Epstein-Barr/enzimología , Herpesvirus Humano 4/metabolismo , Sistema de Señalización de MAP Quinasas , Metaloproteinasa 9 de la Matriz/metabolismo , Neoplasias Nasofaríngeas/enzimología , Proteínas Proto-Oncogénicas c-fos/metabolismo , Proteínas de la Matriz Viral/metabolismo , Carcinoma , Infecciones por Virus de Epstein-Barr/genética , Infecciones por Virus de Epstein-Barr/metabolismo , Infecciones por Virus de Epstein-Barr/virología , Herpesvirus Humano 4/genética , Humanos , Metaloproteinasa 9 de la Matriz/genética , Carcinoma Nasofaríngeo , Neoplasias Nasofaríngeas/genética , Neoplasias Nasofaríngeas/metabolismo , Neoplasias Nasofaríngeas/virología , Proteínas Proto-Oncogénicas c-fos/genética , Factor de Transcripción AP-1/genética , Factor de Transcripción AP-1/metabolismo , Activación Transcripcional , Regulación hacia Arriba , Proteínas de la Matriz Viral/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA