Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 83
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
J Virol ; 96(6): e0180621, 2022 03 23.
Artículo en Inglés | MEDLINE | ID: mdl-35045264

RESUMEN

Human cytomegalovirus (HCMV) modulates numerous cellular pathways to facilitate infection. Iron is essential to many cellular processes and is often incorporated into proteins and enzymes involved in oxidative phosphorylation and DNA synthesis and repair, among others. Despite its prominent role in the cell, little is known about the regulation of iron metabolism during HCMV infection. Herein, we observe modulation of the transferrin receptor (TfR) during infection and a corresponding change in the cellular labile iron pool. TfR and the iron pool are increased early during infection and then return to mock levels at the late stages of infection. We identified the cellular ubiquitin ligase MARCH1 as an important regulator of TfR. MARCH1 plays a proviral role during infection, as its knockdown leads to a decrease in infectious titers. Knockdown of MARCH1 also leads to an increase in ROS, lipid peroxidation, and mitochondrial dysfunction. Inhibiting an early increase in TfR expression during infection also decreases virus production. These findings indicate the importance of tightly regulating iron metabolism during HCMV infection to facilitate efficient virus production. IMPORTANCE Iron is essential for cells, playing important roles in energy generation, DNA replication, and gene expression. During infection, HCMV alters many cellular processes to aid its replication. We found that iron levels are tightly regulated during infection and that dysregulation of iron levels alters the ability to produce infectious virions. We also found that HCMV inactivates many of the cellular safeguards put in place to deal with excess iron. Thus, infected cells become more susceptible to variations in iron levels, which could be exploited as a therapeutic strategy for dealing with HCMV infections.


Asunto(s)
Infecciones por Citomegalovirus , Hierro , Ubiquitina-Proteína Ligasas , Citomegalovirus/fisiología , Infecciones por Citomegalovirus/enzimología , Infecciones por Citomegalovirus/fisiopatología , Humanos , Hierro/metabolismo , Ubiquitina-Proteína Ligasas/genética , Ubiquitina-Proteína Ligasas/metabolismo , Regulación hacia Arriba
2.
PLoS Pathog ; 16(4): e1008426, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-32282833

RESUMEN

Human cytomegalovirus (HCMV) is the most frequent viral cause of congenital defects and can trigger devastating disease in immune-suppressed patients. Cytotoxic lymphocytes (CD8+ T cells and NK cells) control HCMV infection by releasing interferon-γ and five granzymes (GrA, GrB, GrH, GrK, GrM), which are believed to kill infected host cells through cleavage of intracellular death substrates. However, it has recently been demonstrated that the in vivo killing capacity of cytotoxic T cells is limited and multiple T cell hits are required to kill a single virus-infected cell. This raises the question whether cytotoxic lymphocytes can use granzymes to control HCMV infection in a noncytotoxic manner. Here, we demonstrate that (primary) cytotoxic lymphocytes can block HCMV dissemination independent of host cell death, and interferon-α/ß/γ. Prior to killing, cytotoxic lymphocytes induce the degradation of viral immediate-early (IE) proteins IE1 and IE2 in HCMV-infected cells. Intriguingly, both IE1 and/or IE2 are directly proteolyzed by all human granzymes, with GrB and GrM being most efficient. GrB and GrM cleave IE1 after Asp398 and Leu414, respectively, likely resulting in IE1 aberrant cellular localization, IE1 instability, and functional impairment of IE1 to interfere with the JAK-STAT signaling pathway. Furthermore, GrB and GrM cleave IE2 after Asp184 and Leu173, respectively, resulting in IE2 aberrant cellular localization and functional abolishment of IE2 to transactivate the HCMV UL112 early promoter. Taken together, our data indicate that cytotoxic lymphocytes can also employ noncytotoxic ways to control HCMV infection, which may be explained by granzyme-mediated targeting of indispensable viral proteins during lytic infection.


Asunto(s)
Infecciones por Citomegalovirus/enzimología , Citomegalovirus/metabolismo , Granzimas/metabolismo , Proteínas Inmediatas-Precoces/metabolismo , Células Asesinas Naturales/enzimología , Transactivadores/metabolismo , Secuencias de Aminoácidos , Citomegalovirus/genética , Infecciones por Citomegalovirus/virología , Granzimas/genética , Interacciones Huésped-Patógeno , Humanos , Proteínas Inmediatas-Precoces/genética , Proteolisis , Linfocitos T Citotóxicos/enzimología , Transactivadores/genética
3.
Viruses ; 11(5)2019 05 03.
Artículo en Inglés | MEDLINE | ID: mdl-31058862

RESUMEN

Cytomegalovirus (CMV) infections are still a global health problem, because the latent viruses persist in humans and cause recurring disease. Currently, there are no therapies for CMV latent infections and the therapies for active infections are limited by side effects and other problems. It is impossible to eradicate latent viruses in animals. HCMV (human CMV) is specific to human diseases; however, it is difficult to study HCMV due to its host specificity and long life cycle. Fortunately, MCMV (murine CMV) provides an excellent animal model. Here, three specific pairs of transcription activator-like effector nuclease (TALEN) plasmids (MCMV1-2, 3-4, and 5-6) were constructed to target the MCMV M80/80.5 sequence in order to test their efficacy in blocking MCMV lytic replication in NIH3T3 cell culture. The preliminary data showed that TALEN plasmids demonstrate specific targeting and cleavage in the MCMV M80/80.5 sequence and effectively inhibit MCMV growth in cell culture when the plasmid transfection is prior to the viral infection. The most specific pairs of TALEN plasmids (MCMV3-4) were further used to confirm the negative regulation of latent MCMV replication and gene expression in Balb/c mice. The injection of specific TALEN plasmids caused significant inhibition in the copy number level of immediately early gene (ie-1) DNA in five organs of mice, when compared with the controls. The result demonstrated that TALENs potentially provide an effective strategy to remove latent MCMV in animals.


Asunto(s)
Infecciones por Citomegalovirus/enzimología , Citomegalovirus/fisiología , Infecciones por Herpesviridae/enzimología , Infecciones por Herpesviridae/virología , Muromegalovirus/fisiología , Nucleasas de los Efectores Tipo Activadores de la Transcripción/metabolismo , Latencia del Virus , Animales , Citomegalovirus/genética , Infecciones por Citomegalovirus/genética , Infecciones por Citomegalovirus/virología , Modelos Animales de Enfermedad , Infecciones por Herpesviridae/genética , Humanos , Ratones , Ratones Endogámicos BALB C , Muromegalovirus/genética , Muromegalovirus/crecimiento & desarrollo , Nucleasas de los Efectores Tipo Activadores de la Transcripción/genética , Proteínas Virales/genética , Proteínas Virales/metabolismo , Replicación Viral
4.
J Virol ; 93(9)2019 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-30760575

RESUMEN

Human cytomegalovirus (HCMV) is a ubiquitous herpesvirus that causes disease in immunosuppressed populations. HCMV has a complex relationship with innate immune signaling pathways. Specifically, HCMV has been found to block some aspects of inflammatory signaling while benefiting from others. Through analysis of knockout cell lines targeting the NF-κB regulatory kinases IκB kinase α (IKKα) and IKKß, we find that the IKKs are host restriction factors that contribute to cytokine-mediated resistance to viral infection, limit the initiation of HCMV infection, and attenuate viral cell-to-cell spread. The HCMV UL26 protein is a viral immune modulator important for HCMV infection that has been shown to inhibit host cell NF-κB signaling, yet it has remained unclear how UL26-mediated NF-κB modulation contributes to infection. Here, we find that UL26 modulation of NF-κB signaling is separable from its contribution to high-titer viral replication. However, we find that IKKß is required for the induction of cytokine expression associated with ΔUL26 infection. Collectively, our data indicate that the IKKs restrict infection but HCMV targets their signaling to modulate the cellular inflammatory environment.IMPORTANCE Innate immune signaling is a critical defense against viral infection and represents a central host-virus interaction that frequently determines the outcomes of infections. NF-κB signaling is an essential component of innate immunity that is extensively modulated by HCMV, a significant cause of morbidity in neonates and immunosuppressed individuals. However, the roles that various facets of NF-κB signaling play during HCMV infection have remained elusive. We find that the two major regulatory kinases in this pathway, IKKα and IKKß, limit the initiation of infection, viral replication, and cell-to-cell spread. In addition, our results indicate that these kinases contribute differently to the host cell response to infection in the absence of a virally encoded NF-κB inhibitor, UL26. Given the importance of NF-κB in viral infection, elucidating the contributions of various NF-κB constituents to infection is an essential first step toward the possibility of targeting this pathway therapeutically.


Asunto(s)
Infecciones por Citomegalovirus/enzimología , Citomegalovirus/metabolismo , Quinasa I-kappa B/metabolismo , Transducción de Señal , Citomegalovirus/genética , Infecciones por Citomegalovirus/genética , Células HEK293 , Humanos , Quinasa I-kappa B/genética , FN-kappa B/genética , FN-kappa B/metabolismo , Proteínas Virales/genética , Proteínas Virales/metabolismo
5.
Placenta ; 72-73: 10-19, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30501876

RESUMEN

INTRODUCTION: Congenital cytomegalovirus (HCMV) infection may cause significant fetal malformation and in severe cases fetal and neonatal death. Fetal injury may be caused indirectly by the placental response to infection. Dual-specificity tyrosine phosphorylation-regulated kinases (DYRKs) have recently been identified as critical kinases for HCMV replication. In this study we provide first evidence that DYRK1A and DYRK1B are utilised during HCMV placental replication. METHODS: DYRK expression was investigated in AD169- and Merlin-infected TEV-1 trophoblast cells, ex vivo placental explants and naturally infected clinical placentae by immunofluorescence, western blot, co-immunoprecipitation and RT-qPCR. RESULTS: HCMV-infected placental cells showed accumulation and re-localisation of DYRK1A and DYRK1B protein to areas of cytoplasmic virion assembly complexes and nuclear viral replication compartments, respectively. This accumulation was a result of upregulated DYRK1A/B protein expression with HCMV inducing up to a 5.3-fold increase in DYRK1A and up to a 4.7-fold increase in DYRK1B protein, relative to mock-infected TEV-1 cells (p < 0.0001). Increased DYRK protein expression was correlated with DYRK1A/B mRNA upregulation, with HCMV-infected cells showing up to a 3.7-fold increase and 2.9-fold increase in DYRK1A and DYRK1B mRNA levels respectively (p < 0.05). Protein-protein interactions were detected between DYRK1A/1B complexes and HCMV immediate early IE2p86, early pp65 and pUL44 and late pp150 proteins. Treatment of HCMV-infected TEV-1 cells and placental explants with DYRK inhibitors significantly inhibited HCMV replication (p < 0.05) indicating these cellular kinases are required during HCMV placental replication. CONCLUSION: HCMV modulates cellular DYRKs during placental replication which may have implications for congenital HCMV pathogenesis and represent promising antiviral targets.


Asunto(s)
Citomegalovirus/fisiología , Placenta/virología , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Tirosina Quinasas/metabolismo , Replicación Viral/fisiología , Línea Celular , Núcleo Celular/enzimología , Infecciones por Citomegalovirus/complicaciones , Infecciones por Citomegalovirus/congénito , Infecciones por Citomegalovirus/enzimología , Citoplasma/enzimología , Femenino , Técnica del Anticuerpo Fluorescente , Humanos , Placenta/enzimología , Embarazo , Proteínas Serina-Treonina Quinasas/análisis , Proteínas Serina-Treonina Quinasas/genética , Proteínas Tirosina Quinasas/análisis , Proteínas Tirosina Quinasas/genética , ARN Mensajero/análisis , Trofoblastos/enzimología , Trofoblastos/virología , Regulación hacia Arriba , Proteínas Virales/análisis , Proteínas Virales/metabolismo , Quinasas DyrK
6.
J Virol ; 92(6)2018 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-29263260

RESUMEN

While cytomegalovirus (CMV) infections are often limited in host range by lengthy coevolution with a single host species, a few CMVs are known to deviate from this rule. For example, rhesus macaque CMV (RhCMV), a model for human CMV (HCMV) pathogenesis and vaccine development, can replicate in human cells, as well as in rhesus cells. Both HCMV and RhCMV encode species-specific antagonists of the broadly acting host cell restriction factor protein kinase R (PKR). Although the RhCMV antagonist of PKR, rTRS1, has very limited activity against human PKR, here, we show it is essential for RhCMV replication in human cells because it prevents human PKR from phosphorylating the translation initiation factor eIF2α, thereby allowing continued translation and viral replication. Although rTRS1 is necessary for RhCMV replication, it is not sufficient to rescue replication of HCMV lacking its own PKR antagonists in human fibroblasts. However, overexpression of rTRS1 in human fibroblasts enabled HCMV expressing rTRS1 to replicate, indicating that elevated levels or early expression of a weak antagonist can counteract a resistant restriction factor like human PKR. Exploring potential mechanisms that might allow RhCMV to replicate in human cells revealed that RhCMV makes no less double-stranded RNA than HCMV. Rather, in human cells, RhCMV expresses rTRS1 at levels 2 to 3 times higher than those of the HCMV-encoded PKR antagonists during HCMV infection. These data suggest that even a modest increase in expression of this weak PKR antagonist is sufficient to enable RhCMV replication in human cells.IMPORTANCE Rhesus macaque cytomegalovirus (RhCMV) offers a valuable model for studying congenital human cytomegalovirus (HCMV) pathogenesis and vaccine development. Therefore, it is critical to understand variations in how each virus infects and affects its host species to be able to apply insights gained from the RhCMV model to HCMV. While HCMV is capable only of infecting cells from humans and very closely related species, RhCMV displays a wider host range, including human as well as rhesus cells. RhCMV expresses an antagonist of a broadly acting antiviral factor present in all mammalian cells, and its ability to counter both the rhesus and human versions of this host factor is a key component of RhCMV's ability to cross species barriers. Here, we examine the molecular mechanisms that allow this RhCMV antagonist to function against a human restriction factor.


Asunto(s)
Infecciones por Citomegalovirus/enzimología , Citomegalovirus/metabolismo , Fibroblastos/enzimología , Transducción de Señal , eIF-2 Quinasa/metabolismo , Línea Celular , Citomegalovirus/genética , Infecciones por Citomegalovirus/genética , Fibroblastos/patología , Fibroblastos/virología , Humanos , Especificidad de la Especie , eIF-2 Quinasa/antagonistas & inhibidores , eIF-2 Quinasa/genética
7.
Cell Rep ; 20(4): 846-853, 2017 07 25.
Artículo en Inglés | MEDLINE | ID: mdl-28746870

RESUMEN

Herein, we demonstrate that HCMV miR-UL112-5p targets ERAP1, thereby inhibiting the processing and presentation of the HCMV pp65495-503 peptide to specific CTLs. In addition, we show that the rs17481334 G variant, naturally occurring in the ERAP1 3' UTR, preserves ERAP1 from miR-UL112-5p-mediated degradation. Specifically, HCMV miR-UL112-5p binds the 3' UTR of ERAP1 A variant, but not the 3' UTR of ERAP1 G variant, and, accordingly, ERAP1 expression is reduced both at RNA and protein levels only in human fibroblasts homozygous for the A variant. Consistently, HCMV-infected GG fibroblasts were more efficient in trimming viral antigens and being lysed by HCMV-peptide-specific CTLs. Notably, a significantly decreased HCMV seropositivity was detected among GG individuals suffering from multiple sclerosis, a disease model in which HCMV is negatively associated with adult-onset disorder. Overall, our results identify a resistance mechanism to HCMV miR-UL112-5p-based immune evasion strategy with potential implications for individual susceptibility to infection and other diseases.


Asunto(s)
Aminopeptidasas/metabolismo , Citomegalovirus/genética , Variación Genética/genética , MicroARNs/metabolismo , Antígenos de Histocompatibilidad Menor/metabolismo , Regiones no Traducidas 3'/genética , Aminopeptidasas/genética , Linfocitos T CD8-positivos/metabolismo , Infecciones por Citomegalovirus/enzimología , Infecciones por Citomegalovirus/genética , Genotipo , Humanos , MicroARNs/genética , Antígenos de Histocompatibilidad Menor/genética , Esclerosis Múltiple/enzimología , Esclerosis Múltiple/genética , Unión Proteica , ARN Mensajero/genética , ARN Viral/genética , Linfocitos T Citotóxicos/metabolismo
8.
PLoS Pathog ; 13(7): e1006542, 2017 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-28750047

RESUMEN

Congenital human cytomegalovirus (HCMV) infection is the leading cause of neurological disabilities in children worldwide, but the mechanisms underlying these disorders are far from well-defined. HCMV infection has been shown to dysregulate the Notch signaling pathway in human neural progenitor cells (NPCs). As an important downstream effector of Notch signaling, the transcriptional regulator Hairy and Enhancer of Split 1 (Hes1) is essential for governing NPC fate and fetal brain development. In the present study, we report that HCMV infection downregulates Hes1 protein levels in infected NPCs. The HCMV 72-kDa immediate-early 1 protein (IE1) is involved in Hes1 degradation by assembling a ubiquitination complex and promoting Hes1 ubiquitination as a potential E3 ubiquitin ligase, followed by proteasomal degradation of Hes1. Sp100A, an important component of PML nuclear bodies, is identified to be another target of IE1-mediated ubiquitination. A C-terminal acidic region in IE1, spanning amino acids 451 to 475, is required for IE1/Hes1 physical interaction and IE1-mediated Hes1 ubiquitination, but is dispensable for IE1/Sp100A interaction and ubiquitination. Our study suggests a novel mechanism linking downregulation of Hes1 protein to neurodevelopmental disorders caused by HCMV infection. Our findings also complement the current knowledge of herpesviruses by identifying IE1 as the first potential HCMV-encoded E3 ubiquitin ligase.


Asunto(s)
Infecciones por Citomegalovirus/enzimología , Infecciones por Citomegalovirus/virología , Citomegalovirus/metabolismo , Células-Madre Neurales/metabolismo , Factor de Transcripción HES-1/genética , Ubiquitina-Proteína Ligasas/metabolismo , Citomegalovirus/genética , Infecciones por Citomegalovirus/genética , Regulación hacia Abajo , Interacciones Huésped-Patógeno , Humanos , Proteínas Inmediatas-Precoces/genética , Proteínas Inmediatas-Precoces/metabolismo , Células-Madre Neurales/enzimología , Células-Madre Neurales/virología , Unión Proteica , Proteolisis , Factor de Transcripción HES-1/metabolismo , Ubiquitina-Proteína Ligasas/genética , Ubiquitinación
9.
J Virol ; 91(5)2017 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-27974558

RESUMEN

Double-stranded RNAs (dsRNA) produced during human cytomegalovirus (HCMV) infection activate the antiviral kinase protein kinase R (PKR), which potently inhibits virus replication. The HCMV pTRS1 and pIRS1 proteins antagonize PKR to promote HCMV protein synthesis and replication; however, the mechanism by which pTRS1 inhibits PKR is unclear. PKR activation occurs in a three-step cascade. First, binding to dsRNA triggers PKR homodimerizaton. PKR dimers then autophosphorylate, leading to a conformational shift that exposes the binding site for the PKR substrate eIF2α. Consistent with previous in vitro studies, we found that pTRS1 bound and inhibited PKR. pTRS1 binding to PKR was not mediated by an RNA intermediate, and mutations in the pTRS1 RNA binding domain did not affect PKR binding or inhibition. Rather, mutations that disrupted the pTRS1 interaction with PKR ablated the ability of pTRS1 to antagonize PKR activation by dsRNA. pTRS1 did not block PKR dimerization and could bind and inhibit a constitutively dimerized PKR kinase domain. In addition, pTRS1 binding to PKR inhibited PKR kinase activity. Single amino acid point mutations in the conserved eIF2α binding domain of PKR disrupted pTRS1 binding and rendered PKR resistant to inhibition by pTRS1. Consistent with a critical role for the conserved eIF2α contact site in PKR binding, pTRS1 bound an additional eIF2α kinase, heme-regulated inhibitor (HRI), and inhibited eIF2α phosphorylation in response to an HRI agonist. Together our data suggest that pTRS1 inhibits PKR by binding to conserved amino acids in the PKR eIF2α binding site and blocking PKR kinase activity.IMPORTANCE The antiviral kinase PKR plays a critical role in controlling HCMV replication. This study furthered our understanding of how HCMV evades inhibition by PKR and identified new strategies for how PKR activity might be restored during infection to limit HCMV disease.


Asunto(s)
Infecciones por Citomegalovirus/enzimología , Citomegalovirus/fisiología , eIF-2 Quinasa/metabolismo , Secuencias de Aminoácidos , Infecciones por Citomegalovirus/virología , Activación Enzimática , Células HEK293 , Interacciones Huésped-Patógeno , Humanos , Fosforilación , Dominios y Motivos de Interacción de Proteínas , Multimerización de Proteína , Procesamiento Proteico-Postraduccional , ARN Bicatenario/fisiología , ARN Viral/fisiología , Proteínas Virales/fisiología , Replicación Viral
10.
Oncotarget ; 7(30): 47221-47231, 2016 Jul 26.
Artículo en Inglés | MEDLINE | ID: mdl-27363017

RESUMEN

BACKGROUND: Both arginase (ARG2) and human cytomegalovirus (HCMV) have been implicated in tumorigenesis. However, the role of ARG2 in the pathogenesis of glioblastoma (GBM) and the HCMV effects on ARG2 are unknown. We hypothesize that HCMV may contribute to tumorigenesis by increasing ARG2 expression. RESULTS: ARG2 promotes tumorigenesis by increasing cellular proliferation, migration, invasion and vasculogenic mimicry in GBM cells, at least in part due to overexpression of MMP2/9. The nor-NOHA significantly reduced migration and tube formation of ARG2-overexpressing cells. HCMV immediate-early proteins (IE1/2) or its downstream pathways upregulated the expression of ARG2 in U-251 MG cells. Immunostaining of GBM tissue sections confirmed the overexpression of ARG2, consistent with data from subsets of Gene Expression Omnibus. Moreover, higher levels of ARG2 expression tended to be associated with poorer survival in GBM patient by analyzing data from TCGA. METHODS: The role of ARG2 in tumorigenesis was examined by proliferation-, migration-, invasion-, wound healing- and tube formation assays using an ARG2-overexpressing cell line and ARG inhibitor, N (omega)-hydroxy-nor-L-arginine (nor-NOHA) and siRNA against ARG2 coupled with functional assays measuring MMP2/9 activity, VEGF levels and nitric oxide synthase activity. Association between HCMV and ARG2 were examined in vitro with 3 different GBM cell lines, and ex vivo with immunostaining on GBM tissue sections. The viral mechanism mediating ARG2 induction was examined by siRNA approach. Correlation between ARG2 expression and patient survival was extrapolated from bioinformatics analysis on data from The Cancer Genome Atlas (TCGA). CONCLUSIONS: ARG2 promotes tumorigenesis, and HCMV may contribute to GBM pathogenesis by upregulating ARG2.


Asunto(s)
Arginasa/biosíntesis , Citomegalovirus/fisiología , Glioblastoma/virología , Arginasa/genética , Carcinogénesis , Línea Celular Tumoral , Movimiento Celular/fisiología , Proliferación Celular/fisiología , Citomegalovirus/genética , Citomegalovirus/metabolismo , Infecciones por Citomegalovirus/enzimología , Infecciones por Citomegalovirus/patología , Infecciones por Citomegalovirus/virología , Progresión de la Enfermedad , Glioblastoma/irrigación sanguínea , Glioblastoma/enzimología , Glioblastoma/patología , Humanos , Inmunohistoquímica , Neovascularización Patológica/enzimología , Neovascularización Patológica/patología , Neovascularización Patológica/virología , Transfección , Regulación hacia Arriba
11.
Virology ; 489: 194-201, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26773380

RESUMEN

Human cytomegalovirus (HCMV) was recently shown to encode a large number of spliced mRNAs. While the nuclear export of unspliced viral transcripts has been extensively studied, the role of host mRNA export factors in HCMV mRNA trafficking remains poorly defined. We found that the eIF4AIII RNA helicase, a component of the exon junction complex, was necessary for efficient virus replication. Depletion of eIF4AIII limited viral DNA accumulation, export of viral mRNAs from the nucleus, and the production of progeny virus. However eIF4AIII was dispensable for the association of viral transcripts with ribosomes. We found that pateamine A, a natural compound that inhibits both eIF4AI/II and eIF4AIII, has potent antiviral activity and inhibits HCMV replication throughout the virus lytic cycle. Our results demonstrate that eIF4AIII is required for efficient HCMV replication, and suggest that eIF4A family helicases may be a new class of targets for the development of host-directed antiviral therapeutics.


Asunto(s)
Infecciones por Citomegalovirus/enzimología , Citomegalovirus/fisiología , Factor 4A Eucariótico de Iniciación/metabolismo , Transporte Activo de Núcleo Celular , Núcleo Celular/virología , Citomegalovirus/genética , Infecciones por Citomegalovirus/genética , Infecciones por Citomegalovirus/virología , Factor 4A Eucariótico de Iniciación/genética , Interacciones Huésped-Patógeno , Humanos , ARN Viral/genética , ARN Viral/metabolismo , Replicación Viral
12.
Virology ; 489: 75-85, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26716879

RESUMEN

Human cytomegalovirus (HCMV) lacking TRS1 and IRS1 (HCMV[ΔI/ΔT]) cannot replicate in cell culture. Although both proteins can block the protein kinase R (PKR) pathway, they have multiple other activities and binding partners. It remains unknown which functions are essential for HCMV replication. To investigate this issue, we first identified a TRS1 mutant that is unable to bind to PKR. Like HCMV[ΔI/ΔT], a recombinant HCMV containing this mutant (HCMV[TRS1-Mut 1]) did not replicate in wild-type cells. However, HCMV[ΔI/ΔT] did replicate in cells in which PKR expression was reduced by RNA interference. Moreover, HCMV[ΔI/ΔT] and HCMV[TRS1-Mut 1] replicated to similar levels as virus containing wild-type TRS1 in cell lines in which PKR expression was knocked out by CRISPR/Cas9-mediated genome editing. These results demonstrate that the sole essential function of TRS1 is to antagonize PKR and that its other activities do not substantially enhance HCMV replication, at least in cultured human fibroblasts.


Asunto(s)
Infecciones por Citomegalovirus/enzimología , Citomegalovirus/fisiología , Proteínas Virales/metabolismo , eIF-2 Quinasa/metabolismo , Citomegalovirus/genética , Infecciones por Citomegalovirus/genética , Infecciones por Citomegalovirus/virología , Células HeLa , Humanos , Unión Proteica , Proteínas Virales/genética , Replicación Viral , eIF-2 Quinasa/genética
14.
Folia Microbiol (Praha) ; 60(3): 199-209, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25366712

RESUMEN

Herpesvirus infections, such as those induced by human cytomegalovirus (HCMV), induce specific DNA damages. DNA damages can lead to cell mutation, death, apoptosis and immune system activation. Various types of DNA damage are repaired through multiple repair pathways, such as base excision, nucleotide excision, homologous recombination and nonhomologous end joining. Changes in the activity of DNA repair proteins during viral infection can cause disturbances in the DNA repair system and change its mechanisms. This report reviews results from studies, assaying a DNA repair system in HCMV infection.


Asunto(s)
Infecciones por Citomegalovirus/genética , Citomegalovirus/fisiología , Reparación del ADN , Animales , Infecciones por Citomegalovirus/enzimología , Infecciones por Citomegalovirus/metabolismo , Infecciones por Citomegalovirus/microbiología , Daño del ADN , Enzimas Reparadoras del ADN/genética , Enzimas Reparadoras del ADN/metabolismo , Humanos
15.
Viruses ; 8(1)2015 Dec 29.
Artículo en Inglés | MEDLINE | ID: mdl-26729153

RESUMEN

Human cytomegalovirus (HCMV) was reported to downregulate the Wnt/ß-catenin pathway. Induction of Axin1, the negative regulator of the Wnt pathway, has been reported as an important mechanism for inhibition of ß-catenin. Since Tankyrase (TNKS) negatively regulates Axin1, we investigated the effect of HCMV on TNKS expression and poly-ADP ribose polymerase (PARsylation) activity, during virus replication. Starting at 24 h post infection, HCMV stabilized the expression of TNKS and reduced its PARsylation activity, resulting in accumulation of Axin1 and reduction in its PARsylation as well. General PARsylation was not changed in HCMV-infected cells, suggesting specific inhibition of TNKS PARsylation. Similarly, treatment with XAV939, a chemical inhibitor of TNKS' activity, resulted in the accumulation of TNKS in both non-infected and HCMV-infected cell lines. Reduction of TNKS activity or knockdown of TNKS was beneficial for HCMV, evidenced by its improved growth in fibroblasts. Our results suggest that HCMV modulates the activity of TNKS to induce Axin1, resulting in inhibition of the ß-catenin pathway. Since HCMV replication is facilitated by TNKS knockdown or inhibition of its activity, TNKS may serve as an important virus target for control of a variety of cellular processes.


Asunto(s)
Infecciones por Citomegalovirus/enzimología , Citomegalovirus/fisiología , Tanquirasas/metabolismo , Vía de Señalización Wnt , Línea Celular Tumoral , Citomegalovirus/genética , Infecciones por Citomegalovirus/genética , Infecciones por Citomegalovirus/metabolismo , Infecciones por Citomegalovirus/virología , Interacciones Huésped-Patógeno , Humanos , Poli(ADP-Ribosa) Polimerasas/genética , Poli(ADP-Ribosa) Polimerasas/metabolismo , Tanquirasas/genética , Replicación Viral
16.
J Immunol ; 193(3): 1344-52, 2014 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-24973455

RESUMEN

Proteolytic shedding of ligands for the NK group 2D (NKG2D) receptor is a strategy used by tumors to modulate immune recognition by NK cells and cytotoxic T cells. A number of metalloproteases, especially those of the A disintegrin and metalloprotease (ADAM) family, can mediate NKG2D ligand cleavage and this process can be modulated by expression of the thiol isomerase ERp5. In this article, we describe that an increased shedding of the NKG2D ligand MICA is observed postinfection with several strains of human CMV due to an enhanced activity of ADAM17 (TNF-α converting enzyme) and matrix metalloprotease 14 caused by a reduction in the expression of the endogenous inhibitor of metalloproteases tissue inhibitors of metalloproteinase 3 (TIMP3). This decrease in TIMP3 expression correlates with increased expression of a cellular miRNA known to target TIMP3, and we also identify a human CMV-encoded microRNA able to modulate TIMP3 expression. These observations characterize a novel viral strategy to influence the shedding of cell-surface molecules involved in immune response modulation. They also provide an explanation for previous reports of increased levels of various ADAM17 substrates in the serum from patients with CMV disease. Consistent with this hypothesis, we detected soluble MICA in serum of transplant recipients with CMV disease. Finally, these data suggest that it might be worthwhile to prospectively study ADAM17 activity in a larger group of patients to assay whether this might be a useful biomarker to identify patients at risk for development of CMV disease.


Asunto(s)
Infecciones por Citomegalovirus/inmunología , Regulación hacia Abajo/inmunología , Regulación Viral de la Expresión Génica/inmunología , Antígenos de Histocompatibilidad Clase I/metabolismo , MicroARNs/genética , Inhibidor Tisular de Metaloproteinasa-3/antagonistas & inhibidores , Inhibidor Tisular de Metaloproteinasa-3/biosíntesis , Línea Celular Tumoral , Citomegalovirus/genética , Citomegalovirus/inmunología , Infecciones por Citomegalovirus/enzimología , Infecciones por Citomegalovirus/genética , Regulación hacia Abajo/genética , Antígenos de Histocompatibilidad Clase I/sangre , Humanos , Metaloproteinasa 14 de la Matriz/sangre , Metaloproteinasa 14 de la Matriz/metabolismo , MicroARNs/biosíntesis , Cultivo Primario de Células , Especificidad por Sustrato/genética , Especificidad por Sustrato/inmunología , Inhibidor Tisular de Metaloproteinasa-3/sangre , Regulación hacia Arriba/genética , Regulación hacia Arriba/inmunología
17.
J Virol ; 88(18): 10982-5, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24965476

RESUMEN

Human cytomegalovirus (HCMV) kinase UL97 is required for efficient nuclear lamina disruption during nuclear egress. However, cellular protein kinase C (PKC) has been implicated in this process in other systems. Comparing the effects of UL97 and cellular kinase inhibitors on HCMV nuclear egress confirms a role for UL97 in lamina disruption and nuclear egress. A pan-PKC inhibitor did not affect lamina disruption but did reduce the number of cytoplasmic capsids more than the number of nuclear capsids.


Asunto(s)
Núcleo Celular/virología , Infecciones por Citomegalovirus/enzimología , Citomegalovirus/enzimología , Lámina Nuclear/virología , Fosfotransferasas (Aceptor de Grupo Alcohol)/antagonistas & inhibidores , Proteína Quinasa C/antagonistas & inhibidores , Inhibidores de Proteínas Quinasas/farmacología , Liberación del Virus/efectos de los fármacos , Cápside/metabolismo , Línea Celular , Núcleo Celular/efectos de los fármacos , Citomegalovirus/efectos de los fármacos , Citomegalovirus/genética , Citomegalovirus/fisiología , Infecciones por Citomegalovirus/virología , Humanos , Lámina Nuclear/efectos de los fármacos , Fosfotransferasas (Aceptor de Grupo Alcohol)/genética , Fosfotransferasas (Aceptor de Grupo Alcohol)/metabolismo , Proteína Quinasa C/metabolismo , Ensamble de Virus/efectos de los fármacos
18.
Placenta ; 35(6): 345-50, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24746852

RESUMEN

INTRODUCTION: Human cytomegalovirus (HCMV) can cause congenital infection with risk of neurological disability. Maternal-fetal transmission is associated with placental inflammation. 5-lipoxygenase (5-LO) is the key enzyme in the biosynthesis of Leukotrienes (LTs), which are proinflammatory mediators. This study investigated the effect of HCMV infection on 5-LO expression and Leukotriene-B4 (LTB4) induction in human placentae and umbilical vein endothelial cells (HUVEC). METHODS: Seven placentae from fetuses with congenital HCMV infection and brain damage and six controls were stained with HCMV-immediate-early-antigen (HCMV-IEA) and 5-LO by immunohistochemistry. 5-hydroxyeicosatetraenoic acid (5-HETE) and LTB4 were measured in culture supernatant from ex vivo HCMV-infected placental histocultures by liquid chromatography. In vitro, HCMV infected HUVEC cells were analyzed for 5-LO mRNA and protein expression by real time PCR and immunofluorescence staining. RESULTS: HCMV-IEA was abundant in all HCMV infected placentae but absent in control placentae. 5-LO expression was higher in endothelial and smooth muscle cells of HCMV-infected placentae, compared to control placentae. HCMV infection induced an up-regulation of LTB4 in ex vivo placental explants with higher levels of LTB4 at 72 h compared to controls (p = 0.002). In vitro, 5-LO transcript and protein expression were significantly induced in HCMV-infected HUVEC, compared to the control cultures (p = 0.036). CONCLUSION: The presence of HCMV coincided with high 5-LO expression in cells of in vivo HCMV infected placentae. HCMV induced up-regulation of 5-LO in both ex vivo HCMV-infected placental explants and HUVEC. HCMV induced LT-biosynthesis in congenitally infected placentae may have a role in pathogenesis of congenital HCMV disease.


Asunto(s)
Araquidonato 5-Lipooxigenasa/análisis , Infecciones por Citomegalovirus/congénito , Células Endoteliales/química , Leucotrieno B4/análisis , Placenta/química , Venas Umbilicales/química , Araquidonato 5-Lipooxigenasa/genética , Infecciones por Citomegalovirus/enzimología , Infecciones por Citomegalovirus/metabolismo , Células Endoteliales/enzimología , Femenino , Células Endoteliales de la Vena Umbilical Humana , Humanos , Ácidos Hidroxieicosatetraenoicos/análisis , Inmunohistoquímica , Placenta/enzimología , Embarazo , ARN Mensajero/análisis , Venas Umbilicales/enzimología , Regulación hacia Arriba
19.
Exp Gerontol ; 54: 130-7, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24440462

RESUMEN

NK cells represent an important component of the innate immune response against infection and tumors. Age-associated changes in NK cell phenotype have been previously reported that can be responsible of functional NK cell deficiency. The aim of this work was to analyze the effect CMV seropositivity and aging on the distribution of NK cell subsets with a focus on the expression of cytotoxicity-related molecules and on the expression of CD94/NKG2 heterodimers and CD57 on these NK cell subsets. Our results show that CMV seropositivity in young individuals does not significantly affect peripheral blood NK cell percentage and NK cell subsets defined by the use of CD56 and CD16 markers. In contrast a significant increase in the percentage of NK cells is observed in elderly donors, all of them are CMV seropositive, when compared with young CMV seropositive subjects. A decrease in the percentage of CD56bright NK cells, either fully immature CD16 negative or CD16+ and an increase in the CD56-CD16+ subset are also found in the elderly. CMV seropositivity either in healthy young or elderly individuals is associated to the expression of CD94/NKG2C dimers and high expression of CD57on the CD56dimCD16+ NK cell subset. CD56-CD16+ NK cells, which are expanded in the elderly, show a decreased expression of granzymes A and B and an increased expression of CD94/NKG2C and CD57 in CMV seropositive young donors when compared with CMV seronegative young individuals. These results indicate that CMV and age have a different effect on NK cell phenotype and emphasize the relevance of including the determination of CMV serostatus in those studies addressed to analyze the immune response in the elderly.


Asunto(s)
Infecciones por Citomegalovirus/inmunología , Células Asesinas Naturales/virología , Adulto , Anciano , Anciano de 80 o más Años , Antígeno CD56/metabolismo , Antígenos CD57/metabolismo , Infecciones por Citomegalovirus/enzimología , Femenino , Granzimas/metabolismo , Humanos , Células Asesinas Naturales/enzimología , Subgrupos Linfocitarios/enzimología , Subgrupos Linfocitarios/virología , Masculino , Subfamília D de Receptores Similares a Lectina de las Células NK/metabolismo , Perforina/metabolismo , Adulto Joven
20.
J Gen Virol ; 95(Pt 3): 659-670, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24337170

RESUMEN

Despite a rigorous blockade of interferon-γ (IFN-γ) signalling in infected fibroblasts as a mechanism of immune evasion by human cytomegalovirus (HCMV), IFN-γ induced indoleamine-2,3-dioxygenase (IDO) has been proposed to represent the major antiviral restriction factor limiting HCMV replication in epithelial cells. Here we show that HCMV efficiently blocks transcription of IFN-γ-induced IDO mRNA both in infected fibroblasts and epithelial cells even in the presence of a preexisting IFN-induced antiviral state. This interference results in severe suppression of IDO bioactivity in HCMV-infected cells and restoration of vigorous HCMV replication. Depletion of IDO expression nonetheless substantially alleviated the antiviral impact of IFN-γ treatment in both cell types. These findings highlight the effectiveness of this IFN-γ induced effector gene in restricting HCMV productivity, but also the impact of viral counter-measures.


Asunto(s)
Infecciones por Citomegalovirus/enzimología , Citomegalovirus/fisiología , Indolamina-Pirrol 2,3,-Dioxigenasa/genética , Replicación Viral , Línea Celular , Citomegalovirus/genética , Infecciones por Citomegalovirus/genética , Infecciones por Citomegalovirus/virología , Humanos , Indolamina-Pirrol 2,3,-Dioxigenasa/metabolismo , Interferón gamma/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...