Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
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
2.
Ocul Surf ; 17(1): 40-49, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30317007

RESUMO

Herpes simplex virus type 1 (HSV) keratitis is a leading cause of infectious blindness. Clinical disease occurs variably throughout the cornea from epithelium to endothelium and recurrent HSV stromal keratitis is associated with corneal scarring and neovascularization. HSV keratitis can be associated with ocular pain and subsequent neutrophic keratopathy. Host cell interactions with HSV trigger an inflammatory cascade responsible not only for clearance of virus but also for progressive corneal opacification due to inflammatory cell infiltrate, angiogenesis, and corneal nerve loss. Current antiviral therapies target viral replication to decrease disease duration, severity and recurrence, but there are limitations to these agents. Therapies directed towards viral entry into cells, protein synthesis, inflammatory cytokines and vascular endothelial growth factor pathways in animal models represent promising new approaches to the treatment of recurrent HSV keratitis.


Assuntos
Córnea/patologia , Infecções Oculares Virais/patologia , Imunidade Celular , Inflamação/patologia , Ceratite Herpética/patologia , Simplexvirus/patogenicidade , Animais , Córnea/virologia , DNA Viral/análise , Infecções Oculares Virais/virologia , Interações Hospedeiro-Patógeno , Humanos , Ceratite Herpética/virologia
3.
J Virol ; 92(23)2018 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-30232188

RESUMO

Herpes simplex virus 2 (HSV-2) can productively infect many different cell types of human and nonhuman origin. Here we demonstrate interconnected roles for two host enzymes, heparanase (HPSE) and cathepsin L, in HSV-2 release from cells. In vaginal epithelial cells, HSV-2 causes heparan sulfate shedding and upregulation in HPSE levels during the productive phase of infection. We also noted increased levels of cathepsin L and show that regulation of HPSE by cathepsin L via cleavage of HPSE proenzyme is important for infection. Furthermore, inhibition of HPSE by a specific inhibitor, OGT 2115, dramatically reduces HSV-2 release from vaginal epithelial cells. Likewise, we show evidence that the inhibition of cathepsin L is detrimental to the infection. The HPSE increase after infection is mediated by an increased NF-κB nuclear localization and a resultant activation of HPSE transcription. Together these mechanisms contribute to the removal of heparan sulfate from the cell surface and thus facilitate virus release from cells.IMPORTANCE Genital infections by HSV-2 represent one of the most common sexually transmitted viral infections. The virus causes painful lesions and sores around the genitals or rectum. Intermittent release of the virus from infected tissues during sexual activities is the most common cause of transmission. At the molecular level, cell surface heparan sulfate (HS) is known to provide attachment sites for HSV-2. While the removal of HS during HSV-1 release has been shown, not much is known about the host factors and their regulators that contribute to HSV-2 release from natural target cell types. Here we suggest a role for the host enzyme heparanase in HSV-2 release. Our work reveals that in addition to the regulation of transcription by NF-κB, HPSE is also regulated posttranslationally by cathepsin L and that inhibition of heparanase activity directly affects HSV-2 release. We provide unique insights into the host mechanisms controlling HSV-2 egress and spread.


Assuntos
Catepsina L/metabolismo , Glucuronidase/metabolismo , Herpes Simples/virologia , Herpesvirus Humano 2/fisiologia , Vagina/virologia , Liberação de Vírus , Animais , Catepsina L/genética , Células Cultivadas , Chlorocebus aethiops , Feminino , Glucuronidase/genética , Herpes Simples/genética , Herpes Simples/metabolismo , Interações Hospedeiro-Patógeno , Humanos , Regulação para Cima , Vagina/metabolismo , Vagina/patologia , Células Vero , Replicação Viral
4.
Cell Rep ; 20(2): 439-450, 2017 07 11.
Artigo em Inglês | MEDLINE | ID: mdl-28700944

RESUMO

Herpes simplex virus-1 (HSV-1) causes lifelong recurrent pathologies without a cure. How infection by HSV-1 triggers disease processes, especially in the immune-privileged avascular human cornea, remains a major unresolved puzzle. It has been speculated that a cornea-resident molecule must tip the balance in favor of pro-inflammatory and pro-angiogenic conditions observed with herpetic, as well as non-herpetic, ailments of the cornea. Here, we demonstrate that heparanase (HPSE), a host enzyme, is the molecular trigger for multiple pathologies associated with HSV-1 infection. In human corneal epithelial cells, HSV-1 infection upregulates HPSE in a manner dependent on HSV-1 infected cell protein 34.5. HPSE then relocates to the nucleus to regulate cytokine production, inhibits wound closure, enhances viral spread, and thus generates a toxic local environment. Overall, our findings implicate activated HPSE as a driver of viral pathogenesis and call for further attention to this host protein in infection and other inflammatory disorders.


Assuntos
Glucuronidase/metabolismo , Herpesvirus Humano 1/patogenicidade , Ativação Viral/fisiologia , Animais , Linhagem Celular , Feminino , Citometria de Fluxo , Células HeLa , Herpes Simples/metabolismo , Humanos , Imuno-Histoquímica , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Microscopia de Fluorescência , Cicatrização/fisiologia
5.
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
6.
Future Virol ; 10(10): 1145-1154, 2015 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-27066105

RESUMO

HSV type-1 and -2 are widespread pathogens producing lifelong infection with multiple sequelae, including oral, ocular and genital disease. The process of herpesvirus entry is a highly complex process involving numerous viral and cellular factors. Entry begins with attachment of virus to the cell surface followed by interactions between viral glycoproteins and cellular receptors to facilitate capsid penetration. The nucleocapsid is then transported along microtubules to the nuclear membrane, where viral DNA is released for replication in the nucleus. The work reviewed here comprises the most recent advancements in our understanding of the mechanism involved in the herpesvirus entry process.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...