Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 3.041
Filter
1.
FASEB J ; 38(18): e23820, 2024 Sep 30.
Article in English | MEDLINE | ID: mdl-39302257

ABSTRACT

Epstein-Barr virus (EBV), a common gamma herpesvirus, establishes a life-long latent infection in the host to defend against innate immune recognition, which is closely related to a variety of malignant tumors, but its specific mechanism is unclear. BFRF3, an EBV-encoded small capsid protein, is mainly involved in the assembly of the viral capsid structure and the maintenance of its stability. Here, we showed that BFRF3 can inhibit TNF-α-mediated NF-кB promoter activation. Moreover, BFRF3 downregulates NF-кB-mediated promoter activation and transcription of inflammatory cytokines, including IL-6 and IL-8. Dual-luciferase reporter assay demonstrated that BFRF3 restrains NF-кB promoter activity at or below the p65 level, and coimmunoprecipitation analysis revealed that BFRF3 not only interacts with p65 but also binds to its critical truncated Rel homology domain (RHD) and transcriptional activation domain (TAD). However, BFRF3 does not affect the dimerization of p65-p50, but overexpression of BFRF3 reduces the nuclear accumulation of p65, and the phosphorylation of p65 (Ser536) is repressed during BFRF3 transfection and EBV lytic infection, which promotes the proliferation of EBV. Overall, our study suggested that BFRF3 may play a crucial role in antiviral immunity to defend against EBV infection by inhibiting NF-κB activity.


Subject(s)
Capsid Proteins , Herpesvirus 4, Human , NF-kappa B , Signal Transduction , Transcription Factor RelA , Humans , Herpesvirus 4, Human/metabolism , Herpesvirus 4, Human/immunology , Herpesvirus 4, Human/physiology , Capsid Proteins/metabolism , Capsid Proteins/genetics , Transcription Factor RelA/metabolism , NF-kappa B/metabolism , HEK293 Cells , Epstein-Barr Virus Infections/metabolism , Epstein-Barr Virus Infections/virology , Epstein-Barr Virus Infections/immunology , Promoter Regions, Genetic , Tumor Necrosis Factor-alpha/metabolism
3.
Virol J ; 21(1): 224, 2024 Sep 20.
Article in English | MEDLINE | ID: mdl-39304953

ABSTRACT

Nasopharyngeal carcinoma (NPC) is often diagnosed at a very advanced stage due to its location and non-specific initial symptoms. Moreover, no clinically useful serological marker has been established so far for early detection of NPC. In this study, we have investigated the clinical significance of plasma Epstein-Barr virus DNA load along with interleukin-6 (IL-6) and vascular endothelial growth factor (VEGF) levels to evaluate if these three all together can be useful as a strong serological marker for early detection and prediction of treatment response in patients with NPC. Plasma EBV DNA load, IL-6 level, VEGF expressions were measured in 24 patients with NPC at presentation and various time points during and after treatment. There was a positive correlation between high plasma EBV DNA load with higher IL-6 and VEGF expression, which was closely associated with therapeutic response as well. Persistent or recurrent plasma EBV load with higher IL-6 and VEGF levels can potentially predict disease progression and may be useful to select patients for additional therapy and longer follow-up.


Subject(s)
Carcinoma , DNA, Viral , Epstein-Barr Virus Infections , Herpesvirus 4, Human , Interleukin-6 , Nasopharyngeal Carcinoma , Nasopharyngeal Neoplasms , Vascular Endothelial Growth Factor A , Viral Load , Humans , Interleukin-6/blood , Nasopharyngeal Neoplasms/virology , Nasopharyngeal Neoplasms/blood , Nasopharyngeal Neoplasms/diagnosis , Herpesvirus 4, Human/genetics , Female , Male , DNA, Viral/blood , Middle Aged , Vascular Endothelial Growth Factor A/blood , Nasopharyngeal Carcinoma/blood , Nasopharyngeal Carcinoma/virology , Nasopharyngeal Carcinoma/diagnosis , Adult , Prognosis , Carcinoma/virology , Carcinoma/blood , Carcinoma/diagnosis , Epstein-Barr Virus Infections/blood , Epstein-Barr Virus Infections/virology , Epstein-Barr Virus Infections/diagnosis , Biomarkers/blood , Aged , Plasma/virology
4.
PLoS Pathog ; 20(9): e1012525, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39241017

ABSTRACT

Epstein-Barr virus (EBV) uses latency programs to colonize the memory B-cell reservoir, and each program is associated with human malignancies. However, knowledge remains incomplete of epigenetic mechanisms that maintain the highly restricted latency I program, present in memory and Burkitt lymphoma cells, in which EBNA1 is the only EBV-encoded protein expressed. Given increasing appreciation that higher order chromatin architecture is an important determinant of viral and host gene expression, we investigated roles of Wings Apart-Like Protein Homolog (WAPL), a host factor that unloads cohesin to control DNA loop size and that was discovered as an EBNA2-associated protein. WAPL knockout (KO) in Burkitt cells de-repressed LMP1 and LMP2A expression, but not other EBV oncogenes, to yield a viral program reminiscent of EBV latency II, which is rarely observed in B-cells. WAPL KO also increased LMP1/2A levels in latency III lymphoblastoid cells. WAPL KO altered EBV genome architecture, triggering formation of DNA loops between the LMP promoter region and the EBV origins of lytic replication (oriLyt). Hi-C analysis further demonstrated that WAPL KO reprogrammed EBV genomic DNA looping. LMP1 and LMP2A de-repression correlated with decreased histone repressive marks at their promoters. We propose that EBV coopts WAPL to negatively regulate latent membrane protein expression to maintain Burkitt latency I.


Subject(s)
Epstein-Barr Virus Infections , Gene Expression Regulation, Viral , Herpesvirus 4, Human , Viral Matrix Proteins , Virus Latency , Humans , Herpesvirus 4, Human/genetics , Virus Latency/physiology , Viral Matrix Proteins/metabolism , Viral Matrix Proteins/genetics , Epstein-Barr Virus Infections/virology , Epstein-Barr Virus Infections/metabolism , Epstein-Barr Virus Infections/genetics , B-Lymphocytes/virology , B-Lymphocytes/metabolism , Burkitt Lymphoma/virology , Burkitt Lymphoma/genetics , Burkitt Lymphoma/metabolism , Cell Line, Tumor
5.
Immun Inflamm Dis ; 12(9): e70020, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39279442

ABSTRACT

OBJECTIVE: This study aims to enhance the management of Epstein-Barr Virus (EBV) infections by analyzing the correlation between laboratory indicators and clinical manifestations in children, thereby proposing more precise diagnostic and treatment strategies. METHODS: In this retrospective study included 163 pediatric patients with EBV infections treated at the Children's Hospital of Soochow University from December 2017 to December 2019. Data collected through retrospective analysis included gender, age, clinical symptoms, signs, liver function tests, T-cell subset distribution, EBV-DNA copy numbers in plasma, and treatment outcomes. Patients were grouped based on EBV-DNA copy numbers in plasma and hospital stay duration to compare clinical indicators across different groups. RESULTS: The dichotomous results of EBV-DNA copy numbers in plasma showed that the two groups of children were significantly different in the number of days of fever (p = .0022), platelet count (p = .0212), ALT (p = .001), immunoglobulin IgM (p = .0039), IgG (p = .0195), TBiL (p = .025), LDH (p = 0.0001), and length of hospital stay (p < .001) were significantly different, indicating that EBV-DNA copy numbers in plasma may be correlated with these characteristic variables. The dichotomous results of the length of hospital stay showed that the two groups were significantly increased in tonsil enlargement (p = .0024), platelet count (p = .0059), LDH (p = .0394), and ferritin (p = .0106) and EBV-DNA copy numbers in plasma (p = 0.0361) were significantly different, This suggests a potential correlation between EBV-DNA copy numbers in plasma and these clinical indicators. CONCLUSION: Variations in platelet counts and lactate dehydrogenase (LDH) levels in children with EBV infections may serve as indicators of clinical outcomes.


Subject(s)
Herpesvirus 4, Human , Infectious Mononucleosis , Humans , Retrospective Studies , Male , Female , Infectious Mononucleosis/diagnosis , Infectious Mononucleosis/blood , Infectious Mononucleosis/virology , Infectious Mononucleosis/immunology , Child , Child, Preschool , DNA, Viral/blood , Adolescent , Epstein-Barr Virus Infections/blood , Epstein-Barr Virus Infections/virology , Epstein-Barr Virus Infections/diagnosis , Infant , Viral Load
6.
BMC Infect Dis ; 24(1): 937, 2024 Sep 09.
Article in English | MEDLINE | ID: mdl-39251898

ABSTRACT

AIMS: This study aims to evaluate the presence of EBV, HCMV, and BKV genomic sequences in the plasma samples (active infection/viremia) of kidney transplant recipients suspected of rejection and to investigate host and risk factors related to the activation of these viruses in these patients. METHODS: In this cross-sectional single-center study, plasma samples were collected from 98 suspected kidney transplant rejection patients at Labafinejad Hospital, Tehran, Iran, between December 2022 and June 2023. Quantitative real-time PCR assays for HCMV, EBV, and BK were performed using GeneProof Real-time PCR kits. ROC curve analysis was used to determine the viral load cutoff point for each virus. FINDINGS: HCMV active viremia was detected in 18 (18.36%) recipients, EBV active viremia in 7 (7.14%), and BKV active viremia in 5 (5.10%). ROC results indicated viral load cutoff points of 778, 661, and 457 points for HCMV, EBV, and BKV, respectively. The duration of time after transplantation significantly differed between active viremia and no viremia groups (120.5 vs. 46 months, P = 0.014). In the BKV active viremia group, the increase in creatinine compared to baseline creatinine was significantly higher than in the no viremia group (2.7 vs. 0.8, P = 0.017). The odds ratio of HCMV active viremia in patients taking tacrolimus was 2.84 times higher, and the odds of HCMV active viremia in patients taking antithymocyte globulin was 3.01 times higher than in patients not taking these drugs. CONCLUSION: Rapid and timely diagnosis of viral active infections in kidney transplant patients is crucial for effective disease management and implementation of appropriate treatment strategies. Identifying potential risk factors, including host and treatment-related factors that influence transplantation, can facilitate the development of suitable preventive strategies.


Subject(s)
BK Virus , Cytomegalovirus Infections , Cytomegalovirus , Epstein-Barr Virus Infections , Graft Rejection , Herpesvirus 4, Human , Kidney Transplantation , Polyomavirus Infections , Viral Load , Viremia , Humans , Kidney Transplantation/adverse effects , Male , Female , Middle Aged , BK Virus/isolation & purification , BK Virus/genetics , Adult , Cross-Sectional Studies , Polyomavirus Infections/virology , Epstein-Barr Virus Infections/virology , Epstein-Barr Virus Infections/complications , Cytomegalovirus Infections/virology , Cytomegalovirus/isolation & purification , Cytomegalovirus/genetics , Graft Rejection/virology , Herpesvirus 4, Human/genetics , Herpesvirus 4, Human/isolation & purification , Iran/epidemiology , Risk Factors , Tumor Virus Infections/virology , Tumor Virus Infections/blood , Aged , Young Adult , Transplant Recipients/statistics & numerical data
8.
Int J Mol Sci ; 25(15)2024 Jul 24.
Article in English | MEDLINE | ID: mdl-39125633

ABSTRACT

Epstein-Barr virus (EBV) infection and various chemokines, including CCL20, CXCL8 and CXCL10 are considered to participate in the pathogenesis of multiple sclerosis (MS), and several studies point to a direct regulatory effect of EBV on the expression of these chemokines. In our study we hypothesized that serum concentrations of CCL20, CXCL8 and CXCL0 are induced in patients with relapsing-remitting MS (RRMS) in comparison to healthy individuals, and that they are associated with EBV infection. Serum concentrations of CXCL8 and CXCL10 were lower in RRMS patients in relapse in comparison to healthy controls. Although potential effects of corticosteroid therapy introduced in a subgroup of RRMS patients prior to sampling were excluded by subgroup comparison, this possibility has to be considered while interpreting the results. We found an inverse association between serum concentrations of CXCL8 and anti-Epstein-Barr Virus Nuclear Antigen (EBNA) IgG and decreased expression of CXCL8 in peripheral blood mononuclear cells (PBMC) in relapse compared to remission. Lower serum concentrations of CXCL8 and CXCL10 in RRMS patients and decreased peripheral production of CXCL8 in relapse may indicate compensatory anti-inflammatory counter-regulation in MS.


Subject(s)
Chemokine CCL20 , Chemokine CXCL10 , Herpesvirus 4, Human , Interleukin-8 , Multiple Sclerosis, Relapsing-Remitting , Humans , Multiple Sclerosis, Relapsing-Remitting/blood , Multiple Sclerosis, Relapsing-Remitting/immunology , Multiple Sclerosis, Relapsing-Remitting/drug therapy , Multiple Sclerosis, Relapsing-Remitting/virology , Female , Chemokine CXCL10/blood , Adult , Male , Herpesvirus 4, Human/immunology , Chemokine CCL20/blood , Interleukin-8/blood , Antibodies, Viral/blood , Antibodies, Viral/immunology , Epstein-Barr Virus Infections/blood , Epstein-Barr Virus Infections/immunology , Epstein-Barr Virus Infections/virology , Middle Aged , Leukocytes, Mononuclear/metabolism , Leukocytes, Mononuclear/immunology , Case-Control Studies
9.
Transplantation ; 108(9): 1867-1881, 2024 Sep 01.
Article in English | MEDLINE | ID: mdl-39166902

ABSTRACT

Posttransplant lymphoproliferative disorders (PTLDs) are among the most common malignant complications after transplantation, leading to a drastic reduction in patient survival rates. The majority of PTLDs are tightly linked to Epstein-Barr virus (EBV+PTLDs) and are the result of an uncontrolled proliferation of EBV-infected cells. However, although EBV infections are a common finding in transplant recipients, most patients with high EBV loads will never develop EBV+PTLD. Natural killer cells and EBV-specific CD8+ T lymphocytes are critical for controlling EBV-infected cells, and the impairment of these cytotoxic immune responses facilitates the unfettered proliferation of EBV-infected cells. Recent years have seen a considerable increase in available literature aiming to describe novel risk factors associated with the development of EBV+PTLD, which may critically relate to the strength of EBV-specific natural killer cell and EBV-CD8+ T lymphocyte responses. The accumulation of risk factors and the increased risk of developing EBV+PTLD go hand in hand. On the one hand, most of these risk factors, such as the level of immunosuppression or the EBV donor and recipient serologic mismatch, and distinct genetic risk factors are host related and affect cytotoxic EBV-specific immune responses. On the other hand, there is growing evidence that distinct EBV variants may have an increased malignant potential and are thus more likely to induce EBV+PTLD. Here, we aim to review, from a mechanistic point of view, the risk factors for EBV+PTLD in the host and the infecting EBV variants that may explain why only a minority of transplant recipients develop EBV+PTLD.


Subject(s)
Epstein-Barr Virus Infections , Herpesvirus 4, Human , Killer Cells, Natural , Lymphoproliferative Disorders , Organ Transplantation , Humans , Lymphoproliferative Disorders/virology , Lymphoproliferative Disorders/immunology , Lymphoproliferative Disorders/etiology , Epstein-Barr Virus Infections/immunology , Epstein-Barr Virus Infections/virology , Herpesvirus 4, Human/immunology , Risk Factors , Organ Transplantation/adverse effects , Killer Cells, Natural/immunology , Killer Cells, Natural/virology , CD8-Positive T-Lymphocytes/immunology , Animals , Immunosuppressive Agents/adverse effects , Host-Pathogen Interactions
10.
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
11.
Int J Biol Macromol ; 278(Pt 4): 134837, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39179085

ABSTRACT

Nasopharyngeal carcinoma (NPC) is closely related to Epstein-Barr virus (EBV) infection. Long noncoding RNAs (lncRNAs) play important roles in cancers. However, the molecular mechanism underlying the roles of lncRNAs in EBV-associated NPC remains largely unclear. In this study, we confirmed that the expression of the lncRNA brain cytoplasmic 200 (BC200) was significantly increased in EBV-infected NPC cells and tissues. BC200 facilitated the growth and migration of NPC cells, suggesting that it participated in NPC progression by functioning as an oncogene. Mechanistically, BC200 was found to act as a ceRNA by sponging and inhibiting miR-6834-5p. Thymidylate synthetase (TYMS), whose high expression was reported to be an independent indicator of poor prognosis in NPC via an unknown mechanism, was identified as a target gene of miR-6834-5p in the present study. BC200 upregulated TYMS expression in a manner that depends on miR-6834-5p. TYMS was abnormally upregulated in EBV-positive NPC cells and tissues, and its ectopic expression contributed to the proliferation and migration of NPC cells. This study highlights the role of lncRNA BC200, which is upregulated by EBV, in promoting the development of NPC, suggesting that BC200-mediated ceRNA network may be valuable biomarkers for the diagnosis and treatment of EBV-associated NPC.


Subject(s)
Cell Movement , Cell Proliferation , Epstein-Barr Virus Infections , Gene Expression Regulation, Neoplastic , Herpesvirus 4, Human , MicroRNAs , Nasopharyngeal Carcinoma , Nasopharyngeal Neoplasms , RNA, Long Noncoding , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Humans , Nasopharyngeal Carcinoma/genetics , Nasopharyngeal Carcinoma/virology , Nasopharyngeal Carcinoma/metabolism , Nasopharyngeal Carcinoma/pathology , MicroRNAs/genetics , MicroRNAs/metabolism , Herpesvirus 4, Human/genetics , Cell Line, Tumor , Epstein-Barr Virus Infections/genetics , Epstein-Barr Virus Infections/virology , Epstein-Barr Virus Infections/metabolism , Cell Proliferation/genetics , Cell Movement/genetics , Nasopharyngeal Neoplasms/genetics , Nasopharyngeal Neoplasms/virology , Nasopharyngeal Neoplasms/metabolism , Nasopharyngeal Neoplasms/pathology , Thymidylate Synthase/genetics , Thymidylate Synthase/metabolism , Up-Regulation/genetics
12.
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
13.
Virus Genes ; 60(5): 488-500, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39103702

ABSTRACT

Epstein-Barr virus (EBV) infection has a strong correlation with the development of nasopharyngeal carcinoma (NPC). Aquaporin 3 (AQP3), a member of the aquaporin family, plays an important role in tumor development, especially in epithelial-mesenchymal transition. In this study, the expression of AQP3 in EBV-positive NPC cells was significantly lower than that in EBV-negative NPC cells. Western blot and qRT-PCR analysis showed that LMP1 down-regulated the expression of AQP3 by activating the ERK pathway. Cell biology experiments have confirmed that AQP3 affects the development of tumor by promoting cell migration and proliferation in NPC cells. In addition, AQP3 can promote the lysis of EBV in EBV-positive NPC cells. The inhibition of AQP3 expression by EBV through LMP1 may be one of the mechanisms by which EBV maintains latent infection-induced tumor progression.


Subject(s)
Aquaporin 3 , Cell Movement , Down-Regulation , Epstein-Barr Virus Infections , Herpesvirus 4, Human , Nasopharyngeal Carcinoma , Nasopharyngeal Neoplasms , Viral Matrix Proteins , Humans , Viral Matrix Proteins/genetics , Viral Matrix Proteins/metabolism , Nasopharyngeal Carcinoma/virology , Nasopharyngeal Carcinoma/pathology , Nasopharyngeal Carcinoma/metabolism , Nasopharyngeal Carcinoma/genetics , Herpesvirus 4, Human/genetics , Herpesvirus 4, Human/pathogenicity , Aquaporin 3/metabolism , Aquaporin 3/genetics , Epstein-Barr Virus Infections/virology , Nasopharyngeal Neoplasms/virology , Nasopharyngeal Neoplasms/pathology , Nasopharyngeal Neoplasms/genetics , Nasopharyngeal Neoplasms/metabolism , Cell Line, Tumor , Latent Infection/virology , Cell Proliferation , Carcinoma/virology , Carcinoma/genetics
14.
Viruses ; 16(8)2024 Aug 14.
Article in English | MEDLINE | ID: mdl-39205268

ABSTRACT

The acquisition or reactivation of Epstein-Barr virus (EBV) after allogeneic Hematopoietic Stem Cell Transplant (HSCT) can be associated with complications including the development of post-transplant lymphoproliferative disorder (PTLD), which is associated with significant morbidity and mortality. A number of risk factors for PTLD have been defined, including T-cell depletion, and approaches to monitoring EBV, especially in high-risk patients, with the use of preemptive therapy upon viral activation have been described. Newer therapies for the preemption or treatment of PTLD, such as EBV-specific cytotoxic T-cells, hold promise. Further studies to help define risks, diagnosis, and treatment of EBV-related complications are needed in this at-risk population.


Subject(s)
Epstein-Barr Virus Infections , Hematopoietic Stem Cell Transplantation , Herpesvirus 4, Human , Lymphoproliferative Disorders , Transplantation, Homologous , Virus Activation , Hematopoietic Stem Cell Transplantation/adverse effects , Humans , Epstein-Barr Virus Infections/virology , Transplantation, Homologous/adverse effects , Lymphoproliferative Disorders/etiology , Lymphoproliferative Disorders/therapy , Risk Factors
15.
Pathology ; 56(6): 773-785, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39127542

ABSTRACT

Epstein-Barr virus (EBV) is a ubiquitous gammaherpesvirus that has been related to oncogenesis of lymphoid and epithelial malignancies. Although the mechanism of EBV infection of NK and T cells remains enigmatic, it plays a pathogenic role in various EBV+ NK-cell and T-cell lymphoproliferative diseases (LPDs), through promotion of cell activation pathways, inhibition of cell apoptotic pathways, behaving as oncogenes, interacting with host oncogenes or acting epigenetically. The study of NK-cell LPDs, previously hampered by the lack of immunophenotypical and genotypical criteria of NK cells, has become feasible with the recently accepted criteria. EBV+ NK- and T-cell LPDs are mostly of poor prognosis. This review delivers a short history from primeval to recent EBV+ NK- and T-cell LPDs in non-immunocompromised subjects, coupled with increasing interest, and work on the biological and oncogenic roles of EBV.


Subject(s)
Epstein-Barr Virus Infections , Herpesvirus 4, Human , Killer Cells, Natural , Lymphoproliferative Disorders , T-Lymphocytes , Humans , Lymphoproliferative Disorders/virology , Lymphoproliferative Disorders/pathology , Lymphoproliferative Disorders/immunology , Epstein-Barr Virus Infections/immunology , Epstein-Barr Virus Infections/complications , Epstein-Barr Virus Infections/virology , Herpesvirus 4, Human/immunology , Killer Cells, Natural/immunology , Killer Cells, Natural/pathology , Killer Cells, Natural/virology , T-Lymphocytes/immunology , T-Lymphocytes/pathology
16.
Int J Mol Sci ; 25(16)2024 Aug 21.
Article in English | MEDLINE | ID: mdl-39201739

ABSTRACT

The relationship between Toll-like receptors (TLRs) and prostate cancer (PCa) is complex due to the presence of the Epstein-Barr virus (EBV) infection, which has been identified as a predisposing factor for some cancers, including PCa. The present study aims to investigate these complex links by examining the levels of selected TLRs and the potential impact of EBV infection on PCa. Therefore, we examined the serum of patients with PCa. The study compared EBV(+) patients to risk groups, the Gleason score (GS), and the T-trait. Additionally, the correlation between TLR and antibody levels was examined. The results indicated that higher levels of TLR-2 and TLR-9 were observed in more advanced PCa. The findings of this study may contribute to a deeper understanding of the role of viral infections in PCa and provide information on future strategies for the diagnosis, prevention, and treatment of these malignancies.


Subject(s)
Epstein-Barr Virus Infections , Herpesvirus 4, Human , Prostatic Neoplasms , Toll-Like Receptor 2 , Toll-Like Receptor 9 , Humans , Male , Prostatic Neoplasms/blood , Prostatic Neoplasms/virology , Toll-Like Receptor 2/blood , Toll-Like Receptor 2/metabolism , Epstein-Barr Virus Infections/blood , Epstein-Barr Virus Infections/virology , Epstein-Barr Virus Infections/complications , Aged , Middle Aged , Neoplasm Grading
18.
Clin Transplant ; 38(8): e15424, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39136236

ABSTRACT

BACKGROUND: Epstein-Barr virus (EBV) is a herpesvirus linked to nine different human tumors and lymphoproliferative disorders. Immunosuppression promotes EBV-driven malignancies. The most frequent EBV-induced malignancies are lymphomas and nasopharyngeal carcinoma. By promoting smooth muscle proliferation, EBV can induce EBV-associated smooth muscle tumors (EBV-SMT). EBV-SMT is a rare oncological entity for which no current guideline for diagnosis or management exists. Data on posttransplant EBV-SMT (PT-SMT) are scarce in kidney transplant recipients. METHODS: We conducted a national multicentric retrospective study and collected cases among transplantation centers in France. Kidney transplant recipients experiencing histologically proven PT-SMT were included. We collected data on demographic characteristics of patient, history of kidney transplantation, history of PT-SMT, evolution of graft function, and patient survival. RESULTS: Eight patients were included. The median age at PT-SMT diagnosis was 31 years (range 6.5-40). PT-SMT occurred after a median delay of 37.8 months after transplantation (range 6-175). PT-SMT management consisted in immunosuppressive regimen minimization in all patients. Introduction of mTOR inhibitors was performed in two patients. Four patients (50%) needed chemotherapy. Surgical resection was performed in four patients. At last follow-up after PT-SMT diagnosis (median 33 months (range 17-132)), five patients were considered in complete remission, and two patients had died. Two patients experienced graft rejection; two resumed dialysis (25%). All patients with available data presented with impaired graft function at last follow-up. CONCLUSION: PT-SMT is a subacute and progressive disease during kidney transplantation. Even if the risk of developing PT-SMT is low in kidney transplant recipients (0.07% in our cohort), PT-SMT is associated with significant graft loss, possibly due to reduced immunosuppression. Developing guidelines could help transplantation teams better manage these patients.


Subject(s)
Epstein-Barr Virus Infections , Herpesvirus 4, Human , Kidney Transplantation , Postoperative Complications , Smooth Muscle Tumor , Humans , Kidney Transplantation/adverse effects , Retrospective Studies , Male , Female , Smooth Muscle Tumor/virology , Smooth Muscle Tumor/etiology , Smooth Muscle Tumor/pathology , Smooth Muscle Tumor/diagnosis , Adult , Follow-Up Studies , Epstein-Barr Virus Infections/complications , Epstein-Barr Virus Infections/virology , Herpesvirus 4, Human/isolation & purification , Prognosis , France/epidemiology , Adolescent , Young Adult , Child , Postoperative Complications/diagnosis , Graft Rejection/etiology , Kidney Failure, Chronic/surgery , Graft Survival , Risk Factors , Kidney Function Tests , Glomerular Filtration Rate , Survival Rate
19.
Int J Mol Sci ; 25(15)2024 Jul 26.
Article in English | MEDLINE | ID: mdl-39125729

ABSTRACT

Epstein-Barr virus (EBV), a member of the γ-herpesvirus family, is one of the most prevalent and persistent human viruses, infecting up to 90% of the adult population globally. EBV's life cycle includes primary infection, latency, and lytic reactivation, with the virus primarily infecting B cells and epithelial cells. This virus has evolved sophisticated strategies to evade both innate and adaptive immune responses, thereby maintaining a lifelong presence within the host. This persistence is facilitated by the expression of latent genes such as EBV nuclear antigens (EBNAs) and latent membrane proteins (LMPs), which play crucial roles in viral latency and oncogenesis. In addition to their well-known roles in several types of cancer, including nasopharyngeal carcinoma and B-cell lymphomas, recent studies have identified the pathogenic roles of EBV in autoimmune diseases such as multiple sclerosis, rheumatoid arthritis, and systemic lupus erythematosus. This review highlights the intricate interactions between EBV and the host immune system, underscoring the need for further research to develop effective therapeutic and preventive strategies against EBV-associated diseases.


Subject(s)
Autoimmune Diseases , Epstein-Barr Virus Infections , Herpesvirus 4, Human , Immune Evasion , Humans , Herpesvirus 4, Human/immunology , Herpesvirus 4, Human/pathogenicity , Autoimmune Diseases/immunology , Autoimmune Diseases/virology , Epstein-Barr Virus Infections/immunology , Epstein-Barr Virus Infections/virology , Animals , Virus Latency/immunology
20.
J Med Virol ; 96(8): e29834, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39092825

ABSTRACT

Emerging biologic subsets and new prognostic markers are significantly important for aggressive diffuse large B-cell lymphoma (DLBCL). Nevertheless, the high cost of testing limits the availability of these tests in most hospitals, thus making prognostic judgment based on basic immunohistochemical testing, whole blood Epstein-Barr virus DNA (WBEBV) surveillance and clinical features advantageous for hospitals and patients with poor medical conditions. We included 647 DLBCL patients treated in our hospital from January 2009 to March 2023. Non-germinal center B-cell like, Ki-67, and International Prognostic Index (IPI) scores were related to cMYC/B-cell lymphoma 2 (Bcl-2)-double expression. Age, Epstein-Barr virus-encoded small RNA (EBER) positivity, and IPI scores were associated with mortality. The cutoffs for differential overall survival (OS) of age, WBEBV, Bcl-2, and cMYC were 57 years, 1514 copies/mL (baseline), 5.89 × 104 copies/mL (treatment), 40%, and 55%, respectively. EBER positivity was significantly associated with a worse OS. Patients with newly defined DE (Bcl-2 ≥ 40 and cMYC > 55) had a worse prognosis than controls (p = 0.04). We found that cMYC with an optimal cutoff of 47.5 could effectively predict high-grade DLBCL with an area under the curve of 0.912, and the specificity and sensitivity were 70.7% and 100%, respectively. Our study provides valuable insights into the prognostic factors and biomarker cutoffs that influence OS in DLBCL patients, which may guide clinicians in tailoring treatment strategies and improving patient outcomes.


Subject(s)
Epstein-Barr Virus Infections , Herpesvirus 4, Human , Lymphoma, Large B-Cell, Diffuse , Humans , Lymphoma, Large B-Cell, Diffuse/diagnosis , Lymphoma, Large B-Cell, Diffuse/virology , Male , Female , Epstein-Barr Virus Infections/diagnosis , Epstein-Barr Virus Infections/virology , Middle Aged , Prognosis , Aged , Herpesvirus 4, Human/genetics , Herpesvirus 4, Human/isolation & purification , Adult , Aged, 80 and over , Immunohistochemistry/methods , Young Adult , DNA, Viral , Biomarkers, Tumor , Adolescent , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/analysis , Retrospective Studies
SELECTION OF CITATIONS
SEARCH DETAIL