Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 1.242
Filtrer
2.
Int J Mol Sci ; 25(11)2024 May 28.
Article de Anglais | MEDLINE | ID: mdl-38892033

RÉSUMÉ

The Epstein-Barr virus (EBV) is frequently found in endomyocardial biopsies (EMBs) from patients with heart failure, but the detection of EBV-specific DNA has not been associated with progressive hemodynamic deterioration. In this paper, we investigate the use of targeted next-generation sequencing (NGS) to detect EBV transcripts and their correlation with myocardial inflammation in EBV-positive patients with heart failure with reduced ejection fraction (HFrEF). Forty-four HFrEF patients with positive EBV DNA detection and varying degrees of myocardial inflammation were selected. EBV-specific transcripts from EMBs were enriched using a custom hybridization capture-based workflow and, subsequently, sequenced by NGS. The short-read sequencing revealed the presence of EBV-specific transcripts in 17 patients, of which 11 had only latent EBV genes and 6 presented with lytic transcription. The immunohistochemical staining for CD3+ T lymphocytes showed a significant increase in the degree of myocardial inflammation in the presence of EBV lytic transcripts, suggesting a possible influence on the clinical course. These results imply the important role of EBV lytic transcripts in the pathogenesis of inflammatory heart disease and emphasize the applicability of targeted NGS in EMB diagnostics as a basis for specific treatment.


Sujet(s)
Infections à virus Epstein-Barr , Défaillance cardiaque , Herpèsvirus humain de type 4 , Myocardite , Humains , Herpèsvirus humain de type 4/génétique , Défaillance cardiaque/virologie , Défaillance cardiaque/génétique , Défaillance cardiaque/anatomopathologie , Mâle , Femelle , Infections à virus Epstein-Barr/virologie , Infections à virus Epstein-Barr/complications , Infections à virus Epstein-Barr/génétique , Infections à virus Epstein-Barr/anatomopathologie , Adulte d'âge moyen , Myocardite/virologie , Myocardite/anatomopathologie , Sujet âgé , Séquençage nucléotidique à haut débit , Myocarde/anatomopathologie , Myocarde/métabolisme , ADN viral/génétique , Adulte , Biopsie
3.
J Clin Immunol ; 44(7): 153, 2024 Jun 19.
Article de Anglais | MEDLINE | ID: mdl-38896122

RÉSUMÉ

Magnesium transporter 1 (MAGT1) gene loss-of-function variants lead to X-linked MAGT1 deficiency with increased susceptibility to EBV infection and N-glycosylation defect (XMEN), a condition with a variety of clinical and immunological effects. In addition, MAGT1 deficiency has been classified as a congenital disorder of glycosylation (CDG) due to its unique role in glycosylation of multiple substrates including NKG2D, necessary for viral protection. Due to the predisposition for EBV, this etiology has been linked with hemophagocytic lymphohistiocytosis (HLH), however only limited literature exists. Here we present a complex case with HLH and EBV-driven classic Hodgkin lymphoma (cHL) as the presenting manifestation of underlying immune defect. However, the patient's underlying immunodeficiency was not identified until his second recurrence of Hodgkin disease, recurrent episodes of Herpes Zoster, and after he had undergone autologous hematopoietic stem cell transplant (HSCT) for refractory Hodgkin lymphoma. This rare presentation of HLH and recurrent lymphomas without some of the classical immune deficiency manifestations of MAGT1 deficiency led us to review the literature for similar presentations and to report the evolving spectrum of disease in published literature. Our systematic review showcased that MAGT1 predisposes to multiple viruses (including EBV) and adds risk of viral-driven neoplasia. The roles of MAGT1 in the immune system and glycosylation were highlighted through the multiple organ dysfunction showcased by the previously validated Immune Deficiency and Dysregulation Activity (IDDA2.1) score and CDG-specific Nijmegen Pediatric CDG Rating Scale (NPCRS) score for the patient cohort in the systematic review.


Sujet(s)
Infections à virus Epstein-Barr , Maladie de Hodgkin , Lymphohistiocytose hémophagocytaire , Humains , Mâle , Transporteurs de cations , Infections à virus Epstein-Barr/diagnostic , Infections à virus Epstein-Barr/complications , Infections à virus Epstein-Barr/génétique , Transplantation de cellules souches hématopoïétiques , Herpèsvirus humain de type 4 , Maladie de Hodgkin/diagnostic , Maladie de Hodgkin/génétique , Maladie de Hodgkin/étiologie , Lymphohistiocytose hémophagocytaire/diagnostic , Lymphohistiocytose hémophagocytaire/étiologie , Lymphohistiocytose hémophagocytaire/génétique , Récidive
4.
PLoS Pathog ; 20(6): e1011970, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38885264

RÉSUMÉ

Reactivation from latency plays a significant role in maintaining persistent lifelong Epstein-Barr virus (EBV) infection. Mechanisms governing successful activation and progression of the EBV lytic phase are not fully understood. EBV expresses multiple viral microRNAs (miRNAs) and manipulates several cellular miRNAs to support viral infection. To gain insight into the host miRNAs regulating transitions from EBV latency into the lytic stage, we conducted a CRISPR/Cas9-based screen in EBV+ Burkitt lymphoma (BL) cells using anti-Ig antibodies to crosslink the B cell receptor (BCR) and induce reactivation. Using a gRNA library against >1500 annotated human miRNAs, we identified miR-142 as a key regulator of EBV reactivation. Genetic ablation of miR-142 enhanced levels of immediate early and early lytic gene products in infected BL cells. Ago2-PAR-CLIP experiments with reactivated cells revealed miR-142 targets related to Erk/MAPK signaling, including components directly downstream of the B cell receptor (BCR). Consistent with these findings, disruption of miR-142 enhanced SOS1 levels and Mek phosphorylation in response to surface Ig cross-linking. Effects could be rescued by inhibitors of Mek (cobimetinib) or Raf (dabrafenib). Taken together, these results show that miR-142 functionally regulates SOS1/Ras/Raf/Mek/Erk signaling initiated through the BCR and consequently, restricts EBV entry into the lytic cycle.


Sujet(s)
Systèmes CRISPR-Cas , Infections à virus Epstein-Barr , Herpèsvirus humain de type 4 , microARN , Activation virale , Latence virale , Humains , Herpèsvirus humain de type 4/génétique , microARN/génétique , microARN/métabolisme , Infections à virus Epstein-Barr/virologie , Infections à virus Epstein-Barr/génétique , Infections à virus Epstein-Barr/métabolisme , Lymphome de Burkitt/virologie , Lymphome de Burkitt/génétique , Lymphome de Burkitt/métabolisme , Lignée cellulaire tumorale
5.
J Clin Immunol ; 44(7): 155, 2024 Jun 26.
Article de Anglais | MEDLINE | ID: mdl-38922539

RÉSUMÉ

PURPOSE: Moesin (MSN) deficiency is a recently reported combined immunodeficiency, and few cases have been reported to date. We describe a Chinese patient with a novel mutation causing MSN deficiency and a novel phenotype. METHODS: Clinical and immunological data were collected. Whole-exome sequencing was performed to identify gene mutations. MSN protein expression and T cell proliferation and activation were determined by flow cytometry. Cell migration was confirmed with a Transwell assay. Autoantibody levels were analyzed using antigen microarrays. RESULTS: The patient was a 10-year-old boy who presented with recurrent fever, oral ulcers and dermatomyositis-like symptoms, such as periorbital edema, facial swelling, elevated creatine kinase levels, and abnormal electromyography and muscle biopsy results. Epstein-Barr virus (EBV) DNA was detected in the serum, cells and tissues of this patient. He further developed nasal-type NK/T-cell lymphoma. A novel hemizygous mutation (c.68 A > G, p.N23S) in the MSN gene was found. The immunological phenotype of this patient included persistent decreases in T and B lymphocyte counts but normal immunoglobulin IgG levels. The patient had attenuated MSN protein expression and impaired T-cell proliferation and migration. The proportions of Tfh cells and CD21low B cells in the patient were higher than those in the controls. Moreover, 82 IgG and 102 IgM autoantibodies were more abundant in the patient than in the healthy controls. CONCLUSIONS: The novel mutation N23S is pathogenic and leads to a severe clinical phenotype. EBV infection, tumor, and dermatomyositis-like autoimmune symptoms may be associated with MSN deficiency, further expanding the understanding of the disease.


Sujet(s)
Dermatomyosite , Infections à virus Epstein-Barr , Protéines des microfilaments , Mutation , Humains , Mâle , Infections à virus Epstein-Barr/diagnostic , Infections à virus Epstein-Barr/génétique , Infections à virus Epstein-Barr/complications , Infections à virus Epstein-Barr/immunologie , Dermatomyosite/génétique , Dermatomyosite/diagnostic , Dermatomyosite/immunologie , Enfant , Protéines des microfilaments/génétique , Mutation/génétique , Herpèsvirus humain de type 4 , , Déficits immunitaires/génétique , Déficits immunitaires/diagnostic , Autoanticorps/sang , Autoanticorps/immunologie , Phénotype , Lymphocytes T/immunologie
6.
Nat Commun ; 15(1): 4156, 2024 May 16.
Article de Anglais | MEDLINE | ID: mdl-38755141

RÉSUMÉ

Epstein-Barr virus (EBV) uses a biphasic lifecycle of latency and lytic reactivation to infect >95% of adults worldwide. Despite its central role in EBV persistence and oncogenesis, much remains unknown about how EBV latency is maintained. We used a human genome-wide CRISPR/Cas9 screen to identify that the nuclear protein SFPQ was critical for latency. SFPQ supported expression of linker histone H1, which stabilizes nucleosomes and regulates nuclear architecture, but has not been previously implicated in EBV gene regulation. H1 occupied latent EBV genomes, including the immediate early gene BZLF1 promoter. Upon reactivation, SFPQ was sequestered into sub-nuclear puncta, and EBV genomic H1 occupancy diminished. Enforced H1 expression blocked EBV reactivation upon SFPQ knockout, confirming it as necessary downstream of SFPQ. SFPQ knockout triggered reactivation of EBV in B and epithelial cells, as well as of Kaposi's sarcoma-associated herpesvirus in B cells, suggesting a conserved gamma-herpesvirus role. These findings highlight SFPQ as a major regulator of H1 expression and EBV latency.


Sujet(s)
Herpèsvirus humain de type 4 , Histone , Facteur d'épissage associé à PTB , Activation virale , Latence virale , Herpèsvirus humain de type 4/génétique , Herpèsvirus humain de type 4/physiologie , Humains , Histone/métabolisme , Activation virale/génétique , Latence virale/génétique , Facteur d'épissage associé à PTB/métabolisme , Facteur d'épissage associé à PTB/génétique , Régulation de l'expression des gènes viraux , Lymphocytes B/virologie , Lymphocytes B/métabolisme , Infections à virus Epstein-Barr/virologie , Infections à virus Epstein-Barr/génétique , Infections à virus Epstein-Barr/métabolisme , Systèmes CRISPR-Cas , Régions promotrices (génétique)/génétique , Transactivateurs/métabolisme , Transactivateurs/génétique , Génome viral
7.
PLoS Pathog ; 20(5): e1012263, 2024 May.
Article de Anglais | MEDLINE | ID: mdl-38805547

RÉSUMÉ

Genetic variants in Epstein-Barr virus (EBV) have been strongly associated with nasopharyngeal carcinoma (NPC) in South China. However, different results regarding the most significant viral variants, with polymorphisms in EBER2 and BALF2 loci, have been reported in separate studies. In this study, we newly sequenced 100 EBV genomes derived from 61 NPC cases and 39 population controls. Comprehensive genomic analyses of EBV sequences from both NPC patients and healthy carriers in South China were conducted, totaling 279 cases and 227 controls. Meta-analysis of genome-wide association study revealed a 4-bp deletion downstream of EBER2 (coordinates, 7188-7191; EBER-del) as the most significant variant associated with NPC. Furthermore, multiple viral variants were found to be genetically linked to EBER-del forming a risk haplotype, suggesting that multiple viral variants might be associated with NPC pathogenesis. Population structure and phylogenetic analyses further characterized a high risk EBV lineage for NPC revealing a panel of 38 single nucleotide polymorphisms (SNPs), including those in the EBER2 and BALF2 loci. With linkage disequilibrium clumping and feature selection algorithm, the 38 SNPs could be narrowed down to 9 SNPs which can be used to accurately detect the high risk EBV lineage. In summary, our study provides novel insight into the role of EBV genetic variation in NPC pathogenesis by defining a risk haplotype of EBV for downstream functional studies and identifying a single high risk EBV lineage characterized by 9 SNPs for potential application in population screening of NPC.


Sujet(s)
Infections à virus Epstein-Barr , Génome viral , Herpèsvirus humain de type 4 , Cancer du nasopharynx , Tumeurs du rhinopharynx , Femelle , Humains , Mâle , Chine/épidémiologie , Peuples d'Asie de l'Est , Infections à virus Epstein-Barr/virologie , Infections à virus Epstein-Barr/génétique , Variation génétique , Étude d'association pangénomique , Herpèsvirus humain de type 4/génétique , Cancer du nasopharynx/virologie , Cancer du nasopharynx/génétique , Tumeurs du rhinopharynx/virologie , Tumeurs du rhinopharynx/génétique , Phylogenèse , Polymorphisme de nucléotide simple
8.
Nat Microbiol ; 9(6): 1540-1554, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38806670

RÉSUMÉ

Epstein-Barr virus (EBV) is an aetiologic risk factor for the development of multiple sclerosis (MS). However, the role of EBV-infected B cells in the immunopathology of MS is not well understood. Here we characterized spontaneous lymphoblastoid cell lines (SLCLs) isolated from MS patients and healthy controls (HC) ex vivo to study EBV and host gene expression in the context of an individual's endogenous EBV. SLCLs derived from MS patient B cells during active disease had higher EBV lytic gene expression than SLCLs from MS patients with stable disease or HCs. Host gene expression analysis revealed activation of pathways associated with hypercytokinemia and interferon signalling in MS SLCLs and upregulation of forkhead box protein 1 (FOXP1), which contributes to EBV lytic gene expression. We demonstrate that antiviral approaches targeting EBV replication decreased cytokine production and autologous CD4+ T cell responses in this ex vivo model. These data suggest that dysregulation of intrinsic B cell control of EBV gene expression drives a pro-inflammatory, pathogenic B cell phenotype that can be attenuated by suppressing EBV lytic gene expression.


Sujet(s)
Lymphocytes B , Infections à virus Epstein-Barr , Herpèsvirus humain de type 4 , Sclérose en plaques , Humains , Herpèsvirus humain de type 4/génétique , Sclérose en plaques/virologie , Sclérose en plaques/immunologie , Sclérose en plaques/génétique , Sclérose en plaques/métabolisme , Lymphocytes B/immunologie , Lymphocytes B/métabolisme , Lymphocytes B/virologie , Infections à virus Epstein-Barr/virologie , Infections à virus Epstein-Barr/immunologie , Infections à virus Epstein-Barr/génétique , Infections à virus Epstein-Barr/complications , Cytokines/métabolisme , Lymphocytes T CD4+/immunologie , Lymphocytes T CD4+/virologie , Lymphocytes T CD4+/métabolisme , Facteurs de transcription Forkhead/génétique , Facteurs de transcription Forkhead/métabolisme , Transcriptome , Réplication virale , Régulation de l'expression des gènes viraux , Lignée cellulaire , Interactions hôte-pathogène/génétique , Interactions hôte-pathogène/immunologie , Analyse de profil d'expression de gènes , Adulte , Femelle , Mâle
9.
Sci Rep ; 14(1): 10315, 2024 05 05.
Article de Anglais | MEDLINE | ID: mdl-38705879

RÉSUMÉ

Several studies have shown an association between prostate carcinoma (PCa) and Epstein-Barr virus (EBV); however, none of the studies so far have identified the histopathological and genetic markers of cancer aggressiveness associated with EBV in PCa tissues. In this study, we used previously characterized EBV-PCR-positive (n = 39) and EBV-negative (n = 60) PCa tissues to perform an IHC-based assessment of key histopathological and molecular markers of PCa aggressiveness (EMT markers, AR expression, perineural invasion, and lymphocytic infiltration characterization). Additionally, we investigated the differential expression of key oncogenes, EMT-associated genes, and PCa-specific oncomiRs, in EBV-positive and -negative tissues, using the qPCR array. Finally, survival benefit analysis was also performed in EBV-positive and EBV-negative PCa patients. The EBV-positive PCa exhibited a higher percentage (80%) of perineural invasion (PNI) compared to EBV-negative PCa (67.3%) samples. Similarly, a higher lymphocytic infiltration was observed in EBV-LMP1-positive PCa samples. The subset characterization of T and B cell lymphocytic infiltration showed a trend of higher intratumoral and tumor stromal lymphocytic infiltration in EBV-negative tissues compared with EBV-positive tissues. The logistic regression analysis showed that EBV-positive status was associated with decreased odds (OR = 0.07; p-value < 0.019) of CD3 intratumoral lymphocytic infiltration in PCa tissues. The analysis of IHC-based expression patterns of EMT markers showed comparable expression of all EMT markers, except vimentin, which showed higher expression in EBV-positive PCa tissues compared to EBV-negative PCa tissues. Furthermore, gene expression analysis showed a statistically significant difference (p < 0.05) in the expression of CDH1, AR, CHEK-2, CDKN-1B, and CDC-20 and oncomiRs miR-126, miR-152-3p, miR-452, miR-145-3p, miR-196a, miR-183-3p, and miR-146b in EBV-positive PCa tissues compared to EBV-negative PCa tissues. Overall, the survival proportion was comparable in both groups. The presence of EBV in the PCa tissues results in an increased expression of certain oncogenes, oncomiRs, and EMT marker (vimentin) and a decrease in CD3 ITL, which may be associated with the aggressive forms of PCa.


Sujet(s)
Marqueurs biologiques tumoraux , Infections à virus Epstein-Barr , Herpèsvirus humain de type 4 , Tumeurs de la prostate , Humains , Mâle , Tumeurs de la prostate/génétique , Tumeurs de la prostate/anatomopathologie , Tumeurs de la prostate/virologie , Tumeurs de la prostate/mortalité , Tumeurs de la prostate/métabolisme , Herpèsvirus humain de type 4/génétique , Infections à virus Epstein-Barr/virologie , Infections à virus Epstein-Barr/génétique , Infections à virus Epstein-Barr/anatomopathologie , Infections à virus Epstein-Barr/complications , Marqueurs biologiques tumoraux/génétique , Sujet âgé , Régulation de l'expression des gènes tumoraux , Marqueurs génétiques , Adulte d'âge moyen , Lymphocytes TIL/immunologie , Transition épithélio-mésenchymateuse/génétique , Invasion tumorale
10.
Int J Mol Sci ; 25(9)2024 May 03.
Article de Anglais | MEDLINE | ID: mdl-38732219

RÉSUMÉ

Epstein-Barr virus (EBV) is a ubiquitous gammaherpesvirus etiologically associated with benign and malignant diseases. Since the pathogenic mechanisms of EBV are not fully understood, understanding EBV genetic diversity is an ongoing goal. Therefore, the present work describes the genetic diversity of the lytic gene BZLF1 in a sampling of 70 EBV-positive cases from southeastern Brazil. Additionally, together with the genetic regions previously characterized, the aim of the present study was to determine the impact of viral genetic factors that may influence EBV genetic diversity. Accordingly, the phylogenetic analysis of the BZLF1 indicated two main clades with high support, BZ-A and BZ-B (PP > 0.85). Thus, the BZ-A clade was the most diverse clade associated with the main polymorphisms investigated, including the haplotype Type 1 + V3 (p < 0.001). Furthermore, the multigene phylogenetic analysis (MLA) between BZLF1 and the oncogene LMP1 showed specific clusters, revealing haplotypic segregation that previous single-gene phylogenies from both genes failed to demonstrate. Surprisingly, the LMP1 Raji-related variant clusters were shown to be more diverse, associated with BZ-A/B and the Type 2/1 + V3 haplotypes. Finally, due to the high haplotypic diversity of the Raji-related variants, the number of DNA recombination-inducing motifs (DRIMs) was evaluated within the different clusters defined by the MLA. Similarly, the haplotype BZ-A + Raji was shown to harbor a greater number of DRIMs (p < 0.001). These results call attention to the high haplotype diversity of EBV in southeast Brazil and strengthen the hypothesis of the recombinant potential of South American Raji-related variants via the LMP1 oncogene.


Sujet(s)
Infections à virus Epstein-Barr , Variation génétique , Herpèsvirus humain de type 4 , Phylogenèse , Recombinaison génétique , Herpèsvirus humain de type 4/génétique , Humains , Brésil , Infections à virus Epstein-Barr/virologie , Infections à virus Epstein-Barr/génétique , Transactivateurs/génétique , Mâle , Femelle , Haplotypes/génétique , Adulte , Protéines de la matrice virale/génétique , Enfant , Adulte d'âge moyen , Adolescent , Latence virale/génétique , Enfant d'âge préscolaire , Jeune adulte
11.
J Clin Immunol ; 44(4): 103, 2024 Apr 20.
Article de Anglais | MEDLINE | ID: mdl-38642164

RÉSUMÉ

Epstein-Barr virus (EBV) infection can lead to infectious mononucleosis (EBV-IM) and, more rarely, EBV-associated hemophagocytic lymphohistiocytosis (EBV-HLH), which is characterized by a life-threatening hyperinflammatory cytokine storm with immune dysregulation. Interferon-gamma (IFNγ) has been identified as a critical mediator for primary HLH; however, the detailed role of IFNγ and other cytokines in EBV-HLH is not fully understood. In this study, we used single-cell RNA sequencing to characterize the immune landscape of EBV-HLH and compared it with EBV-IM. Three pediatric patients with EBV-HLH with different backgrounds, one with X-linked lymphoproliferative syndrome type 1 (XLP1), two with chronic active EBV disease (CAEBV), and two patients with EBV-IM were enrolled. The TUBA1B + STMN1 + CD8 + T cell cluster, a responsive proliferating cluster with rich mRNA detection, was explicitly observed in EBV-IM, and the upregulation of SH2D1A-the gene responsible for XLP1-was localized in this cluster. This proliferative cluster was scarcely observed in EBV-HLH cases. In EBV-HLH cases with CAEBV, upregulation of LAG3 was observed in EBV-infected cells, which may be associated with an impaired response by CD8 + T cells. Additionally, genes involved in type I interferon (IFN) signaling were commonly upregulated in each cell fraction of EBV-HLH, and activation of type II IFN signaling was observed in CD4 + T cells, natural killer cells, and monocytes but not in CD8 + T cells in EBV-HLH. In conclusion, impaired responsive proliferation of CD8 + T cells and upregulation of type I IFN signaling were commonly observed in EBV-HLH cases, regardless of the patients' background, indicating the key features of EBV-HLH.


Sujet(s)
Infections à virus Epstein-Barr , Lymphohistiocytose hémophagocytaire , Syndromes lymphoprolifératifs , Humains , Enfant , Herpèsvirus humain de type 4 , Lymphohistiocytose hémophagocytaire/diagnostic , Lymphohistiocytose hémophagocytaire/génétique , Infections à virus Epstein-Barr/complications , Infections à virus Epstein-Barr/génétique , Lymphocytes T CD8+ , Interféron gamma/génétique , Syndromes lymphoprolifératifs/diagnostic , Syndromes lymphoprolifératifs/génétique , Syndromes lymphoprolifératifs/complications , Analyse de profil d'expression de gènes
12.
Oncol Res ; 32(5): 999-1009, 2024.
Article de Anglais | MEDLINE | ID: mdl-38686046

RÉSUMÉ

Background: EBV-miR-BARTs exhibit significant relevance in epithelial tumors, particularly in EBV-associated gastric and nasopharyngeal cancers. However, their specific mechanisms in the initiation and progression of gastric cancer remain insufficiently explored. Material and Methods: Initially, EBV-miRNA-BART6-5p and its target gene SMAD4 expression were assessed in EBV-associated gastric cancer tissues and cell lines. Subsequent transfection induced overexpression of EBV-miRNA-BART6-5p in AGS and MKN-45, and downregulation in EBV-positive cells (SUN-719). The subsequent evaluation aimed to observe their impact on gastric cancer cell proliferation, migration, and glycolytic processes, with the TGF-ß/SMAD4 signaling pathway value clarified using a TGF-ß inhibitor. Results: EBV-miRNA-BART6-5p exhibits pronounced upregulation in EBV-associated gastric cancer tissues and EBV-positive cells, while its target gene SMAD4 demonstrates downregulated expression. Upregulation of it can promote the proliferation and migration of gastric cancer cells. Additionally, We found EBV-miRNA-BART6-5p promotes glycolysis of gastric cancer cells. Inhibition of the TGF-ß/SMAD4 signaling pathway resulted in suppressed proliferation and migration of gastric cancer cells, concomitant with a diminished glycolytic capacity. Conclusion: In this study, we found that EBV-miRNA-BART6-5p can target SMAD4, effectively increasing glycolysis in gastric cancer cells by regulating the TGF-ß/SMAD4 signaling pathway, thereby enhancing the proliferation and metastasis of gastric cancer cells. Our findings may offer new insights into the metabolic aspects of gastric cancer.


Sujet(s)
Mouvement cellulaire , Prolifération cellulaire , Régulation de l'expression des gènes tumoraux , Glycolyse , Herpèsvirus humain de type 4 , microARN , Transduction du signal , Protéine Smad-4 , Tumeurs de l'estomac , Facteur de croissance transformant bêta , Humains , Tumeurs de l'estomac/anatomopathologie , Tumeurs de l'estomac/virologie , Tumeurs de l'estomac/génétique , Tumeurs de l'estomac/métabolisme , Protéine Smad-4/génétique , Protéine Smad-4/métabolisme , microARN/génétique , Glycolyse/génétique , Facteur de croissance transformant bêta/métabolisme , Facteur de croissance transformant bêta/génétique , Herpèsvirus humain de type 4/génétique , Lignée cellulaire tumorale , Mouvement cellulaire/génétique , Infections à virus Epstein-Barr/génétique , Infections à virus Epstein-Barr/virologie , Infections à virus Epstein-Barr/métabolisme , Infections à virus Epstein-Barr/complications , Infections à virus Epstein-Barr/anatomopathologie , Métastase tumorale , ARN viral/génétique
13.
Biochim Biophys Acta Mol Basis Dis ; 1870(5): 167132, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38565386

RÉSUMÉ

The Epstein-Barr virus (EBV) is implicated in several cancers, including EBV-associated gastric cancer (EBVaGC). This study focuses on EBV-encoded BALF1 (BamH1 A fragment leftward reading frame 1), a key apoptosis regulator in EBV-related cancers, whose specific impact on EBVaGC was previously unknown. Our findings indicate that BALF1 overexpression in gastric cancer cells significantly enhances their proliferation, migration, and resistance to chemotherapy-induced apoptosis, confirming BALF1's oncogenic potential. A novel discovery is that BALF1 undergoes degradation via the ubiquitin-proteasome pathway. Through analysis of 69 deubiquitinating enzymes (DUBs), ovarian tumor protease (OTU) domain-containing protein 1 (OTUD1) emerged as a vital regulator for maintaining BALF1 protein stability. Furthermore, BALF1 was found to play a role in regulating the stability of the B-cell lymphoma-2 (Bcl-2) protein, increasing its levels through deubiquitination. This mechanism reveals BALF1's multifaceted oncogenic role in gastric cancer, as it contributes both directly and indirectly to cancer progression, particularly by stabilizing Bcl-2, known for its anti-apoptotic characteristics. These insights significantly deepen our understanding of EBV's involvement in the pathogenesis of gastric cancer. The elucidation of OTUD1's role in BALF1 regulation and its influence on Bcl-2 stabilization provide new avenues for therapeutic intervention in EBVaGC, bridging the gap between viral oncogenesis and cellular protein regulation and offering a more holistic view of gastric cancer development under the influence of EBV.


Sujet(s)
Apoptose , Protéines proto-oncogènes c-bcl-2 , Tumeurs de l'estomac , Ubiquitination , Humains , Tumeurs de l'estomac/anatomopathologie , Tumeurs de l'estomac/virologie , Tumeurs de l'estomac/métabolisme , Tumeurs de l'estomac/génétique , Protéines proto-oncogènes c-bcl-2/métabolisme , Protéines proto-oncogènes c-bcl-2/génétique , Lignée cellulaire tumorale , Herpèsvirus humain de type 4/métabolisme , Herpèsvirus humain de type 4/génétique , Protéines virales/métabolisme , Protéines virales/génétique , Prolifération cellulaire , Ubiquitin-specific proteases/métabolisme , Ubiquitin-specific proteases/génétique , Infections à virus Epstein-Barr/virologie , Infections à virus Epstein-Barr/métabolisme , Infections à virus Epstein-Barr/anatomopathologie , Infections à virus Epstein-Barr/génétique , Stabilité protéique , Mouvement cellulaire , Animaux , Enzymes de désubiquitinylation/métabolisme , Enzymes de désubiquitinylation/génétique , Protéines virales régulatrices ou accessoires
14.
PLoS Pathog ; 20(4): e1012132, 2024 Apr.
Article de Anglais | MEDLINE | ID: mdl-38620028

RÉSUMÉ

Epstein-Barr virus (EBV) is an important cause of human lymphomas, including Burkitt lymphoma (BL). EBV+ BLs are driven by Myc translocation and have stringent forms of viral latency that do not express either of the two major EBV oncoproteins, EBNA2 (which mimics Notch signaling) and LMP1 (which activates NF-κB signaling). Suppression of Myc-induced apoptosis, often through mutation of the TP53 (p53) gene or inhibition of pro-apoptotic BCL2L11 (BIM) gene expression, is required for development of Myc-driven BLs. EBV+ BLs contain fewer cellular mutations in apoptotic pathways compared to EBV-negative BLs, suggesting that latent EBV infection inhibits Myc-induced apoptosis. Here we use an EBNA2-deleted EBV virus (ΔEBNA2 EBV) to create the first in vivo model for EBV+ BL-like lymphomas derived from primary human B cells. We show that cord blood B cells infected with both ΔEBNA2 EBV and a Myc-expressing vector proliferate indefinitely on a CD40L/IL21 expressing feeder layer in vitro and cause rapid onset EBV+ BL-like tumors in NSG mice. These LMP1/EBNA2-negative Myc-driven lymphomas have wild type p53 and very low BIM, and express numerous germinal center B cell proteins (including TCF3, BACH2, Myb, CD10, CCDN3, and GCSAM) in the absence of BCL6 expression. Myc-induced activation of Myb mediates expression of many of these BL-associated proteins. We demonstrate that Myc blocks LMP1 expression both by inhibiting expression of cellular factors (STAT3 and Src) that activate LMP1 transcription and by increasing expression of proteins (DNMT3B and UHRF1) known to enhance DNA methylation of the LMP1 promoters in human BLs. These results show that latent EBV infection collaborates with Myc over-expression to induce BL-like human B-cell lymphomas in mice. As NF-κB signaling retards the growth of EBV-negative BLs, Myc-mediated repression of LMP1 may be essential for latent EBV infection and Myc translocation to collaboratively induce human BLs.


Sujet(s)
Lymphocytes B , Lymphome de Burkitt , Infections à virus Epstein-Barr , Herpèsvirus humain de type 4 , Protéines proto-oncogènes c-myc , Latence virale , Animaux , Lymphome de Burkitt/virologie , Lymphome de Burkitt/métabolisme , Lymphome de Burkitt/anatomopathologie , Lymphome de Burkitt/génétique , Humains , Souris , Infections à virus Epstein-Barr/virologie , Infections à virus Epstein-Barr/métabolisme , Infections à virus Epstein-Barr/complications , Infections à virus Epstein-Barr/génétique , Herpèsvirus humain de type 4/génétique , Protéines proto-oncogènes c-myc/métabolisme , Protéines proto-oncogènes c-myc/génétique , Lymphocytes B/virologie , Lymphocytes B/métabolisme , Antigènes nucléaires du virus d'Epstein-Barr/métabolisme , Antigènes nucléaires du virus d'Epstein-Barr/génétique , Apoptose , Protéines virales/métabolisme , Protéines virales/génétique
15.
Nat Commun ; 15(1): 2901, 2024 Apr 04.
Article de Anglais | MEDLINE | ID: mdl-38575571

RÉSUMÉ

Simultaneous multi-target detection and multi-site gene editing are two key factors restricting the development of disease diagnostic and treatment technologies. Despite numerous explorations on the source, classification, functional features, crystal structure, applications and engineering of CRISPR-Cas13a, all reports use the contiguous target RNA activation paradigm that only enables single-target detection in vitro and one-site gene editing in vivo. Here we propose a noncontiguous target RNA activation paradigm of Cas13a and establish a CRISPR-Cas13a Gemini System composed of two Cas13a:crRNA binary complexes, which can provide rapid, simultaneous, highly specific and sensitive detection of two RNAs in a single readout, as well as parallel dual transgene knockdown. CRISPR-Cas13a Gemini System are demonstrated in the detection of two miRNAs (miR-155 and miR-375) for breast cancer diagnosis and two small RNAs (EBER-1 and EBER-2) for Epstein-Barr virus diagnosis using multiple diagnostic platforms, including fluorescence and colorimetric-based lateral flow systems. We also show that CRISPR-Cas13a Gemini System can knockdown two foreign genes (EGFP and mCherry transcripts) in mammalian cells simultaneously. These findings suggest the potential of highly effective and simultaneous detection of multiple biomarkers and gene editing of multiple sites.


Sujet(s)
Infections à virus Epstein-Barr , microARN , Animaux , Humains , ARN/génétique , Clustered regularly interspaced short palindromic repeats/génétique , Systèmes CRISPR-Cas/génétique , Infections à virus Epstein-Barr/génétique , Herpèsvirus humain de type 4/génétique , Mammifères/génétique
16.
Front Immunol ; 15: 1370991, 2024.
Article de Anglais | MEDLINE | ID: mdl-38633254

RÉSUMÉ

Background and aims: X lymphoproliferative syndrome type 1 (XLP1) is a rare inborn error of immunity due to mutations of SH2D1A, encoding for slam-associated protein (SAP). The clinical phenotype includes severe mononucleosis, hemophagocytic lymphohistiocytosis (HLH), and B-cell lymphomas. Methods: We report the case of a child affected with XLP1 who presented with an incomplete HLH, triggered by Epstein-Barr virus (EBV) and treated with rituximab, involving orbits and paranasal sinuses. Results: The lesion was indistinguishable from lymphoma, complicating diagnosis and treatment. In addition, considering the high incidence of lymphoma in patients with XLP1, histology helped define its nature, driving therapeutic choices. Conclusion: We described an unusual presentation of incomplete HLH in a patient affected with XLP1: an EBV-driven infiltration of the orbits and paranasal sinuses. This led us to a challenging differential diagnosis of lymphoma-associated hemophagocytic syndrome, which can be frequently observed in patients with XLP1. Considering the extremely poor prognosis of this clinical finding, we sought for a prompt diagnosis and managed to obtain it and to immediately establish the right treatment on the basis of the pathological finding.


Sujet(s)
Infections à virus Epstein-Barr , Déficits immunitaires , Lymphohistiocytose hémophagocytaire , Lymphomes , Syndromes lymphoprolifératifs , Enfant , Humains , Herpèsvirus humain de type 4 , Rituximab , Infections à virus Epstein-Barr/génétique , Lymphohistiocytose hémophagocytaire/génétique
17.
PLoS Pathog ; 20(4): e1011939, 2024 Apr.
Article de Anglais | MEDLINE | ID: mdl-38683861

RÉSUMÉ

Epstein-Barr virus (EBV) persistently infects 95% of adults worldwide and is associated with multiple human lymphomas that express characteristic EBV latency programs used by the virus to navigate the B-cell compartment. Upon primary infection, the EBV latency III program, comprised of six Epstein-Barr Nuclear Antigens (EBNA) and two Latent Membrane Protein (LMP) antigens, drives infected B-cells into germinal center (GC). By incompletely understood mechanisms, GC microenvironmental cues trigger the EBV genome to switch to the latency II program, comprised of EBNA1, LMP1 and LMP2A and observed in GC-derived Hodgkin lymphoma. To gain insights into pathways and epigenetic mechanisms that control EBV latency reprogramming as EBV-infected B-cells encounter microenvironmental cues, we characterized GC cytokine effects on EBV latency protein expression and on the EBV epigenome. We confirmed and extended prior studies highlighting GC cytokine effects in support of the latency II transition. The T-follicular helper cytokine interleukin 21 (IL-21), which is a major regulator of GC responses, and to a lesser extent IL-4 and IL-10, hyper-induced LMP1 expression, while repressing EBNA expression. However, follicular dendritic cell cytokines including IL-15 and IL-27 downmodulate EBNA but not LMP1 expression. CRISPR editing highlighted that STAT3 and STAT5 were necessary for cytokine mediated EBNA silencing via epigenetic effects at the EBV genomic C promoter. By contrast, STAT3 was instead necessary for LMP1 promoter epigenetic remodeling, including gain of activating histone chromatin marks and loss of repressive polycomb repressive complex silencing marks. Thus, EBV has evolved to coopt STAT signaling to oppositely regulate the epigenetic status of key viral genomic promoters in response to GC cytokine cues.


Sujet(s)
Épigenèse génétique , Infections à virus Epstein-Barr , Régulation de l'expression des gènes viraux , Centre germinatif , Herpèsvirus humain de type 4 , Latence virale , Herpèsvirus humain de type 4/génétique , Herpèsvirus humain de type 4/physiologie , Humains , Centre germinatif/immunologie , Centre germinatif/virologie , Infections à virus Epstein-Barr/virologie , Infections à virus Epstein-Barr/génétique , Infections à virus Epstein-Barr/immunologie , Cytokines/métabolisme , Lymphocytes B/virologie , Lymphocytes B/métabolisme
18.
Virus Res ; 343: 199352, 2024 05.
Article de Anglais | MEDLINE | ID: mdl-38462175

RÉSUMÉ

This study aims to explore the role and regulatory mechanism of Yes-associated protein 1 (YAP1) in the development of Epstein-Barr virus-associated gastric cancer (EBVaGC). Here we showed that EBV can upregulate the expression and activity of YAP1 protein through its encoded latent products EBV-encoded small RNA 1 (EBER1) and latent membrane protein 2A (LMP2A), enhancing the malignant characteristics of EBVaGC cells. In addition, we also showed that overexpression of YAP1 induced the expression of EBV encoding latent and lytic phase genes and proteins in the epithelial cell line AGS-EBV infected with EBV, and increased the copy number of the EBV genome, while loss of YAP1 expression reduced the aforementioned indicators. Moreover, we found that YAP1 enhanced EBV lytic reactivation induced by two known activators, 12-O-tetradecanoylhorbol-13-acetate (TPA) and sodium butyrate (NaB). These results indicated a bidirectional regulatory mechanism between EBV and YAP1 proteins, providing new experimental evidence for further understanding the regulation of EBV infection patterns and carcinogenic mechanisms in gastric cancer.


Sujet(s)
Carcinomes , Infections à virus Epstein-Barr , ARN viral , Tumeurs de l'estomac , Humains , Infections à virus Epstein-Barr/complications , Infections à virus Epstein-Barr/génétique , Herpèsvirus humain de type 4/métabolisme , Tumeurs de l'estomac/anatomopathologie , Protéines membranaires/métabolisme , Facteurs de transcription/génétique , Facteurs de transcription/métabolisme
19.
EBioMedicine ; 102: 105057, 2024 Apr.
Article de Anglais | MEDLINE | ID: mdl-38490101

RÉSUMÉ

BACKGROUND: Nasopharyngeal carcinoma (NPC) is an Epstein-Barr virus (EBV)-associated malignant epithelial tumor endemic to Southern China and Southeast Asia. While previous studies have revealed a low frequency of gene mutations in NPC, its epigenomic aberrations are not fully elucidated apart from DNA hypermethylation. Epigenomic rewiring and enhancer dysregulation, such as enhancer hijacking due to genomic structural changes or extrachromosomal DNA, drive cancer progression. METHODS: We conducted Hi-C, 4C-seq, ChIP-seq, and RNA-seq analyses to comprehensively elucidate the epigenome and interactome of NPC using C666-1 EBV(+)-NPC cell lines, NP69T immortalized nasopharyngeal epithelial cells, clinical NPC biopsy samples, and in vitro EBV infection in HK1 and NPC-TW01 EBV(-) cell lines. FINDINGS: In C666-1, the EBV genome significantly interacted with inactive B compartments of host cells; the significant association of EBV-interacting regions (EBVIRs) with B compartment was confirmed using clinical NPC and in vitro EBV infection model. EBVIRs in C666-1 showed significantly higher levels of active histone modifications compared with NP69T. Aberrant activation of EBVIRs after EBV infection was validated using in vitro EBV infection models. Within the EBVIR-overlapping topologically associating domains, 14 H3K4me3(+) genes were significantly upregulated in C666-1. Target genes of EBVIRs including PLA2G4A, PTGS2 and CITED2, interacted with the enhancers activated in EBVIRs and were highly expressed in NPC, and their knockdown significantly reduced cell proliferation. INTERPRETATION: The EBV genome contributes to NPC tumorigenesis through "enhancer infestation" by interacting with the inactive B compartments of the host genome and aberrantly activating enhancers. FUNDING: The funds are listed in the Acknowledgements section.


Sujet(s)
Carcinomes , Infections à virus Epstein-Barr , Tumeurs du rhinopharynx , Humains , Cancer du nasopharynx/génétique , Herpèsvirus humain de type 4/génétique , Infections à virus Epstein-Barr/complications , Infections à virus Epstein-Barr/génétique , Tumeurs du rhinopharynx/génétique , Tumeurs du rhinopharynx/anatomopathologie , Carcinogenèse/génétique , ADN , Protéines de répression , Transactivateurs
20.
Anticancer Res ; 44(4): 1425-1440, 2024 Apr.
Article de Anglais | MEDLINE | ID: mdl-38537989

RÉSUMÉ

BACKGROUND/AIM: Nasopharyngeal carcinoma (NPC), a common cancer in Southern China, is associated with Epstein-Barr Virus (EBV) infection. Although many therapies for NPC have been established, the definite role of EBV in NPC remains unclear. Therefore, this work focuses on LMP2A, a latent EBV gene, and investigates whether LMP2A is related to peroxiredoxin 1 (PRDX1) in EBV-positive NPC. MATERIALS AND METHODS: The mRNA and protein expression levels of LMP2A, PRDX1, and beta-catenin were compared in patient samples. To identify molecular mechanisms, EBV-negative NP69 and EBV-positive C666-1 NPC cell lines were used. After making an agar cell block for cell slides, the intensity of LMP2A expression was observed visually. To measure the level of reactive oxygen species, both fluorescence microscope and flow cytometry were used. To investigate the intracellular signaling molecular mechanisms with and without the LMP2A gene, reverse transcription polymerase chain reaction and western blotting were used. RESULTS: Both patient samples and cells of nasopharyngeal carcinoma infected with EBV had increased expression of LMP2A compared with controls, and high ROS levels were identified. Cell viability assay showed that LMP2A promoted cell growth by regulating gene expression. Furthermore, LMP2A induced the expression of PRDX1 and beta-catenin. LMP2A also increased the expression of both cyclin B1 and cyclin D1. CONCLUSION: In NPC cells, PRDX1 and beta-catenin were regulated through LMP2A expression, which reduced cell growth through cell cycle-related gene expression. This study suggests that LMP2A could be a target molecule for inhibiting cancer progression in NPC cells infected with EBV.


Sujet(s)
Infections à virus Epstein-Barr , Tumeurs du rhinopharynx , Humains , Cancer du nasopharynx , Infections à virus Epstein-Barr/complications , Infections à virus Epstein-Barr/génétique , Herpèsvirus humain de type 4/génétique , bêta-Caténine/métabolisme , Tumeurs du rhinopharynx/anatomopathologie
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...