Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 127
Filtrar
1.
EMBO Rep ; 25(2): 725-744, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-38177923

RESUMO

Viral infection often trigger an ATM serine/threonine kinase (ATM)-dependent DNA damage response in host cells that suppresses viral replication. Viruses evolved different strategies to counteract this antiviral surveillance system. Here, we report that human herpesvirus 6B (HHV-6B) infection causes genomic instability by suppressing ATM signaling in host cells. Expression of immediate-early protein 1 (IE1) phenocopies this phenotype and blocks homology-directed double-strand break repair. Mechanistically, IE1 interacts with NBS1, and inhibits ATM signaling through two distinct domains. HHV-6B seems to efficiently inhibit ATM signaling as further depletion of either NBS1 or ATM do not significantly boost viral replication in infected cells. Interestingly, viral integration of HHV-6B into the host's telomeres is not strictly dependent on NBS1, challenging current models where integration occurs through homology-directed repair. Given that spontaneous IE1 expression has been detected in cells of subjects with inherited chromosomally-integrated form of HHV-6B (iciHHV-6B), a condition associated with several health conditions, our results raise the possibility of a link between genomic instability and the development of iciHHV-6-associated diseases.


Assuntos
Herpesvirus Humano 6 , Proteínas Imediatamente Precoces , Infecções por Roseolovirus , Humanos , Herpesvirus Humano 6/genética , Herpesvirus Humano 6/metabolismo , Infecções por Roseolovirus/genética , Proteínas Imediatamente Precoces/genética , Proteínas Imediatamente Precoces/metabolismo , Integração Viral , Instabilidade Genômica , Proteínas Mutadas de Ataxia Telangiectasia/genética , Proteínas Mutadas de Ataxia Telangiectasia/metabolismo
2.
J Virol ; 97(9): e0071823, 2023 09 28.
Artigo em Inglês | MEDLINE | ID: mdl-37671864

RESUMO

Nascent nucleocapsids of herpesviruses acquire a primary envelope during their nuclear export by budding through the inner nuclear membrane into the perinuclear space between the inner and outer nuclear membranes. This process is mediated by a conserved viral heterodimeric complex designated the nuclear egress complex, which consists of the nuclear matrix protein and the nuclear membrane protein. In addition to its essential roles during nuclear egress, the nuclear matrix protein has been shown to interact with intracellular signaling pathway molecules including NF-κB and IFN-ß to affect viral or cellular gene expression. The human herpesvirus 6A (HHV-6A) U37 gene encodes a nuclear matrix protein, the role of which has not been analyzed. Here, we show that HHV-6A U37 activates the heat shock element promoter and induces the accumulation of the molecular chaperone Hsp90. Mechanistically, HHV-6A U37 interacts with heat shock transcription factor 1 (HSF1) and induces its phosphorylation at Ser-326. We report that pharmacological inhibition of HSF1, Hsp70, or Hsp90 decreases viral protein accumulation and viral replication. Taken together, our results lead us to propose a model in which HHV-6A U37 activates the heat shock response to support viral gene expression and replication. IMPORTANCE Human herpesvirus 6A (HHV-6A) is a dsDNA virus belonging to the Roseolovirus genus within the Betaherpesvirinae subfamily. It is frequently found in patients with neuroinflammatory disease, although its pathogenetic role, if any, awaits elucidation. The heat shock response is important for cell survival under stressful conditions that disrupt homeostasis. Our results indicate that HHV-6A U37 activates the heat shock element promoter and leads to the accumulation of heat shock proteins. Next, we show that the heat shock response is important for viral replication. Overall, our findings provide new insights into the function of HHV-6A U37 in host cell signaling and identify potential cellular targets involved in HHV-6A pathogenesis and replication.


Assuntos
Fatores de Transcrição de Choque Térmico , Resposta ao Choque Térmico , Herpesvirus Humano 6 , Proteínas da Matriz Viral , Humanos , Fatores de Transcrição de Choque Térmico/metabolismo , Resposta ao Choque Térmico/genética , Herpesvirus Humano 6/metabolismo , Herpesvirus Humano 6/patogenicidade , Proteínas da Matriz Viral/metabolismo , Proteínas de Choque Térmico HSP90/metabolismo , Proteínas de Choque Térmico HSP70/metabolismo , Regiões Promotoras Genéticas , Replicação Viral , Fosforilação , Regulação Viral da Expressão Gênica , Transdução de Sinais
3.
Glycoconj J ; 39(6): 711-724, 2022 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-36227524

RESUMO

The Human Betaherpesviruses HHV-5 and HHV-6 are quite inimical in immunocompromised hosts individually. A co-infection of both has been surmised to be far more disastrous. This can be attributed to a synergetic effect of their combined pathologies. While there have been attempts to develop a vaccine against each virus, no efforts were made to contrive an effective prophylaxis for the highly detrimental co-infection. In this study, an ensemble of viral envelope glycoproteins from both the viruses was utilized to design a multi-epitope vaccine using immunoinformatics tools. A collection of bacterial protein toll-like receptor agonists (BPTAs) was screened to identify a highly immunogenic adjuvant for the vaccine construct. The constructed vaccine was analysed using an array of methodologies ranging from World population coverage analysis to Immune simulation, whose results indicate high vaccine efficacy and stability. Furthermore, codon optimization and in silico cloning analysis were performed to check for efficient expression in a bacterial system. Collectively, these findings demonstrate the potential of the constructed vaccine to elicit an immune response against HHV-5 and HHV-6, thus supporting the viability of in vitro and in vivo studies.


Assuntos
Coinfecção , Herpesvirus Humano 6 , Vacinas , Humanos , Herpesvirus Humano 6/genética , Herpesvirus Humano 6/metabolismo , Citomegalovirus/metabolismo , Epitopos de Linfócito T , Proteínas do Envelope Viral/genética , Proteínas do Envelope Viral/metabolismo , Simulação de Acoplamento Molecular , Vacinas de Subunidades
4.
Mol Cell Neurosci ; 123: 103770, 2022 12.
Artigo em Inglês | MEDLINE | ID: mdl-36055520

RESUMO

Many neurodegenerative diseases have a multifactorial etiology and variable course of progression that cannot be explained by current models. Neurotropic viruses have long been suggested to play a role in these diseases, although their exact contributions remain unclear. Human herpesvirus 6A (HHV-6A) is one of the most common viruses detected in the adult brain, and has been clinically associated with multiple sclerosis (MS), and, more recently, Alzheimer's disease (AD). HHV-6A is a ubiquitous viral pathogen capable of infecting glia and neurons. Primary infection in childhood is followed by the induction of latency, characterized by expression of the U94A viral transcript in the absence of viral replication. Here we examine the effects of U94A on cells of the central nervous system. We found that U94A expression inhibits the migration and impairs cytoplasmic maturation of human oligodendrocyte precursor cells (OPCs) without affecting their viability, a phenotype that may contribute to the failure of remyelination seen in many patients with MS. A subsequent proteomics analysis of U94A expression OPCs revealed altered expression of genes involved in tubulin associated cytoskeletal regulation. As HHV-6A seems to significantly be associated with early AD pathology, we extended our initially analysis of the impact of U94A on human derived neurons. We found that U94A expression inhibits neurite outgrowth of primary human cortical neurons and impairs synapse maturation. Based on these data we suggest that U94A expression by latent HHV-6A in glial cells and neurons renders them susceptible to dysfunction and degeneration. Therefore, latent viral infections of the brain represent a unique pathological risk factor that may contribute to disease processes.


Assuntos
Herpesvirus Humano 6 , Esclerose Múltipla , Células Precursoras de Oligodendrócitos , Humanos , Herpesvirus Humano 6/genética , Herpesvirus Humano 6/metabolismo , Sistema Nervoso Central , Neuroglia
5.
Front Immunol ; 13: 864898, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35444636

RESUMO

Human roseolovirus U20 and U21 are type I membrane glycoproteins that have been implicated in immune evasion by interfering with recognition of classical and non-classical MHC proteins. U20 and U21 are predicted to be type I glycoproteins with extracytosolic immunoglobulin-like domains, but detailed structural information is lacking. AlphaFold and RoseTTAfold are next generation machine-learning-based prediction engines that recently have revolutionized the field of computational three-dimensional protein structure prediction. Here, we review the structural biology of viral immunoevasins and the current status of computational structure prediction algorithms. We use these computational tools to generate structural models for U20 and U21 proteins, which are predicted to adopt MHC-Ia-like folds with closed MHC platforms and immunoglobulin-like domains. We evaluate these structural models and place them within current understanding of the structural basis for viral immune evasion of T cell and natural killer cell recognition.


Assuntos
Herpesvirus Humano 6 , Herpesvirus Humano 7 , Infecções por Roseolovirus , Herpesvirus Humano 6/metabolismo , Herpesvirus Humano 7/metabolismo , Humanos , Modelos Estruturais , Proteínas Virais/metabolismo
6.
Viruses ; 14(1)2022 01 16.
Artigo em Inglês | MEDLINE | ID: mdl-35062364

RESUMO

Human herpesvirus 6B (HHV-6B) is a T-lymphotropic virus and the etiological agent of exanthem subitum. HHV-6B is present in a latent or persistent form after primary infection and is produced in the salivary glands or transmitted to this organ. Infected individuals continue to secrete the virus in their saliva, which is thus considered a source for virus transmission. HHV-6B primarily propagates in T cells because its entry receptor, CD134, is mainly expressed by activated T cells. The virus then spreads to the host's organs, including the salivary glands, nervous system, and liver. However, CD134 expression is not detected in these organs. Therefore, HHV-6B may be entering cells via a currently unidentified cell surface molecule, but the mechanisms for this have not yet been investigated. In this study, we investigated a CD134-independent virus entry mechanism in the parotid-derived cell line HSY. First, we confirmed viral infection in CD134-membrane unanchored HSY cells. We then determined that nectin cell adhesion molecule 2 (nectin-2) mediated virus entry and that HHV-6B-insensitive T-cells transduced with nectin-2 were transformed into virus-permissive cells. We also found that virus entry was significantly reduced in nectin-2 knockout parotid-derived cells. Furthermore, we showed that HHV-6B glycoprotein B (gB) interacted with the nectin-2 V-set domain. The results suggest that nectin-2 acts as an HHV-6B entry-mediated protein.


Assuntos
Herpesvirus Humano 6/metabolismo , Nectinas/genética , Nectinas/metabolismo , Proteínas do Envelope Viral/metabolismo , Ligação Viral , Internalização do Vírus , Linhagem Celular , Técnicas de Inativação de Genes , Herpesvirus Humano 6/classificação , Herpesvirus Humano 6/genética , Humanos
7.
J Virol ; 96(4): e0168821, 2022 02 23.
Artigo em Inglês | MEDLINE | ID: mdl-34878807

RESUMO

Human herpesvirus 6 (HHV-6) belongs to the betaherpesvirus subfamily and is divided into two distinct species, HHV-6A and HHV-6B. HHV-6 can infect nerve cells and is associated with a variety of nervous system diseases. Recently, the association of HHV-6A infection with Alzheimer's disease (AD) has been suggested. The main pathological phenomena of AD are the accumulation of ß-amyloid (Aß), neurofibrillary tangles, and neuroinflammation; however, the specific molecular mechanism of pathogenesis of AD is not completely clear. In this study, we focused on the effect of HHV-6A U4 gene function on Aß expression. Coexpression of HHV-6A U4 with amyloid precursor protein (APP) resulted in inhibition of ubiquitin-mediated proteasomal degradation of APP. Consequently, accumulation of ß-amyloid peptide (Aß), insoluble neurofibrillary tangles, and loss of neural cells may occur. Immunoprecipitation coupled with mass spectrometry (IP-MS) showed that HHV-6A U4 protein interacts with E3 ubiquitin ligase composed of DDB1 and cullin 4B, which is also responsible for APP degradation. We hypothesize that HHV-6A U4 protein competes with APP for binding to E3 ubiquitin ligase, resulting in the inhibition of APP ubiquitin modification and clearance. Finally, this leads to an increase in APP expression and Aß deposition, which are the hallmarks of AD. These findings provide novel evidence for the etiological hypothesis of AD, which can contribute to the further analysis of the role of HHV-6A in AD. IMPORTANCE The association of HHV-6A infection with Alzheimer's disease has attracted increasing attention, although its role and molecular mechanism remain to be established. Our results here indicate that HHV-6A U4 inhibits amyloid precursor protein (APP) degradation. U4 protein interacts with CRLs (cullin-RING E3 ubiquitin-protein ligases), which is also responsible for APP degradation. We propose a model in which U4 competitively binds to CRLs with APP, resulting in APP accumulation and Aß generation. Our findings provide new insights into the etiological hypothesis of HHV-6A in AD that can help further analyses.


Assuntos
Precursor de Proteína beta-Amiloide/metabolismo , Herpesvirus Humano 6/metabolismo , Complexo de Endopeptidases do Proteassoma/metabolismo , Proteínas Virais/metabolismo , Peptídeos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/genética , Linhagem Celular , Proteínas Culina/metabolismo , Proteínas de Ligação a DNA/metabolismo , Expressão Gênica , Herpesvirus Humano 6/genética , Humanos , Ligação Proteica , Ubiquitina/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Ubiquitinação , Proteínas Virais/genética
9.
J Virol ; 95(23): e0126921, 2021 11 09.
Artigo em Inglês | MEDLINE | ID: mdl-34549982

RESUMO

Viral infection induces host cells to mount a variety of immune responses, which may either limit viral propagation or create conditions conducive to virus replication in some instances. In this regard, activation of the NF-κB transcription factor is known to modulate virus replication. Human herpesvirus 6A (HHV-6A), which belongs to the Betaherpesvirinae subfamily, is frequently found in patients with neuroinflammatory diseases, although its role in disease pathogenesis has not been elucidated. In this study, we found that the HHV-6A-encoded U14 protein activates NF-κB signaling following interaction with the NF-κB complex protein, p65. Through induction of nuclear translocation of p65, U14 increases the expression of interleukin-6 (IL-6), IL-8, and monocyte chemoattractant protein 1 transcripts. We also demonstrated that activation of NF-κB signaling is important for HHV-6A replication, since inhibition of this pathway reduced virus protein accumulation and viral genome copy number. Taken together, our results suggest that HHV-6A infection activates the NF-κB pathway and promotes viral gene expression via late gene products, including U14. IMPORTANCE Human herpesvirus 6A (HHV-6A) is frequently found in patients with neuro-inflammation, although its role in the pathogenesis of this disease has not been elucidated. Most viral infections activate the NF-κB pathway, which causes the transactivation of various genes, including those encoding proinflammatory cytokines. Our results indicate that HHV-6A U14 activates the NF-κB pathway, leading to upregulation of proinflammatory cytokines. We also found that activation of the NF-κB transcription factor is important for efficient viral replication. This study provides new insight into HHV-6A U14 function in host cell signaling and identifies potential cellular targets involved in HHV-6A pathogenesis and replication.


Assuntos
Herpesvirus Humano 6/genética , Herpesvirus Humano 6/metabolismo , NF-kappa B/metabolismo , Infecções por Roseolovirus/imunologia , Transdução de Sinais/imunologia , Proteínas Virais/imunologia , Linhagem Celular , Regulação da Expressão Gênica , Genes Virais , Genoma Viral , Humanos , NF-kappa B/genética , Doenças Neuroinflamatórias , Receptor EphB2 , Proteínas Virais/genética , Replicação Viral
10.
J Neuroimmunol ; 358: 577636, 2021 09 15.
Artigo em Inglês | MEDLINE | ID: mdl-34174587

RESUMO

Herpesviruses like Epstein-Barr virus, human herpesvirus (HHV)-6, HHV-1, VZV, and human endogenous retroviruses, have an age-old clinical association with multiple sclerosis (MS). MS is an autoimmune disease of the nervous system wherein the myelin sheath deteriorates. The most popular mode of virus mediated immune system manipulation is molecular mimicry. Numerous herpesvirus antigens are similar to myelin proteins. Other mechanisms described here include the activity of cytokines and autoantibodies produced by the autoreactive T and B cells, respectively, viral déjà vu, epitope spreading, CD46 receptor engagement, impaired remyelination etc. Overall, this review addresses the host-parasite association of viruses with MS.


Assuntos
Autoanticorpos/imunologia , Herpesviridae/imunologia , Esclerose Múltipla/diagnóstico , Esclerose Múltipla/imunologia , Autoanticorpos/sangue , Herpesviridae/metabolismo , Herpesvirus Humano 1/imunologia , Herpesvirus Humano 1/metabolismo , Herpesvirus Humano 3/imunologia , Herpesvirus Humano 3/metabolismo , Herpesvirus Humano 4/imunologia , Herpesvirus Humano 4/metabolismo , Herpesvirus Humano 6/imunologia , Herpesvirus Humano 6/metabolismo , Humanos , Esclerose Múltipla/sangue
11.
J Virol ; 95(14): e0162820, 2021 06 24.
Artigo em Inglês | MEDLINE | ID: mdl-33952641

RESUMO

Like all herpesviruses, the roseoloviruses (HHV6A, -6B, and -7) establish lifelong infection within their host, requiring these viruses to evade host antiviral responses. One common host-evasion strategy is the downregulation of host-encoded, surface-expressed glycoproteins. Roseoloviruses have been shown to evade the host immune response by downregulating NK-activating ligands, class I MHC, and the TCR/CD3 complex. To more globally identify glycoproteins that are differentially expressed on the surface of HHV6A-infected cells, we performed cell surface capture of N-linked glycoproteins present on the surface of T cells infected with HHV6A, and compared these to proteins present on the surface of uninfected T cells. We found that the protein tyrosine phosphatase CD45 is downregulated in T cells infected with HHV6A. We also demonstrated that CD45 is similarly downregulated in cells infected with HHV7. CD45 is essential for signaling through the T cell receptor and, as such, is necessary for developing a fully functional immune response. Interestingly, the closely related betaherpesviruses human cytomegalovirus (HCMV) and murine cytomegalovirus (MCMV) have also separately evolved unique mechanisms to target CD45. While HCMV and MCMV target CD45 signaling and trafficking, HHV6A acts to downregulate CD45 transcripts. IMPORTANCE Human herpesviruses-6 and -7 infect essentially 100% of the world's population before the age of 5 and then remain latent or persistent in their host throughout life. As such, these viruses are among the most pervasive and stealthy of all viruses. Host immune cells rely on the presence of surface-expressed proteins to identify and target virus-infected cells. Here, we investigated the changes that occur to proteins expressed on the cell surface of T cells after infection with human herpesvirus-6A. We discovered that HHV-6A infection results in a reduction of CD45 on the surface of infected T cells and impaired activation in response to T cell receptor stimulation.


Assuntos
Regulação Enzimológica da Expressão Gênica , Regulação Viral da Expressão Gênica , Herpesvirus Humano 6/genética , Herpesvirus Humano 7/genética , Antígenos Comuns de Leucócito/genética , Linfócitos T/virologia , Linhagem Celular , Regulação para Baixo , Células HEK293 , Herpesvirus Humano 6/metabolismo , Herpesvirus Humano 7/metabolismo , Humanos , Estabilidade Proteica , Receptores de Antígenos de Linfócitos T/metabolismo , Linfócitos T/imunologia , Linfócitos T/metabolismo
12.
Am J Reprod Immunol ; 85(4): e13313, 2021 04.
Artigo em Inglês | MEDLINE | ID: mdl-32710571

RESUMO

Molecular diagnostics is a rapidly growing branch of the clinical laboratory and has accelerated the advance of personalized medicine in the fields of pharmacogenomics, pharmacogenetics, and nutrigenomics. The versatility of molecular biology allows it to be effective in several medical fields that include reproduction, immunogenetics, and virology. Implementation of molecular and sequencing technology in reproductive medicine can add another layer of understanding to better define the causes behind infertility and recurrent reproductive loss. In the following, we examine current molecular methods for probing factors behind reproductive pregnancy loss including reverse transcription polymerase chain reaction and next generation sequencing (NGS). We review several current and potential genetic (DNA) and transcriptional (RNA)-based parameters in women with infertility that can be significant in diagnosis and treatment. These molecular factors can be inferred either from genomic DNA or RNA locally within the endometrium. Furthermore, we consider infection-based abnormalities such as human herpesvirus-6 and severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2). Finally, we present future directions as well as data demonstrating the potential role of human endogenous retroviruses in pregnancy loss. We hope these discussions will assist the clinician in delineating some of the intricate molecular factors that can contribute to infertility and recurrent reproductive failures.


Assuntos
Aborto Espontâneo , COVID-19 , Regulação da Expressão Gênica , Herpesvirus Humano 6 , Infertilidade Feminina , Infecções por Roseolovirus , SARS-CoV-2 , Aborto Espontâneo/genética , Aborto Espontâneo/metabolismo , Aborto Espontâneo/virologia , COVID-19/genética , COVID-19/metabolismo , Endométrio/metabolismo , Endométrio/virologia , Feminino , Herpesvirus Humano 6/genética , Herpesvirus Humano 6/metabolismo , Humanos , Infertilidade Feminina/genética , Infertilidade Feminina/metabolismo , Infecções por Roseolovirus/metabolismo , SARS-CoV-2/genética , SARS-CoV-2/metabolismo
13.
Microbiol Immunol ; 64(11): 747-761, 2020 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-32910457

RESUMO

The human herpesvirus 6B (HHV-6B) U79/80 gene belongs to the early gene class and appears as early as 3 hr postinfection. It is one of the most abundantly expressed transcripts and a useful diagnostic marker for viral reactivation. However, the expression mechanisms of the U79/80 gene remain unclear. To identify the viral factor(s) that activates the U79/80 promoter along with other HHV-6B core early gene promoters, p41, DNA polymerase, and U41, we examined the activities of U79/80 and other early gene promoters. In HHV-6B-infected MT-4 cells, U79/80 promoter activity was the highest among early gene promoters. In addition, we identified that HHV-6B immediate-early (IE)2B protein is one of the viral proteins involved in the activation of the U79/80 and other early gene promoters. Although the IE2B could independently activate these early gene promoters, the presence of IE1B significantly augmented the activities of early gene promoters. We also found that IE2B bound three human cytomegalovirus IE2-binding consensus, cis repression signal (CRS), within the U79/80 promoter. Moreover, the U79/80 promoter was activated by cellular factors, which are highly expressed in MT-4 cells, instead of HeLa cells because it was upregulated by mock infection and in the absence of IE2B. These results suggested that the activation mechanism of the U79/80 gene differs from other HHV-6B core early genes, apparently supporting its rapid and abundant expression. Therefore, the U79/80 early gene is an actually suitable biomarker of HHV-6B reactivation.


Assuntos
Herpesvirus Humano 6/genética , Herpesvirus Humano 6/metabolismo , Proteínas Imediatamente Precoces/genética , Proteínas Imediatamente Precoces/metabolismo , Regiões Promotoras Genéticas , Proteínas Virais/genética , Proteínas Virais/metabolismo , Citomegalovirus/genética , DNA Polimerase Dirigida por DNA , Regulação Viral da Expressão Gênica , Células HeLa , Humanos , Transcrição Gênica , Ativação Transcricional
14.
PLoS Pathog ; 16(7): e1008648, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-32678833

RESUMO

A unique glycoprotein is expressed on the virus envelope of human herpesvirus 6B (HHV-6B): the complex gH/gL/gQ1/gQ2 (hereafter referred to as the HHV-6B tetramer). This tetramer recognizes a host receptor expressed on activated T cells: human CD134 (hCD134). This interaction is essential for HHV-6B entry into the susceptible cells and is a determinant for HHV-6B cell tropism. The structural mechanisms underlying this unique interaction were unknown. Herein we solved the interactions between the HHV-6B tetramer and the receptor by using their neutralizing antibodies in molecular and structural analyses. A surface plasmon resonance analysis revealed fast dissociation/association between the tetramer and hCD134, although the affinity was high (KD = 18 nM) and comparable to those for the neutralizing antibodies (anti-gQ1: 17 nM, anti-gH: 2.7 nM). A competition assay demonstrated that the anti-gQ1 antibody competed with hCD134 in the HHV-6B tetramer binding whereas the anti-gH antibody did not, indicating the direct interaction of gQ1 and hCD134. A single-particle analysis by negative-staining electron microscopy revealed the tetramer's elongated shape with a gH/gL part and extra density corresponding to gQ1/gQ2. The anti-gQ1 antibody bound to the tip of the extra density, and anti-gH antibody bound to the putative gH/gL part. These results highlight the interaction of gQ1/gQ2 in the HHV-6B tetramer with hCD134, and they demonstrate common features among viral ligands of the betaherpesvirus subfamily from a macroscopic viewpoint.


Assuntos
Herpesvirus Humano 6/metabolismo , Receptores OX40/metabolismo , Infecções por Roseolovirus/metabolismo , Proteínas do Envelope Viral/metabolismo , Humanos
15.
Viruses ; 12(6)2020 06 26.
Artigo em Inglês | MEDLINE | ID: mdl-32604892

RESUMO

The aim of this study was to investigate the role of human herpesvirus-6 (HHV-6) in autoimmune thyroiditis (AIT) development. We examined the possible involvement of HHV-6 gene expression encoding immunomodulating proteins U12 and U51 in AIT development and their role in the modulation of chemokine signaling. One hundred patients with autoimmune thyroiditis following thyroidectomy were enrolled in this study. Nested polymerase chain reaction (nPCR) was used to detect the HHV-6 sequence in DNA samples. Reverse transcription PCR (RT-PCR) with three different HHV-6 gene targets (U79/80, U51 and U12) was to detect active infection markers. HHV-6 load was identified using a commercial real-time PCR kit. Immunohistochemistry was performed to investigate the expression of the HHV-6 antigen and RANTES (Regulated upon Activation, Normal T Cell Expressed and Secreted) in thyroid gland tissue. Different commercial immunosorbent assay kits were used for the detection of RANTES, IFNγ, IL-6, and TNFα levels in the AIT patient group and controls. We detected 98% presence of the HHV-6 genomic sequence in AIT patients' thyroid gland tissues. Markers of active HHV-6 infection (HHV-6 U79/80, U12 and/or U51 mRNA) were predominant in AIT patients' thyroid tissue samples in comparison with the control group (56% vs. 6%). Evidence from immunofluorescence microscopy showed that HHV-6 can persist in thyrocytes and can interact with RANTES. Visual confirmation of the intense immunofluorescence signal of RANTES detected in thyroid tissues could indicate high expression of this chemokine in the thyroid gland. On the other hand, immunosorbent assays showed very low RANTES levels in AIT patients' peripheral plasma. These results indicate that RANTES level in AIT patients could be influenced by HHV-6 activation, which in turn may aid AIT development.


Assuntos
Quimiocina CCL5/metabolismo , Herpesvirus Humano 6/metabolismo , Infecções por Roseolovirus/patologia , Glândula Tireoide/metabolismo , Tireoidite Autoimune/patologia , Idoso , Feminino , Regulação da Expressão Gênica/genética , Regulação da Expressão Gênica/imunologia , Genoma Viral/genética , Herpesvirus Humano 6/genética , Humanos , Imuno-Histoquímica , Interferon gama/metabolismo , Interleucina-6/metabolismo , Masculino , Pessoa de Meia-Idade , Receptores de Quimiocinas/genética , Receptores Virais/genética , Infecções por Roseolovirus/imunologia , Células Epiteliais da Tireoide/metabolismo , Células Epiteliais da Tireoide/virologia , Fator de Necrose Tumoral alfa/metabolismo
16.
PLoS Pathog ; 16(7): e1008683, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-32658923

RESUMO

Human herpesvirus 6B (HHV-6B) is a betaherpesvirus capable of integrating its genome into the telomeres of host chromosomes. Until now, the cellular and/or viral proteins facilitating HHV-6B integration have remained elusive. Here we show that a cellular protein, the promyelocytic leukemia protein (PML) that forms nuclear bodies (PML-NBs), associates with the HHV-6B immediate early 1 (IE1) protein at telomeres. We report enhanced levels of SUMOylated IE1 in the presence of PML and have identified a putative SUMO Interacting Motif (SIM) within IE1, essential for its nuclear distribution, overall SUMOylation and association with PML to nuclear bodies. Furthermore, using PML knockout cell lines we made the original observation that PML is required for efficient HHV-6B integration into host chromosomes. Taken together, we could demonstrate that PML-NBs are important for IE1 multiSUMOylation and that PML plays an important role in HHV-6B integration into chromosomes, a strategy developed by this virus to maintain its genome in its host over long periods of time.


Assuntos
Herpesvirus Humano 6/metabolismo , Proteínas Imediatamente Precoces/metabolismo , Fosfoproteínas/metabolismo , Proteína da Leucemia Promielocítica/metabolismo , Infecções por Roseolovirus/metabolismo , Telômero/virologia , Linhagem Celular , Herpesvirus Humano 6/genética , Humanos , Infecções por Roseolovirus/genética , Sumoilação , Latência Viral/genética
17.
PLoS Pathog ; 16(6): e1008568, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-32516328

RESUMO

Human herpesvirus 6 (HHV-6) is an important immunosuppressive and immunomodulatory virus worldwide. However, whether and how HHV-6 infection influences the metabolic machinery of the host cell to provide the energy and biosynthetic resources for virus propagation remains unknown. In this study, we identified that HHV-6A infection promotes glucose metabolism in infected T cells, resulting in elevated glycolytic activity with an increase of glucose uptake, glucose consumption and lactate secretion. Furthermore, we explored the mechanisms involved in HHV-6A-mediated glycolytic activation in the infected T cells. We found increased expressions of the key glucose transporters and glycolytic enzymes in HHV-6A-infected T cells. In addition, HHV-6A infection dramatically activated AKT-mTORC1 signaling in the infected T cells and pharmacological inhibition of mTORC1 blocked HHV-6A-mediated glycolytic activation. We also found that direct inhibition of glycolysis by 2-Deoxy-D-glucose (2-DG) or inhibition of mTORC1 activity in HHV-6A-infected T cells effectively reduced HHV-6 DNA replication, protein synthesis and virion production. These results not only reveal the mechanism of how HHV-6 infection affects host cell metabolism, but also suggest that targeting the metabolic pathway could be a new avenue for HHV-6 therapy.


Assuntos
Glicólise , Herpesvirus Humano 6/metabolismo , Infecções por Roseolovirus/metabolismo , Transdução de Sinais , Linfócitos T/metabolismo , Serina-Treonina Quinases TOR/metabolismo , Linhagem Celular , Replicação do DNA/efeitos dos fármacos , DNA Viral/biossíntese , Desoxiglucose/farmacologia , Glucose/metabolismo , Humanos , Ácido Láctico/metabolismo , Alvo Mecanístico do Complexo 1 de Rapamicina/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Infecções por Roseolovirus/tratamento farmacológico , Infecções por Roseolovirus/patologia , Linfócitos T/patologia , Linfócitos T/virologia , Proteínas Virais/biossíntese , Vírion/metabolismo
18.
J Virol ; 94(13)2020 06 16.
Artigo em Inglês | MEDLINE | ID: mdl-32295911

RESUMO

Human herpesviruses 6A and 6B (HHV-6A and HHV-6B, respectively) are two virus species in the betaherpesvirus subfamily that exhibit T cell tropism. CD46 and CD134 are the cellular receptors for HHV-6A and HHV-6B, respectively. Interestingly, the efficiency of HHV-6A/6B entry is different among different types of target cells despite similar receptor expression levels on these cells. Here, we found that the cellular factor gp96 (also known as glucose-regulated protein 94 [GRP94]) is expressed on the cell surface and interacts with viral glycoprotein Q1 (gQ1) during virus entry. gp96 cell surface expression levels are associated with the efficiency of HHV-6A and HHV-6B entry into target cells. Both loss-of-function and gain-of-function experiments indicated that gp96 plays an important role in HHV-6 infection. Our findings provide new insight into the HHV-6 entry process and might suggest novel therapeutic targets for HHV-6 infection.IMPORTANCE Although new clinical importance has been revealed for human herpesviruses 6A (HHV-6A) and 6B, much is still unknown about the life cycles of these viruses in target cells. We identified a novel cellular factor, gp96, that is critical for both HHV-6A and -6B entry into host cells. As gp96 can function as an adjuvant in vaccine development for both infectious agents and cancers, it can be a potential therapeutic target for infection by these two viruses.


Assuntos
Herpesvirus Humano 6/metabolismo , Glicoproteínas de Membrana/metabolismo , Linhagem Celular , Sangue Fetal/metabolismo , Herpesvirus Humano 6/patogenicidade , Humanos , Glicoproteínas de Membrana/genética , Cultura Primária de Células , Ligação Proteica , Infecções por Roseolovirus/virologia , Linfócitos T/virologia , Proteínas do Envelope Viral/metabolismo , Internalização do Vírus
19.
PLoS Pathog ; 16(4): e1008496, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-32320442

RESUMO

Human herpesviruses 6A and 6B (HHV-6A/B) are unique among human herpesviruses in their ability to integrate their genome into host chromosomes. Viral integration occurs at the ends of chromosomes within the host telomeres. The ends of the HHV-6A/B genomes contain telomeric repeats that facilitate the integration process. Here, we report that productive infections are associated with a massive increase in telomeric sequences of viral origin. The majority of the viral telomeric signals can be detected within viral replication compartments (VRC) that contain the viral DNA processivity factor p41 and the viral immediate-early 2 (IE2) protein. Components of the shelterin protein complex present at telomeres, including TRF1 and TRF2 are also recruited to VRC during infection. Biochemical, immunofluorescence coupled with in situ hybridization and chromatin immunoprecipitation demonstrated the binding of TRF2 to the HHV-6A/B telomeric repeats. In addition, approximately 60% of the viral IE2 protein localize at cellular telomeres during infection. Transient knockdown of TRF2 resulted in greatly reduced (13%) localization of IE2 at cellular telomeres (p<0.0001). Lastly, TRF2 knockdown reduced HHV-6A/B integration frequency (p<0.05), while no effect was observed on the infection efficiency. Overall, our study identified that HHV-6A/B IE2 localizes to telomeres during infection and highlight the role of TRF2 in HHV-6A/B infection and chromosomal integration.


Assuntos
Herpesvirus Humano 6/genética , Herpesvirus Humano 6/metabolismo , Proteína 2 de Ligação a Repetições Teloméricas/genética , Integração Viral/genética , Linhagem Celular Tumoral , DNA Viral/genética , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Humanos , Proteínas Imediatamente Precoces/genética , Proteínas Imediatamente Precoces/metabolismo , Infecções por Roseolovirus/genética , Infecções por Roseolovirus/metabolismo , Infecções por Roseolovirus/virologia , Complexo Shelterina , Telômero/genética , Proteínas de Ligação a Telômeros/genética , Proteínas de Ligação a Telômeros/metabolismo , Proteína 2 de Ligação a Repetições Teloméricas/metabolismo , Proteínas Virais/genética , Proteínas Virais/metabolismo , Replicação Viral/genética
20.
Reprod Sci ; 27(3): 779-786, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-32046402

RESUMO

We recently reported that human herpesvirus 6 (HHV-6) infection is frequently present in endometrial tissue of women with unexplained infertility, and that virus infection induces a profound remodulation of miRNA expression in human cells of different origin. Since specific miRNA patterns have been associated with specific pregnancy outcomes, we aimed to analyze the impact of HHV-6A infection on miRNAs expression and trophoblast receptivity in human endometrial cells. To this purpose, a human endometrial cell line (HEC-1A) was infected with HHV-6A and analyzed for alterations in the expression of miRNAs and for permissiveness to the attachment of a human choriocarcinoma trophoblast cell line (JEG-3). The results showed that HHV-6A infection of endometrial cells up-modulates miR22 (26-fold), miR15 (19.5-fold), and miR196-5p (12.1 fold), that are correlated with implant failure, and down-modulates miR18 (11.4 fold), miR101-3p (4.6 fold), miR181-5p (4.9 fold), miR92 (3.3 fold), and miR1207-5p (3.9 fold), characterized by a low expression in preeclampsia. Moreover, HHV-6A-infected endometrial cells infected resulted less permissive to the attachment of trophoblast cells. In conclusion, collected data suggest that HHV-6A infection could modify miRNA expression pattern and control of trophoblast cell adhesion of endometrial cells, undermining a correct trophoblast cell attachment on endometrial cells.


Assuntos
Adesão Celular , Endométrio/virologia , Células Epiteliais/virologia , Herpesvirus Humano 6/metabolismo , MicroRNAs/metabolismo , Infecções por Roseolovirus/metabolismo , Trofoblastos/virologia , Linhagem Celular , Linhagem Celular Tumoral , Coriocarcinoma/metabolismo , Coriocarcinoma/virologia , Endométrio/metabolismo , Células Epiteliais/metabolismo , Feminino , Humanos , Trofoblastos/metabolismo , Neoplasias Uterinas/metabolismo , Neoplasias Uterinas/virologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...