Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.257
Filter
1.
J Clin Immunol ; 44(8): 172, 2024 Aug 07.
Article in English | MEDLINE | ID: mdl-39110273

ABSTRACT

The clinical penetrance of infectious diseases varies considerably among patients with inborn errors of immunity (IEI), even for identical genetic defects. This variability is influenced by pathogen exposure, healthcare access and host-environment interactions. We describe here a patient in his thirties who presented with epidermodysplasia verruciformis (EV) due to infection with a weakly virulent beta-papillomavirus (HPV38) and CD4+ T-cell lymphopenia. The patient was born to consanguineous parents living in the United States. Exome sequencing identified a previously unknown biallelic STK4 stop-gain mutation (p.Trp425X). The patient had no relevant history of infectious disease during childhood other than mild wart-like lesion on the skin, but he developed diffuse large B-cell lymphoma (DLBCL) and EBV viremia with a low viral load in his thirties. Despite his low CD4+ T-cell count, the patient had normal counts of CD3+ cells, predominantly double-negative T cells (67.4%), which turned out to be Vδ2+ γδ T cells. γδ T-cell expansion has frequently been observed in the 33 reported cases with STK4 deficiency. The Vδ2 γδ T cells of this STK4-deficient patient are mostly CD45RA-CD27+CCR7+ central memory γδT cells, and their ability to proliferate in response to T-cell activation was impaired, as was that of CD4+ T cells. In conclusion, γδ T-cell expansion may act as a compensatory mechanism to combat viral infection, providing immune protection in immunocompromised individuals.


Subject(s)
Epidermodysplasia Verruciformis , Protein Serine-Threonine Kinases , Humans , Epidermodysplasia Verruciformis/genetics , Epidermodysplasia Verruciformis/diagnosis , Male , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/deficiency , Adult , Receptors, Antigen, T-Cell, gamma-delta/genetics , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/deficiency , Lymphoma, Large B-Cell, Diffuse/genetics , Lymphoma, Large B-Cell, Diffuse/etiology , Lymphoma, Large B-Cell, Diffuse/immunology , Lymphoma, Large B-Cell, Diffuse/diagnosis , Mutation/genetics , Epstein-Barr Virus Infections/immunology , Epstein-Barr Virus Infections/genetics , Epstein-Barr Virus Infections/complications , Intraepithelial Lymphocytes/immunology , Consanguinity
2.
J Med Virol ; 96(8): e29869, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39165093

ABSTRACT

Epstein-Barr virus (EBV) is a highly successful pathogen that infects ~95% of the adult population and is associated with diverse cancers and autoimmune diseases. The most abundant viral factor in latently infected cells is not a protein but a noncoding RNA called EBV-encoded RNA 1 (EBER1). Even though EBER1 is highly abundant and was discovered over forty years ago, the function of EBER1 has remained elusive. EBER1 interacts with the ribosomal protein L22, which normally suppresses the expression of its paralog L22-like 1 (L22L1). Here we show that when L22 binds EBER1, it cannot suppress L22L1, resulting in L22L1 being expressed and incorporated into ribosomes. We further show that L22L1-containing ribosomes preferentially translate mRNAs involved in the oxidative phosphorylation pathway. Moreover, upregulation of L22L1 is indispensable for growth transformation and immortalization of resting B cells upon EBV infection. Taken together, our results suggest that the function of EBER1 is to modulate host gene expression at the translational level, thus bypassing the need for dysregulating host gene transcription.


Subject(s)
Herpesvirus 4, Human , Oxidative Phosphorylation , RNA, Viral , Ribosomal Proteins , Ribosomal Proteins/genetics , Ribosomal Proteins/metabolism , Humans , Herpesvirus 4, Human/genetics , Herpesvirus 4, Human/physiology , RNA, Viral/genetics , RNA, Viral/metabolism , B-Lymphocytes/virology , Host-Pathogen Interactions/genetics , Epstein-Barr Virus Infections/virology , Epstein-Barr Virus Infections/genetics , Epstein-Barr Virus Infections/metabolism , Ribosomes/metabolism , Ribosomes/genetics , RNA-Binding Proteins
3.
Skin Res Technol ; 30(8): e70002, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39167023

ABSTRACT

BACKGROUND: As a common chronic recurrent inflammatory skin disease, psoriasis is characterized by erythema and scaly skin lesions, with infection as an integral part of the pathogenesis of many diseases. Many previous cases reported the impact of psoriasis on infection. However, the existing research fails to completely clarify the infection factors associated with the potential of these diseases and causality. MATERIALS AND METHODS: Thirteen kinds of pathogens and their immune responses and psoriasis in the phenotype of 46 species of SNPs data were respectively obtained from the GWAS catalog database and the UK biobank database. With the help of R software, three methods of inverse variance weighted (IVW), weighted median (WME), and MR-Egger regression were used to analyze the causality of the dataset. RESULTS: According to the results of IVW analysis, there is a causal relationship between anti-Epstein Barr virus antibody and psoriasis (OR: 1.003, 95% CI: 1.001∼1.006, P = 0.046) with a positive correlation. CONCLUSION: Based on the results of MR analysis, there is a causal relationship between psoriasis and EBV infection, which indicates that EBV infection can increase the risk or severity of psoriasis. Therefore, in clinical scenarios, patients afflicted with psoriasis should be prevented from contracting the infection and recurrence of EBV as well as symptoms of psoriasis. The underlying immunological mechanism also provides a new perspective for experimental research.


Subject(s)
Mendelian Randomization Analysis , Polymorphism, Single Nucleotide , Psoriasis , Psoriasis/genetics , Psoriasis/epidemiology , Humans , Genome-Wide Association Study , Epstein-Barr Virus Infections/genetics , Epstein-Barr Virus Infections/complications , Epstein-Barr Virus Infections/epidemiology , Genetic Predisposition to Disease/genetics , Risk Factors
4.
Sci Rep ; 14(1): 20236, 2024 08 30.
Article in English | MEDLINE | ID: mdl-39215087

ABSTRACT

Systemic lupus erythematosus (SLE) is a multifactorial disease characterized by the convergence of genetic, immunological, and viral elements resulting in a complex interaction of both internal and external factors. The role of the Epstein-Barr virus (EBV) and human endogenous retroviruses (HERV-E) as triggers and maintenance elements in the pathogenesis of SLE has been widely recognized. Previous studies have independently evaluated the effects of EBV and HERV-E in this disease. In this work, for the first time, these viral factors are jointly investigated in SLE patients. This study aimed at assessing the differential expression of immune regulatory genes and the incidence of specific viral pathogens (EBV and HERV-E), alongside the detailed characterization of surface markers in T- and B-lymphocytes in patients with SLE and control participants. A comparative analysis between patients with SLE and control participants was performed, evaluating the expression of phenotypic markers and genes involved in the immune response (TNF-α, IL-2, IL-6, IL-10, IFNG, TLR3), as well as HERV-E gag and EBV viral genes (LMP1 and BZLF1).A significant association between SLE and EBV was found in this study. A notable increase in EBV LMP1 gene expression was observed in patients with SLE . Also, a significant overexpression of HERV-E was observed, in addition to a considerable increase in the distribution of the cell surface marker CD27 + on T- and B-lymphocytes, observed in individuals with SLE compared to the control group. This study provides evidence regarding the role that EBV virus plays in lymphocytes in the context of SLE, highlighting how both the virus and the host gene expression may influence disease pathogenesis by altering immune regulatory pathways mediated by TNF-α, IFN-γ, and IL-10, as well as parallel overexpression of HERV-E gag. The decrease in TLR3 could indicate a compromised antiviral response, which could facilitate viral reactivation and contribute to disease activity.


Subject(s)
Endogenous Retroviruses , Herpesvirus 4, Human , Leukocytes, Mononuclear , Lupus Erythematosus, Systemic , Humans , Lupus Erythematosus, Systemic/immunology , Lupus Erythematosus, Systemic/genetics , Lupus Erythematosus, Systemic/virology , Endogenous Retroviruses/genetics , Herpesvirus 4, Human/immunology , Herpesvirus 4, Human/genetics , Adult , Female , Male , Leukocytes, Mononuclear/immunology , Leukocytes, Mononuclear/virology , Leukocytes, Mononuclear/metabolism , Gene Expression Profiling , Epstein-Barr Virus Infections/immunology , Epstein-Barr Virus Infections/virology , Epstein-Barr Virus Infections/genetics , Middle Aged , B-Lymphocytes/immunology , B-Lymphocytes/virology , Case-Control Studies , T-Lymphocytes/immunology , Cytokines/metabolism , Cytokines/genetics
5.
Virus Genes ; 60(5): 464-474, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39096336

ABSTRACT

Epstein-Barr virus (EBV) is the first human oncogenic virus known to express microRNAs (miRNAs), which are closely associated with the development of various tumors, including nasopharyngeal and gastric cancers. Stearoyl-CoA Desaturase 1 (SCD1) is a key enzyme in fatty acid synthesis, highly expressed in numerous tumors, promoting tumor growth and metastasis, making it a potential therapeutic target. In this study, we found that SCD1 expression in EBV-associated gastric cancer (EBVaGC) was significantly lower than in EBV-negative gastric cancer (EBVnGC) at both cellular and tissue levels. In addition, EBV-miR-BART20-5p targets the 3'-UTR of SCD1, downregulating its expression. Moreover, overexpression of SCD1 in EBVaGC cells promoted cell migration and proliferation while inhibiting autophagy. These results suggest that EBV-encoded miRNA-BART20-5p may contribute to EBVaGC progression by targeting SCD1.


Subject(s)
Autophagy , Cell Movement , Cell Proliferation , Herpesvirus 4, Human , MicroRNAs , Stearoyl-CoA Desaturase , Stomach Neoplasms , Humans , Stomach Neoplasms/virology , Stomach Neoplasms/genetics , Stomach Neoplasms/pathology , MicroRNAs/genetics , Stearoyl-CoA Desaturase/genetics , Autophagy/genetics , Cell Movement/genetics , Herpesvirus 4, Human/genetics , Cell Proliferation/genetics , Cell Line, Tumor , Epstein-Barr Virus Infections/virology , Epstein-Barr Virus Infections/genetics , Gene Expression Regulation, Neoplastic , 3' Untranslated Regions/genetics , RNA, Viral/genetics
6.
J Autoimmun ; 148: 103279, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38972102

ABSTRACT

B cells of people with multiple sclerosis (MS) are more responsive to IFN-γ, corresponding to their brain-homing potential. We studied how a coding single nucleotide polymorphism (SNP) in IFNGR2 (rs9808753) co-operates with Epstein-Barr virus (EBV) infection as MS risk factors to affect the IFN-γ signaling pathway in human B cells. In both cell lines and primary cells, EBV infection positively associated with IFN-γ receptor expression and STAT1 phosphorylation. The IFNGR2 risk SNP selectively promoted downstream signaling via STAT1, particularly in transitional B cells. Altogether, EBV and the IFNGR2 risk SNP independently amplify IFN-γ signaling, potentially driving B cells to enter the MS brain.


Subject(s)
B-Lymphocytes , Epstein-Barr Virus Infections , Interferon gamma Receptor , Interferon-gamma , Multiple Sclerosis , Polymorphism, Single Nucleotide , Receptors, Interferon , Signal Transduction , Humans , Multiple Sclerosis/genetics , Multiple Sclerosis/immunology , Receptors, Interferon/genetics , Receptors, Interferon/metabolism , Interferon-gamma/metabolism , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Epstein-Barr Virus Infections/immunology , Epstein-Barr Virus Infections/genetics , STAT1 Transcription Factor/metabolism , STAT1 Transcription Factor/genetics , Genetic Predisposition to Disease , Herpesvirus 4, Human , Female , Male , Phosphorylation
8.
Viruses ; 16(7)2024 Jun 24.
Article in English | MEDLINE | ID: mdl-39066175

ABSTRACT

Viral coinfection among HIV-positive patients, coupled with the development of AIDS, remains a major public health problem. The synergism between the presence of HIV and other viruses has consequences in relation to changes in the severity of the infection, as well as changes in the natural course of both infections. Several polymorphisms present in genes that encode cytokines have a relevant influence on their transcription and consequently on the production of such immunological molecules. The present study evaluated the influence of SNPs located in the promoter regions of genes encoding the cytokines INF-É£, TNF, IL-6, IL-4, and IL-2, as well as their respective plasma concentrations, in patients infected with HIV and/or EBV in the state of Pará. Additionally, this study described the epidemiological profile and compared CD4+ and CD8+ T lymphocyte counts among the groups studied. The associative analysis between the SNPs and plasma cytokine concentrations in different groups showed statistical relevance for three polymorphisms: rs2069762 (IL2), where the GG genotype demonstrated higher IL-2 levels in HIV mono-infected individuals; rs2243250 (IL4), where the CT genotype showed higher IL-4 levels in the control group; and rs2069705 (IFNG), where the TT genotype showed higher IFN-γ levels in the coinfected group. Regarding SNP associations with CD4+/CD8+ counts, significant findings were observed in HIV mono-infected individuals: the rs2069705 (IFNG) polymorphism was linked to higher CD4+ counts with the CT genotype, and rs1799964 (TNF) was associated with higher CD8+ counts with the CC genotype. Therefore, this study provides evidence that the rs2069705 (IFNG) SNP is associated with elevated IFN-γ levels, which may have pathogenic consequences, as depletion of this cytokine is concerning for people living with HIV due to its antiviral properties.


Subject(s)
Coinfection , Cytokines , Epstein-Barr Virus Infections , HIV Infections , HIV-1 , Herpesvirus 4, Human , Polymorphism, Single Nucleotide , Humans , HIV Infections/genetics , HIV Infections/immunology , HIV Infections/virology , HIV Infections/complications , Brazil/epidemiology , Epstein-Barr Virus Infections/genetics , Epstein-Barr Virus Infections/immunology , Epstein-Barr Virus Infections/virology , Epstein-Barr Virus Infections/complications , Male , Adult , Female , HIV-1/immunology , HIV-1/genetics , Cytokines/genetics , Cytokines/blood , Middle Aged , Coinfection/virology , Coinfection/immunology , Coinfection/genetics , Herpesvirus 4, Human/immunology , Herpesvirus 4, Human/genetics , Genotype , CD8-Positive T-Lymphocytes/immunology , Young Adult , CD4 Lymphocyte Count , Immunogenetics
9.
Int J Mol Sci ; 25(14)2024 Jul 17.
Article in English | MEDLINE | ID: mdl-39063079

ABSTRACT

The Epstein-Barr virus (EBV) has a very high prevalence (>90% in adults), establishes a lifelong latency after primary infection, and exerts an oncogenic potential. This dsDNA virus encodes for various molecules, including microRNAs (miRs), which can be detected in the latent and lytic phases with different expression levels and affect, among others, immune evasion and malignant transformation. In this study, the different EBV miRs are quantified in EBV-positive lymphomas, and the impact on the host cell transcriptome of the most abundant EBV miRs will be analyzed using comparative RNA sequencing analyses. The EBV miRs ebv-miR-BART1, -BART4, -BART17, and -BHRF1-1 were most highly expressed, and their selective overexpression in EBV-negative human cells resulted in a large number of statistically significantly down- and up-regulated host cell genes. Functional analyses showed that these dysregulated target genes are involved in important cellular processes, including growth factor pathways such as WNT, EGF, FGF, and PDGF, as well as cellular processes such as apoptosis regulation and inflammation. Individual differences were observed between these four analyzed EBV miRs. In particular, ebv-miR-BHRF1-1 appears to be more important for malignant transformation and immune evasion than the other EBV miRs.


Subject(s)
Epstein-Barr Virus Infections , Herpesvirus 4, Human , MicroRNAs , Transcriptome , Humans , MicroRNAs/genetics , Herpesvirus 4, Human/genetics , Epstein-Barr Virus Infections/virology , Epstein-Barr Virus Infections/genetics , RNA, Viral/genetics , Gene Expression Regulation, Viral , Host-Pathogen Interactions/genetics , Gene Expression Profiling
10.
J Clin Immunol ; 44(7): 153, 2024 Jun 19.
Article in English | MEDLINE | ID: mdl-38896122

ABSTRACT

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.


Subject(s)
Epstein-Barr Virus Infections , Hodgkin Disease , Lymphohistiocytosis, Hemophagocytic , Humans , Male , Cation Transport Proteins , Epstein-Barr Virus Infections/diagnosis , Epstein-Barr Virus Infections/complications , Epstein-Barr Virus Infections/genetics , Hematopoietic Stem Cell Transplantation , Herpesvirus 4, Human , Hodgkin Disease/diagnosis , Hodgkin Disease/genetics , Hodgkin Disease/etiology , Lymphohistiocytosis, Hemophagocytic/diagnosis , Lymphohistiocytosis, Hemophagocytic/etiology , Lymphohistiocytosis, Hemophagocytic/genetics , Recurrence
11.
Neoplasia ; 55: 101012, 2024 09.
Article in English | MEDLINE | ID: mdl-38875930

ABSTRACT

Increased mutational burden and EBV load have been revealed from normal tissues to Epstein-Barr virus (EBV)-associated gastric carcinomas (EBVaGCs). BPLF1, encoded by EBV, is a lytic cycle protein with deubiquitinating activity has been found to participate in disrupting repair of DNA damage. We first confirmed that BPLF1 gene in gastric cancer (GC) significantly increased the DNA double strand breaks (DSBs). Ubiquitination mass spectrometry identified histones as BPLF1 interactors and potential substrates, and co-immunoprecipitation and in vitro experiments verified that BPLF1 regulates H2Bub by targeting Rad6. Over-expressing Rad6 restored H2Bub but partially reduced γ-H2AX, suggesting that other downstream DNA repair processes were affected. mRNA expression of BRCA2 were significantly down-regulated by next-generation sequencing after over-expression of BPLF1, and over-expression of p65 facilitated the repair of DSBs. We demonstrated BPLF1 may lead to the accumulation of DSBs by two pathways, reducing H2B ubiquitination (H2Bub) and blocking homologous recombination which may provide new ideas for the treatment of gastric cancer.


Subject(s)
DNA Breaks, Double-Stranded , Epstein-Barr Virus Infections , Genomic Instability , Herpesvirus 4, Human , Histones , Stomach Neoplasms , Ubiquitination , Humans , Herpesvirus 4, Human/genetics , Stomach Neoplasms/virology , Stomach Neoplasms/genetics , Stomach Neoplasms/metabolism , Stomach Neoplasms/pathology , Stomach Neoplasms/etiology , Epstein-Barr Virus Infections/virology , Epstein-Barr Virus Infections/metabolism , Epstein-Barr Virus Infections/complications , Epstein-Barr Virus Infections/genetics , Histones/metabolism , Cell Line, Tumor , DNA Repair , Deubiquitinating Enzymes/metabolism , Deubiquitinating Enzymes/genetics , Gene Expression Regulation, Neoplastic , BRCA2 Protein/genetics , BRCA2 Protein/metabolism , Carcinogenesis/genetics , Viral Regulatory and Accessory Proteins
12.
Int J Mol Sci ; 25(11)2024 May 28.
Article in English | MEDLINE | ID: mdl-38892033

ABSTRACT

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.


Subject(s)
Epstein-Barr Virus Infections , Heart Failure , Herpesvirus 4, Human , Myocarditis , Humans , Herpesvirus 4, Human/genetics , Heart Failure/virology , Heart Failure/genetics , Heart Failure/pathology , Male , Female , Epstein-Barr Virus Infections/virology , Epstein-Barr Virus Infections/complications , Epstein-Barr Virus Infections/genetics , Epstein-Barr Virus Infections/pathology , Middle Aged , Myocarditis/virology , Myocarditis/pathology , Aged , High-Throughput Nucleotide Sequencing , Myocardium/pathology , Myocardium/metabolism , DNA, Viral/genetics , Adult , Biopsy
13.
mBio ; 15(7): e0079524, 2024 Jul 17.
Article in English | MEDLINE | ID: mdl-38874417

ABSTRACT

Epstein-Barr virus (EBV) is a ubiquitous human tumor virus that establishes lifelong, persistent infections in B cells. The presence of EBV in cancer cells presents an opportunity to target these cells by reactivating the virus from latency. In this study, we developed a novel approach for EBV reactivation termed clustered regularly interspaced short palindromic repeats (CRISPR)/dCas9-mediated EBV reactivation (CMER) strategy. Using modified CRISPR-associated protein 9 (dCas9) fused with VP64, we designed 10 single guide RNAs (sgRNAs) to target and activate the EBV immediate-early gene promoter. In Akata Burkitt lymphoma cells, 9 out of 10 CMER sgRNAs effectively reactivated EBV. Among these, CMER sgRNA-5 triggered robust reactivation across various cell types, including lymphoma, gastric cancer, and nasopharyngeal carcinoma cells. Importantly, the combination of CMER and ganciclovir selectively eliminated EBV-positive cells, regardless of their cell origin. These findings indicate that targeted virus reactivation by CMER, combined with nucleoside analog therapy, holds promise for EBV-associated cancer treatment. IMPORTANCE: This study explores a novel strategy called clustered regularly interspaced short palindromic repeats (CRISPR)/dCas9-mediated Epstein-Barr virus (EBV) reactivation (CMER) to reactivate the Epstein-Barr virus in cancer cells. EBV is associated with various cancers, and reactivating EBV from latency offers a potential therapeutic strategy. We utilized an enzymatically inactive CRISPR-associated protein 9 (dCas9) fused with VP64 and designed 10 single guide RNAs to target the EBV immediate-early gene promoter. Nine of these sgRNAs effectively reactivated EBV in Burkitt lymphoma cells, with CMER sgRNA-5 demonstrating strong reactivation across different cancer cell types. Combining CMER with ganciclovir selectively eliminated EBV-positive cells, showing promise for EBV-associated cancer treatment.


Subject(s)
CRISPR-Cas Systems , Epstein-Barr Virus Infections , Ganciclovir , Herpesvirus 4, Human , Virus Activation , Humans , Herpesvirus 4, Human/genetics , Herpesvirus 4, Human/drug effects , Herpesvirus 4, Human/physiology , Virus Activation/drug effects , Virus Activation/genetics , Epstein-Barr Virus Infections/virology , Epstein-Barr Virus Infections/genetics , Cell Line, Tumor , Ganciclovir/pharmacology , Virus Latency/genetics , Virus Latency/drug effects , Antiviral Agents/pharmacology , Burkitt Lymphoma/genetics , Burkitt Lymphoma/virology , Clustered Regularly Interspaced Short Palindromic Repeats , CRISPR-Associated Protein 9/genetics
14.
PLoS Pathog ; 20(6): e1011970, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38885264

ABSTRACT

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.


Subject(s)
CRISPR-Cas Systems , Epstein-Barr Virus Infections , Herpesvirus 4, Human , MicroRNAs , Virus Activation , Virus Latency , Humans , Herpesvirus 4, Human/genetics , MicroRNAs/genetics , MicroRNAs/metabolism , Epstein-Barr Virus Infections/virology , Epstein-Barr Virus Infections/genetics , Epstein-Barr Virus Infections/metabolism , Burkitt Lymphoma/virology , Burkitt Lymphoma/genetics , Burkitt Lymphoma/metabolism , Cell Line, Tumor
15.
J Clin Immunol ; 44(7): 155, 2024 Jun 26.
Article in English | MEDLINE | ID: mdl-38922539

ABSTRACT

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.


Subject(s)
Dermatomyositis , Epstein-Barr Virus Infections , Microfilament Proteins , Mutation , Humans , Male , Epstein-Barr Virus Infections/diagnosis , Epstein-Barr Virus Infections/genetics , Epstein-Barr Virus Infections/complications , Epstein-Barr Virus Infections/immunology , Dermatomyositis/genetics , Dermatomyositis/diagnosis , Dermatomyositis/immunology , Child , Microfilament Proteins/genetics , Mutation/genetics , Herpesvirus 4, Human , Exome Sequencing , Immunologic Deficiency Syndromes/genetics , Immunologic Deficiency Syndromes/diagnosis , Autoantibodies/blood , Autoantibodies/immunology , Phenotype , T-Lymphocytes/immunology
16.
Oncogene ; 43(28): 2143-2159, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38778160

ABSTRACT

Angiogenesis is one of the characteristics of malignant tumors, and persistent generation of abnormal tumor blood vessels is an important factor contributing to tumor treatment resistance. Epstein-Barr virus (EBV) is a highly prevalent DNA oncogenic virus that is associated with the development of various epithelial malignancies. However, the relationship between EBV infection and tumor vascular abnormalities as well as its underlying mechanisms is still unclear. In this study, we found that compared to EBV-uninfected tumors, EBV-infected tumors were more angiogenic, but the neovascularization was mostly immature vessels without pericyte attachment in both clinical patient tumor samples and mouse xenograft models; These immature vessels exhibited aberrant functionality, characterized by poor blood perfusion and increased vascular permeability. The vascular abnormalities caused by EBV infection exacerbated tumor hypoxia and was responsible for accelerated tumor growth. Mechanistically, EBV infection upregulated ANXA3-HIF-1α-VEGF pathway. Silencing the ANXA3 gene or neutralizing ANXA3 with an antibody can diminish vascular abnormalities, thereby increasing immune cell infiltration and alleviating treatment resistance. Finally, a new therapy combining ANXA3 blockade and NK cell + PD1 antibody significantly inhibited the growth of EBV-infected xenografts in mice. In conclusion, our study identified a previously unrecognized role for EBV infection in tumor vascular abnormalities and revealed its underlying mechanism that upregulated the ANXA3-HIF-1α-VEGF pathway. ANXA3 is a potential therapeutic target for EBV-infected tumors and ANXA3 blockade to improve vascular conditions, in combination with NK cell + PD1 antibody therapy, holds promise as an effective treatment strategy for EBV-associated epithelial malignancies.


Subject(s)
Epstein-Barr Virus Infections , Herpesvirus 4, Human , Hypoxia-Inducible Factor 1, alpha Subunit , Neovascularization, Pathologic , Vascular Endothelial Growth Factor A , Humans , Animals , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Mice , Epstein-Barr Virus Infections/complications , Epstein-Barr Virus Infections/virology , Epstein-Barr Virus Infections/genetics , Epstein-Barr Virus Infections/pathology , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor A/genetics , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/pathology , Neovascularization, Pathologic/virology , Neovascularization, Pathologic/metabolism , Cell Line, Tumor , Up-Regulation , Signal Transduction , Xenograft Model Antitumor Assays , Female
17.
Nat Commun ; 15(1): 4156, 2024 May 16.
Article in English | MEDLINE | ID: mdl-38755141

ABSTRACT

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.


Subject(s)
Herpesvirus 4, Human , Histones , PTB-Associated Splicing Factor , Virus Activation , Virus Latency , Herpesvirus 4, Human/genetics , Herpesvirus 4, Human/physiology , Humans , Histones/metabolism , Virus Activation/genetics , Virus Latency/genetics , PTB-Associated Splicing Factor/metabolism , PTB-Associated Splicing Factor/genetics , Gene Expression Regulation, Viral , B-Lymphocytes/virology , B-Lymphocytes/metabolism , Epstein-Barr Virus Infections/virology , Epstein-Barr Virus Infections/genetics , Epstein-Barr Virus Infections/metabolism , CRISPR-Cas Systems , Promoter Regions, Genetic/genetics , Trans-Activators/metabolism , Trans-Activators/genetics , Genome, Viral
18.
Nat Microbiol ; 9(6): 1540-1554, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38806670

ABSTRACT

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.


Subject(s)
B-Lymphocytes , Epstein-Barr Virus Infections , Herpesvirus 4, Human , Multiple Sclerosis , Humans , Herpesvirus 4, Human/genetics , Multiple Sclerosis/virology , Multiple Sclerosis/immunology , Multiple Sclerosis/genetics , Multiple Sclerosis/metabolism , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , B-Lymphocytes/virology , Epstein-Barr Virus Infections/virology , Epstein-Barr Virus Infections/immunology , Epstein-Barr Virus Infections/genetics , Epstein-Barr Virus Infections/complications , Cytokines/metabolism , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/virology , CD4-Positive T-Lymphocytes/metabolism , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , Transcriptome , Virus Replication , Gene Expression Regulation, Viral , Cell Line , Host-Pathogen Interactions/genetics , Host-Pathogen Interactions/immunology , Gene Expression Profiling , Adult , Female , Male
19.
Int J Mol Sci ; 25(9)2024 May 03.
Article in English | MEDLINE | ID: mdl-38732219

ABSTRACT

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.


Subject(s)
Epstein-Barr Virus Infections , Genetic Variation , Herpesvirus 4, Human , Phylogeny , Recombination, Genetic , Herpesvirus 4, Human/genetics , Humans , Brazil , Epstein-Barr Virus Infections/virology , Epstein-Barr Virus Infections/genetics , Trans-Activators/genetics , Male , Female , Haplotypes/genetics , Adult , Viral Matrix Proteins/genetics , Child , Middle Aged , Adolescent , Virus Latency/genetics , Child, Preschool , Young Adult
20.
PLoS Pathog ; 20(5): e1012263, 2024 May.
Article in English | MEDLINE | ID: mdl-38805547

ABSTRACT

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.


Subject(s)
Epstein-Barr Virus Infections , Genome, Viral , Herpesvirus 4, Human , Nasopharyngeal Carcinoma , Nasopharyngeal Neoplasms , Female , Humans , Male , China/epidemiology , East Asian People , Epstein-Barr Virus Infections/virology , Epstein-Barr Virus Infections/genetics , Genetic Variation , Genome-Wide Association Study , Herpesvirus 4, Human/genetics , Nasopharyngeal Carcinoma/virology , Nasopharyngeal Carcinoma/genetics , Nasopharyngeal Neoplasms/virology , Nasopharyngeal Neoplasms/genetics , Phylogeny , Polymorphism, Single Nucleotide
SELECTION OF CITATIONS
SEARCH DETAIL