Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 616
Filter
1.
J Virol ; 98(2): e0189923, 2024 Feb 20.
Article in English | MEDLINE | ID: mdl-38294245

ABSTRACT

After Epstein-Barr virus (EBV) genome replication and encapsidation in the nucleus, nucleocapsids are translocated into the cytoplasm for subsequent tegumentation and maturation. The EBV BGLF4 kinase, which induces partial disassembly of the nuclear lamina, and the nuclear egress complex BFRF1/BFLF2 coordinately facilitate the nuclear egress of nucleocapsids. Here, we demonstrate that within EBV reactivated epithelial cells, viral capsids, tegument proteins, and glycoproteins are clustered in the juxtanuclear concave region, accompanied by redistributed cytoplasmic organelles and the cytoskeleton regulator IQ-domain GTPase-activation protein 1 (IQGAP1), close to the microtubule-organizing center (MTOC). The assembly compartment (AC) structure was diminished in BGLF4-knockdown TW01-EBV cells and BGLF4-knockout bacmid-carrying TW01 cells, suggesting that the formation of AC structure is BGLF4-dependent. Notably, glycoprotein gp350/220 was observed by confocal imaging to be distributed in the perinuclear concave region and surrounded by the endoplasmic reticulum (ER) membrane marker calnexin, indicating that the AC may be located within a globular structure derived from ER membranes, adjacent to the outer nuclear membrane. Moreover, the viral capsid protein BcLF1 and tegument protein BBLF1 were co-localized with IQGAP1 near the cytoplasmic membrane in the late stage of replication. Knockdown of IQGAP1 did not affect the AC formation but decreased virion release from both TW01-EBV and Akata+ cells, suggesting IQGAP1-mediated trafficking regulates EBV virion release. The data presented here show that BGLF4 is required for cytoskeletal rearrangement, coordination with the redistribution of cytoplasmic organelles and IQGAP1 for virus maturation, and subsequent IQGAP1-dependent virion release.IMPORTANCEEBV genome is replicated and encapsidated in the nucleus, and the resultant nucleocapsids are translocated to the cytoplasm for subsequent virion maturation. We show that a cytoplasmic AC, containing viral proteins, markers of the endoplasmic reticulum, Golgi, and endosomes, is formed in the juxtanuclear region of epithelial and B cells during EBV reactivation. The viral BGLF4 kinase contributes to the formation of the AC. The cellular protein IQGAP1 is also recruited to the AC and partially co-localizes with the virus capsid protein BcLF1 and tegument protein BBLF1 in EBV-reactivated cells, dependent on the BGLF4-induced cytoskeletal rearrangement. In addition, virion release was attenuated in IQGAP1-knockdown epithelial and B cells after reactivation, suggesting that IQGAP1-mediated trafficking may regulate the efficiency of virus maturation and release.


Subject(s)
Cytoplasm , Herpesvirus 4, Human , Protein Serine-Threonine Kinases , Viral Proteins , Virion , Virus Assembly , Virus Release , ras GTPase-Activating Proteins , Humans , Capsid Proteins/metabolism , Cytoplasm/metabolism , Cytoplasm/virology , Epstein-Barr Virus Infections/metabolism , Epstein-Barr Virus Infections/virology , Herpesvirus 4, Human/chemistry , Herpesvirus 4, Human/genetics , Herpesvirus 4, Human/growth & development , Herpesvirus 4, Human/metabolism , Membrane Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , ras GTPase-Activating Proteins/metabolism , Viral Proteins/metabolism , Virion/chemistry , Virion/growth & development , Virion/metabolism , Virus Assembly/physiology , Endoplasmic Reticulum/metabolism , Endosomes/metabolism , Golgi Apparatus/metabolism
2.
Nat Commun ; 13(1): 187, 2022 01 17.
Article in English | MEDLINE | ID: mdl-35039491

ABSTRACT

Epstein-Barr virus (EBV) persists in human B-cells by maintaining its chromatinized episomes within the nucleus. We have previously shown that cellular factor Poly [ADP-ribose] polymerase 1 (PARP1) binds the EBV genome, stabilizes CTCF binding at specific loci, and that PARP1 enzymatic activity correlates with maintaining a transcriptionally active latency program. To better understand PARP1's role in regulating EBV latency, here we functionally characterize the effect of PARP enzymatic inhibition on episomal structure through in situ HiC mapping, generating a complete 3D structure of the EBV genome. We also map intragenomic contact changes after PARP inhibition to global binding of chromatin looping factors CTCF and cohesin across the EBV genome. We find that PARP inhibition leads to fewer total unique intragenomic interactions within the EBV episome, yet new chromatin loops distinct from the untreated episome are also formed. This study also illustrates that PARP inhibition alters gene expression at the regions where chromatin looping is most effected. We observe that PARP1 inhibition does not alter cohesin binding sites but does increase its frequency of binding at those sites. Taken together, these findings demonstrate that PARP has an essential role in regulating global EBV chromatin structure and latent gene expression.


Subject(s)
Cell Cycle Proteins/genetics , Chromatin/chemistry , Chromosomal Proteins, Non-Histone/genetics , Chromosome Mapping/methods , Genome, Viral , Herpesvirus 4, Human/genetics , Poly (ADP-Ribose) Polymerase-1/genetics , B-Lymphocytes/pathology , B-Lymphocytes/virology , CCCTC-Binding Factor/metabolism , Cell Cycle Proteins/metabolism , Chromatin/metabolism , Chromosomal Proteins, Non-Histone/metabolism , Epstein-Barr Virus Infections/virology , Gene Expression Regulation , Herpesvirus 4, Human/drug effects , Herpesvirus 4, Human/growth & development , Herpesvirus 4, Human/immunology , Host-Pathogen Interactions , Humans , Models, Molecular , Phthalazines/pharmacology , Piperazines/pharmacology , Plasmids/metabolism , Poly (ADP-Ribose) Polymerase-1/metabolism , Protein Binding , Signal Transduction , Transcription, Genetic , Virus Latency/genetics , Cohesins
3.
Am J Trop Med Hyg ; 106(2): 623-625, 2021 11 29.
Article in English | MEDLINE | ID: mdl-34844205

ABSTRACT

Epstein-Barr virus (EBV) is the most common cause of infectious mononucleosis (IM) and IM is a clinical syndrome typically characterized by fever, pharyngitis, and cervical lymph node enlargement. We describe the case of a 19-year-old man with IM complicated by splenic infarction. The patient visited our hospital because of upper abdominal pain without a fever and sore throat. Abdominal computed tomography revealed a low-density area in the spleen, which indicated splenic infarction. The next day, he developed a fever. After diminishing abdominal pain and fever, he developed pharyngitis accompanied by fever. Acute EBV infection was confirmed by serological tests. The patient was successfully managed with no specific therapy. Splenic infarction is a rare complication of IM and this case showed that splenic infarction can precede a fever and pharyngitis.


Subject(s)
Epstein-Barr Virus Infections/pathology , Infectious Mononucleosis/pathology , Spleen/pathology , Splenic Infarction/pathology , Abdominal Pain/physiopathology , Epstein-Barr Virus Infections/diagnostic imaging , Epstein-Barr Virus Infections/virology , Fever/physiopathology , Herpesvirus 4, Human/growth & development , Herpesvirus 4, Human/pathogenicity , Humans , Infectious Mononucleosis/diagnostic imaging , Infectious Mononucleosis/virology , Lymphadenopathy/physiopathology , Male , Pharyngitis/physiopathology , Remission, Spontaneous , Spleen/diagnostic imaging , Spleen/virology , Splenic Infarction/diagnostic imaging , Splenic Infarction/virology , Tomography, X-Ray Computed , Young Adult
4.
Biomolecules ; 11(8)2021 08 04.
Article in English | MEDLINE | ID: mdl-34439817

ABSTRACT

Epstein-Barr virus (EBV) and cytomegalovirus (CMV) are viruses globally distributed that have been associated with the development and prognosis of many pathologies, including hematological diseases. This study aimed to characterize the epidemiological profile of EBV infection and the infection-correlated hepatic manifestations in patients with hematological diseases of the northern Brazilian state of Amazonas. A total of 228 patients were serologically tested for the presence of anti-EBV and anti-CMV IgG antibodies through an enzyme-linked immunosorbent assay. The coinfection with CMV, sociodemographic and laboratory records of all patients were also assessed. The overall prevalence observed among the study population for EBV infection and EBV/CMV coinfection was 85.09% (95% CI: 0.80-0.90) and 78.51% (95% CI: 0.73-0.84), respectively. The age group 31-40 years old were more susceptible to EBV/CMV coinfection (95% CI: 1.59-93.41, p = 0.011), while young people aged 1-10 years old were less affected for both EBV infection (CI 95%; 0.66-0.91, p = 0.001) and EBV/CMV coinfection (95% CI: 0.52-0.81, p < 0.0001). High serum levels of the liver biomarker ferritin were associated with EBV infection (95% CI: 1.03-1.54, p = 0.031) and EBV/CMV coinfection (95% CI: 1.02-1.70, p = 0.038). Our findings indicated that the elevated prevalence of EBV infection is not associated with the hematological diseases or transfusion rates, but with the socioeconomic status of the study population. Also, this study suggests that the EBV infection and its coinfection with CMV are related to the increase of serum ferritin levels.


Subject(s)
Anemia/epidemiology , Antibodies, Viral/blood , Cytomegalovirus Infections/epidemiology , Epstein-Barr Virus Infections/epidemiology , Ferritins/blood , Leukemia/epidemiology , Lymphoma/epidemiology , Adolescent , Adult , Aged , Aged, 80 and over , Anemia/immunology , Anemia/pathology , Anemia/virology , Biomarkers/blood , Blood Transfusion/statistics & numerical data , Brazil/epidemiology , Child , Child, Preschool , Coinfection , Cytomegalovirus/growth & development , Cytomegalovirus/pathogenicity , Cytomegalovirus Infections/immunology , Cytomegalovirus Infections/pathology , Cytomegalovirus Infections/virology , Epstein-Barr Virus Infections/immunology , Epstein-Barr Virus Infections/pathology , Epstein-Barr Virus Infections/virology , Female , Herpesvirus 4, Human/growth & development , Herpesvirus 4, Human/pathogenicity , Humans , Immunoglobulin G/blood , Infant , Infant, Newborn , Leukemia/immunology , Leukemia/pathology , Leukemia/virology , Liver/immunology , Liver/pathology , Liver/virology , Lymphoma/immunology , Lymphoma/pathology , Lymphoma/virology , Male , Middle Aged , Prevalence , Social Class
5.
J Virol ; 95(13): e0246620, 2021 06 10.
Article in English | MEDLINE | ID: mdl-33853959

ABSTRACT

Epstein-Barr virus (EBV) is a ubiquitous herpesvirus responsible for several diseases, including cancers of lymphoid and epithelial cells. EBV cancers typically exhibit viral latency; however, the production and release of EBV through its lytic phase are essential for cancer development. Antiviral agents that specifically target EBV production do not currently exist. Previously, we reported that the proton pump inhibitor tenatoprazole, which blocks the interaction of ubiquitin with the ESCRT-1 factor Tsg101, inhibits production of several enveloped viruses, including EBV. Here, we show that three structurally distinct prazoles impair mature particle formation postreactivation and identify the impact on stages of replication. The prazoles did not impair expression of lytic genes representative of the different kinetic classes but interfered with capsid maturation in the nucleus as well as virion transport from the nucleus. Replacement of endogenous Tsg101 with a mutant Tsg101 refractory to prazole-mediated inhibition rescued EBV release. These findings directly implicate Tsg101 in EBV nuclear egress and identify prazoles as potential therapeutic candidates for conditions that rely on EBV replication, such as chronic active EBV infection and posttransplant lymphoproliferative disorders. IMPORTANCE Production of virions is necessary for the ubiquitous Epstein-Barr virus (EBV) to persist in humans and can set the stage for development of EBV cancers in at-risk individuals. In our attempts to identify inhibitors of the EBV lytic phase, we previously found that a prazole proton pump inhibitor, known to block the interaction of ubiquitin with the ESCRT-1 factor Tsg101, blocks production of EBV. We now find that three structurally distinct prazoles impair maturation of EBV capsids and virion transport from the nucleus and, by interfering with Tsg101, prevent EBV release from lytically active cells. Our findings not only implicate Tsg101 in EBV production but also identify widely used prazoles as candidates to prevent development of posttransplant EBV lymphomas.


Subject(s)
2-Pyridinylmethylsulfinylbenzimidazoles/pharmacology , Antiviral Agents/pharmacology , DNA-Binding Proteins/metabolism , Endosomal Sorting Complexes Required for Transport/metabolism , Rabeprazole/pharmacology , Transcription Factors/metabolism , Virus Release/drug effects , A549 Cells , Cell Line, Tumor , Epstein-Barr Virus Infections/prevention & control , HEK293 Cells , Herpesvirus 4, Human/drug effects , Herpesvirus 4, Human/growth & development , Humans , Proton Pump Inhibitors/pharmacology , Viral Load/drug effects , Virus Activation/drug effects , Virus Latency/drug effects , Virus Replication/drug effects
6.
Dis Mon ; 67(9): 101166, 2021 Sep.
Article in English | MEDLINE | ID: mdl-33663798

ABSTRACT

OBJECTIVES: The effect of antiretroviral therapy (ART) on the oral pathogenic microbes in human immunodeficiency virus-1 seropositive patients remains relatively unexplored. Thus, the present study assessed the effect of ART on the sub-gingival levels of 3 pathogenic microbes. MATERIALS AND METHODS: The study groups consisted of 60 human immunodeficiency virus-1 seropositive patients divided into 3 groups of 20 each. Group 1 had periodontitis and did not start with the ART. Group 2 had periodontitis and started with ART (Tenofovir Disoproxil Fumarate 300 mg + Lamivudine 300 mg + Efavirenz 600 mg) at least 6 months before the study. Group 3 with normal periodontium, and have not started ART. The sub-gingival loads of Cytomegalovirus, Epstein-Barr virus, and the Porphyromonas gingivalis levels were assessed, along with the CD4 counts. RESULTS: The cytomegalovirus load was highest in group 1, followed by groups 2, and 3 (p-value of 0.271). The Epstein-Barr load was highest for group 2, followed by group 3, and 1 (p-value of 0.022). The P.gingivalis load was highest in group 2, followed by groups 1 and 3, (p-value of 0.028). The Epstein-Barr and Cytomegalovirus counts were significantly higher (p-value < 0.02) when the CD4 counts were less than 500 cells/cu3. CONCLUSION: ART did not cause any significant reduction in the sub-gingival levels of any of the 3 examined microbes. Given the lack of any significant effect on the sub-gingival microbial loads by the ART, human immunodeficiency virus patients may require additional anti-microbial agents and regular mechanical plaque removal to maintain their periodontal status.


Subject(s)
Antiretroviral Therapy, Highly Active , Cytomegalovirus/growth & development , HIV Infections , HIV-1/growth & development , Herpesvirus 4, Human/growth & development , Periodontitis , Porphyromonas gingivalis/growth & development , Adult , Anti-HIV Agents/pharmacology , Anti-HIV Agents/therapeutic use , Bacteroidaceae Infections/complications , Bacteroidaceae Infections/microbiology , CD4 Lymphocyte Count , Cytomegalovirus Infections/complications , Cytomegalovirus Infections/virology , Epstein-Barr Virus Infections/complications , Epstein-Barr Virus Infections/virology , Female , HIV Infections/complications , HIV Infections/drug therapy , HIV Infections/virology , Humans , Male , Periodontitis/complications , Periodontitis/microbiology , Periodontitis/virology , Periodontium/drug effects , Periodontium/microbiology , Periodontium/pathology , Periodontium/virology
7.
Nat Commun ; 12(1): 117, 2021 01 05.
Article in English | MEDLINE | ID: mdl-33402692

ABSTRACT

Nasopharyngeal cancer (NPC), endemic in Southeast Asia, lacks effective diagnostic and therapeutic strategies. Even in high-income countries the 5-year survival rate for stage IV NPC is less than 40%. Here we report high somatostatin receptor 2 (SSTR2) expression in multiple clinical cohorts comprising 402 primary, locally recurrent and metastatic NPCs. We show that SSTR2 expression is induced by the Epstein-Barr virus (EBV) latent membrane protein 1 (LMP1) via the NF-κB pathway. Using cell-based and preclinical rodent models, we demonstrate the therapeutic potential of SSTR2 targeting using a cytotoxic drug conjugate, PEN-221, which is found to be superior to FDA-approved SSTR2-binding cytostatic agents. Furthermore, we reveal significant correlation of SSTR expression with increased rates of survival and report in vivo uptake of the SSTR2-binding 68Ga-DOTA-peptide radioconjugate in PET-CT scanning in a clinical trial of NPC patients (NCT03670342). These findings reveal a key role in EBV-associated NPC for SSTR2 in infection, imaging, targeted therapy and survival.


Subject(s)
Epstein-Barr Virus Infections , Gene Expression Regulation, Neoplastic , Nasopharyngeal Carcinoma , Nasopharyngeal Neoplasms , Neoplasm Recurrence, Local , Receptors, Somatostatin , Viral Matrix Proteins , Animals , Female , Humans , Male , Mice , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Epstein-Barr Virus Infections/drug therapy , Epstein-Barr Virus Infections/genetics , Epstein-Barr Virus Infections/mortality , Epstein-Barr Virus Infections/virology , Herpesvirus 4, Human/drug effects , Herpesvirus 4, Human/growth & development , Herpesvirus 4, Human/pathogenicity , Host-Pathogen Interactions/genetics , Lymphatic Metastasis , Mice, Nude , Molecular Targeted Therapy , Nasopharyngeal Carcinoma/drug therapy , Nasopharyngeal Carcinoma/genetics , Nasopharyngeal Carcinoma/mortality , Nasopharyngeal Carcinoma/virology , Nasopharyngeal Neoplasms/drug therapy , Nasopharyngeal Neoplasms/genetics , Nasopharyngeal Neoplasms/mortality , Nasopharyngeal Neoplasms/virology , Neoplasm Recurrence, Local/drug therapy , Neoplasm Recurrence, Local/genetics , Neoplasm Recurrence, Local/mortality , Neoplasm Recurrence, Local/virology , NF-kappa B/genetics , NF-kappa B/metabolism , Octreotide/pharmacology , Positron Emission Tomography Computed Tomography , Receptors, Somatostatin/antagonists & inhibitors , Receptors, Somatostatin/genetics , Receptors, Somatostatin/metabolism , Signal Transduction , Survival Analysis , Viral Matrix Proteins/antagonists & inhibitors , Viral Matrix Proteins/genetics , Viral Matrix Proteins/metabolism , Xenograft Model Antitumor Assays
8.
Semin Cell Dev Biol ; 111: 135-147, 2021 03.
Article in English | MEDLINE | ID: mdl-32631785

ABSTRACT

Oncogenic DNA viruses establish lifelong infections in humans, and they cause cancers, often in immunocompromised patients, despite anti-viral immune surveillance targeted against viral antigens. High-throughput sequencing techniques allowed the field to identify novel viral non-coding RNAs (ncRNAs). ncRNAs are ideal factors for DNA viruses to exploit; they are non-immunogenic to T cells, thus viral ncRNAs can manipulate host cells without evoking adaptive immune responses. Viral ncRNAs may still trigger the host innate immune response, but many viruses encode decoys/inhibitors to counter-act and evade recognition. In addition, ncRNAs can be secreted to the extracellular space and influence adjacent cells to create a pro-viral microenvironment. In this review, we present recent progress in understanding interactions between oncoviruses and ncRNAs including small and long ncRNAs, microRNAs, and recently identified viral circular RNAs. In addition, potential clinical applications for ncRNA will be discussed. Extracellular ncRNAs are suggested to be diagnostic and prognostic biomarkers and, with the realization of the importance of viral ncRNAs in tumorigenesis, approaches to target critical viral ncRNAs are emerging. Further understanding of viral utilization of ncRNAs will advance anti-viral therapeutics beyond conventional medication and vaccination.


Subject(s)
Immune Evasion/genetics , MicroRNAs/genetics , Neoplasms/genetics , RNA, Circular/genetics , RNA, Long Noncoding/genetics , RNA, Viral/genetics , Virus Diseases/genetics , Alphapapillomavirus/genetics , Alphapapillomavirus/growth & development , Alphapapillomavirus/pathogenicity , Antiviral Agents/therapeutic use , Carcinogenesis/genetics , Carcinogenesis/immunology , Carcinogenesis/pathology , Gene Expression Regulation , Herpesvirus 4, Human/genetics , Herpesvirus 4, Human/growth & development , Herpesvirus 4, Human/pathogenicity , Herpesvirus 8, Human/genetics , Herpesvirus 8, Human/growth & development , Herpesvirus 8, Human/pathogenicity , Human T-lymphotropic virus 1/genetics , Human T-lymphotropic virus 1/growth & development , Human T-lymphotropic virus 1/pathogenicity , Humans , Immunity, Innate , MicroRNAs/antagonists & inhibitors , MicroRNAs/immunology , Neoplasms/immunology , Neoplasms/therapy , Neoplasms/virology , Oligonucleotides, Antisense/therapeutic use , RNA, Circular/immunology , RNA, Long Noncoding/immunology , RNA, Viral/immunology , Signal Transduction , Virus Diseases/immunology , Virus Diseases/therapy , Virus Diseases/virology
9.
Kaohsiung J Med Sci ; 37(4): 346-347, 2021 04.
Article in English | MEDLINE | ID: mdl-33340392
10.
Nat Commun ; 11(1): 5405, 2020 10 26.
Article in English | MEDLINE | ID: mdl-33106493

ABSTRACT

Epstein-Barr virus (EBV) is a γ-herpesvirus associated with the occurrence of several human malignancies. BBRF2 and BSRF1 are two EBV tegument proteins that have been suggested to form a hetero-complex and mediate viral envelopment, but the molecular basis of their interaction and the functional mechanism of this complex remains unknown. Here, we present crystal structures of BBRF2 alone and in complex with BSRF1. BBRF2 has a compact globular architecture featuring a central ß-sheet that is surrounded by 10 helices, it represents a novel fold distinct from other known protein structures. The central portion of BSRF1 folds into two tightly associated antiparallel α-helices, forming a composite four-helix bundle with two α-helices from BBRF2 via a massive hydrophobic network. In vitro, a BSRF1-derived peptide binds to BBRF2 and reduces the number of viral genome copies in EBV-positive cells. Exogenous BBRF2 and BSRF1 co-localize at the Golgi apparatus. Furthermore, BBRF2 binds capsid and capsid-associated proteins, whereas BSRF1 associates with glycoproteins. These findings indicate that the BBRF2-BSRF1 complex tethers EBV nucleocapsids to the glycoprotein-enriched Golgi membrane, facilitating secondary envelopment.


Subject(s)
Herpesvirus 4, Human/metabolism , Capsid/chemistry , Capsid/metabolism , Epstein-Barr Virus Infections/virology , Genome, Viral , Herpesvirus 4, Human/chemistry , Herpesvirus 4, Human/genetics , Herpesvirus 4, Human/growth & development , Humans , Protein Binding , Protein Conformation, alpha-Helical
11.
J Virol ; 94(21)2020 10 14.
Article in English | MEDLINE | ID: mdl-32796077

ABSTRACT

Epstein-Barr virus (EBV) is one of nine human herpesviruses that persist latently to establish permanent residence in their hosts. Periodic activation into the lytic/replicative phase allows such viruses to propagate and spread, but can also cause disease in the host. This lytic phase is also essential for EBV to cause infectious mononucleosis and cancers, including B lymphocyte-derived Burkitt lymphoma and immunocompromise-associated lymphoproliferative diseases/lymphomas as well as epithelial cell-derived nasopharyngeal cell carcinoma. In the absence of anti-EBV agents, however, therapeutic options for EBV-related diseases are limited. In earlier work, we discovered that through the activities of the viral protein kinase conserved across herpesviruses and two cellular proteins, ATM and KAP1, a lytic cycle amplification loop is established, and disruption of this loop disables the EBV lytic cascade. We therefore devised a high-throughput screening assay, screened a small-molecule-compound library, and identified 17 candidates that impair the release of lytically replicated EBV. The identified compounds will (i) serve as lead compounds or may be modified to inhibit EBV and potentially other herpesviruses, and (ii) be developed into anticancer agents, as functions of KAP1 and ATM are tightly linked to cancer. Importantly, our screening strategy may also be used to screen additional compound libraries for antiherpesviral and anticancer drugs.IMPORTANCE Epstein-Barr virus, which is nearly ubiquitous in humans, is causal to infectious mononucleosis, chronic active EBV infection, and lymphoid and epithelial cancers. However, EBV-specific antiviral agents are not yet available. To aid in the identification of compounds that may be developed as antivirals, we pursued a mechanism-based approach. Since many of these diseases rely on EBV's lytic phase, we developed a high-throughput assay that is able to measure a key step that is essential for successful completion of EBV's lytic cascade. We used this assay to screen a library of small-molecule compounds and identified inhibitors that may be pursued for their anti-EBV and possibly even antiherpesviral potential, as this key mechanism appears to be common to several human herpesviruses. Given the prominent role of this mechanism in both herpesvirus biology and cancer, our screening assay may be used as a platform to identify both antiherpesviral and anticancer drugs.


Subject(s)
Antiviral Agents/pharmacology , Ataxia Telangiectasia Mutated Proteins/genetics , Herpesvirus 4, Human/drug effects , Protein Kinases/genetics , Small Molecule Libraries/pharmacology , Trans-Activators/genetics , Tripartite Motif-Containing Protein 28/genetics , Antiviral Agents/chemistry , Ataxia Telangiectasia Mutated Proteins/metabolism , B-Lymphocytes/drug effects , B-Lymphocytes/metabolism , B-Lymphocytes/virology , Burkitt Lymphoma/drug therapy , Burkitt Lymphoma/pathology , Burkitt Lymphoma/virology , Cell Line, Tumor , Epstein-Barr Virus Infections/drug therapy , Epstein-Barr Virus Infections/pathology , Epstein-Barr Virus Infections/virology , Gene Expression Regulation , Herpesvirus 4, Human/genetics , Herpesvirus 4, Human/growth & development , Herpesvirus 4, Human/metabolism , High-Throughput Screening Assays , Host-Pathogen Interactions/drug effects , Host-Pathogen Interactions/genetics , Humans , Lysogeny/drug effects , Phosphorylation , Protein Kinases/metabolism , Signal Transduction , Small Molecule Libraries/chemistry , Trans-Activators/metabolism , Tripartite Motif-Containing Protein 28/metabolism , Virus Activation/drug effects , Virus Latency/drug effects , Virus Replication
12.
J Virol ; 94(18)2020 08 31.
Article in English | MEDLINE | ID: mdl-32641480

ABSTRACT

We previously reported that the cellular transcription factor hypoxia-inducible factor 1α (HIF-1α) binds a hypoxia response element (HRE) located within the promoter of Epstein-Barr virus's (EBV's) latent-lytic switch BZLF1 gene, Zp, inducing viral reactivation. In this study, EBV-infected cell lines derived from gastric cancers and Burkitt lymphomas were incubated with HIF-1α-stabilizing drugs: the iron chelator deferoxamine (Desferal [DFO]), a neddylation inhibitor (pevonedistat [MLN-4924]), and a prolyl hydroxylase inhibitor (roxadustat [FG-4592]). DFO and MLN-4924, but not FG-4592, induced accumulation of both lytic EBV proteins and phosphorylated p53 in cell lines that contain a wild-type p53 gene. FG-4592 also failed to activate transcription from Zp in a reporter assay despite inducing accumulation of HIF-1α and transcription from another HRE-containing promoter. Unexpectedly, DFO failed to induce EBV reactivation in cell lines that express mutant or no p53 or when p53 expression was knocked down with short hairpin RNAs (shRNAs). Likewise, HIF-1α failed to activate transcription from Zp when p53 was knocked out by CRISPR-Cas9. Importantly, DFO induced binding of p53 as well as HIF-1α to Zp in chromatin immunoprecipitation (ChIP) assays, but only when the HRE was present. Nutlin-3, a drug known to induce accumulation of phosphorylated p53, synergized with DFO and MLN-4924 in inducing EBV reactivation. Conversely, KU-55933, a drug that inhibits ataxia telangiectasia mutated, thereby preventing p53 phosphorylation, inhibited DFO-induced EBV reactivation. Lastly, activation of Zp transcription by DFO and MLN-4924 mapped to its HRE. Thus, we conclude that induction of BZLF1 gene expression by HIF-1α requires phosphorylated, wild-type p53 as a coactivator, with HIF-1α binding recruiting p53 to Zp.IMPORTANCE EBV, a human herpesvirus, is latently present in most nasopharyngeal carcinomas, Burkitt lymphomas, and some gastric cancers. To develop a lytic-induction therapy for treating patients with EBV-associated cancers, we need a way to efficiently reactivate EBV into lytic replication. EBV's BZLF1 gene product, Zta, usually controls this reactivation switch. We previously showed that HIF-1α binds the BZLF1 gene promoter, inducing Zta synthesis, and HIF-1α-stabilizing drugs can induce EBV reactivation. In this study, we determined which EBV-positive cell lines are reactivated by classes of HIF-1α-stabilizing drugs. We found, unexpectedly, that HIF-1α-stabilizing drugs only induce reactivation when they also induce accumulation of phosphorylated, wild-type p53. Fortunately, p53 phosphorylation can also be provided by drugs such as nutlin-3, leading to synergistic reactivation of EBV. These findings indicate that some HIF-1α-stabilizing drugs may be helpful as part of a lytic-induction therapy for treating patients with EBV-positive malignancies that contain wild-type p53.


Subject(s)
Herpesvirus 4, Human/genetics , Host-Pathogen Interactions/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Trans-Activators/genetics , Tumor Suppressor Protein p53/genetics , Cell Line, Tumor , Cyclopentanes/pharmacology , Deferoxamine/pharmacology , Enzyme Inhibitors/pharmacology , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Epithelial Cells/virology , Gene Expression Regulation , Glycine/analogs & derivatives , Glycine/pharmacology , Herpesvirus 4, Human/drug effects , Herpesvirus 4, Human/growth & development , Herpesvirus 4, Human/metabolism , Host-Pathogen Interactions/drug effects , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/agonists , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Imidazoles/pharmacology , Iron Chelating Agents/pharmacology , Isoquinolines/pharmacology , Lymphocytes/drug effects , Lymphocytes/metabolism , Lymphocytes/virology , Morpholines/pharmacology , Piperazines/pharmacology , Prolyl-Hydroxylase Inhibitors/pharmacology , Promoter Regions, Genetic , Protein Binding/drug effects , Pyrimidines/pharmacology , Pyrones/pharmacology , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Response Elements , Signal Transduction , Trans-Activators/metabolism , Tumor Suppressor Protein p53/antagonists & inhibitors , Tumor Suppressor Protein p53/metabolism , Virus Activation/drug effects
13.
J Virol ; 94(17)2020 08 17.
Article in English | MEDLINE | ID: mdl-32581094

ABSTRACT

The Epstein-Barr virus (EBV) BHLF1 gene encodes an abundant linear and several circular RNAs believed to perform noncoding functions during virus replication, although an open reading frame (ORF) is retained among an unknown percentage of EBV isolates. Evidence suggests that BHLF1 is also transcribed during latent infection, which prompted us to investigate the contribution of this locus to latency. Analysis of transcripts transiting BHLF1 revealed that its transcription is widespread among B-cell lines supporting the latency I or III program of EBV protein expression and is more complex than originally presumed. EBV-negative Burkitt lymphoma cell lines infected with either wild-type or two different BHLF1 mutant EBVs were initially indistinguishable in supporting latency III. However, cells infected with BHLF1- virus ultimately transitioned to the more restrictive latency I program, whereas cells infected with wild-type virus either sustained latency III or transitioned more slowly to latency I. Upon infection of primary B cells, which require latency III for growth in vitro, both BHLF1- viruses exhibited variably reduced immortalization potential relative to the wild-type virus. Finally, in transfection experiments, efficient protein expression from an intact BHLF1 ORF required the EBV posttranscriptional regulator protein SM, whose expression is limited to the replicative cycle. Thus, one way in which BHLF1 may contribute to latency is through a mechanism, possibly mediated or regulated by a long noncoding RNA, that supports latency III critical for the establishment of EBV latency and lifelong persistence within its host, whereas any retained protein-dependent function of BHLF1 may be restricted to the replication cycle.IMPORTANCE Epstein-Barr virus (EBV) has significant oncogenic potential that is linked to its latent infection of B lymphocytes, during which virus replication is not supported. The establishment of latent infection, which is lifelong and can precede tumor development by years, requires the concerted actions of nearly a dozen EBV proteins and numerous small non-protein-coding RNAs. Elucidating how these EBV products contribute to latency is crucial for understanding EBV's role in specific malignancies and, ultimately, for clinical intervention. Historically, EBV genes that contribute to virus replication have been excluded from consideration of a role in latency, primarily because of the general incompatibility between virus production and cell survival. However, here, we provide evidence that the genetic locus containing one such gene, BHLF1, indeed contributes to key aspects of EBV latency, including its ability to promote the continuous growth of B lymphocytes, thus providing significant new insight into EBV biology and oncogenic potential.


Subject(s)
B-Lymphocytes/virology , Herpesvirus 4, Human/genetics , Herpesvirus 4, Human/metabolism , Viral Proteins/genetics , Viral Proteins/metabolism , Virus Latency/physiology , Burkitt Lymphoma , Cell Line , Epstein-Barr Virus Infections/virology , Epstein-Barr Virus Nuclear Antigens/genetics , Epstein-Barr Virus Nuclear Antigens/metabolism , Gene Expression Regulation, Viral , HEK293 Cells , Herpesvirus 4, Human/growth & development , Humans , RNA, Long Noncoding/genetics , Transcriptome , Virus Replication
14.
Adv Exp Med Biol ; 1225: 127-135, 2020.
Article in English | MEDLINE | ID: mdl-32030652

ABSTRACT

Epstein Barr virus (EBV) and Kaposi sarcoma-associated herpesvirus (KSHV) constitute the human γ-herpesviruses and two of the seven human tumor viruses. In addition to their viral oncogenes that primarily belong to the latent infection programs of these viruses, they encode proteins that condition the microenvironment. Many of these are early lytic gene products and are only expressed in a subset of infected cells of the tumor mass. In this chapter I will describe their function and the evidence that targeting them in addition to the latent oncogenes could be beneficial for the treatment of EBV- and KSHV-associated malignancies.


Subject(s)
Herpesvirus 4, Human/growth & development , Herpesvirus 4, Human/pathogenicity , Herpesvirus 8, Human/growth & development , Herpesvirus 8, Human/pathogenicity , Neoplasms/drug therapy , Neoplasms/virology , Oncogenes , Tumor Microenvironment , Virus Replication , Herpesvirus 4, Human/drug effects , Herpesvirus 4, Human/genetics , Herpesvirus 8, Human/drug effects , Herpesvirus 8, Human/genetics , Humans , Oncogenes/drug effects , Virus Replication/drug effects
15.
J Virol ; 94(10)2020 05 04.
Article in English | MEDLINE | ID: mdl-32102884

ABSTRACT

The Epstein-Barr virus (EBV) causes human cancers, and epidemiological studies have shown that lytic replication is a risk factor for some of these tumors. This fits with the observation that EBV M81, which was isolated from a Chinese patient with nasopharyngeal carcinoma, induces potent virus production and increases the risk of genetic instability in infected B cells. To find out whether this property extends to viruses found in other parts of the world, we investigated 22 viruses isolated from Western patients. While one-third of the viruses hardly replicated, the remaining viruses showed variable levels of replication, with three isolates replicating at levels close to that of M81 in B cells. We cloned one strongly replicating virus into a bacterial artificial chromosome (BAC); the resulting recombinant virus (MSHJ) retained the properties of its nonrecombinant counterpart and showed similarities to M81, undergoing lytic replication in vitro and in vivo after 3 weeks of latency. In contrast, B cells infected with the nonreplicating Western B95-8 virus showed early but abortive replication accompanied by cytoplasmic BZLF1 expression. Sequencing confirmed that rMSHJ is a Western virus, being genetically much closer to B95-8 than to M81. Spontaneous replication in rM81- and rMSHJ-infected B cells was dependent on phosphorylated Btk and was inhibited by exposure to ibrutinib, opening the way to clinical intervention in patients with abnormal EBV replication. As rMSHJ contains the complete EBV genome and induces lytic replication in infected B cells, it is ideal to perform genetic analyses of all viral functions in Western strains and their associated diseases.IMPORTANCE The Epstein-Barr virus (EBV) infects the majority of the world population but causes different diseases in different countries. Evidence that lytic replication, the process that leads to new virus progeny, is linked to cancer development is accumulating. Indeed, viruses such as M81 that were isolated from Far Eastern nasopharyngeal carcinomas replicate strongly in B cells. We show here that some viruses isolated from Western patients, including the MSHJ strain, share this property. Moreover, replication of both M81 and of MSHJ was sensitive to ibrutinib, a commonly used drug, thereby opening an opportunity for therapeutic intervention. Sequencing of MSHJ showed that this virus is quite distant from M81 and is much closer to nonreplicating Western viruses. We conclude that Western EBV strains are heterogeneous, with some viruses being able to replicate more strongly and therefore being potentially more pathogenic than others, and that the virus sequence information alone cannot predict this property.


Subject(s)
B-Lymphocytes/virology , Epstein-Barr Virus Infections/virology , Herpesvirus 4, Human/genetics , Virus Replication/physiology , Animals , B-Lymphocytes/pathology , Cell Line , Cloning, Molecular , DNA, Viral , Disease Models, Animal , Genome, Viral , HEK293 Cells , Herpesvirus 4, Human/growth & development , Herpesvirus 4, Human/isolation & purification , Humans , Nasopharyngeal Neoplasms/virology , Trans-Activators/genetics
16.
Arch Virol ; 165(2): 313-320, 2020 Feb.
Article in English | MEDLINE | ID: mdl-31813023

ABSTRACT

Epstein-Barr virus (EBV) is a ubiquitous human herpesvirus that transforms primary B lymphocytes, yielding lymphoblastoid cell lines (LCLs). EBV-encoded nuclear antigen 2 (EBNA2) and EBV-encoded nuclear antigen leader protein (EBNALP) are the first viral products expressed after EBV infection of primary B lymphocytes and are essential for EBV-induced B-lymphocyte growth transformation. EBNA2 functions as a transcriptional activator of viral and cellular genes, with EBNALP as a coactivator for EBNA2-mediated transcriptional activation. We previously reported that mutant EBNALP with a C-terminal 10-amino-acid truncation loses the ability to coactivate, and has a dominant-negative effect on wild-type-EBNALP-mediated coactivation. However, the functional relevance of EBNALP in maintenance of LCL cell growth has not been investigated. To address this, we have constructed LCL-derived cell clones in which this dominant-negative form of EBNALP (DNLP) is conditionally expressed by the Cre-loxP system. We used these cells to evaluate the effect of DNLP expression on EBV-induced cell proliferation. After drug treatment, the DNLP-expressing LCL clones showed reduced cell proliferation and viability. These results indicate that EBNALP is critical for maintaining LCL growth and EBV-induced cell proliferation.


Subject(s)
B-Lymphocytes/virology , Cell Proliferation , Cell Transformation, Viral , Epstein-Barr Virus Nuclear Antigens/metabolism , Gene Expression , Herpesvirus 4, Human/growth & development , Mutant Proteins/metabolism , Viral Proteins/metabolism , Cell Line , Epstein-Barr Virus Nuclear Antigens/genetics , Humans , Mutant Proteins/genetics , Viral Proteins/genetics
17.
Am J Dermatopathol ; 41(12): 927-930, 2019 Dec.
Article in English | MEDLINE | ID: mdl-31599749

ABSTRACT

Cutaneous manifestations of Epstein-Barr virus (EBV)-driven B-cell lymphoid proliferations occur rarely as a result of severe immunodeficiency. To date, only a few cases of extranodal EBV-associated B-cell lymphomas arising in patients with angioimmunoblastic T-cell lymphoma (AITL) have been reported, and less common is a cutaneous presentation. AITL is a rare aggressive tumor that carries a poor prognosis and prompt diagnosis, and recognition of EBV-associated diffuse B-cell lymphoma is essential in these patients to instigate the correct treatment.


Subject(s)
Epstein-Barr Virus Infections/virology , Herpesvirus 4, Human/growth & development , Immunoblastic Lymphadenopathy/immunology , Immunocompromised Host , Lymphoma, Large B-Cell, Diffuse/virology , Lymphoma, T-Cell/immunology , Skin Neoplasms/immunology , Skin Neoplasms/virology , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Cyclophosphamide/adverse effects , Doxorubicin/adverse effects , Epstein-Barr Virus Infections/immunology , Herpesvirus 4, Human/immunology , Humans , Immunoblastic Lymphadenopathy/drug therapy , Immunoblastic Lymphadenopathy/pathology , Lymphoma, Large B-Cell, Diffuse/drug therapy , Lymphoma, Large B-Cell, Diffuse/immunology , Lymphoma, Large B-Cell, Diffuse/pathology , Lymphoma, T-Cell/drug therapy , Lymphoma, T-Cell/pathology , Male , Middle Aged , Prednisone/adverse effects , Skin Neoplasms/drug therapy , Skin Neoplasms/pathology , Treatment Outcome , Vincristine/adverse effects , Virus Replication
18.
PLoS One ; 14(9): e0222847, 2019.
Article in English | MEDLINE | ID: mdl-31568538

ABSTRACT

The Epstein-Barr virus (EBV) induces B-cell proliferation with high efficiency through expression of latent proteins and microRNAs. This process takes place in vivo soon after infection, presumably to expand the virus reservoir, but can also induce pathologies, e.g. an infectious mononucleosis (IM) syndrome after primary infection or a B-cell lymphoproliferation in immunosuppressed individuals. In this paper, we investigated the growth characteristics of EBV-infected B-cells isolated from transplant recipients or patients with IM. We found that these cells grew and withstood apoptosis at highly variable rates, suggesting that the expansion rate of the infected B-cells widely varies between individuals, thereby influencing the size of the B-cell reservoir and the ability to form tumors in infected individuals. All viruses investigated were type 1 and genetically close to western strains. EBV-infected B-cells expressed the transforming EBV latent genes and microRNAs (miRNAs) at variable levels. We found that the B-cell growth rates positively correlated with the BHRF1 miRNA levels. Comparative studies showed that infected B-cells derived from transplant recipients with iEBVL on average expressed higher levels of EBV miR-BHRF1 miRNAs and grew more rapidly than B-cells from IM patients, suggesting infection by more transforming viruses. Altogether, these findings suggest that EBV infection has a highly variable impact on the B-cell compartment that probably reflects the genetic diversity of both the virus and the host. It also demonstrates the unexpected finding that B-cells from different individuals can grow at different speed under the influence of the same virus infection.


Subject(s)
B-Lymphocytes/virology , Gene Expression Regulation, Viral , Genes, Viral , Herpesvirus 4, Human/genetics , Immunocompromised Host , Infectious Mononucleosis/virology , MicroRNAs/genetics , Adult , Aged , B-Lymphocytes/immunology , B-Lymphocytes/pathology , Cell Line, Transformed , Cell Proliferation , Female , Hematopoietic Stem Cell Transplantation , Herpesvirus 4, Human/growth & development , Herpesvirus 4, Human/metabolism , Host-Pathogen Interactions/genetics , Host-Pathogen Interactions/immunology , Humans , Infectious Mononucleosis/immunology , Infectious Mononucleosis/pathology , Kidney Transplantation , Male , MicroRNAs/immunology , Middle Aged , Primary Cell Culture , Viral Proteins/genetics , Viral Proteins/immunology
19.
Nat Rev Microbiol ; 17(11): 691-700, 2019 11.
Article in English | MEDLINE | ID: mdl-31477887

ABSTRACT

Epstein-Barr virus (EBV) was the first tumour virus identified in humans. The virus is primarily associated with lymphomas and epithelial cell cancers. These tumours express latent EBV antigens and the oncogenic potential of individual latent EBV proteins has been extensively explored. Nevertheless, it was presumed that the pro-proliferative and anti-apoptotic functions of these oncogenes allow the virus to persist in humans; however, recent evidence suggests that cellular transformation is not required for virus maintenance. Vice versa, lytic EBV replication was assumed to destroy latently infected cells and thereby inhibit tumorigenesis, but at least the initiation of the lytic cycle has now been shown to support EBV-driven malignancies. In addition to these changes in the roles of latent and lytic EBV proteins during tumorigenesis, the function of non-coding RNAs has become clearer, suggesting that they might mainly mediate immune escape rather than cellular transformation. In this Review, these recent findings will be discussed with respect to the role of EBV-encoded oncogenes in viral persistence and the contributions of lytic replication as well as non-coding RNAs in virus-driven tumour formation. Accordingly, early lytic EBV antigens and attenuated viruses without oncogenes and microRNAs could be harnessed for immunotherapies and vaccination.


Subject(s)
Herpesvirus 4, Human/growth & development , Viral Proteins/metabolism , Virus Latency , Virus Replication , Carcinogenesis , Cell Survival , Cell Transformation, Neoplastic , Epithelial Cells/virology , Humans , Lymphocytes/virology
20.
Nat Microbiol ; 4(12): 2475-2486, 2019 12.
Article in English | MEDLINE | ID: mdl-31501540

ABSTRACT

The Epstein-Barr virus M81 strain, isolated from a nasopharyngeal carcinoma, induces potent spontaneous virus production in infected B cells. We found that the M81 non-coding Epstein-Barr-encoded RNA EBER2, which carries polymorphisms that are mainly restricted to viruses found in endemic nasopharyngeal carcinomas, markedly stimulated this process. M81 EBER2 increased CXCL8 expression, and this chemokine enhanced spontaneous lytic replication levels in M81-infected B cells. Both events resulted from the endocytosis of extracellular vesicles containing EBER2 that were generated by neighbouring M81-infected B cells, thereby generating a paracrine loop. These effects were strictly dependent on a functional Toll-like receptor 7 (TLR7), a sensor of single-stranded RNA located in the endosome of these cells. These unique properties of M81 EBER2 could be ascribed to its unusually high expression level and to the ability of its single-stranded region to activate TLR7; both of these properties were dependent on M81-specific polymorphisms. Thus, M81 induced chronic inflammation in its target cells and this resulted in increased virus production. These observations provide a mechanistic molecular link between M81 virus replication-a central viral function and a cancer risk factor-and the production of a chemokine involved in inflammation and carcinogenesis.


Subject(s)
Epstein-Barr Virus Infections/virology , Herpesvirus 4, Human/growth & development , Herpesvirus 4, Human/genetics , Nasopharyngeal Carcinoma/metabolism , Nasopharyngeal Carcinoma/virology , Nasopharyngeal Neoplasms/virology , RNA, Untranslated/genetics , Virus Replication , B-Lymphocytes/virology , Chemokines/metabolism , Epstein-Barr Virus Infections/immunology , HEK293 Cells , Host-Pathogen Interactions , Humans , Interleukin-8/metabolism , Nasopharyngeal Carcinoma/genetics , Nasopharyngeal Neoplasms/genetics , Nasopharyngeal Neoplasms/metabolism , Oncogenic Viruses , RNA, Viral , Toll-Like Receptor 7/metabolism , Virus Cultivation
SELECTION OF CITATIONS
SEARCH DETAIL
...