Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 146
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
Angew Chem Int Ed Engl ; 62(41): e202309838, 2023 10 09.
Artículo en Inglés | MEDLINE | ID: mdl-37555536

RESUMEN

Herpes simplex virus (HSV-1) employs heparan sulfate (HS) as receptor for cell attachment and entry. During late-stage infection, the virus induces the upregulation of human heparanase (Hpse) to remove cell surface HS allowing viral spread. We hypothesized that inhibition of Hpse will prevent viral release thereby representing a new therapeutic strategy for HSV-1. A range of HS-oligosaccharides was prepared to examine the importance of chain length and 2-O-sulfation of iduronic moieties for Hpse inhibition. It was found that hexa- and octasaccharides potently inhibited the enzyme and that 2-O-sulfation of iduronic acid is tolerated. Computational studies provided a rationale for the observed structure-activity relationship. Treatment of human corneal epithelial cells (HCEs) infected with HSV-1 with the hexa- and octasaccharide blocked viral induced shedding of HS which significantly reduced spread of virions. The compounds also inhibited migration and proliferation of immortalized HCEs thereby providing additional therapeutic properties.


Asunto(s)
Glucuronidasa , Herpes Simple , Herpesvirus Humano 1 , Humanos , Glucuronidasa/antagonistas & inhibidores , Glucuronidasa/metabolismo , Heparitina Sulfato/farmacología , Herpes Simple/enzimología , Herpes Simple/virología , Herpesvirus Humano 1/metabolismo , Oligosacáridos/farmacología , Oligosacáridos/metabolismo
2.
PLoS One ; 17(2): e0264001, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35167620

RESUMEN

The role played by certain domestic species such as dogs as a translational model in comparative oncology shows great interest to develop new therapeutic strategies in brain tumors. Gliomas are a therapeutic challenge that represents the most common form of malignant primary brain tumors in humans and the second most common form in dogs. Gene-directed enzyme/prodrug therapy using adipose mesenchymal stem cells (Ad-MSCs) expressing the herpes simplex virus thymidine kinase (TK) has proven to be a promising alternative in glioblastoma therapy, through its capacity to migrate and home to the tumor and delivering local cytotoxicity avoiding other systemic administration. In this study, we demonstrate the possibility for canine Ad-MSCs (cAd-MSCs) to be genetically engineered efficiently with a lentiviral vector to express TK (TK-cAd-MSCs) and in combination with ganciclovir (GCV) prodrug demonstrated its potential antitumor efficacy in vitro and in vivo in a mice model with the human glioblastoma cell line U87. TK-cAd-MSCs maintained cell proliferation, karyotype stability, and MSCs phenotype. Genetic modification significantly affects its secretory profile, both the analyzed soluble factors and exosomes. TK-cAd-MSCs showed a high secretory profile of some active antitumor immune response cytokines and a threefold increase in the amount of secreted exosomes, with changes in their protein cargo. We also found that the prodrug protein is not released directly into the culture medium by TK-cAd-MSCs. We believe that our work provides new perspectives for glioblastoma gene therapy in dogs and a better understanding of this therapy in view of its possible implantation in humans.


Asunto(s)
Neoplasias Encefálicas/terapia , Ganciclovir/administración & dosificación , Glioblastoma/terapia , Herpes Simple/enzimología , Trasplante de Células Madre Mesenquimatosas/métodos , Células Madre Mesenquimatosas/citología , Timidina Quinasa/genética , Animales , Neoplasias Encefálicas/genética , Línea Celular Tumoral , Técnicas de Cocultivo , Perros , Ganciclovir/farmacología , Genes Transgénicos Suicidas , Terapia Genética , Glioblastoma/genética , Herpes Simple/genética , Humanos , Lentivirus/genética , Células Madre Mesenquimatosas/metabolismo , Ratones , Timidina Quinasa/metabolismo , Transducción Genética , Proteínas Virales/genética , Proteínas Virales/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
3.
mBio ; 12(6): e0279221, 2021 12 21.
Artículo en Inglés | MEDLINE | ID: mdl-34749529

RESUMEN

Under pathological conditions like herpes simplex virus 1 (HSV-1) infection, host-pathogen interactions lead to major reconstruction of the host protein network, which contributes to the dysregulation of signaling pathways and disease onset. Of note is the upregulation of a multifunctional host protein, heparanase (HPSE), following infection, which serves as a mediator in HSV-1 replication. In this study, we identify a novel function of HPSE and highlight it as a key regulator of ß-catenin signal transduction. The regulatory role of HPSE on the activation, nuclear translocation, and signal transduction of ß-catenin disrupts cellular homeostasis and establishes a pathogenic environment that promotes viral replication. Under normal physiological conditions, ß-catenin is bound to a group of proteins, referred to as the destruction complex, and targeted for ubiquitination and, ultimately, degradation. We show that virus-induced upregulation of HPSE leads to the activation of Akt and subsequent stabilization and activation of ß-catenin through (i) the release of ß-catenin from the destruction complex, and (ii) direct phosphorylation of ß-catenin at Ser552. This study also provides an in-depth characterization of the proviral role of ß-catenin signaling during HSV-1 replication using physiologically relevant cell lines and in vivo models of ocular infection. Furthermore, pharmacological inhibitors of this pathway generated a robust antiviral state against multiple laboratory and clinical strains of HSV-1. Collectively, our findings assign a novel regulatory role to HPSE as a driver of ß-catenin signaling in HSV-1 infection. IMPORTANCE Heparanase (HPSE) and ß-catenin have independently been implicated in regulating key pathophysiological processes, including neovascularization, angiogenesis, and inflammation; however, the relationship between the two proteins has remained elusive thus far. For that reason, characterizing this relationship is crucial and can lead to the development of novel therapeutics. For HSV-1 specifically, current antivirals are not able to abolish the virus from the host, leaving patients susceptible to episodes of viral reactivation. Identifying a host-based intervention can provide a better alternative with enhanced efficacy and sustained relief.


Asunto(s)
Glucuronidasa/metabolismo , Herpes Simple/enzimología , Herpesvirus Humano 1/fisiología , Proteínas Proto-Oncogénicas c-akt/metabolismo , beta Catenina/metabolismo , Secuencias de Aminoácidos , Línea Celular , Glucuronidasa/genética , Herpes Simple/genética , Herpes Simple/metabolismo , Herpes Simple/virología , Herpesvirus Humano 1/genética , Interacciones Huésped-Patógeno , Humanos , Fosforilación , Proteínas Proto-Oncogénicas c-akt/genética , Activación Viral , Replicación Viral , Vía de Señalización Wnt , beta Catenina/química , beta Catenina/genética
4.
mBio ; 12(2)2021 03 30.
Artículo en Inglés | MEDLINE | ID: mdl-33785613

RESUMEN

Retinoic acid-inducible gene I (RIG-I) is a sensor that recognizes cytosolic double-stranded RNA derived from microbes to induce host immune response. Viruses, such as herpesviruses, deploy diverse mechanisms to derail RIG-I-dependent innate immune defense. In this study, we discovered that mouse RIG-I is intrinsically resistant to deamidation and evasion by herpes simplex virus 1 (HSV-1). Comparative studies involving human and mouse RIG-I indicate that N495 of human RIG-I dictates species-specific deamidation by HSV-1 UL37. Remarkably, deamidation of the other site, N549, hinges on that of N495, and it is catalyzed by cellular phosphoribosylpyrophosphate amidotransferase (PPAT). Specifically, deamidation of N495 enables RIG-I to interact with PPAT, leading to subsequent deamidation of N549. Collaboration between UL37 and PPAT is required for HSV-1 to evade RIG-I-mediated antiviral immune response. This work identifies an immune regulatory role of PPAT in innate host defense and establishes a sequential deamidation event catalyzed by distinct deamidases in immune evasion.IMPORTANCE Herpesviruses are ubiquitous pathogens in human and establish lifelong persistence despite host immunity. The ability to evade host immune response is pivotal for viral persistence and pathogenesis. In this study, we investigated the evasion, mediated by deamidation, of species-specific RIG-I by herpes simplex virus 1 (HSV-1). Our findings uncovered a collaborative and sequential action between viral deamidase UL37 and a cellular glutamine amidotransferase, phosphoribosylpyrophosphate amidotransferase (PPAT), to inactivate RIG-I and mute antiviral gene expression. PPAT catalyzes the rate-limiting step of the de novo purine synthesis pathway. This work describes a new function of cellular metabolic enzymes in host defense and viral immune evasion.


Asunto(s)
Amidofosforribosiltransferasa/metabolismo , Proteína 58 DEAD Box/metabolismo , Herpes Simple/enzimología , Herpesvirus Humano 1/enzimología , Proteínas Estructurales Virales/metabolismo , Replicación Viral , Amidofosforribosiltransferasa/genética , Secuencias de Aminoácidos , Animales , Proteína 58 DEAD Box/química , Proteína 58 DEAD Box/genética , Herpes Simple/genética , Herpes Simple/virología , Herpesvirus Humano 1/genética , Herpesvirus Humano 1/fisiología , Interacciones Huésped-Patógeno , Humanos , Ratones , Unión Proteica , Especificidad de la Especie , Proteínas Estructurales Virales/genética
5.
Biochem J ; 478(1): 261-279, 2021 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-33355669

RESUMEN

Herpes simplex virus 1 (HSV-1) is a human DNA virus that causes cold sores, keratitis, meningitis, and encephalitis. Ubiquitination is a post-translational protein modification essential for regulation of cellular events, such as proteasomal degradation, signal transduction, and protein trafficking. The process is also involved in events for establishing viral infection and replication. The first step in ubiquitination involves ubiquitin (Ub) binding with Ub-activating enzyme (E1, also termed UBE1) via a thioester linkage. Our results show that HSV-1 infection alters protein ubiquitination pattern in host cells, as evidenced by MS spectra and co-immunoprecipitation assays. HSV-1 induced ubiquitination of UBE1a isoform via an isopeptide bond with Lys604. Moreover, we show that ubiquitination of K604 in UBE1a enhances UBE1a activity; that is, the activity of ubiquitin-transfer to E2 enzyme. Subsequently, we investigated the functional role of UBE1a and ubiquitination of K604 in UBE1a. We found that UBE1-knockdown increased HSV-1 DNA replication and viral production. Furthermore, overexpression of UBE1a, but not a UBE1a K604A mutant, suppressed viral replication. Furthermore, we found that UBE1a and ubiquitination at K604 in UBE1a retarded expression of HSV-1 major capsid protein, ICP5. Our findings show that UBE1a functions as an antiviral factor that becomes activated upon ubiquitination at Lys604.


Asunto(s)
Antivirales/metabolismo , Herpes Simple/metabolismo , Herpesvirus Humano 1/metabolismo , Interacciones Huésped-Patógeno/efectos de los fármacos , Enzimas Activadoras de Ubiquitina/metabolismo , Animales , Supervivencia Celular/genética , Chlorocebus aethiops , Cromatografía Liquida , Doxiciclina/farmacología , Células HeLa , Herpes Simple/enzimología , Herpes Simple/genética , Herpesvirus Humano 1/genética , Interacciones Huésped-Patógeno/genética , Humanos , Unión Proteica , Dominios Proteicos , Procesamiento Proteico-Postraduccional/efectos de los fármacos , ARN Interferente Pequeño , Espectrometría de Masas en Tándem , Tetraciclina/farmacología , Transfección , Enzimas Activadoras de Ubiquitina/genética , Ubiquitinación/efectos de los fármacos , Células Vero , Replicación Viral/efectos de los fármacos
6.
Nat Commun ; 11(1): 1338, 2020 03 12.
Artículo en Inglés | MEDLINE | ID: mdl-32165633

RESUMEN

Macrophages have important protective functions during infection with herpes simplex virus type 1 (HSV-1). However, molecular mechanisms that restrict viral propagation and protect from severe disease are unclear. Here we show that macrophages take up HSV-1 via endocytosis and transport the virions into multivesicular bodies (MVBs). In MVBs, acid ceramidase (aCDase) converts ceramide into sphingosine and increases the formation of sphingosine-rich intraluminal vesicles (ILVs). Once HSV-1 particles reach MVBs, sphingosine-rich ILVs bind to HSV-1 particles, which restricts fusion with the limiting endosomal membrane and prevents cellular infection. Lack of aCDase in macrophage cultures or in Asah1-/- mice results in replication of HSV-1 and Asah1-/- mice die soon after systemic or intravaginal inoculation. The treatment of macrophages with sphingosine enhancing compounds blocks HSV-1 propagation, suggesting a therapeutic potential of this pathway. In conclusion, aCDase loads ILVs with sphingosine, which prevents HSV-1 capsids from penetrating into the cytosol.


Asunto(s)
Ceramidasa Ácida/metabolismo , Herpes Simple/enzimología , Herpes Simple/prevención & control , Herpesvirus Humano 1/fisiología , Macrófagos/enzimología , Cuerpos Multivesiculares/virología , Ceramidasa Ácida/genética , Animales , Femenino , Herpes Simple/virología , Humanos , Macrófagos/virología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Replicación Viral
7.
Cancer Gene Ther ; 26(5-6): 174-178, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30393375

RESUMEN

We conducted a phase 1 trial for single-dose intravenous Ad5CRT, a replication-defective adenovirus vector expressing HSVtk (herpes simplex virus thymidine kinase) modulated by a specific trans-splicing ribozyme that targets human telomerase reverse transcriptase (hTERT)-encoding RNAs. Dose-limiting toxicities (DLTs) were evaluated in 15 patients at dose levels of 0.1-2 × 1012 virus particles. Patients well tolerated study treatment. During the DLT evaluation period, none of the 15 patients developed any grade 4 toxicities or treatment discontinuation that was related to agents investigated by this trial. The most frequent treatment-related adverse event was fever/chill (26.7%). Of the 18 patients, no patients achieved a partial or complete response, and the median progression-free survival for 18 patients was 1.1 months (95% CI, 1.0-1.3) and the results suggest no clinical benefit from this treatment. Ad5CRT's circulating virus half-life was approximately 10 min. Maximum tolerated dose was 2 × 1012 virus particles. Single-dose intravenous Ad5CRT was feasible and well tolerated in patients with gastrointestinal cancer liver metastasis. Ad5CRT did not provide meaningful clinical benefit, and the reason for the lack of efficacy was not entirely clear because no pharmocodynamic assessment was made.


Asunto(s)
Neoplasias Gastrointestinales/terapia , Terapia Genética/métodos , Neoplasias Hepáticas/secundario , Neoplasias Hepáticas/terapia , Adenoviridae/genética , Administración Intravenosa , Adulto , Anciano , Anciano de 80 o más Años , Femenino , Neoplasias Gastrointestinales/enzimología , Neoplasias Gastrointestinales/genética , Neoplasias Gastrointestinales/patología , Vectores Genéticos , Herpes Simple/enzimología , Herpes Simple/genética , Humanos , Neoplasias Hepáticas/enzimología , Neoplasias Hepáticas/genética , Masculino , Persona de Mediana Edad , Metástasis de la Neoplasia , Telomerasa/metabolismo , Timidina Quinasa/genética , Timidina Quinasa/metabolismo , Adulto Joven
8.
J Biol Chem ; 291(25): 13206-15, 2016 Jun 17.
Artículo en Inglés | MEDLINE | ID: mdl-27129230

RESUMEN

Viral infection or lipopolysaccharide (LPS) treatment induces expression of a large array of genes, the products of which play a critical role in host antipathogen immunity and inflammation. We have previously reported that the expression of ubiquitin-specific protease 25 (USP25) is significantly up-regulated after viral infection or LPS treatment, and this is essential for innate immune signaling. However, the mechanism behind this phenomenon is unclear. In this study, we found that viral infection-induced up-regulation of Usp25 is diminished in cells lacking interferon regulatory factor 7 (IRF7) or interferon α receptor 1 (IFNAR1) but not p65. Sendai virus- or type I interferon-induced up-regulation of Usp25 requires de novo protein synthesis of IRF7. Furthermore, IRF7 directly binds to the two conserved IRF binding sites on the USP25 promoter to drive transcription of Usp25, and mutation of these two sites abolished Sendai virus-induced IRF7-mediated activation of the USP25 promoter. Our study has uncovered a previously unknown mechanism by which viral infection or LPS induces up-regulation of USP25.


Asunto(s)
Factor 7 Regulador del Interferón/fisiología , Interferón Tipo I/fisiología , Ubiquitina Tiolesterasa/genética , Animales , Células Cultivadas , Inducción Enzimática/inmunología , Herpes Simple/enzimología , Herpesvirus Humano 1/fisiología , Lipopolisacáridos/farmacología , Ratones Noqueados , Regiones Promotoras Genéticas , Unión Proteica , Transducción de Señal , Transcripción Genética , Ubiquitina Tiolesterasa/metabolismo , Regulación hacia Arriba/inmunología
9.
Virus Res ; 205: 63-72, 2015 Jul 02.
Artículo en Inglés | MEDLINE | ID: mdl-26031763

RESUMEN

Herpes simplex virus type-1 (HSV-1) is ubiquitous and is able to establish a lifelong persistent latent infection in neurons of infected individuals. It has been estimated that in approximately 70% of the population over 50 years old, the virus enters the brain and infects neurons, and possibly undergoes recurrent reactivation episodes during lifetime, especially in immunodepressed individuals. We previously showed that the sensors AMP-dependent kinase (AMPK) and Sirtuin 1 (Sirt1), involved in survival pathways and neuroprotection, were affected during the course of HSV-1 infection. To evaluate if natural activators of the AMPK/Sirt1 axis, such as Resveratrol and Quercetin could reduce viral propagation and/or counteract the effects of neuronal infection, we analyzed progeny virion production, neuronal viability and neurodegenerative events during HSV-1 infection. We found that the activators of AMPK/Sirt1 axis, increased the viability of infected neurons, significantly reduced the viral titer in the supernatant and the expression of viral genes. More importantly, pretreatment of neurons with Resveratrol or Quercetin significantly reduced the levels of caspase-3 cleaved- and hyperphosphorylated tau associated with HSV-1 infection. These results suggest that activators of the AMPK/Sirt1 axis could be potentially useful in reducing the risk of HSV-1 productive infection in neurons and the cellular damage associated with reactivation episodes.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Suplementos Dietéticos/análisis , Activadores de Enzimas/farmacología , Herpes Simple/enzimología , Herpesvirus Humano 1/fisiología , Neuronas/virología , Sirtuina 1/metabolismo , Proteínas Quinasas Activadas por AMP/genética , Herpes Simple/genética , Herpes Simple/virología , Herpesvirus Humano 1/efectos de los fármacos , Herpesvirus Humano 1/genética , Humanos , Neuronas/efectos de los fármacos , Quercetina/farmacología , Resveratrol , Sirtuina 1/genética , Estilbenos/farmacología
10.
Dongwuxue Yanjiu ; 36(3): 142-51, 2015 May 18.
Artículo en Inglés | MEDLINE | ID: mdl-26018857

RESUMEN

Herpes simplex virus type 1 (HSV-1) enters productive infection after infecting epithelial cells, where it controls the host nucleus to make viral proteins, starts viral DNA synthesis and assembles infectious virions. In this process, replicating viral genomes are organized into replication centers to facilitate viral growth. HSV-1 is known to use host factors, including host chromatin and host transcription regulators, to transcribe its genes; however, the invading virus also encounters host defense and stress responses to inhibit viral growth. Recently, we found that HSV-1 replication centers recruit host factor CTCF but exclude γH2A.X. Thus, HSV-1 replication centers may selectively recruit cellular factors needed for viral growth, while excluding host factors that are deleterious for viral transcription or replication. Here we report that the viral replication centers selectively excluded modified histone H3, including heterochromatin mark H3K9me3, H3S10P and active chromatin mark H3K4me3, but not unmodified H3. We found a dynamic association between the viral replication centers and host RNA polymerase II. The centers also recruited components of the DNA damage response pathway, including 53BP1, BRCA1 and host antiviral protein SP100. Importantly, we found that ATM kinase was needed for the recruitment of CTCF to the viral centers. These results suggest that the HSV-1 replication centers took advantage of host signaling pathways to actively recruit or exclude host factors to benefit viral growth.


Asunto(s)
Replicación del ADN , Herpes Simple/metabolismo , Herpesvirus Humano 1/genética , Proteínas Virales/metabolismo , Antígenos Nucleares/genética , Antígenos Nucleares/metabolismo , Autoantígenos/genética , Autoantígenos/metabolismo , Factor de Unión a CCCTC , Línea Celular , Herpes Simple/enzimología , Herpes Simple/genética , Herpes Simple/virología , Herpesvirus Humano 1/metabolismo , Histonas/genética , Histonas/metabolismo , Interacciones Huésped-Patógeno , Humanos , Unión Proteica , ARN Polimerasa II/genética , ARN Polimerasa II/metabolismo , Proteínas Represoras/genética , Proteínas Represoras/metabolismo , Proteínas Virales/genética
11.
Nat Commun ; 6: 6985, 2015 Apr 27.
Artículo en Inglés | MEDLINE | ID: mdl-25912399

RESUMEN

Herpesviruses exemplified by herpes simplex virus-1 (HSV-1) attach to cell surface heparan sulfate (HS) for entry into host cells. However, during a productive infection, the HS moieties on parent cells can trap newly exiting viral progenies and inhibit their release. Here we demonstrate that a HS-degrading enzyme of the host, heparanase (HPSE), is upregulated through NF-kB and translocated to the cell surface upon HSV-1 infection for the removal of HS to facilitate viral release. We also find a significant increase in HPSE release in vivo during infection of murine corneas and that knockdown of HPSE in vivo inhibits virus shedding. Overall, we propose that HPSE acts as a molecular switch for turning a virus-permissive 'attachment mode' of host cells to a virus-deterring 'detachment mode'. Since many human viruses use HS as an attachment receptor, the HPSE-HS interplay may delineate a common mechanism for virus release.


Asunto(s)
Glucuronidasa/metabolismo , Heparitina Sulfato/metabolismo , Herpes Simple/enzimología , Herpesvirus Humano 1/fisiología , Virión/fisiología , Animales , Chlorocebus aethiops , Femenino , Células HEK293 , Células HeLa , Herpes Simple/virología , Interacciones Huésped-Patógeno , Humanos , Ratones , Ratones Endogámicos BALB C , Regulación hacia Arriba , Células Vero , Liberación del Virus
12.
J Virol ; 89(10): 5747-50, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25762734

RESUMEN

Herpes simplex virus 1 (HSV-1) can establish lifelong latency in human trigeminal ganglia. Latently infected ganglia contain CD8(+) T cells, which secrete granzyme B and are thus capable of inducing neuronal apoptosis. Using immunohistochemistry and single-cell reverse transcription-quantitative PCR (RT-qPCR), higher frequency and transcript levels of caspase-3 were found in HSV-1-negative compared to HSV-1-positive ganglia and neurons, respectively. No terminal deoxynucleotidyltransferase-mediated dUTP-biotin nick end labeling (TUNEL) assay-positive neurons were detected. The infiltrating T cells do not induce apoptosis in latently infected neurons.


Asunto(s)
Herpes Simple/patología , Herpes Simple/virología , Herpesvirus Humano 1/fisiología , Herpesvirus Humano 1/patogenicidad , Ganglio del Trigémino/virología , Apoptosis , Linfocitos T CD8-positivos/patología , Caspasa 3/metabolismo , Herpes Simple/enzimología , Herpesvirus Humano 1/genética , Interacciones Huésped-Patógeno , Humanos , MicroARNs/genética , MicroARNs/metabolismo , Neuronas/enzimología , Neuronas/patología , Neuronas/virología , Latencia del Virus
13.
J Virol ; 89(6): 3417-20, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25552720

RESUMEN

Herpes simplex virus 1 (HSV-1) genomes are associated with the repressive heterochromatic marks H3K9me2/me3 and H3K27me3 during latency. Previous studies have demonstrated that inhibitors of H3K9me2/me3 histone demethylases reduce the ability of HSV-1 to reactivate from latency. Here we demonstrate that GSK-J4, a specific inhibitor of the H3K27me3 histone demethylases UTX and JMJD3, inhibits HSV-1 reactivation from sensory neurons in vitro. These results indicate that removal of the H3K27me3 mark plays a key role in HSV-1 reactivation.


Asunto(s)
Herpes Simple/enzimología , Herpesvirus Humano 1/fisiología , Histona Demetilasas/metabolismo , Histonas/metabolismo , Histona Demetilasas con Dominio de Jumonji/metabolismo , Neuronas/metabolismo , Ganglio del Trigémino/metabolismo , Activación Viral , Secuencias de Aminoácidos , Animales , Regulación hacia Abajo/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Herpes Simple/genética , Herpes Simple/metabolismo , Herpes Simple/virología , Herpesvirus Humano 1/efectos de los fármacos , Herpesvirus Humano 1/genética , Histona Demetilasas/antagonistas & inhibidores , Histona Demetilasas/genética , Histonas/química , Histonas/genética , Humanos , Histona Demetilasas con Dominio de Jumonji/antagonistas & inhibidores , Histona Demetilasas con Dominio de Jumonji/genética , Metilación , Ratones , Neuronas/enzimología , Neuronas/virología , Ganglio del Trigémino/enzimología , Ganglio del Trigémino/virología , Activación Viral/efectos de los fármacos , Latencia del Virus/efectos de los fármacos
14.
Virology ; 449: 88-95, 2014 Jan 20.
Artículo en Inglés | MEDLINE | ID: mdl-24418541

RESUMEN

Actin and its regulators are critical for neuronal function. Infection with herpes simplex virus 1 (HSV-1) remodels neuronal cell actin dynamics, which may relate virus-induced pathological processes in the nervous system. We previously demonstrated that cofilin is an actin regulator that participates in HSV-1-induced actin dynamics in neuronal cells, but how HSV-1 regulates cofilin has remained unclear. In the present study, we demonstrated the HSV-1-induced the inactivation of cofilin and the accumulation of phosphorylated cofilin in the nucleus, which together benefited viral replication. This consistent cofilin inactivation was achieved by the downregulation of slingshot 1 (SSH1). Notably, virus-induced SSH1 downregulation depended on the ubiquitin-proteasome system. Cofilin inactivation is therefore critical for HSV-1 replication during neuronal infection and is maintained by SSH1 downregulation. Moreover, these results provide new insight into the HSV-1-induced neurological pathogenesis and suggest potential new strategies to inhibit HSV-1 replication.


Asunto(s)
Cofilina 1/metabolismo , Herpes Simple/enzimología , Herpesvirus Humano 1/fisiología , Neuronas/virología , Complejo de la Endopetidasa Proteasomal/metabolismo , Ubiquitina/metabolismo , Replicación Viral , Núcleo Celular/genética , Núcleo Celular/metabolismo , Cofilina 1/genética , Regulación hacia Abajo , Herpes Simple/genética , Herpes Simple/virología , Herpesvirus Humano 1/genética , Humanos , Neuronas/metabolismo , Fosfoproteínas Fosfatasas , Transporte de Proteínas
15.
Antivir Ther ; 19(1): 15-29, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-23813409

RESUMEN

BACKGROUND: Herpes simplex virus type-1 (HSV-1) is the leading cause of infectious blindness worldwide. Through a multistep process, HSV-1 enters into naturally susceptible human corneal epithelial (HCE) cells where it establishes an optimal environment for viral replication and spread. HSV-1 employment of cytoskeletal proteins, kinases, and cell signalling pathways is crucial for the entry process. METHODS: Here we demonstrate that non-muscle myosin IIA (NM-IIA) and/or a myosin activating kinase, myosin light chain kinase (MLCK), can be targeted for the development of new and effective therapies against HSV-1. HCE cells were incubated with MLCK inhibitors ML-7 and ML-9 and NM-IIA inhibitor blebbistatin. Following the application of inhibitors, HSV-1 entry and spread to neighbouring HCE cells was evaluated. RESULTS: Upon application of MLCK inhibitors ML-7 and ML-9 and NM-IIA inhibitor blebbistatin, HSV-1 entry into HCE cells was significantly decreased. Furthermore, dramatic impairment of glycoprotein-mediated membrane fusion was seen in cells treated with MLCK inhibitors, thus establishing a role for MLCK activation in cell-to-cell fusion and multinucleated syncytial cell formation. These results also indicate that the activation of motor protein NM-IIA by MLCK is crucial for cytoskeletal changes required for HSV-1 infection of corneal cells. CONCLUSIONS: We provide new evidence that NM-IIA and MLCK can be used as effective antiviral targets against ocular herpes.


Asunto(s)
Antivirales/farmacología , Herpes Simple/enzimología , Herpesvirus Humano 1/efectos de los fármacos , Herpesvirus Humano 1/fisiología , Quinasa de Cadena Ligera de Miosina/antagonistas & inhibidores , Animales , Antivirales/uso terapéutico , Línea Celular , Células Epiteliales/metabolismo , Células Epiteliales/virología , Espacio Extracelular/metabolismo , Células Gigantes/efectos de los fármacos , Herpes Simple/tratamiento farmacológico , Humanos , Espacio Intracelular/metabolismo , Proteínas Motoras Moleculares/antagonistas & inhibidores , Proteínas Motoras Moleculares/metabolismo , Cadenas Pesadas de Miosina/antagonistas & inhibidores , Cadenas Pesadas de Miosina/metabolismo , Unión Proteica , Ensayo de Placa Viral , Internalización del Virus/efectos de los fármacos
16.
Proc Natl Acad Sci U S A ; 110(51): E5006-15, 2013 Dec 17.
Artículo en Inglés | MEDLINE | ID: mdl-24297925

RESUMEN

Herpes simplex virus 1 (HSV-1) infection triggers specific metabolic changes in its host cell. To explore the interactions between cellular metabolism and HSV-1 infection, we performed an siRNA screen of cellular metabolic genes, measuring their effect on viral replication. The screen identified multiple enzymes predicted to influence HSV-1 replication, including argininosuccinate synthetase 1 (AS1), which consumes aspartate as part of de novo arginine synthesis. Knockdown of AS1 robustly enhanced viral genome replication and the production of infectious virus. Using high-resolution liquid chromatography-mass spectrometry, we found that the metabolic phenotype induced by knockdown of AS1 in human fibroblasts mimicked multiple aspects of the metabolic program observed during HSV-1 infection, including an increase in multiple nucleotides and their precursors. Together with the observation that AS1 protein and mRNA levels decrease during wild-type infection, this work suggests that reduced AS1 activity is partially responsible for the metabolic program induced by infection.


Asunto(s)
Argininosuccinato Sintasa/metabolismo , Fibroblastos/enzimología , Herpes Simple/enzimología , Herpesvirus Humano 1/fisiología , Replicación Viral/fisiología , Animales , Argininosuccinato Sintasa/genética , Chlorocebus aethiops , Fibroblastos/patología , Fibroblastos/virología , Técnicas de Silenciamiento del Gen , Genoma Viral/fisiología , Herpes Simple/genética , Herpes Simple/patología , Humanos , ARN Mensajero/genética , ARN Mensajero/metabolismo , Células Vero
17.
Antiviral Res ; 100(1): 44-53, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23867132

RESUMEN

Pyrithione (PT), known as a zinc ionophore, is effective against several pathogens from the Streptococcus and Staphylococcus genera. The antiviral activity of PT was also reported against a number of RNA viruses. In this paper, we showed that PT could effectively inhibit herpes simplex virus types 1 and 2 (HSV-1 and HSV-2). PT inhibited HSV late gene (Glycoprotein D, gD) expression and the production of viral progeny, and this action was dependent on Zn(2+). Further studies showed that PT suppressed the expression of HSV immediate early (IE) gene, the infected cell polypeptide 4 (ICP4), but had less effect on another regulatory IE protein, ICP0. It was found that PT treatment could interfere with cellular ubiquitin-proteasome system (UPS), leading to the inhibition of HSV-2-induced IκB-α degradation to inhibit NF-κB activation and enhanced promyelocytic leukemia protein (PML) stability in nucleus. However, PT did not show direct inhibition of 26S proteasome activity. Instead, it induced Zn(2+) influx, which facilitated the dysregulation of UPS and the accumulation of intracellular ubiquitin-conjugates. UPS inhibition by PT caused disruption of IκB-α degradation and NF-κB activation thus leading to marked reduction of viral titer.


Asunto(s)
Herpes Simple/enzimología , Herpesvirus Humano 1/efectos de los fármacos , Herpesvirus Humano 2/efectos de los fármacos , Ionóforos/farmacología , FN-kappa B/genética , Complejo de la Endopetidasa Proteasomal/metabolismo , Piridinas/farmacología , Tionas/farmacología , Zinc/farmacología , Antivirales/farmacología , Regulación hacia Abajo/efectos de los fármacos , Herpes Simple/genética , Herpes Simple/metabolismo , Herpes Simple/virología , Herpesvirus Humano 1/genética , Herpesvirus Humano 1/fisiología , Herpesvirus Humano 2/genética , Herpesvirus Humano 2/fisiología , Humanos , FN-kappa B/metabolismo , Complejo de la Endopetidasa Proteasomal/genética , Replicación Viral/efectos de los fármacos
18.
Molecules ; 18(5): 5104-24, 2013 May 02.
Artículo en Inglés | MEDLINE | ID: mdl-23644977

RESUMEN

The efficient syntheses of 5-(2-hydroxyethyl)- and 5-(3-hydroxypropyl)-substituted pyrimidine derivatives bearing 2,3-dihydroxypropyl, acyclovir-, ganciclovir- and penciclovir-like side chains are reported. A synthetic approach that included the alkylation of an N-anionic-5-substituted pyrimidine intermediate (method A) provided the target acyclonucleosides in significantly higher overall yields in comparison to those obtained by method B using sylilation reaction. The phosphorylation assays of novel compounds as potential substrates for thymidine kinase of herpes simplex virus type 1 (HSV-1 TK) showed that solely pyrimidine 5-substituted acyclonucleosides with a penciclovir-like side chain acted as a fraudulent substrates of HSV-1 TK. Moreover, the uracil derivative with penciclovir-like side chain with less bulky 2-hydroxyethyl substituent at C-5 proved to be a better substrate than the corresponding one with a 3-hydroxypropyl substituent. Therefore, this acyclonucleoside was selected as a lead compound for the development of a positron emission tomography HSV-1 TK activity imaging agent.


Asunto(s)
Aciclovir/análogos & derivados , Antivirales , Ganciclovir , Herpesvirus Humano 1/enzimología , Nucleósidos de Pirimidina , Timidina Quinasa/metabolismo , Aciclovir/síntesis química , Aciclovir/química , Aciclovir/farmacología , Línea Celular , Fibroblastos , Ganciclovir/síntesis química , Ganciclovir/química , Ganciclovir/farmacología , Guanina , Herpes Simple/diagnóstico por imagen , Herpes Simple/enzimología , Humanos , Tomografía de Emisión de Positrones/métodos , Nucleósidos de Pirimidina/síntesis química , Nucleósidos de Pirimidina/química , Nucleósidos de Pirimidina/farmacología , Radiografía
19.
Curr Opin Hematol ; 19(6): 427-33, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22918402

RESUMEN

PURPOSE OF REVIEW: Preserving the beneficial effects of donor T cells against tumor and pathogens while avoiding noxious graft-versus-host disease (GvHD) is the 'holy grail' of allogeneic hematopoietic stem cell transplantation (HSCT). The suicide gene strategy allows the selective elimination of genetically modified donor T cells during GvHD. This review summarizes the results obtained in recent years in the clinical trials of suicide gene therapy using the paradigmatic herpes simplex virus thymidine kinase (TK) suicide gene. RECENT FINDINGS: T cells genetically modified to express the TK suicide gene, TK-cells, are safe and preserve most of their functional features; when infused into patients they are capable of conferring substantial protection against infections and tumor recurrence, and are promptly eliminated in the case of GvHD, with complete resolution of the adverse reaction in all treated cases. Unexpectedly, TK-cells also have the indirect effect of promoting patient thymopoiesis, contributing to the renewal of a host-tolerant immune repertoire. SUMMARY: Suicide gene therapy with TK-cells is a promising approach to overcome the risk of GvHD in allogeneic HSCT, especially from partially incompatible donors, and is currently under evaluation in a multicentric phase III clinical trial.


Asunto(s)
Terapia Genética/métodos , Trasplante de Células Madre Hematopoyéticas/métodos , Linfocitos T/trasplante , Timidina Quinasa/genética , Animales , Genes Transgénicos Suicidas , Enfermedad Injerto contra Huésped/tratamiento farmacológico , Enfermedad Injerto contra Huésped/prevención & control , Haploidia , Herpes Simple/enzimología , Herpes Simple/genética , Humanos , Timidina Quinasa/biosíntesis
20.
J Virol ; 86(15): 8259-68, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22623791

RESUMEN

Herpes simplex virus 1 infection triggers multiple changes in the metabolism of host cells, including a dramatic decrease in the levels of NAD(+). In addition to its role as a cofactor in reduction-oxidation reactions, NAD(+) is required for certain posttranslational modifications. Members of the poly(ADP-ribose) polymerase (PARP) family of enzymes are major consumers of NAD(+), which they utilize to form poly(ADP-ribose) (PAR) chains on protein substrates in response to DNA damage. PAR chains can subsequently be removed by the enzyme poly(ADP-ribose) glycohydrolase (PARG). We report here that the HSV-1 infection-induced drop in NAD(+) levels required viral DNA replication, was associated with an increase in protein poly(ADP-ribosyl)ation (PARylation), and was blocked by pharmacological inhibition of PARP-1/PARP-2 (PARP-1/2). Neither virus yield nor the cellular metabolic reprogramming observed during HSV-1 infection was altered by the rescue or further depletion of NAD(+) levels. Expression of the viral protein ICP0, which possesses E3 ubiquitin ligase activity, was both necessary and sufficient for the degradation of the 111-kDa PARG isoform. This work demonstrates that HSV-1 infection results in changes to NAD(+) metabolism by PARP-1/2 and PARG, and as PAR chain accumulation can induce caspase-independent apoptosis, we speculate that the decrease in PARG levels enhances the auto-PARylation-mediated inhibition of PARP, thereby avoiding premature death of the infected cell.


Asunto(s)
Glicósido Hidrolasas/metabolismo , Herpes Simple/enzimología , Herpesvirus Humano 1/metabolismo , Poli(ADP-Ribosa) Polimerasas/metabolismo , Proteolisis , Células Cultivadas , Replicación del ADN/genética , ADN Viral/genética , ADN Viral/metabolismo , Glicósido Hidrolasas/genética , Herpes Simple/genética , Herpesvirus Humano 1/genética , Humanos , Proteínas Inmediatas-Precoces/genética , Proteínas Inmediatas-Precoces/metabolismo , NAD/genética , NAD/metabolismo , Poli(ADP-Ribosa) Polimerasa-1 , Poli(ADP-Ribosa) Polimerasas/genética , Ubiquitina-Proteína Ligasas/genética , Ubiquitina-Proteína Ligasas/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA