Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 146
Filtrar
1.
Angew Chem Int Ed Engl ; 62(41): e202309838, 2023 10 09.
Artigo em Inglês | MEDLINE | ID: mdl-37555536

RESUMO

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.


Assuntos
Glucuronidase , Herpes Simples , Herpesvirus Humano 1 , Humanos , Glucuronidase/antagonistas & inibidores , Glucuronidase/metabolismo , Heparitina Sulfato/farmacologia , Herpes Simples/enzimologia , Herpes Simples/virologia , Herpesvirus Humano 1/metabolismo , Oligossacarídeos/farmacologia , Oligossacarídeos/metabolismo
2.
PLoS One ; 17(2): e0264001, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35167620

RESUMO

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.


Assuntos
Neoplasias Encefálicas/terapia , Ganciclovir/administração & dosagem , Glioblastoma/terapia , Herpes Simples/enzimologia , Transplante de Células-Tronco Mesenquimais/métodos , Células-Tronco Mesenquimais/citologia , Timidina Quinase/genética , Animais , Neoplasias Encefálicas/genética , Linhagem Celular Tumoral , Técnicas de Cocultura , Cães , Ganciclovir/farmacologia , Genes Transgênicos Suicidas , Terapia Genética , Glioblastoma/genética , Herpes Simples/genética , Humanos , Lentivirus/genética , Células-Tronco Mesenquimais/metabolismo , Camundongos , Timidina Quinase/metabolismo , Transdução Genética , Proteínas Virais/genética , Proteínas Virais/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto
3.
mBio ; 12(6): e0279221, 2021 12 21.
Artigo em Inglês | MEDLINE | ID: mdl-34749529

RESUMO

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.


Assuntos
Glucuronidase/metabolismo , Herpes Simples/enzimologia , Herpesvirus Humano 1/fisiologia , Proteínas Proto-Oncogênicas c-akt/metabolismo , beta Catenina/metabolismo , Motivos de Aminoácidos , Linhagem Celular , Glucuronidase/genética , Herpes Simples/genética , Herpes Simples/metabolismo , Herpes Simples/virologia , Herpesvirus Humano 1/genética , Interações Hospedeiro-Patógeno , Humanos , Fosforilação , Proteínas Proto-Oncogênicas c-akt/genética , Ativação Viral , Replicação Viral , Via de Sinalização Wnt , beta Catenina/química , beta Catenina/genética
4.
mBio ; 12(2)2021 03 30.
Artigo em Inglês | MEDLINE | ID: mdl-33785613

RESUMO

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.


Assuntos
Amidofosforribosiltransferase/metabolismo , Proteína DEAD-box 58/metabolismo , Herpes Simples/enzimologia , Herpesvirus Humano 1/enzimologia , Proteínas Estruturais Virais/metabolismo , Replicação Viral , Amidofosforribosiltransferase/genética , Motivos de Aminoácidos , Animais , Proteína DEAD-box 58/química , Proteína DEAD-box 58/genética , Herpes Simples/genética , Herpes Simples/virologia , Herpesvirus Humano 1/genética , Herpesvirus Humano 1/fisiologia , Interações Hospedeiro-Patógeno , Humanos , Camundongos , Ligação Proteica , Especificidade da Espécie , Proteínas Estruturais Virais/genética
5.
Biochem J ; 478(1): 261-279, 2021 01 15.
Artigo em Inglês | MEDLINE | ID: mdl-33355669

RESUMO

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.


Assuntos
Antivirais/metabolismo , Herpes Simples/metabolismo , Herpesvirus Humano 1/metabolismo , Interações Hospedeiro-Patógeno/efeitos dos fármacos , Enzimas Ativadoras de Ubiquitina/metabolismo , Animais , Sobrevivência Celular/genética , Chlorocebus aethiops , Cromatografia Líquida , Doxiciclina/farmacologia , Células HeLa , Herpes Simples/enzimologia , Herpes Simples/genética , Herpesvirus Humano 1/genética , Interações Hospedeiro-Patógeno/genética , Humanos , Ligação Proteica , Domínios Proteicos , Processamento de Proteína Pós-Traducional/efeitos dos fármacos , RNA Interferente Pequeno , Espectrometria de Massas em Tandem , Tetraciclina/farmacologia , Transfecção , Enzimas Ativadoras de Ubiquitina/genética , Ubiquitinação/efeitos dos fármacos , Células Vero , Replicação Viral/efeitos dos fármacos
6.
Nat Commun ; 11(1): 1338, 2020 03 12.
Artigo em Inglês | MEDLINE | ID: mdl-32165633

RESUMO

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.


Assuntos
Ceramidase Ácida/metabolismo , Herpes Simples/enzimologia , Herpes Simples/prevenção & controle , Herpesvirus Humano 1/fisiologia , Macrófagos/enzimologia , Corpos Multivesiculares/virologia , Ceramidase Ácida/genética , Animais , Feminino , Herpes Simples/virologia , Humanos , Macrófagos/virologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Replicação Viral
7.
Cancer Gene Ther ; 26(5-6): 174-178, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-30393375

RESUMO

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.


Assuntos
Neoplasias Gastrointestinais/terapia , Terapia Genética/métodos , Neoplasias Hepáticas/secundário , Neoplasias Hepáticas/terapia , Adenoviridae/genética , Administração Intravenosa , Adulto , Idoso , Idoso de 80 Anos ou mais , Feminino , Neoplasias Gastrointestinais/enzimologia , Neoplasias Gastrointestinais/genética , Neoplasias Gastrointestinais/patologia , Vetores Genéticos , Herpes Simples/enzimologia , Herpes Simples/genética , Humanos , Neoplasias Hepáticas/enzimologia , Neoplasias Hepáticas/genética , Masculino , Pessoa de Meia-Idade , Metástase Neoplásica , Telomerase/metabolismo , Timidina Quinase/genética , Timidina Quinase/metabolismo , Adulto Jovem
8.
J Biol Chem ; 291(25): 13206-15, 2016 Jun 17.
Artigo em Inglês | MEDLINE | ID: mdl-27129230

RESUMO

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.


Assuntos
Fator Regulador 7 de Interferon/fisiologia , Interferon Tipo I/fisiologia , Ubiquitina Tiolesterase/genética , Animais , Células Cultivadas , Indução Enzimática/imunologia , Herpes Simples/enzimologia , Herpesvirus Humano 1/fisiologia , Lipopolissacarídeos/farmacologia , Camundongos Knockout , Regiões Promotoras Genéticas , Ligação Proteica , Transdução de Sinais , Transcrição Gênica , Ubiquitina Tiolesterase/metabolismo , Regulação para Cima/imunologia
9.
Virus Res ; 205: 63-72, 2015 Jul 02.
Artigo em Inglês | MEDLINE | ID: mdl-26031763

RESUMO

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.


Assuntos
Proteínas Quinases Ativadas por AMP/metabolismo , Suplementos Nutricionais/análise , Ativadores de Enzimas/farmacologia , Herpes Simples/enzimologia , Herpesvirus Humano 1/fisiologia , Neurônios/virologia , Sirtuína 1/metabolismo , Proteínas Quinases Ativadas por AMP/genética , Herpes Simples/genética , Herpes Simples/virologia , Herpesvirus Humano 1/efeitos dos fármacos , Herpesvirus Humano 1/genética , Humanos , Neurônios/efeitos dos fármacos , Quercetina/farmacologia , Resveratrol , Sirtuína 1/genética , Estilbenos/farmacologia
10.
Dongwuxue Yanjiu ; 36(3): 142-51, 2015 May 18.
Artigo em Inglês | MEDLINE | ID: mdl-26018857

RESUMO

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.


Assuntos
Replicação do DNA , Herpes Simples/metabolismo , Herpesvirus Humano 1/genética , Proteínas Virais/metabolismo , Antígenos Nucleares/genética , Antígenos Nucleares/metabolismo , Autoantígenos/genética , Autoantígenos/metabolismo , Fator de Ligação a CCCTC , Linhagem Celular , Herpes Simples/enzimologia , Herpes Simples/genética , Herpes Simples/virologia , Herpesvirus Humano 1/metabolismo , Histonas/genética , Histonas/metabolismo , Interações Hospedeiro-Patógeno , Humanos , Ligação Proteica , RNA Polimerase II/genética , RNA Polimerase II/metabolismo , Proteínas Repressoras/genética , Proteínas Repressoras/metabolismo , Proteínas Virais/genética
11.
Nat Commun ; 6: 6985, 2015 Apr 27.
Artigo em Inglês | MEDLINE | ID: mdl-25912399

RESUMO

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.


Assuntos
Glucuronidase/metabolismo , Heparitina Sulfato/metabolismo , Herpes Simples/enzimologia , Herpesvirus Humano 1/fisiologia , Vírion/fisiologia , Animais , Chlorocebus aethiops , Feminino , Células HEK293 , Células HeLa , Herpes Simples/virologia , Interações Hospedeiro-Patógeno , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Regulação para Cima , Células Vero , Liberação de Vírus
12.
J Virol ; 89(10): 5747-50, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25762734

RESUMO

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.


Assuntos
Herpes Simples/patologia , Herpes Simples/virologia , Herpesvirus Humano 1/fisiologia , Herpesvirus Humano 1/patogenicidade , Gânglio Trigeminal/virologia , Apoptose , Linfócitos T CD8-Positivos/patologia , Caspase 3/metabolismo , Herpes Simples/enzimologia , Herpesvirus Humano 1/genética , Interações Hospedeiro-Patógeno , Humanos , MicroRNAs/genética , MicroRNAs/metabolismo , Neurônios/enzimologia , Neurônios/patologia , Neurônios/virologia , Latência Viral
13.
J Virol ; 89(6): 3417-20, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25552720

RESUMO

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.


Assuntos
Herpes Simples/enzimologia , Herpesvirus Humano 1/fisiologia , Histona Desmetilases/metabolismo , Histonas/metabolismo , Histona Desmetilases com o Domínio Jumonji/metabolismo , Neurônios/metabolismo , Gânglio Trigeminal/metabolismo , Ativação Viral , Motivos de Aminoácidos , Animais , Regulação para Baixo/efeitos dos fármacos , Inibidores Enzimáticos/farmacologia , Herpes Simples/genética , Herpes Simples/metabolismo , Herpes Simples/virologia , Herpesvirus Humano 1/efeitos dos fármacos , Herpesvirus Humano 1/genética , Histona Desmetilases/antagonistas & inibidores , Histona Desmetilases/genética , Histonas/química , Histonas/genética , Humanos , Histona Desmetilases com o Domínio Jumonji/antagonistas & inibidores , Histona Desmetilases com o Domínio Jumonji/genética , Metilação , Camundongos , Neurônios/enzimologia , Neurônios/virologia , Gânglio Trigeminal/enzimologia , Gânglio Trigeminal/virologia , Ativação Viral/efeitos dos fármacos , Latência Viral/efeitos dos fármacos
14.
Virology ; 449: 88-95, 2014 Jan 20.
Artigo em Inglês | MEDLINE | ID: mdl-24418541

RESUMO

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.


Assuntos
Cofilina 1/metabolismo , Herpes Simples/enzimologia , Herpesvirus Humano 1/fisiologia , Neurônios/virologia , Complexo de Endopeptidases do Proteassoma/metabolismo , Ubiquitina/metabolismo , Replicação Viral , Núcleo Celular/genética , Núcleo Celular/metabolismo , Cofilina 1/genética , Regulação para Baixo , Herpes Simples/genética , Herpes Simples/virologia , Herpesvirus Humano 1/genética , Humanos , Neurônios/metabolismo , Fosfoproteínas Fosfatases , Transporte Proteico
15.
Antivir Ther ; 19(1): 15-29, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-23813409

RESUMO

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.


Assuntos
Antivirais/farmacologia , Herpes Simples/enzimologia , Herpesvirus Humano 1/efeitos dos fármacos , Herpesvirus Humano 1/fisiologia , Quinase de Cadeia Leve de Miosina/antagonistas & inibidores , Animais , Antivirais/uso terapêutico , Linhagem Celular , Células Epiteliais/metabolismo , Células Epiteliais/virologia , Espaço Extracelular/metabolismo , Células Gigantes/efeitos dos fármacos , Herpes Simples/tratamento farmacológico , Humanos , Espaço Intracelular/metabolismo , Proteínas Motores Moleculares/antagonistas & inibidores , Proteínas Motores Moleculares/metabolismo , Cadeias Pesadas de Miosina/antagonistas & inibidores , Cadeias Pesadas de Miosina/metabolismo , Ligação Proteica , Ensaio de Placa Viral , Internalização do Vírus/efeitos dos fármacos
16.
Proc Natl Acad Sci U S A ; 110(51): E5006-15, 2013 Dec 17.
Artigo em Inglês | MEDLINE | ID: mdl-24297925

RESUMO

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.


Assuntos
Argininossuccinato Sintase/metabolismo , Fibroblastos/enzimologia , Herpes Simples/enzimologia , Herpesvirus Humano 1/fisiologia , Replicação Viral/fisiologia , Animais , Argininossuccinato Sintase/genética , Chlorocebus aethiops , Fibroblastos/patologia , Fibroblastos/virologia , Técnicas de Silenciamento de Genes , Genoma Viral/fisiologia , Herpes Simples/genética , Herpes Simples/patologia , Humanos , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Células Vero
17.
Antiviral Res ; 100(1): 44-53, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23867132

RESUMO

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.


Assuntos
Herpes Simples/enzimologia , Herpesvirus Humano 1/efeitos dos fármacos , Herpesvirus Humano 2/efeitos dos fármacos , Ionóforos/farmacologia , NF-kappa B/genética , Complexo de Endopeptidases do Proteassoma/metabolismo , Piridinas/farmacologia , Tionas/farmacologia , Zinco/farmacologia , Antivirais/farmacologia , Regulação para Baixo/efeitos dos fármacos , Herpes Simples/genética , Herpes Simples/metabolismo , Herpes Simples/virologia , Herpesvirus Humano 1/genética , Herpesvirus Humano 1/fisiologia , Herpesvirus Humano 2/genética , Herpesvirus Humano 2/fisiologia , Humanos , NF-kappa B/metabolismo , Complexo de Endopeptidases do Proteassoma/genética , Replicação Viral/efeitos dos fármacos
18.
Molecules ; 18(5): 5104-24, 2013 May 02.
Artigo em Inglês | MEDLINE | ID: mdl-23644977

RESUMO

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.


Assuntos
Aciclovir/análogos & derivados , Antivirais , Ganciclovir , Herpesvirus Humano 1/enzimologia , Nucleosídeos de Pirimidina , Timidina Quinase/metabolismo , Aciclovir/síntese química , Aciclovir/química , Aciclovir/farmacologia , Linhagem Celular , Fibroblastos , Ganciclovir/síntese química , Ganciclovir/química , Ganciclovir/farmacologia , Guanina , Herpes Simples/diagnóstico por imagem , Herpes Simples/enzimologia , Humanos , Tomografia por Emissão de Pósitrons/métodos , Nucleosídeos de Pirimidina/síntese química , Nucleosídeos de Pirimidina/química , Nucleosídeos de Pirimidina/farmacologia , Radiografia
19.
Curr Opin Hematol ; 19(6): 427-33, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-22918402

RESUMO

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.


Assuntos
Terapia Genética/métodos , Transplante de Células-Tronco Hematopoéticas/métodos , Linfócitos T/transplante , Timidina Quinase/genética , Animais , Genes Transgênicos Suicidas , Doença Enxerto-Hospedeiro/tratamento farmacológico , Doença Enxerto-Hospedeiro/prevenção & controle , Haploidia , Herpes Simples/enzimologia , Herpes Simples/genética , Humanos , Timidina Quinase/biossíntese
20.
Biofizika ; 57(2): 232-42, 2012.
Artigo em Russo | MEDLINE | ID: mdl-22594278

RESUMO

Data obtained show that antiviral activities of bis-linked netropsin derivatives are targeted by specific complexes formed by helicase UL9 of herpes simplex virus type 1 with viral DNA replication origins, represented by two OriS sites and one OriL site. According to the results of footprinting studies bis-netropsins get bound selectively to an A+T-cluster which separates interaction sites I and II for helicase UL9 in OriS. Upon binding to DNA bis-netropsins stabilize a structure of the A+T-cluster and inhibit thermal fluctuation-induced opening of AT- base pairs which is needed for local unwinding of DNA by helicase UL9. Kinetics of ATP-dependent DNA unwinding in the presence and absence of Pt-bis-netropsin are studied by measuring the efficiency of Forster resonance energy transfer (FRET) between the fluorescent probes attached covalently to 3?- and 5?-ends of the oligonucleotides in the minimal OriS duplex. Pt-bis-netropsin and related molecules inhibit unwinding of OriS duplex by helicase UL9. Pt-bis-netropsin is also able to reduce the rate of unwinding of the AT- rich hairpin formed by the upper strand in the minimal OriS duplex. The antiviral activities and toxicity of bis-linked netropsin derivatives are studied in cell cultured experiments and experiments with animals infected by herpes virus.


Assuntos
Antivirais/farmacologia , Replicação do DNA/efeitos dos fármacos , DNA Viral/metabolismo , Proteínas de Ligação a DNA , Herpes Simples , Herpesvirus Humano 1/enzimologia , Netropsina/farmacologia , Proteínas Virais , Animais , Chlorocebus aethiops , Proteínas de Ligação a DNA/antagonistas & inibidores , Proteínas de Ligação a DNA/metabolismo , Herpes Simples/tratamento farmacológico , Herpes Simples/enzimologia , Camundongos , Camundongos Endogâmicos BALB C , Netropsina/análogos & derivados , Células Vero , Proteínas Virais/antagonistas & inibidores , Proteínas Virais/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...